Academic Appointments


Administrative Appointments


  • Director, Center for Translational Medicine, Stanford University School of Medicine (2019 - Present)
  • Chief, Division of Immunology and Rheumatology, Stanford University School of Medicine (2013 - 2019)

Current Research and Scholarly Interests


Autoimmunity
Chronic inflammatory disease
Metabolic control of immune function

2023-24 Courses


Stanford Advisees


Graduate and Fellowship Programs


All Publications


  • Stem-like CD4+ T cells in perivascular tertiary lymphoid structures sustain autoimmune vasculitis. Science translational medicine Sato, Y., Jain, A., Ohtsuki, S., Okuyama, H., Sturmlechner, I., Takashima, Y., Le, K. C., Bois, M. C., Berry, G. J., Warrington, K. J., Goronzy, J. J., Weyand, C. M. 2023; 15 (712): eadh0380

    Abstract

    Autoimmune vasculitis of the medium and large elastic arteries can cause blindness, stroke, aortic arch syndrome, and aortic aneurysm. The disease is often refractory to immunosuppressive therapy and progresses over decades as smoldering aortitis. How the granulomatous infiltrates in the vessel wall are maintained and how tissue-infiltrating T cells and macrophages are replenished are unknown. Single-cell and whole-tissue transcriptomic studies of immune cell populations in vasculitic arteries identified a CD4+ T cell population with stem cell-like features. CD4+ T cells supplying the tissue-infiltrating and tissue-damaging effector T cells survived in tertiary lymphoid structures around adventitial vasa vasora, expressed the transcription factor T cell factor 1 (TCF1), had high proliferative potential, and gave rise to two effector populations, Eomesodermin (EOMES)+ cytotoxic T cells and B cell lymphoma 6 (BCL6)+ T follicular helper-like cells. TCF1hiCD4+ T cells expressing the interleukin 7 receptor (IL-7R) sustained vasculitis in serial transplantation experiments. Thus, TCF1hiCD4+ T cells function as disease stem cells and promote chronicity and autonomy of autoimmune tissue inflammation. Remission-inducing therapies will require targeting stem-like CD4+ T cells instead of only effector T cells.

    View details for DOI 10.1126/scitranslmed.adh0380

    View details for PubMedID 37672564

  • Immunology of Giant Cell Arteritis. Circulation research Weyand, C. M., Goronzy, J. J. 2023; 132 (2): 238-250

    Abstract

    Giant cell arteritis is an autoimmune disease of medium and large arteries, characterized by granulomatous inflammation of the three-layered vessel wall that results in vaso-occlusion, wall dissection, and aneurysm formation. The immunopathogenesis of giant cell arteritis is an accumulative process in which a prolonged asymptomatic period is followed by uncontrolled innate immunity, a breakdown in self-tolerance, the transition of autoimmunity from the periphery into the vessel wall and, eventually, the progressive evolution of vessel wall inflammation. Each of the steps in pathogenesis corresponds to specific immuno-phenotypes that provide mechanistic insights into how the immune system attacks and damages blood vessels. Clinically evident disease begins with inappropriate activation of myeloid cells triggering the release of hepatic acute phase proteins and inducing extravascular manifestations, such as muscle pains and stiffness diagnosed as polymyalgia rheumatica. Loss of self-tolerance in the adaptive immune system is linked to aberrant signaling in the NOTCH pathway, leading to expansion of NOTCH1+CD4+ T cells and the functional decline of NOTCH4+ T regulatory cells (Checkpoint 1). A defect in the endothelial cell barrier of adventitial vasa vasorum networks marks Checkpoint 2; the invasion of monocytes, macrophages and T cells into the arterial wall. Due to the failure of the immuno-inhibitory PD-1 (programmed cell death protein 1)/PD-L1 (programmed cell death ligand 1) pathway, wall-infiltrating immune cells arrive in a permissive tissues microenvironment, where multiple T cell effector lineages thrive, shift toward high glycolytic activity, and support the development of tissue-damaging macrophages, including multinucleated giant cells (Checkpoint 3). Eventually, the vascular lesions are occupied by self-renewing T cells that provide autonomy to the disease process and limit the therapeutic effectiveness of currently used immunosuppressants. The multi-step process deviating protective to pathogenic immunity offers an array of interception points that provide opportunities for the prevention and therapeutic management of this devastating autoimmune disease.

    View details for DOI 10.1161/CIRCRESAHA.122.322128

    View details for PubMedID 36656970

  • Hyperactivity of the CD155 immune checkpoint suppresses anti-viral immunity in patients with coronary artery disease. Nature cardiovascular research Zhao, T. V., Hu, Z., Ohtsuki, S., Jin, K., Wu, B., Berry, G. J., Frye, R. L., Goronzy, J. J., Weyand, C. M. 2022; 1 (7): 634-648

    Abstract

    Pre-existent cardiovascular disease is a risk factor for weak anti-viral immunity, but underlying mechanisms remain undefined. Here, we report that patients with coronary artery disease (CAD) have macrophages (Mϕ) that actively suppress the induction of helper T cells reactive to two viral antigens: the SARS-CoV2 Spike protein and the Epstein-Barr virus (EBV) glycoprotein 350. CAD Mϕ overexpressed the methyltransferase METTL3, promoting the accumulation of N⁶-methyladenosine (m6A) in Poliovirus receptor (CD155) mRNA. m6A modifications of positions 1635 and 3103 in the 3'UTR of CD155 mRNA stabilized the transcript and enhanced CD155 surface expression. As a result, the patients' Mϕ abundantly expressed the immunoinhibitory ligand CD155 and delivered negative signals to CD4+ T cells expressing CD96 and/or TIGIT receptors. Compromised antigen-presenting function of METTL3hi CD155hi Mϕ diminished anti-viral T cell responses in vitro and in vivo. LDL and its oxidized form induced the immunosuppressive Mϕ phenotype. Undifferentiated CAD monocytes had hypermethylated CD155 mRNA, implicating post-transcriptional RNA modifications in the bone-marrow in shaping anti-viral immunity in CAD.

    View details for DOI 10.1038/s44161-022-00096-8

    View details for PubMedID 36860353

  • The transcription factor RFX5 coordinates antigen-presenting function and resistance to nutrient stress in synovial macrophages. Nature metabolism Hu, Z., Zhao, T. V., Huang, T., Ohtsuki, S., Jin, K., Goronzy, I. N., Wu, B., Abdel, M. P., Bettencourt, J. W., Berry, G. J., Goronzy, J. J., Weyand, C. M. 2022

    Abstract

    Tissue macrophages (Mϕ) are essential effector cells in rheumatoid arthritis (RA), contributing to autoimmune tissue inflammation through diverse effector functions. Their arthritogenic potential depends on their proficiency to survive in the glucose-depleted environment of the inflamed joint. Here, we identify a mechanism that links metabolic adaptation to nutrient stress with the efficacy of tissue Mϕ to activate adaptive immunity by presenting antigen to tissue-invading T cells. Specifically, Mϕ populating the rheumatoid joint produce and respond to the small cytokine CCL18, which protects against cell death induced by glucose withdrawal. Mechanistically, CCL18 induces the transcription factor RFX5 that selectively upregulates glutamate dehydrogenase 1 (GLUD1), thus enabling glutamate utilization to support energy production. In parallel, RFX5 enhances surface expression of HLA-DR molecules, promoting Mϕ-dependent expansion of antigen-specific T cells. These data place CCL18 at the top of a RFX5-GLUD1 survival pathway and couple adaptability to nutrient conditions in the tissue environment to antigen-presenting function in autoimmune tissue inflammation.

    View details for DOI 10.1038/s42255-022-00585-x

    View details for PubMedID 35739396

  • Mitochondrial aspartate regulates TNF biogenesis and autoimmune tissue inflammation. Nature immunology Wu, B., Zhao, T. V., Jin, K., Hu, Z., Abdel, M. P., Warrington, K. J., Goronzy, J. J., Weyand, C. M. 2021

    Abstract

    Misdirected immunity gives rise to the autoimmune tissue inflammation of rheumatoid arthritis, in which excess production of the cytokine tumor necrosis factor (TNF) is a central pathogenic event. Mechanisms underlying the breakdown of self-tolerance are unclear, but T cells in the arthritic joint have a distinctive metabolic signature of ATPlo acetyl-CoAhi proinflammatory effector cells. Here we show that a deficiency in the production of mitochondrial aspartate is an important abnormality in these autoimmune T cells. Shortage of mitochondrial aspartate disrupted the regeneration of the metabolic cofactor nicotinamide adenine dinucleotide, causing ADP deribosylation of the endoplasmic reticulum (ER) sensor GRP78/BiP. As a result, ribosome-rich ER membranes expanded, promoting co-translational translocation and enhanced biogenesis of transmembrane TNF. ERrich T cells were the predominant TNF producers in the arthritic joint. Transfer of intact mitochondria into T cells, as well as supplementation of exogenous aspartate, rescued the mitochondria-instructed expansion of ER membranes and suppressed TNF release and rheumatoid tissue inflammation.

    View details for DOI 10.1038/s41590-021-01065-2

    View details for PubMedID 34811544

  • The immunology of rheumatoid arthritis. Nature immunology Weyand, C. M., Goronzy, J. J. 2020

    Abstract

    The immunopathogenesis of rheumatoid arthritis (RA) spans decades, beginning with the production of autoantibodies against post-translationally modified proteins (checkpoint 1). After years of asymptomatic autoimmunity and progressive immune system remodeling, tissue tolerance erodes and joint inflammation ensues as tissue-invasive effector T cells emerge and protective joint-resident macrophages fail (checkpoint 2). The transition of synovial stromal cells into autoaggressive effector cells converts synovitis from acute to chronic destructive (checkpoint 3). The loss of T cell tolerance derives from defective DNA repair, causing abnormal cell cycle dynamics, telomere fragility and instability of mitochondrial DNA. Mitochondrial and lysosomal anomalies culminate in the generation of short-lived tissue-invasive effector T cells. This differentiation defect builds on a metabolic platform that shunts glucose away from energy generation toward the cell building and motility programs. The next frontier in RA is the development of curative interventions, for example, reprogramming T cell defects during the period of asymptomatic autoimmunity.

    View details for DOI 10.1038/s41590-020-00816-x

    View details for PubMedID 33257900

  • NOTCH-induced rerouting of endosomal trafficking disables regulatory T-cells in vasculitis. The Journal of clinical investigation Jin, K., Wen, Z., Wu, B., Zhang, H., Qiu, J., Wang, Y., Warrington, K. J., Berry, G., Goronzy, J. J., Weyand, C. M. 2020

    Abstract

    The aorta and the large conductive arteries are immunoprivileged tissues and are protected against inflammatory attack. A breakdown of the immunoprivilege leads to autoimmune vasculitis, such as giant cell arteritis (GCA), in which CD8+ T regulatory (Treg) cells fail to contain CD4+ T cells and macrophages, resulting in the formation of tissue-destructive granulomatous lesions. Here, we report that the molecular defect of malfunctioning CD8+ Treg cells lies in aberrant NOTCH4 signaling that deviates endosomal trafficking and minimizes exosome production. By transcriptionally controlling the profile of RAB GTPases, NOTCH4 signaling restricted membrane translocation and vesicular secretion of the enzyme NADPH oxidase 2 (NOX2). Specifically, NOTCH4hiCD8+ Treg cells increased RAB5A and RAB11A expression and suppressed RAB7A, culminating in the accumulation of early and recycling endosomes and trapping of NOX2 in an intracellular, non-secretory compartment. RAB7AloCD8+ Treg cells failed in the surface translocation and the exosomal release of NOX2. NOTCH4hi RAB5Ahi RAB7Alo RAB11Ahi CD8+ Treg cells left adaptive immunity unopposed, enabling a breakdown in tissue tolerance and aggressive vessel wall inflammation. Inhibiting NOTCH4 signaling corrected the defect and protected arteries from inflammatory insult. The study implicates NOTCH4-dependent transcriptional control of RAB proteins and intracellular vesicle trafficking in autoimmune disease and in vascular inflammation.

    View details for DOI 10.1172/JCI136042

    View details for PubMedID 32960812

  • Succinyl-CoA Ligase Deficiency in Pro-inflammatory and Tissue-Invasive T Cells. Cell metabolism Wu, B. n., Qiu, J. n., Zhao, T. V., Wang, Y. n., Maeda, T. n., Goronzy, I. N., Akiyama, M. n., Ohtsuki, S. n., Jin, K. n., Tian, L. n., Goronzy, J. J., Weyand, C. M. 2020; 32 (6): 967–80.e5

    Abstract

    Autoimmune T cells in rheumatoid arthritis (RA) have a defect in mitochondrial oxygen consumption and ATP production. Here, we identified suppression of the GDP-forming β subunit of succinate-CoA ligase (SUCLG2) as an underlying abnormality. SUCLG2-deficient T cells reverted the tricarboxylic acid (TCA) cycle from the oxidative to the reductive direction, accumulated α-ketoglutarate, citrate, and acetyl-CoA (AcCoA), and differentiated into pro-inflammatory effector cells. In AcCoAhi RA T cells, tubulin acetylation stabilized the microtubule cytoskeleton and positioned mitochondria in a perinuclear location, resulting in cellular polarization, uropod formation, T cell migration, and tissue invasion. In the tissue, SUCLG2-deficient T cells functioned as cytokine-producing effector cells and were hyperinflammatory, a defect correctable by replenishing the enzyme. Preventing T cell tubulin acetylation by tubulin acetyltransferase knockdown was sufficient to inhibit synovitis. These data link mitochondrial failure and AcCoA oversupply to autoimmune tissue inflammation.

    View details for DOI 10.1016/j.cmet.2020.10.025

    View details for PubMedID 33264602

  • N-myristoyltransferase deficiency impairs activation of kinase AMPK and promotes synovial tissue inflammation NATURE IMMUNOLOGY Wen, Z., Jin, K., Shen, Y., Yang, Z., Li, Y., Wu, B., Tian, L., Shoor, S., Roche, N. E., Goronzy, J. J., Weyand, C. M. 2019; 20 (3): 313-+
  • N-myristoyltransferase deficiency impairs activation of kinase AMPK and promotes synovial tissue inflammation. Nature immunology Wen, Z., Jin, K., Shen, Y., Yang, Z., Li, Y., Wu, B., Tian, L., Shoor, S., Roche, N. E., Goronzy, J. J., Weyand, C. M. 2019

    Abstract

    N-myristoyltransferase (NMT) attaches the fatty acid myristate to the N-terminal glycine of proteins to sort them into soluble and membrane-bound fractions. Function of the energy-sensing AMP-activated protein kinase, AMPK, is myristoylation dependent. In rheumatoid arthritis (RA), pathogenic T cells shift glucose away from adenosine tri-phosphate production toward synthetic and proliferative programs, promoting proliferation, cytokine production, and tissue invasion. We found that RA T cells had a defect in NMT1 function, which prevented AMPK activation and enabled unopposed mTORC1 signaling. Lack of the myristate lipid tail disrupted the lysosomal translocation and activation of AMPK. Instead, myristoylation-incompetent RA T cells hyperactivated the mTORC1 pathway and differentiated into pro-inflammatory TH1 and TH17 helper T cells. In vivo, NMT1 loss caused robust synovial tissue inflammation, whereas forced NMT1 overexpression rescued AMPK activation and suppressed synovitis. Thus, NMT1 has tissue-protective functions by facilitating lysosomal recruitment of AMPK and dampening mTORC1 signaling.

    View details for PubMedID 30718913

  • Glucose metabolism controls disease-specific signatures of macrophage effector functions JCI INSIGHT Watanabe, R., Hilhorst, M., Zhang, H., Zeisbrich, M., Berry, G. J., Wallis, B. B., Harrison, D. G., Giacomini, J. C., Goronzy, J. J., Weyand, C. M. 2018; 3 (20)
  • A Mitochondrial Checkpoint in Autoimmune Disease. Cell metabolism Weyand, C. M., Goronzy, J. J. 2018; 28 (2): 185–86

    Abstract

    Cell-fate decisions in innate and adaptive immune cells are metabolically regulated and modified by tissue-derived signals, thereby balancing protective and pathogenic immunity. Reporting in Science, Ling etal. (2018) have identified C1q as a mitochondrial activator that suppresses exuberant anti-self and anti-viral CD8 Tcell effector responses to prevent autoimmune disease.

    View details for PubMedID 30089238

  • Inhibition of JAK-STAT Signaling Suppresses Pathogenic Immune Responses in Medium and Large Vessel Vasculitis CIRCULATION Zhang, H., Watanabe, R., Berry, G. J., Tian, L., Goronzy, J. J., Weyand, C. M. 2018; 137 (18): 1934–48

    Abstract

    Giant cell arteritis, a chronic autoimmune disease of the aorta and its large branches, is complicated by aneurysm formation, dissection, and arterial occlusions. Arterial wall dendritic cells attract CD4+ T cells and macrophages to form prototypic granulomatous infiltrates. Vasculitic lesions contain a diverse array of effector T cells that persist despite corticosteroid therapy and sustain chronic, smoldering vasculitis. Transmural inflammation induces microvascular neoangiogenesis and results in lumen-occlusive intimal hyperplasia. We have examined whether persistent vessel wall inflammation is maintained by lesional T cells, including the newly identified tissue-resident memory T cells, and whether such T cells are sensitive to the cytokine-signaling inhibitor tofacitinib, a Janus kinase (JAK) inhibitor targeting JAK3 and JAK1.Vascular inflammation was induced in human arteries engrafted into immunodeficient mice that were reconstituted with T cells and monocytes from patients with giant cell arteritis. Mice carrying inflamed human arteries were treated with tofacitinib or vehicle. Vasculitic arteries were examined for gene expression (reverse transcription polymerase chain reaction), protein expression (immunohistochemistry), and infiltrating cell populations (flow cytometry).Tofacitinib effectively suppressed innate and adaptive immunity in the vessel wall. Lesional T cells responded to tofacitinib with reduced proliferation rates (<10%) and minimal production of the effector molecules interferon-γ, interleukin-17, and interleukin-21. Tofacitinib disrupted adventitial microvascular angiogenesis, reduced outgrowth of hyperplastic intima, and minimized CD4+CD103+ tissue-resident memory T cells.Cytokine signaling dependent on JAK3 and JAK1 is critically important in chronic inflammation of medium and large arteries. The JAK inhibitor tofacitinib effectively suppresses tissue-resident memory T cells and inhibits core vasculitogenic effector pathways.

    View details for PubMedID 29254929

    View details for PubMedCentralID PMC5930040

  • Immunoinhibitory checkpoint deficiency in medium and large vessel vasculitis PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Zhang, H., Watanabe, R., Berry, G. J., Vaglio, A., Liao, Y. J., Warrington, K. J., Goronzy, J. J., Weyand, C. M. 2017; 114 (6): E970-E979

    Abstract

    Giant cell arteritis (GCA) causes autoimmune inflammation of the aorta and its large branches, resulting in aortic arch syndrome, blindness, and stroke. CD4(+) T cells and macrophages form organized granulomatous lesions in the walls of affected arteries, destroy the tunica media, and induce ischemic organ damage through rapid intimal hyperplasia and luminal occlusion. Pathogenic mechanisms remain insufficiently understood; specifically, it is unknown whether the unopposed activation of the immune system is because of deficiency of immunoinhibitory checkpoints. Transcriptome analysis of GCA-affected temporal arteries revealed low expression of the coinhibitory ligand programmed death ligand-1 (PD-L1) concurrent with enrichment of the programmed death-1 (PD-1) receptor. Tissue-residing and ex vivo-generated dendritic cells (DC) from GCA patients were PD-L1(lo), whereas the majority of vasculitic T cells expressed PD-1, suggesting inefficiency of the immunoprotective PD-1/PD-L1 immune checkpoint. DC-PD-L1 expression correlated inversely with clinical disease activity. In human artery-SCID chimeras, PD-1 blockade exacerbated vascular inflammation, enriched for PD-1(+) effector T cells, and amplified tissue production of multiple T-cell effector cytokines, including IFN-γ, IL-17, and IL-21. Arteries infiltrated by PD-1(+) effector T cells developed microvascular neoangiogenesis as well as hyperplasia of the intimal layer, implicating T cells in the maladaptive behavior of vessel wall endogenous cells. Thus, in GCA, a breakdown of the tissue-protective PD1/PD-L1 checkpoint unleashes vasculitic immunity and regulates the pathogenic remodeling of the inflamed arterial wall.

    View details for DOI 10.1073/pnas.1616848114

    View details for Web of Science ID 000393422200011

    View details for PubMedID 28115719

    View details for PubMedCentralID PMC5307483

  • The microvascular niche instructs T cells in large vessel vasculitis via the VEGF-Jagged1-Notch pathway. Science translational medicine Wen, Z. n., Shen, Y. n., Berry, G. n., Shahram, F. n., Li, Y. n., Watanabe, R. n., Liao, Y. J., Goronzy, J. J., Weyand, C. M. 2017; 9 (399)

    Abstract

    Microvascular networks in the adventitia of large arteries control access of inflammatory cells to the inner wall layers (media and intima) and thus protect the immune privilege of the aorta and its major branches. In autoimmune vasculitis giant cell arteritis (GCA), CD4 T helper 1 (TH1) and TH17 cells invade into the wall of the aorta and large elastic arteries to form tissue-destructive granulomas. Whether the disease microenvironment provides instructive cues for vasculitogenic T cells is unknown. We report that adventitial microvascular endothelial cells (mvECs) perform immunoregulatory functions by up-regulating the expression of the Notch ligand Jagged1. Vascular endothelial growth factor (VEGF), abundantly present in GCA patients' blood, induced Jagged1 expression, allowing mvECs to regulate effector T cell induction via the Notch-mTORC1 (mammalian target of rapamycin complex 1) pathway. We found that circulating CD4 T cells in GCA patients have left the quiescent state, actively signal through the Notch pathway, and differentiate into TH1 and TH17 effector cells. In an in vivo model of large vessel vasculitis, exogenous VEGF functioned as an effective amplifier to recruit and activate vasculitogenic T cells. Thus, systemic VEGF co-opts endothelial Jagged1 to trigger aberrant Notch signaling, biases responsiveness of CD4 T cells, and induces pathogenic effector functions. Adventitial microvascular networks function as an instructive tissue niche, which can be exploited to target vasculitogenic immunity in large vessel vasculitis.

    View details for PubMedID 28724574

  • Metabolic control of the scaffold protein TKS5 in tissue-invasive, proinflammatory T cells. Nature immunology Shen, Y. n., Wen, Z. n., Li, Y. n., Matteson, E. L., Hong, J. n., Goronzy, J. J., Weyand, C. M. 2017; 18 (9): 1025–34

    Abstract

    Pathogenic T cells in individuals with rheumatoid arthritis (RA) infiltrate non-lymphoid tissue sites, maneuver through extracellular matrix and form lasting inflammatory microstructures. Here we found that RA T cells abundantly express the podosome scaffolding protein TKS5, which enables them to form tissue-invasive membrane structures. TKS5 overexpression was regulated by the intracellular metabolic environment of RA T cells-specifically, by reduced glycolytic flux that led to deficiencies in ATP and pyruvate. ATP(lo)pyruvate(lo) conditions triggered fatty acid biosynthesis and the formation of cytoplasmic lipid droplets. Restoration of pyruvate production or inhibition of fatty acid synthesis corrected the tissue-invasiveness of RA T cells in vivo and reversed their proarthritogenic behavior. Thus, metabolic control of T cell locomotion provides new opportunities to interfere with T cell invasion into specific tissue sites.

    View details for PubMedID 28737753

    View details for PubMedCentralID PMC5568495

  • Deficient Activity of the Nuclease MRE11A Induces T Cell Aging and Promotes Arthritogenic Effector Functions in Patients with Rheumatoid Arthritis. Immunity Li, Y., Shen, Y., Hohensinner, P., Ju, J., Wen, Z., Goodman, S. B., Zhang, H., Goronzy, J. J., Weyand, C. M. 2016; 45 (4): 903-916

    Abstract

    Immune aging manifests with a combination of failing adaptive immunity and insufficiently restrained inflammation. In patients with rheumatoid arthritis (RA), T cell aging occurs prematurely, but the mechanisms involved and their contribution to tissue-destructive inflammation remain unclear. We found that RA CD4(+) T cells showed signs of aging during their primary immune responses and differentiated into tissue-invasive, proinflammatory effector cells. RA T cells had low expression of the double-strand-break repair nuclease MRE11A, leading to telomeric damage, juxtacentromeric heterochromatin unraveling, and senescence marker upregulation. Inhibition of MRE11A activity in healthy T cells induced the aging phenotype, whereas MRE11A overexpression in RA T cells reversed it. In human-synovium chimeric mice, MRE11A(low) T cells were tissue-invasive and pro-arthritogenic, and MRE11A reconstitution mitigated synovitis. Our findings link premature T cell aging and tissue-invasiveness to telomere deprotection and heterochromatin unpacking, identifying MRE11A as a therapeutic target to combat immune aging and suppress dysregulated tissue inflammation.

    View details for DOI 10.1016/j.immuni.2016.09.013

    View details for PubMedID 27742546

  • NADPH oxidase deficiency underlies dysfunction of aged CD8(+) Tregs JOURNAL OF CLINICAL INVESTIGATION Wen, Z., Shimojima, Y., Shirai, T., Li, Y., Ju, J., Yang, Z., Tian, L., Goronzy, J. J., Weyand, C. M. 2016; 126 (5): 1953-1967

    Abstract

    Immune aging results in progressive loss of both protective immunity and T cell-mediated suppression, thereby conferring susceptibility to a combination of immunodeficiency and chronic inflammatory disease. Here, we determined that older individuals fail to generate immunosuppressive CD8+CCR7+ Tregs, a defect that is even more pronounced in the age-related vasculitic syndrome giant cell arteritis. In young, healthy individuals, CD8+CCR7+ Tregs are localized in T cell zones of secondary lymphoid organs, suppress activation and expansion of CD4 T cells by inhibiting the phosphorylation of membrane-proximal signaling molecules, and effectively inhibit proliferative expansion of CD4 T cells in vitro and in vivo. We identified deficiency of NADPH oxidase 2 (NOX2) as the molecular underpinning of CD8 Treg failure in the older individuals and in patients with giant cell arteritis. CD8 Tregs suppress by releasing exosomes that carry preassembled NOX2 membrane clusters and are taken up by CD4 T cells. Overexpression of NOX2 in aged CD8 Tregs promptly restored suppressive function. Together, our data support NOX2 as a critical component of the suppressive machinery of CD8 Tregs and suggest that repairing NOX2 deficiency in these cells may protect older individuals from tissue-destructive inflammatory disease, such as large-vessel vasculitis.

    View details for DOI 10.1172/JCI84181

    View details for Web of Science ID 000375182100029

    View details for PubMedID 27088800

    View details for PubMedCentralID PMC4855948

  • Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Science translational medicine Yang, Z., Shen, Y., Oishi, H., Matteson, E. L., Tian, L., Goronzy, J. J., Weyand, C. M. 2016; 8 (331): 331ra38-?

    Abstract

    In patients with rheumatoid arthritis (RA), CD4(+)T cells hyperproliferate during clonal expansion, differentiating into cytokine-producing effector cells that contribute to disease pathology. However, the metabolic underpinnings of this hyperproliferation remain unclear. In contrast to healthy T cells, naïve RA T cells had a defect in glycolytic flux due to the up-regulation of glucose-6-phosphate dehydrogenase (G6PD). Excess G6PD shunted glucose into the pentose phosphate pathway, resulting in NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) accumulation and reactive oxygen species (ROS) consumption. With surplus reductive equivalents, RA T cells insufficiently activated the redox-sensitive kinase ataxia telangiectasia mutated (ATM), bypassed the G2/M cell cycle checkpoint, and hyperproliferated. Moreover, insufficient ATM activation biased T cell differentiation toward the T helper 1 (TH1) and TH17 lineages, imposing a hyperinflammatory phenotype. We have identified several interventions that replenish intracellular ROS, which corrected the abnormal proliferative behavior of RA T cells and successfully suppressed synovial inflammation. Thus, rebalancing glucose utilization and restoring oxidant signaling may provide a therapeutic strategy to prevent autoimmunity in RA.

    View details for DOI 10.1126/scitranslmed.aad7151

    View details for PubMedID 27009267

  • The glycolytic enzyme PKM2 bridges metabolic and inflammatory dysfunction in coronary artery disease JOURNAL OF EXPERIMENTAL MEDICINE Shirai, T., Nazarewicz, R. R., Wallis, B. B., Yanes, R. E., Watanabe, R., Hilhorst, M., Tian, L., Harrison, D. G., Giacomini, J. C., Assimes, T. L., Goronzy, J. J., Weyand, C. M. 2016; 213 (3): 337-354

    Abstract

    Abnormal glucose metabolism and enhanced oxidative stress accelerate cardiovascular disease, a chronic inflammatory condition causing high morbidity and mortality. Here, we report that in monocytes and macrophages of patients with atherosclerotic coronary artery disease (CAD), overutilization of glucose promotes excessive and prolonged production of the cytokines IL-6 and IL-1β, driving systemic and tissue inflammation. In patient-derived monocytes and macrophages, increased glucose uptake and glycolytic flux fuel the generation of mitochondrial reactive oxygen species, which in turn promote dimerization of the glycolytic enzyme pyruvate kinase M2 (PKM2) and enable its nuclear translocation. Nuclear PKM2 functions as a protein kinase that phosphorylates the transcription factor STAT3, thus boosting IL-6 and IL-1β production. Reducing glycolysis, scavenging superoxide and enforcing PKM2 tetramerization correct the proinflammatory phenotype of CAD macrophages. In essence, PKM2 serves a previously unidentified role as a molecular integrator of metabolic dysfunction, oxidative stress and tissue inflammation and represents a novel therapeutic target in cardiovascular disease.

    View details for DOI 10.1084/jem.20150900

    View details for PubMedID 26926996

  • Blocking the NOTCH Pathway Inhibits Vascular Inflammation in Large-Vessel Vasculitis CIRCULATION Piggott, K., Deng, J., Warrington, K., Younge, B., Kubo, J. T., Desai, M., Goronzy, J. J., Weyand, C. M. 2011; 123 (3): 309-U180

    Abstract

    Giant cell arteritis is a granulomatous vasculitis of the aorta and its branches that causes blindness, stroke, and aortic aneurysm. CD4 T cells are key pathogenic regulators, instructed by vessel wall dendritic cells to differentiate into vasculitic T cells. The unique pathways driving this dendritic cell-T-cell interaction are incompletely understood, but may provide novel therapeutic targets for a disease in which the only established therapy is long-term treatment with high doses of corticosteroids.Immunohistochemical and gene expression analyses of giant cell arteritis-affected temporal arteries revealed abundant expression of the NOTCH receptor and its ligands, Jagged1 and Delta1. Cleavage of the NOTCH intracellular domain in wall-infiltrating T cells indicated ongoing NOTCH pathway activation in large-vessel vasculitis. NOTCH activation did not occur in small-vessel vasculitis affecting branches of the vasa vasorum tree. We devised 2 strategies to block NOTCH pathway activation: γ-secretase inhibitor treatment, preventing nuclear translocation of the NOTCH intracellular domain, and competing for receptor-ligand interactions through excess soluble ligand, Jagged1-Fc. In a humanized mouse model, NOTCH pathway disruption had strong immunosuppressive effects, inhibiting T-cell activation in the early and established phases of vascular inflammation. NOTCH inhibition was particularly effective in downregulating Th17 responses, but also markedly suppressed Th1 responses.Blocking NOTCH signaling depleted T cells from the vascular infiltrates, implicating NOTCH- NOTCH ligand interactions in regulating T-cell retention and survival in vessel wall inflammation. Modulating the NOTCH signaling cascade emerges as a promising new strategy for immunosuppressive therapy of large-vessel vasculitis.

    View details for DOI 10.1161/CIRCULATIONAHA.110.936203

    View details for Web of Science ID 000286507800018

    View details for PubMedID 21220737

    View details for PubMedCentralID PMC3056570

  • DNA-dependent protein kinase catalytic subunit mediates T-cell loss in rheumatoid arthritis EMBO MOLECULAR MEDICINE Shao, L., Goronzy, J. J., Weyand, C. M. 2010; 2 (10): 415-427

    Abstract

    In the autoimmune syndrome rheumatoid arthritis (RA), T cells and T-cell precursors have age-inappropriate shortening of telomeres and accumulate deoxyribonucleic acid (DNA) double strand breaks. Whether damaged DNA elicits DNA repair activity and how this affects T-cell function and survival is unknown. Here, we report that naïve and resting T cells from RA patients are susceptible to undergo apoptosis. In such T cells, unrepaired DNA stimulates a p53-ataxia telangiectasia mutated-independent pathway involving the non-homologous-end-joining protein DNA-protein kinase catalytic subunit (DNA-PKcs). Upregulation of DNA-PKcs transcription, protein expression and phosphorylation in RA T cells co-occurs with diminished expression of the Ku70/80 heterodimer, limiting DNA repair capacity. Inhibition of DNA-PKcs kinase activity or gene silencing of DNA-PKcs protects RA T cells from apoptosis. DNA-PKcs induces T-cell death by activating the JNK pathway and upregulating the apoptogenic BH3-only proteins Bim and Bmf. In essence, in RA, the DNA-PKcs-JNK-Bim/Bmf axis transmits genotoxic stress into shortened survival of naïve resting T cells, imposing chronic proliferative turnover of the immune system and premature immunosenescence. Therapeutic blockade of the DNA-PK-dependent cell-death machinery may rejuvenate the immune system in RA.

    View details for DOI 10.1002/emmm.201000096

    View details for Web of Science ID 000283891400005

    View details for PubMedID 20878914

    View details for PubMedCentralID PMC3017722

  • Th17 and Th1 T-Cell Responses in Giant Cell Arteritis CIRCULATION Deng, J., Younge, B. R., Olshen, R. A., Goronzy, J. J., Weyand, C. M. 2010; 121 (7): 906-U107

    Abstract

    In giant cell arteritis (GCA), vasculitic damage of the aorta and its branches is combined with a syndrome of intense systemic inflammation. Therapeutically, glucocorticoids remain the gold standard because they promptly and effectively suppress acute manifestations; however, they fail to eradicate vessel wall infiltrates. The effects of glucocorticoids on the systemic and vascular components of GCA are not understood.The immunoprofile of untreated and glucocorticoid-treated GCA was examined in peripheral blood and temporal artery biopsies with protein quantification assays, flow cytometry, quantitative real-time polymerase chain reaction, and immunohistochemistry. Plasma interferon-gamma and interleukin (IL)-17 and frequencies of interferon-gamma-producing and IL-17-producing T cells were markedly elevated before therapy. Glucocorticoid treatment suppressed the Th17 but not the Th1 arm in the blood and the vascular lesions. Analysis of monocytes/macrophages in the circulation and in temporal arteries revealed glucocorticoid-mediated suppression of Th17-promoting cytokines (IL-1beta, IL-6, and IL-23) but sparing of Th1-promoting cytokines (IL-12). In human artery-severe combined immunodeficiency mouse chimeras, in which patient-derived T cells cause inflammation of engrafted human temporal arteries, glucocorticoids were similarly selective in inhibiting Th17 cells and leaving Th1 cells unaffected.Two pathogenic pathways mediated by Th17 and Th1 cells contribute to the systemic and vascular manifestations of GCA. IL-17-producing Th17 cells are sensitive to glucocorticoid-mediated suppression, but interferon-gamma-producing Th1 responses persist in treated patients. Targeting steroid-resistant Th1 responses will be necessary to resolve chronic smoldering vasculitis. Monitoring Th17 and Th1 frequencies can aid in assessing disease activity in GCA.

    View details for DOI 10.1161/CIRCULATIONAHA.109.872903

    View details for Web of Science ID 000274797500011

    View details for PubMedID 20142449

    View details for PubMedCentralID PMC2837465

  • Immune aging - A mechanism in autoimmune disease. Seminars in immunology Zheng, Y., Liu, Q., Goronzy, J. J., Weyand, C. M. 2023; 69: 101814

    Abstract

    Evidence is emerging that the process of immune aging is a mechanism leading to autoimmunity. Over lifetime, the immune system adapts to profound changes in hematopoiesis and lymphogenesis, and progressively restructures in face of an ever-expanding exposome. Older adults fail to generate adequate immune responses against microbial infections and tumors, but accumulate aged T cells, B cells and myeloid cells. Age-associated B cells are highly efficient in autoantibody production. T-cell aging promotes the accrual of end-differentiated effector T cells with potent cytotoxic and pro-inflammatory abilities and myeloid cell aging supports a low grade, sterile and chronic inflammatory state (inflammaging). In pre-disposed individuals, immune aging can lead to frank autoimmune disease, manifesting with chronic inflammation and irreversible tissue damage. Emerging data support the concept that autoimmunity results from aging-induced failure of fundamental cellular processes in immune effector cells: genomic instability, loss of mitochondrial fitness, failing proteostasis, dwindling lysosomal degradation and inefficient autophagy. Here, we have reviewed the evidence that malfunctional mitochondria, disabled lysosomes and stressed endoplasmic reticula induce pathogenic T cells and macrophages that drive two autoimmune diseases, rheumatoid arthritis (RA) and giant cell arteritis (GCA). Recognizing immune aging as a risk factor for autoimmunity will open new avenues of immunomodulatory therapy, including the repair of malfunctioning mitochondria and lysosomes.

    View details for DOI 10.1016/j.smim.2023.101814

    View details for PubMedID 37542986

  • Deficiency of the CD155-CD96 immune checkpoint controls IL-9 production in giant cell arteritis. Cell reports. Medicine Ohtsuki, S., Wang, C., Watanabe, R., Zhang, H., Akiyama, M., Bois, M. C., Maleszewski, J. J., Warrington, K. J., Berry, G. J., Goronzy, J. J., Weyand, C. M. 2023; 4 (4): 101012

    Abstract

    Loss of function of inhibitory immune checkpoints, unleashing pathogenic immune responses, is a potential risk factor for autoimmune disease. Here, we report that patients with the autoimmune vasculitis giant cell arteritis (GCA) have a defective CD155-CD96 immune checkpoint. Macrophages from patients with GCA retain the checkpoint ligand CD155 in the endoplasmic reticulum (ER) and fail to bring it to the cell surface. CD155low antigen-presenting cells induce expansion of CD4+CD96+ T cells, which become tissue invasive, accumulate in the blood vessel wall, and release the effector cytokine interleukin-9 (IL-9). In a humanized mouse model of GCA, recombinant human IL-9 causes vessel wall destruction, whereas anti-IL-9 antibodies efficiently suppress innate and adaptive immunity in the vasculitic lesions. Thus, defective surface translocation of CD155 creates antigen-presenting cells that deviate T cell differentiation toward Th9 lineage commitment and results in the expansion of vasculitogenic effector T cells.

    View details for DOI 10.1016/j.xcrm.2023.101012

    View details for PubMedID 37075705

  • CISH impairs lysosomal function in activated T cells resulting in mitochondrial DNA release and inflammaging. Nature aging Jin, J., Mu, Y., Zhang, H., Sturmlechner, I., Wang, C., Jadhav, R. R., Xia, Q., Weyand, C. M., Goronzy, J. J. 2023

    Abstract

    Chronic systemic inflammation is one of the hallmarks of the aging immune system. Here we show that activated T cells from older adults contribute to inflammaging by releasing mitochondrial DNA (mtDNA) into their environment due to an increased expression of the cytokine-inducible SH2-containing protein (CISH). CISH targets ATP6V1A, an essential component of the proton pump V-ATPase, for proteasomal degradation, thereby impairing lysosomal function. Impaired lysosomal activity caused intracellular accumulation of multivesicular bodies and amphisomes and the export of their cargos, including mtDNA. CISH silencing in T cells from older adults restored lysosomal activity and prevented amphisomal release. In antigen-specific responses in vivo, CISH-deficient CD4+ T cells released less mtDNA and induced fewer inflammatory cytokines. Attenuating CISH expression may present a promising strategy to reduce inflammation in an immune response of older individuals.

    View details for DOI 10.1038/s43587-023-00399-w

    View details for PubMedID 37118554

    View details for PubMedCentralID 7584388

  • TRIB2 safeguards naive Tcell homeostasis during aging. Cell reports Cao, W., Sturmlechner, I., Zhang, H., Jin, J., Hu, B., Jadhav, R. R., Fang, F., Weyand, C. M., Goronzy, J. J. 2023; 42 (3): 112195

    Abstract

    Naive CD4+ Tcells are more resistant to age-related loss than naive CD8+ Tcells, suggesting mechanisms that preferentially protect naive CD4+ Tcells during aging. Here, we show that TRIB2 is more abundant in naive CD4+ than CD8+ Tcells and counteracts quiescence exit by suppressing AKT activation. TRIB2 deficiency increases AKT activity and accelerates proliferation and differentiation in response to interleukin-7 (IL-7) in humans and during lymphopenia in mice. TRIB2 transcription is controlled by the lineage-determining transcription factors ThPOK and RUNX3. Ablation of Zbtb7b (encoding ThPOK) and Cbfb (obligatory RUNT cofactor) attenuates the difference in lymphopenia-induced proliferation between naive CD4+ and CD8+ cells. In older adults, ThPOK and TRIB2 expression wanes in naive CD4+ Tcells, causing loss of naivety. These findings assign TRIB2 a key role in regulating Tcell homeostasis and provide a model to explain the lesser resilience of CD8+ Tcells to undergo changes with age.

    View details for DOI 10.1016/j.celrep.2023.112195

    View details for PubMedID 36884349

  • Aging-associated HELIOS deficiency in naive CD4+ T cells alters chromatin remodeling and promotes effector cell responses. Nature immunology Zhang, H., Jadhav, R. R., Cao, W., Goronzy, I. N., Zhao, T. V., Jin, J., Ohtsuki, S., Hu, Z., Morales, J., Greenleaf, W. J., Weyand, C. M., Goronzy, J. J. 2022

    Abstract

    Immune aging combines cellular defects in adaptive immunity with the activation of pathways causing a low-inflammatory state. Here we examined the influence of age on the kinetic changes in the epigenomic and transcriptional landscape induced by T cell receptor (TCR) stimulation in naive CD4+ T cells. Despite attenuated TCR signaling in older adults, TCR activation accelerated remodeling of the epigenome and induced transcription factor networks favoring effector cell differentiation. We identified increased phosphorylation of STAT5, at least in part due to aberrant IL-2 receptor and lower HELIOS expression, as upstream regulators. Human HELIOS-deficient, naive CD4+ T cells, when transferred into human-synovium-mouse chimeras, infiltrated tissues more efficiently. Inhibition of IL-2 or STAT5 activity in T cell responses of older adults restored the epigenetic response pattern to the one seen in young adults. In summary, reduced HELIOS expression in non-regulatory naive CD4+ T cells in older adults directs T cell fate decisions toward inflammatory effector cells that infiltrate tissue.

    View details for DOI 10.1038/s41590-022-01369-x

    View details for PubMedID 36510022

    View details for PubMedCentralID 7494270

  • Mitochondria as disease-relevant organelles in rheumatoid arthritis. Clinical and experimental immunology Weyand, C. M., Wu, B., Huang, T., Hu, Z., Goronzy, J. J. 2022

    Abstract

    Mitochondria are the controllers of cell metabolism and are recognized as decision makers in cell death pathways, organizers of cytoplasmic signaling networks, managers of cellular stress responses and regulators of nuclear gene expression. Cells of the immune system are particularly dependent on mitochondrial resources, as they must swiftly respond to danger signals with activation, trafficking, migration, and generation of daughter cells. Analogously, faulty immune responses that lead to autoimmunity and tissue inflammation rely on mitochondria to supply energy, cell building blocks and metabolic intermediates. Emerging data endorse the concept that mitochondrial fitness, and the lack of it, is of particular relevance in the autoimmune disease rheumatoid arthritis (RA) where deviations of bioenergetic and biosynthetic flux affect T cells during early and late stages of disease. During early stages of RA, mitochondrial deficiency allows naïve RA T cells to lose self-tolerance, biasing fundamental choices of the immune system towards immune-mediated tissue damage and away from host protection. During late stages of RA, mitochondrial abnormalities shape the response patterns of RA effector T cells engaged in the inflammatory lesions, enabling chronicity of tissue damage and tissue remodeling. In the inflamed joint, autoreactive T cells partner with metabolically reprogrammed tissue macrophages that specialize in antigen-presentation and survive by adapting to the glucose-deplete tissue microenvironment. Here, we summarize recent data on dysfunctional mitochondria and mitochondria-derived signals relevant in the RA disease process that offer novel opportunities to deter autoimmune tissue inflammation by metabolic interference.

    View details for DOI 10.1093/cei/uxac107

    View details for PubMedID 36420636

  • T cell aging as a risk factor for autoimmunity. Journal of autoimmunity Liu, Q., Zheng, Y., Goronzy, J. J., Weyand, C. M. 2022: 102947

    Abstract

    Immune aging is a complex process rendering the host susceptible to cancer, infection, and insufficient tissue repair. Many autoimmune diseases preferentially occur during the second half of life, counterintuitive to the concept of excess adaptive immunity driving immune-mediated tissue damage. T cells are particularly susceptible to aging-imposed changes, as they are under extreme proliferative pressure to fulfill the demands of clonal expansion and of homeostatic T cell repopulation. T cells in older adults have a footprint of genetic and epigenetic changes, lack mitochondrial fitness, and fail to maintain proteostasis, diverging them from host protection to host injury. Here, we review recent progress in understanding how the human T-cell system ages and the evidence detailing how T cell aging contributes to autoimmune conditions. T cell aging is now recognized as a risk determinant in two prototypic autoimmune syndromes; rheumatoid arthritis and giant cell arteritis. The emerging concept adds susceptibility to autoimmune and autoinflammatory disease to the spectrum of aging-imposed adaptations and opens new opportunities for immunomodulatory therapy by restoring the functional intactness of aging T cells.

    View details for DOI 10.1016/j.jaut.2022.102947

    View details for PubMedID 36357240

  • Immune Responses and Disease Marker Changes Following SARS-CoV-2 mRNA Vaccination in a Cohort of Rheumatic Disease Patients An, Z., Zhou, X., Li, Y., Jaquith, J., McCarthy-Fruin, K., Warrington, K. J., Duarte-Garcia, A., Thanarajasingam, U., Weyand, C. M., Zeng, H. WILEY. 2022: 1501-1502
  • IL-4 prevents adenosine-mediated immunoregulation by inhibiting CD39 expression. JCI insight Fang, F., Cao, W., Mu, Y., Okuyama, H., Li, L., Qiu, J., Weyand, C. M., Goronzy, J. J. 2022; 7 (12)

    Abstract

    The ectonucleotidase CD39 functions as a checkpoint in purinergic signaling on effector T cells. By depleting eATP and initiating the generation of adenosine, it impairs memory cell development and contributes to T cell exhaustion, thereby causing defective tumor immunity and deficient T cell responses in older adults who have increased CD39 expression. Tuning enzymatic activity of CD39 and targeting the transcriptional regulation of ENTPD1 can be used to modulate purinergic signaling. Here, we describe that STAT6 phosphorylation downstream of IL-4 signaling represses CD39 expression on activated T cells by inducing a transcription factor network including GATA3, GFI1, and YY1. GATA3 suppresses ENTPD1 transcription through prevention of RUNX3 recruitment to the ENTPD1 promoter. Conversely, pharmacological STAT6 inhibition decreases T cell effector functions via increased CD39 expression, resulting in the defective signaling of P2X receptors by ATP and stimulation of A2A receptors by adenosine. Our studies suggest that inhibiting the STAT6 pathway to increase CD39 expression has the potential to treat autoimmune disease while stimulation of the pathway could improve T cell immunity.

    View details for DOI 10.1172/jci.insight.157509

    View details for PubMedID 35730568

  • Reduced chromatin accessibility to CD4 T cell super-enhancers encompassing susceptibility loci of rheumatoid arthritis. EBioMedicine Jadhav, R. R., Hu, B., Ye, Z., Sheth, K., Li, X., Greenleaf, W. J., Weyand, C. M., Goronzy, J. J. 1800; 76: 103825

    Abstract

    BACKGROUND: Rheumatoid arthritis (RA) is an inflammatory disease that manifests as a preclinical stage of systemic autoimmunity followed by chronic progressive synovitis. Disease-associated genetic SNP variants predominantly map to non-coding, regulatory regions of functional importance in CD4 T cells, implicating these cells as key regulators. A better understanding of the epigenome of CD4 T cells holds the promise of providing information on the interaction between genetic susceptibility and exogenous factors.METHODS: We mapped regions of chromatin accessibility using ATAC-seq in peripheral CD4 T cell subsets of patients with RA (n=18) and compared them to T cells from patients with psoriatic arthritis (n=11) and age-matched healthy controls (n=10). Transcripts of selected genes were quantified using qPCR.FINDINGS: RA-associated epigenetic signatures were identified that in part overlapped between central and effector memory CD4 T cells and that were to a lesser extent already present in naive cells. Sites more accessible in RA were highly enriched for the motif of the transcription factor (TF) CTCF suggesting differences in the three-dimensional chromatin structure. Unexpectedly, sites with reduced chromatin accessibility were enriched for motifs of TFs pertinent for T cell function. Most strikingly, super-enhancers encompassing RA-associated SNPs were less accessible. Analysis of selected transcripts and published DNA methylation patterns were consistent with this finding. The preferential loss in accessibility at these super-enhancers was seen in patients with high and low disease activity and on a variety of immunosuppressive treatment modalities.INTERPRETATION: Disease-associated genes are epigenetically less poised to respond in CD4 T cells from patients with established RA.FUNDING: This work was supported by I01 BX001669 from the Veterans Administration.

    View details for DOI 10.1016/j.ebiom.2022.103825

    View details for PubMedID 35085847

  • Large-vessel vasculitis. Nature reviews. Disease primers Pugh, D., Karabayas, M., Basu, N., Cid, M. C., Goel, R., Goodyear, C. S., Grayson, P. C., McAdoo, S. P., Mason, J. C., Owen, C., Weyand, C. M., Youngstein, T., Dhaun, N. 1800; 7 (1): 93

    Abstract

    Large-vessel vasculitis (LVV) manifests as inflammation of the aorta and its major branches and is the most common primary vasculitis in adults. LVV comprises two distinct conditions, giant cell arteritis and Takayasu arteritis, although the phenotypic spectrum of primary LVV is complex. Non-specific symptoms often predominate and so patients with LVV present to a range of health-care providers and settings. Rapid diagnosis, specialist referral and early treatment are key to good patient outcomes. Unfortunately, disease relapse remains common and chronic vascular complications are a source of considerable morbidity. Although accurate monitoring of disease activity is challenging, progress in vascular imaging techniques and the measurement of laboratory biomarkers may facilitate better matching of treatment intensity with disease activity. Further, advances in our understanding of disease pathophysiology have paved the way for novel biologic treatments that target important mediators of disease in both giant cell arteritis and Takayasu arteritis. This work has highlighted the substantial heterogeneity present within LVV and the importance of an individualized therapeutic approach. Future work will focus on understanding the mechanisms of persisting vascular inflammation, which will inform the development of increasingly sophisticated imaging technologies. Together, these will enable better disease prognostication, limit treatment-associated adverse effects, and facilitate targeted development and use of novel therapies.

    View details for DOI 10.1038/s41572-021-00327-5

    View details for PubMedID 34992251

  • Age as a risk factor in vasculitis. Seminars in immunopathology Gloor, A. D., Berry, G. J., Goronzy, J. J., Weyand, C. M. 2022

    Abstract

    Two vasculitides, giant cell arteritis (GCA) and Takayasu arteritis (TAK), are recognized as autoimmune and autoinflammatory diseases that manifest exclusively within the aorta and its large branches. In both entities, the age of the affected host is a critical risk factor. TAK manifests during the 2nd-4th decade of life, occurring while the immune system is at its height of performance. GCA is a disease of older individuals, with infrequent cases during the 6th decade and peak incidence during the 8th decade of life. In both vasculitides, macrophages and T cells infiltrate into the adventitia and media of affected vessels, induce granulomatous inflammation, cause vessel wall destruction, and reprogram vascular cells to drive adventitial and neointimal expansion. In GCA, abnormal immunity originates in an aged immune system and evolves within the aged vascular microenvironment. One hallmark of the aging immune system is the preferential loss of CD8+ T cell function. Accordingly, in GCA but not in TAK, CD8+ effector T cells play a negligible role and anti-inflammatory CD8+ T regulatory cells are selectively impaired. Here, we review current evidence of how the process of immunosenescence impacts the risk for GCA and how fundamental differences in the age of the immune system translate into differences in the granulomatous immunopathology of TAK versus GCA.

    View details for DOI 10.1007/s00281-022-00911-1

    View details for PubMedID 35141865

  • The influence of three-dimensional structure on naive T cell homeostasis and aging. Frontiers in aging Lambert, S., Cao, W., Zhang, H., Colville, A., Liu, J., Weyand, C. M., Goronzy, J. J., Gustafson, C. E. 2022; 3: 1045648

    Abstract

    A breakdown in cellular homeostasis is thought to drive naive T cell aging, however the link between naive T cell homeostasis and aging in humans is poorly understood. To better address this, we developed a lymphoid organoid system that maintains resting naive T cells for more than 2weeks, in conjunction with high CD45RA expression. Deep phenotypic characterization of naive T cells across age identified reduced CD45RA density as a hallmark of aging. A conversion from CD45RAhigh naive cells to a CD45RAlow phenotype was reproduced within our organoid system by structural breakdown, but not by stromal cell aging or reduced lymphocyte density, and mediated by alternative CD45 splicing. Together, these data suggest that external influences within the lymph node microenvironment may cause phenotypic conversion of naive T cells in older adults.

    View details for DOI 10.3389/fragi.2022.1045648

    View details for PubMedID 36419548

  • T-Cell Aging-Associated Phenotypes in Autoimmune Disease. Frontiers in aging Zhao, T. V., Sato, Y., Goronzy, J. J., Weyand, C. M. 2022; 3: 867950

    Abstract

    The aging process causes profound restructuring of the host immune system, typically associated with declining host protection against cancer and infection. In the case of T cells, aging leads to the accumulation of a diverse set of T-cell aging-associated phenotypes (TASP), some of which have been implicated in driving tissue inflammation in autoimmune diseases. T cell aging as a risk determinant for autoimmunity is exemplified in two classical autoimmune conditions: rheumatoid arthritis (RA), a disease predominantly affecting postmenopausal women, and giant cell arteritis (GCA), an inflammatory vasculopathy exclusively occurring during the 6th-9th decade of life. Pathogenic T cells in RA emerge as a consequence of premature immune aging. They have shortening and fragility of telomeric DNA ends and instability of mitochondrial DNA. As a result, they produce a distinct profile of metabolites, disproportionally expand their endoplasmic reticulum (ER) membranes and release excess amounts of pro-inflammatory effector cytokines. Characteristically, they are tissue invasive, activate the inflammasome and die a pyroptotic death. Patients with GCA expand pathogenic CD4+ T cells due to aberrant expression of the co-stimulatory receptor NOTCH1 and the failure of the PD-1/PD-L1 immune checkpoint. In addition, GCA patients lose anti-inflammatory Treg cells, promoting tissue-destructive granulomatous vasculitis. In summary, emerging data identify T cell aging as a risk factor for autoimmune disease and directly link TASPs to the breakdown of T cell tolerance and T-cell-induced tissue inflammation.

    View details for DOI 10.3389/fragi.2022.867950

    View details for PubMedID 35821833

  • Regulatory T Cells in Autoimmune Vasculitis. Frontiers in immunology Jin, K., Parreau, S., Warrington, K. J., Koster, M. J., Berry, G. J., Goronzy, J. J., Weyand, C. M. 2022; 13: 844300

    Abstract

    Blood vessels are indispensable for host survival and are protected from inappropriate inflammation by immune privilege. This protection is lost in patients with autoimmune vasculitides, a heterogeneous group of diseases causing damage to arteries, arterioles, and capillaries. Vasculitis leads to vascular wall destruction and/or luminal occlusion, resulting in hemorrhage and tissue ischemia. Failure in the quantity and quality of immunosuppressive regulatory T cells (Treg) has been implicated in the breakdown of the vascular immune privilege. Emerging data suggest that Treg deficiencies are disease-specific, affecting distinct pathways in distinct vasculitides. Mechanistic studies have identified faulty CD8+ Tregs in Giant Cell Arteritis (GCA), a vasculitis of the aorta and the large aortic branch vessels. Specifically, aberrant signaling through the NOTCH4 receptor expressed on CD8+ Treg cells leads to rerouting of intracellular vesicle trafficking and failure in the release of immunosuppressive exosomes, ultimately boosting inflammatory attack to medium and large arteries. In Kawasaki's disease, a medium vessel vasculitis targeting the coronary arteries, aberrant expression of miR-155 and dysregulated STAT5 signaling have been implicated in undermining CD4+ Treg function. Explorations of mechanisms leading to insufficient immunosuppression and uncontrolled vascular inflammation hold the promise to discover novel therapeutic interventions that could potentially restore the immune privilege of blood vessels and pave the way for urgently needed innovations in vasculitis management.

    View details for DOI 10.3389/fimmu.2022.844300

    View details for PubMedID 35296082

  • The cell-surface 5'-nucleotidase CD73 defines a functional T memory cell subset that declines with age. Cell reports Fang, F., Cao, W., Zhu, W., Lam, N., Li, L., Gaddam, S., Wang, Y., Kim, C., Lambert, S., Zhang, H., Hu, B., Farber, D. L., Weyand, C. M., Goronzy, J. J. 2021; 37 (6): 109981

    Abstract

    Memory Tcells exhibit considerable diversity that determines their ability to be protective. Here, we examine whether changes in Tcell heterogeneity contribute to the age-associated failure of immune memory. By screening for age-dependent Tcell-surface markers, we identify CD4 and CD8 memory Tcell subsets that are unrelated to previously defined subsets of central and effector memory cells. Memory Tcells expressing the ecto-5'-nucleotidase CD73 constitute a functionally distinct subset of memory Tcells that declines with age. They resemble long-lived, polyfunctional memory cells but are also poised to display effector functions and to develop into cells resembling tissue-resident memory Tcells (TRMs). Upstream regulators of differential chromatin accessibility and transcriptomes include transcription factors that facilitate CD73 expression and regulate TRM differentiation. CD73 is not just a surrogate marker of these regulatory networks but is directly involved in Tcell survival.

    View details for DOI 10.1016/j.celrep.2021.109981

    View details for PubMedID 34758299

  • Immune And Inflammatory Mechanisms Mediate Cardiovascular Diseases From Head To Toe. Cardiovascular research Libby, P., Mallat, Z., Weyand, C. 2021

    Abstract

    Practitioners have long recognized the involvement of inflammation in certain acute cardiovascular diseases such as endocarditis, myocarditis, and pericarditis. Attention to the participation of immune and inflammatory mechanisms in chronic cardiovascular diseases has generally lagged. Yet, these pathways contribute to a broad swath of clinically important cardiovascular conditions, both acute and chronic. Understanding of these complex mechanisms can aid specialists in cardiovascular research and practice immeasurably by providing new concepts and illuminating new diagnostic and therapeutic strategies. The collection of essays presented in this focused issue of Cardiovascular Research aims to promote this goal.

    View details for DOI 10.1093/cvr/cvab332

    View details for PubMedID 34698765

  • Understanding T cell aging to improve anti-viral immunity. Current opinion in virology Zhang, H., Weyand, C. M., Goronzy, J. J., Gustafson, C. E. 2021; 51: 127-133

    Abstract

    T cells are a critical component of the immune system and required for protection against viral and bacterial infections. However, the capacity of these cells to provide sufficient protection declines with age, leading to an increased susceptibility to and mortality from infection in older individuals. In many cases, it also contributes to poor vaccine-induced immunity. Understanding the basic biology behind T cell aging is key to unraveling these defects and, in turn, designing more effective vaccines and therapeutics for the older population. Here, we will discuss recent studies that have provided significant insight into the features of T cell aging, how these features may contribute to poor immune responses with advancing age and newer avenues of research that may further enhance anti-viral immunity in older individuals.

    View details for DOI 10.1016/j.coviro.2021.09.017

    View details for PubMedID 34688983

  • Activation of mTORC1 at late endosomes misdirects T cell fate decision in older individuals. Science immunology Jin, J., Kim, C., Xia, Q., Gould, T. M., Cao, W., Zhang, H., Li, X., Weiskopf, D., Grifoni, A., Sette, A., Weyand, C. M., Goronzy, J. J. 2021; 6 (60)

    Abstract

    The nutrient-sensing mammalian target of rapamycin (mTOR) is integral to cell fate decisions after T cell activation. Sustained mTORC1 activity favors the generation of terminally differentiated effector T cells instead of follicular helper and memory T cells. This is particularly pertinent for T cell responses of older adults who have sustained mTORC1 activation despite dysfunctional lysosomes. Here, we show that lysosome-deficient T cells rely on late endosomes rather than lysosomes as an mTORC1 activation platform, where mTORC1 is activated by sensing cytosolic amino acids. T cells from older adults have an increased expression of the plasma membrane leucine transporter SLC7A5 to provide a cytosolic amino acid source. Hence, SLC7A5 and VPS39 deficiency (a member of the HOPS complex promoting early to late endosome conversion) substantially reduced mTORC1 activities in T cells from older but not young individuals. Late endosomal mTORC1 is independent of the negative-feedback loop involving mTORC1-induced inactivation of the transcription factor TFEB that controls expression of lysosomal genes. The resulting sustained mTORC1 activation impaired lysosome function and prevented lysosomal degradation of PD-1 in CD4+ T cells from older adults, thereby inhibiting their proliferative responses. VPS39 silencing of human T cells improved their expansion to pertussis and to SARS-CoV-2 peptides in vitro. Furthermore, adoptive transfer of CD4+ Vps39-deficient LCMV-specific SMARTA cells improved germinal center responses, CD8+ memory T cell generation, and recall responses to infection. Thus, curtailing late endosomal mTORC1 activity is a promising strategy to enhance T cell immunity.

    View details for DOI 10.1126/sciimmunol.abg0791

    View details for PubMedID 34145066

  • miR-181a-regulated pathways in T-cell differentiation and aging. Immunity & ageing : I & A Kim, C., Ye, Z., Weyand, C. M., Goronzy, J. J. 2021; 18 (1): 28

    Abstract

    MicroRNAs (miRNAs) are regulatory noncoding RNAs important for many aspects of cellular processes including cell differentiation and proliferation. Functions of numerous miRNAs have been identified in T cells, with miR-181a regulating T cell activation thresholds during thymic T cell development and during activation of peripheral T cells. Intriguingly, miR-181a is implicated in defective antiviral and vaccine responses in older individuals, as its expression declines in naive T cells with increasing age. Here, we review the pathways that are regulated by miR-181a and that explain the unique role of miR-181a in T cell development, T cell activation and antiviral T cell responses. These studies provide a framework for understanding how a decline in miR-181a expression in T cells could contribute to age-related defects in adaptive immunity. We furthermore review the mechanisms that cause the age-related decline in miR-181a expression and discuss the potential of restoring miR-181a expression or targeting miR-181a-regulated pathways to improve impaired T cell responses in older individuals.

    View details for DOI 10.1186/s12979-021-00240-1

    View details for PubMedID 34130717

  • Influence of Lymph Node Microenvironment on Naive T cell Aging Gustafson, C. E., Liu, J., Colville, A. J., Cao, W., Zhang, H., Weyand, C. M., Goronzy, J. J. AMER ASSOC IMMUNOLOGISTS. 2021
  • Hallmarks of the aging T cell system. The FEBS journal Zhang, H., Weyand, C. M., Goronzy, J. J. 2021

    Abstract

    The adaptive immune system has the enormous challenge to protect the host through the generation and differentiation of pathogen-specific short-lived effector T cells while in parallel developing long-lived memory cells to control future encounters with the same pathogen. A complex regulatory network is needed to preserve a population of naive cells over lifetime that exhibit sufficient diversity of antigen receptors to respond to new antigens, while also sustaining immune memory. In parallel, cells need to maintain their proliferative potential and the plasticity to differentiate into different functional lineages. Initial signs of waning immune competence emerge after 50 years of age, with increasing clinical relevance in the 7th -10th decade of life. Morbidity and mortality from infections increase, as drastically exemplified by the current COVID-19 pandemic. Many vaccines, such as for the influenza virus, are poorly effective to generate protective immunity in older individuals. Age-associated changes occur at the level of the T cell population as well as the functionality of its cellular constituents. The system highly relies on the self-renewal of naive and memory T cells, which is robust but eventually fails. Genetic and epigenetic modifications contribute to functional differences in responsiveness and differentiation potential. To some extent, these changes arise from defective maintenance; to some, they represent successful, but not universally beneficial adaptations to the aging host. Interventions that can compensate for the age-related defects and improve immune responses in older adults are increasingly within reach.

    View details for DOI 10.1111/febs.15770

    View details for PubMedID 33590946

  • The GSK3beta-beta-catenin-TCF1 pathway improves naive T cell activation in old adults by upregulating miR-181a. NPJ aging and mechanisms of disease Ye, Z., Gould, T. M., Zhang, H., Jin, J., Weyand, C. M., Goronzy, J. J. 2021; 7 (1): 4

    Abstract

    MicroRNAs play an important role in the regulation of T cell development, activation, and differentiation. One of the most abundant microRNAs in lymphocytes is miR-181a, which controls T cell receptor (TCR) activation thresholds in thymic selection as well as in peripheral T cell responses. We previously found that miR-181a levels decline in T cells in the elderly. In this study, we identified TCF1 as a transcriptional regulator of pri-miR-181a. A decline in TCF1 levels in old individuals accounted for the reduced miR-181a expression impairing TCR signaling. Inhibition of GSK3SS restored expression of miR-181a by inducing TCF1 in T cells from old adults. GSK3SS inhibition enhanced TCR signaling to increase downstream expression of activation markers and production of IL-2. The effect involved the upregulation of miR-181a and the inhibition of DUSP6 expression. Thus, inhibition of GSK3SS can restore responses of old T cells by inducing miR-181a expression through TCF1.

    View details for DOI 10.1038/s41514-021-00056-9

    View details for PubMedID 33558531

  • Metabolic Control of Autoimmunity and Tissue Inflammation in Rheumatoid Arthritis. Frontiers in immunology Qiu, J., Wu, B., Goodman, S. B., Berry, G. J., Goronzy, J. J., Weyand, C. M. 2021; 12: 652771

    Abstract

    Like other autoimmune diseases, rheumatoid arthritis (RA) develops in distinct stages, with each phase of disease linked to immune cell dysfunction. HLA class II genes confer the strongest genetic risk to develop RA. They encode for molecules essential in the activation and differentiation of T cells, placing T cells upstream in the immunopathology. In Phase 1 of the RA disease process, T cells lose a fundamental function, their ability to be self-tolerant, and provide help for autoantibody-producing B cells. Phase 2 begins many years later, when mis-differentiated T cells gain tissue-invasive effector functions, enter the joint, promote non-resolving inflammation, and give rise to clinically relevant arthritis. In Phase 3 of the RA disease process, abnormal innate immune functions are added to adaptive autoimmunity, converting synovial inflammation into a tissue-destructive process that erodes cartilage and bone. Emerging data have implicated metabolic mis-regulation as a fundamental pathogenic pathway in all phases of RA. Early in their life cycle, RA T cells fail to repair mitochondrial DNA, resulting in a malfunctioning metabolic machinery. Mitochondrial insufficiency is aggravated by the mis-trafficking of the energy sensor AMPK away from the lysosomal surface. The metabolic signature of RA T cells is characterized by the shunting of glucose toward the pentose phosphate pathway and toward biosynthetic activity. During the intermediate and terminal phase of RA-imposed tissue inflammation, tissue-residing macrophages, T cells, B cells and stromal cells are chronically activated and under high metabolic stress, creating a microenvironment poor in oxygen and glucose, but rich in metabolic intermediates, such as lactate. By sensing tissue lactate, synovial T cells lose their mobility and are trapped in the tissue niche. The linkage of defective DNA repair, misbalanced metabolic pathways, autoimmunity, and tissue inflammation in RA encourages metabolic interference as a novel treatment strategy during both the early stages of tolerance breakdown and the late stages of tissue inflammation. Defining and targeting metabolic abnormalities provides a new paradigm to treat, or even prevent, the cellular defects underlying autoimmune disease.

    View details for DOI 10.3389/fimmu.2021.652771

    View details for PubMedID 33868292

  • Histone deficiency and accelerated replication stress in T cell aging. The Journal of clinical investigation Kim, C., Jin, J., Ye, Z., Jadhav, R. R., Gustafson, C. E., Hu, B., Cao, W., Tian, L., Weyand, C. M., Goronzy, J. J. 2021; 131 (11)

    Abstract

    With increasing age, individuals are more vulnerable to viral infections such as with influenza or the SARS-CoV-2 virus. One age-associated defect in human T cells is the reduced expression of miR-181a. miR-181ab1 deficiency in peripheral murine T cells causes delayed viral clearance after infection, resembling human immune aging. Here we show that naive T cells from older individuals as well as miR-181ab1-deficient murine T cells develop excessive replication stress after activation, due to reduced histone expression and delayed S-phase cell cycle progression. Reduced histone expression was caused by the miR-181a target SIRT1 that directly repressed transcription of histone genes by binding to their promoters and reducing histone acetylation. Inhibition of SIRT1 activity or SIRT1 silencing increased histone expression, restored cell cycle progression, diminished the replication-stress response, and reduced the production of inflammatory mediators in replicating T cells from old individuals. Correspondingly, treatment with SIRT1 inhibitors improved viral clearance in mice with miR-181a-deficient T cells after LCMV infection. In conclusion, SIRT1 inhibition may be beneficial to treat systemic viral infection in older individuals by targeting antigen-specific T cells that develop replication stress due to miR-181a deficiency.

    View details for DOI 10.1172/JCI143632

    View details for PubMedID 34060486

  • Arachidonic acid-regulated calcium signaling in T cells from patients with rheumatoid arthritis promotes synovial inflammation. Nature communications Ye, Z., Shen, Y., Jin, K., Qiu, J., Hu, B., Jadhav, R. R., Sheth, K., Weyand, C. M., Goronzy, J. J. 2021; 12 (1): 907

    Abstract

    Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) are two distinct autoimmune diseases that manifest with chronic synovial inflammation. Here, we show that CD4+ T cells from patients with RA and PsA have increased expression of the pore-forming calcium channel component ORAI3, thereby increasing the activity of the arachidonic acid-regulated calcium-selective (ARC) channel and making T cells sensitive to arachidonic acid. A similar increase does not occur in T cells from patients with systemic lupus erythematosus. Increased ORAI3 transcription in RA and PsA T cells is caused by reduced IKAROS expression, a transcriptional repressor of the ORAI3 promoter. Stimulation of the ARC channel with arachidonic acid induces not only a calcium influx, but also the phosphorylation of components of the T cell receptor signaling cascade. In a human synovium chimeric mouse model, silencing ORAI3 expression in adoptively transferred T cells from patients with RA attenuates tissue inflammation, while adoptive transfer of T cells from healthy individuals with reduced expression of IKAROS induces synovitis. We propose that increased ARC activity due to reduced IKAROS expression makes T cells more responsive and contributes to chronic inflammation in RA and PsA.

    View details for DOI 10.1038/s41467-021-21242-z

    View details for PubMedID 33568645

  • Lysosomes in T Cell Immunity and Aging. Frontiers in aging Jin, J., Zhang, H., Weyand, C. M., Goronzy, J. J. 2021; 2: 809539

    Abstract

    Lysosomes were initially recognized as degradation centers that regulate digestion and recycling of cellular waste. More recent studies document that the lysosome is an important signaling hub that regulates cell metabolism. Our knowledge of the role of lysosomes in immunity is mostly derived from innate immune cells, especially lysosomal degradation-specialized cells such as macrophages and dendritic cells. Their function in adaptive immunity is less understood. However, with the recent emphasis on metabolic regulation of T cell differentiation, lysosomes are entering center stage in T cell immunology. In this review, we will focus on the role of lysosomes in adaptive immunity and discuss recent findings on lysosomal regulation of T cell immune responses and lysosomal dysfunction in T cell aging.

    View details for DOI 10.3389/fragi.2021.809539

    View details for PubMedID 35822050

  • The Transcription Factor TCF1 in T Cell Differentiation and Aging. International journal of molecular sciences Kim, C., Jin, J., Weyand, C. M., Goronzy, J. J. 2020; 21 (18)

    Abstract

    The transcription factor T cell factor 1 (TCF1), a pioneer transcription factor as well as a downstream effector of WNT/beta-catenin signaling, is indispensable for T cell development in the thymus. Recent studies have highlighted the additional critical role of TCF1 in peripheral T cell responses to acute and chronic infections as well as cancer. Here, we review the regulatory functions of TCF1 in the differentiation of T follicular helper cells, memory T cells and recently described stem-like exhausted T cells, where TCF1 promotes less differentiated stem-like cell states by controlling common gene-regulatory networks. These studies also provide insights into the mechanisms of defective T cell responses in older individuals. We discuss alterations in TCF1 expression and related regulatory networks with age and their consequences for T cell responses to infections and vaccination. The increasing understanding of the pathways regulating TCF1 expression and function in aged T cells holds the promise of enabling the design of therapeutic interventions aiming at improving T cell responses in older individuals.

    View details for DOI 10.3390/ijms21186497

    View details for PubMedID 32899486

  • Pathogenesis of Giant Cell Arteritis and Takayasu Arteritis-Similarities and Differences. Current rheumatology reports Watanabe, R., Berry, G. J., Liang, D. H., Goronzy, J. J., Weyand, C. M. 2020; 22 (10): 68

    Abstract

    PURPOSE OF REVIEW: Giant cell arteritis (GCA) and Takayasu arteritis (TAK) are auto-inflammatory and autoimmune diseases with a highly selective tissue tropism for medium and large arteries. In both diseases, CD4+ T cells and macrophages form granulomatous lesions within the arterial wall, a tissue site normally protected by immune privilege. Vascular lesions can be accompanied by an extravascular component, typically an intense hepatic acute phase response that produces well-known laboratory abnormalities, e.g., elevated ESR and CRP. It is unclear whether GCA and TAK lie on a spectrum of disease or whether they represent fundamentally different disease processes.RECENT FINDINGS: GCA and TAK share many clinical features, but there are substantial differences in genetics, epidemiology, disease mechanisms, response to treatment, and treatment complications that give rise to different disease trajectories. A significant difference lies in the composition of the wall-infiltrating immune cell compartment, which in TAK includes a significant population of CD8+ T cells as well as natural killer cells, specifying disparate disease effector pathways mediating tissue damage and vessel wall remodeling. Despite the similarities in tissue tropism and histomorphology, GCA and TAK are two distinct vasculitides that rely on separate disease mechanisms and require disease-specific approaches in diagnosis and management.

    View details for DOI 10.1007/s11926-020-00948-x

    View details for PubMedID 32845392

  • T Cell Aging in Hypertension. Cardiovascular research Weyand, C. M., Goronzy, J. J. 2020

    View details for DOI 10.1093/cvr/cvaa185

    View details for PubMedID 32609335

  • Homeostatic maintenance of human T cells in bioengineered secondary lymphoid organoids - a model for studying age-related immune decline Gustafson, C., Lambert, S., Weyand, C. M., Goronzy, J. J. AMER ASSOC IMMUNOLOGISTS. 2020
  • Influence of immune aging on vaccine responses. The Journal of allergy and clinical immunology Gustafson, C. E., Kim, C., Weyand, C. M., Goronzy, J. J. 2020; 145 (5): 1309–21

    Abstract

    Impaired vaccine responses in older individuals are associated with alterations in both the quantity and quality of the T-cell compartment with age. As reviewed herein, the T-cell response to vaccination requires a fine balance between the generation of inflammatory effector T cells versus follicular helper T (TFH) cells that mediate high-affinity antibody production in tandem with the induction of long-lived memory cells for effective recall immunity. During aging, we find that this balance is tipped where T cells favor short-lived effector but not memory or TFH responses. Consistently, vaccine-induced antibodies commonly display a lower protective capacity. Mechanistically, multiple, potentially targetable, changes in T cells have been identified that contribute to these age-related defects, including posttranscription regulation, T-cell receptor signaling, and metabolic function. Although research into the induction of tissue-specific immunity by vaccines and with age is still limited, current mechanistic insights provide a framework for improved design of age-specific vaccination strategies that require further evaluation in a clinical setting.

    View details for DOI 10.1016/j.jaci.2020.03.017

    View details for PubMedID 32386655

  • FOXO1 deficiency impairs proteostasis in aged T cells. Science advances Jin, J., Li, X., Hu, B., Kim, C., Cao, W., Zhang, H., Weyand, C. M., Goronzy, J. J. 2020; 6 (17): eaba1808

    Abstract

    T cell differentiation involves the dynamic regulation of FOXO1 expression, which rapidly declines after activation and is subsequently restored. Reexpression is impaired in naïve CD4+ T cell responses from older individuals. Here, we show that FOXO1 promotes lysosome function through the induction of the key transcription factor for lysosomal proteins, TFEB. Subdued FOXO1 reexpression in activated CD4+ T cells impairs lysosomal activity, causing an expansion of multivesicular bodies (MVBs). Expansion of the MVB compartment induces the sequestration of glycogen synthase kinase 3β (GSK3β), thereby suppressing protein turnover and enhancing glycolytic activity. As a consequence, older activated CD4+ T cells develop features reminiscent of senescent cells. They acquire an increased cell mass, preferentially differentiate into short-lived effector T cells, and secrete exosomes that harm cells in the local environment through the release of granzyme B.

    View details for DOI 10.1126/sciadv.aba1808

    View details for PubMedID 32494657

    View details for PubMedCentralID PMC7176426

  • FOXO1 deficiency impairs proteostasis in aged T cells SCIENCE ADVANCES Jin, J., Li, X., Hu, B., Kim, C., Cao, W., Zhang, H., Weyand, C. M., Goronzy, J. J. 2020; 6 (17)
  • Immune cell repertoires in breast cancer patients after adjuvant chemotherapy. JCI insight Gustafson, C. E., Jadhav, R., Cao, W., Qi, Q., Pegram, M., Tian, L., Weyand, C. M., Goronzy, J. J. 2020; 5 (4)

    Abstract

    Adjuvant chemotherapy in breast cancer patients causes immune cell depletion at an age when the regenerative capacity is compromised. Successful regeneration requires the recovery of both quantity and quality of immune cell subsets. Although immune cell numbers rebound within a year after treatment, it is unclear whether overall compositional diversity is recovered. We investigated the regeneration of immune cell complexity by comparing peripheral blood mononuclear cells from breast cancer patients ranging from 1-5 years after chemotherapy with those of age-matched healthy controls using mass cytometry and T cell receptor sequencing. These data reveal universal changes in patients' CD4+ T cells that persisted for years and consisted of expansion of Th17-like CD4 memory populations with incomplete recovery of CD4+ naive T cells. Conversely, CD8+ T cells fully recovered within a year. Mechanisms of T cell regeneration, however, were unbiased, as CD4+ and CD8+ T cell receptor diversity remained high. Likewise, terminal differentiated effector memory cells were not expanded, indicating that regeneration was not driven by recognition of latent viruses. These data suggest that, while CD8+ T cell immunity is successfully regenerated, the CD4 compartment may be irreversibly affected. Moreover, the bias of CD4 memory toward inflammatory effector cells may impact responses to vaccination and infection.

    View details for DOI 10.1172/jci.insight.134569

    View details for PubMedID 32102986

  • Immunometabolism in the development of rheumatoid arthritis. Immunological reviews Weyand, C. M., Goronzy, J. J. 2020

    Abstract

    In rheumatoid arthritis (RA), breakdown of self-tolerance and onset of clinical disease are separated in time and space, supporting a multi-hit model in which emergence of autoreactive T cells is a pinnacle pathogenic event. Determining factors in T cell differentiation and survival include antigen recognition, but also the metabolic machinery that provides energy and biosynthetic molecules for cell building. Studies in patients with RA have yielded a disease-specific metabolic signature, which enables naive CD4 T cells to differentiate into pro-inflammatory helper T cells that are prone to invade into tissue and elicit inflammation through immunogenic cell death. A typifying property of RA CD4 T cells is the shunting of glucose away from glycolytic breakdown and mitochondrial processing toward the pentose phosphate pathway, favoring anabolic over catabolic reactions. Key defects have been localized to the mitochondria and the lysosome; including instability of mitochondrial DNA due to the lack of the DNA repair nuclease MRE11A and inefficient lysosomal tethering of AMPK due to deficiency of N-myristoyltransferase 1 (NMT1). The molecular taxonomy of the metabolically reprogrammed RA T cells includes glycolytic enzymes (glucose-6-phosphate dehydrogenase, phosphofructokinase), DNA repair molecules (MRE11A, ATM), regulators of protein trafficking (NMT1), and the membrane adapter protein TSK5. As the mechanisms determining abnormal T cell behavior in RA are unraveled, opportunities will emerge to interject autoimmune T cells by targeting their metabolic checkpoints.

    View details for DOI 10.1111/imr.12838

    View details for PubMedID 31984519

  • Metabolic Fitness of T Cells in Autoimmune Disease. Immunometabolism Wu, B., Goronzy, J. J., Weyand, C. M. 2020; 2 (2)

    Abstract

    Rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) are relatively common autoimmune diseases, often considered prototypic examples for how protective immunity switches to destructive immunity. The autoantigens recognized in RA and SLE are distinct, clinical manifestations are partially overlapping. A shared feature is the propensity of the adaptive immune system to respond inappropriately, with T cell hyper-responsiveness a pinnacle pathogenic defect. Upon antigen recognition, T cells mobilize a multi-pranged metabolic program, enabling them to massively expand and turn into highly mobile effector cells. Current evidence supports that T cells from patients with RA or SLE adopt metabolic programs different from healthy T cells, in line with the concept that autoimmune effector functions rely on specified pathways of energy sensing, energy generation and energy utilization. Due to misrouting of the energy sensor AMPK, RA T cells have a defect in balancing catabolic and anabolic processes and deviate towards a cell-building program. They supply biosynthetic precursors by shunting glucose away from glycolytic breakdown towards the pentose phosphate pathway and upregulate lipogenesis, enabling cellular motility and tissue invasiveness. Conversely, T cells from SLE patients are committed to high glycolytic flux, overusing the mitochondrial machinery and imposing oxidative stress. Typically, disease-relevant effector functions in SLE are associated with inappropriate activation of the key metabolic regulator mTORC1. Taken together, disease-specific metabolic signatures in RA and SLE represent vulnerabilities that are therapeutically targetable to suppress pathogenic immune responses.

    View details for DOI 10.20900/immunometab20200017

    View details for PubMedID 32477606

  • Distinct Age-Related Epigenetic Signatures in CD4 and CD8 T Cells. Frontiers in immunology Hu, B., Jadhav, R. R., Gustafson, C. E., Le Saux, S., Ye, Z., Li, X., Tian, L., Weyand, C. M., Goronzy, J. J. 2020; 11: 585168

    Abstract

    Healthy immune aging is in part determined by how well the sizes of naive T cell compartments are being maintained with advancing age. Throughout adult life, replenishment largely derives from homeostatic proliferation of existing naive and memory T cell populations. However, while the subpopulation composition of CD4 T cells is relatively stable, the CD8 T cell compartment undergoes more drastic changes with loss of naive CD8 T cells and accumulation of effector T cells, suggesting that CD4 T cells are more resilient to resist age-associated changes. To determine the epigenetic basis for these differences in behaviors, we compared chromatin accessibility maps of CD4 and CD8 T cell subsets from young and old individuals and related the results to the expressed transcriptome. The dominant age-associated signatures resembled hallmarks of differentiation, which were more pronounced for CD8 naive and memory than the corresponding CD4 T cell subsets, indicating that CD8 T cells are less able to keep cellular quiescence upon homeostatic proliferation. In parallel, CD8 T cells from old adults, irrespective of their differentiation state, displayed greater reduced accessibility to genes of basic cell biological function, including genes encoding ribosomal proteins. One possible mechanism is the reduced expression of the transcription factors YY1 and NRF1. Our data suggest that chromatin accessibility signatures can be identified that distinguish CD4 and CD8 T cells from old adults and that may confer the higher resilience of CD4 T cells to aging.

    View details for DOI 10.3389/fimmu.2020.585168

    View details for PubMedID 33262764

  • Large and Medium-Vessel Vasculitides AUTOIMMUNE DISEASES, 6TH EDITION Weyand, C. M., Goronzy, J. J., Rose, N. R., Mackay, I. R. 2020: 1313–34
  • Ecto-NTPDase CD39 is a negative checkpoint that inhibits follicular helper cell generation. The Journal of clinical investigation Cao, W. n., Fang, F. n., Gould, T. n., Li, X. n., Kim, C. n., Gustafson, C. n., Lambert, S. n., Weyand, C. M., Goronzy, J. J. 2020

    Abstract

    Vaccination is a mainstay in preventive medicine, reducing morbidity and mortality from infection, largely by generating pathogen-specific neutralizing antibodies. However, standard immunization strategies are insufficient with increasing age due to immunological impediments, including defects in T follicular helper (Tfh) cells. Here, we found that Tfh generation is inversely linked to the expression of the ecto-NTPDase CD39 that modifies purinergic signaling. The lineage-determining transcription factor BCL6 inhibited CD39 expression, while increased Tfh frequencies were found in individuals with a germline polymorphism preventing transcription of ENTPD1, encoding CD39. In in vitro human and in vivo mouse studies, Tfh generation and germinal center responses were enhanced by reducing CD39 expression through the inhibition of the cAMP/PKA/p-CREB pathway, or by blocking adenosine signaling downstream of CD39 using the selective adenosine A2a receptor antagonist istradefylline. Thus, purinergic signaling in differentiating T cells can be targeted to improve vaccine responses, in particular in older individuals who have increased CD39 expression.

    View details for DOI 10.1172/JCI132417

    View details for PubMedID 32452837

  • Cellular Signaling Pathways in Medium and Large Vessel Vasculitis. Frontiers in immunology Watanabe, R., Berry, G. J., Liang, D. H., Goronzy, J. J., Weyand, C. M. 2020; 11: 587089

    Abstract

    Autoimmune and autoinflammatory diseases of the medium and large arteries, including the aorta, cause life-threatening complications due to vessel wall destruction but also by wall remodeling, such as the formation of wall-penetrating microvessels and lumen-stenosing neointima. The two most frequent large vessel vasculitides, giant cell arteritis (GCA) and Takayasu arteritis (TAK), are HLA-associated diseases, strongly suggestive for a critical role of T cells and antigen recognition in disease pathogenesis. Recent studies have revealed a growing spectrum of effector functions through which T cells participate in the immunopathology of GCA and TAK; causing the disease-specific patterning of pathology and clinical outcome. Core pathogenic features of disease-relevant T cells rely on the interaction with endothelial cells, dendritic cells and macrophages and lead to vessel wall invasion, formation of tissue-damaging granulomatous infiltrates and induction of the name-giving multinucleated giant cells. Besides antigen, pathogenic T cells encounter danger signals in their immediate microenvironment that they translate into disease-relevant effector functions. Decisive signaling pathways, such as the AKT pathway, the NOTCH pathway, and the JAK/STAT pathway modify antigen-induced T cell activation and emerge as promising therapeutic targets to halt disease progression and, eventually, reset the immune system to reestablish the immune privilege of the arterial wall.

    View details for DOI 10.3389/fimmu.2020.587089

    View details for PubMedID 33072134

  • Innate and Adaptive Immunity in Giant Cell Arteritis. Frontiers in immunology Akiyama, M. n., Ohtsuki, S. n., Berry, G. J., Liang, D. H., Goronzy, J. J., Weyand, C. M. 2020; 11: 621098

    Abstract

    Autoimmune diseases can afflict every organ system, including blood vessels that are critically important for host survival. The most frequent autoimmune vasculitis is giant cell arteritis (GCA), which causes aggressive wall inflammation in medium and large arteries and results in vaso-occlusive wall remodeling. GCA shares with other autoimmune diseases that it occurs in genetically predisposed individuals, that females are at higher risk, and that environmental triggers are suspected to beget the loss of immunological tolerance. GCA has features that distinguish it from other autoimmune diseases and predict the need for tailored diagnostic and therapeutic approaches. At the core of GCA pathology are CD4+ T cells that gain access to the protected tissue niche of the vessel wall, differentiate into cytokine producers, attain tissue residency, and enforce macrophages differentiation into tissue-destructive effector cells. Several signaling pathways have been implicated in initiating and sustaining pathogenic CD4+ T cell function, including the NOTCH1-Jagged1 pathway, the CD28 co-stimulatory pathway, the PD-1/PD-L1 co-inhibitory pathway, and the JAK/STAT signaling pathway. Inadequacy of mechanisms that normally dampen immune responses, such as defective expression of the PD-L1 ligand and malfunction of immunosuppressive CD8+ T regulatory cells are a common theme in GCA immunopathology. Recent studies are providing a string of novel mechanisms that will permit more precise pathogenic modeling and therapeutic targeting in GCA and will fundamentally inform how abnormal immune responses in blood vessels lead to disease.

    View details for DOI 10.3389/fimmu.2020.621098

    View details for PubMedID 33717054

    View details for PubMedCentralID PMC7947610

  • Neutrophil Extracellular Traps Induce Tissue-Invasive Macrophages in Granulomatosis with Polyangiitis Dominated by Ear, Nose and Throat Manifestations Akiyama, M., Ibrahim, N., Zeisbrich, M., Hwang, P., Goronzy, J., Weyand, C. WILEY. 2019
  • Lysosomal Placement of the Energy Sensors AMPK and mTORC1 Controls Tissue Inflammation in Rheumatoid Arthritis Wen, Z., Jin, K., Li, Y., Wu, B., Goronzy, J., Weyand, C. WILEY. 2019
  • Loss-of-function of the DNA Repair Nuclease MRE11A Induces Mitochondrial Failure and Tissue Inflammation in Rheumatoid Arthritis Li, Y., Shen, Y., Jin, K., Wen, Z., Cao, W., Wu, B., Wen, R., Tian, L., Berry, G., Goronzy, J., Weyand, C. WILEY. 2019
  • Metabolic Signatures of Pathogenic T Cells in Medium and Large Vessel Vasculitis Akiyama, M., Zhang, H., Watanabe, R., Maeda, T., Berry, G., Goronzy, J., Weyand, C. WILEY. 2019
  • GM-CSF Is a Pro-Inflammatory Cytokine in Experimental Vasculitis of Medium and Large Arteries Watanabe, R., Zhang, H., Maeda, T., Akiyama, M., Gandhi, R., Paolini, J., Berry, G., Weyand, C. WILEY. 2019
  • Cytokines, growth factors and proteases in medium and large vessel vasculitis CLINICAL IMMUNOLOGY Weyand, C. M., Watanabe, R., Zhang, H., Akiyama, M., Berry, G. J., Goronzy, J. J. 2019; 206: 33–41
  • Geographical variations in ocular and extra-ocular manifestations in Behcet's disease. European journal of rheumatology Shahram, F., Mahlen, M. T., Akhlaghi, M., Davatchi, F., Liao, Y. J., Weyand, C. M. 2019: 1–8

    Abstract

    OBJECTIVE: Behcet's disease (BD) is a rare vasculitis that results in multi-organ inflammatory disease. At-risk populations are most prevalent in the Middle East and East Asia. Clinical data on BD in Western countries, especially in the United States, are scarce. We have compared clinical patterning of BD vasculitis in two geographically defined patient cohorts in the Western United States and Iran.METHODS: Comparative analysis of a retrospective cohort of 56 patients with BD evaluated at Stanford University Hospital between 2000 and 2016 and a cohort of 163 patients from the BD Registry at Tehran University of Medical Sciences. Clinical, demographic, laboratory, and treatment data were available. Comparisons were performed using descriptive statistics, Student's t-test, and chi2-test.RESULTS: The Stanford patients with BD were significantly younger at disease onset, had a higher proportion of females, and had longer disease duration than Iranian patients with BD. Genital ulcers, skin, joint, neurological, vascular, cardiopulmonary manifestations were all significantly more common in the Stanford cohort and 38% of Stanford patients had four or more organ systems involved compared with approximately 10% of Iranian patients. In contrast, Stanford patients had fewer ocular lesions (Stanford 21.4% vs. Iran 53.4% p<0.05), with the biggest difference seen for retinal vasculitis.CONCLUSION: Patients with BD from the Western US have a more severe disease course when compared to Iranian patients with BD, as demonstrated by earlier onset and a higher rate of multi-organ involvement. The high risk of Iranian patients with BD developing vasculitis of ocular structures suggests distinct pathomechanisms driving ocular versus extra-ocular BD.

    View details for DOI 10.5152/eurjrheum.2019.18215

    View details for PubMedID 31329543

  • The DNA Repair Nuclease MRE11A Functions as a Mitochondrial Protector and Prevents T Cell Pyroptosis and Tissue Inflammation. Cell metabolism Li, Y., Shen, Y., Jin, K., Wen, Z., Cao, W., Wu, B., Wen, R., Tian, L., Berry, G. J., Goronzy, J. J., Weyand, C. M. 2019

    Abstract

    In the autoimmune disease rheumatoid arthritis (RA), CD4+ Tcells promote pro-inflammatory effector functions by shunting glucose away from glycolysis and ATP production. Underlying mechanisms remain unknown, and here we implicate the DNA repair nuclease MRE11A in the cells' bioenergetic failure. MRE11A deficiency in RA Tcells disrupted mitochondrial oxygen consumption and suppressed ATP generation. Also, MRE11A loss of function caused leakage of mitochondrial DNA (mtDNA) into the cytosol, triggering inflammasome assembly, caspase-1 activation, and pyroptotic cell death. Caspase-1 activation was frequent in lymph-node-residing Tcells in RA patients. Invivo, pharmacologic and genetic inhibition of MRE11A resulted in tissuedeposition of mtDNA, caspase-1 proteolysis, andaggressive tissue inflammation. Conversely, MRE11A overexpression restored mitochondrial fitness and shielded tissue from inflammatory attack. Thus, the nuclease MRE11A regulates a mitochondrial protection program, and MRE11A deficiency leads to DNA repair defects, energy production, and failure and loss of tissue homeostasis.

    View details for DOI 10.1016/j.cmet.2019.06.016

    View details for PubMedID 31327667

  • Transcription factor networks in aged naive CD4 T cells bias lineage differentiation. Aging cell Hu, B., Li, G., Ye, Z., Gustafson, C. E., Tian, L., Weyand, C. M., Goronzy, J. J. 2019: e12957

    Abstract

    With reducedthymic activity, the population of naive T cells in humans is maintained by homeostatic proliferation throughout adult life. In young adults, naive CD4 T cells have enormous proliferative potential and plasticity to differentiate into different lineages. Here, we explored whether naive CD4 T-cell aging is associated with a partial loss of this unbiased multipotency. We find that naive CD4 T cells from older individuals have developed a propensity to develop into TH9 cells. Two major mechanisms contribute to this predisposition. First, responsiveness to transforming growth factor beta (TGFbeta) stimulation is enhanced with age due to an upregulation of the TGFbetaR3 receptor that results in increased expression of the transcription factor PU.1. Secondly, aged naive CD4 T cells display altered transcription factor profiles in response to T-cell receptor stimulation, including enhanced expression of BATF and IRF4 and reduced expression of ID3 and BCL6. These transcription factors are involved in TH9 differentiation as well as IL9 transcription suggesting that the aging-associated changes in the transcription factor profile favor TH9 commitment.

    View details for DOI 10.1111/acel.12957

    View details for PubMedID 31264370

  • OSTEOPOROSIS AND RISK OF GIANT CELL ARTERITIS: A COMPARATIVE STUDY Le Pendu, C., Tchouante, P., Ly, K., Alexandra, J., Aslanbekova, N., Benali, K., Gonzalez-Chiappe, S., Lechtman, S., Sene, D., Weyand, C., Sacre, K., Mahr, A. BMJ PUBLISHING GROUP. 2019: 1760
  • CD28 AS A POTENTIAL THERAPEUTIC TARGET FOR GIANT CELL ARTERITIS Watanabe, R., Zhang, H., Berry, G., Nadler, S., Goronzy, J., Weyand, C. BMJ PUBLISHING GROUP. 2019: 146
  • CD28 Signaling Controls Metabolic Fitness of Pathogenic T Cells in Medium and Large Vessel Vasculitis JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY Zhang, H., Watanabe, R., Berry, G. J., Nadler, S. G., Goronzy, J. J., Weyand, C. M. 2019; 73 (14): 1811–23

    Abstract

    In giant cell arteritis, vessel-wall infiltrating CD4 T cells and macrophages form tissue-destructive granulomatous infiltrates, and the artery responds with a maladaptive response to injury, leading to intramural neoangiogenesis, intimal hyperplasia, and luminal occlusion. Lesion-residing T cells receive local signals, which represent potential therapeutic targets.The authors examined how CD28 signaling affects vasculitis induction and maintenance, and which pathogenic processes rely on CD28-mediated T-cell activation.Vasculitis was induced by transferring peripheral blood mononuclear cells from giant cell arteritis patients into immunodeficient NSG mice engrafted with human arteries. Human artery-NSG chimeras were treated with anti-CD28 domain antibody or control antibody. Treatment effects and immunosuppressive mechanisms were examined in vivo and in vitro applying tissue transcriptome analysis, immunohistochemistry, flow cytometry, and immunometabolic analysis.Blocking CD28-dependent signaling markedly reduced tissue-infiltrating T cells and effectively suppressed vasculitis. Mechanistic studies implicated CD28 in activating AKT signaling, T-cell proliferation and differentiation of IFN-γ and IL-21-producing effector T cells. Blocking CD28 was immunosuppressive by disrupting T-cell metabolic fitness; specifically, the ability to utilize glucose. Expression of the glucose transporter Glut1 and of glycolytic enzymes as well as mitochondrial oxygen consumption were all highly sensitive to CD28 blockade. Also, induction and maintenance of CD4+CD103+ tissue-resident memory T cells, needed to replenish the vasculitic infiltrates, depended on CD28 signaling. CD28 blockade effectively suppressed vasculitis-associated remodeling of the vessel wall.CD28 stimulation provides a metabolic signal required for pathogenic effector functions in medium and large vessel vasculitis. Disease-associated glycolytic activity in wall-residing T-cell populations can be therapeutically targeted by blocking CD28 signaling.

    View details for PubMedID 30975299

  • OSTEOPOROSIS AND RISK OF GIANT CELL ARTERITIS: A COMPARATIVE STUDY Le Pendu, C., Tchouante, P., Ly, K., Alexandra, J., Aslanbekova, N., Benali, K., Gonzalez-Chiappe, S., Lechtman, S., Sene, D., Weyand, C., Sacre, K., Mahr, A. OXFORD UNIV PRESS. 2019: 66
  • Cytokines, growth factors and proteases in medium and large vessel vasculitis. Clinical immunology (Orlando, Fla.) Weyand, C. M., Watanabe, R., Zhang, H., Akiyama, M., Berry, G. J., Goronzy, J. J. 2019

    Abstract

    Giant cell arteritis and Takayasu arteritis are autoimmune vasculitides that cause aneurysm formation and tissue infarction. Extravascular inflammation consists of an intense acute phase response. Deeper understanding of pathogenic events in the vessel wall has highlighted the loss of tissue protective mechanisms, the intrusion of immune cells into "forbidden territory", and the autonomy of self-renewing vasculitic infiltrates. Adventitial vasa vasora critically control vessel wall access and drive differentiation of tissue-invasive T cells. Selected T cells establish tissue residency and build autonomous, self-sufficient inflammatory lesions. Pathogenic effector T cells intrude and survive due to failed immune checkpoint inhibition. Vasculitis-sustaining T cells and macrophages provide a broad portfolio of effector functions, involving heterogeneous populations of pro-inflammatory T cells and diverse macrophage subsets that ultimately induce wall capillarization and intimal hyperplasia. Redirecting diagnostic and therapeutic strategies from control of extravascular inflammatory markers to suppression of vascular inflammation will improve disease management.

    View details for PubMedID 30772599

  • Functional pathways regulated by microRNA networks in CD8 T-cell aging AGING CELL Gustafson, C. E., Cavanagh, M. M., Jin, J., Weyand, C. M., Goronzy, J. J. 2019; 18 (1)

    View details for DOI 10.1111/acel.12879

    View details for Web of Science ID 000459022900033

  • Neutrophil Extracellular Traps Induce Tissue-Invasive Monocytes in Granulomatosis With Polyangiitis. Frontiers in immunology Akiyama, M. n., Zeisbrich, M. n., Ibrahim, N. n., Ohtsuki, S. n., Berry, G. J., Hwang, P. H., Goronzy, J. J., Weyand, C. M. 2019; 10: 2617

    Abstract

    Objective: Granulomatosis with polyangiitis (GPA) is a multi-organ vasculitic syndrome typically associated with neutrophil extracellular trap (NET) formation and aggressive tissue inflammation. Manifestations in head and neck (H&N) GPA include septal perforations, saddle-nose deformities, bony erosions of the orbital and sinus walls, middle ear damage and epiglottitis, indicative of bone, cartilage, and connective tissue destruction. Whether H&N-centric lesions engage disease pathways distinctive from the ischemic tissue damage in the lungs, kidneys, skin, and peripheral nerves is unknown. We have compared inflammatory responses triggered by neutrophilic NETs in patients with H&N GPA and systemic GPA (sGPA). Methods: Neutrophils and monocytes were isolated from the peripheral blood of patients with H&N GPA, sGPA, and age/gender matched healthy individuals. Neutrophil NETosis was induced. NETs were isolated and cocultured with monocytes. Gene induction was quantified by RT-PCR, protein upregulation by flow cytometry. Tissue invasiveness of monocytes was measured in a 3D collagen matrix system. Expression of MMP-9 in tissue-residing macrophages was assessed by immunohistochemistry in tissue biopsies. Results: Neutrophils from H&N GPA patients showed more intense NETosis with higher frequencies of netting neutrophils (P < 0.001) and release of higher amounts of NETs (P < 0.001). Isolated NETs from H&N GPA functioned as an inducer of danger-associated molecular patterns in monocytes; specifically, alarmin S100A9. NET-induced upregulation of monocyte S100A9 required recognition of DNA. S100A9 release resulted in the induction of metalloproteinases, including MMP-9, and enabled monocytes to invade into extracellular matrix. Anti-MMP-9 treatment attenuated the tissue invasiveness of monocytes primed with NETs from H&N GPA patients. MMP-9-producing macrophages dominated the tissue infiltrates in naso-sinal biopsies from H&N GPA patients. Conclusion: Distinct disease patterns in GPA are associated with differences in NET formation and NET content. H&N GPA patients with midline cartilaginous and bony lesions are highly efficient in generating NETs. H&N GPA neutrophils trigger the induction of the alarmin S100A9, followed by production of MMP-9, endowing monocytes with tissue-invasive capabilities.

    View details for DOI 10.3389/fimmu.2019.02617

    View details for PubMedID 31798577

    View details for PubMedCentralID PMC6874157

  • Defects in Antiviral T Cell Responses Inflicted by Aging-Associated miR-181a Deficiency. Cell reports Kim, C. n., Jadhav, R. R., Gustafson, C. E., Smithey, M. J., Hirsch, A. J., Uhrlaub, J. L., Hildebrand, W. H., Nikolich-Žugich, J. n., Weyand, C. M., Goronzy, J. J. 2019; 29 (8): 2202–16.e5

    Abstract

    Generation of protective immunity to infections and vaccinations declines with age. Studies in healthy individuals have implicated reduced miR-181a expression in T cells as contributing to this defect. To understand the impact of miR-181a expression on antiviral responses, we examined LCMV infection in mice with miR-181ab1-deficient T cells. We found that miR-181a deficiency delays viral clearance, thereby biasing the immune response in favor of CD4 over CD8 T cells. Antigen-specific CD4 T cells in mice with miR-181a-deficient T cells expand more and have a broader TCR repertoire with preferential expansion of high-affinity T cells than in wild-type mice. Importantly, generation of antigen-specific miR-181a-deficient CD8 effector T cells is particularly impaired, resulting in lower frequencies of CD8 T cells in the liver even at time points when the infection has been cleared. Consistent with the mouse model, CD4 memory T cells in individuals infected with West Nile virus at older ages tend to be more frequent and of higher affinity.

    View details for DOI 10.1016/j.celrep.2019.10.044

    View details for PubMedID 31747595

  • Mechanisms underlying T cell ageing. Nature reviews. Immunology Goronzy, J. J., Weyand, C. M. 2019

    Abstract

    T cell ageing has a pivotal role in rendering older individuals vulnerable to infections and cancer and in impairing the response to vaccination. Easy accessibility to peripheral human T cells as well as an expanding array of tools to examine T cell biology have provided opportunities to examine major ageing pathways and their consequences for T cell function. Here, we review emerging concepts of how the body attempts to maintain a functional T cell compartment with advancing age, focusing on three fundamental domains of the ageing process, namely self-renewal, control of cellular quiescence and cellular senescence. Understanding these critical elements in successful T cell ageing will allow the design of interventions to prevent or reverse ageing-related T cell failure.

    View details for DOI 10.1038/s41577-019-0180-1

    View details for PubMedID 31186548

  • Chronic inflammation in the etiology of disease across the life span. Nature medicine Furman, D. n., Campisi, J. n., Verdin, E. n., Carrera-Bastos, P. n., Targ, S. n., Franceschi, C. n., Ferrucci, L. n., Gilroy, D. W., Fasano, A. n., Miller, G. W., Miller, A. H., Mantovani, A. n., Weyand, C. M., Barzilai, N. n., Goronzy, J. J., Rando, T. A., Effros, R. B., Lucia, A. n., Kleinstreuer, N. n., Slavich, G. M. 2019; 25 (12): 1822–32

    Abstract

    Although intermittent increases in inflammation are critical for survival during physical injury and infection, recent research has revealed that certain social, environmental and lifestyle factors can promote systemic chronic inflammation (SCI) that can, in turn, lead to several diseases that collectively represent the leading causes of disability and mortality worldwide, such as cardiovascular disease, cancer, diabetes mellitus, chronic kidney disease, non-alcoholic fatty liver disease and autoimmune and neurodegenerative disorders. In the present Perspective we describe the multi-level mechanisms underlying SCI and several risk factors that promote this health-damaging phenotype, including infections, physical inactivity, poor diet, environmental and industrial toxicants and psychological stress. Furthermore, we suggest potential strategies for advancing the early diagnosis, prevention and treatment of SCI.

    View details for DOI 10.1038/s41591-019-0675-0

    View details for PubMedID 31806905

  • Determinants governing T cell receptor α/β-chain pairing in repertoire formation of identical twins. Proceedings of the National Academy of Sciences of the United States of America Tanno, H. n., Gould, T. M., McDaniel, J. R., Cao, W. n., Tanno, Y. n., Durrett, R. E., Park, D. n., Cate, S. J., Hildebrand, W. H., Dekker, C. L., Tian, L. n., Weyand, C. M., Georgiou, G. n., Goronzy, J. J. 2019

    Abstract

    The T cell repertoire in each individual includes T cell receptors (TCRs) of enormous sequence diversity through the pairing of diverse TCR α- and β-chains, each generated by somatic recombination of paralogous gene segments. Whether the TCR repertoire contributes to susceptibility to infectious or autoimmune diseases in concert with disease-associated major histocompatibility complex (MHC) polymorphisms is unknown. Due to a lack in high-throughput technologies to sequence TCR α-β pairs, current studies on whether the TCR repertoire is shaped by host genetics have so far relied only on single-chain analysis. Using a high-throughput single T cell sequencing technology, we obtained the largest paired TCRαβ dataset so far, comprising 965,523 clonotypes from 15 healthy individuals including 6 monozygotic twin pairs. Public TCR α- and, to a lesser extent, TCR β-chain sequences were common in all individuals. In contrast, sharing of entirely identical TCRαβ amino acid sequences was very infrequent in unrelated individuals, but highly increased in twins, in particular in CD4 memory T cells. Based on nucleotide sequence identity, a subset of these shared clonotypes appeared to be the progeny of T cells that had been generated during fetal development and had persisted for more than 50 y. Additional shared TCRαβ in twins were encoded by different nucleotide sequences, implying that genetic determinants impose structural constraints on thymic selection that favor the selection of TCR α-β pairs with entire sequence identities.

    View details for DOI 10.1073/pnas.1915008117

    View details for PubMedID 31879353

  • The metabolic signature of T cells in rheumatoid arthritis. Current opinion in rheumatology Weyand, C. M., Wu, B. n., Goronzy, J. J. 2019

    Abstract

    Rheumatoid arthritis (RA) is a prototypic autoimmune disease manifesting as chronic inflammation of the synovium and leading to acceleration of cardiovascular disease and shortening of life expectancy. The basic defect causing autoimmunity has remained elusive, but recent insights have challenged the notion that autoantigen is the core driver.Emerging data have added metabolic cues involved in the proper maintenance and activation of immune cells as pathogenic regulators. Specifically, studies have unveiled metabolic pathways that enforce T cell fate decisions promoting tissue inflammation; including T cell tissue invasiveness, T cell cytokine release, T cell-dependent macrophage activation and inflammatory T cell death. At the center of the metabolic abnormalities lies the mitochondria, which is consistently underperforming in RA T cells. The mitochondrial defect results at least partially from insufficient DNA repair and leads to lipid droplet accumulation, formation of invasive membrane ruffles, inflammasome activation and pyroptotic T cell death.T cells in patients with RA, even naïve T cells never having been involved in inflammatory lesions, have a unique metabolic signature and the changes in intracellular metabolites drive pathogenic T cell behavior. Recognizing the role of metabolic signals in cell fate decisions opens the possibility for immunomodulation long before the end stage synovial inflammation encountered in clinical practice.

    View details for DOI 10.1097/BOR.0000000000000683

    View details for PubMedID 31895885

  • Metabolic reprogramming in memory CD4 T cell responses of old adults. Clinical immunology (Orlando, Fla.) Yanes, R. E., Zhang, H. n., Shen, Y. n., Weyand, C. M., Goronzy, J. J. 2019

    Abstract

    To determine whether aging affects the ability of T cells to undergo metabolic reprogramming upon activation, we compared CD4 T cell responses after polyclonal in vitro stimulation. Compared to younger adults, CD4 memory T cells from healthy older individuals exhibited a higher upregulation of oxidative phosphorylation with increased production of reactive oxygen species and intracellular and secreted ATP. Increased ATP secretion led to increased purinergic signaling and P2X7-dependent increases in cytoplasmic calcium. The increased mitochondrial activity was not due to a difference in activation-induced mitochondrial biogenesis. Expression of carnitine palmitoyl transferase 1 was higher, conversely that of fatty acid synthase was reduced in older T cells, resulting in increased fatty acid oxidation, while depleting intracellular lipid stores. The aged CD4 memory T cells therefore maintain a more catabolic state in lipid metabolism, while their ability to upregulate glycolysis upon activation is preserved.

    View details for DOI 10.1016/j.clim.2019.07.003

    View details for PubMedID 31279855

  • Activation of miR-21-Regulated Pathways in Immune Aging Selects against Signatures Characteristic of Memory T Cells CELL REPORTS Kim, C., Hu, B., Jadhav, R. R., Jin, J., Zhang, H., Cavanagh, M. M., Akondy, R. S., Ahmed, R., Weyand, C. M., Goronzy, J. J. 2018; 25 (8): 2148-+
  • Activation of miR-21-Regulated Pathways in Immune Aging Selects against Signatures Characteristic of Memory T Cells. Cell reports Kim, C., Hu, B., Jadhav, R. R., Jin, J., Zhang, H., Cavanagh, M. M., Akondy, R. S., Ahmed, R., Weyand, C. M., Goronzy, J. J. 2018; 25 (8): 2148

    Abstract

    Induction of protective vaccine responses, governed by the successful generation of antigen-specific antibodies and long-lived memory Tcells, is increasingly impaired with age. Regulation of the Tcell proteome by a dynamic network of microRNAs is crucial to Tcell responses. Here, we show that activation-induced upregulation of miR-21 biases the transcriptome of differentiating Tcells away from memory Tcells and toward inflammatory effector Tcells. Such a transcriptome bias is also characteristic of Tcell responses in older individuals who have increased miR-21 expression and is reversed by antagonizing miR-21. miR-21 targets negative feedback circuits in several signaling pathways. The concerted, sustained activity of these signaling pathways in miR-21high Tcells disfavors the induction of transcription factor networks involved in memory cell differentiation. Our data suggest that curbing miR-21 upregulation or activity in older individuals may improve their ability to mount effective vaccine responses.

    View details for PubMedID 30463012

  • Glucose metabolism controls disease-specific signatures of macrophage effector functions. JCI insight Watanabe, R., Hilhorst, M., Zhang, H., Zeisbrich, M., Berry, G. J., Wallis, B. B., Harrison, D. G., Giacomini, J. C., Goronzy, J. J., Weyand, C. M. 2018; 3 (20)

    Abstract

    BACKGROUND: In inflammatory blood vessel diseases, macrophages represent a key component of the vascular infiltrates and are responsible for tissue injury and wall remodeling.METHODS: To examine whether inflammatory macrophages in the vessel wall display a single distinctive effector program, we compared functional profiles in patients with either coronary artery disease (CAD) or giant cell arteritis (GCA).RESULTS: Unexpectedly, monocyte-derived macrophages from the 2 patient cohorts displayed disease-specific signatures and differed fundamentally in metabolic fitness. Macrophages from CAD patients were high producers for T cell chemoattractants (CXCL9, CXCL10), the cytokines IL-1beta and IL-6, and the immunoinhibitory ligand PD-L1. In contrast, macrophages from GCA patients upregulated production of T cell chemoattractants (CXCL9, CXCL10) but not IL-1beta and IL-6, and were distinctly low for PD-L1 expression. Notably, disease-specific effector profiles were already identifiable in circulating monocytes. The chemokinehicytokinehiPD-L1hi signature in CAD macrophages was sustained by excess uptake and breakdown of glucose, placing metabolic control upstream of inflammatory function.CONCLUSIONS: We conclude that monocytes and macrophages contribute to vascular inflammation in a disease-specific and discernible pattern, have choices to commit to different functional trajectories, are dependent on glucose availability in their immediate microenvironment, and possess memory in their lineage commitment.FUNDING: Supported by the NIH (R01 AR042527, R01 HL117913, R01 AI108906, P01 HL129941, R01 AI108891, R01 AG045779 U19 AI057266, R01 AI129191), I01 BX001669, and the Cahill Discovery Fund.

    View details for PubMedID 30333306

  • Epigenetics of Tcell aging JOURNAL OF LEUKOCYTE BIOLOGY Goronzy, J. J., Hu, B., Kim, C., Jadhav, R. R., Weyand, C. M. 2018; 104 (4): 691–99
  • T follicular helper cell development and functionality in immune ageing. Clinical science (London, England : 1979) Gustafson, C. E., Weyand, C. M., Goronzy, J. J. 2018; 132 (17): 1925–35

    Abstract

    By 2050, there will be over 1.6 billion adults aged 65 years and older, making age-related diseases and conditions a growing public health concern. One of the leading causes of death in the ageing population is pathogenic infections (e.g. influenza, Streptococcus pneumoniae). This age-dependent susceptibility to infection has been linked to a reduced ability of the ageing immune system to mount protective responses against infectious pathogens, as well as to vaccines against these pathogens. The primary immune response that promotes protection is the production of antibodies by B cells - a response that is directly mediated by T follicular helper (TFH) cells within germinal centers (GCs) in secondary lymphoid tissues. In this review, we will summarize the current knowledge on the development and functionality of TFH cells, the use of circulating TFH (cTFH) cells as vaccine biomarkers, and the influence of age on these processes. Moreover, we will discuss the strategies for overcoming TFH cell dysfunction to improve protective antibody responses in the ageing human population.

    View details for PubMedID 30185614

  • T follicular helper cell development and functionality in immune ageing CLINICAL SCIENCE Gustafson, C. E., Weyand, C. M., Goronzy, J. J. 2018; 132 (17): 1925–35

    View details for DOI 10.1042/CS20171157

    View details for Web of Science ID 000443729100004

  • Redox-sensitive signaling in inflammatory T cells and in autoimmune disease FREE RADICAL BIOLOGY AND MEDICINE Weyand, C. M., Shen, Y., Goronzy, J. J. 2018; 125: 36–43
  • Hypertension and increased endothelial mechanical stretch promote monocyte differentiation and activation: roles of STAT3, interleukin 6 and hydrogen peroxide CARDIOVASCULAR RESEARCH Loperena, R., Van Beusecum, J. P., Itani, H. A., Engel, N., Laroumanie, F., Xiao, L., Elijovich, F., Laffer, C. L., Gnecco, J. S., Noonan, J., Maffia, P., Jasiewicz-Honkisz, B., Czesnikiewicz-Guzik, M., Mikolajczyk, T., Sliwa, T., Dikalov, S., Weyand, C. M., Guzik, T. J., Harrison, D. G. 2018; 114 (11): 1547–63

    Abstract

    Monocytes play an important role in hypertension. Circulating monocytes in humans exist as classical, intermediate, and non-classical forms. Monocyte differentiation can be influenced by the endothelium, which in turn is activated in hypertension by mechanical stretch. We sought to examine the role of increased endothelial stretch and hypertension on monocyte phenotype and function.Human monocytes were cultured with confluent human aortic endothelial cells undergoing either 5% or 10% cyclical stretch. We also characterized circulating monocytes in normotensive and hypertensive humans. In addition, we quantified accumulation of activated monocytes and monocyte-derived cells in aortas and kidneys of mice with Angiotensin II-induced hypertension. Increased endothelial stretch enhanced monocyte conversion to CD14++CD16+ intermediate monocytes and monocytes bearing the CD209 marker and markedly stimulated monocyte mRNA expression of interleukin (IL)-6, IL-1β, IL-23, chemokine (C-C motif) ligand 4, and tumour necrosis factor α. STAT3 in monocytes was activated by increased endothelial stretch. Inhibition of STAT3, neutralization of IL-6 and scavenging of hydrogen peroxide prevented formation of intermediate monocytes in response to increased endothelial stretch. We also found evidence that nitric oxide (NO) inhibits formation of intermediate monocytes and STAT3 activation. In vivo studies demonstrated that humans with hypertension have increased intermediate and non-classical monocytes and that intermediate monocytes demonstrate evidence of STAT3 activation. Mice with experimental hypertension exhibit increased aortic and renal infiltration of monocytes, dendritic cells, and macrophages with activated STAT3.These findings provide insight into how monocytes are activated by the vascular endothelium during hypertension. This is likely in part due to a loss of NO signalling and increased release of IL-6 and hydrogen peroxide by the dysfunctional endothelium and a parallel increase in STAT activation in adjacent monocytes. Interventions to enhance bioavailable NO, reduce IL-6 or hydrogen peroxide production or to inhibit STAT3 may have anti-inflammatory roles in hypertension and related conditions.

    View details for PubMedID 29800237

    View details for PubMedCentralID PMC6106108

  • MMP (Matrix Metalloprotease)-9-Producing Monocytes Enable T Cells to Invade the Vessel Wall and Cause Vasculitis CIRCULATION RESEARCH Watanabe, R., Maeda, T., Zhang, H., Berry, G. J., Zeisbrich, M., Brockett, R., Greenstein, A. E., Tian, L., Goronzy, J. J., Weyand, C. M. 2018; 123 (6): 700–715
  • A Mitochondrial Checkpoint in Autoimmune Disease CELL METABOLISM Weyand, C. M., Goronzy, J. J. 2018; 28 (2): 185–86
  • Regulation of miR-181a expression in T cell aging. Nature communications Ye, Z., Li, G., Kim, C., Hu, B., Jadhav, R. R., Weyand, C. M., Goronzy, J. J. 2018; 9 (1): 3060

    Abstract

    MicroRNAs have emerged as key regulators in T cell development, activation, and differentiation, with miR-181a having a prominent function. By targeting several signaling pathways, miR-181a is an important rheostat controlling T cell receptor (TCR) activation thresholds in thymic selection as well as peripheral T cell responses. A decline in miR-181a expression, due to reduced transcription of pri-miR-181a, accounts for T cell activation defects that occur with older age. Here we examine the transcriptional regulation of miR-181a expression and find a putative pri-miR-181a enhancer around position 198,904,300 on chromosome 1, which is regulated by a transcription factor complex including YY1. The decline in miR-181a expression correlates with reduced transcription of YY1 in older individuals. Partial silencing of YY1 in T cells from young individuals reproduces the signaling defects seen in older T cells. In conclusion, YY1 controls TCR signaling by upregulating miR-181a and dampening negative feedback loops mediated by miR-181a targets.

    View details for PubMedID 30076309

  • Regulation of miR-181a expression in T cell aging NATURE COMMUNICATIONS Ye, Z., Li, G., Kim, C., Hu, B., Jadhav, R. R., Weyand, C. M., Goronzy, J. J. 2018; 9
  • Matrix Metalloprotease-9 (MMP-9)-Producing Monocytes Enable T Cells to Invade the Vessel Wall and Cause Vasculitis. Circulation research Watanabe, R., Maeda, T., Zhang, H., Berry, G. J., Zeisbrich, M., Brockett, R. D., Greenstein, A. E., Tian, L., Goronzy, J., Weyand, C. M. 2018

    Abstract

    Rationale: Giant cell arteritis (GCA), a primary vasculitis of medium and large arteries, is associated with vessel wall damage, elastic membrane fragmentation and vascular remodeling. Proteinases are believed to contribute to pathogenesis by degrading extracellular matrix and causing tissue injury. Objective: The matrix metalloproteinase 9 (MMP-9), a type IV collagenase, is produced in the vasculitic lesions of GCA. It is unknown which pathogenic processes are MMP-9 dependent. Methods and Results: The tissue transcriptome of GCA-affected temporal arteries contained high amounts of MMP-9 transcripts and immunostaining for pro-MMP-9 localized the enzyme to wall-infiltrating macrophages. MMP-2 and MMP-9 transcripts were also abundant in monocytes and monocyte-derived macrophages from GCA patients. Patient-derived monocytes outperformed healthy monocytes in passing through engineered basement membranes. GCA CD4+ T cells required MMP-9-producing monocytes to penetrate through matrix built from type IV collagen. In vivo functions of MMP-9 were tested in a human artery-SCID chimera model by blocking enzyme activity with a highly specific monoclonal antibody or by injecting recombinant MMP-9. Inhibiting MMP-9 activity profoundly suppressed vascular injury, decreased the density of inflammatory infiltrates (p<0.001), reduced intramural neoangiogenesis (p<0.001) and prevented intimal layer hyperplasia (p<0.001). Recombinant MMP-9 amplified all domains of vasculitogenic activity, promoted assembly of T cell infiltrates (p<0.05), intensified formation of new microvessels (p<0.001) and worsened intimal thickening (p<0.001). Systemic delivery of N-acetyl-proline-glycine-proline (ac-PGP), a matrikine produced by MMP-9-mediated gelatinolysis, had limited vasculitogenic effects. Conclusions: In large vessel vasculitis, MMP-9 controls the access of monocytes and T cells to the vascular wall. T cells depend on MMP-9-producing monocytes to pass through collagen IV-containing basement membrane. Invasion of vasculitogenic T cells and monocytes, formation of neoangiogenic networks and neointimal growth all require the enzymatic activity of MMP-9; identifying this protease as a potential therapeutic target to restore the immunoprivilege of the arterial wall in large vessel vasculitis.

    View details for PubMedID 29970365

  • Hypermetabolic macrophages in rheumatoid arthritis and coronary artery disease due to glycogen synthase kinase 3b inactivation ANNALS OF THE RHEUMATIC DISEASES Zeisbrich, M., Yanes, R. E., Zhang, H., Watanabe, R., Li, Y., Brosig, L., Hong, J., Wallis, B. B., Giacomini, J. C., Assimes, T. L., Goronzy, J. J., Weyand, C. M. 2018; 77 (7): 1053–62
  • Epigenetics of Tcell aging. Journal of leukocyte biology Goronzy, J. J., Hu, B., Kim, C., Jadhav, R. R., Weyand, C. M. 2018

    Abstract

    T cells are a heterogeneous population of cells that differ in their differentiation stages. Functional states are reflected in the epigenome that confers stability in cellular identity and is therefore important for naive as well as memory Tcell function. In many cellular systems, changes in chromatin structure due to alterations in histone expression, histone modifications and DNA methylation are characteristic of the aging process and cause or at least contribute to cellular dysfunction in senescence. Here, we review the epigenetic changes in Tcells that occur with age and discuss them in the context of canonical epigenetic marks in aging model systems as well as recent findings of chromatin accessibility changes in Tcell differentiation. Remarkably, transcription factor networks driving Tcell differentiation account for many of the age-associated modifications in chromatin structures suggesting that loss of quiescence and activation of differentiation pathways are major components of Tcell aging.

    View details for PubMedID 29947427

  • DNA damage, metabolism and aging in pro-inflammatory T cells Rheumatoid arthritis as a model system EXPERIMENTAL GERONTOLOGY Li, Y., Goronzy, J. J., Weyand, C. M. 2018; 105: 118–27

    Abstract

    The aging process is the major driver of morbidity and mortality, steeply increasing the risk to succumb to cancer, cardiovascular disease, infection and neurodegeneration. Inflammation is a common denominator in age-related pathologies, identifying the immune system as a gatekeeper in aging overall. Among immune cells, T cells are long-lived and exposed to intense replication pressure, making them sensitive to aging-related abnormalities. In successful T cell aging, numbers of naïve cells, repertoire diversity and activation thresholds are preserved as long as possible; in maladaptive T cell aging, protective T cell functions decline and pro-inflammatory effector cells are enriched. Here, we review in the model system of rheumatoid arthritis (RA) how maladaptive T cell aging renders the host susceptible to chronic, tissue-damaging inflammation. In T cells from RA patients, known to be about 20years pre-aged, three interconnected functional domains are altered: DNA damage repair, metabolic activity generating energy and biosynthetic precursor molecules, and shaping of plasma membranes to promote T cell motility. In each of these domains, key molecules and pathways have now been identified, including the glycolytic enzymes PFKFB3 and G6PD; the DNA repair molecules ATM, DNA-PKcs and MRE11A; and the podosome marker protein TKS5. Some of these molecules may help in defining targetable pathways to slow the T cell aging process.

    View details for PubMedID 29101015

    View details for PubMedCentralID PMC5871568

  • Redox-sensitive signaling in inflammatory T cells and in autoimmune disease. Free radical biology & medicine Weyand, C. M., Shen, Y., Goronzy, J. J. 2018

    Abstract

    Reactive oxygen species (ROS) are byproducts of oxygen metabolism best known for their damaging potential, but recent evidence has exposed their role as secondary messengers, which regulate cell function through redox-activatable signaling systems. In immune cells, specifically in T cells, redox-sensitive signaling pathways have been implicated in controlling several functional domains; including cell cycle progression, T effector cell differentiation, tissue invasion and inflammatory behavior. T cells from patients with the autoimmune disease rheumatoid arthritis (RA) have emerged as a valuable model system to examine the functional impact of ROS on T cell function. Notably, RA T cells are distinguished from healthy T cells based on reduced ROS production and undergo "reductive stress". Upstream defects leading to the ROSlow status of RA T cells are connected to metabolic reorganization. RA T cells shunt glucose away from pyruvate and ATP production towards the pentose phosphate pathway, where they generate NADPH and consume cellular ROS. Downstream consequences of the ROSlow conditions in RA T cells include insufficient activation of the DNA repair kinase ATM, bypassing of the G2/M cell cycle checkpoint and biased differentiation of T cells into IFN-gamma and IL-17-producing inflammatory cells. Also, ROSlow T cells rapidly invade into peripheral tissue due to dysregulated lipogenesis, excessive membrane ruffling, and overexpression of a motility module dominated by the scaffolding protein Tks5. These data place ROS into a pinnacle position in connecting cellular metabolism and protective versus auto-aggressive T cell immunity. Therapeutic interventions for targeted ROS enhancement instead of ROS depletion should be developed as a novel strategy to treat autoimmune tissue inflammation.

    View details for PubMedID 29524605

  • Refractory Giant Cell Arteritis Complicated by Vision Loss From Optic Atrophy and Maculopathy Associated With Pachymeningitis JOURNAL OF NEURO-OPHTHALMOLOGY Uribe, J. A., Aggarwal, I., Witthayaweerasak, J., Liao, Y., Berry, G. J., Sab, U. K., Weyand, C. M. 2018; 38 (1): 17–23

    Abstract

    We describe a 75-year-old woman who experienced vision loss in her left eye due to biopsy-proven giant cell arteritis (GCA). She subsequently developed pachymeningitis causing refractory headaches and bilateral optic neuropathy and maculopathy.Case report with literature review.Eighteen months after the initial diagnosis of GCA, imaging studies in our patient demonstrated pachymeningeal enhancement, and meningeal biopsy confirmed lymphoplasmacytic tissue infiltrates with low frequencies of IgG4+ plasma cells. Laboratory investigation revealed the presence of 3 antiretinal antibodies and antimyeloperoxidase antibodies, consistent with autoimmune retinopathy. Treatment with B-cell-depleting anti-CD20 antibodies suppressed meningeal inflammation and prevented further vision loss.This case illustrates that bilateral vision loss and chronic headaches in patients with GCA may result from retina-directed autoimmunity and pachymeningitis.

    View details for PubMedID 29059089

    View details for PubMedCentralID PMC5811389

  • The immunoinhibitory PD-1/PD-L1 pathway in inflammatory blood vessel disease Weyand, C. M., Berry, G. J., Goronzy, J. J. WILEY. 2018: 565–75
  • The immunoinhibitory PD-1/PD-L1 pathway in inflammatory blood vessel disease. Journal of leukocyte biology Weyand, C. M., Berry, G. J., Goronzy, J. J. 2018; 103 (3): 565–75

    Abstract

    Because of their vital function, the wall structures of medium and large arteries are immunoprivileged and protected from inflammatory attack. That vascular immunoprivilege is broken in atherosclerosis and in vasculitis, when wall-invading T cells and macrophages (Mϕ) promote tissue injury and maladaptive repair. Historically, tissue-residing T cells were studied for their antigen specificity, but recent progress has refocused attention to antigen-nonspecific regulation, which determines tissue access, persistence, and functional differentiation of T cells. The coinhibitory receptor PD-1, expressed on T cells, delivers negative signals when engaged by its ligand PD-L1, expressed on dendritic cells, Mϕ, and endothelial cells to attenuate T cell activation, effector functions, and survival. Through mitigating signals, the PD-1 immune checkpoint maintains tissue tolerance. In line with this concept, dendritic cells and Mϕs from patients with the vasculitic syndrome giant cell arteritis (GCA) are PD-L1lo ; including vessel-wall-embedded DCs that guard the vascular immunoprivilege. GCA infiltrates in the arterial walls are filled with PD-1+ T cells that secrete IFN-gamma, IL-17, and IL-21; drive inflammation-associated angiogenesis; and facilitate intimal hyperplasia. Conversely, chronic tissue inflammation in the atherosclerotic plaque is associated with an overreactive PD-1 checkpoint. Plaque-residing Mϕs are PD-L1hi , a defect induced by their addiction to glucose and glycolytic breakdown. PD-L1hi Mϕs render patients with coronary artery disease immunocompromised and suppress antiviral immunity, including protective anti-varicella zoster virus T cells. Thus, immunoinhibitory signals affect several domains of vascular inflammation; failing PD-L1 in vasculitis enables unopposed immunostimulation and opens the flood gates for polyfunctional inflammatory T cells, and excess PD-L1 in the atherosclerotic plaque disables tissue-protective T cell immunity.

    View details for PubMedID 28848042

  • Hypermetabolic macrophages in rheumatoid arthritis and coronary artery disease due to glycogen synthase kinase 3b inactivation. Annals of the rheumatic diseases Zeisbrich, M., Yanes, R. E., Zhang, H., Watanabe, R., Li, Y., Brosig, L., Hong, J., Wallis, B. B., Giacomini, J. C., Assimes, T. L., Goronzy, J. J., Weyand, C. M. 2018

    Abstract

    OBJECTIVES: Accelerated atherosclerotic disease typically complicates rheumatoid arthritis (RA), leading to premature cardiovascular death. Inflammatory macrophages are key effector cells in both rheumatoid synovitis and the plaques of coronary artery disease (CAD). Whether both diseases share macrophage-dependent pathogenic mechanisms is unknown.METHODS: Patients with RA or CAD (at least one myocardial infarction) and healthy age-matched controls were recruited into the study. Peripheral blood CD14+ monocytes were differentiated into macrophages. Metabolic profiles were assessed by Seahorse Analyzer, intracellular ATP concentrations were quantified and mitochondrial protein localisation was determined by confocal image analysis.RESULTS: In macrophages from patients with RA or CAD, mitochondria consumed more oxygen, generated more ATP and built tight interorganelle connections with the endoplasmic reticulum, forming mitochondria-associated membranes (MAM). Calcium transfer through MAM sites sustained mitochondrial hyperactivity and was dependent on inactivation of glycogen synthase kinase 3b (GSK3b), a serine/threonine kinase functioning as a metabolic switch. In patient-derived macrophages, inactivated pGSK3b-Ser9 co-precipitated with the mitochondrial fraction. Immunostaining of atherosclerotic plaques and synovial lesions confirmed that most macrophages had inactivated GSK3b. MAM formation and GSK3b inactivation sustained production of the collagenase cathepsin K, a macrophage effector function closely correlated with clinical disease activity in RA and CAD.CONCLUSIONS: Re-organisation of the macrophage metabolism in patients with RA and CAD drives unopposed oxygen consumption and ultimately, excessive production of tissue-destructive enzymes. The underlying molecular defect relates to the deactivation of GSK3b, which controls mitochondrial fuel influx and as such represents a potential therapeutic target for anti-inflammatory therapy.

    View details for PubMedID 29431119

  • Functional pathways regulated by microRNA networks in CD8 T-cell aging. Aging cell Gustafson, C. E., Cavanagh, M. M., Jin, J. n., Weyand, C. M., Goronzy, J. J. 2018: e12879

    Abstract

    One of the most prominent immunological changes during human aging is the alteration in CD8 T-cell subset distribution, predominated by a loss of naïve CD8 T cells. The molecular mechanisms that contribute to the loss of naïve CD8 T-cells during aging remain unclear. Considering that many CD8 T-cell functions are influenced by microRNAs (miRNAs), we explored miRNA expression profiling to identify novel dysfunctions that contribute to naïve CD8 T-cell loss during aging. Here, we describe age-dependent miRNA expression changes in naïve, central memory, and effector memory CD8 T-cell subsets. Changes in old naïve CD8 T-cells partially resembled those driven by an underlying shift in cellular differentiation toward a young central memory phenotype. Pathways enriched for targets of age-dependent miRNAs included FOXO1, NF-κB, and PI3K-AKT signaling. Transcriptome analysis of old naïve CD8 T-cells yielded corresponding patterns that correlated to those seen with reduced FOXO1 or altered NF-κB activities. Of particular interest, IL-7R expression, controlled by FOXO1 signaling, declines on naïve CD8 T cells with age and directly correlates with the frequencies of naïve CD8 T cells. Thus, age-associated changes in miRNA networks may ultimately contribute to the failure in CD8 T-cell homeostasis exemplified by the loss in naïve cells.

    View details for PubMedID 30488559

  • Comparative Analysis of the Macrophage Glycolytic Machinery in Giant Cell Arteritis (GCA) and in Coronary Artery Disease (CAD) Weyand, C. M., Watanabe, R., Shirai, T., Zhang, H., Berry, G., Goronzy, J. WILEY. 2017
  • Metabolic Activity Sustains Macrophage Cytokine Production in Rheumatoid Arthritis and Coronary Artery Disease Weyand, C. M., Zeisbrich, M., Brosig, L., Wallis, B., Roche, N., Lin, J., Goronzy, J. WILEY. 2017
  • Tissue-Invasive T Cells in Rheumatoid Arthritis Weyand, C. M., Shen, Y., Li, Y., Matteson, E. L., Goodman, S., Goronzy, J. WILEY. 2017
  • The Microvascular Niche Instructs Pathogenic T Cells in Medium and Large Vessel Vasculitis Weyand, C. M., Wen, Z., Shen, Y., Berry, G., Liao, J., Goronzy, J. WILEY. 2017
  • Immune Checkpoint Function of CD85j in CD8 T Cell Differentiation and Aging FRONTIERS IN IMMUNOLOGY Gustafson, C. E., Qi, Q., Hutter-Saunders, J., Gupta, S., Jadhav, R., Newell, E., Maecker, H., Weyand, C. M., Goronzy, J. J. 2017; 8: 692

    Abstract

    Aging is associated with an increased susceptibility to infection and a failure to control latent viruses thought to be driven, at least in part, by alterations in CD8 T cell function. The aging T cell repertoire is characterized by an accumulation of effector CD8 T cells, many of which express the negative regulatory receptor CD85j. To define the biological significance of CD85j expression on CD8 T cells and to address the question whether presence of CD85j in older individuals is beneficial or detrimental for immune function, we examined the specific attributes of CD8 T cells expressing CD85j as well as the functional role of CD85j in antigen-specific CD8 T cell responses during immune aging. Here, we show that CD85j is mainly expressed by terminally differentiated effector (TEMRAs) CD8 T cells, which increase with age, in cytomegalovirus (CMV) infection and in males. CD85j+ CMV-specific cells demonstrate clonal expansion. However, TCR diversity is similar between CD85j+ and CD85j- compartments, suggesting that CD85j does not directly impact the repertoire of antigen-specific cells. Further phenotypic and functional analyses revealed that CD85j identifies a specific subset of CMV-responsive CD8 T cells that coexpress a marker of senescence (CD57) but retain polyfunctional cytokine production and expression of cytotoxic mediators. Blocking CD85j binding enhanced proliferation of CMV-specific CD8 T cells upon antigen stimulation but did not alter polyfunctional cytokine production. Taken together, these data demonstrate that CD85j characterizes a population of "senescent," but not exhausted antigen-specific effector CD8 T cells and indicates that CD85j is an important checkpoint regulator controlling expansion of virus-specific T cells during aging. Inhibition of CD85j activity may be a mechanism to promote stronger CD8 T cell effector responses during immune aging.

    View details for PubMedID 28659925

  • Metabolic signatures of T-cells and macrophages in rheumatoid arthritis CURRENT OPINION IN IMMUNOLOGY Weyand, C. M., Zeisbrich, M., Goronzy, J. J. 2017; 46: 112–20

    Abstract

    In most autoimmune diseases, a decade-long defect in self-tolerance eventually leads to clinically relevant, tissue-destructive inflammatory disease. The pathogenic potential of chronic persistent immune responses during the pre-clinical and clinical phase is ultimately linked to the bioenergetic fitness of innate and adaptive immune cells. Chronic immune cell stimulation, high cellular turn-over, structural damage to the host tissue and maladaptive wound healing, all require a reliable supply of nutrients, oxygen, and biosynthetic precursors. Here, we use the model system of rheumatoid arthritis (RA) to discuss immunometabolism from the vantage point of T-cells and macrophages that encounter fundamentally different metabolic stress scenarios in the RA host. We outline the general principle that both insufficient nutrient supply, as well as nutrient excess generate cellular stress responses and guide immune function. ATPlow, NADPHhigh, ROSlow T-cells hyperproliferate and are forced into premature senescence. ATPhigh, ROShigh macrophages dimerize the glycolytic enzyme pyruvate kinase to amplify STAT3-dependent inflammatory effector functions. A corollary of this model is that simple nutraceutical interventions will be insufficient to re-educate the immune system in RA. Instead, interference with cell-type-exclusive and differentiation-stage-dependent metabolic setpoints will be needed to reprogram arthritogenic pathways.

    View details for PubMedID 28538163

    View details for PubMedCentralID PMC5554742

  • Immune checkpoint dysfunction in large and medium vessel vasculitis. American journal of physiology. Heart and circulatory physiology Watanabe, R., Zhang, H., Berry, G., Goronzy, J. J., Weyand, C. M. 2017; 312 (5): H1052-H1059

    Abstract

    Giant cell arteritis (GCA) is a granulomatous vasculitis of the aorta and its medium-sized branch vessels. CD4 T cells, macrophages, and dendritic cells (DCs) build granulomatous infiltrates that injure the vessel wall and elicit a maladaptive response to injury. Pathological consequences include fragmentation of elastic membranes, destruction of the medial layer, microvascular neoangiogenesis, massive outgrowth of myofibroblasts, and lumen-occlusive intimal hyperplasia. Antigens have been suspected to drive the local activation of vasculitogenic CD4 T cells, but recent data have suggested a more generalized defect in the threshold setting of such T cells, rendering them hyperreactive. Under physiological conditions, immune checkpoints provide negative signals to curb T cell activation and prevent inflammation-associated tissue destruction. This protective mechanism is disrupted in GCA. Vessel wall DCs fail to express the immunoinhibitory ligand programmed cell death ligand-1, leaving lesional T cells unchecked. Consequently, programmed cell death protein-1-positive CD4 T cells can enter the immunoprivileged vessel wall, where they produce a broad spectrum of inflammatory cytokines (interferon-γ, IL-17, and IL-21) and have a direct role in driving intimal hyperplasia and intramural neoangiogenesis. The deficiency of the programmed cell death protein-1 immune checkpoint in GCA, promoting unopposed T cell immunity, contrasts with checkpoint hyperactivity in cancer patients in whom excessive programmed cell death ligand-1 expression paralyzes the function of antitumor T cells. Excessive checkpoint activity is the principle underlying cancer-immune evasion and is therapeutically targeted by immunotherapy with checkpoint inhibitors. Such checkpoint inhibitors, which unleash anticancer T cells and induce immune-related toxicity, may lead to drug-induced vasculitis.

    View details for DOI 10.1152/ajpheart.00024.2017

    View details for PubMedID 28314758

  • Immunometabolism in early and late stages of rheumatoid arthritis NATURE REVIEWS RHEUMATOLOGY Weyand, C. M., Goronzy, J. J. 2017; 13 (5): 1-11

    Abstract

    One of the fundamental traits of immune cells in rheumatoid arthritis (RA) is their ability to proliferate, a property shared with the joint-resident cells that form the synovial pannus. The building of biomass imposes high demands for energy and biosynthetic precursors, implicating metabolic control as a basic disease mechanism. During preclinical RA, when autoreactive T cells expand and immunological tolerance is broken, the main sites of disease are the secondary lymphoid tissues. Naive CD4(+) T cells from patients with RA have a distinct metabolic signature, characterized by dampened glycolysis, low ATP levels and enhanced shunting of glucose into the pentose phosphate pathway. Equipped with high levels of NADPH and depleted of intracellular reactive oxygen species, such T cells hyperproliferate and acquire proinflammatory effector functions. During clinical RA, immune cells coexist with stromal cells in the acidic milieu of the inflamed joint. This microenvironment is rich in metabolic intermediates that are released into the extracellular space to shape cell-cell communication and the functional activity of tissue-resident cells. Increasing awareness of how metabolites regulate signalling pathways, guide post-translational modifications and condition the tissue microenvironment will help to connect environmental factors with the pathogenic behaviour of T cells in RA.

    View details for DOI 10.1038/nrrheum.2017.49

    View details for Web of Science ID 000399784500006

  • Immunometabolism in early and late stages of rheumatoid arthritis. Nature reviews. Rheumatology Weyand, C. M., Goronzy, J. J. 2017; 13 (5): 291-301

    Abstract

    One of the fundamental traits of immune cells in rheumatoid arthritis (RA) is their ability to proliferate, a property shared with the joint-resident cells that form the synovial pannus. The building of biomass imposes high demands for energy and biosynthetic precursors, implicating metabolic control as a basic disease mechanism. During preclinical RA, when autoreactive T cells expand and immunological tolerance is broken, the main sites of disease are the secondary lymphoid tissues. Naive CD4(+) T cells from patients with RA have a distinct metabolic signature, characterized by dampened glycolysis, low ATP levels and enhanced shunting of glucose into the pentose phosphate pathway. Equipped with high levels of NADPH and depleted of intracellular reactive oxygen species, such T cells hyperproliferate and acquire proinflammatory effector functions. During clinical RA, immune cells coexist with stromal cells in the acidic milieu of the inflamed joint. This microenvironment is rich in metabolic intermediates that are released into the extracellular space to shape cell-cell communication and the functional activity of tissue-resident cells. Increasing awareness of how metabolites regulate signalling pathways, guide post-translational modifications and condition the tissue microenvironment will help to connect environmental factors with the pathogenic behaviour of T cells in RA.

    View details for DOI 10.1038/nrrheum.2017.49

    View details for PubMedID 28360422

  • PKC-epsilon and TLR4 synergistically regulate resistin-mediated inflammation in human macrophages ATHEROSCLEROSIS Zuniga, M. C., Raghuraman, G., Hitchner, E., Weyand, C., Robinson, W., Zhou, W. 2017; 259: 51–59

    Abstract

    Resistin has been associated with atherosclerotic inflammation and cardiovascular complications. We and others have previously shown that PKC-epsilon (PKCε) is involved in resistin-induced smooth muscle cell (VSMC) dysfunction at a high pathological concentration. This study aimed to evaluate the role and potential pathways of resistin at a physiological concentration, in atherosclerosis-related inflammation.Plasma from patients with atherosclerosis was analyzed for resistin concentration. Patients were divided into tertiles based on resistin levels and cytokines were compared between tertiles. Macrophages were then treated with resistin in the presence or absence of PKCε inhibitor and/or TLR4 blocking-antibody, and their inflammatory state was evaluated with ELISA, RT-PCR, immunocytochemistry, and Western blot.We observed significant associations between plasma resistin levels and TNF-α, IL-6, IL-12, MIP-1α, MIP-1β, and CD40L. Our in vitro analyses revealed that resistin activated PKCε via TLR4. This was followed by NF-kB activation and induction of a pro-inflammatory phenotype in macrophages, significantly upregulating CD40, downregulating CD206 and stimulating gene expression and secretion of the inflammatory cytokines, for which we found association in our plasma analysis. Resistin also induced persistent TRAM and CD40L upregulation up to 36 h after resistin treatment. PKCε and TLR4 inhibitors suppressed gene expression to levels similar to control, especially when used in combination.Resistin, at a physiological concentration, exacerbates the inflammatory response of macrophages. PKCε is a key upstream mediator in resistin-induced inflammation that may interact synergistically with TLR4 to promote NF-kB activation, while TRAM is an important signal. PKCε and TRAM may represent novel molecular targets for resistin-associated chronic atherosclerotic inflammation.

    View details for PubMedID 28286252

    View details for PubMedCentralID PMC5395299

  • Response to Comment on "Diversification of the antigen-specific T cell receptor repertoire after varicella zoster vaccination". Science translational medicine Goronzy, J., Qi, Q., Weyand, C. 2017; 9 (382)

    Abstract

    In our repertoire studies of varicella zoster virus-specific T cells, we used very stringent computational criteria to keep contamination with T cell receptor sequences from bystander-activated T cells to a minimum.

    View details for PubMedID 28330866

  • Response to Comment on "Diversification of the antigen-specific T cell receptor repertoire after varicella zoster vaccination" SCIENCE TRANSLATIONAL MEDICINE Goronzy, J., Qi, Q., Weyand, C. 2017; 9 (382)
  • Successful and Maladaptive T Cell Aging IMMUNITY Goronzy, J. J., Weyand, C. M. 2017; 46 (3): 364-378

    Abstract

    Throughout life, the T cell system adapts to shifting resources and demands, resulting in a fundamentally restructured immune system in older individuals. Here we review the cellular and molecular features of an aged immune system and discuss the trade-offs inherent to these adaptive mechanisms. Processes include homeostatic proliferation that maintains compartment size at the expense of partial loss in stemness and incomplete differentiation and the activation of negative regulatory programs, which constrain effector T cell expansion and prevent increasing oligoclonality but also interfere with memory cell generation. We propose that immune failure occurs when adaptive strategies developed by the aging T cell system fail and also discuss how, in some settings, the programs associated with T cell aging culminates in a maladaptive response that directly contributes to chronic inflammatory disease.

    View details for DOI 10.1016/j.immuni.2017.03.010

    View details for Web of Science ID 000396818100010

    View details for PubMedID 28329703

  • Clinical and pathological evolution of giant cell arteritis: a prospective study of follow-up temporal artery biopsies in 40 treated patients. Modern pathology Maleszewski, J. J., Younge, B. R., Fritzlen, J. T., Hunder, G. G., Goronzy, J. J., Warrington, K. J., Weyand, C. M. 2017

    Abstract

    Although clinical signs and symptoms of giant cell arteritis improve promptly after starting glucocorticoid therapy, reports have suggested that the vascular inflammation may persist. To assess the duration and quality of histopathologic changes in treated patients, we prospectively obtained second temporal artery biopsies in patients treated for 3 to 12 months after their first diagnostic biopsy. Forty patients (28 women, 12 men, median age 77 years) agreed to have a second temporal artery biopsy randomly assigned to 3, 6, 9, or 12 months subsequent to the first. Clinical and laboratory evaluation of the patient cohort revealed a typical rapid response and continued suppression of clinical manifestations as a result of glucocorticoid treatment. Histopathologic findings, evaluated in a blinded manner by a cardiovascular pathologist, showed unequivocal findings of vasculitis in 7/10 patients with second temporal artery biopsy at 3 months, 9/12 at 6 months, 4/9 at 9 months, and 4/9 at 12 months. Lymphocytes were present in all positive initial biopsies and remained the dominant cell population in chronically treated patients. Granulomatous inflammation decreased in a time-dependent manner from 78 to 100% at initial biopsy to 50% at 9 months and 25% at 12 months. The increased medial fibrosis noted in the second biopsies (60 vs 33% in primary temporal artery biopsies) suggested that the finding may represent a chronic finding in arteritis. In summary, the response to glucocorticoids in giant cell arteritis was frequently discordant. Clinical manifestations were readily suppressed, but vascular changes were gradual and often incomplete.Modern Pathology advance online publication, 3 March 2017; doi:10.1038/modpathol.2017.10.

    View details for DOI 10.1038/modpathol.2017.10

    View details for PubMedID 28256573

  • Epigenomics of human CD8 T cell differentiation and aging. Science immunology Moskowitz, D. M., Zhang, D. W., Hu, B., Le Saux, S., Yanes, R. E., Ye, Z., Buenrostro, J. D., Weyand, C. M., Greenleaf, W. J., Goronzy, J. J. 2017; 2 (8)

    Abstract

    The efficacy of the adaptive immune response declines dramatically with age, but the cell-intrinsic mechanisms driving immune aging in humans remain poorly understood. Immune aging is characterized by a loss of self-renewing naïve cells and the accumulation of differentiated but dysfunctional cells within the CD8 T cell compartment. Using ATAC-seq, we inferred the transcription factor binding activities correlated with naive and central and effector memory CD8 T cell states in young adults. Integrating our results with RNA-seq, we identified transcription networks associated with CD8 T cell differentiation, with prominent roles implicated for BATF, ETS1, Eomes, and Sp1. Extending our analysis to aged humans, we found that the differences between the memory and naive subsets were largely preserved across age, but that naive and central memory cells from older individuals exhibited a shift toward more differentiated patterns of chromatin openness. Additionally, aged naive cells displayed a loss in chromatin accessibility at gene promoters, largely associated with a decrease in NRF1 binding. This shift was implicated in a marked drop-off in the ability of the aged naive cells to transcribe respiratory chain genes, which may explain the reduced capacity of oxidative phosphorylation in older naïve cells. Our findings identify BATF- and NRF1-driven gene regulation as potential targets for delaying CD8 T cell aging and restoring function.

    View details for DOI 10.1126/sciimmunol.aag0192

    View details for PubMedID 28439570

  • Epigenomics of human CD8 T cell differentiation and aging SCIENCE IMMUNOLOGY Moskowitz, D. M., Zhang, D. W., Hu, B., Le Saux, S., Yanes, R. E., Ye, Z., Buenrostro, J. D., Weyand, C. M., Greenleaf, W. J., Goronzy, J. J. 2017; 2 (8)
  • The life cycle of a T cell after vaccination - where does immune ageing strike? CLINICAL AND EXPERIMENTAL IMMUNOLOGY Kim, C., Fang, F., Weyand, C. M., Goronzy, J. J. 2017; 187 (1): 71-81

    Abstract

    Vaccination is the optimal intervention to prevent the increased morbidity and mortality from infection in older individuals and to maintain immune health during ageing. To optimize benefits from vaccination, strategies have to be developed that overcome the defects in an adaptive immune response that occur with immune ageing. Most current approaches are concentrated on activating the innate immune system by adjuvants to improve the induction of a T cell response. This review will focus upon T cell-intrinsic mechanisms that control how a T cell is activated, expands rapidly to differentiate into short-lived effector cells and into memory precursor cells, with short-lived effector T cells then mainly undergoing apoptosis and memory precursor cells surviving as long-lived memory T cells. Insights into each step of this longitudinal course of a T cell response that takes place over a period of several weeks is beginning to allow identifying interventions that can improve this process of T cell memory generation and specifically target defects that occur with ageing.

    View details for DOI 10.1111/cei.12829

    View details for Web of Science ID 000393964800010

    View details for PubMedCentralID PMC5167048

  • Pyruvate controls the checkpoint inhibitor PD-L1 and suppresses T cell immunity. The Journal of clinical investigation Watanabe, R. n., Shirai, T. n., Namkoong, H. n., Zhang, H. n., Berry, G. J., Wallis, B. B., Schaefgen, B. n., Harrison, D. G., Tremmel, J. A., Giacomini, J. C., Goronzy, J. J., Weyand, C. M. 2017; 127 (7): 2725–38

    Abstract

    Patients with coronary artery disease (CAD) are at high risk for reactivation of the varicella zoster virus (VZV) and development of herpes zoster (HZ). Here, we found that macrophages from patients with CAD actively suppress T cell activation and expansion, leading to defective VZV-specific T cell immunity. Monocyte-derived and plaque-infiltrating macrophages from patients with CAD spontaneously expressed high surface density of the immunoinhibitory ligand programmed death ligand-1 (PD-L1), thereby providing negative signals to programmed death-1+ (PD-1+) T cells. We determined that aberrant PD-L1 expression in patient-derived macrophages was metabolically controlled. Oversupply of the glycolytic intermediate pyruvate in mitochondria from CAD macrophages promoted expression of PD-L1 via induction of the bone morphogenetic protein 4/phosphorylated SMAD1/5/IFN regulatory factor 1 (BMP4/p-SMAD1/5/IRF1) signaling pathway. Thus, CAD macrophages respond to nutrient excess by activating the immunoinhibitory PD-1/PD-L1 checkpoint, leading to impaired T cell immunity. This finding indicates that metabolite-based immunotherapy may be a potential strategy for restoring adaptive immunity in CAD.

    View details for PubMedID 28604383

  • Lymphocyte generation and population homeostasis throughout life SEMINARS IN HEMATOLOGY Yanes, R. E., Gustafson, C. E., Weyand, C. M., Goronzy, J. J. 2017; 54 (1): 33-38

    Abstract

    Immune aging is a multi-faceted process that manifests as reduced competence to fight infections and malignant cells, as well as diminished tissue repair, unprovoked inflammation, and increased autoreactivity. The aging adaptive immune system, with its high complexity in functional cell subpopulations and diversity of B- and T-cell receptors, has to cope with the challenge of maintaining homeostasis while responding to exogenous stimuli and compensating for reduced generative capacity. With thymic involution, naïve T cells begin to function as quasi-stem cells and maintain the compartment through peripheral homeostatic proliferation that shapes the T-cell repertoire through peripheral selection and the activation of differentiation pathways. Similarly, reduced generation of early B-cell progenitors alters the composition of the peripheral B-cell compartment with the emergence of a unique, auto-inflammatory B-cell subset, termed age-associated B cells (ABCs). These changes in T- and B-cell composition and function are core manifestations of immune aging.

    View details for DOI 10.1053/j.seminhematol.2016.10.003

    View details for Web of Science ID 000393445800006

    View details for PubMedID 28088985

  • Aging of the Immune System. Mechanisms and Therapeutic Targets. Annals of the American Thoracic Society Weyand, C. M., Goronzy, J. J. 2016; 13: S422-S428

    Abstract

    Beginning with the sixth decade of life, the human immune system undergoes dramatic aging-related changes, which continuously progress to a state of immunosenescence. The aging immune system loses the ability to protect against infections and cancer and fails to support appropriate wound healing. Vaccine responses are typically impaired in older individuals. Conversely, inflammatory responses mediated by the innate immune system gain in intensity and duration, rendering older individuals susceptible to tissue-damaging immunity and inflammatory disease. Immune system aging functions as an accelerator for other age-related pathologies. It occurs prematurely in some clinical conditions, most prominently in patients with the autoimmune syndrome rheumatoid arthritis (RA); and such patients serve as an informative model system to study molecular mechanisms of immune aging. T cells from patients with RA are prone to differentiate into proinflammatory effector cells, sustaining chronic-persistent inflammatory lesions in the joints and many other organ systems. RA T cells have several hallmarks of cellular aging; most importantly, they accumulate damaged DNA. Because of deficiency of the DNA repair kinase ataxia telangiectasia mutated, RA T cells carry a higher burden of DNA double-strand breaks, triggering cell-indigenous stress signals that shift the cell's survival potential and differentiation pattern. Immune aging in RA T cells is also associated with metabolic reprogramming; specifically, with reduced glycolytic flux and diminished ATP production. Chronic energy stress affects the longevity and the functional differentiation of older T cells. Altered metabolic patterns provide opportunities to therapeutically target the immune aging process through metabolic interference.

    View details for DOI 10.1513/AnnalsATS.201602-095AW

    View details for PubMedID 28005419

    View details for PubMedCentralID PMC5291468

  • Editorial: Vascular inflammation in Systemic Autoimmunity FRONTIERS IN IMMUNOLOGY Ramirez, G. A., Weyand, C., Vaglio, A., Manfredi, A. A. 2016; 7: 471

    View details for PubMedID 27917171

  • Adaptive Immunity Dysregulation in Acute Coronary Syndromes From Cellular and Molecular Basis to Clinical Implications JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY Flego, D., Liuzzo, G., Weyand, C. M., Crea, F. 2016; 68 (19): 2107-2117

    Abstract

    Although the early outcome of acute coronary syndrome (ACS) has considerably improved in the last decade, cardiovascular diseases still represent the main cause of morbidity and mortality worldwide. This is mainly because recurrence of ACS eventually leads to the pandemics of heart failure and sudden cardiac death, thus calling for a reappraisal of the mechanisms responsible for coronary instability. This review discusses recent advances in our understanding of how adaptive immunity contributes to the pathogenesis of ACS and the clinical implications that arise from these new pathogenic concepts.

    View details for DOI 10.1016/j.jacc.2016.08.036

    View details for Web of Science ID 000386829300012

    View details for PubMedID 27810051

  • Defective T Memory Cell Differentiation after Varicella Zoster Vaccination in Older Individuals. PLoS pathogens Qi, Q., Cavanagh, M. M., Le Saux, S., Wagar, L. E., Mackey, S., Hu, J., Maecker, H., Swan, G. E., Davis, M. M., Dekker, C. L., Tian, L., Weyand, C. M., Goronzy, J. J. 2016; 12 (10)

    Abstract

    Vaccination with attenuated live varicella zoster virus (VZV) can prevent zoster reactivation, but protection is incomplete especially in an older population. To decipher the molecular mechanisms underlying variable vaccine responses, T- and B-cell responses to VZV vaccination were examined in individuals of different ages including identical twin pairs. Contrary to the induction of VZV-specific antibodies, antigen-specific T cell responses were significantly influenced by inherited factors. Diminished generation of long-lived memory T cells in older individuals was mainly caused by increased T cell loss after the peak response while the expansion of antigen-specific T cells was not affected by age. Gene expression in activated CD4 T cells at the time of the peak response identified gene modules related to cell cycle regulation and DNA repair that correlated with the contraction phase of the T cell response and consequently the generation of long-lived memory cells. These data identify cell cycle regulatory mechanisms as targets to reduce T cell attrition in a vaccine response and to improve the generation of antigen-specific T cell memory, in particular in an older population.

    View details for DOI 10.1371/journal.ppat.1005892

    View details for PubMedID 27764254

    View details for PubMedCentralID PMC5072604

  • Switching from Anabolic to Catabolic Metabolism - a Novel Immunomodulatory Therapy in RA Yang, Z., Shen, Y., Matteson, E. L., Hosgur, E., Hong, J., Goronzy, J., Weyand, C. M. WILEY. 2016
  • Pro-inflammatory and anti-inflammatory T cells in giant cell arteritis. Joint, bone, spine : revue du rhumatisme Watanabe, R., Hosgur, E., Zhang, H., Wen, Z., Berry, G., Goronzy, J. J., Weyand, C. M. 2016

    Abstract

    Giant cell arteritis is an autoimmune disease defined by explicit tissue tropism to the walls of medium and large arteries. Pathognomic inflammatory lesions are granulomatous in nature, emphasizing the functional role of CD4T cells and macrophages. Evidence for a pathogenic role of antibodies and immune complexes is missing. Analysis of T cell populations in giant cell arteritis, both in the tissue lesions and in the circulation, has supported a model of broad, polyclonal T cell activation, involving an array of functional T cell lineages. The signature of T cell cytokines produced by vasculitic lesions is typically multifunctional, including IL-2, IFN-γ, IL-17, IL-21, and GM-CSF, supportive for a general defect in T cell regulation. Recent data describing the lack of a lymph node-based population of anti-inflammatory T cells in giant cell arteritis patients offers a fresh look at the immunopathology of this vasculitis. Due to defective CD8(+)NOX2(+) regulatory T cells, giant cell arteritis patients appear unable to curtail clonal expansion within the CD4T cell compartment, resulting in widespread CD4T cell hyperimmunity. Why unopposed expansion of committed CD4 effector T cells would lead to invasion of the walls of medium and large arteries needs to be explored in further investigations.

    View details for DOI 10.1016/j.jbspin.2016.07.005

    View details for PubMedID 27663755

  • Activation of Human T Cells in Hypertension Studies of Humanized Mice and Hypertensive Humans HYPERTENSION Itani, H. A., McMaster, W. G., Saleh, M. A., Nazarewicz, R. R., Mikolajczyk, T. P., Kaszuba, A. M., Konior, A., Prejbisz, A., Januszewicz, A., Norlander, A. E., Chen, W., Bonami, R. H., Marshall, A. F., Poffenberger, G., Weyand, C. M., Madhur, M. S., Moore, D. J., Harrison, D. G., Guzik, T. J. 2016; 68 (1): 123-?

    Abstract

    Emerging evidence supports an important role for T cells in the genesis of hypertension. Because this work has predominantly been performed in experimental animals, we sought to determine whether human T cells are activated in hypertension. We used a humanized mouse model in which the murine immune system is replaced by the human immune system. Angiotensin II increased systolic pressure to 162 versus 116 mm Hg for sham-treated animals. Flow cytometry of thoracic lymph nodes, thoracic aorta, and kidney revealed increased infiltration of human leukocytes (CD45(+)) and T lymphocytes (CD3(+) and CD4(+)) in response to angiotensin II infusion. Interestingly, there was also an increase in the memory T cells (CD3(+)/CD45RO(+)) in the aortas and lymph nodes. Prevention of hypertension using hydralazine and hydrochlorothiazide prevented the accumulation of T cells in these tissues. Studies of isolated human T cells and monocytes indicated that angiotensin II had no direct effect on cytokine production by T cells or the ability of dendritic cells to drive T-cell proliferation. We also observed an increase in circulating interleukin-17A producing CD4(+) T cells and both CD4(+) and CD8(+) T cells that produce interferon-γ in hypertensive compared with normotensive humans. Thus, human T cells become activated and invade critical end-organ tissues in response to hypertension in a humanized mouse model. This response likely reflects the hypertensive milieu encountered in vivo and is not a direct effect of the hormone angiotensin II.

    View details for DOI 10.1161/HYPERTENSIONAHA.116.07237

    View details for PubMedID 27217403

  • The life cycle of a T cell after vaccination - where does immune ageing strike? Clinical and experimental immunology Kim, C., Fang, F., Weyand, C. M., Goronzy, J. J. 2016

    Abstract

    Vaccination is the optimal intervention to prevent the increased morbidity and mortality from infection in older individuals and to maintain immune health during ageing. To optimize benefits from vaccination, strategies have to be developed that overcome the defects in an adaptive immune response that occur with immune ageing. Most current approaches are concentrated on activating the innate immune system by adjuvants to improve the induction of a T cell response. This review will focus upon T cell-intrinsic mechanisms that control how a T cell is activated, expands rapidly to differentiate into short-lived effector cells and into memory precursor cells, with short-lived effector T cells then mainly undergoing apoptosis and memory precursor cells surviving as long-lived memory T cells. Insights into each step of this longitudinal course of a T cell response that takes place over a period of several weeks is beginning to allow identifying interventions that can improve this process of T cell memory generation and specifically target defects that occur with ageing.

    View details for DOI 10.1111/cei.12829

    View details for PubMedID 27324743

    View details for PubMedCentralID PMC5167048

  • Giant Cell Arteritis: From Pathogenesis to Therapeutic Management. Current treatment options in rheumatology Watanabe, R., Goronzy, J. J., Berry, G., Liao, Y. J., Weyand, C. M. 2016; 2 (2): 126-137

    View details for PubMedID 27298757

  • Systems analysis of microRNAs reveals a novel link between CD8 T cell dysfunction and TGF-beta signaling in immune aging Gustafson, C., Cavanagh, M., Hui, B., Zhang, D., Weyand, C. M., Goronzy, J. J. AMER ASSOC IMMUNOLOGISTS. 2016
  • Immune activation caused by vascular oxidation promotes fibrosis and hypertension. journal of clinical investigation Wu, J., Saleh, M. A., Kirabo, A., Itani, H. A., Montaniel, K. R., Xiao, L., Chen, W., Mernaugh, R. L., Cai, H., Bernstein, K. E., Goronzy, J. J., Weyand, C. M., Curci, J. A., Barbaro, N. R., Moreno, H., Davies, S. S., Roberts, L. J., Madhur, M. S., Harrison, D. G. 2016; 126 (4): 1607-?

    View details for DOI 10.1172/JCI87425

    View details for PubMedID 27035819

  • Diversification of the antigen-specific T cell receptor repertoire after varicella zoster vaccination. Science translational medicine Qi, Q., Cavanagh, M. M., Le Saux, S., Namkoong, H., Kim, C., Turgano, E., Liu, Y., Wang, C., Mackey, S., Swan, G. E., Dekker, C. L., Olshen, R. A., Boyd, S. D., Weyand, C. M., Tian, L., Goronzy, J. J. 2016; 8 (332): 332ra46-?

    Abstract

    Diversity and size of the antigen-specific T cell receptor (TCR) repertoire are two critical determinants for successful control of chronic infection. Varicella zoster virus (VZV) that establishes latency during childhood can escape control mechanisms, in particular with increasing age. We examined the TCR diversity of VZV-reactive CD4 T cells in individuals older than 50 years by studying three identical twin pairs and three unrelated individuals before and after vaccination with live attenuated VZV. Although all individuals had a small number of dominant T cell clones, the breadth of the VZV-specific repertoire differed markedly. A genetic influence was seen for the sharing of individual TCR sequences from antigen-reactive cells but not for repertoire richness or the selection of dominant clones. VZV vaccination favored the expansion of infrequent VZV antigen-reactive TCRs, including those from naïve T cells with lesser boosting of dominant T cell clones. Thus, vaccination does not reinforce the in vivo selection that occurred during chronic infection but leads to a diversification of the VZV-reactive T cell repertoire. However, a single-booster immunization seems insufficient to establish new clonal dominance. Our results suggest that repertoire analysis of antigen-specific TCRs can be an important readout to assess whether a vaccination was able to generate memory cells in clonal sizes that are necessary for immune protection.

    View details for DOI 10.1126/scitranslmed.aaf1725

    View details for PubMedID 27030598

  • Expression of CD39 on Activated T Cells Impairs their Survival in Older Individuals CELL REPORTS Fang, F., Yu, M., Cavanagh, M. M., Saunders, J. H., Qi, Q., Ye, Z., Le Saux, S., Sultan, W., Turgano, E., Dekker, C. L., Tian, L., Weyand, C. M., Goronzy, J. J. 2016; 14 (5): 1218-1231

    Abstract

    In an immune response, CD4(+) T cells expand into effector T cells and then contract to survive as long-lived memory cells. To identify age-associated defects in memory cell formation, we profiled activated CD4(+) T cells and found an increased induction of the ATPase CD39 with age. CD39(+) CD4(+) T cells resembled effector T cells with signs of metabolic stress and high susceptibility to undergo apoptosis. Pharmacological inhibition of ATPase activity dampened effector cell differentiation and improved survival, suggesting that CD39 activity influences T cell fate. Individuals carrying a low-expressing CD39 variant responded better to vaccination with an increase in vaccine-specific memory T cells. Increased inducibility of CD39 after activation may contribute to the impaired vaccine response with age.

    View details for DOI 10.1016/j.celrep.2016.01.002

    View details for Web of Science ID 000369616100022

  • Expression of CD39 on Activated T Cells Impairs their Survival in Older Individuals. Cell reports Fang, F., Yu, M., Cavanagh, M. M., Hutter Saunders, J., Qi, Q., Ye, Z., Le Saux, S., Sultan, W., Turgano, E., Dekker, C. L., Tian, L., Weyand, C. M., Goronzy, J. J. 2016; 14 (5): 1218-1231

    Abstract

    In an immune response, CD4(+) T cells expand into effector T cells and then contract to survive as long-lived memory cells. To identify age-associated defects in memory cell formation, we profiled activated CD4(+) T cells and found an increased induction of the ATPase CD39 with age. CD39(+) CD4(+) T cells resembled effector T cells with signs of metabolic stress and high susceptibility to undergo apoptosis. Pharmacological inhibition of ATPase activity dampened effector cell differentiation and improved survival, suggesting that CD39 activity influences T cell fate. Individuals carrying a low-expressing CD39 variant responded better to vaccination with an increase in vaccine-specific memory T cells. Increased inducibility of CD39 after activation may contribute to the impaired vaccine response with age.

    View details for DOI 10.1016/j.celrep.2016.01.002

    View details for PubMedID 26832412

  • Immune activation caused by vascular oxidation promotes fibrosis and hypertension JOURNAL OF CLINICAL INVESTIGATION Wu, J., Saleh, M. A., Kirabo, A., Itani, H. A., Montaniel, K. R., Xiao, L., Chen, W., Mernaugh, R. L., Cai, H., Bernstein, K. E., Goronzy, J. J., Weyand, C. M., Curci, J. A., Barbaro, N. R., Moreno, H., Davies, S. S., Roberts, L. J., Madhur, M. S., Harrison, D. G. 2016; 126 (1): 50-67

    Abstract

    Vascular oxidative injury accompanies many common conditions associated with hypertension. In the present study, we employed mouse models with excessive vascular production of ROS (tg(sm/p22phox) mice, which overexpress the NADPH oxidase subunit p22(phox) in smooth muscle, and mice with vascular-specific deletion of extracellular SOD) and have shown that these animals develop vascular collagen deposition, aortic stiffening, renal dysfunction, and hypertension with age. T cells from tg(sm/p22phox) mice produced high levels of IL-17A and IFN-γ. Crossing tg(sm/p22phox) mice with lymphocyte-deficient Rag1(-/-) mice eliminated vascular inflammation, aortic stiffening, renal dysfunction, and hypertension; however, adoptive transfer of T cells restored these processes. Isoketal-protein adducts, which are immunogenic, were increased in aortas, DCs, and macrophages of tg(sm/p22phox) mice. Autologous pulsing with tg(sm/p22phox) aortic homogenates promoted DCs of tg(sm/p22phox) mice to stimulate T cell proliferation and production of IFN-γ, IL-17A, and TNF-α. Treatment with the superoxide scavenger tempol or the isoketal scavenger 2-hydroxybenzylamine (2-HOBA) normalized blood pressure; prevented vascular inflammation, aortic stiffening, and hypertension; and prevented DC and T cell activation. Moreover, in human aortas, the aortic content of isoketal adducts correlated with fibrosis and inflammation severity. Together, these results define a pathway linking vascular oxidant stress to immune activation and aortic stiffening and provide insight into the systemic inflammation encountered in common vascular diseases.

    View details for DOI 10.1172/JCI80761

    View details for Web of Science ID 000367765600010

    View details for PubMedCentralID PMC4701546

  • Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition) AUTOPHAGY Klionsky, D. J., Abdelmohsen, K., Abe, A., Abedin, M. J., Abeliovich, H., Arozena, A. A., Adachi, H., Adams, C. M., Adams, P. D., Adeli, K., Adhihetty, P. J., Adler, S. G., Agam, G., Agarwal, R., Aghi, M. K., Agnello, M., Agostinis, P., Aguilar, P. V., Aguirre-Ghiso, J., Airoldi, E. M., Ait-Si-Ali, S., Akematsu, T., Akporiaye, E. T., Al-Rubeai, M., Albaiceta, G. M., Albanese, C., Albani, D., Albert, M. L., Aldudo, J., Alguel, H., Alirezaei, M., Alloza, I., Almasan, A., Almonte-Beceril, M., Alnemri, E. S., Alonso, C., Altan-Bonnet, N., Altieri, D. C., Alvarez, S., Alvarez-Erviti, L., Alves, S., Amadoro, G., Amano, A., Amantini, C., Ambrosio, S., Amelio, I., Amer, A. O., Amessou, M., Amon, A., An, Z., Anania, F. A., Andersen, S. U., Andley, U. P., Andreadi, C. K., Andrieu-Abadie, N., Anel, A., Ann, D. K., Anoopkumar-Dukie, S., Antonioli, M., Aoki, H., Apostolova, N., Aquila, S., Aquilano, K., Araki, K., Arama, E., Aranda, A., Araya, J., Arcaro, A., Arias, E., Arimoto, H., Ariosa, A. R., Armstrong, J. L., Arnould, T., Arsov, I., Asanuma, K., Askanas, V., Asselin, E., Atarashi, R., Atherton, S. S., Atkin, J. D., Attardi, L. D., Auberger, P., Auburger, G., Aurelian, L., Autelli, R., Avagliano, L., Avantaggiati, M. L., Avrahami, L., Awale, S., Azad, N., Bachetti, T., Backer, J. M., Bae, D., Bae, J., Bae, O., Bae, S. H., Baehrecke, E. H., Baek, S., Baghdiguian, S., Bagniewska-Zadworna, A., Bai, H., Bai, J., Bai, X., Bailly, Y., Balaji, K. N., Balduini, W., Ballabio, A., Balzan, R., Banerjee, R., Banhegyi, G., Bao, H., Barbeau, B., Barrachina, M. D., Barreiro, E., Bartel, B., Bartolome, A., Bassham, D. C., Bassi, M. T., Bast, R. C., Basu, A., Batista, M. T., Batoko, H., Battino, M., Bauckman, K., Baumgarner, B. L., Bayer, K. U., Beale, R., Beaulieu, J., Beck, G. R., Becker, C., Beckham, J. D., Bedard, P., Bednarski, P. J., Begley, T. J., Behl, C., Behrends, C., Behrens, G. M., Behrns, K. E., Bejarano, E., Belaid, A., Belleudi, F., Benard, G., Berchem, G., Bergamaschi, D., Bergami, M., Berkhout, B., Berliocchi, L., Bernard, A., Bernard, M., Bernassola, F., Bertolotti, A., Bess, A. S., Besteiro, S., Bettuzzi, S., Bhalla, S., Bhattacharyya, S., Bhutia, S. K., Biagosch, C., Bianchi, M. W., Biard-Piechaczyk, M., Billes, V., Bincoletto, C., Bingol, B., Bird, S. W., Bitoun, M., Bjedov, I., Blackstone, C., Blanc, L., Blanco, G. A., Blomhoff, H. K., Boada-Romero, E., Boeckler, S., Boes, M., Boesze-Battaglia, K., Boise, L. H., Bolino, A., Boman, A., Bonaldo, P., Bordi, M., Bosch, J., Botana, L. M., Botti, J., Bou, G., Bouche, M., Bouchecareilh, M., Boucher, M., Boulton, M. E., Bouret, S. G., Boya, P., Boyer-Guittaut, M., Bozhkov, P. V., Brady, N., Braga, V. M., Brancolini, C., Braus, G. H., Bravo-San Pedro, J. M., Brennan, L. A., Bresnick, E. H., Brest, P., Bridges, D., Bringer, M., Brini, M., Brito, G. C., Brodin, B., Brookes, P. S., Brown, E. J., Brown, K., Broxmeyer, H. E., Bruhat, A., Brum, P. C., Brumell, J. H., Brunetti-Pierri, N., Bryson-Richardson, R. J., Buch, S., Buchan, A. M., Budak, H., Bulavin, D. V., Bultman, S. J., Bultynck, G., Bumbasirevic, V., Burelle, Y., Burke, R. E., Burmeister, M., Buetikofer, P., Caberlotto, L., Cadwell, K., Cahova, M., Cai, D., Cai, J., Cai, Q., Calatayud, S., Camougrand, N., Campanella, M., Campbell, G. R., Campbell, M., Campello, S., Candau, R., Caniggia, I., Cantoni, L., Cao, L., Caplan, A. B., Caraglia, M., Cardinali, C., Cardoso, S. M., Carew, J. S., Carleton, L. A., Carlin, C. R., Carloni, S., Carlsson, S. R., Carmona-Gutierrez, D., Carneiro, L. A., Carnevali, O., Carra, S., Carrier, A., Carroll, B., Casas, C., Casas, J., Cassinelli, G., Castets, P., Castro-Obregon, S., Cavallini, G., Ceccherini, I., Cecconi, F., Cederbaum, A. I., Cena, V., Cenci, S., Cerella, C., Cervia, D., Cetrullo, S., Chaachouay, H., Chae, H., Chagin, A. S., Chai, C., Chakrabarti, G., Chamilos, G., Chan, E. Y., Chan, M. T., Chandra, D., Chandra, P., Chang, C., Chang, R. C., Chang, T. Y., Chatham, J. C., Chatterjee, S., Chauhan, S., Che, Y., Cheetham, M. E., Cheluvappa, R., Chen, C., Chen, G., Chen, G., Chen, G., Chen, H., Chen, J. W., Chen, J., Chen, M., Chen, M., Chen, P., Chen, Q., Chen, Q., Chen, S., Chen, S., Chen, S. S., Chen, W., Chen, W., Chen, W. Q., Chen, W., Chen, X., Chen, Y., Chen, Y., Chen, Y., Chen, Y., Chen, Y., Chen, Y., Chen, Y., Chen, Y., Chen, Z., Chen, Z., Cheng, A., Cheng, C. H., Cheng, H., Cheong, H., Cherry, S., Chesney, J., Cheung, C. H., Chevet, E., Chi, H. C., Chi, S., Chiacchiera, F., Chiang, H., Chiarelli, R., Chiariello, M., Chieppa, M., Chin, L., Chiong, M., Chiu, G. N., Cho, D., Cho, S., Cho, W. C., Cho, Y., Cho, Y., Choi, A. M., Choi, E., Choi, E., Choi, J., Choi, M. E., Choi, S., Chou, T., Chouaib, S., Choubey, D., Choubey, V., Chow, K., Chowdhury, K., Chu, C. T., Chuang, T., Chun, T., Chung, H., Chung, T., Chung, Y., Chwae, Y., Cianfanelli, V., Ciarcia, R., Ciechomska, I. A., Ciriolo, M. R., Cirone, M., Claerhout, S., Clague, M. J., Claria, J., Clarke, P. G., Clarke, R., Clementi, E., Cleyrat, C., Cnop, M., Coccia, E. M., Cocco, T., Codogno, P., Coers, J., Cohen, E. E., Colecchia, D., Coletto, L., Coll, N. S., Colucci-Guyon, E., Comincini, S., Condello, M., Cook, K. L., Coombs, G. H., Cooper, C. D., Cooper, J. M., Coppens, I., Corasaniti, M. T., Corazzari, M., Corbalan, R., Corcelle-Termeau, E., Cordero, M. D., Corral-Ramos, C., Corti, O., Cossarizza, A., Costelli, P., Costes, S., Costes, S., Coto-Montes, A., Cottet, S., Couve, E., Covey, L. R., Cowart, L. A., Cox, J. S., Coxon, F. P., Coyne, C. B., Cragg, M. S., Craven, R. J., Crepaldi, T., Crespo, J. L., Criollo, A., Crippa, V., Cruz, M. T., Cuervo, A. M., Cuezva, J. M., Cui, T., Cutillas, P. R., Czaja, M. J., Czyzyk-Krzeska, M. F., Dagda, R. K., Dahmen, U., Dai, C., Dai, W., Dai, Y., Dalby, K. N., Valle, L. D., Dalmasso, G., D'Amelio, M., Damme, M., Darfeuille-Michaud, A., Dargemont, C., Darley-Usmar, V. M., Dasarathy, S., Dasgupta, B., Dash, S., Dass, C. R., Davey, H. M., Davids, L. M., Davila, D., Davis, R. J., Dawson, T. M., Dawson, V. L., Daza, P., de Belleroche, J., de Figueiredo, P., Bressan Queiroz De Figueiredo, R. C., de la Fuente, J., De Martino, L., De Matteis, A., De Meyer, G. R., De Milito, A., De Santi, M., de Souza, W., De Tata, V., De Zio, D., Debnath, J., Dechant, R., Decuypere, J., Deegan, S., Dehay, B., Del Bello, B., Del Re, D. P., Delage-Mourroux, R., Delbridge, L. M., Deldicque, L., Delorme-Axford, E., Deng, Y., Dengjel, J., Denizot, M., Dent, P., Der, C. J., Deretic, V., Derrien, B., Deutsch, E., Devarenne, T. P., Devenish, R. J., Di Bartolomeo, S., Di Daniele, N., Di Domenico, F., Di Nardo, A., Di Paola, S., Di Pietro, A., Di Renzo, L., Diantonio, A., Diaz-Araya, G., Diaz-Laviada, I., Diaz-Meco, M. T., Diaz-Nido, J., Dickey, C. A., Dickson, R. C., Diederich, M., Digard, P., Dikic, I., Dinesh-Kumar, S. P., Ding, C., Ding, W., Ding, Z., Dini, L., Distler, J. H., Diwan, A., Djavaheri-Mergny, M., Dmytruk, K., Dobson, R. C., Doetsch, V., Dokladny, K., Dokudovskaya, S., Donadelli, M., Dong, X. C., Dong, X., Dong, Z., Donohue, T. M., Doran, K. S., D'Orazi, G., Dorn, G. W., Dosenko, V., Dridi, S., Drucker, L., Du, J., Du, L., Du, L., Du Toit, A., Dua, P., Duan, L., Duann, P., Dubey, V. K., Duchen, M. R., Duchosal, M. A., Duez, H., Dugail, I., Dumit, V. I., Duncan, M. C., Dunlop, E. A., Dunn, W. A., Dupont, N., Dupuis, L., Duran, R. V., Durcan, T. M., Duvezin-Caubet, S., Duvvuri, U., Eapen, V., Ebrahimi-Fakhari, D., Echard, A., Eckhart, L., Edelstein, C. L., Edinger, A. L., Eichinger, L., Eisenberg, T., Eisenberg-Lerner, A., Eissa, N. T., El-Deiry, W. S., El-Khoury, V., Elazar, Z., Eldar-Finkelman, H., Elliott, C. J., Emanuele, E., Emmenegger, U., Engedal, N., Engelbrecht, A., Engelender, S., Enserink, J. M., Erdmann, R., Erenpreisa, J., Eri, R., Eriksen, J. L., Erman, A., Escalante, R., Eskelinen, E., Espert, L., Esteban-Martinez, L., Evans, T. J., Fabri, M., Fabrias, G., Fabrizi, C., Facchiano, A., Faergeman, N. J., Faggioni, A., Fairlie, W. D., Fan, C., Fan, D., Fan, J., Fang, S., Fanto, M., Fanzani, A., Farkas, T., Faure, M., Favier, F. B., Fearnhead, H., Federici, M., Fei, E., Felizardo, T. C., Feng, H., Feng, Y., Feng, Y., Ferguson, T. A., Fernandez, A. F., Fernandez-Barrena, M. G., Fernandez-Checa, J. C., Fernandez-Lopez, A., Fernandez-Zapico, M. E., Feron, O., Ferraro, E., Ferreira-Halder, C. V., Fesus, L., Feuer, R., Fiesel, F. C., Filippi-Chiela, E. C., Filomeni, G., Fimia, G. M., Fingert, J. H., Finkbeiner, S., Finkel, T., Fiorito, F., Fisher, P. B., Flajolet, M., Flamigni, F., Florey, O., Florio, S., Floto, R. A., Folini, M., Follo, C., Fon, E. A., Fornai, F., Fortunato, F., Fraldi, A., Franco, R., Francois, A., Francois, A., Frankel, L. B., Fraser, I. D., Frey, N., Freyssenet, D. G., Frezza, C., Friedman, S. L., Frigo, D. E., Fu, D., Fuentes, J. M., Fueyo, J., Fujitani, Y., Fujiwara, Y., Fujiya, M., Fukuda, M., Fulda, S., Fusco, C., Gabryel, B., Gaestel, M., Gailly, P., Gajewska, M., Galadari, S., Galili, G., Galindo, I., Galindo, M. F., Galliciotti, G., Galluzzi, L., Galluzzi, L., Galy, V., Gammoh, N., Gandy, S., Ganesan, A. K., Ganesan, S., Ganley, I. G., Gannage, M., Gao, F., Gao, F., Gao, J., Garcia Nannig, L., Vescovi, E. G., Garcia-Macia, M., Garcia-Ruiz, C., Garg, A. D., Garg, P. K., Gargini, R., Gassen, N. C., Gatica, D., Gatti, E., Gavard, J., Gavathiotis, E., Ge, L., Ge, P., Ge, S., Gean, P., Gelmetti, V., Genazzani, A. A., Geng, J., Genschik, P., Gerner, L., Gestwicki, J. E., Gewirtz, D. A., Ghavami, S., Ghigo, E., Ghosh, D., Giammarioli, A. M., Giampieri, F., Giampietri, C., Giatromanolaki, A., Gibbings, D. J., Gibellini, L., Gibson, S. B., Ginet, V., Giordano, A., Giorgini, F., Giovannetti, E., Girardin, S. E., Gispert, S., Giuliano, S., Gladson, C. L., Glavic, A., Gleave, M., Godefroy, N., Gogal, R. M., Gokulan, K., Goldman, G. H., Goletti, D., Goligorsky, M. S., Gomes, A. V., Gomes, L. C., Gomez, H., Gomez-Manzano, C., Gomez-Sanchez, R., Goncalves, D. A., Goncu, E., Gong, Q., Gongora, C., Gonzalez, C. B., Gonzalez-Alegre, P., Gonzalez-Cabo, P., Ana Gonzalez-Polo, R., Goping, I. S., Gorbea, C., Gorbunov, N. V., Goring, D. R., Gorman, A. M., Gorski, S. M., Goruppi, S., Goto-Yamada, S., Gotor, C., Gottlieb, R. A., Gozes, I., Gozuacik, D., Graba, Y., Graef, M., Granato, G. E., Grant, G. D., Grant, S., Gravina, G. L., Green, D. R., Greenhough, A., Greenwood, M. T., Grimaldi, B., Gros, F., Grose, C., Groulx, J., Gruber, F., Grumati, P., Grune, T., Guan, J., Guan, K., Guerra, B., Guillen, C., Gulshan, K., Gunst, J., Guo, C., Guo, L., Guo, M., Guo, W., Guo, X., Gust, A. A., Gustafsson, A. B., Gutierrez, E., Gutierrez, M. G., Gwak, H., Haas, A., Haber, J. E., Hadano, S., Hagedorn, M., Hahn, D. R., Halayko, A. J., Hamacher-Brady, A., Hamada, K., Hamai, A., Hamann, A., Hamasaki, M., Hamer, I., Hamid, Q., Hammond, E. M., Han, F., Han, W., Handa, J. T., Hanover, J. A., Hansen, M., Harada, M., Harhaji-Trajkovic, L., Harper, J. W., Harrath, A. H., Harris, A. L., Harris, J., Hasler, U., Hasselblatt, P., Hasui, K., Hawley, R. G., Hawley, T. S., He, C., He, C. Y., He, F., He, G., He, R., He, X., He, Y., He, Y., Heath, J. K., Hebert, M., Heinzen, R. A., Helgason, G. V., Hensel, M., Henske, E. P., Her, C., Herman, P. K., Hernandez, A., Hernandez, C., Hernandez-Tiedra, S., Hetz, C., Hiesinger, P. R., Higaki, K., Hilfiker, S., Hill, B. G., Hill, J. A., Hill, W. D., Hino, K., Hofius, D., Hofman, P., Hoeglinger, G. U., Hoehfeld, J., Holz, M. K., Hong, Y., Hood, D. A., Hoozemans, J. J., Hoppe, T., Hsu, C., Hsu, C., Hsu, L., Hu, D., Hu, G., Hu, H., Hu, H., Hu, M. C., Hu, Y., Hu, Z., Hua, F., Hua, Y., Huang, C., Huang, H., Huang, K., Huang, K., Huang, S., Huang, S., Huang, W., Huang, Y., Huang, Y., Huang, Y., Huber, T. B., Huebbe, P., Huh, W., Hulmi, J. J., Hur, G. M., Hurley, J. H., Husak, Z., Hussain, S. N., Hussain, S., Hwang, J. j., Hwang, S., Hwang, T. I., Ichihara, A., Imai, Y., Imbriano, C., Inomata, M., Into, T., Iovane, V., Iovanna, J. L., Iozzo, R. V., Ip, N. Y., Irazoqui, J. E., Iribarren, P., Isaka, Y., Isakovic, A. J., Ischiropoulos, H., Isenberg, J. S., Ishaq, M., Ishida, H., Ishii, I., Ishmael, J. E., Isidoro, C., Isobe, K., Isono, E., Issazadeh-Navikas, S., Itahana, K., Itakura, E., Ivanov, A. I., Iyer, A. K., Izquierdo, J. M., Izumi, Y., Izzo, V., Jaeaettelae, M., Jaber, N., Jackson, D. J., Jackson, W. T., Jacob, T. G., Jacques, T. S., Jagannath, C., Jain, A., Jana, N. R., Jang, B. K., Jani, A., Janji, B., Jannig, P. R., Jansson, P. J., Jean, S., Jendrach, M., Jeon, J., Jessen, N., Jeung, E., Jia, K., Jia, L., Jiang, H., Jiang, H., Jiang, L., Jiang, T., Jiang, X., Jiang, X., Jiang, X., Jiang, Y., Jiang, Y., Jimenez, A., Jin, C., Jin, H., Jin, L., Jin, M., Jin, S., Jinwal, U. K., Jo, E., Johansen, T., Johnson, D. E., Johnson, G. V., Johnson, J. D., Jonasch, E., Jones, C., Joosten, L. A., Jordan, J., Joseph, A., Joseph, B., Joubert, A. M., Ju, D., Ju, J., Juan, H., Juenemann, K., Juhasz, G., Jung, H. S., Jung, J. U., Jung, Y., Jungbluth, H., Justice, M. J., Jutten, B., Kaakoush, N. O., Kaarniranta, K., Kaasik, A., Kabuta, T., Kaeffer, B., Kagedal, K., Kahana, A., Kajimura, S., Kakhlon, O., Kalia, M., Kalvakolanu, D. V., Kamada, Y., Kambas, K., Kaminskyy, V. O., Kampinga, H. H., Kandouz, M., Kang, C., Kang, R., Kang, T., Kanki, T., Kanneganti, T., Kanno, H., Kanthasamy, A. G., Kantorow, M., Kaparakis-Liaskos, M., Kapuy, O., Karantza, V., Karim, M. R., Karmakar, P., Kaser, A., Kaushik, S., Kawula, T., Kaynar, A. M., Ke, P., Ke, Z., Kehrl, J. H., Keller, K. E., Kemper, J. K., Kenworthy, A. K., Kepp, O., Kern, A., Kesari, S., Kessel, D., Ketteler, R., Kettelhut, I. D., Khambu, B., Khan, M. M., Khandelwal, V. K., Khare, S., Kiang, J. G., Kiger, A. A., Kihara, A., Kim, A. L., Kim, C. H., Kim, D. R., Kim, D., Kim, E. K., Kim, H. Y., Kim, H., Kim, J., Kim, J. H., Kim, J. C., Kim, J. H., Kim, K. W., Kim, M. D., Kim, M., Kim, P. K., Kim, S. W., Kim, S., Kim, Y., Kim, Y., Kimchi, A., Kimmelman, A. C., Kimura, T., King, J. S., Kirkegaard, K., Kirkin, V., Kirshenbaum, L. A., Kishi, S., Kitajima, Y., Kitamoto, K., Kitaoka, Y., Kitazato, K., Kley, R. A., Klimecki, W. T., Klinkenberg, M., Klucken, J., Knaevelsrud, H., Knecht, E., Knuppertz, L., Ko, J., Kobayashi, S., Koch, J. C., Koechlin-Ramonatxo, C., Koenig, U., Ko, Y. H., Koehler, K., Kohlwein, S. D., Koike, M., Komatsu, M., Kominami, E., Kong, D., Kong, H. J., Konstantakou, E. G., Kopp, B. T., Korcsmaros, T., Korhonen, L., Korolchuk, V. I., Koshkina, N. V., Kou, Y., Koukourakis, M. I., Koumenis, C., Kovacs, A. L., Kovacs, T., Kovacs, W. J., Koya, D., Kraft, C., Krainc, D., Kramer, H., Kravic-Stevovic, T., Krek, W., Kretz-Remy, C., Krick, R., Krishnamurthy, M., Kriston-Vizi, J., Kroemer, G., Kruer, M. C., Kruger, R., Ktistakis, N. T., Kuchitsu, K., Kuhn, C., Kumar, A. P., Kumar, A., Kumar, A., Kumar, D., Kumar, D., Kumar, R., Kumar, S., Kundu, M., Kung, H., Kuno, A., Kuo, S., Kuret, J., Kurz, T., Kwok, T., Kwon, T. K., Kwon, Y. T., Kyrmizi, I., La Spada, A. R., Lafont, F., Lahm, T., Lakkaraju, A., Lam, T., Lamark, T., Lancel, S., Landowski, T. H., Lane, D. J., Lane, J. D., Lanzi, C., Lapaquette, P., Lapierre, L. R., Laporte, J., Laukkarinen, J., Laurie, G. W., Lavandero, S., Lavie, L., LaVoie, M. J., Law, B. Y., Law, H. K., Law, K. B., Layfield, R., Lazo, P. A., Le Cam, L., Le Roch, K. G., Le Stunff, H., Leardkamolkarn, V., Lecuit, M., Lee, B., Lee, C., Lee, E. F., Lee, G. M., Lee, H., Lee, H., Lee, J. K., Lee, J., Lee, J., Lee, J. H., Lee, M., Lee, M., Lee, P. J., Lee, S. W., Lee, S., Lee, S., Lee, S. Y., Lee, S. H., Lee, S. S., Lee, S., Lee, S., Lee, Y., Lee, Y. J., Lee, Y. H., Leeuwenburgh, C., Lefort, S., Legouis, R., Lei, J., Lei, Q., Leib, D. A., Leibowitz, G., Lekli, I., Lemaire, S. D., Lemasters, J. J., Lemberg, M. K., Lemoine, A., Leng, S., Lenz, G., Lenzi, P., Lerman, L. O., Barbato, D. L., Leu, J. I., Leung, H. Y., Levine, B., Lewis, P. A., Lezoualc'h, F., Li, C., Li, F., Li, F., Li, J., Li, K., Li, L., Li, M., Li, M., Li, Q., Li, R., Li, S., Li, W., Li, W., Li, X., Li, Y., Lian, J., Liang, C., Liang, Q., Liao, Y., Liberal, J., Liberski, P. P., Lie, P., Lieberman, A. P., Lim, H. J., Lim, K., Lim, K., Lima, R. T., Lin, C., Lin, C., Lin, F., Lin, F., Lin, F., Lin, K., Lin, K., Lin, P., Lin, T., Lin, W., Lin, Y., Lin, Y., Linden, R., Lindholm, D., Lindqvist, L. M., Lingor, P., Linkermann, A., Liotta, L. A., Lipinski, M. M., Lira, V. A., Lisanti, M. P., Liton, P. B., Liu, B., Liu, C., Liu, C., Liu, F., Liu, H., Liu, J., Liu, J., Liu, J., Liu, K., Liu, L., Liu, L., Liu, Q., Liu, R., Liu, S., Liu, S., Liu, W., Liu, X., Liu, X., Liu, X., Liu, X., Liu, X., Liu, X., Liu, Y., Liu, Y., Liu, Z., Liu, Z., Liuzzi, J. P., Lizard, G., Ljujic, M., Lodhi, I. J., Logue, S. E., Lokeshwar, B. L., Long, Y. C., Lonial, S., Loos, B., Lopez-Otin, C., Lopez-Vicario, C., Lorente, M., Lorenzi, P. L., Lorincz, P., Los, M., Lotze, M. T., Lovat, P. E., Lu, B., Lu, B., Lu, J., Lu, Q., Lu, S., Lu, S., Lu, Y., Luciano, F., Luckhart, S., Lucocq, J. M., Ludovico, P., Lugea, A., Lukacs, N. W., Lum, J. J., Lund, A. H., Luo, H., Luo, J., Luo, S., Luparello, C., Lyons, T., Ma, J., Ma, Y., Ma, Y., Ma, Z., Machado, J., Machado-Santelli, G. M., Macian, F., MacIntosh, G. C., MacKeigan, J. P., Macleod, K. F., MacMicking, J. D., MacMillan-Crow, L. A., Madeo, F., Madesh, M., Madrigal-Matute, J., Maeda, A., Maeda, T., Maegawa, G., Maellaro, E., Maes, H., Magarinos, M., Maiese, K., Maiti, T. K., Maiuri, L., Maiuri, M. C., Maki, C. G., Malli, R., Malorni, W., Maloyan, A., Mami-Chouaib, F., Man, N., Mancias, J. D., Mandelkow, E., Mandell, M. A., Manfredi, A. A., Manie, S. N., Manzoni, C., Mao, K., Mao, Z., Mao, Z., Marambaud, P., Marconi, A. M., Marelja, Z., Marfe, G., Margeta, M., Margittai, E., Mari, M., Mariani, F. V., Marin, C., Marinelli, S., Marino, G., Markovic, I., Marquez, R., Martelli, A. M., Martens, S., Martin, K. R., Martin, S. J., Martin, S., Martin-Acebes, M. A., Martin-Sanz, P., Martinand-Mari, C., Martinet, W., Martinez, J., Martinez-Lopez, N., Martinez-Outschoorn, U., Martinez-Velazquez, M., Martinez-Vicente, M., Martins, W. K., Mashima, H., Mastrianni, J. A., Matarese, G., Matarrese, P., Mateo, R., Matoba, S., Matsumoto, N., Matsushita, T., Matsuura, A., Matsuzawa, T., Mattson, M. P., Matus, S., Maugeri, N., Mauvezin, C., Mayer, A., Maysinger, D., Mazzolini, G. D., McBrayer, M. K., McCall, K., McCormick, C., McInerney, G. M., McIver, S. C., McKenna, S., McMahon, J. J., McNeish, I. A., Mechta-Grigoriou, F., Medema, J. P., Medina, D. L., Megyeri, K., Mehrpour, M., Mehta, J. L., Mei, Y., Meier, U., Meijer, A. J., Melendez, A., Melino, G., Melino, S., Tenorio de Melo, E. J., Mena, M. A., Meneghini, M. D., Menendez, J. A., Menezes, R., Meng, L., Meng, L., Meng, S., Menghini, R., Menko, A. S., Menna-Barreto, R. F., Menon, M. B., Meraz-Rios, M. A., Merla, G., Merlini, L., Merlot, A. M., Meryk, A., Meschini, S., Meyer, J. N., Mi, M., Miao, C., Micale, L., Michaeli, S., Michiels, C., Migliaccio, A. R., Mihailidou, A. S., Mijaljica, D., Mikoshiba, K., Milan, E., Miller-Fleming, L., Mills, G. B., Mills, I. G., Minakaki, G., Minassian, B. A., Ming, X., Minibayeva, F., Minina, E. A., Mintern, J. D., Minucci, S., Miranda-Vizuete, A., Mitchell, C. H., Miyamoto, S., Miyazawa, K., Mizushima, N., Mnich, K., Mograbi, B., Mohseni, S., Moita, L. F., Molinari, M., Molinari, M., Moller, A. B., Mollereau, B., Mollinedo, F., Monick, M. M., Monick, M. M., Montagnaro, S., Montell, C., Moore, D. J., Moore, M. N., Mora-Rodriguez, R., Moreira, P. I., Morel, E., Morelli, M. B., Moreno, S., Morgan, M. J., Moris, A., Moriyasu, Y., Morrison, J. L., Morrison, L. A., Morselli, E., Moscat, J., Moseley, P. L., Mostowy, S., Motori, E., Mottet, D., Mottram, J. C., Moussa, C. E., Mpakou, V. E., Mukhtar, H., Levy, J. M., Muller, S., Munoz-Moreno, R., Munoz-Pinedo, C., Muenz, C., Murphy, M. E., Murray, J. T., Murthy, A., Mysorekar, I. U., Nabi, I. R., Nabissi, M., Nader, G. A., Nagahara, Y., Nagai, Y., Nagata, K., Nagelkerke, A., Nagy, P., Naidu, S. R., Nair, S., Nakano, H., Nakatogawa, H., Nanjundan, M., Napolitano, G., Naqvi, N. I., Nardacci, R., Narendra, D. P., Narita, M., Nascimbeni, A. C., Natarajan, R., Navegantes, L. C., Nawrocki, S. T., Nazarko, T. Y., Nazarko, V. Y., Neill, T., Neri, L. M., Netea, M. G., Netea-Maier, R. T., Neves, B. M., Ney, P. A., Nezis, I. P., Nguyen, H. T., Huu Phuc Nguyen, H. P., Nicot, A., Nilsen, H., Nilsson, P., Nishimura, M., Nishino, I., Niso-Santano, M., Niu, H., Nixon, R. A., Njar, V. C., Noda, T., Noegel, A. A., Nolte, E. M., Norberg, E., Norga, K. K., Noureini, S. K., Notomi, S., Notterpek, L., Nowikovsky, K., Nukina, N., Nuernberger, T., O'Donnell, V. B., O'Donovan, T., O'Dwyer, P. J., Oehme, I., Oeste, C. L., Ogawa, M., Ogretmen, B., Ogura, Y., Oh, Y. J., Ohmuraya, M., Ohshima, T., Ojha, R., Okamoto, K., Okazaki, T., Oliver, F. J., Ollinger, K., Olsson, S., Orban, D. P., Ordonez, P., Orhon, I., Orosz, L., O'Rourke, E. J., Orozco, H., Ortega, A. L., Ortona, E., Osellame, L. D., Oshima, J., Oshima, S., Osiewacz, H. D., Otomo, T., Otsu, K., Ou, J. J., Outeiro, T. F., Ouyang, D., Ouyang, H., Overholtzer, M., Ozbun, M. A., Ozdinler, P. H., Ozpolat, B., Pacelli, C., Paganetti, P., Page, G., Pages, G., Pagnini, U., Pajak, B., Pak, S. C., Pakos-Zebrucka, K., Pakpour, N., Palkova, Z., Palladino, F., Pallauf, K., Pallet, N., Palmieri, M., Paludan, S. R., Palumbo, C., Palumbo, S., Pampliega, O., Pan, H., Pan, W., Panaretakis, T., Pandey, A., Pantazopoulou, A., Papackova, Z., Papademetrio, D. L., Papassideri, I., Papini, A., Parajuli, N., Pardo, J., Parekh, V. V., Parenti, G., Park, J., Park, J., Park, O. K., Parker, R., Parlato, R., Parys, J. B., Parzych, K. R., Pasquet, J., Pasquier, B., Pasumarthi, K. B., Patschan, D., Patterson, C., Pattingre, S., Pattison, S., Pause, A., Pavenstaedt, H., Pavone, F., Pedrozo, Z., Pena, F. J., Penalva, M. A., Pende, M., Peng, J., Penna, F., Penninger, J. M., Pensalfini, A., Pepe, S., Pereira, G. J., Pereira, P. C., Perez-De La Cruz, V., Esther Perez-Perez, M., Perez-Rodriguez, D., Perez-Sala, D., Perier, C., Perl, A., Perlmutter, D. H., Perrotta, I., Pervaiz, S., Pesonen, M., Pessin, J. E., Peters, G. J., Petersen, M., Petrache, I., Petrof, B. J., Petrovski, G., Phang, J. M., Piacentini, M., Pierdominici, M., Pierre, P., Pierrefite-Carle, V., Pietrocola, F., Pimentel-Muinos, F. X., Pinar, M., Pineda, B., Pinkas-Kramarski, R., Pinti, M., Pinton, P., Piperdi, B., Piret, J. M., Platanias, L. C., Platta, H. W., Plowey, E. D., Poggeler, S., Poirot, M., Polic, P., Poletti, A., Poon, A. H., Popelka, H., Popova, B., Poprawa, I., Poulose, S. M., Poulton, J., Powers, S. K., Powers, T., Pozuelo-Rubio, M., Prak, K., Prange, R., Prescott, M., Priault, M., Prince, S., Proia, R. L., Proikas-Cezanne, T., Prokisch, H., Promponas, V. J., Przyklenk, K., Puertollano, R., Pugazhenthi, S., Puglielli, L., Pujol, A., Puyal, J., Pyeon, D., Qi, X., Qian, W., Qin, Z., Qiu, Y., Qu, Z., Quadrilatero, J., Quinn, F., Raben, N., Rabinowich, H., Radogna, F., Ragusa, M. J., Rahmani, M., Raina, K., Ramanadham, S., Ramesh, R., Rami, A., Randall-Demllo, S., Randow, F., Rao, H., Rao, V. A., Rasmussen, B. B., Rasse, T. M., Ratovitski, E. A., Rautou, P., Ray, S. K., Razani, B., Reed, B. H., Reggiori, F., Rehm, M., Reichert, A. S., Rein, T., Reiner, D. J., Reits, E., Ren, J., Ren, X., Renna, M., Reusch, J. E., Revuelta, J. L., Reyes, L., Rezaie, A. R., Richards, R. I., Richardson, D. R., Richetta, C., Riehle, M. A., Rihn, B. H., Rikihisa, Y., Riley, B. E., Rimbach, G., Rippo, M. R., Ritis, K., Rizzi, F., Rizzo, E., Roach, P. J., Robbins, J., Roberge, M., Roca, G., Roccheri, M. C., Rocha, S., Rodrigues, C. M., Rodriguez, C. I., Rodriguez de Cordoba, S., Rodriguez-Muela, N., Roelofs, J., Rogov, V. V., Rohn, T. T., Rohrer, B., Romanelli, D., Romani, L., Silvia Romano, P., Roncero, M. I., Luis Rosa, J., Rosello, A., Rosen, K. V., Rosenstiel, P., Rost-Roszkowska, M., Roth, K. A., Roue, G., Rouis, M., Rouschop, K. M., Ruan, D. T., Ruano, D., Rubinsztein, D. C., Rucker, E. B., Rudich, A., Rudolf, E., Rudolf, R., Ruegg, M. A., Ruiz-Roldan, C., Ruparelia, A. A., Rusmini, P., Russ, D. W., Russo, G. L., Russo, G., Russo, R., Rusten, T. E., Ryabovol, V., Ryan, K. M., Ryter, S. W., Sabatini, D. M., Sacher, M., Sachse, C., Sack, M. N., Sadoshima, J., Saftig, P., Sagi-Eisenberg, R., Sahni, S., Saikumar, P., Saito, T., Saitoh, T., Sakakura, K., Sakoh-Nakatogawa, M., Sakuraba, Y., Salazar-Roa, M., Salomoni, P., Saluja, A. K., Salvaterra, P. M., Salvioli, R., Samali, A., Sanchez, A. M., Sanchez-Alcazar, J. A., Sanchez-Prieto, R., Sandri, M., Sanjuan, M. A., Santaguida, S., Santambrogio, L., Santoni, G., dos Santos, C. N., Saran, S., Sardiello, M., Sargent, G., Sarkar, P., Sarkar, S., Sarrias, M. R., Sarwal, M. M., Sasakawa, C., Sasaki, M., Sass, M., Sato, K., Sato, M., Satriano, J., Savaraj, N., Saveljeva, S., Schaefer, L., Schaible, U. E., Scharl, M., Schatzl, H. M., Schekman, R., Scheper, W., Schiavi, A., Schipper, H. M., Schmeisser, H., Schmidt, J., Schmitz, I., Schneider, B. E., Schneider, E. M., Schneider, J. L., Schon, E. A., Schoenenberger, M. J., Schoenthal, A. H., Schorderet, D. F., Schroeder, B., Schuck, S., Schulze, R. J., Schwarten, M., Schwarz, T. L., Sciarretta, S., Scotto, K., Scovassi, A. I., Screaton, R. A., Screen, M., Seca, H., Sedej, S., Segatori, L., Segev, N., Seglen, P. O., Segui-Simarro, J. M., Segura-Aguilar, J., Seiliez, I., Seki, E., Sell, C., Semenkovich, C. F., Semenza, G. L., Sen, U., Serra, A. L., Serrano-Puebla, A., Sesaki, H., Setoguchi, T., Settembre, C., Shacka, J. J., Shajahan-Haq, A. N., Shapiro, I. M., Sharma, S., She, H., Shen, C. J., Shen, C., Shen, H., Shen, S., Shen, W., Sheng, R., Sheng, X., Sheng, Z., Shepherd, T. G., Shi, J., Shi, Q., Shi, Q., Shi, Y., Shibutani, S., Shibuya, K., Shidoji, Y., Shieh, J., Shih, C., Shimada, Y., Shimizu, S., Shin, D. W., Shinohara, M. L., Shintani, M., Shintani, T., Shioi, T., Shirabe, K., Shiri-Sverdlov, R., Shirihai, O., Shore, G. C., Shu, C., Shukla, D., Sibirny, A. A., Sica, V., Sigurdson, C. J., Sigurdsson, E. M., Sijwali, P. S., Sikorska, B., Silveira, W. A., Silvente-Poirot, S., Silverman, G. A., Simak, J., Simmet, T., Simon, A. K., Simon, H., Simone, C., Simons, M., Simonsen, A., Singh, R., Singh, S. V., Singh, S. K., Sinha, D., Sinha, S., Sinicrope, F. A., Sirko, A., Sirohi, K., Sishi, B. J., Sittler, A., Siu, P. M., Sivridis, E., Skwarska, A., Slack, R., Slaninova, I., Slavov, N., Smaili, S. S., Smalley, K. S., Smith, D. R., Soenen, S. J., Soleimanpour, S. A., Solhaug, A., Somasundaram, K., Son, J. H., Sonawane, A., Song, C., Song, F., Song, H. K., Song, J., Song, W., Soo, K. Y., Sood, A. K., Soong, T. W., Soontornniyomkij, V., Sorice, M., Sotgia, F., Soto-Pantoja, D. R., Sotthibundhu, A., Sousa, M. J., Spaink, H. P., Span, P. N., Spang, A., Sparks, J. D., Speck, P. G., Spector, S. A., Spies, C. D., Springer, W., St Clair, D., Stacchiotti, A., Staels, B., Stang, M. T., Starczynowski, D. T., Starokadomskyy, P., Steegborn, C., Steele, J. W., Stefanis, L., Steffan, J., Stellrecht, C. M., Stenmark, H., Stepkowski, T. M., Stern, S. T., Stevens, C., Stockwell, B. R., Stoka, V., Storchova, Z., Stork, B., Stratoulias, V., Stravopodis, D. J., Strnad, P., Strohecker, A. M., Stroem, A., Stromhaug, P., Stulik, J., Su, Y., Su, Z., Subauste, C. S., Subramaniam, S., Sue, C. M., Suh, S. W., Sui, X., Sukseree, S., Sulzer, D., Sun, F., Sun, J., Sun, J., Sun, S., Sun, Y., Sun, Y., Sun, Y., Sundaramoorthy, V., Sung, J., Suzuki, H., Suzuki, K., Suzuki, N., Suzuki, T., Suzuki, Y. J., Swanson, M. S., Swanton, C., Swaerd, K., Swarup, G., Sweeney, S. T., Sylvester, P. W., Szatmari, Z., Szegezdi, E., Szlosarek, P. W., Taegtmeyer, H., Tafani, M., Taillebourg, E., Tait, S. W., Takacs-Vellai, K., Takahashi, Y., Takats, S., Takemura, G., Takigawa, N., Talbot, N. J., Tamagno, E., Tamburini, J., Tan, C., Tan, L., Tan, M. L., Tan, M., Tan, Y., Tanaka, K., Tanaka, M., Tang, D., Tang, D., Tang, G., Tanida, I., Tanji, K., Tannous, B. A., Tapia, J. A., Tasset-Cuevas, I., Tatar, M., Tavassoly, I., Tavernarakis, N., Taylor, A., Taylor, G. S., Taylor, G. A., Taylor, J. P., Taylor, M. J., Tchetina, E. V., Tee, A. R., Teixeira-Clerc, F., Telang, S., Tencomnao, T., Teng, B., Teng, R., Terro, F., Tettamanti, G., Theiss, A. L., Theron, A. E., Thomas, K. J., Thome, M. P., Thomes, P. G., Thorburn, A., Thorner, J., Thum, T., Thumm, M., Thurston, T. L., Tian, L., Till, A., Ting, J. P., Titorenko, V. I., Toker, L., Toldo, S., Tooze, S. A., Topisirovic, I., Torgersen, M. L., Torosantucci, L., Torriglia, A., Torrisi, M. R., Tournier, C., Towns, R., Trajkovic, V., Travassos, L. H., Triola, G., Tripathi, D. N., Trisciuoglio, D., Troncoso, R., Trougakos, I. P., Truttmann, A. C., Tsai, K., Tschan, M. P., Tseng, Y., Tsukuba, T., Tsung, A., Tsvetkov, A. S., Tu, S., Tuan, H., Tucci, M., Tumbarello, D. A., Turk, B., Turk, V., Turner, R. F., Tveita, A. A., Tyagi, S. C., Ubukata, M., Uchiyama, Y., Udelnow, A., Ueno, T., Umekawa, M., Umemiya-Shirafuji, R., Underwood, B. R., Ungermann, C., Ureshino, R. P., Ushioda, R., Uversky, V. N., Uzcategui, N. L., Vaccari, T., Vaccaro, M. I., Vachova, L., Vakifahmetoglu-Norberg, H., Valdor, R., Valente, E. M., Vallette, F., Valverde, A. M., Van den Berghe, G., Van Den Bosch, L., van den Brink, G. R., van der Goot, F. G., van der Klei, I. J., van der Laan, L. J., van Doorn, W. G., van Egmond, M., van Golen, K. L., Van Kaer, L., Campagne, M. v., Vandenabeele, P., Vandenberghe, W., Vanhorebeek, I., Varela-Nieto, I., Vasconcelos, M. H., Vasko, R., Vavvas, D. G., Vega-Naredo, I., Velasco, G., Velentzas, A. D., Velentzas, P. D., Vellai, T., Vellenga, E., Vendelbo, M. H., Venkatachalam, K., Ventura, N., Ventura, S., Veras, P. S., Verdier, M., Vertessy, B. G., Viale, A., Vidal, M., Vieira, H. L., Vierstra, R. D., Vigneswaran, N., Vij, N., Vila, M., Villar, M., Villar, V. H., Villarroya, J., Vindis, C., Viola, G., Viscomi, M. T., Vitale, G., Vogl, D. T., Voitsekhovskaja, O. V., von Haefen, C., von Schwarzenberg, K., Voth, D. E., Vouret-Craviari, V., Vuori, K., Vyas, J. M., Waeber, C., Walker, C. L., Walker, M. J., Walter, J., Wan, L., Wan, X., Wang, B., Wang, C., Wang, C., Wang, C., Wang, C., Wang, C., Wang, D., Wang, F., Wang, F., Wang, G., Wang, H., Wang, H., Wang, H., Wang, H., Wang, H., Wang, J., Wang, J., Wang, M., Wang, M., Wang, P., Wang, P., Wang, R. C., Wang, S., Wang, T., Wang, X., Wang, X., Wang, X., Wang, X., Wang, X., Wang, Y., Wang, Y., Wang, Y., Wang, Y., Wang, Y., Wang, Y., Wang, Y. T., Wang, Y., Wang, Z., Wappner, P., Ward, C., Ward, D. M., Warnes, G., Watada, H., Watanabe, Y., Watase, K., Weaver, T. E., Weekes, C. D., Wei, J., Weide, T., Weihl, C. C., Weindl, G., Weis, S. N., Wen, L., Wen, X., Wen, Y., Westermann, B., Weyand, C. M., White, A. R., White, E., Whitton, J. L., Whitworth, A. J., Wiels, J., Wild, F., Wildenberg, M. E., Wileman, T., Wilkinson, D. S., Wilkinson, S., Willbold, D., Williams, C., Williams, K., Williamson, P. R., Winklhofer, K. F., Witkin, S. S., Wohlgemuth, S. E., Wollert, T., Wolvetang, E. J., Wong, E., Wong, G. W., Wong, R. W., Wong, V. K., Woodcock, E. A., Wright, K. L., Wu, C., Wu, D., Wu, G. S., Wu, J., Wu, J., Wu, M., Wu, M., Wu, S., Wu, W. K., Wu, Y., Wu, Z., Xavier, C. P., Xavier, R. J., Xia, G., Xia, T., Xia, W., Xia, Y., Xiao, H., Xiao, J., Xiao, S., Xiao, W., Xie, C., Xie, Z., Xie, Z., Xilouri, M., Xiong, Y., Xu, C., Xu, C., Xu, F., Xu, H., Xu, H., Xu, J., Xu, J., Xu, J., Xu, L., Xu, X., Xu, Y., Xu, Y., Xu, Z., Xu, Z., Xue, Y., Yamada, T., Yamamoto, A., Yamanaka, K., Yamashina, S., Yamashiro, S., Yan, B., Yan, B., Yan, X., Yan, Z., Yanagi, Y., Yang, D., Yang, J., Yang, L., Yang, M., Yang, P., Yang, P., Yang, Q., Yang, W., Yang, W. Y., Yang, X., Yang, Y., Yang, Y., Yang, Z., Yang, Z., Yao, M., Yao, P. J., Yao, X., Yao, Z., Yao, Z., Yasui, L. S., Ye, M., Yedvobnick, B., Yeganeh, B., Yeh, E. S., Yeyati, P. L., Yi, F., Yi, L., Yin, X., Yip, C. K., Yoo, Y., Yoo, Y. H., Yoon, S., Yoshida, K., Yoshimori, T., Young, K. H., Yu, H., Yu, J. J., Yu, J., Yu, J., Yu, L., Yu, W. H., Yu, X., Yu, Z., Yuan, J., Yuan, Z., Yue, B. Y., Yue, J., Yue, Z., Zacks, D. N., Zacksenhaus, E., Zaffaroni, N., Zaglia, T., Zakeri, Z., Zecchini, V., Zeng, J., Zeng, M., Zeng, Q., Zervos, A. S., Zhang, D. D., Zhang, F., Zhang, G., Zhang, G., Zhang, H., Zhang, H., Zhang, H., Zhang, H., Zhang, J., Zhang, J., Zhang, J., Zhang, J., Zhang, J., Zhang, L., Zhang, L., Zhang, L., Zhang, L., Zhang, M., Zhang, X., Zhang, X. D., Zhang, Y., Zhang, Y., Zhang, Y., Zhang, Y., Zhang, Y., Zhao, M., Zhao, W., Zhao, X., Zhao, Y. G., Zhao, Y., Zhao, Y., Zhao, Y., Zhao, Z., Zhao, Z. J., Zheng, D., Zheng, X., Zheng, X., Zhivotovsky, B., Zhong, Q., Zhou, G., Zhou, G., Zhou, H., Zhou, S., Zhou, X., Zhu, H., Zhu, H., Zhu, W., Zhu, W., Zhu, X., Zhu, Y., Zhuang, S., Zhuang, X., Ziparo, E., Zois, C. E., Zoladek, T., Zong, W., Zorzano, A., Zughaier, S. M. 2016; 12 (1): 1-222

    View details for DOI 10.1080/15548627.2015.1100356

    View details for PubMedID 26799652

  • High-throughput sequencing insights into T-cell receptor repertoire diversity in aging GENOME MEDICINE Goronzy, J. J., Qi, Q., Olshen, R. A., Weyand, C. M. 2015; 7: 117

    Abstract

    Decline in T-cell generation leading to T-cell receptor repertoire contraction is a cornerstone of immune system aging, and consequent disorders. High-throughput sequencing enables in-depth immune repertoire characterization, but blood samples are too small to capture its total diversity. New computational models could enable accurate estimation of this diversity.

    View details for PubMedID 26582264

  • Accelerated atherosclerosis in patients with chronic inflammatory rheumatologic conditions. International journal of clinical rheumatology Hong, J., Maron, D. J., Shirai, T., Weyand, C. M. 2015; 10 (5): 365-381

    Abstract

    Atherosclerosis is a complex inflammatory disease involving aberrant immune and tissue healing responses, which begins with endothelial dysfunction and ends with plaque development, instability and rupture. The increased risk for coronary artery disease in patients with rheumatologic diseases highlights how aberrancy in the innate and adaptive immune system may be central to development of both disease states and that atherosclerosis may be on a spectrum of immune-mediated conditions. Recognition of the tight association between chronic inflammatory disease and complications of atherosclerosis will impact the understanding of underlying pathogenic mechanisms and change diagnostic and therapeutic approaches in patients with rheumatologic syndromes as well as patients with coronary artery disease. In this review, we provide a summary of the role of the immune system in atherosclerosis, discuss the proposed mechanisms of accelerated atherosclerosis seen in association with rheumatologic diseases, evaluate the effect of immunosuppression on atherosclerosis and provide updates on available risk assessment tools, biomarkers and imaging modalities.

    View details for PubMedID 27042216

  • Failure in Nutrient Sensing Supports mTOR Hyperactivity and Proinflammatory Functions in T Cells from Patients with Rheumatoid Arthritis Yang, Z., Matteson, E. L., Hong, J., Goronzy, J., Weyand, C. M. WILEY-BLACKWELL. 2015
  • Enhanced Oxidant Signaling in Inflammatory Macrophages in Rheumatoid Arthritis (RA) and in Coronary Artery Disease (CAD) Shirai, T., Schaefgen, B., Wallis, B., Matteson, E. L., Assimes, T. L., Harrison, D. G., Goronzy, J., Weyand, C. M. WILEY-BLACKWELL. 2015
  • Age-Associated Failure To Adjust Type I IFN Receptor Signaling Thresholds after T Cell Activation JOURNAL OF IMMUNOLOGY Li, G., Ju, J., Weyand, C. M., Goronzy, J. J. 2015; 195 (3): 865-874

    Abstract

    With increasing age, naive CD4 T cells acquire intrinsic defects that compromise their ability to respond and differentiate. Type I IFNs, pervasive constituents of the environment in which adaptive immune responses occur, are known to regulate T cell differentiation and survival. Activated naive CD4 T cells from older individuals have reduced responses to type I IFN, a defect that develops during activation and that is not observed in quiescent naive CD4 T cells. Naive CD4 T cells from young adults upregulate the expression of STAT1 and STAT5 after activation, lowering their threshold to respond to type I IFN stimulation. The heightened STAT signaling is critical to maintain the expression of CD69 that regulates lymphocyte egress and the ability to produce IL-2 and to survive. Although activation of T cells from older adults also induces transcription of STAT1 and STAT5, failure to exclude SHP-1 from the signaling complex blunts their type I IFN response. In summary, our data show that type I IFN signaling thresholds in naive CD4 T cells after activation are dynamically regulated to respond to environmental cues for clonal expansion and memory cell differentiation. Naive CD4 T cells from older adults have a defect in this threshold calibration. Restoring their ability to respond to type I IFN emerges as a promising target to restore T cell responses and to improve the induction of T cell memory.

    View details for DOI 10.4049/jimmunol.1402389

    View details for Web of Science ID 000358070400016

    View details for PubMedCentralID PMC4506866

  • Age-Associated Failure To Adjust Type I IFN Receptor Signaling Thresholds after T Cell Activation. Journal of immunology (Baltimore, Md. : 1950) Li, G., Ju, J., Weyand, C. M., Goronzy, J. J. 2015; 195 (3): 865-74

    Abstract

    With increasing age, naive CD4 T cells acquire intrinsic defects that compromise their ability to respond and differentiate. Type I IFNs, pervasive constituents of the environment in which adaptive immune responses occur, are known to regulate T cell differentiation and survival. Activated naive CD4 T cells from older individuals have reduced responses to type I IFN, a defect that develops during activation and that is not observed in quiescent naive CD4 T cells. Naive CD4 T cells from young adults upregulate the expression of STAT1 and STAT5 after activation, lowering their threshold to respond to type I IFN stimulation. The heightened STAT signaling is critical to maintain the expression of CD69 that regulates lymphocyte egress and the ability to produce IL-2 and to survive. Although activation of T cells from older adults also induces transcription of STAT1 and STAT5, failure to exclude SHP-1 from the signaling complex blunts their type I IFN response. In summary, our data show that type I IFN signaling thresholds in naive CD4 T cells after activation are dynamically regulated to respond to environmental cues for clonal expansion and memory cell differentiation. Naive CD4 T cells from older adults have a defect in this threshold calibration. Restoring their ability to respond to type I IFN emerges as a promising target to restore T cell responses and to improve the induction of T cell memory.

    View details for DOI 10.4049/jimmunol.1402389

    View details for PubMedID 26091718

    View details for PubMedCentralID PMC4506866

  • Large-Scale and Comprehensive Immune Profiling and Functional Analysis of Normal Human Aging PLOS ONE Whiting, C. C., Siebert, J., Newman, A. M., Du, H., Alizadeh, A. A., Goronzy, J., Weyand, C. M., Krishnan, E., Fathman, C. G., Maecker, H. T. 2015; 10 (7)

    Abstract

    While many age-associated immune changes have been reported, a comprehensive set of metrics of immune aging is lacking. Here we report data from 243 healthy adults aged 40-97, for whom we measured clinical and functional parameters, serum cytokines, cytokines and gene expression in stimulated and unstimulated PBMC, PBMC phenotypes, and cytokine-stimulated pSTAT signaling in whole blood. Although highly heterogeneous across individuals, many of these assays revealed trends by age, sex, and CMV status, to greater or lesser degrees. Age, then sex and CMV status, showed the greatest impact on the immune system, as measured by the percentage of assay readouts with significant differences. An elastic net regression model could optimally predict age with 14 analytes from different assays. This reinforces the importance of multivariate analysis for defining a healthy immune system. These data provide a reference for others measuring immune parameters in older people.

    View details for DOI 10.1371/journal.pone.0133627

    View details for Web of Science ID 000358547600123

  • Autophagy in autoimmune disease JOURNAL OF MOLECULAR MEDICINE-JMM Yang, Z., Goronzy, J. J., Weyand, C. M. 2015; 93 (7): 707-717

    Abstract

    Autophagy is a protective and life-sustaining process in which cytoplasmic components are packaged into double-membrane vesicles and targeted to lysosomes for degradation. This process of cellular self-digestion is an essential stress response and is cytoprotective by removing damaged organelles and proteins that threaten the cell's survival. Key outcomes include energy generation and recycling of metabolic precursors. In the immune system, autophagy regulates processes such as antigen uptake and presentation, removal of pathogens, survival of short- and long-lived immune cells, and cytokine-dependent inflammation. In all cases, a window of optimal autophagic activity appears critical to balance catabolic, reparative, and inflammation-inducing processes. Dysregulation of autophagosome formation and autophagic flux can have deleterious consequences, ranging from a failure to "clean house" to the induction of autophagy-induced cell death. Abnormalities in the autophagic pathway have been implicated in numerous autoimmune diseases. Genome-wide association studies have linked polymorphisms in autophagy-related genes with predisposition for tissue-destructive inflammatory disease, specifically in inflammatory bowel disease and systemic lupus erythematosus. Although the precise mechanisms by which dysfunctional autophagy renders the host susceptible to continuous inflammation remain unclear, autophagy's role in regulating the long-term survival of adaptive immune cells has recently surfaced as a defect in multiple sclerosis and rheumatoid arthritis. Efforts are underway to identify autophagy-inducing and autophagy-suppressing pharmacologic interventions that can be added to immunosuppressive therapy to improve outcomes of patients with autoimmune disease.

    View details for DOI 10.1007/s00109-015-1297-8

    View details for Web of Science ID 000356530700002

    View details for PubMedID 26054920

    View details for PubMedCentralID PMC4486076

  • Naive T Cell Maintenance and Function in Human Aging JOURNAL OF IMMUNOLOGY Goronzy, J. J., Fang, F., Cavanagh, M. M., Qi, Q., Weyand, C. M. 2015; 194 (9): 4073-4080

    Abstract

    In studies of immune aging, naive T cells frequently take center stage. Describing the complexity of the human naive T cell repertoire remains a daunting task; however, emerging data suggest that homeostatic mechanisms are robust enough to maintain a large and diverse CD4 T cell repertoire with age. Compartment shrinkage and clonal expansions are challenges for naive CD8 T cells. In addition to population aspects, identification of potentially targetable cellular defects is receiving renewed interest. The last decade has seen remarkable progress in identifying genetic and biochemical pathways that are pertinent for aging in general and that are instructive to understand naive T cell dysfunction. One hallmark sets naive T cell aging apart from most other tissues except stem cells: they initiate but do not complete differentiation programs toward memory cells. Maintaining quiescence and avoiding differentiation may be the ultimate challenge to maintain the functions unique for naive T cells.

    View details for DOI 10.4049/jimmunol.1500046

    View details for Web of Science ID 000353727400005

    View details for PubMedID 25888703

    View details for PubMedCentralID PMC4452284

  • Macrophages in vascular inflammation - From atherosclerosis to vasculitis AUTOIMMUNITY Shirai, T., Hilhorst, M., Harrison, D. G., Goronzy, J. J., Weyand, C. M. 2015; 48 (3): 139-151

    Abstract

    The spectrum of vascular inflammatory disease ranges from atherosclerosis and hypertension, widespread conditions affecting large proportions of the population, to the vasculitides, rare syndromes leading to fast and irreversible organ failure. Atherosclerosis progresses over decades, inevitably proceeding through multiple phases of disease and causes its major complications when the vessel wall lesion ruptures, giving rise to lumen-occlusive atherothrombosis. Vasculitides of medium and large arteries progress rapidly, causing tissue ischemia through lumen-occlusive intimal hyperplasia. In both disease entities, macrophages play a decisive role in pathogenesis, but function in the context of other immune cells that direct their differentiation and their functional commitments. In atherosclerosis, macrophages are involved in the removal of lipids and tissue debris and make a critical contribution to tissue damage and wall remodeling. In several of the vasculitides, macrophages contribute to granuloma formation, a microstructural platform optimizing macrophage-T-cell interactions, antigen containment and inflammatory amplification. By virtue of their versatility and plasticity, macrophages are able to promote a series of pathogenic functions, ranging from the release of cytokines and enzymes, the production of reactive oxygen species, presentation of antigen and secretion of tissue remodeling factors. However, as short-lived cells that lack memory, macrophages are also amendable to reprogramming, making them promising targets for anti-inflammatory interventions.

    View details for DOI 10.3109/08916934.2015.1027815

    View details for Web of Science ID 000353523900002

    View details for PubMedID 25811915

  • Cardiorheumatology: cardiac involvement in systemic rheumatic disease NATURE REVIEWS CARDIOLOGY Prasad, M., Hermann, J., Gabriel, S. E., Weyand, C. M., Mulvagh, S., Mankad, R., Oh, J. K., Matteson, E. L., Lerman, A. 2015; 12 (3): 168-176

    Abstract

    Autoimmune rheumatic diseases can affect the cardiac vasculature, valves, myocardium, pericardium, and conduction system, leading to a plethora of cardiovascular manifestations that can remain clinically silent or lead to substantial cardiovascular morbidity and mortality. Although the high risk of cardiovascular pathology in patients with autoimmune inflammatory rheumatological diseases is not owing to atherosclerosis alone, this particular condition contributes substantially to cardiovascular morbidity and mortality-the degree of coronary atherosclerosis observed in patients with rheumatic diseases can be as accelerated, diffuse, and extensive as in patients with diabetes mellitus. The high risk of atherosclerosis is not solely attributable to traditional cardiovascular risk factors: dysfunctional immune responses, a hallmark of patients with rheumatic disorders, are thought to cause chronic tissue-destructive inflammation. Prompt recognition of cardiovascular abnormalities is needed for timely and appropriate management, and aggressive control of traditional risk factors remains imperative in patients with rheumatic diseases. Moreover, therapies directed towards inflammatory process are crucial to reduce cardiovascular disease morbidity and mortality. In this Review, we examine the multiple cardiovascular manifestations in patients with rheumatological disorders, their underlying pathophysiology, and available management strategies, with particular emphasis on the vascular aspects of the emerging field of 'cardiorheumatology'.

    View details for Web of Science ID 000350107600006

    View details for PubMedID 25533796

  • T-cell metabolism in autoimmune disease ARTHRITIS RESEARCH & THERAPY Yang, Z., Matteson, E. L., Goronzy, J. J., Weyand, C. M. 2015; 17

    Abstract

    Cancer cells have long been known to fuel their pathogenic growth habits by sustaining a high glycolytic flux, first described almost 90 years ago as the so-called Warburg effect. Immune cells utilize a similar strategy to generate the energy carriers and metabolic intermediates they need to produce biomass and inflammatory mediators. Resting lymphocytes generate energy through oxidative phosphorylation and breakdown of fatty acids, and upon activation rapidly switch to aerobic glycolysis and low tricarboxylic acid flux. T cells in patients with rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) have a disease-specific metabolic signature that may explain, at least in part, why they are dysfunctional. RA T cells are characterized by low adenosine triphosphate and lactate levels and increased availability of the cellular reductant NADPH. This anti-Warburg effect results from insufficient activity of the glycolytic enzyme phosphofructokinase and differentiates the metabolic status in RA T cells from those in cancer cells. Excess production of reactive oxygen species and a defect in lipid metabolism characterizes metabolic conditions in SLE T cells. Owing to increased production of the glycosphingolipids lactosylceramide, globotriaosylceramide and monosialotetrahexosylganglioside, SLE T cells change membrane raft formation and fail to phosphorylate pERK, yet hyperproliferate. Borrowing from cancer metabolomics, the metabolic modifications occurring in autoimmune disease are probably heterogeneous and context dependent. Variations of glucose, amino acid and lipid metabolism in different disease states may provide opportunities to develop biomarkers and exploit metabolic pathways as therapeutic targets.

    View details for DOI 10.1186/s13075-015-0542-4

    View details for Web of Science ID 000351577600002

    View details for PubMedCentralID PMC4324046

  • Serpin Treatment Suppresses Inflammatory Vascular Lesions in Temporal Artery Implants (TAI) from Patients with Giant Cell Arteritis PLOS ONE Chen, H., Zheng, D., Ambadapadi, S., Davids, J., Ryden, S., Samy, H., Bartee, M., Sobel, E., Dai, E., Liu, L., Macaulay, C., Yachnis, A., Weyand, C., Thoburn, R., Lucas, A. 2015; 10 (2)

    Abstract

    Giant cell arteritis (GCA) and Takayasu's disease are inflammatory vasculitic syndromes (IVS) causing sudden blindness and widespread arterial obstruction and aneurysm formation. Glucocorticoids and aspirin are mainstays of treatment, predominantly targeting T cells. Serp-1, a Myxomavirus-derived serpin, blocks macrophage and T cells in a wide range of animal models. Serp-1 also reduced markers of myocardial injury in a Phase IIa clinical trial for unstable coronary disease. In recent work, we detected improved survival and decreased arterial inflammation in a mouse Herpesvirus model of IVS. Here we examine Serp-1 treatment of human temporal artery (TA) biopsies from patients with suspected TA GCA arteritis after implant (TAI) into the aorta of immunodeficient SCID (severe combined immunodeficiency) mice. TAI positive for arteritis (GCApos) had significantly increased inflammation and plaque when compared to negative TAI (GCAneg). Serp-1 significantly reduced intimal inflammation and CD11b+ cell infiltrates in TAI, with reduced splenocyte Th1, Th17, and Treg. Splenocytes from mice with GCApos grafts had increased gene expression for interleukin-1 beta (IL-1β), IL-17, and CD25 and decreased Factor II. Serp-1 decreased IL-1β expression. In conclusion, GCApos TAI xenografts in mice provide a viable disease model and have increased intimal inflammation as expected and Serp-1 significantly reduces vascular inflammatory lesions with reduced IL-1β.

    View details for DOI 10.1371/journal.pone.0115482

    View details for Web of Science ID 000349444900018

    View details for PubMedID 25658487

    View details for PubMedCentralID PMC4319900

  • Visual Manifestations in Giant Cell Arteritis: Trend over 5 Decades in a Population-based Cohort. journal of rheumatology Singh, A. G., Kermani, T. A., Crowson, C. S., Weyand, C. M., Matteson, E. L., Warrington, K. J. 2015; 42 (2): 309-315

    Abstract

    To evaluate clinical characteristics, treatment, and outcomes of patients with visual changes from giant cell arteritis (GCA) and to examine trends over the last 5 decades.We reviewed the medical records of a population-based cohort of patients with GCA diagnosed between 1950 and 2004. The clinical, ophthalmological, and laboratory features of patients with visual manifestations attributable to GCA were compared to patients without visual complications. Trends over time were examined using logistic regression modeling adjusted for age and sex.In a cohort of 204 cases of GCA (mean age 76.0 ± 8.2 yrs, 80% female), visual changes from GCA were observed in 47 patients (23%), and 4.4% suffered complete vision loss. A higher proportion of patients with visual manifestations reported jaw claudication than did patients without visual changes (55% vs 38%, p = 0.04). Over a period of 55 years, we observed a significant decline in the incidence of visual symptoms due to GCA. There was a lower incidence of ischemic optic neuropathy in the 1980-2004 cohort vs 1950-1979 (6% vs 15%, p = 0.03). Patients diagnosed in later decades were more likely to recover from visual symptoms (HR 1.34, 95% CI 1.06-1.71). Chances of recovery were poor in patients with anterior ischemic optic neuropathy or complete vision loss.Incidence of visual symptoms has declined over the past 5 decades, and chances of recovery from visual symptoms have improved. However, complete loss of vision is essentially irreversible. Jaw claudication is associated with higher likelihood of development of visual symptoms.

    View details for DOI 10.3899/jrheum.140188

    View details for PubMedID 25512481

  • Large-Scale and Comprehensive Immune Profiling and Functional Analysis of Normal Human Aging. PloS one Whiting, C. C., Siebert, J., Newman, A. M., Du, H., Alizadeh, A. A., Goronzy, J., Weyand, C. M., Krishnan, E., Fathman, C. G., Maecker, H. T. 2015; 10 (7)

    Abstract

    While many age-associated immune changes have been reported, a comprehensive set of metrics of immune aging is lacking. Here we report data from 243 healthy adults aged 40-97, for whom we measured clinical and functional parameters, serum cytokines, cytokines and gene expression in stimulated and unstimulated PBMC, PBMC phenotypes, and cytokine-stimulated pSTAT signaling in whole blood. Although highly heterogeneous across individuals, many of these assays revealed trends by age, sex, and CMV status, to greater or lesser degrees. Age, then sex and CMV status, showed the greatest impact on the immune system, as measured by the percentage of assay readouts with significant differences. An elastic net regression model could optimally predict age with 14 analytes from different assays. This reinforces the importance of multivariate analysis for defining a healthy immune system. These data provide a reference for others measuring immune parameters in older people.

    View details for DOI 10.1371/journal.pone.0133627

    View details for PubMedID 26197454

  • T-cell metabolism in autoimmune disease. Arthritis research & therapy Yang, Z., Matteson, E. L., Goronzy, J. J., Weyand, C. M. 2015; 17: 29-?

    Abstract

    Cancer cells have long been known to fuel their pathogenic growth habits by sustaining a high glycolytic flux, first described almost 90 years ago as the so-called Warburg effect. Immune cells utilize a similar strategy to generate the energy carriers and metabolic intermediates they need to produce biomass and inflammatory mediators. Resting lymphocytes generate energy through oxidative phosphorylation and breakdown of fatty acids, and upon activation rapidly switch to aerobic glycolysis and low tricarboxylic acid flux. T cells in patients with rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) have a disease-specific metabolic signature that may explain, at least in part, why they are dysfunctional. RA T cells are characterized by low adenosine triphosphate and lactate levels and increased availability of the cellular reductant NADPH. This anti-Warburg effect results from insufficient activity of the glycolytic enzyme phosphofructokinase and differentiates the metabolic status in RA T cells from those in cancer cells. Excess production of reactive oxygen species and a defect in lipid metabolism characterizes metabolic conditions in SLE T cells. Owing to increased production of the glycosphingolipids lactosylceramide, globotriaosylceramide and monosialotetrahexosylganglioside, SLE T cells change membrane raft formation and fail to phosphorylate pERK, yet hyperproliferate. Borrowing from cancer metabolomics, the metabolic modifications occurring in autoimmune disease are probably heterogeneous and context dependent. Variations of glucose, amino acid and lipid metabolism in different disease states may provide opportunities to develop biomarkers and exploit metabolic pathways as therapeutic targets.

    View details for DOI 10.1186/s13075-015-0542-4

    View details for PubMedID 25890351

  • Giant-cell arteritis and polymyalgia rheumatica. New England journal of medicine Weyand, C. M., Goronzy, J. J. 2014; 371 (17): 1653-?

    View details for DOI 10.1056/NEJMc1409206

    View details for PubMedID 25337759

  • Distinct Profiles of Proinflammatory Macrophages in Rheumatoid Arthritis and Coronary Artery Disease. Shirai, T., Matteson, E. L., Harrison, D. G., Wallis, B. B., Assimes, T. L., Goronzy, J. J., Weyand, C. M. WILEY-BLACKWELL. 2014: S187
  • T cell-macrophage interactions and granuloma formation in vasculitis FRONTIERS IN IMMUNOLOGY Hilhorst, M., Shirai, T., Berry, G., Goronzy, J. J., Weyand, C. M. 2014; 5: 1-14
  • Diversity and clonal selection in the human T-cell repertoire PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Qi, Q., Liu, Y., Cheng, Y., Glanville, J., Zhang, D., Lee, J., Olshen, R. A., Weyand, C. M., Boyd, S. D., Goronzy, J. J. 2014; 111 (36): 13139-13144

    Abstract

    T-cell receptor (TCR) diversity, a prerequisite for immune system recognition of the universe of foreign antigens, is generated in the first two decades of life in the thymus and then persists to an unknown extent through life via homeostatic proliferation of naïve T cells. We have used next-generation sequencing and nonparametric statistical analysis to estimate a lower bound for the total number of different TCR beta (TCRB) sequences in human repertoires. We arrived at surprisingly high minimal estimates of 100 million unique TCRB sequences in naïve CD4 and CD8 T-cell repertoires of young adults. Naïve repertoire richness modestly declined two- to fivefold in healthy elderly. Repertoire richness contraction with age was even less pronounced for memory CD4 and CD8 T cells. In contrast, age had a major impact on the inequality of clonal sizes, as estimated by a modified Gini-Simpson index clonality score. In particular, large naïve T-cell clones that were distinct from memory clones were found in the repertoires of elderly individuals, indicating uneven homeostatic proliferation without development of a memory cell phenotype. Our results suggest that a highly diverse repertoire is maintained despite thymic involution; however, peripheral fitness selection of T cells leads to repertoire perturbations that can influence the immune response in the elderly.

    View details for DOI 10.1073/pnas.1409155111

    View details for Web of Science ID 000341625600045

    View details for PubMedCentralID PMC4246948

  • Diversity and clonal selection in the human T-cell repertoire. Proceedings of the National Academy of Sciences of the United States of America Qi, Q., Liu, Y., Cheng, Y., Glanville, J., Zhang, D., Lee, J., Olshen, R. A., Weyand, C. M., Boyd, S. D., Goronzy, J. J. 2014; 111 (36): 13139-13144

    Abstract

    T-cell receptor (TCR) diversity, a prerequisite for immune system recognition of the universe of foreign antigens, is generated in the first two decades of life in the thymus and then persists to an unknown extent through life via homeostatic proliferation of naïve T cells. We have used next-generation sequencing and nonparametric statistical analysis to estimate a lower bound for the total number of different TCR beta (TCRB) sequences in human repertoires. We arrived at surprisingly high minimal estimates of 100 million unique TCRB sequences in naïve CD4 and CD8 T-cell repertoires of young adults. Naïve repertoire richness modestly declined two- to fivefold in healthy elderly. Repertoire richness contraction with age was even less pronounced for memory CD4 and CD8 T cells. In contrast, age had a major impact on the inequality of clonal sizes, as estimated by a modified Gini-Simpson index clonality score. In particular, large naïve T-cell clones that were distinct from memory clones were found in the repertoires of elderly individuals, indicating uneven homeostatic proliferation without development of a memory cell phenotype. Our results suggest that a highly diverse repertoire is maintained despite thymic involution; however, peripheral fitness selection of T cells leads to repertoire perturbations that can influence the immune response in the elderly.

    View details for DOI 10.1073/pnas.1409155111

    View details for PubMedID 25157137

  • Clinical practice. Giant-cell arteritis and polymyalgia rheumatica. New England journal of medicine Weyand, C. M., Goronzy, J. J. 2014; 371 (1): 50-57

    View details for DOI 10.1056/NEJMcp1214825

    View details for PubMedID 24988557

  • Giant-Cell Arteritis and Polymyalgia Rheumatica NEW ENGLAND JOURNAL OF MEDICINE Weyand, C. M., Goronzy, J. J. 2014; 371 (1): 50-57
  • Mechanisms shaping the naïve T cell repertoire in the elderly - Thymic involution or peripheral homeostatic proliferation? Experimental gerontology Qi, Q., Zhang, D. W., Weyand, C. M., Goronzy, J. J. 2014; 54: 71-74

    Abstract

    The ability of the human immune system to repel infections is drastically diminished with age. Elderly individuals are more susceptible to new threats and are less able to control endogenous infections. The thymus, which is the sole source of new T cells, has been proposed as a target for regenerative efforts to improve immune competence, as thymic activity is dramatically reduced after puberty. In this review, we review the role of the thymus in the maintenance of T cell homeostasis throughout life and contrast the differences in mice and humans. We propose that in humans, lack of thymic T cell generation does not explain a decline in T cell receptor diversity nor would thymic rejuvenation restore diversity. Initial studies using next generation sequencing are beginning to establish lower boundaries of T cell receptor diversity. With increasing sequencing depth and the development of new statistical models, we are now in the position to test this model and to assess the impact of age on T cell diversity and clonality.

    View details for DOI 10.1016/j.exger.2014.01.005

    View details for PubMedID 24440389

  • Targets of Immune Regeneration in Rheumatoid Arthritis MAYO CLINIC PROCEEDINGS Hohensinner, P. J., Goronzy, J. J., Weyand, C. M. 2014; 89 (4): 563-575

    Abstract

    Many of the aging-related morbidities, including cancer, cardiovascular disease, neurodegenerative disease, and infectious susceptibility, are linked to a decline in immune competence with a concomitant rise in proinflammatory immunity, placing the process of immune aging at the center of aging biology. Immune aging affects individuals older than 50 years and is accelerated in patients with the autoimmune disease rheumatoid arthritis. Immune aging results in a marked decline in protective immune responses and a parallel increase in tissue inflammatory responses. By studying immune cells in patients with rheumatoid arthritis, several of the molecular underpinnings of the immune aging process have been delineated, such as the loss of telomeres and inefficiencies in the repair of damaged DNA. Aging T cells display a series of abnormalities, including the unopposed up-regulation of cytoplasmic phosphatases and the loss of glycolytic competence, that alter their response to stimulating signals and undermine their longevity. Understanding the connection between accelerated immune aging and autoimmunity remains an area of active research. With increasing knowledge of the molecular pathways that cause immunosenescence, therapeutic interventions can be designed to slow or halt the seemingly inevitable deterioration of protective immunity with aging.

    View details for DOI 10.1016/j.mayocp.2014.01.020

    View details for Web of Science ID 000333528700021

    View details for PubMedID 24684878

  • The glycolytic enzyme PFKFB3/phosphofructokinase regulates autophagy. Autophagy Yang, Z., Goronzy, J. J., Weyand, C. M. 2014; 10 (2): 382-383

    Abstract

    T lymphocytes, the master regulators of immunity, have an unusual lifestyle. Equipped with a clonally distributed receptor they remain resting for long periods of time but go into overdrive when encountering antigen. Antigen recognition triggers an activation program that results in massive proliferation, differentiation into effector/memory cells, egress from lymphoid storage sites, and production of an array of cytokines. To adapt to the sudden demand for energy and biosynthetic macromolecules, T cells resort to aerobic glycolysis, relying on the Warburg effect to provide sufficient ATP and precursor molecules. Metabolic adaptation to the biosynthetic needs includes upregulation of autophagy, a catabolic process resulting in the degradation of cytoplasmic contents. The close connection between a metabolic switch, proliferative expansion, and functional differentiation connects the metabolic conditions in the cell to normal and pathogenic immunity.

    View details for DOI 10.4161/auto.27345

    View details for PubMedID 24351650

  • T cell-macrophage interactions and granuloma formation in vasculitis. Frontiers in immunology Hilhorst, M., Shirai, T., Berry, G., Goronzy, J. J., Weyand, C. M. 2014; 5: 432-?

    Abstract

    Granuloma formation, bringing into close proximity highly activated macrophages and T cells, is a typical event in inflammatory blood vessel diseases, and is noted in the name of several of the vasculitides. It is not known whether specific properties of the microenvironment in the blood vessel wall or the immediate surroundings of blood vessels contribute to granuloma formation and, in some cases, generation of multinucleated giant cells. Granulomas provide a specialized niche to optimize macrophage-T cell interactions, strongly activating both cell types. This is mirrored by the intensity of the systemic inflammation encountered in patients with vasculitis, often presenting with malaise, weight loss, fever, and strongly upregulated acute phase responses. As a sophisticated and highly organized structure, granulomas can serve as an ideal site to induce differentiation and maturation of T cells. The granulomas possibly seed aberrant Th1 and Th17 cells into the circulation, which are known to be the main pathogenic cells in vasculitis. Through the induction of memory T cells, aberrant innate immune responses can imprint the host immune system for decades to come and promote chronicity of the disease process. Improved understanding of T cell-macrophage interactions will redefine pathogenic models in the vasculitides and provide new avenues for immunomodulatory therapy.

    View details for DOI 10.3389/fimmu.2014.00432

    View details for PubMedID 25309534

    View details for PubMedCentralID PMC4162471

  • T-cell aging in rheumatoid arthritis CURRENT OPINION IN RHEUMATOLOGY Weyand, C. M., Yang, Z., Goronzy, J. J. 2014; 26 (1): 93-100

    Abstract

    With progressive age, the immune system and the propensity for abnormal immunity change fundamentally. Individuals greater than 50 years of age are not only more susceptible to infection and cancer, but also at higher risk for chronic inflammation and immune-mediated tissue damage. The process of immunosenescence is accelerated in rheumatoid arthritis (RA).Premature T-cell senescence occurs not only in RA, but also has been involved in morbidity and mortality of chronic HIV infection. Senescent cells acquire the 'senescence-associated secretory phenotype', which promotes and sustains tissue inflammation. Molecular mechanisms underlying T-cell aging are beginning to be understood. In addition to the contraction of T-cell diversity because of uneven clonal expansion, senescent T cells have defects in balancing cytoplasmic kinase and phosphatase activities, changing their activation thresholds. Also, leakiness in repairing DNA lesions and uncapped telomeres imposes genomic stress. Age-induced changes in the tissue microenvironment may alter the T-cell responses.Gain-of-function and loss-of-function in senescent T cells undermine protective immunity and create the conditions for chronic tissue inflammation, a combination typically encountered in RA. Genetic programs involved in T-cell signaling and DNA repair are of high interest in the search for underlying molecular defects.

    View details for DOI 10.1097/BOR.0000000000000011

    View details for Web of Science ID 000328818500014

    View details for PubMedID 24296720

  • Regulatory T Cells and the Immune Aging Process: A Mini-Review GERONTOLOGY Jagger, A., Shimojima, Y., Goronzy, J. J., Weyand, C. M. 2014; 60 (2): 130-137

    Abstract

    Constant exposure to new and persisting antigens and the need to replace cellular attrition with newly built cells lead to profound remodeling of the immune system after the age of 50 years. The impact of the immunosenescence process varies amongst the different cellular subsets represented within the immune system. Emerging data suggest that progressive aging significantly affects frequencies, subset distribution and functional competence of regulatory T cells (Tregs). Given the central role of Tregs in immune homeostasis, age-related loss of Treg function would be predicted to cause excessive immunity, encountered in elderly humans as a syndrome of chronic, smoldering inflammation as well as the age-related increase in the risk for autoimmunity. Conversely, age-dependent gain of Treg activity would result in failing immunity, such as the rising risk of malignancies and infections amongst the elderly. Emerging data suggest that some Treg populations, specifically naturally occurring Tregs, seem to accumulate with advancing age, whereas inducible Tregs appear to be less available in the older host. More studies are necessary to elucidate functional competence of old Tregs, with an emphasis on comparing the efficacy of young and old Tregs for defined functional domains. Mechanisms of declining Treg inducibility are not understood, but may provide an opportunity for targeted immunomodulation in the elderly. On the horizon is the potential to develop novel therapeutic interventions that target Tregs to make the elderly more efficient in fighting cancers and infections and dampen the risk for senescence-associated inflammation.

    View details for DOI 10.1159/000355303

    View details for Web of Science ID 000331773700006

    View details for PubMedID 24296590

  • Immune mechanisms in medium and large-vessel vasculitis NATURE REVIEWS RHEUMATOLOGY Weyand, C. M., Goronzy, J. J. 2013; 9 (12): 731-740

    Abstract

    Vasculitis of the medium and large arteries, most often presenting as giant cell arteritis (GCA), is an infrequent, but potentially fatal, type of immune-mediated vascular disease. The site of the aberrant immune reaction, the mural layers of the artery, is strictly defined by vascular dendritic cells, endothelial cells, vascular smooth muscle cells and fibroblasts, which engage in an interaction with T cells and macrophages to, ultimately, cause luminal stenosis or aneurysmal wall damage of the vessel. A multitude of effector cytokines, all known as critical mediators in host-protective immunity, have been identified in vasculitic lesions. Two dominant cytokine clusters--the IL-6-IL-17 axis and the IL-12-IFN-γ axis--have been linked to disease activity. These two clusters seem to serve different roles in the vasculitic process. The IL-6-IL-17 cluster is highly responsive to standard corticosteroid therapy, whereas the IL-12-IFN-γ cluster is resistant to steroid-mediated immunosuppression. The information exchange between vascular and immune cells and stabilization of the vasculitic process involves members of the Notch receptor and ligand family. Focusing on elements in the tissue context of GCA, instead of broadly suppressing host immunity, might enable a more tailored therapeutic approach that avoids unwanted adverse effects of aggressive immunosuppression.

    View details for DOI 10.1038/nrrheum.2013.161

    View details for Web of Science ID 000328126000006

    View details for PubMedID 24189842

    View details for PubMedCentralID PMC4277683

  • JAK/STAT-Signaling in Giant Cell Arteritis Hartmann, B., Mohan, S. V., Goronzy, J. J., Weyand, C. M. LIPPINCOTT WILLIAMS & WILKINS. 2013
  • Phosphofructokinase deficiency impairs ATP generation, autophagy, and redox balance in rheumatoid arthritis T cells. journal of experimental medicine Yang, Z., Fujii, H., Mohan, S. V., Goronzy, J. J., Weyand, C. M. 2013; 210 (10): 2119-2134

    Abstract

    In the HLA class II-associated autoimmune syndrome rheumatoid arthritis (RA), CD4 T cells are critical drivers of pathogenic immunity. We have explored the metabolic activity of RA T cells and its impact on cellular function and fate. Naive CD4 T cells from RA patients failed to metabolize equal amounts of glucose as age-matched control cells, generated less intracellular ATP, and were apoptosis-susceptible. The defect was attributed to insufficient induction of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), a regulatory and rate-limiting glycolytic enzyme known to cause the Warburg effect. Forced overexpression of PFKFB3 in RA T cells restored glycolytic flux and protected cells from excessive apoptosis. Hypoglycolytic RA T cells diverted glucose toward the pentose phosphate pathway, generated more NADPH, and consumed intracellular reactive oxygen species (ROS). PFKFB3 deficiency also constrained the ability of RA T cells to resort to autophagy as an alternative means to provide energy and biosynthetic precursor molecules. PFKFB3 silencing and overexpression identified a novel extraglycolytic role of the enzyme in autophagy regulation. In essence, T cells in RA patients, even those in a naive state, are metabolically reprogrammed with insufficient up-regulation of the glycolytic activator PFKFB3, rendering them energy-deprived, ROS- and autophagy-deficient, apoptosis-sensitive, and prone to undergo senescence.

    View details for DOI 10.1084/jem.20130252

    View details for PubMedID 24043759

    View details for PubMedCentralID PMC3782046

  • Understanding immunosenescence to improve responses to vaccines NATURE IMMUNOLOGY Goronzy, J. J., Weyand, C. M. 2013; 14 (5): 428-436

    Abstract

    In the older adult, the benefits of vaccination to prevent infectious disease are limited, mainly because of the adaptive immune system's inability to generate protective immunity. The age-dependent decrease in immunological competence, often referred to as 'immunosenescence', results from the progressive deterioration of innate and adaptive immune responses. Most insights into mechanisms of immunological aging have been derived from studies of mouse models. In this Review, we explore how well such models are applicable to understanding the aging process throughout the 80-100 years of human life and discuss recent advances in identifying and characterizing the mechanisms that underlie age-associated defective adaptive immunity in humans.

    View details for DOI 10.1038/ni.2588

    View details for Web of Science ID 000317732100007

    View details for PubMedID 23598398

  • IL-7-and IL-15-Mediated TCR Sensitization Enables T Cell Responses to Self-Antigens JOURNAL OF IMMUNOLOGY Deshpande, P., Cavanagh, M. M., Le Saux, S., Singh, K., Weyand, C. M., Goronzy, J. J. 2013; 190 (4): 1416-1423

    Abstract

    Regulation of the ERK pathway is intimately involved in determining whether TCR stimulation is productive or induces anergy. T cells from patients with rheumatoid arthritis (RA) have increased ERK responsiveness, which may be relevant for disease pathogenesis. Inflammatory cytokines such as TNF-α did not reproduce the TCR hypersensitivity typical for RA in T cells from healthy individuals. In contrast, priming with the homeostatic cytokines (HCs) IL-7 and IL-15 amplified ERK phosphorylation to TCR stimulation 2- to 3-fold. The underlying mechanism involved a priming of the SOS-dependent amplification loop of RAS activation. The sensitization of the TCR signaling pathway has downstream consequences, such as increased proliferation and preferential Th1 differentiation. Importantly, priming with IL-7 or IL-15 enabled T cell responses to autoantigens associated with RA. Production of HCs is induced in lymphopenic conditions, which have been shown to predispose for autoimmunity and which appear to be present in the preclinical stages of RA. We propose that HCs, possibly induced by lymphopenia, decrease the signaling threshold for TCR activation and are thereby partly responsible for autoimmunity in RA.

    View details for DOI 10.4049/jimmunol.1201620

    View details for Web of Science ID 000314825400005

    View details for PubMedID 23325887

    View details for PubMedCentralID PMC3574821

  • The janus head of T cell aging - autoimmunity and immunodeficiency. Frontiers in immunology Goronzy, J. J., Li, G., Yang, Z., Weyand, C. M. 2013; 4: 131-?

    Abstract

    Immune aging is best known for its immune defects that increase susceptibility to infections and reduce adaptive immune responses to vaccination. In parallel, the aged immune system is prone to autoimmune responses and many autoimmune diseases increase in incidence with age or are even preferentially encountered in the elderly. Why an immune system that suboptimally responds to exogenous antigen fails to maintain tolerance to self-antigens appears to be perplexing. In this review, we will discuss age-associated deviations in the immune repertoire and the regulation of signaling pathways that may shed light on this conundrum.

    View details for DOI 10.3389/fimmu.2013.00131

    View details for PubMedID 23761790

    View details for PubMedCentralID PMC3671290

  • The Janus head of T cell aging - autoimmunity and immunodeficiency FRONTIERS IN IMMUNOLOGY Goronzy, J. J., Li, G., Yang, Z., Weyand, C. M. 2013; 4

    Abstract

    Immune aging is best known for its immune defects that increase susceptibility to infections and reduce adaptive immune responses to vaccination. In parallel, the aged immune system is prone to autoimmune responses and many autoimmune diseases increase in incidence with age or are even preferentially encountered in the elderly. Why an immune system that suboptimally responds to exogenous antigen fails to maintain tolerance to self-antigens appears to be perplexing. In this review, we will discuss age-associated deviations in the immune repertoire and the regulation of signaling pathways that may shed light on this conundrum.

    View details for DOI 10.3389/fimmu.2013.00131

    View details for Web of Science ID 000209374100130

    View details for PubMedCentralID PMC3671290

  • Decline in miR-181a expression with age impairs T cell receptor sensitivity by increasing DUSP6 activity NATURE MEDICINE Li, G., Yu, M., Lee, W., Tsang, M., Krishnan, E., Weyand, C. M., Goronzy, J. J. 2012; 18 (10): 1518-U113

    Abstract

    The ability of the human immune system to respond to vaccination declines with age. We identified an age-associated defect in T cell receptor (TCR)-induced extracellular signal-regulated kinase (ERK) phosphorylation in naive CD4(+) T cells, whereas other signals, such as ζ chain-associated protein kinase 70 (ZAP70) and phospholipase C-γ1 phosphorylation, were not impaired. The defective ERK signaling was caused by the dual specific phosphatase 6 (DUSP6), whose protein expression increased with age due to a decline in repression by miR-181a. Reconstitution of miR-181a lowered DUSP6 expression in naive CD4(+) T cells in elderly individuals. DUSP6 repression using miR-181a or specific siRNA and DUSP6 inhibition by the allosteric inhibitor (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one improved CD4(+) T cell responses, as seen by increased expression of activation markers, improved proliferation and supported preferential T helper type 1 cell differentiation. DUSP6 is a potential intervention target for restoring T cell responses in the elderly, which may augment the effectiveness of vaccination.

    View details for DOI 10.1038/nm.2963

    View details for PubMedID 23023500

  • Visual Manifestations in Giant Cell Arteritis: Trend Over Five Decades Singh, A. G., Crowson, C. S., Kermani, T. A., Weyand, C. M., Matteson, E. L., Warrington, K. J. WILEY-BLACKWELL. 2012: S993
  • Metabolic Reprogramming of Autoimmune T Cells in Rheumatoid Arthritis Annual Scientific Meeting of the American-College-of-Rheumatology (ACR) and Association-of-Rheumatology-Health-Professionals (ARHP) Yang, Z., Fujii, H., Mohan, S., Goronzy, J. J., Weyand, C. M. WILEY-BLACKWELL. 2012: S1130–S1131
  • Signaling pathways in aged T cells - A reflection of T cell differentiation, cell senescence and host environment SEMINARS IN IMMUNOLOGY Goronzy, J. J., Li, G., Yu, M., Weyand, C. M. 2012; 24 (5): 365-372

    Abstract

    With increasing age, the ability of the immune system to protect against new antigenic challenges or to control chronic infections erodes. Decline in thymic function and cumulating antigenic experiences of acute and chronic infections threaten T cell homeostasis, but insufficiently explain the failing immune competence and the increased susceptibility for autoimmunity. Alterations in signaling pathways in the aging T cells account for many of the age-related defects. Signaling threshold calibrations seen with aging frequently built on mechanisms that are operational in T cell development and T cell differentiation or are adaptations to the changing environment in the aging host. Age-related changes in transcription of receptors and signaling molecules shift the balance towards inhibitory pathways, most dominantly seen in CD8 T cells and to a lesser degree in CD4 T cells. Prominent examples are the expression of negative regulatory receptors of the CD28 and the TNF receptor superfamilies as well the expression of various cytoplasmic and nuclear dual-specific phosphatases.

    View details for DOI 10.1016/j.smim.2012.04.003

    View details for Web of Science ID 000311017700009

    View details for PubMedID 22560928

    View details for PubMedCentralID PMC3435478

  • The Immunopathology of Giant Cell Arteritis: Diagnostic and Therapeutic Implications JOURNAL OF NEURO-OPHTHALMOLOGY Weyand, C. M., Liao, Y. J., Goronzy, J. J. 2012; 32 (3): 259-265

    Abstract

    Giant cell arteritis (GCA) is an important cause of preventable blindness, most commonly due to anterior ischemic optic neuropathy. Ischemic tissue injury is the end result of a process that begins within the walls of susceptible arteries in which local dendritic cells (DCs) recruit and activate CD4 T cells that, in turn, direct the activity of effector macrophages. In response to the immune attack, the blood vessel forms lumen-stenosing intima. Multiple cascades of excessive T-cell reactivity contribute to the autoimmune features of giant cell arteritis with TH1 and TH17 immunity responsible for the early phase and TH1 immunity promoting chronic-smoldering inflammation. These cascades are only partially overlapping, supporting the concept that a multitude of instigators induce and sustain vascular inflammation. The artery actively participates in the abnormal immune response through endogenous immune sentinels, so-called vascular DCs embedded in the adventitia. Advancing age, the strongest of all risk factors for GCA, contributes to both, the dysfunction of the immune system and the vascular system. Expansion of the therapeutic armamentarium for GCA needs to focus on approaches that mitigate the impact of the aging artery and adapt to the needs of the immunosenescent host.

    View details for DOI 10.1097/WNO.0b013e318268aa9b

    View details for Web of Science ID 000308187800018

    View details for PubMedID 22914691

  • CD8(+) CD45RA(+) CCR7(+) FOXP3(+) T Cells with Immunosuppressive Properties: A Novel Subset of Inducible Human Regulatory T Cells JOURNAL OF IMMUNOLOGY Suzuki, M., Jagger, A. L., Konya, C., Shimojima, Y., Pryshchep, S., Goronzy, J. J., Weyand, C. M. 2012; 189 (5): 2118-2130

    Abstract

    CD8 T cells stimulated with a suboptimal dose of anti-CD3 Abs (100 pg/ml) in the presence of IL-15 retain a naive phenotype with expression of CD45RA, CD28, CD27, and CCR7 but acquire new functions and differentiate into immunosuppressive T cells. CD8+CCR7+ regulatory T cells (Tregs) express FOXP3 and prevent CD4 T cells from responding to TCR stimulation and entering the cell cycle. Naive CD4 T cells are more susceptible to inhibition than memory cells. The suppressive activity of CD8+CCR7+ Tregs is not mediated by IL-10, TGF-β, CTLA-4, CCL4, or adenosine and relies on interference with very early steps of the TCR signaling cascade. Specifically, CD8+CCR7+ Tregs prevent TCR-induced phosphorylation of ZAP70 and dampen the rise of intracellular calcium in CD4 T cells. The inducibility of CD8+CCR7+ Tregs is correlated with the age of the individual with PBLs of donors older than 60 y yielding low numbers of FOXP3(low) CD8 Tregs. Loss of CD8+CCR7+ Tregs in the elderly host may be of relevance in the aging immune system as immunosenescence is associated with a state of chronic smoldering inflammation.

    View details for DOI 10.4049/jimmunol.1200122

    View details for Web of Science ID 000308083600009

    View details for PubMedID 22821963

    View details for PubMedCentralID PMC3424334

  • Chronic inflammation and aging: DNA damage tips the balance CURRENT OPINION IN IMMUNOLOGY Cavanagh, M. M., Weyand, C. M., Goronzy, J. J. 2012; 24 (4): 488-493

    Abstract

    The aged immune system, typically hyporesponsive to infection and vaccination, can be hyperresponsive in the context of inflammatory pathology. Here we review current work examining the mechanisms behind the amplified inflammatory profile of aged adaptive immunity, and the reciprocal relationship between chronic inflammation and immune aging. Aged hematopoietic stem cells are driven to differentiate following accumulated DNA damage, thus depleting the stem cell pool and increasing the number of damaged effector cells in the circulation. Chronic DNA damage responses in lymphocytes as well as senescent cells of other lineages initiate the production of inflammatory mediators. In addition, aged lymphocytes become less reliant on specific antigen for stimulation and more prone to activation through innate receptors. When these lymphocytes are exposed to inflammatory signals produced by senescent tissues, the bias toward inflammation exacerbates destruction without necessarily improving immunity.

    View details for DOI 10.1016/j.coi.2012.04.003

    View details for Web of Science ID 000308898700020

    View details for PubMedID 22565047

    View details for PubMedCentralID PMC3423478

  • K-RAS GTPase- and B-RAF kinase-mediated T-cell tolerance defects in rheumatoid arthritis PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Singh, K., Deshpande, P., Li, G., Yu, M., Pryshchep, S., Cavanagh, M., Weyand, C. M., Goronzy, J. J. 2012; 109 (25): E1629-E1637

    Abstract

    Autoantibodies to common autoantigens and neoantigens, such as IgG Fc and citrullinated peptides, are immunological hallmarks of rheumatoid arthritis (RA). We examined whether a failure in maintaining tolerance is mediated by defects in T-cell receptor activation threshold settings. RA T cells responded to stimulation with significantly higher ERK phosphorylation (P < 0.001). Gene expression arrays of ERK pathway members suggested a higher expression of KRAS and BRAF, which was confirmed by quantitative PCR (P = 0.003), Western blot, and flow cytometry (P < 0.01). Partial silencing of KRAS and BRAF lowered activation-induced phosphorylated ERK levels (P < 0.01). In individual cells, levels of these signaling molecules correlated with ERK phosphorylation, attesting that their concentrations are functionally important. In confocal studies, B-RAF/K-RAS clustering was increased in RA T cells 2 min after T-cell receptor stimulation (P < 0.001). Overexpression of B-RAF and K-RAS in normal CD4 T cells amplified polyclonal T-cell proliferation and facilitated responses to citrullinated peptides. We propose that increased expression of B-RAF and K-RAS lowers T-cell activation thresholds in RA T cells, enabling responses to autoantigens.

    View details for DOI 10.1073/pnas.1117640109

    View details for Web of Science ID 000306061400009

    View details for PubMedID 22615393

    View details for PubMedCentralID PMC3382540

  • Immune aging and autoimmunity CELLULAR AND MOLECULAR LIFE SCIENCES Goronzy, J. J., Weyand, C. M. 2012; 69 (10): 1615-1623

    Abstract

    Age is an important risk for autoimmunity, and many autoimmune diseases preferentially occur in the second half of adulthood when immune competence has declined and thymic T cell generation has ceased. Many tolerance checkpoints have to fail for an autoimmune disease to develop, and several of those are susceptible to the immune aging process. Homeostatic T cell proliferation which is mainly responsible for T cell replenishment during adulthood can lead to the selection of T cells with increased affinity to self- or neoantigens and enhanced growth and survival properties. These cells can acquire a memory-like phenotype, in particular under lymphopenic conditions. Accumulation of end-differentiated effector T cells, either specific for self-antigen or for latent viruses, have a low activation threshold due to the expression of signaling and regulatory molecules and generate an inflammatory environment with their ability to be cytotoxic and to produce excessive amounts of cytokines and thereby inducing or amplifying autoimmune responses.

    View details for DOI 10.1007/s00018-012-0970-0

    View details for Web of Science ID 000303509800007

    View details for PubMedID 22466672

  • Signal inhibition by the dual-specific phosphatase 4 impairs T cell-dependent B-cell responses with age PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Yu, M., Li, G., Lee, W., Yuan, M., Cui, D., Weyand, C., Goronzy, J. J. 2012; 109 (15): E879-E888

    Abstract

    T cell-dependent B-cell responses decline with age, suggesting defective CD4 T-cell function. CD4 memory T cells from individuals older than 65 y displayed increased and sustained transcription of the dual-specific phosphatase 4 (DUSP4) that shortened expression of CD40-ligand (CD40L) and inducible T-cell costimulator (ICOS) (both P < 0.001) and decreased production of IL-4, IL-17A, and IL-21 (all P < 0.001) after in vitro activation. In vivo after influenza vaccination, activated CD4 T cells from elderly individuals had increased DUSP4 transcription (P = 0.002), which inversely correlated with the expression of CD40L (r = 0.65, P = 0.002), ICOS (r = 0.57, P = 0.008), and IL-4 (r = 0.66, P = 0.001). In CD4 KO mice reconstituted with DUSP4 OT-II T cells, DUSP4 had a negative effect on the expansion of antigen-specific B cells (P = 0.003) and the production of ova-specific antibodies (P = 0.03) after immunization. Silencing of DUSP4 in memory CD4 T cells improved CD40L (P < 0.001), IL-4 (P = 0.007), and IL-21 (P = 0.04) expression significantly more in the elderly than young adults. Consequently, the ability of CD4 memory T cells to support B-cell differentiation that was impaired in the elderly (P = 0.004) was restored. Our data suggest that increased DUSP4 expression in activated T cells in the elderly in part accounts for defective adaptive immune responses.

    View details for DOI 10.1073/pnas.1109797109

    View details for PubMedID 22434910

  • Dampened ERK signaling in hematopoietic progenitor cells in rheumatoid arthritis CLINICAL IMMUNOLOGY Colmegna, I., Pryshchep, S., Oishi, H., Goronzy, J. J., Weyand, C. M. 2012; 143 (1): 73-82

    Abstract

    In rheumatoid arthritis (RA), hematopoietic progenitor cells (HPC) have age-inappropriate telomeric shortening suggesting premature senescence and possible restriction of proliferative capacity. In response to hematopoietic growth factors RA-derived CD34(+) HPC expanded significantly less than age-matched controls. Cell surface receptors for stem cell factor (SCF), Flt 3-Ligand, IL-3 and IL-6 were intact in RA HPC but the cells had lower transcript levels of cell cycle genes, compatible with insufficient signal strength in the ERK pathway. Cytokine-induced phosphorylation of ERK1/2 was diminished in RA HPC whereas phosphorylated STAT3 and STAT5 molecules accumulated to a similar extent as in controls. Confocal microscopy demonstrated that the membrane-proximal colocalization of K-Ras and B-Raf was less efficient in RA-derived CD34(+) cells. Thus, hyporesponsiveness of RA HPC to growth factors results from dampening of the ERK signaling pathways; with a defect localized in the very early steps of the ERK signaling cascade.

    View details for DOI 10.1016/j.clim.2012.01.007

    View details for Web of Science ID 000301908800010

    View details for PubMedID 22342385

    View details for PubMedCentralID PMC3303971

  • Dynamic Immune Cell Accumulation During Flow-Induced Atherogenesis in Mouse Carotid Artery An Expanded Flow Cytometry Method ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY Alberts-Grill, N., Rezvan, A., Son, D. J., Qiu, H., Kim, C. W., Kemp, M. L., Weyand, C. M., Jo, H. 2012; 32 (3): 623-U217

    Abstract

    Inflammation plays a central role in atherosclerosis. However, the detailed changes in the composition and quantity of leukocytes in the arterial wall during atherogenesis are not fully understood in part because of the lack of suitable methods and animal models.We developed a 10-fluorochrome, 13-parameter flow cytometry method to quantitate 7 major leukocyte subsets in a single digested arterial wall sample. Apolipoprotein E-deficient mice underwent left carotid artery (LCA) partial ligation and were fed a high-fat diet for 4 to 28 days. Monocyte/macrophages, dendritic cells, granulocytes, natural killer cells, and CD4 T cells significantly infiltrated the LCA as early as 4 days. Monocyte/macrophages and dendritic cells decreased between 7 and 14 days, whereas T-cell numbers remained steady. Leukocyte numbers peaked at 7 days, preceding atheroma formation at 14 days. B cells entered LCA by 14 days. Control right carotid and sham-ligated LCAs showed no significant infiltrates. Polymerase chain reaction and ELISA arrays showed that expression of proinflammatory cytokines and chemokines peaked at 7 and 14 days postligation, respectively.This is the first quantitative description of leukocyte number and composition over the life span of murine atherosclerosis. These results show that disturbed flow induces rapid and dynamic leukocyte accumulation in the arterial wall during the initiation and progression of atherosclerosis.

    View details for DOI 10.1161/ATVBAHA.111.242180

    View details for Web of Science ID 000300639300017

    View details for PubMedID 22247254

    View details for PubMedCentralID PMC3336102

  • Mechanisms of immunosenescence: lessons from models of accelerated immune aging YEAR IN IMMUNOLOGY Le Saux, S., Weyand, C. M., Goronzy, J. J. 2012; 1247: 69-82

    Abstract

    With increasing age, the ability of the adaptive immune system to respond to vaccines and to protect from infection declines. In parallel, the production of inflammatory mediators increases. While cross-sectional studies have been successful in defining age-dependent immunological phenotypes, studies of accelerated immune aging in human subpopulations have been instrumental in obtaining mechanistic insights. The immune system depends on its regenerative capacity; however, the T cell repertoire, once established, is relatively robust to aging and only decompensates when additionally stressed. Such stressors include chronic infections such as CMV and HIV, even when viral replication is controlled, and autoimmune diseases. Reduced regenerative capacity, chronic immune activation in the absence of cell exhaustion, T cell memory inflation, and accumulation of highly potent effector T cells in these patients synergize to develop an immune phenotype that is characteristic of the elderly. Studies of accelerated immune aging in autoimmune diseases have identified an unexpected link to chronic DNA damage responses that are known to be important in aging, but so far had not been implicated in immune aging.

    View details for DOI 10.1111/j.1749-6632.2011.06297.x

    View details for Web of Science ID 000305676600007

    View details for PubMedID 22224726

  • Telomere dysfunction, autoimmunity and aging. Aging and disease Hohensinner, P. J., Goronzy, J. J., Weyand, C. M. 2011; 2 (6): 524-537

    Abstract

    Immune aging is associated with loss of critical immune functions, such as host protection from infection and malignancy. Unexpectedly, immunosenescence also renders the host susceptible to inflammation, which may translate into tissue-damaging disease as the senescent immune system loses its ability to maximize inflammatory protection while minimizing inflammatory injury. On the other hand, chronic inflammation associated with immune-mediated disease represents a profound stress factor for the immune system, affecting cellular turn-over, replication and exhaustion. Immune cell longevity is tightly connected to the functional integrity of telomeres which are regulated by cell multiplication, exposure to oxidative stress and DNA repair mechanisms. Lymphocytes are amongst the few cell types that can actively elongate telomeres through the action of telomerase. In patients with the autoimmune disease rheumatoid arthritis (RA), telomerase deficiency is associated with prematurity of immune aging. Patients with RA have other defects in DNA repair mechanisms, including the kinase Ataxia telangiectasia mutated (ATM), critically involved in the repair of DNA double strand breaks. ATM deficiency in RA shortens lymphocyte survival. Dynamics of telomeric length and structure are beginning to be understood and have distinct patterns in different autoimmune diseases, suggesting a multitude of molecular mechanisms defining the interface between chronic immune stimulation and progressive aging of the immune system.

    View details for PubMedID 22396899

  • Telomere Dysfunction, Autoimmunity and Aging AGING AND DISEASE Hohensinner, P. J., Goronzy, J. J., Weyand, C. M. 2011; 2 (6): 524-537

    Abstract

    Immune aging is associated with loss of critical immune functions, such as host protection from infection and malignancy. Unexpectedly, immunosenescence also renders the host susceptible to inflammation, which may translate into tissue-damaging disease as the senescent immune system loses its ability to maximize inflammatory protection while minimizing inflammatory injury. On the other hand, chronic inflammation associated with immune-mediated disease represents a profound stress factor for the immune system, affecting cellular turn-over, replication and exhaustion. Immune cell longevity is tightly connected to the functional integrity of telomeres which are regulated by cell multiplication, exposure to oxidative stress and DNA repair mechanisms. Lymphocytes are amongst the few cell types that can actively elongate telomeres through the action of telomerase. In patients with the autoimmune disease rheumatoid arthritis (RA), telomerase deficiency is associated with prematurity of immune aging. Patients with RA have other defects in DNA repair mechanisms, including the kinase Ataxia telangiectasia mutated (ATM), critically involved in the repair of DNA double strand breaks. ATM deficiency in RA shortens lymphocyte survival. Dynamics of telomeric length and structure are beginning to be understood and have distinct patterns in different autoimmune diseases, suggesting a multitude of molecular mechanisms defining the interface between chronic immune stimulation and progressive aging of the immune system.

    View details for Web of Science ID 000208950700009

    View details for PubMedCentralID PMC3295061

  • Serpin Treatment Reduces Inflammation in Transplanted Temporal Artery Biopsy Specimens from Patients with Suspected Giant Cell Arteritis in Immunodeficient Mice Chen, H., Zheng, D., Dai, E., Liu, L., Samy, H., Staples, N., Beaver, T., Hess, P., Martin, T., Klodel, C., Ryden, S., Weyand, C. M., Reeves, W., Thoburn, R., Sobel, E., Lucas, A. R. LIPPINCOTT WILLIAMS & WILKINS. 2011
  • The Role of CD8+T Cells, IP-10 and MMP12 in Hypertension Thabet, S. R., Wu, J., Chen, W., Marvar, P., Gongora, M. C., Madhur, M. S., Blinder, Y., Weyand, C., Harrison, D. G. LIPPINCOTT WILLIAMS & WILKINS. 2011: E49–E50
  • Finding Balance: T cell Regulatory Receptor Expression during Aging. Aging and disease Cavanagh, M. M., Qi, Q., Weyand, C. M., Goronzy, J. J. 2011; 2 (5): 398-413

    Abstract

    Aging is associated with a variety of changes to immune responsiveness. Reduced protection against infection, reduced responses to vaccination and increased risk of autoimmunity are all hallmarks of advanced age. Here we consider how changes in the expression of regulatory receptors on the T cell surface contribute to altered immunity during aging.

    View details for PubMedID 22396890

  • Finding Balance: T cell Regulatory Receptor Expression during Aging AGING AND DISEASE Cavanagh, M. M., Qi, Q., Weyand, C. M., Goronzy, J. J. 2011; 2 (5): 398-413

    Abstract

    Aging is associated with a variety of changes to immune responsiveness. Reduced protection against infection, reduced responses to vaccination and increased risk of autoimmunity are all hallmarks of advanced age. Here we consider how changes in the expression of regulatory receptors on the T cell surface contribute to altered immunity during aging.

    View details for Web of Science ID 000208950500006

    View details for PubMedCentralID PMC3295076

  • Magnetic Resonance Angiography in Extracranial Giant Cell Arteritis JCR-JOURNAL OF CLINICAL RHEUMATOLOGY Koenigkam-Santos, M., Sharma, P., Kalb, B., Oshinski, J. N., Weyand, C. M., Goronzy, J. J., Martin, D. R. 2011; 17 (6): 306-310

    Abstract

    Noninvasive diagnosis of giant cell arteritis (GCA) remains challenging, particularly with regard to evaluation of extracranial arterial disease.The objective of the study was to retrospectively review extracranial involvement in patients with GCA and/or polymyalgia rheumatica (PMR), evaluated with magnetic resonance imaging (MRI), especially 3-dimensional contrast-enhanced magnetic resonance angiography images of the aortic arch and its branches.Clinical information, biopsy status, and MRI examinations of 28 patients with GCA/PMR were reviewed. Patient images were mixed randomly with 20 normal control images and were independently reviewed by 2 radiologists. Interobserver agreement for detection of arterial stenosis was determined by the k coefficient.Both readers described vascular alterations in keeping with extracranial GCA in 19 of 28 patients (67%) with good interobserver agreement (k = 0.73) and with even higher agreement on diagnosing nonocclusive versus occlusive disease (k = 1.00). The most common lesions were bilateral axillary stenosis or obstructions, observed by both readers in 8 patients (28%). Among the 19 patients with magnetic resonance angiography lesions in the subclavian/axillary arteries, 12 (75%) had biopsy-proven GCA, but only 5 (41%) of these patients had clinical features of large artery disease.In our series review, MRI could provide accurate information on involvement of the aortic arch and its branches in extracranial GCA, depicting different degrees of stenosis. Our analysis also illustrates that occult large artery vasculitis should be considered in patients without biopsy-proven GCA, patients with classic GCA but without clinical signs of large artery disease, and in patients initially diagnosed as having PMR.

    View details for DOI 10.1097/RHU.0b013e31822acec6

    View details for Web of Science ID 000294483200003

    View details for PubMedID 21869711

  • Inflammation and cardiac outcome CURRENT OPINION IN INFECTIOUS DISEASES Hohensinner, P. J., Niessner, A., Huber, K., Weyand, C. M., Wojta, J. 2011; 24 (3): 259-264

    Abstract

    Inflammation is a key component in cardiovascular disease. Controlling inflammatory events and their subsequent processes holds the potential for novel therapeutic treatment options. Cytokines are the propagators of inflammation. In this review we will discuss important cytokines including IL-6, TNF-α, MCP-1, fractalkine, M-CSF and GDF-15, and their effect on cardiac outcome.Recent studies have shed light on the role of IL-6 in cardiovascular disease. Long-term IL-6 levels are highly associated with coronary heart disease. Molecular studies indicate that a permanent prolongation of STAT signaling in cardiac myocytes might be a potential reason for the detrimental effects of IL-6. TNF-α was long considered to have detrimental effects on myocardial function but recent studies show cardioprotective mechanisms for TNF-α. Macrophage modulating cytokines emerge as interesting molecular targets to treat cardiovascular disease. Especially, the two different subtypes of monocytes, a pro-inflammatory and a reparative subset, and their different chemotactic properties might be possible drug targets. Finally, we discuss GDF-15, which emerges as a novel biomarker in cardiovascular disease reflecting information from several pathological pathways.Cytokines are the main proximal mediators of inflammation and hold the potential of being good molecular targets for novel treatment regimes. Cytokines might be valuable biomarkers, adding information about the pathologic pathways in cardiovascular disease.

    View details for DOI 10.1097/QCO.0b013e328344f50f

    View details for Web of Science ID 000289773600014

    View details for PubMedID 21378564

  • Role of Increased Guanosine Triphosphate Cyclohydrolase-1 Expression and Tetrahydrobiopterin Levels upon T Cell Activation JOURNAL OF BIOLOGICAL CHEMISTRY Chen, W., Li, L., Brod, T., Saeed, O., Thabet, S., Jansen, T., Dikalov, S., Weyand, C., Goronzy, J., Harrison, D. G. 2011; 286 (16): 13846-13851

    Abstract

    Tetrahydrobiopterin (BH(4)) is an essential co-factor for the nitric-oxide (NO) synthases, and in its absence these enzymes produce superoxide (O(2)(·-)) rather than NO. The rate-limiting enzyme for BH(4) production is guanosine triphosphate cyclohydrolase-1 (GTPCH-1). Because endogenously produced NO affects T cell function, we sought to determine whether antigen stimulation affected T cell GTPCH-1 expression and ultimately BH(4) levels. Resting T cells had minimal expression of inducible NOS (NOS2), endothelial NOS (NOS3), and GTPCH-1 protein and nearly undetectable levels of BH(4). Anti-CD3 stimulation of T cells robustly stimulated the coordinated expression of NOS2, NOS3, and GTPCH-1 and markedly increased both GTPCH-1 activity and T cell BH(4) levels. The newly expressed GTPCH-1 was phosphorylated on serine 72 and pharmacological inhibition of casein kinase II reduced GTPCH-1 phosphorylation and blunted the increase in T cell BH(4). Inhibition of GTPCH-1 with diaminohydroxypyrimidine (1 mmol/liter) prevented T cell BH(4) accumulation, reduced NO production, and increased T cell O(2)(·-) production, due to both NOS2 and NOS3 uncoupling. GTPCH-1 inhibition also promoted TH(2) polarization in memory CD4 cells. Ovalbumin immunization of mice transgenic for an ovalbumin receptor (OT-II mice) confirmed a marked increase in T cell BH(4) in vivo. These studies identify a previously unidentified consequence of T cell activation, promoting BH(4) levels, NO production, and modulating T cell cytokine production.

    View details for DOI 10.1074/jbc.M110.191023

    View details for Web of Science ID 000289556200007

    View details for PubMedID 21343293

    View details for PubMedCentralID PMC3077585

  • Regulation of T cell receptor signaling by activation-induced zinc influx JOURNAL OF EXPERIMENTAL MEDICINE Yu, M., Lee, W., Tomar, D., Pryshchep, S., Czesnikiewicz-Guzik, M., Lamar, D. L., Li, G., Singh, K., Tian, L., Weyand, C. M., Goronzy, J. J. 2011; 208 (4): 775-785

    Abstract

    Zinc is a trace element that is essential for innate and adaptive immune responses. In addition to being a structural element of many proteins, zinc also functions as a neurotransmitter and an intracellular messenger. Temporal or spatial changes in bioavailable zinc may influence the activity of several enzymes, including kinases and phosphatases. We provide evidence that zinc functions as an ionic signaling molecule after T cell activation. Cytoplasmic zinc concentrations increased within 1 min after T cell receptor (TCR) triggering, in particular in the subsynaptic compartment. The increase depended on the extracellular zinc concentrations and was inhibited by silencing zinc transporter Zip6. Increased zinc influx reduced the recruitment of SHP-1 to the TCR activation complex, augmented ZAP70 phosphorylation and sustained calcium influx. By calibrating TCR activation thresholds, increased extracellular zinc bioavailability facilitated the induction of T cell proliferative responses to suboptimal stimuli.

    View details for DOI 10.1084/jem.20100031

    View details for PubMedID 21422171

  • The role of CD8+T lymphocytes, IP-10 and MMP12 in hypertension Thabet, S., Wu, J., Blinder, Y., Chen, W., Madhur, M. S., Gongora, M. C., Weyand, C., Harrison, D. G. FEDERATION AMER SOC EXP BIOL. 2011
  • Mechanisms of defective T cell-dependent B cell responses with age Yu, M., Li, G., Lee, W., Yuan, M., Weyand, C., Goronzy, J. AMER ASSOC IMMUNOLOGISTS. 2011
  • Regulation of T-cell receptor signaling by activation-induced zinc influx Lee, W., Yu, M., Tomar, D., Li, G., Czesnikiewicz-Guzik, M., Pryshchep, S., Lamar, D., Singh, K., Weyand, C., Goronzy, J. AMER ASSOC IMMUNOLOGISTS. 2011
  • Extra-telomeric function of telomerase in rheumatoid arthritis Shao, L., Fujii, H., Grisar, J., Jorg, G., Weyand, C. AMER ASSOC IMMUNOLOGISTS. 2011
  • Inflammation, Immunity, and Hypertension HYPERTENSION Harrison, D. G., Guzik, T. J., Lob, H. E., Madhur, M. S., Marvar, P. J., Thabet, S. R., Vinh, A., Weyand, C. M. 2011; 57 (2): 132-140
  • Haematopoietic stem and progenitor cells in rheumatoid arthritis RHEUMATOLOGY Colmegna, I., Weyand, C. M. 2011; 50 (2): 252-260

    Abstract

    RA is the prototypic chronic inflammatory disease, characterized by progressive articular cartilage and bone destruction. The systemic nature of RA is evidenced by the increased risk of atherosclerosis and lymphoproliferative disorders. Components of both the innate and adaptive immune system are implicated in the pathophysiology of the articular and extra-articular manifestations of the disease. A fundamental process in the onset of RA is the breakdown in self-tolerance. Accelerated ageing of immune cells (immunosenescence) appears to be a major mechanism favouring the disruption of tolerance. Telomere erosion, a hallmark of immunosenescence, is present in lymphoid (naïve and memory T cells) and myeloid (granulocytes) cells in RA. The premature ageing process also involves the haematopoietic stem and progenitor cells (CD34(+) HSPC), thus extending the RA immunopathogenesis to include early events in the shaping of the immune system. This review summarizes current concepts of HSPC ageing and its impact on immune regeneration, highlighting the phenotypic and functional similarities between elderly and RA HSPC.

    View details for DOI 10.1093/rheumatology/keq298

    View details for Web of Science ID 000286464100004

    View details for PubMedID 20837497

    View details for PubMedCentralID PMC3021951

  • Giant cell arteritis: immune and vascular aging as disease risk factors ARTHRITIS RESEARCH & THERAPY Mohan, S. V., Liao, Y. J., Kim, J. W., Goronzy, J. J., Weyand, C. M. 2011; 13 (4)

    Abstract

    Susceptibility for giant cell arteritis increases with chronological age, in parallel with age-related restructuring of the immune system and age-induced remodeling of the vascular wall. Immunosenescence results in shrinkage of the naïve T-cell pool, contraction of T-cell diversity, and impairment of innate immunity. Aging of immunocompetent cells forces the host to take alternative routes for protective immunity and confers risk for pathogenic immunity that causes chronic inflammatory tissue damage. Dwindling immunocompetence is particularly relevant as the aging host is forced to cope with an ever growing infectious load. Immunosenescence coincides with vascular aging during which the arterial wall undergoes dramatic structural changes and medium and large arteries lose their pliability and elasticity. On the molecular level, elastic fibers deteriorate and matrix proteins accumulate biochemical modifications. Thus, the aging process impacts the two major biologic systems that liaise to promote giant cell arteritis; the immune system and the vessel wall niche.

    View details for DOI 10.1186/ar3358

    View details for Web of Science ID 000297150200044

    View details for PubMedID 21861860

    View details for PubMedCentralID PMC3239337

  • IFN-gamma and IL-17: the two faces of T-cell pathology in giant cell arteritis CURRENT OPINION IN RHEUMATOLOGY Weyand, C. M., Younge, B. R., Goronzy, J. J. 2011; 23 (1): 43-49

    Abstract

    Granuloma formation in giant cell arteritis (GCA) emphasizes the role of adaptive immunity and highlights the role of antigen-specific T cells. Recent data demonstrate that at least two separate lineages of CD4 T cells participate in vascular inflammation, providing an important clue that multiple disease instigators may initiate pathogenic immunity.IFN-γ-producing Th1 cells and IL-17-producing Th17 cells have been implicated in GCA. Patients with biopsy-positive GCA underwent two consecutive temporal artery biopsies, one prior to therapy and one while on corticosteroids. In untreated patients, Th1 and Th17 cells co-existed in the vascular lesions. Following therapy, Th17 cells were essentially lost, whereas Th1 cells persisted almost unaffected. In the peripheral blood of untreated patients Th17 frequencies were increased eight-fold, but normalized with therapy. Blood Th1 cells were doubled in frequency, independent of therapy. Corticosteroids functioned by selectively suppressing IL-1β, IL-6 and IL-23-releasing antigen-presenting cells (APCs), disrupting induction of Th17 cells.At least two distinct CD4 T-cell subsets promote vascular inflammation in GCA. In early disease, APCs promote differentiation of Th17 as well as Th1 cells. Chronic disease is characterized by persistent Th1-inducing signals, independent of IL-17-mediated inflammation. More than one disease instigator may trigger APCs to induce multiple T-cell lineages. Cocktails of therapies will be needed for appropriate disease control.

    View details for DOI 10.1097/BOR.0b013e32833ee946

    View details for Web of Science ID 000284868700008

    View details for PubMedID 20827207

    View details for PubMedCentralID PMC3081721

  • Inhibition and Genetic Ablation of the B7/CD28 T-Cell Costimulation Axis Prevents Experimental Hypertension CIRCULATION Vinh, A., Chen, W., Blinder, Y., Weiss, D., Taylor, W. R., Goronzy, J. J., Weyand, C. M., Harrison, D. G., Guzik, T. J. 2010; 122 (24): 2529-2537

    Abstract

    The pathogenesis of hypertension remains poorly understood, and treatment is often unsuccessful. Recent evidence suggests that the adaptive immune response plays an important role in this disease. Various hypertensive stimuli cause T-cell activation and infiltration into target organs such as the vessel and the kidney, which promotes vascular dysfunction and blood pressure elevation. Classically, T-cell activation requires T-cell receptor ligation and costimulation. The latter often involves interaction between B7 ligands (CD80 and CD86) on antigen-presenting cells with the T-cell coreceptor CD28. This study was therefore performed to examine the role of this pathway in hypertension.Angiotensin II-induced hypertension increased the presence of activated (CD86(+)) dendritic cells in secondary lymphatic tissues. Blockade of B7-dependent costimulation with CTLA4-Ig reduced both angiotensin II- and deoxycorticosterone acetate (DOCA)-salt-induced hypertension. Activation of circulating T cells, T-cell cytokine production, and vascular T-cell accumulation caused by these hypertensive stimuli were abrogated by CTLA4-Ig. Furthermore, in mice lacking B7 ligands, angiotensin II caused minimal blood pressure elevation and vascular inflammation, and these effects were restored by transplantation with wild-type bone marrow.T-cell costimulation via B7 ligands is essential for development of experimental hypertension, and inhibition of this process could have therapeutic benefit in the treatment of this disease.

    View details for DOI 10.1161/CIRCULATIONAHA.109.930446

    View details for Web of Science ID 000285243200011

    View details for PubMedID 21126972

    View details for PubMedCentralID PMC3064430

  • Statins reduce endothelial cell apoptosis via inhibition of TRAIL expression on activated CD4 T cells in acute coronary syndrome ATHEROSCLEROSIS Sato, K., Nuki, T., Gomita, K., Weyand, C. M., Hagiwara, N. 2010; 213 (1): 33-39

    Abstract

    Statins reduce cardiovascular-related morbidity and mortality, but their effects on inflammation in atherosclerosis are not fully understood. We investigated whether statins can modulate cytotoxic functions of CD4 T cells in acute coronary syndrome (ACS).Fresh CD4 T cells were isolated from 55 patients with ACS without statin treatment on admission and from 34 age-matched controls. CD4 T cells collected from ACS patients induced endothelial cell apoptosis significantly more than control T cells. The TNF-related apoptosis-inducing ligand (TRAIL) receptor DR5 was strongly upregulated on endothelial cells, and TRAIL-specific antibodies effectively blocked CD4 T cell-mediated apoptosis, indicating that T cell-mediated endothelial death was dependent on the TRAIL pathway. Expression of both the activating antigen CD69 and TRAIL was enhanced on ACS T cells. In in-vitro assays rosuvastatin, fluvastatin, and pitavastatin directly blocked CD4 T cell-mediated endothelial cell apoptosis and reduced T cell-expression of CD69 and TRAIL through TCR-induced Extracellar signal-Regulated Kinases (ERK) activation.Activated CD4 T cells expressing TRAIL are enriched in the blood of ACS patients and induce endothelial injury, which may contribute to the destabilization of the plaque. Early statin therapy may suppress T cell-mediated endothelial cell damage in atherosclerotic plaques and thus prevent cardiovascular events.

    View details for DOI 10.1016/j.atherosclerosis.2010.03.034

    View details for Web of Science ID 000283356400058

    View details for PubMedID 20430391

    View details for PubMedCentralID PMC2914144

  • Th17 Cells in Takayasu Arteritis Scientific Sessions on Arteriosclerosis, Thrombosis and Vascular Biology Deng, J., Younge, B. R., Goronzy, J., Weyand, C. M. LIPPINCOTT WILLIAMS & WILKINS. 2010: E198–E198
  • Central and Peripheral Mechanisms of T-Lymphocyte Activation and Vascular Inflammation Produced by Angiotensin II-Induced Hypertension CIRCULATION RESEARCH Marvar, P. J., Thabet, S. R., Guzik, T. J., Lob, H. E., McCann, L. A., Weyand, C., Gordon, F. J., Harrison, D. G. 2010; 107 (2): 263-U186

    Abstract

    We have previously found that T lymphocytes are essential for development of angiotensin II-induced hypertension; however, the mechanisms responsible for T-cell activation in hypertension remain undefined.We sought to study the roles of the CNS and pressure elevation in T-cell activation and vascular inflammation caused by angiotensin II.To prevent the central actions of angiotensin II, we created anteroventral third cerebral ventricle (AV3V) lesions in mice. The elevation in blood pressure in response to angiotensin II was virtually eliminated by AV3V lesions, as was activation of circulating T cells and the vascular infiltration of leukocytes. In contrast, AV3V lesioning did not prevent the hypertension and T-cell activation caused by the peripheral acting agonist norepinephrine. To determine whether T-cell activation and vascular inflammation are attributable to central influences or are mediated by blood pressure elevation, we administered hydralazine (250 mg/L) in the drinking water. Hydralazine prevented the hypertension and abrogated the increase in circulating activated T cells and vascular infiltration of leukocytes caused by angiotensin II.We conclude that the central and pressor effects of angiotensin II are critical for T-cell activation and development of vascular inflammation. These findings also support a feed-forward mechanism in which modest degrees of blood pressure elevation lead to T-cell activation, which in turn promotes inflammation and further raises blood pressure, leading to severe hypertension.

    View details for DOI 10.1161/CIRCRESAHA.110.217299

    View details for Web of Science ID 000280201200013

    View details for PubMedID 20558826

  • Immune Aging and Rheumatoid Arthritis RHEUMATIC DISEASE CLINICS OF NORTH AMERICA Goronzy, J. J., Shao, L., Weyand, C. M. 2010; 36 (2): 297-?

    Abstract

    Immunologic models of rheumatoid arthritis (RA) have to take into account that the disease occurs at an age when immunocompetence is declining and in a host whose immune system shows evidence of accelerated immune aging. By several immune aging biomarkers, the immune system in patients with RA is prematurely aged by more than 20 years. One major pathogenetic mechanism is a defect in telomere maintenance and DNA repair that causes accelerated cell death. These findings in RA are reminiscent of murine autoimmunity models, in which lymphopenia was identified as a major risk factor for autoimmunity. Progress in the understanding of how accelerated immune aging is pathogenetically involved in RA may allow development of new therapeutic approaches that go beyond the use of anti-inflammatory agents and eventually could open new avenues for preventive intervention.

    View details for DOI 10.1016/j.rdc.2010.03.001

    View details for Web of Science ID 000279254800006

    View details for PubMedID 20510235

    View details for PubMedCentralID PMC2914095

  • Promoter choice and translational repression determine cell type-specific cell surface density of the inhibitory receptor CD85j expressed on different hematopoietic lineages BLOOD Lamar, D. L., Weyand, C. M., Goronzy, J. J. 2010; 115 (16): 3278-3286

    Abstract

    CD85j (ILT2/LILRB1/LIR-1) is an inhibitory receptor that recognizes major histocompatibility complex (MHC) class Ia and Ib alleles that are widely expressed on all cell types. On ligand recognition, CD85j diminishes kinase activity by recruiting phosphatases to motifs within its cytoplasmic domain. Within the hematopoietic system, CD85j is expressed with cell-specific patterns and cell surface densities that reflect the different roles of cell contact-mediated inhibition in these lineages. While monocytes ubiquitously have high cell surface expression, B lymphocytes start to express CD85j at intermediate levels during early B-cell maturation and natural killer (NK) cells and T cells exhibit a low level of expression on only a subset of cells. The cell-specific expression pattern is accomplished by 2 complementing but not independent mechanisms. Lymphocytes and monocytes use distinct promoters to drive CD85j expression. The lymphocyte promoter maps 13 kilobases (kb) upstream of the monocyte promoter; its use results in the inclusion of a distant exon into the 5'-untranslated region. A short sequence stretch within this exon has the unique function of repressing CD85j protein translation and is responsible for the subdued expression in lymphocytes. These cell-specific mechanisms allow tailoring of CD85j levels to the distinct roles it plays in different hematopoietic lineages.

    View details for DOI 10.1182/blood-2009-09-243493

    View details for Web of Science ID 000276956500014

    View details for PubMedID 20194892

    View details for PubMedCentralID PMC2858479

  • Loss of miR-181a-mediated DUSP6 repression increases the TCR threshold of naive CD4 T cells in the elderly Li, G., Yu, M., Lee, W., Weyand, C., Goronzy, J. AMER ASSOC IMMUNOLOGISTS. 2010
  • Insufficient Deactivation of the Protein Tyrosine Kinase Lck Amplifies T-Cell Responsiveness in Acute Coronary Syndrome CIRCULATION RESEARCH Pryshchep, S., Goronzy, J. J., Parashar, S., Weyand, C. M. 2010; 106 (4): 769-U269

    Abstract

    In the vulnerable atherosclerotic plaque, T cells may destabilize the tissue structure through direct cell-injurious effector functions. T cells transmit environmental signals, such as recognition of antigen, into cellular responses through regulated phosphorylation of cytoplasmic proteins, with the Src family kinase Lck (lymphocyte-specific protein tyrosine kinase) in critical membrane-proximal position of the T-cell receptor (TCR) signaling cascade. The balance between protein phosphorylation and dephosphorylation defines the signal transduction threshold and determines appropriate T-cell responses.We have examined whether abnormal calibration of intracellular signaling pathways renders acute coronary syndrome (ACS) patients susceptible to disproportionate T-cell responses.Intracellular signaling cascades were quantified in CD4 T cells from ACS patients and control individuals after stimulation with major histocompatibility complex class II-superantigen complexes. ACS T cells mobilized more intracellular calcium and accumulated higher levels of phosphotyrosine than control T cells. Proximal steps in TCR signaling, such as recruitment of ZAP-70 and clustering of TCR complexes in the immune synapse, were abnormally enhanced in ACS T cells. Acceleration of the signaling cascade derived from a proximal defect in ACS T cells, which failed to phosphorylate Lck at Tyr505, extending activation of the Src kinase. Abnormalities in TCR signaling did not correlate with systemic inflammation as measured by C-reactive protein.An intrinsic abnormality in the signaling machinery of ACS T cells resulting in the accumulation of active Lck lowers the TCR threshold and renders lymphocytes hyperreactive and capable of unwanted immune responses.

    View details for DOI 10.1161/CIRCRESAHA.109.206052

    View details for Web of Science ID 000275190500018

    View details for PubMedID 20035083

    View details for PubMedCentralID PMC2859828

  • Induction of Hypertension and Peripheral Inflammation by Reduction of Extracellular Superoxide Dismutase in the Central Nervous System HYPERTENSION Lob, H. E., Marvar, P. J., Guzik, T. J., Sharma, S., McCann, L. A., Weyand, C., Gordon, F. J., Harrison, D. G. 2010; 55 (2): 277-U143

    Abstract

    The circumventricular organs (CVOs) lack a well-formed blood-brain barrier and produce superoxide in response to angiotensin II and other hypertensive stimuli. This increase in central superoxide has been implicated in the regulation of blood pressure. The extracellular superoxide dismutase (SOD3) is highly expressed in cells associated with CVOs and particularly with tanycytes lining this region. To understand the role of SOD3 in the CVOs in blood pressure regulation, we performed intracerebroventricular injection an adenovirus encoding Cre-recombinase (5x10(8) particles per milliliter) in mice with loxP sites flanking the SOD3 coding region (SOD3(loxp/loxp) mice). An adenovirus encoding red-fluorescent protein was injected as a control. Deletion of CVO SOD3 increased baseline blood pressure modestly and markedly augmented the hypertensive response to low-dose angiotensin II (140 ng/kg per day), whereas intracerebroventricular injection of adenovirus encoding red-fluorescent protein had minimal effects on these parameters. Adenovirus encoding Cre-recombinase-treated mice exhibited increased sympathetic modulation of heart rate and blood pressure variability, increased vascular superoxide production, and T-cell activation as characterized by increased circulating CD69(+)/CD3(+) cells. Deletion of CVO SOD3 also markedly increased vascular T-cell and leukocyte infiltration caused by angiotensin II. We conclude that SOD3 in the CVO plays a critical role in the regulation of blood pressure, and its loss promotes T-cell activation and vascular inflammation, in part by modulating sympathetic outflow. These findings provide insight into how central signals produce vascular inflammation in response to hypertensive stimuli, such as angiotensin II.

    View details for DOI 10.1161/HYPERTENSIONAHA.109.142646

    View details for Web of Science ID 000273802500018

    View details for PubMedID 20008675

  • Telomeres and Immunological Diseases of Aging GERONTOLOGY Andrews, N. P., Fujii, H., Goronzy, J. J., Weyand, C. M. 2010; 56 (4): 390-403

    Abstract

    A defining feature of the eukaryotic genome is the presence of linear chromosomes. This arrangement, however, poses several challenges with regard to chromosomal replication and maintenance. To prevent the loss of coding sequences and to suppress gross chromosomal rearrangements, linear chromosomes are capped by repetitive nucleoprotein structures, called telomeres. Each cell division results in a progressive shortening of telomeres that, below a certain threshold, promotes genome instability, senescence, and apoptosis. Telomeric erosion, maintenance, and repair take center stage in determining cell fate. Cells of the immune system are under enormous proliferative demand, stressing telomeric intactness. Lymphocytes are capable of upregulating telomerase, an enzyme that can elongate telomeric sequences and, thus, prolong cellular lifespan. Therefore, telomere dynamics are critical in preserving immune function and have become a focus for studies of immunosenescence and autoimmunity. In this review, we describe the role of telomeric nucleoproteins in shaping telomere architecture and in suppressing DNA damage responses. We summarize new insights into the regulation of telomerase activity, hereditary disorders associated with telomere dysfunction, the role of telomere loss in immune aging, and the impact of telomere dysfunction in chronic inflammatory disease.

    View details for DOI 10.1159/000268620

    View details for Web of Science ID 000278673900005

    View details for PubMedID 20016137

    View details for PubMedCentralID PMC2917738

  • Vasculitis in Systemic Sclerosis INTERNATIONAL JOURNAL OF RHEUMATOLOGY Kao, L., Weyand, C. 2010: 385938

    Abstract

    Systemic sclerosis (SSc) is a multiorgan connective tissue disease characterized by autoantibody production and fibroproliferative stenosis of the microvasculature. The vascoluopathy associated with SSc is considered to be noninflammatory, yet frank vasculitis can complicate SSc, posing diagnostic and therapeutic challenges. Here, we have reviewed the literature for reports of small-, medium-, and large-vessel vasculitis occurring in SSc. Amongst 88 reported cases of vasculitis in SSc, patients with ANCA-associated vasculitis appear to present a unique subclass in that they combined typical features of SSc with the renal manifestation of ANCA-associated glomerulonephritis. Other vasculitic syndromes, including large-vessel vasculitis, Behcet's disease, cryoglobulinemia, and polyarteritis nodosa, are rarely encountered in SSc patients. ANCA-associated vasculitis needs to be considered as a differential diagnosis in SSc patients presenting with renal insufficiency, as renal manifestations may result from distinct disease processes and require appropriate diagnostic testing and treatment.

    View details for PubMedID 20976300

    View details for PubMedCentralID PMC2952802

  • Dendritic cells in atherosclerotic disease CLINICAL IMMUNOLOGY Niessner, A., Weyand, C. M. 2010; 134 (1): 25-32

    Abstract

    Atherosclerosis has been considered a syndrome of dysregulated lipid storage until recent evidence has emphasized the critical contribution of the immune system. Dendritic cells (DC) are positioned at the interface of the innate and adaptive immune system. Recognition of danger signals in atheromas leads to DC activation. Activated DC regulate effector T cells which can kill plaque-resident cells and damage the plaque structure. Two types of DC have been identified in atherosclerotic lesions; classical myeloid DC (mDC) which mainly recognize bacterial signatures and plasmacytoid DC (pDC) which specialize in sensing viral fragments and have the unique potential of producing large amounts of type I interferon (IFN). In human atheromas, type I IFN upregulates expression of the cytotoxic molecule TRAIL which leads to apoptosis of plaque-resident cells. This review will elucidate the role of DC in atherogenesis and particularly in plaque rupture, the underlying pathophysiologic cause of myocardial infarction.

    View details for DOI 10.1016/j.clim.2009.05.006

    View details for Web of Science ID 000273696600003

    View details for PubMedID 19520615

  • ERK-Dependent T Cell Receptor Threshold Calibration in Rheumatoid Arthritis JOURNAL OF IMMUNOLOGY Singh, K., Deshpande, P., Pryshchep, S., Colmegna, I., Liarski, V., Weyand, C. M., Goronzy, J. J. 2009; 183 (12): 8258-8267

    Abstract

    Immune responses to citrullinated neoantigens and clinical efficacy of costimulation blockade indicate a general defect in maintaining T cell tolerance in rheumatoid arthritis (RA). To examine whether TCR threshold calibration contributes to disease pathogenesis, signaling in RA T cells was quantified. RA patients had a selective increase in ERK phosphorylation compared with demographically matched controls due to a mechanism distal of Ras activation. Increased ERK responses included naive and memory CD4 and CD8 T cells and did not correlate with disease activity. The augmented ERK activity delayed SHP-1 recruitment to the TCR synapse and sustained TCR-induced Zap70 and NF-kappaB signaling, facilitating responses to suboptimal stimulation. Increased responsiveness of the ERK pathway was also a characteristic finding in the SKG mouse model of RA where it preceded clinical symptoms. Treatment with subtherapeutic doses of a MEK-1/2 inhibitor delayed arthritis onset and reduced severity, suggesting that increased ERK phosphorylation predisposes for autoimmunity and can be targeted to prevent disease.

    View details for DOI 10.4049/jimmunol.0901784

    View details for Web of Science ID 000272861300071

    View details for PubMedID 20007589

    View details for PubMedCentralID PMC2828269

  • Epigenetic regulation of killer immunoglobulin-like receptor expression in T cells BLOOD Li, G., Yu, M., Weyand, C. M., Goronzy, J. J. 2009; 114 (16): 3422-3430

    Abstract

    With increasing age, T cells gain expression of killer immunoglobulin-like receptors (KIRs) that transmit negative signals and dampen the immune response. KIR expression is induced in CD4 and CD8 T cells by CpG DNA demethylation suggesting epigenetic control. To define the mechanisms that underlie the age-associated preferential KIR expression in CD8 T cells, we examined KIR2DL3 promoter methylation patterns. With age, CD8 T cells developed a patchy and stochastic promoter demethylation even in cells that did not express the KIR2DL3-encoded CD158b protein; complete demethylation of the minimal KIR2DL3 promoter was characteristic for CD158b-expressing cells. In contrast, the promoter in CD4 T cells was fully methylated irrespective of age. The selectivity for CD8 T cells correlated with lower DNMT1 recruitment to the KIR2DL3 promoter which further diminished with age. In contrast, binding of the polycomb protein EZH2 known to be involved in DNMT1 recruitment was not different. Our data suggest that CD8 T cells endure increasing displacement of DNMT1 from the KIR promoter with age, possibly because of an active histone signature. The ensuing partial demethylation lowers the threshold for transcriptional activation and renders CD8 T cells more susceptible to express KIR, thereby contributing to the immune defect in the elderly.

    View details for DOI 10.1182/blood-2009-01-200170

    View details for Web of Science ID 000270834500012

    View details for PubMedID 19628706

    View details for PubMedCentralID PMC2765678

  • Rejuvenating the immune system in rheumatoid arthritis NATURE REVIEWS RHEUMATOLOGY Weyand, C. M., Fujii, H., Shao, L., Goronzy, J. J. 2009; 5 (10): 583-588

    Abstract

    In rheumatoid arthritis (RA), the aging process of the immune system is accelerated. Formerly, this phenomenon was suspected to be a consequence of chronic inflammatory activity. However, newer data strongly suggest that deficiencies in maintaining telomeres and overall DNA stability cause excessive apoptosis of RA T cells, imposing proliferative pressure and premature aging on the system. Already during the early stages of their life cycle, and long before they participate in the inflammatory process, RA T cells are lost owing to increased apoptotic susceptibility. A search for underlying mechanisms has led to the discovery of defective pathways of repairing broken DNA and elongating and protecting telomeric sequences at the chromosomal ends. Two enzymatic machineries devoted to DNA repair and maintenance have been implicated. RA T cells fail to induce sufficient amounts of the telomeric repair enzyme telomerase, leaving telomeric ends uncapped and thus susceptible to damage. Of equal importance, RA T cells produce low levels of the DNA repair enzyme ataxia telangiectasia mutated and the complex of nucleoproteins that sense and fix DNA double-strand breaks. The inability to repair damaged DNA renders naive T cells vulnerable to apoptosis, exhausts T-cell regeneration and reshapes the T cell repertoire. Therapeutic attempts to reset the immune systems of patients with RA and prevent premature immunosenescence should include restoration of DNA repair capability.

    View details for DOI 10.1038/nrrheum.2009.180

    View details for Web of Science ID 000270417600013

    View details for PubMedID 19798035

  • Treating autoimmune disease by targeting CD8(+) T suppressor cells EXPERT OPINION ON BIOLOGICAL THERAPY Konya, C., Goronzy, J. J., Weyand, C. M. 2009; 9 (8): 951-965

    Abstract

    Current treatments for autoimmune disease are hampered by the non-specificity of immunomodulatory interventions, having to accept broad suppression of immunoresponsiveness with potentially serious side effects, such as infection or malignancy. The development of antigen-specific approaches, downregulating pathogenic immune responses while maintaining protective immunity, would be a major step forward. One possible approach involves the targeting of physiological regulatory mechanisms, such as inhibitory CD8 T cells that are now recognized to fine-tune many aspects of immune responses. CD8 T suppressor (Ts) cells may directly inhibit other T cells or condition antigen-presenting cells in such a way that immune amplification steps are dampened. The promise of CD8 Ts cells lies in their potential to disrupt host-injurious immune responses in a targeted fashion. For therapeutic purposes, such CD8 Ts cells could either be generated in vitro and transferred into the host or their numbers and activity could be modulated by treating the patient with established or novel immunomodulators. Emerging evidence shows that several subsets of CD8 Ts cells exist. While there is still considerable uncertainty about the molecular mechanisms through which CD8 Ts cells can reset immune responses to protect the host, their potential diagnostic and therapeutic use is intriguing and has generated renewed interest.

    View details for DOI 10.1517/14712590903020759

    View details for Web of Science ID 000268660100002

    View details for PubMedID 19522557

    View details for PubMedCentralID PMC2790919

  • Clinicopathologic Correlations in Giant Cell Arteritis A Retrospective Study of 107 Cases OPHTHALMOLOGY Zhou, L., Luneau, K., Weyand, C. M., Biousse, V., Newman, N. J., Grossniklaus, H. E. 2009; 116 (8): 1574-1580

    Abstract

    To correlate the pathologic findings of temporal artery biopsies in patients clinically defined as positive, presumed, or negative for giant cell arteritis (GCA).Retrospective case series.Patients evaluated for GCA.Temporal artery biopsies examined between 1989 and 2007 were studied. Clinical information and residual tissue for immunohistochemical staining was identified in 107 patients. Clinical information was used to make a diagnosis of "positive," "presumed," or "negative" GCA. The biopsies were reviewed in a masked fashion and classified as "positive," "indeterminate," or "negative" based on published, classic pathologic diagnosis (CPD) criteria. All biopsies were immunostained for CD3 and CD68 and graded as "negative," "mildly" (+), "moderately" (++), or "markedly" (+++) positive. Clinical and pathologic results were correlated and a modified pathologic diagnosis classification (MPD) scheme was developed. The modified scheme was compared in a masked fashion with the final clinical diagnosis and positive and negative predictive values (PVs) were calculated.Pathologic diagnosis and final clinical diagnosis.Using the MPD classification, there were 25%, 16%, and 61% positive, indeterminate, and negative biopsies, respectively. There was excellent correlation between the modified pathologic criteria and final clinical diagnosis (correlation coefficient 0.997; P<0.0001; kappa = 0.81). The positive PVs for CPD and MPD were 85% and 96%, respectively. The negative PVs for CPD and MPD were 64% and 61%, respectively. Positive and negative biopsies strongly correlated with clinical diagnoses of positive and negative for GCA, respectively, whereas indeterminate cases moderately correlated with presumed GCA. The diagnosis did not change from the original biopsy in 11 patients who had a second biopsy. Immunostaining for CD 68 was helpful in several indeterminate cases.We recommend using the modified histologic classification of temporal artery biopsies. There are indeterminate cases that cannot be further defined using current pathologic classification criteria. A second biopsy has very limited value. Immunostaining for CD68 may be helpful in indeterminate cases, although the diagnosis in these cases is based on clinical judgment.

    View details for DOI 10.1016/j.ophtha.2009.02.027

    View details for Web of Science ID 000268710200025

    View details for PubMedID 19500846

  • Deficiency of the DNA repair enzyme ATM in rheumatoid arthritis JOURNAL OF EXPERIMENTAL MEDICINE Shao, L., Fujii, H., Colmegna, I., Oishi, H., Goronzy, J. J., Weyand, C. M. 2009; 206 (6): 1435-1449

    Abstract

    In rheumatoid arthritis (RA), dysfunctional T cells sustain chronic inflammatory immune responses in the synovium. Even unprimed T cells are under excessive replication pressure, suggesting an intrinsic defect in T cell regeneration. In naive CD4 CD45RA(+) T cells from RA patients, DNA damage load and apoptosis rates were markedly higher than in controls; repair of radiation-induced DNA breaks was blunted and delayed. DNA damage was highest in newly diagnosed untreated patients. RA T cells failed to produce sufficient transcripts and protein of the DNA repair kinase ataxia telangiectasia (AT) mutated (ATM). NBS1, RAD50, MRE11, and p53 were also repressed. ATM knockdown mimicked the biological effects characteristic for RA T cells. Conversely, ATM overexpression reconstituted DNA repair capabilities, response patterns to genotoxic stress, and production of MRE11 complex components and rescued RA T cells from apoptotic death. In conclusion, ATM deficiency in RA disrupts DNA repair and renders T cells sensitive to apoptosis. Apoptotic attrition of naive T cells imposes lymphopenia-induced proliferation, leading to premature immunosenescence and an autoimmune-biased T cell repertoire. Restoration of DNA repair mechanisms emerges as an important therapeutic target in RA.

    View details for DOI 10.1084/jem.20082251

    View details for Web of Science ID 000267133700020

    View details for PubMedID 19451263

    View details for PubMedCentralID PMC2715066

  • Telomerase insufficiency in rheumatoid arthritis PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Fujii, H., Shao, L., Colmegna, I., Goronzy, J. J., Weyand, C. M. 2009; 106 (11): 4360-4365

    Abstract

    In rheumatoid arthritis (RA), chronically stimulated T lymphocytes sustain tissue-destructive joint inflammation. Both naïve and memory T cells in RA are prematurely aged with accelerated loss of telomeres suggesting excessive proliferative pressure or inadequate telomeric maintenance. Upon stimulation, RA naïve CD4 T cells are defective in up-regulating telomerase activity (P < 0.0001) due to insufficient induction of the telomerase component human telomerase reverse transcriptase (hTERT); T cell activation and cell cycle progression are intact. Telomerase insufficiency does not affect memory T cells or CD34 hematopoietic stem cells and is present in untreated patients and independent from disease activity. Knockdown of hTERT in primary human T cells increases apoptotic propensity (P = 0.00005) and limits clonal burst (P = 0.0001) revealing a direct involvement of telomerase in T cell fate decisions. Naïve RA CD4 T cells stimulated through the T cell receptor are highly susceptible to apoptosis, expanding to smaller clonal size. Overexpression of ectopic hTERT in naïve RA T cells conveys apoptotic resistance (P = 0.008) and restores proliferative expansion (P < 0.0001). Telomerase insufficiency in RA results in excessive T cell loss, undermining homeostatic control of the naive T cell compartment and setting the stage for lymphopenia-induced T cell repertoire remodeling. Restoring defective telomerase activity emerges as a therapeutic target in resetting immune abnormalities in RA.

    View details for DOI 10.1073/pnas.0811332106

    View details for Web of Science ID 000264278800056

    View details for PubMedID 19255426

    View details for PubMedCentralID PMC2657451

  • Pathogenesis of medium- and large-vessel vasculitis ZEITSCHRIFT FUR RHEUMATOLOGIE Weyand, C. M., Goronzy, J. J. 2009; 68 (2): 100-?

    Abstract

    Giant cell arteritis (GCA), is a systemic vasculitis which preferentially targets large and medium branches of the upper-body aorta. Typical clinical manifestations result from arterial stenosis/occlusion causing blindness, stroke and aortic arch syndrome. Aortic involvement leads to dissection and aneurysm. On the cellular and molecular level, GCA is a sequel of abnormal innate and adaptive immune responses that occur in the specialized tissue niche of the arterial wall. Based on recent pathogenic studies, a novel disease model for GCA is emerging. It is now understood that the series of pathogenic events begins with dendritic cells (DC) indigenous to the artery's outer wall, leading to inflammatory vasculopathy. Placed close to the vasa vasorum, vascular DC are highly sensitive in recognizing pathogen-associated motifs assigning immune monitoring functions to blood vessels. Thus the large vessels are actively involved in immune monitoring. Each vascular territory expresses a unique profile of pathogen-sensing receptors, emphasizing functional diversity amongst structurally similar arteries. Innate immune stimulators can transform vascular DC into efficient antigen-presenting cells, attracting, activating, and instructing T lymphocytes to acquire tissue-invasive features. Macrophages provide critical tissue-damaging effector functions, directly injuring wall-residing cells and promoting a remodeling process that leads to intimal hyperplasia and luminal occlusion. Novel diagnostic and therapeutic approaches to GCA need to focus on the key position of vascular DC and the signals that break the immunoprivileged state of the vessel wall.

    View details for DOI 10.1007/s00393-008-0374-6

    View details for Web of Science ID 000266196500002

    View details for PubMedID 19224227

  • Toll-Like Receptors 4 and 5 Induce Distinct Types of Vasculitis CIRCULATION RESEARCH Deng, J., Ma-Krupa, W., Gewirtz, A. T., Younge, B. R., Goronzy, J. J., Weyand, C. M. 2009; 104 (4): 488-U139

    Abstract

    Large vessel vasculitides, such as Takayasu arteritis and giant cell arteritis, affect vital arteries and cause clinical complications by either luminal occlusion or vessel wall destruction. Inflammatory infiltrates, often with granulomatous arrangements, are distributed as a panarteritis throughout all of the artery's wall layers or cluster in the adventitia as a perivasculitis. Factors determining the architecture and compartmentalization of vasculitis are unknown. Human macrovessels are populated by indigenous dendritic cells (DCs) positioned in the adventitia. Herein, we report that these vascular DCs sense bacterial pathogens and regulate the patterning of the emerging arteritis. In human temporal artery-SCID chimeras, lipopolysaccharides stimulating Toll-like receptor (TLR)4 and flagellin stimulating TLR5 trigger vascular DCs and induce T-cell recruitment and activation. However, the architecture of the evolving inflammation is ligand-specific; TLR4 ligands cause transmural panarteritis and TLR5 ligands promote adventitial perivasculitis. Underlying mechanisms involve selective recruitment of functional T cell subsets. Specifically, TLR4-mediated DC stimulation markedly enhances production of the chemokine CCL20, biasing recruitment toward CCL20-responsive CCR6(+) T cells. In adoptive transfer experiments, CCR6(+) T cells produce an arteritis pattern with media-invasive T cells damaging vascular smooth muscle cells. Also, CCR6(+) T cells dominate the vasculitic infiltrates in patients with panarteritic giant cell arteritis. Thus, depending on the original danger signal, vascular DCs edit the emerging immune response by differentially recruiting specialized T effector cells and direct the disease process toward distinct types of vasculitis.

    View details for DOI 10.1161/CIRCRESAHA.108.185777

    View details for Web of Science ID 000263771600011

    View details for PubMedID 19150884

    View details for PubMedCentralID PMC2731717

  • Regulation of T-cell function by endogenously produced angiotensin II 10th Annual Meeting for the New Research in Cardiovascular and Kidney Diseases Hoch, N. E., Guzik, T. J., Chen, W., Deans, T., Maalouf, S. A., Gratze, P., Weyand, C., Harrison, D. G. AMER PHYSIOLOGICAL SOC. 2009: R208–R216

    Abstract

    The adaptive immune response and, in particular, T cells have been shown to be important in the genesis of hypertension. In the present study, we sought to determine how the interplay between ANG II, NADPH oxidase, and reactive oxygen species modulates T cell activation and ultimately causes hypertension. We determined that T cells express angiotensinogen, the angiotensin I-converting enzyme, and renin and produce physiological levels of ANG II. AT1 receptors were primarily expressed intracellularly, and endogenously produced ANG II increased T-cell activation, expression of tissue homing markers, and production of the cytokine TNF-alpha. Inhibition of T-cell ACE reduced TNF-alpha production, indicating endogenously produced ANG II has a regulatory role in this process. Studies with specific antagonists and T cells from AT1R and AT2R-deficient mice indicated that both receptor subtypes contribute to TNF-alpha production. We found that superoxide was a critical mediator of T-cell TNF-alpha production, as this was significantly inhibited by polyethylene glycol (PEG)-SOD, but not PEG-catalase. Thus, T cells contain an endogenous renin-angiotensin system that modulates T-cell function, NADPH oxidase activity, and production of superoxide that, in turn, modulates TNF-alpha production. These findings contribute to our understanding of how ANG II and T cells enhance inflammation in cardiovascular disease.

    View details for DOI 10.1152/ajpregu.90521.2008

    View details for Web of Science ID 000262781200003

    View details for PubMedID 19073907

  • Developments in the scientific understanding of rheumatoid arthritis ARTHRITIS RESEARCH & THERAPY Goronzy, J. J., Weyand, C. M. 2009; 11 (5)

    Abstract

    Rheumatoid arthritis (RA) is recognized to be an autoimmune disease that causes preclinical systemic abnormalities and eventually leads to synovial inflammation and destruction of the joint architecture. Recently identified genetic risk factors and novel insights from animal models of spontaneous arthritis have lent support to the concept that thymic selection of an autoreactive T-cell repertoire is an important risk factor for this disease. With advancing age, defects in the homeostatic control of the T-cell pool and in the setting of signaling thresholds lead to the accumulation of pro-inflammatory T-effector cell populations and loss of tolerance to neo-antigens, such as citrullinated peptides. As the breakdown of tolerance to modified self-antigens can precede synovitis by decades, repair of homeostatic defects may open a unique window of opportunity for preventive interventions in RA. The end result of RA, destruction of cartilage and bone, appears to be driven by cytokine- and cell contact-induced activation of synoviocytes and monocytic cells, some of which differentiate into tissue-destructive osteoclasts. Targeting mediators involved in this process has greatly improved the management of this chronic inflammatory syndrome.

    View details for DOI 10.1186/ar2758

    View details for Web of Science ID 000273338400016

    View details for PubMedID 19835638

    View details for PubMedCentralID PMC2787299

  • Vascular damage in giant cell arteritis AUTOIMMUNITY Piggott, K., Biousse, V., Newman, N. J., Goronzy, J. J., Weyand, C. M. 2009; 42 (7): 596-604

    Abstract

    Immune-mediated damage to medium-sized arteries results in wall remodeling with intimal hyperplasia, luminal stenosis and tissue ischemia. In the case of the aorta, vasculitis may result in dissection, aneurysm or rupture. The response-to-injury program of the blood vessel is a concerted action between the immune system and wall-resident cells, involving the release of growth and angiogenic factors from macrophages and giant cells and the migration and hyperproliferation of vascular smooth muscle cells. Innate immune cells, specifically, dendritic cells (DC) positioned in the vessel wall, have been implicated in the earliest steps of vasculitis. Pathogen-derived molecular patterns are capable of activating vascular DC and initiating adaptive immune responses. The pattern of the emerging vessel wall inflammation is ultimately determined by the initial insult. Ligands to toll-like receptor (TLR) 4, such as lipopolysaccharides, facilitate the recruitment of CD4 T cells that invade deep into the wall and distribute in a panarteritic pattern. Conversely, ligands for TLR5 condition vascular DC to support perivasculitic infiltrates. In essence, both innate and adaptive immune reactions collaborate to render the arterial wall susceptible to inflammatory damage. Unique features of the tissue microenvironment, including specialized DC, shape the course of the inflammatory response. Differences in vascular damage pattern encountered in different patients may relate to distinct instigators of vasculitis.

    View details for DOI 10.1080/08916930903002495

    View details for Web of Science ID 000270287500006

    View details for PubMedID 19657775

  • Age-Dependent Signature of Metallothionein Expression in Primary CD4 T Cell Responses Is Due to Sustained Zinc Signaling REJUVENATION RESEARCH Lee, W., Cui, D., Czesnikiewicz-Guzik, M., Vencio, R. Z., Shmulevich, I., Aderem, A., Weyand, C. M., Goronzy, J. J. 2008; 11 (6): 1001-1011

    Abstract

    The ability to mount adaptive immune responses to vaccinations and viral infections declines with increasing age. To identify mechanisms leading to immunosenescence, primary CD4 T cell responses were examined in 60- to 75-year-old individuals lacking overt functional defects. Transcriptome analysis indicated a selective defect in zinc homeostasis. CD4 T cell activation was associated with zinc influx via the zinc transporter Zip6, leading to increased free cytoplasmic zinc and activation of negative feedback loops, including the induction of zinc-binding metallothioneins. In young adults, activation-induced cytoplasmic zinc concentrations declined after 2 days to below prestimulation levels. In contrast, activated naïve CD4 T cells from older individuals failed to downregulate cytoplasmic zinc, resulting in excessive induction of metallothioneins. Activation-induced metallothioneins regulated the redox state in activated T cells and accounted for an increased proliferation of old CD4 T cells, suggesting that regulation of T cell zinc homeostasis functions as a compensatory mechanism to preserve the replicative potential of naïve CD4 T cells with age.

    View details for DOI 10.1089/rej.2008.0747

    View details for Web of Science ID 000262886400003

    View details for PubMedID 19072254

    View details for PubMedCentralID PMC2848531

  • Inhibitory CD8(+) T cells in autoimmune disease HUMAN IMMUNOLOGY Suzuki, M., Konya, C., Goronzy, J. J., Weyand, C. M. 2008; 69 (11): 781-789

    Abstract

    Rheumatologists have long been focused on developing novel immunotherapeutic agents to manage such prototypic autoimmune diseases as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). The ultimate challenge in providing immunosuppressive treatment for patients with RA and SLE has derived from the dilemma that both protective and harmful immune responses result from adaptive immune responses, mediated by highly diverse, antigen-specific T and B cells endowed with powerful effector functions and the ability for long-lasting memory. As regulatory/suppressor T cells can suppress immunity against any antigen, including self-antigens, they emerge as an ideal therapeutic target. Several distinct subtypes of CD8(+) suppressor cells (Ts) have been described that could find application in treating RA or SLE. In a xenograft model of human synovium, CD8(+)CD28(-)CD56(+) T cells effectively suppressed rheumatoid inflammation. Underlying mechanisms involve conditioning of antigen presenting cells (APC). Adoptively transferred CD8(+) T cells characterized by IL-16 secretion have also exhibited disease-inhibitory effects. In mice with polyarthritis, CD8(+) Ts suppressed inflammation by IFNgamma-mediated modulation of the tryptophan metabolism in APC. In SLE animal models, CD8(+) Ts induced by a synthetic peptide exerted suppressive activity mainly via the TGFbeta-Foxp3-PD1 pathway. CD8(+) Ts induced by histone peptides were found to downregulate disease activity by secreting TGFbeta. In essence, disease-specific approaches may be necessary to identify CD8(+) Ts optimally suited to treat immune dysfunctions in different autoimmune syndromes.

    View details for DOI 10.1016/j.humimm.2008.08.283

    View details for Web of Science ID 000261565500018

    View details for PubMedID 18812196

    View details for PubMedCentralID PMC2614126

  • Is hypertension an immunologic disease? Current cardiology reports Harrison, D. G., Guzik, T. J., Goronzy, J., Weyand, C. 2008; 10 (6): 464-469

    Abstract

    Several studies published in the past three decades have suggested that the adaptive immune system contributes to hypertension. Recent studies have shown that T cells play a crucial role in the blood pressure elevation caused by angiotensin II and in response to sodium and volume challenge. Hypertensive stimuli cause effector T cells to enter visceral fat, in particular perivascular fat, where they release cytokines that promote vasoconstriction. Similarly, effector T cells accumulate in the kidney in hypertension and contribute to renal dysfunction, promoting sodium and volume retention. These findings provide some insight into the relationship between inflammation and hypertension and suggest that efforts to reduce T-cell activation may be useful in preventing or treating this disease.

    View details for PubMedID 18950555

  • T-cell co-stimulatory pathways in autoimmunity ARTHRITIS RESEARCH & THERAPY Goronzy, J. J., Weyand, C. M. 2008; 10

    Abstract

    T-cell activation and differentiation depend on the signal strength received by the T-cell receptor and on signals provided by co-stimulatory molecules. The most prominent co-stimulatory molecule is CD28, which controls the activation of naïve and memory T cells by antigen presented on professional antigen-presenting cells. Blocking of the CD28-CD80/86 pathway has been an appealing strategy for inducing tolerance in autoimmune diseases where the disease-inducing autoantigens are not known. Although CD28 has maintained its unique position, the past decade has witnessed the recognition that a large number of regulatory molecules on T cells must be stimulated to generate a fully protective immune response. These regulatory receptors differ in their preferential expression on T-cell subsets, in the ligands that they recognize, and in the signaling pathways that they trigger. They have in common the fact that they provide information on the cellular environment in which the T-cell response occurs. By intercepting these signals, we may be able to influence disease-relevant T-cell responses in autoimmune diseases while potentially minimizing broad immunosuppression.

    View details for DOI 10.1186/ar2414

    View details for Web of Science ID 000264562800003

    View details for PubMedID 19007423

    View details for PubMedCentralID PMC2582810

  • T cells in arteritis and atherosclerosis CURRENT OPINION IN LIPIDOLOGY Weyand, C. M., Younge, B. R., Goronzy, J. J. 2008; 19 (5): 469-477

    Abstract

    Inflammatory vasculopathies, spanning from atherosclerosis to vasculitides, are driven by innate and adaptive immune responses. Instructed by antigen-presenting cells, T cells have unsurpassed skills to orchestrate protective and pathogenic immunity. Pro-inflammatory and anti-inflammatory T cells regulate master pathogenic pathways, providing a framework for novel immunotherapeutic strategies.The multilayered wall of macrovessels creates a unique tissue niche; professional antigen-presenting cells, specifically dendritic cells, are superior in triggering and maintaining T-cell responses in this tissue milieu. Plaque-residing dendritic cells sense pathogen-derived motifs and edit inflammatory responses. T cells respond to antigen but antigen-nonspecific factors setting cellular response thresholds may be equally important. Dysregulated signal transduction pathways emerge as highly relevant in biasing T cells toward hyperresponsiveness. In the inflamed atheroma and in arteritic lesions, pathogenic T cells coordinate multiple injury pathways. Besides inducing tissue-damaging macrophage functions, they directly inflict cellular injury within the arterial wall. Distinctively, selected T cells induce smooth muscle cell apoptosis, most prominently by upregulating the death-receptor ligand TRAIL.Innate sentinels, specifically dendritic cells, populate normal arteries, intramural vasculitic lesions, and the inflamed atheroma. They sense microbial motifs and instruct T cells toward pro-inflammatory and tissue-destructive effector functions. Microenvironmental factors imposed by the unique structure of the arterial wall appear to be highly conserved across disease entities, modulating inflammation in atherosclerosis and arteritis.

    View details for DOI 10.1097/MOL.0b013e32830bfdc2

    View details for Web of Science ID 000259349800005

    View details for PubMedID 18841594

    View details for PubMedCentralID PMC2682337

  • Vessel-specific Toll-like receptor profiles in human medium and large arteries CIRCULATION Pryshchep, O., Ma-Krupa, W., Younge, B. R., Goronzy, J. J., Weyand, C. M. 2008; 118 (12): 1276-1284

    Abstract

    Inflammatory vasculopathies, ranging from the vasculitides (Takayasu arteritis, giant cell arteritis, and polyarteritis nodosa) to atherosclerosis, display remarkable target tissue tropisms for selected vascular beds. Molecular mechanisms directing wall inflammation to restricted anatomic sites within the vascular tree are not understood. We have examined the ability of 6 different human macrovessels (aorta and subclavian, carotid, mesenteric, iliac, and temporal arteries) to initiate innate and adaptive immune responses by comparing pathogen-sensing and T-cell-stimulatory capacities.Gene expression analysis for pathogen-sensing Toll-like receptors (TLRs) 1 to 9 showed vessel-specific profiles, with TLR2 and TLR4 ubiquitously present, TLR7 and TLR9 infrequent, and TLR1, TLR3, TLR5, TLR6, and TLR8 expressed in selective patterns. Experiments with vessel walls stripped of the intimal or adventitial layer identified dendritic cells at the media-adventitia junction as the dominant pathogen sensors. In human artery-severe combined immunodeficiency (SCID) mouse chimeras, adoptively transferred human T cells initiated vessel wall inflammation if wall-embedded dendritic cells were conditioned with TLR ligands. Wall-infiltrating T cells displayed vessel-specific activation profiles with differential production of CD40L, lymphotoxin-alpha, and interferon-gamma. Vascular bed-specific TLR fingerprints were functionally relevant, as exemplified by differential responsiveness of iliac and subclavian vessels to TLR5 but not TLR4 ligands.Populated by indigenous dendritic cells, medium and large human arteries have immune-sensing and T-cell-stimulatory functions. Each vessel in the macrovascular tree exhibits a distinct TLR profile and supports selective T-cell responses, imposing vessel-specific risk for inflammatory vasculopathies.

    View details for DOI 10.1161/CIRCULATIONAHA.108.789172

    View details for Web of Science ID 000259224800009

    View details for PubMedID 18765390

    View details for PubMedCentralID PMC2748975

  • Epigenetic mechanisms of age-dependent KIR2DL4 expression in T cells JOURNAL OF LEUKOCYTE BIOLOGY Li, G., Weyand, C. M., Goronzy, J. J. 2008; 84 (3): 824-834

    Abstract

    Killer Ig-like receptor (KIR) expression is mostly restricted to NK cells controlling their activation. With increasing age, KIRs are expressed on T cells and contribute to age-related diseases. We examined epigenetic mechanisms that determine the competency of T cells to transcribe KIR2DL4. Compared with Jurkat cells and CD4(+)CD28(+) T cells from young individuals, DNA methyltransferase (DNMT) inhibition was strikingly more effective in T cells from elderly adults and the CD4(+)CD28(-) T cell line HUT78 to induce KIR2DL4 transcription. In these susceptible cells, the KIR2DL4 promoter was partially demethylated, and dimethylated H3-Lys 4 was increased, and all other histone modifications were characteristic for an inactive promoter. In comparison, NK cells had a fully demethylated KIR2DL4 promoter and the full spectrum of histone modifications indicative of active transcription with H3 and H4 acetylation, di- and trimethylated H3-Lys 4, and reduced, dimethylated H3-Lys 9. These results suggest that an increased competency of T cells to express KIR2DL4 with aging is conferred by a selective increase in H3-Lys 4 dimethylation and limited DNA demethylation. The partially accessible promoter is sensitive to DNMT inhibition, which is sufficient to induce full transcription without further histone acetylation and methylation.

    View details for DOI 10.1189/jlb.0807583

    View details for Web of Science ID 000258525500029

    View details for PubMedID 18586981

    View details for PubMedCentralID PMC2516893

  • The treatment of giant cell arteritis. Reviews in neurological diseases Fraser, J. A., Weyand, C. M., Newman, N. J., Biousse, V. 2008; 5 (3): 140-152

    Abstract

    Although giant cell arteritis (GCA) is a well-known vasculitis sensitive to corticosteroid-mediated immunosuppression, numerous issues of long-term therapeutic management remain unresolved. Because GCA encompasses a broad spectrum of clinical subtypes, ranging from devastating visual loss and neurological deficits to isolated systemic symptoms, the treatment of GCA must be adjusted to each case, and recommendations vary widely in the literature. This article systematically reviews the treatment options for patients with neuro-ophthalmic and neurological complications of GCA, as well as the evidence for possible adjuvant therapies for patients with GCA. Although there is no randomized controlled clinical trial specifically evaluating GCA patients with ocular and neurological complications, we recommend that GCA patients with acute visual loss or brain ischemia be admitted to the hospital for high-dose intravenous methyl-prednisolone, close monitoring, and prevention of steroid-induced complications. Aspirin may also be helpful in these cases. The evidence supporting the use of steroid-sparing immunomodulatory agents such as methotrexate for long-term management remains debated.

    View details for PubMedID 18838954

  • TLR-mediated induction of negative regulatory ligands on dendritic cells JOURNAL OF MOLECULAR MEDICINE-JMM Groschel, S., Piggott, K. D., Vaglio, A., Ma-Krupa, W., Singh, K., Goronzy, J. J., Weyand, C. M. 2008; 86 (4): 443-455

    Abstract

    Dendritic cells (DCs) shape T-cell response patterns and determine early, intermediate, and late outcomes of immune recognition events. They either facilitate immunostimulation or induce tolerance, possibly determined by initial DC activation signals, such as binding Toll-like receptor (TLR) ligands. Here, we report that DC stimulation through the TLR3 ligand dsRNA [poly(I:C)] limits CD4 T-cell proliferation, curtailing adaptive immune responses. CD4+ T cells instructed by either lipopolysaccharide (LPS) or poly(I:C)-conditioned DCs promptly upregulated the activation marker CD69. Whereas LPS-pretreated DCs subsequently sustained T-cell clonal expansion, proliferation of CD4+ T cells exposed to poly(I:C)-pretreated DCs was markedly suppressed. This proliferative defect required DC-T cell contact, was independent of IFN-alpha, and was overcome by exogenous IL-2, indicating T-cell anergy. Coinciding with the downregulation, CD4+ T cells expressed the inhibitory receptor PD-1. Antibodies blocking the PD-1 ligand PD-L1 restored proliferation. dsRNA-stimulated DCs preferentially induced PD-L1, whereas poly(I:C) and LPS both upregulated the costimulatory molecule CD86 to a comparable extent. Poly(dA-dT), a ligand targeting the cytoplasmic RNA helicase pattern-recognition pathway, failed to selectively induce PD-L1 upregulation, assigning this effect to the TLR3 pathway. Poly(I:C)-conditioned DCs promoted accumulation of phosphorylated SHP-2, the intracellular phosphatase mediating PD-1 inhibitory effects. The ability of dsRNA to bias DC differentiation toward providing inhibitory signals to interacting CD4+ T cells may be instrumental in viral immune evasion. Conversely, TLR3 ligands may have therapeutic value in silencing pathogenic immune responses.

    View details for DOI 10.1007/s00109-008-0310-x

    View details for Web of Science ID 000255093600010

    View details for PubMedID 18253710

    View details for PubMedCentralID PMC2556182

  • T cell subset-specific susceptibility to aging CLINICAL IMMUNOLOGY Czesnikiewicz-Guzik, M., Lee, W., Cui, D., Hiruma, Y., Lamar, D. L., Yang, Z., Ouslander, J. G., Weyand, C. M., Goronzy, J. J. 2008; 127 (1): 107-118

    Abstract

    With increasing age, the competence of the immune system to fight infections and tumors declines. Age-dependent changes have been mostly described for human CD8 T cells, raising the question of whether the response patterns for CD4 T cells are different. Gene expression arrays of memory CD4 T cells yielded a similar age-induced fingerprint as has been described for CD8 T cells. In cross-sectional studies, the phenotypic changes were not qualitatively different for CD4 and CD8 T cells, but occurred much more frequently in CD8 T cells. Homeostatic stability partially explained this lesser age sensitivity of CD4 T cells. With aging, naïve and central memory CD8 T cells were lost at the expense of phenotypically distinct CD8 effector T cells, while effector CD4 T cells did not accumulate. However, phenotypic shifts on central memory T cells were also more pronounced in CD8 T cells. This distinct stability in cell surface marker expression can be reproduced in vitro. The data show that CD8 T cells are age sensitive by at least two partially independent mechanisms: fragile homeostatic control and gene expression instability in a large set of regulatory cell surface molecules.

    View details for DOI 10.1016/j.clim.2007.12.002

    View details for Web of Science ID 000254501700013

    View details for PubMedID 18222733

    View details for PubMedCentralID PMC2435295

  • Defective proliferative capacity and accelerated telomeric loss of hematopoietic progenitor cells in rheumatoid arthritis ARTHRITIS AND RHEUMATISM Colmegna, I., Diaz-Borjon, A., Fujii, H., Schaefer, L., Goronzy, J. J., Weyand, C. M. 2008; 58 (4): 990-1000

    Abstract

    In rheumatoid arthritis (RA), telomeres of lymphoid and myeloid cells are age-inappropriately shortened, suggesting excessive turnover of hematopoietic precursor cells (HPCs). The purpose of this study was to examine the functional competence (proliferative capacity, maintenance of telomeric reserve) of CD34+ HPCs in RA.Frequencies of peripheral blood CD34+,CD45+ HPCs from 63 rheumatoid factor-positive RA patients and 48 controls matched for age, sex, and ethnicity were measured by flow cytometry. Proliferative burst, cell cycle dynamics, and induction of lineage-restricted receptors were tested in purified CD34+ HPCs after stimulation with early hematopoietins. Telomere sequences were quantified by real-time polymerase chain reaction. HPC functions were correlated with the duration, activity, and severity of RA as well as its treatment.In healthy donors, CD34+ HPCs accounted for 0.05% of nucleated cells; their numbers were strictly age dependent and declined at a rate of 1.3% per year. In RA patients, CD34+ HPC frequencies were age-independently reduced to 0.03%. Upon growth factor stimulation, control HPCs passed through 5 replication cycles over 4 days. In contrast, RA-derived HPCs completed only 3 generations. Telomeres of RA CD34+ HPCs were age-inappropriately shortened by 1,600 bp. All HPC defects were independent of disease duration, disease activity, and smoking status, and were present to the same degree in untreated patients.In RA, circulating bone marrow-derived progenitor cells were diminished, and concentrations stagnated at levels typical of those in old control subjects. HPCs from RA patients displayed growth factor nonresponsiveness and sluggish cell cycle progression; marked telomere shortening indicated proliferative stress-induced senescence. Defective HPC function independent of disease activity markers suggests bone marrow failure as a potential pathogenic factor in RA.

    View details for DOI 10.1002/art.23287

    View details for Web of Science ID 000255345200010

    View details for PubMedID 18383391

    View details for PubMedCentralID PMC2820283

  • Vessel wall-embedded dendritic cells induce T-Cell autoreactivity and initiate vascular inflammation CIRCULATION RESEARCH Han, J. W., Shimada, K., Ma-Krupa, W., Johnson, T. L., Nerem, R. M., Goronzy, J. J., Weyand, C. M. 2008; 102 (5): 546-553

    Abstract

    Human medium-sized and large arteries are targeted by inflammation with innate and adaptive immune responses occurring within the unique microspace of the vessel wall. How 3D spatial arrangements influence immune recognition and cellular response thresholds and which cell populations sense immunoactivating ligands and function as antigen-presenting cells are incompletely understood. To mimic the 3D context of human arteries, bioartificial arteries were engineered from collagen type I matrix, human vascular smooth muscle cells (VSMCs), and human endothelial cells and populated with cells implicated in antigen presentation and T-cell stimulation, including monocytes, macrophages, and myeloid dendritic cells (DCs). Responsiveness of wall-embedded antigen-presenting cells was probed with the Toll-like receptor ligand lipopolysaccharide, and inflammation was initiated by adding autologous CD4(+) T cells. DCs colonized the outermost VSMC layer, recapitulating their positioning at the media-adventitia border of normal arteries. Wall-embedded DCs responded to the microbial product lipopolysaccharide by entering the maturation program and upregulating the costimulatory ligand CD86. Activated DCs effectively stimulated autologous CD4 T cells, which produced the proinflammatory cytokine interferon-gamma and infiltrated deeply into the VSMC layer, causing matrix damage. Lipopolysaccharide-triggered macrophages were significantly less efficacious in recruiting T cells and promoting T-cell stimulation. CD14(+) monocytes, even when preactivated, failed to support initial steps of vascular wall inflammation. Innate immune cells, including monocytes, macrophages, and DCs, display differential functions in the vessel wall. DCs are superior in sensing pathogen-derived motifs and are highly efficient in breaking T-cell tolerance, guiding T cells toward proinflammatory and tissue-invasive behavior.

    View details for DOI 10.1161/CIRCRESAHA.107.161653

    View details for Web of Science ID 000253989500008

    View details for PubMedID 18202318

  • Synoviocyte stimulation by the LFA-1-intercellular adhesion molecule-2-Ezrin-Akt pathway in rheumatoid arthritis JOURNAL OF IMMUNOLOGY Singh, K., Colmegna, I., He, X., Weyand, C. M., Goronzy, J. J. 2008; 180 (3): 1971-1978

    Abstract

    In rheumatoid arthritis (RA), the synovium is infiltrated by mononuclear cells that influence the proliferation and activation of fibroblast-like synoviocytes (FLS) through soluble mediators as well as cell-to-cell contact. To identify receptor-ligand pairs involved in this cross-talk, we cocultured T cells with FLS lines isolated from synovial tissues from RA patients. Coculture with T cells induced phosphorylation of Akt (Ser(473)) and its downstream mediators, GSK-3alpha/GSK-beta, FoxO1/3a, and mouse double minute-2, and enhanced FLS proliferation. T cell-mediated phospho-Akt up-regulation was unique for FLS as no such effect was observed upon interaction of T cells with dendritic cells and B cells. Akt activation was induced by all functional T cell subsets independent of MHC/Ag recognition and was also found with other leukocyte populations, suggesting the involvement of a common leukocyte cell surface molecule. Akt phosphorylation, enhanced in vitro FLS proliferation, and enhanced FLS IL-6 production was inhibited by blocking Abs to CD11a and ICAM-2 whereas Abs to ICAM-1 had a lesser effect. Selective involvement of the LFA-1-ICAM-2 pathway was confirmed by the finding of increased ezrin phosphorylation at Tyr(353) that is known to be downstream of ICAM-2 and supports cell survival through Akt activation. CD28(-) T cells, which are overrepresented in RA patients, have high CD11a cell surface expression and induce Akt phosphorylation in FLS more potently than their CD28(+) counterparts. These findings identify ICAM-2 as a potential therapeutic target to inhibit FLS activation in RA, allowing for a more selective intervention than broad LFA-1 inhibition.

    View details for Web of Science ID 000252632900075

    View details for PubMedID 18209096

  • Synergistic proinflammatory effects of the antiviral cytokine interferon-alpha and toll-like receptor 4 ligands in the atherosclerotic plaque CIRCULATION Niessner, A., Shin, M. S., Pryshchep, O., Goronzy, J. J., Chaikof, E. L., Weyand, C. M. 2007; 116 (18): 2043-2052

    Abstract

    Interferon (IFN)-alpha is a pluripotent inflammatory cytokine typically induced by viral infections. In rupture-prone atherosclerotic plaques, plasmacytoid dendritic cells produce IFN-alpha. In the present study we explored the contribution of IFN-alpha to inflammation and tissue injury in the plaque microenvironment.In 53% of carotid plaques (n=30), CD123+ plasmacytoid dendritic cells clustered together with CD11c+ myeloid dendritic cells, a distinct dendritic cell subset specialized in sensing danger signals from bacteria and tissue breakdown. Tissue concentrations of IFN-alpha and tumor necrosis factor (TNF)-alpha transcripts were tightly correlated (r=0.76, P<0.001), suggesting a regulatory role of IFN-alpha in TNF-alpha production. Plaque tissue stimulation with CpG ODN, a Toll-like receptor (TLR) 9 ligand, increased IFN-alpha production (57.8+/-23.7 versus 25.9+/-8.6 pg/mL; P<0.001), whereas the TLR4 ligand lipopolysaccharide induced TNF-alpha secretion (225.1+/-3.0 versus 0.7+/-0.2 pg/mL; P<0.001). Treating plaque tissue with IFN-alpha markedly enhanced lipopolysaccharide-triggered TNF-alpha secretion (559.0+/-25.9 versus 225.1+/-3.0 pg/mL; P<0.001). IFN-alpha pretreatment also amplified the effects of lipopolysaccharide on interleukin-12, interleukin-23, and matrix metalloproteinase-9, suggesting that the antiviral cytokine sensitized myeloid dendritic cells and macrophages toward TLR4 ligands. Mechanistic studies demonstrated that IFN-alpha modulated the myeloid dendritic cell response pattern by upregulating TLR4 expression (P<0.001) involving both the STAT (signal transducer and activator of transcription) and the PI(3)K pathway.In the atherosclerotic plaque, IFN-alpha functions as an inflammatory amplifier. It sensitizes antigen-presenting cells toward pathogen-derived TLR4 ligands by upregulating TLR4 expression and intensifies TNF-alpha, interleukin-12, and matrix metalloproteinase-9 production, all implicated in plaque destabilization. Thus, IFN-alpha-inducing pathogens, even when colonizing distant tissue sites, threaten the stability of inflamed atherosclerotic plaque.

    View details for DOI 10.1161/CIRCULATIONAHA.107.697789

    View details for Web of Science ID 000250518000008

    View details for PubMedID 17938289

  • Fractalkine mediates T cell-dependent proliferation of synovial fibroblasts in rheumatoid arthritis ARTHRITIS AND RHEUMATISM Sawai, H., Park, Y. W., He, X., Goronzy, J. J., Weyand, C. M. 2007; 56 (10): 3215-3225

    Abstract

    In rheumatoid arthritis (RA), synovial fibroblasts proliferate excessively, eventually eroding bone and cartilage. The aim of this study was to examine the mechanisms through which CD4 T cells, the dominant lymphocyte population in patients with rheumatoid synovitis, regulate synoviocyte proliferation.Fibroblast-like synoviocyte (FLS) lines were established from rheumatoid synovium. CD4 T cells from patients with RA and age-matched control subjects were cultured on FLS monolayers. FLS proliferation was quantified by cytometry, using carboxyfluorescein succinimidyl ester staining or microscopic enumeration of PKH26-stained FLS. Surface expression of the fractalkine (FKN) receptor CX(3)CR1 was monitored by fluorescence-activated cell sorting. The induction of CX(3)CR1 and its ligand FKN in FLS was quantified by real-time polymerase chain reaction.The proliferation of FLS was significantly increased in the presence of CD4 T cells from patients with RA compared with control T cells. CD4+,CD28- T cells were particularly effective in supporting FLS growth, inducing a 25-fold expansion compared with a 5-fold expansion induced by CD4+,CD28+ T cells. The growth-promoting activity of CD4+,CD28- T cells was mediated through CX(3)CR1, a chemokine receptor expressed on both T cells and FLS. Anti-CX(3)CR1 antibodies inhibited T cell production of tumor necrosis factor alpha (TNFalpha) and suppressed FLS proliferation. TNFalpha amplified the expansion of FLS by enhancing their expression of CX(3)CR1 and FKN.FKN-CX(3)CR1 receptor-ligand interactions regulate FLS growth and FLS-dependent T cell function. FLS stimulate autocrine growth by releasing FKN and triggering the activity of their own CX(3)CR1. This growth-promotion loop is amplified by TNFalpha produced by CX(3)CR1-expressing T cells upon stimulation by FKN-expressing FLS. These data assign a critical role to FKN and its receptor in fibroblast proliferation and pannus formation in RA.

    View details for DOI 10.1002/art.22919

    View details for Web of Science ID 000250264800008

    View details for PubMedID 17907166

  • Role of the T cell in the genesis of angiotensin II-induced hypertension and vascular dysfunction JOURNAL OF EXPERIMENTAL MEDICINE Guzik, T. J., Hoch, N. E., Brown, K. A., McCann, L. A., Rahman, A., Dikalov, S., Goronzy, J., Weyand, C., Harrison, D. G. 2007; 204 (10): 2449-2460

    Abstract

    Hypertension promotes atherosclerosis and is a major source of morbidity and mortality. We show that mice lacking T and B cells (RAG-1-/- mice) have blunted hypertension and do not develop abnormalities of vascular function during angiotensin II infusion or desoxycorticosterone acetate (DOCA)-salt. Adoptive transfer of T, but not B, cells restored these abnormalities. Angiotensin II is known to stimulate reactive oxygen species production via the nicotinamide adenosine dinucleotide phosphate (NADPH) oxidase in several cells, including some immune cells. Accordingly, adoptive transfer of T cells lacking the angiotensin type I receptor or a functional NADPH oxidase resulted in blunted angiotensin II-dependent hypertension and decreased aortic superoxide production. Angiotensin II increased T cell markers of activation and tissue homing in wild-type, but not NADPH oxidase-deficient, mice. Angiotensin II markedly increased T cells in the perivascular adipose tissue (periadventitial fat) and, to a lesser extent the adventitia. These cells expressed high levels of CC chemokine receptor 5 and were commonly double negative (CD3+CD4-CD8-). This infiltration was associated with an increase in intercellular adhesion molecule-1 and RANTES in the aorta. Hypertension also increased T lymphocyte production of tumor necrosis factor (TNF) alpha, and treatment with the TNFalpha antagonist etanercept prevented the hypertension and increase in vascular superoxide caused by angiotensin II. These studies identify a previously undefined role for T cells in the genesis of hypertension and support a role of inflammation in the basis of this prevalent disease. T cells might represent a novel therapeutic target for the treatment of high blood pressure.

    View details for DOI 10.1084/jem.20070657

    View details for Web of Science ID 000249870100020

    View details for PubMedID 17875676

    View details for PubMedCentralID PMC2118469

  • Unchecked CD70 expression on T cells lowers threshold for T cell activation in rheumatoid arthritis JOURNAL OF IMMUNOLOGY Lee, W., Yang, Z., Li, G., Weyand, C. A., Goronzy, J. J. 2007; 179 (4): 2609-2615

    Abstract

    Rheumatoid arthritis (RA) is characterized by premature immune aging with accumulation of degenerate T cells deficient for CD28. Gene expression profiling of CD4(+)CD28(-) and CD4(+)CD28(+) T cells to discover disease-promoting activities of CD28(-) T cells identified expression of CD70 as a most striking difference. Hence, CD70 was significantly more expressed in CD4 T cells from RA patients compared with age-matched controls (p < 0.006). The underlying mechanism was a failure to repress CD70 expression after activation-dependent induction. This defect in RA was not related to differential promoter demethylation. CD70 on bystander CD4(+)CD28(-) T cells functioned by lowering the threshold for T cell activation; admixture of CD4(+)CD28(-) T cells augmented TCR-induced responses of autologous naive CD4(+)CD28(+) T cells, particularly of low-avidity T cells. The data support a model in which CD70 expressed on T cells causes degeneracy in T cell responses and undermines tolerance mechanisms that normally control T cell autoreactivity.

    View details for Web of Science ID 000248959200067

    View details for PubMedID 17675524

    View details for PubMedCentralID PMC2832914

  • Immunopathologic aspects of rheumatoid arthritis: Who is the conductor and who plays the immunologic instrument? Symposium on Rheumatoid Arthritis - New Targeted Treatments Weyand, C. M. J RHEUMATOL PUBL CO. 2007: 9–14

    Abstract

    The inflammatory process of rheumatoid arthritis (RA) resembles a symphony orchestra playing a piece of music--not a song that anyone wants to hear, but a song nevertheless. Each cellular player has a distinct role, and all must coordinate in order to play their discordant "music" successfully. Rheumatoid synovitis consists of resident cells and invading immune cells that together arrange the inflammatory process in RA. There are 3 major types of synovitis that RA can comprise: germinal center synovitis, aggregate synovitis, and diffuse synovitis. Germinal centers are highly organized complex structures that are functionally competent. Aggregates are B cells and T cells arranged in defined follicles, yet they lack germinal center reactions. Diffuse synovitis is the least organized but can still cause significant damage. For each of these types of synovitis, the cellular players and their molecular instruments vary significantly. Differences in lymphoid microorganizations draw attention to the process of lymphoid organogenesis as a fundamental pathway of rheumatoid synovitis, a process that lends stability and sustainability to dysfunctional immune responses. This article will address how tissue-resident and invading cells, in particular T cells, B cells, dendritic cells, and synoviocytes, are brought together in different "symphonic" arrangements and how this process of lymphoid organization affects disease outcome and therapeutic options in RA.

    View details for Web of Science ID 000248275500003

  • The diagnosis of giant cell arteritis. Reviews in neurological diseases Melson, M. R., Weyand, C. M., Newman, N. J., Biousse, V. 2007; 4 (3): 128-142

    Abstract

    Giant cell arteritis (GCA) is the most common primary vasculitis in adults older than 50 years. The potential of GCA to cause bilateral, sequential vision loss makes it often a true neuro-ophthalmic emergency. Approximately one fifth of patients with GCA will present with ophthalmic complaints alone. The diagnosis of GCA requires a high index of suspicion and a systematic approach to diagnostic testing. The combination of abnormal laboratory markers of systemic inflammation and unilateral temporal artery biopsy is usually diagnostic. Additional testing with other diagnostic modalities may be required in cases in which clinical suspicion remains high despite a negative initial workup. We systematically review the diagnostic modalities used in suspected GCA patients who present with neuro-ophthalmic symptoms and signs.

    View details for PubMedID 17943065

  • Aging and T-cell diversity 1st European Congress of Aging Research in Immunology - Impact of Genomics Goronzy, J. J., Lee, W., Weyand, C. M. PERGAMON-ELSEVIER SCIENCE LTD. 2007: 400–406

    Abstract

    Naïve and memory CD4 and CD8 T cells constitute a highly dynamic system with constant homeostatic and antigen-driven proliferation, influx, and loss of T cells. Thymic activity dwindles with age and essentially ceases in the later decades of life, severely constraining the generation of new T cells. Homeostatic control mechanisms are very effective at maintaining a large and diverse subset of naïve CD4 T cells through the 7th decade of life, but eventually and abruptly fail at about the age of 75 years. In contrast, the CD8 T cell compartment is more unstable, with progressive diminution of naïve T cells and increasing loss of diversity during mid adulthood. Vaccination strategies need to aim at developing a broad repertoire of memory T cells before the critical time period when the naïve CD4 T-cell repertoire collapses. Research efforts need to aim at understanding T-cell homeostatic control mechanisms to ultimately expand the time period of repertoire stability.

    View details for DOI 10.1016/j.exger.2006.11.016

    View details for Web of Science ID 000246532900005

    View details for PubMedID 17218073

    View details for PubMedCentralID PMC2680153

  • Infliximab for maintenance of glucocorticosteroid-induced remission of giant cell arteritis - A randomized trial ANNALS OF INTERNAL MEDICINE Hoffman, G. S., Cid, M. C., Rendt-Zagar, K. E., Merkel, P. A., Weyand, C. M., Stone, J. H., Salvarani, C., Xu, W., Visvanathan, S., Rahman, M. U. 2007; 146 (9): 621-630

    Abstract

    Tumor necrosis factor-alpha is present in arteries in giant cell arteritis.To evaluate the efficacy of infliximab, an anti-tumor necrosis factor-alpha agent, in giant cell arteritis.Randomized, controlled trial.22 sites in the United States, the United Kingdom, Belgium, Italy, and Spain.44 patients with newly diagnosed giant cell arteritis that was in glucocorticosteroid-induced remission.Participants were randomly assigned in a 2:1 ratio to receive infliximab (5 mg/kg of body weight) or placebo. Sixteen patients were assigned to glucocorticosteroid plus placebo, and 28 patients to glucocorticosteroid plus infliximab.End points were measured through week 22, when an interim analysis resulted in early stopping of the planned 54-week trial. Primary end points were the number of patients who remained free of relapse through week 22 and adverse events. Secondary end points were time to first relapse, biomarkers, cumulative glucocorticosteroid dose, and the number of patients who remained relapse-free while the glucocorticosteroid dosage was tapered to 10 mg/d.Infliximab therapy did not increase the proportion of patients without relapse at week 22 compared with placebo (43% vs. 50%, respectively; difference, -7 percentage points [95% CI, -38 to 23 percentage points; P = 0.65), nor did it increase the proportion of patients whose glucocorticosteroid dosages were tapered to 10 mg/d without relapse (61% vs. 75%, respectively; difference, -14 percentage points [CI, -42 to 14 percentage points]; P = 0.31). The incidence of infection was 71% with infliximab and 56% with placebo (difference, 15 percentage points [CI, -14 to 45 percentage points]).The sample was too small to rule out modest effects of infliximab and included only patients with a new diagnosis. Only one dose of infliximab was evaluated, and the study was terminated early.This trial is too small to draw definitive conclusions, but it provides evidence that using infliximab as maintenance therapy in patients in glucocorticoid-induced remission of newly diagnosed giant cell arteritis is of no benefit and may be harmful. If infliximab has benefit, it is unlikely to be great. ClinicalTrials.gov registration number: NCT00076726.

    View details for Web of Science ID 000246070500001

    View details for PubMedID 17470830

  • Functional profile of activated dendritic cells in unstable atherosclerotic plaque BASIC RESEARCH IN CARDIOLOGY Erbel, C., Sato, K., Meyer, F. B., Kopecky, S. L., Frye, R. L., Goronzy, J. J., Weyand, C. M. 2007; 102 (2): 123-132

    Abstract

    Unstable atherosclerotic plaque typically contains an infiltrate of activated macrophages and activated T cells. This study established a functional profile of plaque-residing dendritic cells (DC) to examine whether they can function as Ag-presenting cells to facilitate in situ T-cell activation.Carotid artery plaque tissues were collected from 19 asymptomatic and 38 symptomatic patients undergoing endarterectomy. Matched samples of normal coronary artery wall, stable nonruptured plaque, and eroded unstable plaque were harvested from patients with fatal myocardial infarction. Quantitative PCR and immunohistochemistry were used to analyze the tissues for markers of DC activation (CD83, CD86, CCL19,CCL21) and correlate them with T-cell activation (IFN-gamma,TNF-alpha).Carotid artery plaques from patients with ischemic symptoms compared to asymptomatic patients were characterized by the presence of high amount of T-cells (P<0.01) and tissue production of high levels of the T-cell cytokines IFN-gamma (P=0.001) and TNF-alpha (P=0.006). Plaque tissues from patients with ischemic complications contained elevated levels of CD83 (P<0.001), a marker of DC activation, and the DC chemokines CCL19 (P=0.001) and CCL21 (P<0.02). Unstable coronary artery plaques were similarly correlated compared to carotid plaques from symptomatic patients with the accumulation of T cells (P=0.001) and the production of T cell chemokines IFN-gamma (P=0.001) and TNF-alpha (P=0.002). Immunohistochemistry confirmed the presence of CD83(+) DC in the shoulder region of unstable plaques, where they produced the T cell-attracting chemokines CCL19 and CCL21. Mapping of activated DC demonstrated close contact between mature DC and T cells expressing the activation marker CD40 ligand (CD40L).Activated and fully mature DC are represented in the inflammatory infiltrate characteristic for unstable carotid and coronary atheroma. Such DC produce chemokines, and thus can regulate the cell traffic into the lesion. Through the expression of the costimulatory ligand CD86, plaque-residing DC can augment T-cell stimulation and provide optimal stimulation conditions for T lymphocytes, resembling the microenvironment in organized lymphoid tissues.

    View details for DOI 10.1007/s00395-006-0636-x

    View details for Web of Science ID 000243802600003

    View details for PubMedID 17136419

  • Occurrence of giant cell arteritis...suddenly. Transactions of the American Ophthalmological Society Younge, B. R., Weyand, C. M., Goronzy, J. J. 2007; 105: 141-144

    Abstract

    To define the kinetics and mechanisms of frank arteritis onset in patients with giant cell arteritis.Cytokines were analyzed from tissue of a patient before and after the development of arteritis.A temporal artery biopsy specimen from a patient with giant cell arteritis showed no pathologic changes on microscopic examination, but there was evidence of early tissue activation of inflammatory markers. A specimen from the contralateral artery 12 days later had high levels of IL-18 transcripts and abundant transcripts for CCL19. Also, CD83 and IL-1 were present, confirming that the vascular dendritic cells had fully matured. This second biopsy specimen showed floridly positive giant cell arteritis on histopathologic examination.Partial activation of vascular dendritic cells is typically seen in patients with polymyalgia rheumatica in whom no inflammatory infiltrates are seen on histomorphologic examination. Dendritic cells become activated at an early stage of arteritis, beginning the pathologically evident arteritis, and are fully matured in microscopically florid arteritis.

    View details for PubMedID 18427603

    View details for PubMedCentralID PMC2258118

  • Immune-mediated mechanisms in atherosclerosis: Prevention and treatment of clinical manifestations CURRENT PHARMACEUTICAL DESIGN Niessner, A., Goronzy, J. J., Weyand, C. M. 2007; 13 (36): 3701-3710

    Abstract

    Accumulation of inflammatory cells identifies atherosclerotic plaque at risk for rupture. Typically, activated immune cells occupy the rupture-prone areas of the atherosclerotic lesion. These cells are an appealing therapeutic target for novel strategies of plaque stabilization. Biologic consequences of plaque inflammation ultimately depend not only on the cellular players populating the lesion but also on triggers of immune activation originating from within the plaque or arriving from the circulation, and immune effector mechanisms that mediate cellular damage and plaque destabilization. Recent studies have provided insights into particular immune mechanisms in the atherosclerotic plaque that contribute to plaque vulnerability. This knowledge provides the basis for potential immunomodulatory therapies in cardiovascular disease. These therapeutic approaches can be classified as (1) immunomodulatory effects of existing therapies, (2) therapies targeting inflammatory triggers, and (3) agents inhibiting specific immune mechanisms.

    View details for Web of Science ID 000253584200009

    View details for PubMedID 18220809

  • Pathogen-sensing plasmacytoid dendritic cells stimulate cytotoxic T-cell function in the atherosclerotic plaque through interferon-alpha CIRCULATION Niessner, A., Sato, K., Chaikof, E. L., Colmegna, I., Goronzy, J. J., Weyand, C. M. 2006; 114 (23): 2482-2489

    Abstract

    Unstable atherosclerotic plaque is characterized by an infiltrate of inflammatory cells. Both macrophages and T cells have been implicated in mediating the tissue injury leading to plaque rupture; however, signals regulating their activation remain unidentified. Infectious episodes have been suspected to render plaques vulnerable to rupture. We therefore explored whether plasmacytoid dendritic cells (pDCs) that specialize in sensing bacterial and viral products can regulate effector functions of plaque-residing T cells and thus connect host infection and plaque instability.pDCs were identified in 53% of carotid atheromas (n=30) in which they localized to the shoulder region and produced the potent immunoregulatory cytokine interferon (INF)-alpha. IFN-alpha transcript concentrations in atheroma tissues correlated strongly with plaque instability (P<0.0001). Plaque-residing pDCs responded to pathogen-derived motifs, CpG-containing oligodeoxynucleotides binding to toll-like receptor 9, with enhanced IFN-alpha transcription (P=0.03) and secretion (P=0.007). IFN-alpha emerged as a potent regulator of T-cell function, even in the absence of antigen recognition. Specifically, IFN-alpha induced a 10-fold increase of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on the surface of CD4 T cells (P<0.0001) and enabled them to effectively kill vascular smooth muscle cells (P=0.0003).pDCs in atherosclerotic plaque sense microbial motifs and amplify cytolytic T-cell functions, thus providing a link between host-infectious episodes and acute immune-mediated complications of atherosclerosis.

    View details for DOI 10.1161/CIRCULATIONAHA.106.642801

    View details for Web of Science ID 000243477500012

    View details for PubMedID 17116765

  • Siravastatin suppresses endotoxin-induced upregulation of toll-like receptors 4 and 2 in vivo ATHEROSCLEROSIS Niessner, A., Steiner, S., Speidl, W. S., Pleiner, J., Seidinger, D., Maurer, G., Goronzy, J. J., Weyand, C. M., Kopp, C. W., Huber, K., Wolzt, M., Wojta, J. 2006; 189 (2): 408-413

    Abstract

    In addition to lipid lowering effects, statins appear to have pleiotropic immunomodulatory properties. As they particularly affect monocyte functions, we tested the influence of statin treatment on the monocyte activating toll-like receptors (TLR) 4 and 2 in response to lipopolysaccharides (LPS) in vivo. In this double-blind, placebo-controlled study, 20 healthy, male subjects were randomized to receive either simvastatin (80 mg/day) or placebo for 4 days before intravenous LPS administration (20 IU/kg). Simvastatin did not influence the increase in TLR transcripts after LPS administration measured in mRNA isolated from whole blood by quantitative RT-PCR. In contrast, the parallel upregulation of TLR4 and TLR2 on the surface of monocytes determined by flow cytometry was attenuated by more than half after LPS challenge (P<0.02). Suppressed TLR4 and TLR2 expression was associated with diminished circulating concentrations of tumor necrosis factor-alpha and monocyte chemoattractant protein-1. In conclusion, high-dose simvastatin pretreatment blunted TLR4 and TLR2 expression on monocytes in a human endotoxemia model on a posttranscriptional level. This suppressive effect of statins on key receptors of the innate immunity which was associated with a reduction of effector cytokines reveals a potential mechanism for their beneficial effects in sepsis and cardiovascular disease.

    View details for DOI 10.1016/j.atherosclerosis.2005.12.022

    View details for Web of Science ID 000242305800022

    View details for PubMedID 16443229

  • Treatment of chronic inflammatory diseases with biologic agents: Opportunities and risks for the elderly EXPERIMENTAL GERONTOLOGY Diaz-Borjon, A., Weyand, C. M., Goronzy, J. J. 2006; 41 (12): 1250-1255

    Abstract

    The treatment armamentarium in rheumatic inflammatory diseases has drastically increased in the last years. Earlier uses of conventional disease-modifying antirheumatic drugs (DMARDs), along with the arrival of newer therapies including the so-called "biologic" agents, have provided better long-term outcomes for patients suffering from these illnesses. Biologic agents have shown efficacy for several diseases and failed in others. Due to a high prevalence of some of these diseases in the elderly population, this age group may also benefit, although treatment will have to be tailored to its special needs. In this mini review, we will discuss the use of these medications in rheumatic diseases with a significant prevalence in the elderly, their proven and potential uses, and the considerations that need to be taken into account when using them in this population.

    View details for DOI 10.1016/j.exger.2006.10.010

    View details for Web of Science ID 000243219800012

    View details for PubMedID 17125948

    View details for PubMedCentralID PMC2705938

  • Immunosuppression in atherosclerosis mobilizing the opposition within CIRCULATION Goronzy, J. J., Weyand, C. M. 2006; 114 (18): 1901-1904
  • Treatment of giant cell arteritis using induction therapy with high-dose glucocorticoids - A double-blind, placebo-controlled, randomized prospective clinical trial ARTHRITIS AND RHEUMATISM Mazlumzadeh, M., Hunder, G. G., Easley, K. A., Calamia, K. T., Matteson, E. L., Griffing, W. L., Younge, B. R., Weyand, C. M., Goronzy, J. J. 2006; 54 (10): 3310-3318

    Abstract

    Glucocorticoid (GC) therapy for giant cell arteritis (GCA) is effective but requires prolonged administration, resulting in adverse side effects. The goal of the current study was to test the hypothesis that induction treatment with high-dose pulse intravenous (IV) methylprednisolone permits a shorter course of therapy.Twenty-seven patients with biopsy-proven GCA were enrolled in a randomized, double-blind, placebo-controlled study to receive IV methylprednisolone (15 mg/kg of ideal body weight/day) or IV saline for 3 consecutive days. All patients were started on 40 mg/day prednisone and followed the same tapering schedule as long as disease activity was controlled. The numbers of patients with disease in remission after 36, 52, and 78 weeks of treatment and taking

    View details for DOI 10.1002/art.22163

    View details for Web of Science ID 000241260800032

    View details for PubMedID 17009270

  • Surgical pathology of noninfectious ascending aortitis: A study of 45 cases with emphasis on an isolated variant 93rd Annual Meeting of the United-States-and-Canadian-Academy-of-Pathology Miller, D. V., Isotalo, P. A., Weyand, C. M., Edwards, W. D., Aubry, M., Tazelaar, H. D. LIPPINCOTT WILLIAMS & WILKINS. 2006: 1150–58

    Abstract

    Aortitis is emerging as an important cause of ascending aortic aneurysm in the elderly. Its features have not been described in a surgical population.Retrospective clinicopathologic review of 45 cases of active noninfectious aortitis among 513 consecutive ascending aortic resections (1985 to 1999).Clinical data were collected from medical records. Histopathologic features were recorded during review of slides stained with hematoxylin-eosin and Verhoeff-van Gieson. Cases were categorized by predefined clinical criteria. Clinicopathologic features were compared among groups, with emphasis on unsuspected aortitis without systemic arteritis.The 2 largest groups were isolated aortitis (47%) and giant cell arteritis (31%). Other aortitis groups included Takayasu (14%), rheumatoid (4%), and unclassified (4%). Patients with isolated aortitis and giant cell arteritis were generally women (80%; mean age 73 y). All 6 with Takayasu arteritis were women (mean age 26). Although giant cell arteritis and isolated aortitis were histologically indistinguishable, their clinical courses differed substantially. Among 21 patients with isolated aortitis (2 treated with corticosteroids), only 10% later developed aortic aneurysms. In contrast, of 14 patients with giant cell arteritis (11 treated with corticosteroids), 21% subsequently developed aneurysms (P=0.09).Aortitis primarily affected women. Patients with isolated aortitis and giant cell arteritis were generally older than 50 years and, by definition, those with Takayasu arteritis were younger. In patients with isolated aortitis, outcomes were generally good, despite the absence of anti-inflammatory therapy. Accordingly, a conservative approach may be warranted for managing this subset of patients with aortitis.

    View details for Web of Science ID 000240410000011

    View details for PubMedID 16931960

  • Association of HLA-C3 and smoking with vasculitis in patients with rheumatoid arthritis ARTHRITIS AND RHEUMATISM Turesson, C., Schaid, D. J., Weyand, C. M., Jacobsson, L. T., Goronzy, J. J., Petersson, I. F., Dechant, S. A., Nyahll-Wahlin, B., Truedsson, L., Sturfelt, G., Matteson, E. L. 2006; 54 (9): 2776-2783

    Abstract

    To compare HLA-C genotypes and smoking habits in patients with vasculitis or other severe extraarticular manifestations of rheumatoid arthritis (ExRA) with those in RA patients without extraarticular disease.Patients were recruited from a large research database of patients with RA at the Mayo Clinic, from 2 Swedish cohorts of prevalent RA cases, and from a regional Swedish early RA cohort. Patients with severe ExRA (n = 159) and control patients with RA but no history of ExRA (non-ExRA controls) (n = 178) were matched for duration of RA and for clinical center. Data on smoking at RA onset, rheumatoid factor (RF) status, and antinuclear antibodies (ANAs) were extracted from the medical records. Polymerase chain reaction-based HLA-C genotyping was performed using a sequence-specific primer kit.The distribution of HLA-C alleles was significantly different between patients with RA-associated vasculitis and non-ExRA controls (P = 0.014). This was mainly due to a positive association of the HLA-C3 allele with vasculitis (allele frequency 0.411 in vasculitis patients versus 0.199 in non-ExRA controls; P < 0.001) and a decreased frequency of HLA-C7 (0.122 and 0.243, respectively; P = 0.018). The association between HLA-C3 and vasculitis was not due to linkage disequilibrium with HLA-DRB1. Smoking (P = 0.001), RF positivity (P < 0.0001), and presence of ANAs (P < 0.0001) were all associated with ExRA. HLA-C3 and smoking were both significant predictors of vasculitis in a multivariate model.Vasculitis in RA is associated with HLA-C3. Smoking is an independent predictor of vasculitis and other types of severe ExRA. Our results suggest that these variables are among the genetic and environmental factors that contribute significantly to the pathomechanisms of systemic RA.

    View details for DOI 10.1002/art.22057

    View details for Web of Science ID 000240872900008

    View details for PubMedID 16947780

  • Uncoupling of T-cell effector functions by inhibitory killer immunoglobulin-like receptors BLOOD Henel, G., Singh, K., Cui, D., Pryshchep, S., Lee, W., Weyand, C. M., Goronzy, J. J. 2006; 107 (11): 4449-4457

    Abstract

    Killer immunoglobulin-like receptors (KIRs) are a family of regulatory cell-surface molecules expressed on natural killer (NK) cells and memory T-cell subsets. Their ability to prevent the formation of an activation platform and to inhibit NK cell activation is the basis of the missing self model of NK cell function. The benefits of KIR expression for T-cell biology are unclear. We studied how KIR2DL2 regulates T-cell function. Engagement of KIR2DL2 by the ligand human leukocyte antigen (HLA)-Cw3 did not affect conjugate formation between CD4(+)KIR2DL2(+) T cells and superantigen-pulsed target cells or the development of mature immune synapses with lipid rafts. KIR2DL2 and the corresponding HLA-C ligand were initially recruited to the peripheral supramolecular activation cluster (pSMAC). Consequently, KIR2DL2 engagement did not inhibit the phosphorylation of early signaling proteins and T-cell-receptor (TCR)-mediated cytotoxicity or granule exocytosis. After 15-30 minutes, KIR2DL2 moved to the central supramolecular activation cluster (cSMAC), colocalizing with CD3. TCR synapses dissociated, and phosphorylated phospholipase C (PLC)-gamma1, Vav1, and extracellular signal-regulated kinase 1/2 (ERK1/2) were reduced 90 minutes after stimulation. Gene array studies documented that the inhibition of late signaling events by KIR2DL2 affected transcriptional gene activation. We propose that KIRs on memory T cells operate to uncouple effector functions by modifying the transcriptional profile while leaving granule exocytosis unabated.

    View details for Web of Science ID 000237877300041

    View details for PubMedID 16469873

    View details for PubMedCentralID PMC1895796

  • T cell recognition and killing of vascular smooth muscle cells in acute coronary syndrome CIRCULATION RESEARCH Pryshchep, S., Sato, K., Goronzy, J. J., Weyand, C. M. 2006; 98 (9): 1168-1176

    Abstract

    Loss of vascular smooth muscle cells (VSMCs) has been proposed to destabilize the atherosclerotic plaque and contribute to plaque rupture, superimposed thrombosis, and acute coronary syndromes (ACSs). We examined whether VSMCs are susceptible to T cell-induced apoptosis and found that CD4 T cells are highly effective in establishing cell-cell contact with VSMCs and triggering apoptotic death. Visualization of the T cell-VSMC contact zone on the single-cell level revealed that both patient-derived and control CD4 T cells reorganized their cell membrane to assemble an immunologic synapse with the VSMCs. Within 4 to 10 minutes, the membrane proximal signaling molecule ZAP-70 was recruited and phosphorylated. However, only patient-derived CD4 T cells sustained an intact immunologic synapse beyond 10 minutes and generated intracellular calcium signals. CD4 T cells that maintained a synaptic contact and appeared to be responsible for VSMC apoptosis accounted for approximately 20% of the circulating memory T cell population in ACS patients and were rare in the blood of age-matched controls. CD4 T cells from ACS patients were also hyperresponsive to T cell receptor-mediated stimulation when triggered by a superantigen and non-VSMC target cells. Lowered setting of the T cell activation threshold, attributable to excessive amplification of proximal CD3-mediated signals, may contribute to CD4 T cell-mediated killing of VSMCs and promote plaque instability.

    View details for DOI 10.1161/01.RES.0000220649.10013.5c

    View details for Web of Science ID 000237471600012

    View details for PubMedID 16601227

  • Pathomechanisms in rheumatoid arthritis - time for a string theory? JOURNAL OF CLINICAL INVESTIGATION Weyand, C. M., Goronzy, J. J. 2006; 116 (4): 869-871

    Abstract

    RA is a quintessential autoimmune disease with a growing number of cells, mediators, and pathways implicated in this tissue-injurious inflammation. Now Kuhn and colleagues have provided convincing evidence that autoantibodies reacting with citrullinated proteins, known for their sensitivity and specificity as biomarkers in RA, enhance tissue damage in collagen-induced arthritis (see the related article beginning on page 961). This study adds yet another soldier to the growing army of autoaggressive mechanisms that underlie RA. With great success researchers have dismantled the pathogenic subunits of RA, adding gene to gene, molecule to molecule, and pathway to pathway in an ever more complex scheme of dysfunction. The complexity of the emerging disease model leaves us speechless. It seems that with this wealth of data available, we need to develop a new theory for this disease. We may want to seek guidance from our colleagues in physics and mathematics who have successfully integrated their knowledge of elementary particles and the complexity of their interacting forces by formulating the string theory.

    View details for DOI 10.1172/JCI28300

    View details for Web of Science ID 000236556100008

    View details for PubMedID 16585957

    View details for PubMedCentralID PMC1421369

  • T-cell-targeted therapies in rheumatoid arthritis NATURE CLINICAL PRACTICE RHEUMATOLOGY Weyand, C. M., Goronzy, J. J. 2006; 2 (4): 201-210

    Abstract

    T cells regulate the disease process in rheumatoid arthritis (RA) on multiple levels and represent a logical choice for anti-inflammatory therapy. In the inflamed joint they promote neoangiogenesis and lymphoid organogenesis, and stimulate synoviocyte proliferation and development of bone-eroding osteoclasts. The design of T-cell-targeted therapies for RA needs to take into account the uniqueness of T-cell generation, turnover and differentiation in affected patients. Patients accumulate 'old' T cells that respond to alternate regulatory signals because of an accelerated immune aging process; any therapeutic interventions that increase the replicative stress of T cells should, therefore, be avoided. Instead, therapeutic approaches that raise the threshold for T-cell activation are more promising. As a rule, antigen-derived signals synergize with co-stimulatory signals to stimulate T cells; such co-stimulatory signals are now targeted in novel immunosuppressive therapies. An example is abatacept (soluble cytotoxic-T-lymphocyte-associated protein 4-immunoglobulin), which binds with high affinity to CD80/CD86 and effectively suppresses inflammatory activity in RA. The therapeutic benefits gained by disrupting T-cell co-stimulation indicate that the pathogenesis of RA is driven by a more generalized abnormality in T-cell activation thresholds rather than a highly selective action of arthritogenic antigens.

    View details for DOI 10.1038/ncprheum0142

    View details for Web of Science ID 000236332300005

    View details for PubMedID 16932686

  • Interleukin 12 induces T-cell recruitment into the atherosclerotic plaque CIRCULATION RESEARCH Zhang, X. Y., Niessner, A., Nakajima, T., Ma-Krupa, W., Kopecky, S. L., Frye, R. L., Goronzy, J. J., Weyand, C. M. 2006; 98 (4): 524-531

    Abstract

    CD4 T cells, through the release of cytokines as well as direct effector functions, have been implicated in promoting inflammation of the atherosclerotic plaque. Plaque-infiltrating CD4 T cells include a specialized subset of (CD4+)CD28- T cells that express a unique profile of regulatory receptors and are responsive to novel microenvironmental cues. Here we report that (CD4+)CD28- T cells, either isolated from the plaque tissue or from the blood of patients with acute coronary syndrome (ACS), spontaneously express interleukin (IL)-12 receptors, even in the absence of antigenic stimulation. (CD4+)CD28- IL-12R+ cells responded to IL-12 stimulation with the upregulation of the chemokine receptor CCR5 and the C-type lectin receptor CD161, both implicated in regulating tissue homing of effector T cells. IL-12 treatment of (CD4+)CD28- T cells enhanced their chemotaxis and transendothelial migration toward the chemokine CCL5. In vivo relevance for the role of IL-12 in regulating the recruitment of (CD4+)CD28- T cells into the atheroma was examined in human atheroma-SCID mouse chimeras. Exposure of nonstimulated (CD4+)CD28- T cells to IL-12 was sufficient to amplify T-cell accumulation within the inflamed plaque, and coadministration of anti-CCR5 antibodies blocked T-cell recruitment into the plaque. Thus, (CD4+)CD28- T cells functionally resemble NK cells, which have proinflammatory activity even in the unprimed state and respond to any IL-12-inducing host infection with a shift in tissue trafficking and accrual in inflammatory lesions.

    View details for DOI 10.1161/01.RES.0000204452.46568.57

    View details for Web of Science ID 000235728300017

    View details for PubMedID 16424368

  • Telomeres, immune aging and autoimmunity EXPERIMENTAL GERONTOLOGY Goronzy, J. J., Fujii, H., Weyand, C. M. 2006; 41 (3): 246-251

    Abstract

    Telomere length is important in constraining the replicative potential of cells; cellular systems that are dependent on cell replenishment for renewal or on cell proliferation for functionality are highly sensitive to telomeric erosion. Cell replication invariably leads to telomere loss, which, in some cellular systems, is partially compensated for by telomerase activity. In addition to this typical telomere loss, several mechanisms of sporadic telomere loss exist. Heterogeneity in age-dependent telomere loss can be a consequence of increased cellular turnover during a lifetime, accelerated telomeric DNA damage, or defects in telomere repair. The immune system is a prime example of a highly dynamic cellular system, for which telomere maintenance is pivotal. Immune competence is strictly dependent on rapid expansions of clonal T- and B-cell populations, and telomere loss may contribute to defective immune responses in the elderly. Equally interestingly, accelerated T-cell aging combined with telomeric shortening may predispose for autoimmune responses and thereby explain the increased susceptibility for chronic inflammatory diseases in the elderly.

    View details for DOI 10.1016/j.exger.2005.12.002

    View details for Web of Science ID 000236653300002

    View details for PubMedID 16427234

  • Lymphoma in rheumatoid arthritis: An immune system set up for failure ARTHRITIS AND RHEUMATISM Weyand, C. M., Goronzy, J. J., Kurtin, P. J. 2006; 54 (3): 685-689

    View details for DOI 10.1002/art.21674

    View details for Web of Science ID 000236019700001

    View details for PubMedID 16508924

  • TRAIL-expressing T cells induce apoptosis of vascular smooth muscle cells in the atherosclerotic plaque JOURNAL OF EXPERIMENTAL MEDICINE Sato, K., Niessner, A., Kopecky, S. L., Frye, R. L., Goronzy, J. J., Weyand, C. M. 2006; 203 (1): 239-250

    Abstract

    Acute coronary syndromes (ACS) are precipitated by a rupture of the atherosclerotic plaque, often at the site of T cell and macrophage infiltration. Here, we show that plaque-infiltrating CD4 T cells effectively kill vascular smooth muscle cells (VSMC). VSMCs sensitive to T cell-mediated killing express the death receptor DR5 (TNF-related apoptosis-inducing ligand [TRAIL] receptor 2), and anti-TRAIL and anti-DR5 antibodies block T cell-mediated apoptosis. CD4 T cells that express TRAIL upon stimulation are expanded in patients with ACS and more effectively induce VSMC apoptosis. Adoptive transfer of plaque-derived CD4 T cells into immunodeficient mice that are engrafted with human atherosclerotic plaque results in apoptosis of VSMCs, which was prevented by coadministration of anti-TRAIL antibody. These data identify that the death pathway is triggered by TRAIL-producing CD4 T cells as a direct mechanism of VSMC apoptosis, a process which may lead to plaque destabilization.

    View details for Web of Science ID 000235003600025

    View details for PubMedID 16418392

    View details for PubMedCentralID PMC2118078

  • Tissue trafficking patterns of effector memory CD4+T cells in rheumatoid arthritis ARTHRITIS AND RHEUMATISM Zhang, X. Y., Nakajima, T., Goronzy, J. J., Weyand, C. M. 2005; 52 (12): 3839-3849

    Abstract

    Clonal populations of CD4+,CD28- T cells accumulating in rheumatoid arthritis functionally resemble end-differentiated, nondividing, short-lived effector memory cells that reside in peripheral tissues. We undertook this study to examine the tissue niche for CD4+,CD28- T cells and the signals regulating their survival and tissue homing patterns.Chemokine receptor expression on CD4+,CD28- T cell clones and peripheral blood lymphocytes was assessed by multicolor cytometry. In vitro chemotaxis and transendothelial migration were examined in a Transwell system. In vivo tissue-homing patterns were established by adoptively transferring fluorescence-labeled T cell clones into SCID mice engrafted either with rheumatoid synovium or with human lymph nodes.CD4+,CD28- T cell clones adoptively transferred into human tissue-SCID mouse chimeras infiltrated rheumatoid synovium but preferentially homed to lymph nodes. Such T cells coexpressed the chemokine receptors CCR7, CCR5, and CXCR4 and migrated in response to both inflammatory chemokines (CCL5) and homing chemokines (CXCL12). T cell receptor crosslinking abrogated chemotactic responsiveness. In contrast, interleukin-12 stimulation induced the up-regulation of CCR5 and a shift in the in vivo homing pattern away from the lymph nodes toward the inflamed synovium.CD4+,CD28- T cells resemble both short-lived effector memory cells and long-lived central memory cells, and they find a niche both in inflamed synovium and in lymph nodes. Nonspecific cytokine stimulation, not antigen recognition, triggers the transition from the lymph node to the synovium. By maintaining CCR7 expression, these end-differentiated T cells can home to lymphoid organs, enhance their survival, support clonal expansion, and perpetuate autoreactivity.

    View details for DOI 10.1002/art.21482

    View details for Web of Science ID 000234131500021

    View details for PubMedID 16329093

  • Fractalkine receptor polymorphisms V249I and T280M as genetic risk factors for restenosis THROMBOSIS AND HAEMOSTASIS Niessner, A., Marculescu, R., Kvakan, H., Haschemi, A., Endler, G., Weyand, C. M., Maurer, G., Mannhalter, C., Wojta, J., Wagner, O., Huber, K. 2005; 94 (6): 1251-1256

    Abstract

    The chemokine fractalkine (FKN) recruits leukocytes into lesions of the arterial wall, which may lead to restenosis after stenting. FKN also regulates proliferation of smooth muscle cells, another mechanism pivotal to neointimal thickening. We assessed the hypothesis that functionally important polymorphisms of the FKN receptor CX3CR1 influence restenosis after coronary stenting. Three hundred and sixty-five patients undergoing coronary stenting were genotyped for the CX3CR1 polymorphisms V2491 and T280M. Restenosis occurred in 25% of patients, and recurrent (> 1) restenosis at the target lesion in 8%. The allele 1249 was associated with an increased risk of restenosis (adjusted odds ratio 2.4, 95% confidence interval: 1.3-4.2, P = 0.003) and recurrent restenosis (odds ratio 2.7, 95% confidence interval: 1.3-5.9, P = 0.011). Particularly, patients with 1249 lacking the allele M280 were at an elevated risk of restenosis (P = 0.006) and, accordingly, the haplotype containing 1249 but not M280 was more frequent in patients with restenosis (P = 0.001). In conclusion, the CX3CR1 1249 allele is associated with an increased risk of restenosis while the CX3CR1 M280 allele might counteract the harmful influence of 1249. These findings show the importance of the chemokine FKN and genetic variations of its receptor for restenosis after coronary stenting. Recognition of these inherited risk modifiers may help to individualize treatment of coronary stenosis.

    View details for DOI 10.1160/TH05-06-0417

    View details for Web of Science ID 000234214900023

    View details for PubMedID 16411402

  • BLyS and APRIL in rheumatoid arthritis JOURNAL OF CLINICAL INVESTIGATION Seyler, T. M., Park, Y. W., Takemura, S., Bram, R. J., Kurtin, P. J., Goronzy, J. J., Weyand, C. M. 2005; 115 (11): 3083-3092

    Abstract

    The cytokines B lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL) enhance autoimmune disease by sustaining B cell activation. In RA, B cells contribute to the formation of 3 functionally distinct types of lymphoid microarchitectures in the inflamed synovium: ectopic GCs; T cell-B cell aggregates lacking GC reactions; and unorganized, diffuse infiltrates. We examined 72 tissues representing the 3 types of synovitis for BLyS and APRIL production and for expression of APRIL/BLyS receptors. Biologic effects of BLyS and APRIL were explored by treating human synovium-SCID mouse chimeras with the APRIL and BLyS decoy receptor transmembrane activator and CAML interactor:Fc (TACI:Fc). GC+ synovitis had the highest levels of APRIL, produced exclusively by CD83+ DCs. BLyS was present in similar levels in all tissue types and derived exclusively from CD68+ macrophages. In GC+ synovitis, treatment with TACI:Fc resulted in GC destruction and marked inhibition of IFN-gamma and Ig transcription. In contrast, inhibition of APRIL and BLyS in aggregate and diffuse synovitis left Ig levels unaffected and enhanced IFN-gamma production. These differential immunomodulatory effects correlated with the presence of TACI+ T cells in aggregate and diffuse synovitis and their absence in GC+ synovitis. We propose that BLyS and APRIL regulate B cell as well as T cell function and have pro- and antiinflammatory activities in RA.

    View details for DOI 10.1172/JCI25265

    View details for Web of Science ID 000233022100018

    View details for PubMedID 16239971

    View details for PubMedCentralID PMC1257539

  • T cell development and receptor diversity during aging CURRENT OPINION IN IMMUNOLOGY Goronzy, J. J., Weyand, C. M. 2005; 17 (5): 468-475

    Abstract

    The T cell system is a complex and highly dynamic system that is amazingly robust over many decades of human life. Its functional competence is determined not only by its size but also by its diversity. Homeostatic control mechanisms have to secure sufficient T cell replenishment while preventing loss of clonal diversity. Major homeostatic challenges include profound expansion and shrinkage of T cell clonotypes upon antigenic triggering and, more importantly, age-related changes in T cell regeneration. The ability of the thymus to rebuild a diverse repertoire ceases in the fifth decade of life. Emerging data suggest that the end of the 7th decade of life defines a critical time period when T cell homeostasis is no longer guaranteed and diversity of the naïve T-cell repertoire collapses. Thus, failure of T cell homeostasis appears to result from cumulative defects of T cell generation. Elucidation of the underlying mechanisms will allow for extending this turning point to later in life; ultimately, interventions have to aim at restoring thymic function and complementary modes of T cell reconstitution.

    View details for DOI 10.1016/j.coi.2005.07.020

    View details for Web of Science ID 000231905800004

    View details for PubMedID 16098723

  • Modulation of CD28 expression with anti-tumor necrosis factor alpha therapy in rheumatoid arthritis ARTHRITIS AND RHEUMATISM Bryl, E., Vallejo, A. N., Matteson, E. L., Witkowski, J. M., Weyand, C. M., Goronzy, J. J. 2005; 52 (10): 2996-3003

    Abstract

    The immune system of patients with rheumatoid arthritis (RA) is characterized by the accumulation of CD4+ T cells deficient in CD28 expression and the up-regulation of tumor necrosis factor alpha (TNFalpha). Previous in vitro studies have shown that TNFalpha induces transcriptional silencing of the CD28 gene. Because reduced expression of CD28 in T cells compromises immunocompetence, we examined whether CD28 expression is reduced in patients with RA in vivo and whether the reduction is related to TNFalpha.Patients with RA and age-matched individuals were recruited. Peripheral blood mononuclear cells were stained for CD3, CD4, CD8, CD28, TNF receptor I (TNFRI), and TNFRII, and analyzed by quantitative flow cytometry. The number of CD28 and TNFR molecules was monitored in a subgroup of patients with RA undergoing treatment with anti-TNFalpha.In addition to higher frequencies of CD28null T cells, patients with RA had significantly reduced numbers of CD28 and TNFRI molecules on CD4+,CD28+ T cells. Normal expression could be restored in vitro by overnight culture, suggesting that CD28 in patients was modulated by exogenous factors. In contrast, treatment with TNFalpha in vitro resulted in further down-regulation. CD28 expression was normalized in patients undergoing TNFalpha-neutralizing therapy.Overproduction of TNFalpha in RA induces a global down-regulation of CD28 in CD4+ T cells and may cause reduced sensitivity to costimulatory signals in T cell responses.

    View details for Web of Science ID 000232548800006

    View details for PubMedID 16200579

  • Lipopolysaccharide, CD14 and Toll-like receptors: an emerging link between innate immunity and atherosclerotic disease. Future cardiology Shimada, K., Daida, H., Ma-Krupa, W., Goronzy, J. J., Weyand, C. M. 2005; 1 (5): 657-674

    Abstract

    Atherosclerosis and its clinical complications are now understood to be an inflammatory syndrome in which an ongoing systemic inflammatory response is combined with the accumulation of immune cells in the atherosclerotic plaque. Both arms of the immune system, innate and adaptive, have been implicated in contributing to essentially all stages of atherosclerosis, from initiation to progression and, ultimately, atherothrombotic complications. Innate immunity is the first line of defense against invading microorganisms. The recognition units of the innate immune system are designed to respond to molecular patterns shared by a variety of infectious microorganisms, such as bacterial lipopolysaccharide. Numerous basic and clinical studies have provided evidence that responsiveness to lipopolysaccharide may be correlated to the risk of atherosclerotic disease. The molecular basis of this connection appears to lie in Toll-like receptors that are expressed on cells of the innate immune system, bind to lipopolysaccharide, and thus determine the strength of antibacterial immune responses in the host. Variations in the function of Toll-like receptors and their signaling pathways are now suspected to play a critical role in determining the risk of atherosclerosis. This review summarizes recent research advances exploring the role of innate immunity, particularly lipopolysaccharide, CD14 and Toll-like receptors, in the initiation and development of atherosclerotic disease.

    View details for DOI 10.2217/14796678.1.5.657

    View details for PubMedID 19804106

  • Distinct transcriptional control mechanisms of killer immunoglobulin-like receptors in natural killer (NK) and in T cells JOURNAL OF BIOLOGICAL CHEMISTRY Xu, J., Vallejo, A. N., Jiang, Y., Weyand, C. M., Goronzy, J. J. 2005; 280 (25): 24277-24285

    Abstract

    Killer immunoglobulin-like receptors (KIR) are expressed by natural killer (NK) cells and by subsets of CD4+ and CD8+ T cells, which are therefore thought to be subject to similar regulatory mechanisms. Here, we show that the transcriptional machinery to express KIR is limited to NK and T cells; however, the KIR transcriptional control differs between these two types of lymphocytes. T cells selectively express transcriptional activators binding to positions -52 to -61 of the KIR promoter, whereas an AML site around position-98 is relevant for transcription in NK cells. Although KIR expression is restricted to subsets of memory T cells, our studies demonstrate that transcriptional activators for KIRs are not acquired during T cell differentiation but are already present in naïve T cells, suggesting a basic role of KIRs in T cell biology. We suggest that the regulated expression of KIRs in T cells profoundly influences peripheral tolerance and antigen-specific immune responses.

    View details for DOI 10.1074/jbc.M500727200

    View details for Web of Science ID 000229880000105

    View details for PubMedID 15863493

  • B cells as a therapeutic target in autoimmune diseases EXPERT OPINION ON THERAPEUTIC TARGETS Park, Y. W., Pryshchep, S., Seyler, T. M., Goronzy, J. J., Weyand, C. M. 2005; 9 (3): 431-445

    Abstract

    Historically, the pathogenic role of B cells in autoimmune disease has been attributed to the formation of autoantibodies which, as soluble immunoglobulins or immunocomplexes, can trigger cellular damage and initiate the inflammatory cascade. Recent results from clinical trials applying B cell-directed therapeutics in rheumatoid arthritis and systemic lupus erythematosus have challenged such traditional views and encouraged novel ideas about the disease involvement of B cells. Suppression of disease activity, often disconnected from effects on autoantibody titers, has supported the notion that B cells may promote autoimmune disease by serving as antigen-presenting cells that sustain T cell activation. Likewise, B cells have been implicated in supporting the process of ectopic lymphoid neogenesis, a mechanism that stabilises pathogenic immune responses in target tissues and thus contributes to disease chronicity. As a general rule, clinical effects of B cell-directed therapeutics have often been unanticipated and unpredicted by experimental models, emphasis-ing the need to explore and verify disease principles in the patient.

    View details for DOI 10.1517/14728222.9.3.431

    View details for Web of Science ID 000229916000002

    View details for PubMedID 15948665

  • B cells in rheumatoid synovitis ARTHRITIS RESEARCH & THERAPY Weyand, C. M., Seyler, T., Goronzy, J. J. 2005; 7: S9-S12

    Abstract

    In rheumatoid arthritis, T cells, B cells, macrophages, and dendritic cells invade the synovial membranes, establishing complex microstructures that promote inflammatory/tissue destructive lesions. B cell involvement has been considered to be limited to autoantibody production. However, recent studies suggest that B cells support rheumatoid disease through other mechanisms. A critical element of rheumatoid synovitis is the process of ectopic lymphoid neogenesis, with highly efficient lymphoid architectures established in a nonlymphoid tissue site. Rheumatoid synovitis recapitulates the pathways of lymph node formation, and B cells play a key role in this process. Furthermore, studies of rheumatoid lesions implanted in immunodeficient mice suggest that T cell activation in synovitis is B cell dependent, indicating the role played by B cells in presenting antigens and providing survival signals.

    View details for DOI 10.1186/ar1737

    View details for Web of Science ID 000231215500003

    View details for PubMedID 15960820

    View details for PubMedCentralID PMC2833971

  • The influence of age on T cell generation and TCR diversity JOURNAL OF IMMUNOLOGY Naylor, K., Li, G. J., Vallejo, A. N., Lee, W. W., Koetz, K., Bryl, E., Witkowski, J., Fulbright, J., Weyand, C. M., Goronzy, J. J. 2005; 174 (11): 7446-7452

    Abstract

    The ability to mount protective immune responses depends on the diversity of T cells. T cell diversity may be compromised by the declining thymic output of new T cells. The aging process imposes a threat to diversity, because thymic function deteriorates. In this study we have examined the relationship between thymic production, homeostatic T cell proliferation and TCR beta-chain diversity in young (approximately 25 years), middle-aged ( approximately 60 years), and elderly adults (approximately 75 years). TCR excision circles (TREC) as a marker of thymic output exponentially decreased by >95% between 25 and 60 years of age. The frequency of Ki67(+) cycling CD4 T cells remained steady, and surprisingly, the diversity of the naive CD4 T cell repertoire was maintained at approximately 2 x 10(7) different TCR beta-chains. After the age of 70 years, TRECs only slightly declined, but homeostatic proliferation doubled. The diversity of the T cell pool drastically contracted to 200,000 TCR beta-chains. Also, the phenotypic distinction between naive and memory CD4 T cells became fuzzy. The collapse in CD4 T cell diversity during the seventh and eighth decades indicates substantial T cell loss and implies that therapeutic measures to improve vaccine responses will have to include strategies for T cell replenishment.

    View details for Web of Science ID 000229298400109

    View details for PubMedID 15905594

  • Cell-based immunotherapy with suppressor CD8(+) T cells in rheumatoid arthritis JOURNAL OF IMMUNOLOGY Davila, E., Kang, Y. M., Park, Y. W., Sawai, H., He, X. W., Pryshchep, S., Goronzy, J. J., Weyand, C. M. 2005; 174 (11): 7292-7301

    Abstract

    The chronic persistence of rheumatoid synovitis, an inflammation driven by activated T cells, macrophages, and fibroblasts causing irreversible joint damage, suggests a failure in physiologic mechanisms that down-regulate and terminate chronic immune responses. In vitro CD8(+)CD28(-)CD56(+) T cells tolerize APCs, prevent the priming of naive CD4(+) T cells, and suppress memory CD4(+) T cell responses. Therefore, we generated CD8(+)CD28(-)CD56(+) T cell clones from synovial tissues, expanded them in vitro, and adoptively transferred them into NOD-SCID mice engrafted with synovial tissues from patients with rheumatoid arthritis. Adoptively transferred CD8(+)CD28(-)CD56(+) T cells displayed strong anti-inflammatory activity. They inhibited production of IFN-gamma, TNF-alpha, and chemokines in autologous and HLA class I-matched heterologous synovitis. Down-regulation of costimulatory ligands CD80 and CD86 on synovial fibroblasts was identified as one mechanism of immunosuppression. We propose that rheumatoid synovitis can be suppressed by cell-based immunotherapy with immunoregulatory CD8(+) T cells.

    View details for Web of Science ID 000229298400091

    View details for PubMedID 15905576

  • Opposite effects of CX3CRI receptor polymorphisms V2491 and T280M on the development of acute coronary syndrome - A possible implication of fractalkine in inflammatory activation THROMBOSIS AND HAEMOSTASIS Niessner, A., Marculescu, R., Haschemi, A., Endler, G., Zorn, G., Weyand, C. M., Maurer, G., Mannhalter, C., Wojta, J., Wagner, O., Huber, K. 2005; 93 (5): 949-954

    Abstract

    Several lines of evidence suggest that the chemokine fractalkine (FKN) and its receptor CX3CR1 contribute to the accumulation of leukocytes in the atherosclerotic plaque. The M280 allele of the CX3CR1T280M polymorphism modulates leukocyte recruitment and is associated with lower prevalence of cardiovascular disease. The influence of V249I, another CX3CR1 polymorphism, is discussed controversially. We investigated the association of the alleles M280 and I249 of CX3CR1 with coronary artery disease (CAD) and with acute coronary syndrome (ACS). Additionally, we assessed their association with the soluble ligand FKN and inflammatory activation measured by high sensitivity C-reactive protein (hsCRP). The genotypes of the V249I and T280M polymorphisms were determined in 1152 patients with suspected CAD.720 (62.5%) individuals showed significant CAD with an ACS prevalence of 59.3%. Using multivariate regression, we found a harmful influence of I249 (adjusted OR=1.8, P<0.03) and a protective effect of M280 (adjusted OR=0.6, P<0.04) on the occurrence of ACS in patients with CAD. Correspondingly, patients with I249 but without M280 (17%) were at elevated risk of ACS (OR=1.6, P<0.04). During ACS these patients (carrying only I249) had significantly higher circulating concentrations of FKN and high sensitivity C-reactive protein (1.9- and 1.6-fold). We found no association of the I249 or the M280 allele with the occurrence of CAD. In conclusion, I249 and M280 have opposite effects on the occurrence of ACS. The presence of I249 not "balanced" by M280 confers an elevated risk of ACS. A FKN-mediated enhanced inflammatory activation might explain this increased risk.

    View details for DOI 10.1160/TH04-11-0735

    View details for Web of Science ID 000229087200025

    View details for PubMedID 15886814

  • T cell costimulation by fractalkine-expressing synoviocytes in rheumatoid arthritis ARTHRITIS AND RHEUMATISM Sawai, H., Park, Y. W., Roberson, J., Imai, T., Goronzy, J. J., Weyand, C. A. 2005; 52 (5): 1392-1401

    Abstract

    Patients with rheumatoid arthritis (RA) accumulate prematurely aged T cells that have acquired a new profile of regulatory receptors. Many of the de novo-expressed receptors are typically found on natural killer cells, including CX(3)CR1, the receptor for the chemokine fractalkine (FKN). This study explored whether interactions between CX(3)CR1 and FKN are relevant for T cell functions in rheumatoid synovitis.FKN expression was examined by real-time polymerase chain reaction and immunohistochemistry. CX(3)CR1 expression on peripheral blood T cells was analyzed by flow cytometry. T cell activation was quantified by determining proliferative responses, interferon-gamma (IFNgamma) secretion, and granule release. Fibroblast-like synoviocyte (FLS)/T cell adhesion was measured by the retention of 5-carboxyfluorescein diacetate succinimidyl ester-labeled T cells on FLS monolayers.FKN was expressed on cultured synovial fibroblasts and hyperplastic synoviocytes in the rheumatoid tissue. Among CD4+ T cells, only senescent CD28- T cells were positive for CX(3)CR1 (P < 0.001). Such CD4+,CD28-,CX(3)CR1+ T cells strongly adhered to FLS, with soluble FKN blocking the interaction. FKN expressed on FLS costimulated T cell-activating signals and amplified proliferation, IFNgamma production, and expulsion of cytoplasmic granules.Senescent CD4+ T cells that accumulate in rheumatoid arthritis aberrantly express CX(3)CR1. FKN, which is membrane-anchored on synoviocytes, enhances CD4+ T cell adhesion, provides survival signals, and costimulates the production of proinflammatory cytokines as well as the release of granules. By virtue of their altered receptor profile, senescent CD4+ T cells receive strong stimulatory signals from nonprofessional antigen-presenting cells in the synovial microenvironment.

    View details for DOI 10.1002/art.21140

    View details for Web of Science ID 000229004600007

    View details for PubMedID 15880821

  • Toll-like receptors in giant cell arteritis 12th Annual International Congress of Immunology/4th Annual Conference of the Federation-of-Clinical-Immunology-Societies (FOCIS) Wei, M. K., Kwan, M., Goronzy, J. J., Weyand, C. M. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2005: 38–46

    Abstract

    Giant cell arteritis, a primary vasculitis of medium-sized and large arteries, causes vessel occlusion through fast and concentric intimal hyperplasia. Contextual parameters, especially the topography of the arterial wall, have emerged as critical pathogenic elements. Experimental data support the concept that the disease is initiated in the most outer layer of the arterial wall, the adventitia. CD4 T cells are recruited to the adventitia, undergo local activation and subsequently orchestrate macrophage differentiation. T cells and macrophages infiltrate into all wall layers and acquire different effector functions dependent on cues in their immediate microenvironment. The end result is myofibroblastic proliferation, luminal stenosis, and tissue ischemia. Adaptive immune responses in the adventitia are triggered by a population of indigenous dendritic cells (DC) placed at the adventitia-media junction. These arterial DCs have a unique surface receptor profile, including a series of Toll-like receptors (TLR). Responsiveness of such arterial DCs to blood-borne stimuli has been studied in human arteries engrafted into immunodeficient mice. Ligands of TLR4 are able to start maturation of adventitial DCs which fail to leave the peripheral tissue site. Instead, these adventitial DCs produce chemokines, recruit T cells, and support their local activation. These data identify tissue-residing DCs as gatekeepers in vasculitis and support the model that TLR ligands function as instigators of vessel wall inflammation.

    View details for DOI 10.1016/j.clin.2005.02.009

    View details for Web of Science ID 000229096800007

    View details for PubMedID 15870019

  • Rheumatoid arthritis IMMUNOLOGICAL REVIEWS Goronzy, J. J., Weyand, C. M. 2005; 204: 55-73

    Abstract

    Therapeutic efficacy of depleting B cells or blocking T-cell costimulation in rheumatoid arthritis (RA) has confirmed the critical pathogenic role of adaptive immune responses. Yet, RA preferentially affects elderly individuals, in whom adaptive immunity to exogenous antigens begins to fail. Here, we propose that senescence of the immune system is a risk factor for RA, with chronic inflammation resulting from the accumulation of degenerate T cells that have a low threshold for activation and utilize a spectrum of novel receptors to respond to microenvironmental cues. The process of immunosenescence is accelerated in RA and precedes the onset of disease, the acceleration, in part, being conferred by the HLA-DR4 haplotype. Naive CD4(+) T cells in RA are contracted in diversity and restricted in clonal burst. Senescence of effector CD4(+) T cells is associated with the loss of CD28 and the de novo expression of KIR2DS2, NKG2D, and CX(3)CR1, all of which function as costimulatory molecules and reduce the threshold for T-cell activation. The synovial microenvironment promotes chronic persistent immune responses by facilitating ectopic lymphoid neogenesis, such as the formation of aberrant germinal centers. With the propensity to develop complex lymphoid architectures and to provide optimal activation conditions for senescent CD4(+) T cells, the synovium becomes a natural target for pathogenic immune responses in prematurely aged individuals.

    View details for Web of Science ID 000227672500005

    View details for PubMedID 15790350

  • Vascular dendritic cells in giant cell arteritis Conference on Human Immunology Weyand, C. M., Ma-Krupa, W., Pryshchep, O., Groschel, S., Bernardino, R., Goronzy, J. J. NEW YORK ACAD SCIENCES. 2005: 195–208

    Abstract

    Giant cell arteritis (GCA) is a granulomatous vasculitis that selectively targets medium-sized and large arteries, especially the cranial branches of the aorta. The inflammatory activity of vascular lesions is driven by adaptive immune responses, with CD4 T cells undergoing clonal expansion in the vessel wall and releasing interferon gamma. Recent studies have described a distinctive population of dendritic cells (DCs) localized at the adventitia-media border of normal medium-sized arteries that appear to play a critical role in the initiation of vasculitis. Immune effector functions of this DC population are being examined in human artery-severe combined immunodeficient (SCID) mouse chimeras. In their constitutive form, CD11c+ fascin+ adventitial DCs are not recognized by alloreactive T cells. Triggering with Toll-like receptor (TLR) ligands is sufficient to break this state of tolerance and initiate DC activation, T-cell recruitment, T-cell activation, and T-cell retention in the arterial wall. Systemic administration of ligands for TLR2 or -4 in human artery-SCID chimeras drives differentiation of adventitial DCs into chemokine-producing effector cells with high-level expression of both CD83 and CD86 and mediates T-cell regulatory function through release of interleukin 18. In established vasculitis, fully matured DCs retain antigen-presenting function; antibody-mediated DC depletion disrupts T-cell and macrophage activation and has marked anti-inflammatory effects. We conclude that adventitial DCs, an indigenous cell population of the arterial wall, are responsive to pathogen-derived macromolecules and have gatekeeper function in regulating T-cell recruitment and retention to the arterial adventitia. A switch of adventitial DCs from being nonstimulatory to T-cell activating emerges as a critical event in the initiation of vasculitis.

    View details for DOI 10.1196/annals.1358.023

    View details for Web of Science ID 000236473100020

    View details for PubMedID 16461802

  • Costimulatory pathways in rheumatoid synovitis and T-cell senescence Conference on Human Immunology Goronzy, J. J., Henel, G., Sawai, H., Singh, K., Lee, E. B., Pryshchep, S., Weyand, C. M. NEW YORK ACAD SCIENCES. 2005: 182–194

    Abstract

    The pathogenesis of rheumatoid arthritis (RA) is determined by a complex interaction of genetic and environmental factors. Of all risk factors, age has the largest impact. RA occurs most often during the postmenopausal period of life, with incidence rates peaking in the eighth decade. While age is generally accepted as an etiologic factor for failure of immunocompetence, much less is understood about the role of T-cell senescence in autoimmunity. We have hypothesized that senescent T cells are particularly prone to be activated in specialized microenvironments, such as the synovial membrane. CD4 T cells in the senescence program were identified by the loss of CD28. Gene expression profiling documented that CD28- T cells have acquired a spectrum of regulatory receptors that are usually seen only on NK cells. Such regulatory receptors include stimulatory and inhibitory members of the killer immunoglobulin-like receptor (KIR) family, the stimulatory c-type lectin receptor NKG2D, and CX3CR1, the receptor for the chemokine fractalkine. Synovial fibroblasts express the relevant ligands, thus providing stimulatory signals to tissue-infiltrating T cells. The signaling pathways of these regulatory receptors are complex and dependent on the individual T cells, some of which express important adapter molecules such as DAP10 and DAP12. Inhibitory KIRs on T cells are often only partially functional. Our data suggest that, by virtue of altered receptor profiles, conventional tolerance mechanisms can be evaded in the aging host. By acquiring a new set of regulatory receptors, senescent CD4 T cells become responsive to novel environmental cues and find ideal stimulatory conditions in the synovial microenvironment.

    View details for DOI 10.1196/annals.1358.022

    View details for Web of Science ID 000236473100019

    View details for PubMedID 16461801

  • Increased CD4+ T cell infiltrates in rheumatoid arthritis-associated interstitial pneumonitis compared with idiopathic interstitial pneumonitis ARTHRITIS AND RHEUMATISM Turesson, C., Matteson, E. L., Colby, T. V., Vuk-Pavlovic, Z., Vassallo, R., Weyand, C. M., Tazelaar, H. D., Limper, A. H. 2005; 52 (1): 73-79

    Abstract

    To study lymphocyte markers in rheumatoid arthritis (RA)-associated interstitial pneumonitis (IP) compared with idiopathic IP.Paraffin-embedded lung biopsy specimens from patients with RA (n = 15) and from those without RA (n = 16), all of whom had a diagnosis of either nonspecific IP or usual IP, were studied. Tissue sections from each patient were reviewed by a pathologist, who was blinded to the clinical data. Age and pulmonary function test results were similar in RA and non-RA patients. After high-temperature antigen unmasking, sections were incubated with mouse monoclonal antibodies directed against CD3, CD4, CD8, CD16, and CD20. All slides were coded, and digital images (100x magnification) of the entire tissue area were obtained. Staining was quantified using computer-assisted image analysis.Staining for CD4 was more prominent in patients with RA than in the non-RA comparison group (median 9.3 cells/mm(2), interquartile range [IQR] 5.5-27.3 versus 0.6 cells/mm(2), IQR 0.2-1.9; P = 0.002). CD4+ cell counts were increased in RA patients with nonspecific IP as well as in RA patients with usual IP, with no major difference between these groups. Results were similar for quantification of CD3 (P = 0.012). There was a less striking trend toward more CD8+ cells in RA patients (P = 0.27 versus those with non-RA lung disease).IP lesions in patients with RA are characterized by an increased number of CD4+ cells, as compared with that in patients with idiopathic IP. This finding suggests that CD4+ T cells are critical for the development of pulmonary manifestations in RA, and may have implications for the treatment of RA-associated lung disease.

    View details for DOI 10.1002/art.20765

    View details for Web of Science ID 000226507700010

    View details for PubMedID 15641082

  • The impact of HLA-DRB1 genes on extra-articular disease manifestations in rheumatoid arthritis ARTHRITIS RESEARCH & THERAPY Turesson, C., Schaid, D. J., Weyand, C. M., Jacobsson, L. T., Goronzy, J. J., Petersson, I. F., Sturfelt, G., Nyhall-Wahlin, B. M., Truedsson, L., Dechant, S. A., Matteson, E. L. 2005; 7 (6): R1386-R1393

    Abstract

    The objective of this study was to examine HLA-DRB1 and HLA-DQB1 genotypes in patients with severe extra-articular rheumatoid arthritis (ExRA) and to compare them with the genotypes of rheumatoid arthritis (RA) patients without extra-articular manifestations. Patients with severe ExRA were recruited from a large research database of patients with RA, from two cohorts of prevalent RA cases, and from a regional multicenter early RA cohort. Cases with ExRA manifestations (n = 159) were classified according to predefined criteria. Controls (n = 178) with RA but no ExRA were selected from the same sources. Cases and controls were matched for duration of RA and for clinical center. PCR based HLA-DRB1 and HLA-DQB1 genotyping was performed using the Biotest SSP kit, with additional sequencing in order to distinguish DRB1*04 subtypes. Associations between alleles and disease phenotypes were tested using multiple simulations of random distributions of alleles. There was no difference in global distribution of HLA-DRB1 and HLA-DQB1 alleles between patients with ExRA and controls. DRB1*0401 (P = 0.003) and 0401/0401 homozygosity (P = 0.002) were more frequent in Felty's syndrome than in controls. The presence of two HLA-DRB1*04 alleles encoding the shared epitope (SE) was associated with ExRA (overall odds ratio 1.79, 95% confidence interval 1.04-3.08) and with rheumatoid vasculitis (odds ratio 2.44, 95% confidence interval 1.22-4.89). In this large sample of patients with ExRA, Felty's syndrome was the only manifestation that was clearly associated with HLA-DRB1*0401. Other ExRA manifestations were not associated with individual alleles but with DRB1*04 SE double dose genotypes. This confirms that SE genes contribute to RA disease severity and ExRA. Other genetic and environmental factors may have a more specific impact on individual ExRA manifestations.

    View details for DOI 10.1186/ar1837

    View details for Web of Science ID 000234272000032

    View details for PubMedID 16277691

    View details for PubMedCentralID PMC1297586

  • Rheumatoid arthritis is an independent risk factor for multi-vessel coronary artery disease: a case control study ARTHRITIS RESEARCH & THERAPY Warrington, K. J., Kent, P. D., Frye, R. L., Lymp, J. F., Kopecky, S. L., Goronzy, J. J., Weyand, C. M. 2005; 7 (5): R984-R991

    Abstract

    The risk for cardiovascular (CV) disease is increased in rheumatoid arthritis (RA) but data on the burden of coronary atherosclerosis in patients with RA are lacking. We conducted a retrospective case-control study of Olmsted County (MN, USA) residents with RA and new-onset coronary artery disease (CAD) (n = 75) in comparison with age-and sex-matched controls with newly diagnosed CAD (n = 128). Angiographic scores of the first coronary angiogram and data on CV risk factors and CV events on follow-up were obtained by chart abstraction. Patients with RA were more likely to have multi-vessel coronary involvement at first coronary angiogram compared with controls (P = 0.002). Risk factors for CAD including diabetes, hypertension, hyperlipidemia, and smoking history were not significantly different in the two cohorts. RA remained a significant risk factor for multi-vessel disease after adjustment for age, sex and history of hyperlipidemia. The overall rate of CV events was similar in RA patients and controls; however, there was a trend for increased CV death in patients with RA. In a nested cohort of patients with RA and CAD (n = 27), we measured levels of pro-inflammatory CD4+CD28null T cells by flow cytometry. These T cells have been previously implicated in the pathogenesis of CAD and RA. Indeed, CD4+CD28null T cells were significantly higher in patients with CAD and co-existent RA than in controls with stable angina (P = 0.001) and reached levels found in patients with acute coronary syndromes. Patients with RA are at increased risk for multi-vessel CAD, although the risk of CV events was not increased in our study population. Expansion of CD4+CD28null T cells in these patients may contribute to the progression of atherosclerosis.

    View details for DOI 10.1186/ar1775

    View details for Web of Science ID 000231020200008

    View details for PubMedID 16207339

    View details for PubMedCentralID PMC1257428

  • Thrombospondin 2 functions as an endogenous regulator of angiogenesis and inflammation in rheumatoid arthritis AMERICAN JOURNAL OF PATHOLOGY Park, Y. W., Kang, Y. M., Butterfield, J., Detmar, M., Goronzy, J. J., Weyand, C. M. 2004; 165 (6): 2087-2098

    Abstract

    Thrombospondin 2 (TSP2), a matricellular protein with a primary role in modulating cell-matrix interactions, has been implicated in tissue repair and foreign body responses. Here we show that TSP2 has regulatory function in the chronic inflammatory lesions of rheumatoid arthritis. Tissue TSP2, produced by synovial fibroblasts, endothelial cells, and macrophages correlated not only with the intensity of angiogenesis but also with the architecture of lymphoid infiltrates. Synovial tissues with diffuse inflammatory infiltrates had high levels of TSP2, whereas synovial tissues with ectopic germinal center reactions and T cell-B cell aggregates produced low levels. Cell-based gene therapy with TSP2 was used to examine the in vivo effects of the matrix protein on neoangiogenesis and lymphoid organization. Human synovium-severe combined immunodeficiency (SCID) mouse chimeras were treated with TSP2-transfected fibroblasts deposited into the peritoneum. Overexpression of TSP2 led to the accumulation of TSP2 protein in the inflamed synovium and resulted in a prompt inhibition of lesional vascularization. Beside its anti-angiogenic activity, TSP2 also suppressed the production of the proinflammatory mediators, interferon-gamma and tumor necrosis factor-alpha, and induced the depletion of tissue-residing T cells. We propose that TSP2 is an endogenous regulator of angiogenesis and autoimmune inflammation in the synovium and represents a protective mechanism preventing ectopic lympho-organogenesis and persistent inflammation in this tissue site.

    View details for Web of Science ID 000225381100023

    View details for PubMedID 15579451

    View details for PubMedCentralID PMC1618704

  • Genetics of rheumatoid arthritis: Is there a pattern predicting extraarticular manifestations? ARTHRITIS & RHEUMATISM-ARTHRITIS CARE & RESEARCH Turesson, C., Weyand, C. M., Matteson, E. L. 2004; 51 (5): 853-863

    View details for DOI 10.1002/art.20693

    View details for Web of Science ID 000224502600022

    View details for PubMedID 15478157

  • Stimulatory killer Ig-like receptors modulate T cell activation through DAP12-dependent and DAP12-independent mechanisms JOURNAL OF IMMUNOLOGY Snyder, M. R., Nakajima, T., Leibson, P. J., Weyand, C. M., Goronzy, J. J. 2004; 173 (6): 3725-3731

    Abstract

    Stimulatory killer Ig-like receptors (KIRs) are expressed by various lymphocytes, including NK cells and subsets of T cells. In NK cells, KIRs associate with the adapter molecule KARAP/DAP12, which confers the ability to function as an independent activation unit. The function of KIRs and killer cell activating receptor-associated protein (KARAP)/DAP12 in T cells is unclear. By flow cytometry, we demonstrated that CD4+CD28null T cells heterogeneously express KIRs and/or KARAP/DAP12. In clones that lacked expression of KARAP/DAP12, the stimulatory KIR KIR2DS2 signaled through the JNK pathway, but did not activate the ERK pathway. However, in the presence of KARAP/DAP12, stimulation through KIR2DS2 led to phosphorylation of both JNK and ERK. Transfection experiments confirmed that KIR2DS2-mediated ERK phosphorylation was dependent on KARAP/DAP12. The differential signaling of KIR2DS2 through association with alternative adapter molecules resulted in differential regulation of cellular activity. In clones that lacked expression of KARAP/DAP12, stimulation of KIR2DS2 did not induce cytotoxicity. However, KIR2DS2 did augment suboptimal TCR stimulation, leading to enhanced IFN-gamma production. In clones that expressed KARAP/DAP12, KIR2DS2 directly activated both cytotoxicity and IFN-gamma production without the need for TCR-derived signals. The function of stimulatory KIRs in T cells is determined by the expression of the appropriate adapter molecule. Expression of KARAP/DAP12 is sufficient to convert a costimulatory KIR into a stimulatory molecule. These differing functions mediated by alternative signaling pathways have implications for the pathogenesis of diseases such as rheumatoid arthritis and acute coronary syndromes, in which aberrant expression of KIRs on T cells is frequently observed.

    View details for Web of Science ID 000223878000019

    View details for PubMedID 15356118

  • Stem cell aging and autoimmunity in rheumatoid arthritis TRENDS IN MOLECULAR MEDICINE Weyand, C. M., Goronzy, J. R. 2004; 10 (9): 426-433

    View details for DOI 10.1016/j.molmed.2004.07.006

    View details for Web of Science ID 000224235200003

    View details for PubMedID 15350894

  • VEGF gene polymorphisms and susceptibility to rheumatoid arthritis RHEUMATOLOGY Han, S. W., Kim, G. W., Seo, J. S., Kim, S. J., Sa, K. H., Park, J. Y., Lee, J., Kim, S. Y., Goronzy, J. J., Weyand, C. M., Kang, Y. M. 2004; 43 (9): 1173-1177

    Abstract

    To investigate polymorphisms of the VEGF gene in patients with rheumatoid arthritis (RA), their relationship to clinical features and the radiographic progression of joint disease.One hundred and forty patients with RA and 149 healthy unrelated controls were recruited. We examined four polymorphisms of the VEGF gene which are reported to be associated with production of vascular endothelial growth factor (VEGF), using polymerase chain reaction (PCR) restriction fragment length polymorphism assay and amplification refractory mutation system (ARMS) PCR. Haplotypes were predicted by Bayesian algorithm using the Phase program.All four polymorphisms were in Hardy-Weinberg equilibrium in both patients and controls. The frequency of the 936 T allele, which has been associated with lower production of VEGF, was significantly increased in RA patients compared with controls (22.7 vs 13.4%, P = 0.002). The frequencies of two haplotypes (CGCT and AAGT) which were predicted using the Phase program were significantly increased in RA patients compared with controls [33 vs 14%, odds ratio (OR) 2.636, 95% confidence interval (CI) 1.38-5.04 for CGCT; 17 vs 6%, OR 3.08, 95% CI 1.20-7.92 for AAGT]. The carriers of the susceptible haplotypes in RA patients had a younger age at disease onset but did not show a difference in the progression rate of radiographic joint destruction.Our data suggest that the VEGF gene may play a role in the development of RA

    View details for DOI 10.1093/rheumatology/keh281

    View details for Web of Science ID 000223588600017

    View details for PubMedID 15213335

  • Tumor necrosis factor-alpha and CD80 modulate CD28 expression through a similar mechanism of T-cell receptor-independent inhibition of transcription JOURNAL OF BIOLOGICAL CHEMISTRY LEWIS, D. E., Merched-Sauvage, M., Goronzy, J. J., Weyand, C. M., Vallejo, A. N. 2004; 279 (28): 29130-29138

    Abstract

    Replicative senescence of human T cells is characterized by the loss of CD28 expression, exemplified by the clonal expansion of CD28(null) T cells during repeated stimulation in vitro as well as in chronic inflammatory and infectious diseases and in the normal course of aging. Because CD28 is the major costimulatory receptor for the induction of T cell-mediated immunity, the mechanism(s) underlying CD28 loss is of paramount interest. Current models of replicative senescence involve protracted procedures to generate CD28(null) cells from CD28(+) precursors; hence, a T-cell line model was used to examine the dynamics of CD28 expression. Here, we show the versatility of the JT and Jtag cell lines in tracking CD28(null) <--> CD28(hi) phenotypic transitions. JT and Jtag cells were CD28(null) and CD28(lo), respectively, but expressed high levels of CD28 when exposed to phorbol 12-myristate 13-acetate. This was a result of the reconstitution of the CD28 gene transcriptional initiator (INR). Tumor necrosis factor-alpha reduced CD28 expression because of the inhibition of INR-driven transcription. Ligation of CD28 by an antibody or by CD80 also down-regulated CD28 transcription through the same mechanism, providing evidence that CD28 can generate a T cell receptor-independent signal with a unique biological outcome. Collectively, these data unequivocally demonstrate the critical role of the INR in the regulation of CD28 expression. T cell lines with transient expression of CD28 are invaluable in the dissection of the biochemical processes involved in the transactivation of the CD28 INR, the silencing of which is a key event in the ontogenesis of senescent T cells.

    View details for DOI 10.1074/jbc.M402194200

    View details for Web of Science ID 000222445300038

    View details for PubMedID 15128741

  • Lymphotoxin beta-mediated stimulation of synoviocytes in rheumatoid arthritis ARTHRITIS AND RHEUMATISM Braun, A., Takemura, S., Vallejo, A. N., Goronzy, J. J., Weyand, C. M. 2004; 50 (7): 2140-2150

    Abstract

    Lymphotoxin beta (LTbeta), a cytokine produced by T cells and B cells, plays a central role in the normal development of lymph nodes and is critical in the formation of ectopic germinal center reactions in rheumatoid synovitis. Because resident fibroblast-like synoviocytes (FLS) express receptors for LTbeta, we examined the consequences of FLS activation by LTbeta.FLS from patients with rheumatoid arthritis were isolated and examined for the expression of LTbeta receptor. FLS were incubated with LTalpha1beta2 and assayed for the production of cytokines and chemokines and the up-regulation of adhesion molecules.Exposure of FLS to recombinant LTalpha1beta2 resulted in the production of multiple inflammatory cytokines and metalloproteinases, implicating FLS as amplifiers of the inflammatory process in the inflamed joint. Additionally, LTalpha1beta2 was found to up-regulate the expression of cell adhesion molecules, rendering FLS to efficient adhesion substrates for T cells. LTalpha1beta2 also induced production of the chemokines CCL2 and CCL5, which elicited transmigration activity of T cells. Upon stimulation with LTalpha1beta2, FLS did not acquire characteristics of follicular dendritic cells.These data document that FLS are involved in multiple stages of the inflammatory process, including the recruitment and retention of lymphocytes in the synovial microenvironment. We propose that the heterotypic interaction between LTbeta-producing lymphocytes and responding FLS contributes to the establishment of complex lymphoid microstructures, and that this may be one element that defines susceptibility of the synovial membrane to lymphoid organogenesis.

    View details for DOI 10.1002/art.20356

    View details for Web of Science ID 000222820300012

    View details for PubMedID 15248211

  • T-cell regulation in rheumatoid arthritis CURRENT OPINION IN RHEUMATOLOGY Goronzy, J. J., Weyand, C. M. 2004; 16 (3): 212-217

    Abstract

    Rheumatoid arthritis (RA) is characterized by a chronic T-cell response that has escaped normal control mechanisms. This review summarizes recent insights in pathways that are functional in RA and that favor continuous and pathogenic T-cell activation.T-cell activation is ultimately determined by positive signals from costimulatory molecules and negative signals from regulatory T cells. Blockade of the classic costimulatory pathway, CD28-CD80 or CD86, is beneficial in RA. Additional pathways that predominantly control the activation of memory and effector T cells are functionally important in synovial inflammation. Some of these costimulatory molecules (such as stimulatory killer cell immunoglobulin-like receptors and NKG2D) appear to be relatively specific for RA and not to play a role in normal immune responses. In addition to this predominance of positive signals, age-disproportionate decline in thymic activity in RA may lead to a diminution of regulatory T cells and loss of their negative signals.The successful treatment trial of RA with CTLA-4Ig clearly documents the importance of T-cell costimulation in RA disease activity. Novel costimulatory pathways may be of even greater significance than CD28 in RA and may represent promising new therapeutic targets. The finding of reduced thymic activity in RA is exciting and will stimulate further studies of T-cell homeostasis and the function of regulatory cells.

    View details for Web of Science ID 000221051800007

    View details for PubMedID 15103247

  • How aggressive should initial therapy for rheumatoid arthritis be? Factors associated with response to 'non-aggressive' DMARD treatment and perspective from a 2-yr open label trial RHEUMATOLOGY Matteson, E. L., Weyand, C. M., Fulbright, J. W., Christianson, T. J., McClelland, R. L., Goronzy, J. J. 2004; 43 (5): 619-625

    Abstract

    To determine what baseline factors might be associated with response to an initial mild treatment regimen in patients with early rheumatoid arthritis (RA).Open label 2-yr study of 111 consecutive patients with early RA of duration less than 1 yr. None of the patients had previously received disease-modifying anti-rheumatic drugs (DMARDs). All patients were assigned to receive hydroxychloroquine (HCQ) at enrollment, and could also take non-steroidal anti-inflammatory drugs (NSAIDs) and prednisone. At any point during follow-up, patients not fulfilling the American College of Rheumatology (ACR) 50 criteria for improvement and/or who were taking prednisone > 10 mg/day were considered treatment failures and therapy changed to methotrexate (MTX), 7.5-20 mg/week. Clinical, laboratory and immunogenetic factors potentially predictive of treatment assignment at month 24 were evaluated.After 24 months of follow-up, a majority of patients (56/94) were either still on solo DMARD therapy with HCQ (n = 49) or off DMARD therapy with controlled/quiescent disease (n = 4), and 38 patients were taking MTX (including 11 in combination with other DMARDs). At month 24, all but 9 patients met ACR50 criteria for treatment response. Features present at enrollment which were predictors of MTX therapy at month 24 were high pain score, baseline rheumatoid factor titre > 1:40, higher number of swollen joints, and poor patient global assessment. The presence of HLA-C7xx at enrollment was also predictive of need for MTX therapy.This study suggests that even milder treatment with HCQ is greatly beneficial in patients with early RA. There continue to be very few consistently reliable predictors of treatment needs in patients with this disease.

    View details for DOI 10.1093/rheumatology/keh135

    View details for Web of Science ID 000221297900014

    View details for PubMedID 14983105

  • T-cell senescence: a culprit of immune abnormalities in chronic inflammation and persistent infection TRENDS IN MOLECULAR MEDICINE Vallejo, A. N., Weyand, C. M., Goronzy, J. J. 2004; 10 (3): 119-124

    Abstract

    Long-lived clonal T cells deficient in CD28 expression are commonly found in patients with inflammatory syndromes and persistent infections. Considering that CD28 loss is the most consistent immunological marker of aging, we propose that, in pathological states, CD28(null) T cells represent prematurely senescent cells resulting from persistent immune activation. These unusual lymphocytes have aberrant functions that contribute to disease-related immune abnormalities, and the degree of accumulation of CD28(null) T cells predicts the severity of clinical manifestations. We suggest that understanding of the biological properties of T cells that have reached replicative senescence will influence the future management of certain diseases. Indeed, studies on the molecular basis for the loss of CD28 are already providing information on methods to functionally rescue senescent T cells.

    View details for DOI 10.1016/j.molmed.2004.01.002

    View details for Web of Science ID 000220461200010

    View details for PubMedID 15102354

  • Biology of T lymphocytes RHEUMATIC DISEASE CLINICS OF NORTH AMERICA Vallejo, A. N., Davila, E., Weyand, C. M., Goronzy, J. J. 2004; 30 (1): 135-?

    Abstract

    T cells constitute one arm of the adaptive immune system. The accumulating information on various aspects of T-cell biology shows the intricacies in the regulation of immune responses. How we translate the cellular and molecular details of this regulation into innovation and development of therapies for disease management remains a fundamental, but exciting, challenge.

    View details for DOI 10.1016/S0889-857X(03)00114-5

    View details for Web of Science ID 000220293300008

    View details for PubMedID 15061572

  • Activation of arterial wall dendritic cells and breakdown of self-tolerance in giant cell arteritis JOURNAL OF EXPERIMENTAL MEDICINE Ma-Krupa, W., Jeon, M. S., Spoerl, S., Tedder, T. E., Goronzy, J. J., Weyand, C. M. 2004; 199 (2): 173-183

    Abstract

    Giant cell arteritis (GCA) is a granulomatous and occlusive vasculitis that causes blindness, stroke, and aortic aneurysm. CD4(+) T cells are selectively activated in the adventitia of affected arteries. In human GCA artery-severe combined immunodeficiency (SCID) mouse chimeras, depletion of CD83(+) dendritic cells (DCs) abrogated vasculitis, suggesting that DCs are critical antigen-presenting cells in GCA. Healthy medium-size arteries possessed an indigenous population of DCs at the adventitia-media border. Adoptive T cell transfer into temporal artery-SCID mouse chimeras demonstrated that DCs in healthy arteries were functionally immature, but gained T cell stimulatory capacity after injection of lipopolysaccharide. In patients with polymyalgia rheumatica (PMR), a subclinical variant of GCA, adventitial DCs were mature and produced the chemokines CCL19 and CCL21, but vasculitic infiltrates were lacking. Human histocompatibility leukocyte antigen class II-matched healthy arteries, PMR arteries, and GCA arteries were coimplanted into SCID mice. Immature DCs in healthy arteries failed to stimulate T cells, but DCs in PMR arteries could attract, retain, and activate T cells that originated from the GCA lesions. We propose that in situ maturation of DCs in the adventitia is an early event in the pathogenesis of GCA. Activation of adventitial DCs initiates and maintains T cell responses in the artery and breaks tissue tolerance in the perivascular space.

    View details for DOI 10.1084/jem.20030850

    View details for Web of Science ID 000188369700004

    View details for PubMedID 14734523

    View details for PubMedCentralID PMC2211768

  • The double life of NK receptors: stimulation or co-stimulation? TRENDS IN IMMUNOLOGY Snyder, M. R., Weyand, C. M., Goronzy, J. J. 2004; 25 (1): 25-32

    Abstract

    Stimulatory killer immunoglobulin-like receptors, NKG2D and stimulatory receptors of the CD94-NKG2 family have duplicity in function. On natural killer (NK) cells, these receptors act as independent and competent recognition units. Stimulatory NK receptors also appear on subsets of effector T cells, particularly those that have replicated extensively. When expressed on T cells, they amplify signals mediated through the T-cell antigen receptor and, thus, function as co-stimulatory, but not direct stimulatory, molecules. One mechanism responsible for this dichotomy is the differential expression of adaptor molecules. This duplicity in function, which is not seen for other co-stimulatory molecules, is responsible for the unique context information provided by the NK receptors, and it could explain their involvement in chronic inflammation and autoimmunity.

    View details for DOI 10.1016/j.it.2003.10.011

    View details for Web of Science ID 000188431800007

    View details for PubMedID 14698281

  • Immunopathways in giant cell arteritis and polymyalgia rheumatica AUTOIMMUNITY REVIEWS Weyand, C. M., Ma-Krupa, W., Goronzy, J. J. 2004; 3 (1): 46-53

    Abstract

    Giant cell arteritis (GCA), a vasculitis that targets medium- and large-size arteries, is ranked as a medical emergency because of its potential to cause blindness and stroke. The typical lesions, granulomas in the vessel wall, are formed by IFN-gamma-producing CD4+ T cells and macrophages. CD4+ T cells undergo in situ activation in the adventitia, where they interact with indigenous dendritic cells. Tissue injury is mediated by several distinct sets of macrophages that are committed to diverse effector functions. The dominant tissue injury in the media results from oxidative stress and leads to smooth muscle cell apoptosis and nitration of endothelial cells. Macrophage-derived growth factors are instrumental in driving the response-to-injury program of the artery that causes intimal hyperplasia and vessel occlusion. Clinical manifestations are those of tissue ischemia or a syndrome of exuberant systemic inflammation. The vascular and the systemic components of GCA contribute differentially to the disease, leading to distinct clinical phenotypes of this arteritis. Immunologically most interesting is polymyalgia rheumatica, in which the systemic component is combined with aborted vasculitis, suggesting a role for artery-specific tolerance mechanisms.

    View details for DOI 10.1016/S1568-9972(03)00064-8

    View details for Web of Science ID 000189125500007

    View details for PubMedID 14871649

  • Prognostic markers of radiographic progression in early rheumatoid arthritis ARTHRITIS AND RHEUMATISM Goronzy, J. J., Matteson, E. L., Fulbright, J. W., Warrington, K. J., Chang-Miller, A., Hunder, G. G., Mason, T. G., Nelson, A. M., Valente, R. M., Crowson, C. S., Erlich, H. A., Reynolds, R. L., Swee, R. G., O'Fallon, W. M., Weyand, C. M. 2004; 50 (1): 43-54

    Abstract

    To identify prognostic markers that are predictive of progressive erosive disease in patients with early rheumatoid arthritis (RA).The study involved an inception cohort of 111 consecutive patients with RA and a disease duration of <1 year. Patients were treated according to an algorithm designed to avoid overtreatment of mild disease and to accelerate treatment in patients who had continuous disease activity. Patients were evaluated for the presence of clinical and laboratory disease activity markers. We determined the frequency of CD4+,CD28(null) T cells by flow cytometry, HLA-DRB1 gene polymorphisms by polymerase chain reaction (PCR)/sequencing, and 26 single-nucleotide polymorphisms in 19 candidate genes by multiplex PCR and hybridization to an immobilized probe array. Data were analyzed using proportional odds models to identify prognostic markers predictive of erosive progression over 2 years on serial hand/wrist radiographs.After 2 years, disease activity in 52% of the cohort was controlled by treatment with hydroxychloroquine and nonsteroidal agents. Forty-eight percent of the patients did not develop erosions. Older age, presence of erosions at baseline, presence of rheumatoid factor, rheumatoid factor titer, and HLA-DRB1*04 alleles, particularly homozygosity for HLA-DRB1*04, were univariate predictors of radiographic progression. Promising novel markers were the frequency of CD4+,CD28(null) T cells as an immunosenescence indicator, and a polymorphism in the uteroglobin gene.Clinical disease activity in patients with early RA can frequently be controlled with nonaggressive treatment, but this is not always sufficient to prevent new erosions. Rheumatoid factor titer, HLA-DRB1 polymorphisms, age, and immunosenescence markers are predictors of poor radiographic outcome. A polymorphism in the uteroglobin gene may identify patients who have a low risk of erosive disease.

    View details for DOI 10.1002/art.11445

    View details for Web of Science ID 000188085600006

    View details for PubMedID 14730598

  • HLA-DRB1 haplotype did not affect the medium-term results of total knee arthroplasty in patients with rheumatoid arthritis. Modern rheumatology Takemura, S., Toda, Y., Goronzy, J. J., Weyand, C. M., Ogawa, R., Iida, H. 2004; 14 (1): 37-42

    Abstract

    This study investigated whether the HLA-DRB1 "susceptible allele" (SA) genotype is predictive for total knee arthroplasty (TKA) failure in patients with rheumatoid arthritis (RA). The results of 49 TKAs (30 RA patients) with an average follow-up of 7.9 years (range 5-15 years) were analyzed using a 12-item questionnaire and the Knee Society system. HLA-DRB1 alleles were used to estimate the severity of RA and divide the patients into three categories depending upon the gene dose of SA (SA+/+, SA+/-, and SA-/-). For all three categories, the 12-item questionnaire had significantly improved postoperatively, but without significant difference. We divided the 12 items of the questionnaire into two groups: knee-relevant parameters and general parameters. Patients in all three groups improved similarly in knee-relevant parameters. In contrast, those homozygous for SA (SA+/+) benefited less in general parameters. The average radiolucency score was 1.87 mm, with no difference being detected among the three groups. The HLA-DRB1 genotype did not affect the survival of the knee implants. Overall, patients without the RA-associated HLA gene benefited most from TKA as they improved not only in knee function, but also in parameters of general functional status.

    View details for PubMedID 17028803

  • Premature telomeric loss in rheumatoid arthritis is genetically determined and involves both myeloid and lymphoid cell lineages PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Schonland, S. O., Lopez, C., Widmann, T., Zimmer, J., Bryl, E., Goronzy, J. J., Weyand, C. M. 2003; 100 (23): 13471-13476

    Abstract

    In rheumatoid arthritis, peripheral blood T cells have age-inappropriate telomeric erosion. We examined whether HLA-DRB1*04 alleles, the major susceptibility genes for this disease, confer risk for T cell senescence. In healthy individuals, HLA-DRB1*04 alleles were associated with excessive loss of telomeres in CD4+ T cells. Accelerated telomeric erosion occurred during the first two decades of life and was followed by reduced homeostatic T cell proliferation during adulthood. Premature telomeric loss also affected granulocytes, suggesting that the hematopoietic stem cell is the primary target. Telomeric repair mechanisms were intact in HLA-DRB1*04+ donors. We propose that HLA-DRB1*04 alleles or genes in linkage disequilibrium regulate stem cell replication and contribute to the accumulation of senescent and autoreactive T cells in rheumatoid arthritis.

    View details for Web of Science ID 000186573700065

    View details for PubMedID 14578453

    View details for PubMedCentralID PMC263838

  • Giant-cell arteritis and polymyalgia rheumatica ANNALS OF INTERNAL MEDICINE Weyand, C. M., Goronzy, J. J. 2003; 139 (6): 505-515

    Abstract

    Giant-cell arteritis is an immune-mediated disease characterized by granulomatous infiltrates in the wall of medium-size and large arteries. The immunopathology consists of 2 components. Excessive cytokine production (for example, of interleukin-1 and interleukin-6) induces systemic inflammation with an exuberant acute-phase response. In parallel, interferon-gamma, which is released by T cells captured in the arterial wall, activates tissue-injurious macrophages. In response to the immune injury, the artery generates hyperplasia of the intima that leads to luminal occlusion and subsequent tissue ischemia. Despite the systemic character of the disease, distinct vascular territories are preferentially affected. On the basis of the predominant involvement, clinical subtypes can be distinguished: cranial giant-cell arteritis with ischemic complications in the eye, the face, and the central nervous system; large-vessel giant-cell arteritis with occlusions in the subclavian or axillary vessels; aortic giant-cell arteritis; giant-cell arteritis presenting as an intense systemic inflammatory syndrome with nonstenosing vasculitis; and "isolated" polymyalgia rheumatica with myalgias, systemic inflammation, and subclinical vasculitis. Temporal artery biopsy remains the diagnostic procedure of choice to detect arteritis in cranial vessels. In other vascular territories, giant-cell arteritis is most commonly diagnosed by vascular imaging. Laboratory studies characteristically document the marked elevations of nonspecific acute-phase reactants, such as C-reactive protein and erythrocyte sedimentation rate. Cytokines, such as interleukin-6, that induce the acute-phase reaction are currently being explored as more sensitive biological markers of disease activity. Corticosteroids are highly effective in suppressing systemic inflammation, but they do not eliminate the immune responses in the vessel wall. In general, the clinical outcome of giant-cell arteritis is excellent, and efforts must now concentrate on tailoring therapies to the needs of the individual patient.

    View details for Web of Science ID 000185324100008

    View details for PubMedID 13679329

  • Synoviocyte-mediated expansion of inflammatory T cells in rheumatoid synovitis is dependent on CD47-thrombospondin 1 interaction JOURNAL OF IMMUNOLOGY Vallejo, A. N., Yang, H. Y., Klimiuk, P. A., Weyand, C. M., Goronzy, J. J. 2003; 171 (4): 1732-1740

    Abstract

    Fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis elicit spontaneous proliferation of autologous T cells in an HLA-DR and CD47 costimulation-dependent manner. T cell costimulation through CD47 is attributed to specific interaction with thrombospondin-1 (TSP1), a CD47 ligand displayed on FLS. CD47 binding by FLS has broad biological impact that includes adhesion and the triggering of specific costimulatory signals. TSP1(+) FLS are highly adhesive to T cells and support their aggregation and growth in situ. Long-term cultures of T cells and FLS form heterotypic foci that are amenable to propagation without exogenous growth factors. T cell adhesion and aggregate formation on TSP1(+) FLS substrates are inhibited by CD47-binding peptides. In contrast, FLS from arthroscopy controls lack adhesive or T cell growth-promoting activities. CD47 stimulation transduces a costimulatory signal different from that of CD28, producing a gene expression profile that included induction of ferritin L chain, a component of the inflammatory response. Ferritin L chain augments CD3-induced proliferation of T cells. Collectively, these results demonstrate the active role of FLS in the recruitment, activation, and expansion of T cells in a CD47-dependent manner. Because TSP1 is abundantly expressed in the rheumatoid synovium, CD47-TSP1 interaction is proposed to be a key component of an FLS/T cell regulatory circuit that perpetuates the inflammatory process in the rheumatoid joint.

    View details for Web of Science ID 000184667400018

    View details for PubMedID 12902472

  • Homeostatic control of T-cell generation in neonates BLOOD Schonland, S. O., Zimmer, J. K., Lopez-Benitez, C. M., Widmann, T., Ramin, K. D., Goronzy, J. J., Weyand, C. M. 2003; 102 (4): 1428-1434

    Abstract

    T cells are produced through 2 mechanisms, thymopoiesis and proliferative expansion of postthymic T cells. Thymic output generates diversity of the pool, and proliferation achieves optimal clonal size of each individual T cell. To determine the contribution of these 2 mechanisms to the formation of the initial T-cell repertoire, we examined neonates of 30 to 40 weeks' gestation. Peripheral T cells were in a state of high proliferative turnover. In premature infants, 10% of T cells were dividing; the proliferation rates then declined but were still elevated in mature newborns. Throughout the third trimester, concentrations of T-cell-receptor excision circles (TRECs) were 10 per 100 T cells. Stability of TREC frequencies throughout the period of repertoire generation suggested strict regulation of clonal size to approximately 10 to 20 cells. Neonatal naive CD4+ and CD8+ T cells were explicitly responsive to IL-7; growth-promoting properties of IL-15 were selective for newborn CD8+ T cells. Neonatal T cells expressed telomerase and, in spite of the high turnover, built up a telomeric reserve. Thus, proliferative expansion, facilitated by increased cytokine responsiveness, and thymopoiesis complement each other as mechanisms of T-cell production in neonates. Maintaining optimal clonal size instead of filling the space in a lymphopenic host appears to regulate homeostatic T-cell proliferation during fetal development.

    View details for Web of Science ID 000184651600046

    View details for PubMedID 12714521

  • Immunosenescence, autoimmunity, and rheumatoid arthritis EXPERIMENTAL GERONTOLOGY Weyand, C. M., Fulbright, J. W., Goronzy, J. J. 2003; 38 (8): 833-841

    Abstract

    Current disease models of autoimmune syndromes, such as rheumatoid arthritis, propose that chronic inflammation is caused by 'forbidden T-cell clones' that recognize disease-inducing antigens and drive tissue-injurious immune reactions. Reappraisal of disease incidence data, however, emphasizes that rheumatoid arthritis is a syndrome of the elderly that occurs with highest likelihood in individuals in whom the processes of T-cell generation and T-cell repertoire formation are compromised. Thymic T-cell production declines rapidly with advancing age. Multiple mechanisms, including antigen-driven clonal expansion and homeostasis-driven autoproliferation of post-thymic T cells, impose replicative stress on T cells and induce the biological program of cellular senescence. T-cell immunosenescence is associated with profound changes in T-cell functional profile and leads to accumulation of CD4+ T cells that have lost CD28 but have gained killer immunoglobulin-like receptors and cytolytic capability and produce large amounts of interferon-gamma. In patients with rheumatoid arthritis, T-cell immunosenescence occurs prematurely, probably due to a deficiency in the ability to generate sufficient numbers of novel T cells. We propose that autoimmunity in rheumatoid arthritis is a consequence of immunodegeneration that is associated with age-inappropriate remodeling of the T-cell pool.

    View details for DOI 10.1016/S0531-5565(03)00090-1

    View details for Web of Science ID 000185452800003

    View details for PubMedID 12915205

  • De novo expression of killer immunoglobulin-like receptors and signaling proteins regulates the cytotoxic function of CD4 T cells in acute coronary syndromes CIRCULATION RESEARCH Nakajima, T., Goek, O., Zhang, X. Y., Kopecky, S. L., Frye, R. L., Goronzy, J. J., Weyand, C. M. 2003; 93 (2): 106-113

    Abstract

    The inflammatory infiltrate in atherosclerotic plaque is composed of T cells and macrophages. CD4+ T cells with a unique phenotype, CD4+CD28null, are preferentially recruited into culprit lesions. These T cells are distinct from classic CD4+CD28+ T cells in gene expression and function, including their ability to mediate cytolysis. In this study, we have investigated the regulation of CD4+CD28null T-cell cytolytic function. In patients with acute coronary syndromes (ACS), CD4+CD28null T cells express killer immunoglobulin-like receptors (KIRs). KIRs encompass a polymorphic family of receptors that recognize HLA class I molecules and have been implicated in self-tolerance. CD4+CD28null T-cell clones from patients with ACS and age-matched controls were compared for their KIR-expression profile. T-cell clones derived from the patients expressed a broader spectrum of KIRs (P<0.001) with preference for the stimulatory variant, CD158j. Additionally, CD4+ T-cell clones from patients but not those from controls acquired de novo expression of the DAP12 molecule, an adapter chain that transmits CD158j-derived signals. Cumulative expression of CD158j and DAP12 endowed cytolytic competence on CD4+CD28null T cells, allowing them to kill in the absence of T-cell receptor triggering. Our data demonstrate that CD4+CD28null T cells in ACS are characterized by a unique gene expression profile. Consequently, these T cells acquire cytolytic capability that can bypass the need for T-cell receptor triggering and, thus, impose a threat to self-tolerance.

    View details for DOI 10.1161/01.RES.0000082333.58263.58

    View details for Web of Science ID 000184346500005

    View details for PubMedID 12816883

  • Medium- and large-vessel vasculitis. New England journal of medicine Weyand, C. M., Goronzy, J. J. 2003; 349 (2): 160-169

    View details for PubMedID 12853590

  • Mechanisms of disease: Medium- and large-vessel vasculitis NEW ENGLAND JOURNAL OF MEDICINE Weyand, C. M., Goronzy, J. J. 2003; 349 (2): 160-169
  • The power of the third dimension: tissue architecture and autoimmunity in rheumatoid arthritis CURRENT OPINION IN RHEUMATOLOGY Weyand, C. M., Kang, Y. M., Kurtin, P. J., Goronzy, J. J. 2003; 15 (3): 259-266

    Abstract

    Lymphoid organs are the anatomic solution to the challenge of responding to minute amounts of antigen with powerful effector mechanisms. By arranging interacting cells in complex three-dimensional topographies lymphoid organs provide an optimal match between form and function. This principle is exploited in ectopic lymphoid structures that characteristically appear in rheumatoid synovitis. Synovial tissue T cells and B cells cooperate in different types of lymphoid organizations. Dendritic cell networks in the inflamed synovial membrane optimize antigen collection, storage, processing, and presentation. Synovial tissue cells participate in lymphocyte recruitment and the formation of tissue architectures that amplify immune responses. Recent data support the concept that the tissue organization in the rheumatoid joint fosters a breakdown in self-tolerance by promoting a phase transition from self-limited immune responses to self-perpetuating autoimmune responses.

    View details for Web of Science ID 000182544200013

    View details for PubMedID 12707579

  • CD28 loss in senescent CD4+ T cells: reversal by interleukin-12 stimulation BLOOD Warrington, K. J., Vallejo, A. N., Weyand, C. M., Goronzy, J. J. 2003; 101 (9): 3543-3549

    Abstract

    CD28 is the quintessential costimulatory molecule expressed on CD4(+) and CD8(+) T cells. During chronic infections and the normal aging process, CD28 expression is lost, compromising the functional activity of T cells. CD28 loss is promoted by replicative stress, particularly in the presence of tumor necrosis factor-alpha, owing to an inoperative CD28 initiator element. It is currently unknown whether CD28 loss is irreversible. The present study examined cytokines for their ability to reinduce CD28 expression. CD4(+)CD28(null) T cells constitutively expressed interleukin-12 (IL-12) alpha and beta receptors, which were functional and allowed for the up-regulation of the signal transducer and activator of transcription-4 (STAT-4)-dependent gene CD161. Costimulation of the T-cell and IL-12 receptors induced the transcription of CD28 in approximately 50% of CD4(+)CD28(null) T-cell clones and lines. IL-12 by itself did not restore CD28 expression. Up-regulation of CD28 after IL-12 exposure correlated with the reassembly of the CD28-initiator protein complex. The re-expressed CD28 was functional and restored the ability of CD4(+)CD28(null) T cells to express CD25 and CD40 ligand. Our data suggest that IL-12 may, in part, functionally rescue senescent CD4(+) T cells.

    View details for DOI 10.1182/blood-2002-08-2574

    View details for Web of Science ID 000182625600042

    View details for PubMedID 12506015

  • Selective activation of the c-Jun NH2-terminal protein kinase signaling pathway by stimulatory KIR in the absence of KARAP/DAP12 in CD4(+) T cells JOURNAL OF EXPERIMENTAL MEDICINE Snyder, M. R., Lucas, M., Vivier, E., Weyand, C. M., Goronzy, J. J. 2003; 197 (4): 437-449

    Abstract

    Activation of CD4(+) T cells is governed by interplay between stimulatory and inhibitory receptors; predominance of stimulatory signals favors autoimmune reactions. In patients with rheumatoid arthritis, expression of the critical costimulatory molecule, CD28, is frequently lost. Instead, CD4(+)CD28(null) T cells express killer immunoglobulin-like receptors (KIRs) with a preferential expression of the stimulatory receptor, CD158j. The frequency of CD4(+)CD28(null) T cells in rheumatoid arthritis (RA) correlates with the risk for more severe disease. Moreover, the KIR2DS2 gene, which encodes for CD158j, is a genetic risk factor for rheumatoid vasculitis. CD158j signals through the adaptor molecule, KARAP/DAP12, to positively regulate cytotoxic activity in NK cells. However, the majority of CD4(+)CD28(null) T cell clones lacked the expression of KARAP/DAP12. Despite the absence of KARAP/DAP12, CD158j was functional and augmented interferon-gamma production after T cell receptor stimulation. Cross-linking of CD158j resulted in selective phosphorylation of c-Jun NH(2)-terminal protein kinase (JNK) and its upstream kinase, MKK4 that led to the expression of ATF-2 and c-Jun, all in the absence of extracellular signal-regulated kinase (ERK)1/2 phosphorylation. Mutation of the lysine residue within the transmembrane domain of CD158j abolished JNK activation, suggesting that an alternate adaptor molecule was being used. CD4(+)CD28(null) T cells expressed DAP10 and inhibition of phosphatidylinositol 3-kinase, which acts downstream of DAP10, inhibited JNK activation; however, no interaction of DAP10 with CD158j could be detected. Our data suggest that CD158j in T cells functions as a costimulatory molecule through the JNK pathway independent of KARAP/DAP12 and DAP10. Costimulation by CD158j may contribute to the autoreactivity of CD4(+)CD28(null) T cells in RA.

    View details for DOI 10.1084/jem.20020383

    View details for Web of Science ID 000181179400005

    View details for PubMedID 12591902

    View details for PubMedCentralID PMC2193867

  • Ectopic germinal center formation in rheumatoid synovitis Conference on Immune Mechanisms and Disease Weyand, C. M., Goronzy, J. J. NEW YORK ACAD SCIENCES. 2003: 140–149

    Abstract

    Synovial inflammation in rheumatoid arthritis is closely related to the formation of ectopic lymphoid microstructures. In synovial tissue from some patients, one finds seemingly diffuse infiltrates; in others, T cells and B cells cluster in aggregates with interdigitating dendritic cells (DCs) but no follicular DCs (FDCs). In a third group, T cell/B cell follicles with germinal center (GC) reactions are generated. Within a given patient, aggregates and GCs are mutually exclusive and stable over time. Because antigen storage capacity, lymphoid density, and three-dimensional topography of GCs optimize immune responses, synovial GCs should play a crucial role in the breakdown of self-tolerance. We have identified factors critical for ectopic GCs, thereby transforming the synovial inflammatory process. Tissues with GCs produced 10- to 20-fold higher amounts of the chemokines CXCL13 and CCL21. CXCL13 derived from three sources, endothelial cells, synovial fibroblasts, and FDC networks. The level of CXCL13 transcripts strongly predicted GCs; however, some tissues had high levels of CXCL13 but lacked GCs. Tissue expression of LT-beta emerged as a second key factor. LT-beta protein was detected on follicular center and mantle zone B cells. Multivariate regression analysis identified CXCL13 and LT-beta as the only cytokines predicting GCs. Remarkably, LT-alpha did not contribute independently. The contribution of B cells to ectopic lymphoid organogenesis was not limited to LT-beta production. Rather, synovial tissue B cells were critical in regulating T cell activation. In adoptive transfer experiments in human synovium-SCID mouse chimeras, activation of synovium-derived CD4 T cells was strictly dependent on T cell/B cell follicles. Depletion of synovial tissue B cells abrogated T cell function, and non-B cell antigen-presenting cells could not maintain T cell stimulation. Unexpectedly, GC function in the rheumatoid lesion was also dependent on CD8 T cells. The majority of T cell receptors derived from CD8 T cells were shared between distinct GCs. Depletion of CD8 T cells disrupted synovial GCs, FDC networks disappeared, and transcription of LT-beta, IgG, and Igkappa declined. Follicle-sustaining CD8 T cells were located at the edge of or within the mantle zone. Cell-cell communication in the mantle zone, including CD8 T cells, appears to be critical for ectopic GC formation in rheumatoid synovitis.

    View details for Web of Science ID 000183319000016

    View details for PubMedID 12727633

  • T-cell senescence and contraction of T-cell repertoire diversity catalysts of autoimmunity and chronic inflammation ARTHRITIS RESEARCH & THERAPY Goronzy, J. J., Weyand, C. M. 2003; 5 (5): 225-234

    Abstract

    Rheumatoid arthritis (RA), like many other autoimmune syndromes, is a disease of adults, with the highest incidence rates reported in the elderly. The immune system undergoes profound changes with advancing age that are beginning to be understood and that need to be incorporated into the pathogenetic models of RA. The age-related decline in thymic function causes extensive remodeling of the T-cell system. Age-dependent changes in T-cell homeostasis are accelerated in patients with RA. The repertoire of naive and memory T cells is less diverse, possibly as a result of thymic insufficiency, and it is biased towards autoreactive cells. Presenescent T cells emerge that are resistant to apoptosis and that often expand to large clonal populations. These cells are under the regulatory control of nonconventional costimulatory molecules, display potent effector functions, and appear to be critical in the synovial and extra-articular manifestations of RA.

    View details for DOI 10.1186/ar974

    View details for Web of Science ID 000184729800005

    View details for PubMedCentralID PMC193735

  • Aging, autoimmunity and arthritis: T-cell senescence and contraction of T-cell repertoire diversity - catalysts of autoimmunity and chronic inflammation. Arthritis research & therapy Goronzy, J. J., Weyand, C. M. 2003; 5 (5): 225-234

    Abstract

    Rheumatoid arthritis (RA), like many other autoimmune syndromes, is a disease of adults, with the highest incidence rates reported in the elderly. The immune system undergoes profound changes with advancing age that are beginning to be understood and that need to be incorporated into the pathogenetic models of RA. The age-related decline in thymic function causes extensive remodeling of the T-cell system. Age-dependent changes in T-cell homeostasis are accelerated in patients with RA. The repertoire of naive and memory T cells is less diverse, possibly as a result of thymic insufficiency, and it is biased towards autoreactive cells. Presenescent T cells emerge that are resistant to apoptosis and that often expand to large clonal populations. These cells are under the regulatory control of nonconventional costimulatory molecules, display potent effector functions, and appear to be critical in the synovial and extra-articular manifestations of RA.

    View details for PubMedID 12932282

  • B cells as a therapeutic target in autoimmune disease ARTHRITIS RESEARCH & THERAPY Goronzy, J. J., Weyand, C. M. 2003; 5 (3): 131-135

    Abstract

    Depleting B cells with anti-CD20 monoclonal antibodies emerges as a new therapeutic strategy in autoimmune diseases. Preliminary clinical studies suggest therapeutic benefits in patients with classic autoantibody-mediated syndromes, such as autoimmune cytopenias. Treatment responses in rheumatoid arthritis have opened the discussion about whether mechanisms beyond the removal of potentially pathogenic antibodies are effective in B-cell depletion. B cells may modulate T-cell activity through capturing and presenting antigens or may participate in the neogenesis of lymphoid microstructures that amplify and deviate immune responses. Studies exploring which mechanisms are functional in which subset of patients hold the promise of providing new and rational treatment approaches for autoimmune syndromes.

    View details for DOI 10.1186/ar751

    View details for Web of Science ID 000182395200010

    View details for PubMedID 12723978

    View details for PubMedCentralID PMC165047

  • Molecular basis for the loss of CD28 expression in senescent T cells JOURNAL OF BIOLOGICAL CHEMISTRY Vallejo, A. N., Bryl, E., Klarskov, K., Naylor, S., Weyand, C. M., Goronzy, J. J. 2002; 277 (49): 46940-46949

    Abstract

    CD28(null) T cells are the most consistent biological indicator of the aging immune system in humans and are predictors of immunoincompetence in the elderly. The loss of CD28 is the result of an inoperative transcriptional initiator (INR), which consists of two nonoverlapping alpha and beta motifs that have distinct protein binding profiles but function as a unit. In CD28(null) T cells, there is a coordinate loss of alpha-/beta-bound complexes, hence the alphabeta-INR is inactive. In the present work therefore, studies were conducted to identify the components of such complexes that may account for the trans-activation of the alphabeta-INR. By affinity chromatography and tandem mass spectrometry, two proteins, namely, nucleolin and the A isoform of heterogeneous nuclear ribonucleoprotein-D0 (hnRNP-D0A), were identified to be among the key components of the site alpha complex. In DNA binding assays, specific antibodies indicated their antigenic presence in alpha-bound complexes. Transcription assays showed that they are both required in the trans-activation of alphabeta-INR-driven DNA templates. Because CD28 is T cell-restricted, and nucleolin and hnRNP-D0A are ubiquitous proteins, these results support the notion that cell-specific functions can be regulated by commonly expressed proteins. The present data also provide evidence for INR-regulated transcription that is independent of the known components of the basal transcription complex.

    View details for DOI 10.1074/jbc.M207352200

    View details for Web of Science ID 000179663700015

    View details for PubMedID 12324461

  • Trapping of misdirected dendritic cells in the granulomatous lesions of giant cell arteritis AMERICAN JOURNAL OF PATHOLOGY Krupa, W. M., Dewan, M., Jeon, M. S., Kurtin, P. J., Younge, B. R., Goronzy, J. J., Weyand, C. M. 2002; 161 (5): 1815-1823

    Abstract

    Immature dendritic cells (DCs) are scattered throughout peripheral tissues and act as sentinels that sample the antigenic environment. After activation, they modify their chemokine receptor profile and migrate toward lymphoid tissues. On arrival, they have matured into chemokine-producing DCs that express co-stimulatory molecules and can prime naive T cells. Normal temporal arteries contain immature DCs that are located at the media-adventitia border. In temporal arteries affected by giant cell arteritis, DCs are highly enriched and activated and have matured into fully differentiated cells producing the chemokines, CCL18, CCL19, and CCL21. In keeping with their advanced maturation, DCs in the granulomatous lesions possess the chemokine receptor, CCR7. CCR7 binds CCL19 and CCL21, causing the highly activated DCs to be trapped in the peripheral tissue site. The co-stimulatory molecule, CD86, which is critical for DC/T-cell interaction, is expressed by a subset of DCs captured in the arterial wall. DC/T-cell interaction does not involve interleukin-12; transcripts for interleukin-12 p40 are absent in the vasculitic infiltrates. We propose that differentiation of DCs and the autocrine and paracrine actions of chemokines in granulomatous lesions misdirect DCs away from their usual journey to lymphoid organs and are critical in maintaining T-cell activation and granuloma formation in giant cell arteritis.

    View details for Web of Science ID 000179197000030

    View details for PubMedID 12414528

    View details for PubMedCentralID PMC1850804

  • Reactive nitrogen intermediates in giant cell arteritis - Selective nitration of neocapillaries AMERICAN JOURNAL OF PATHOLOGY Borkowski, A., Younge, B. R., Szweda, L., Mock, B., Bjornsson, J., Moeller, K., Goronzy, J. J., Weyand, C. M. 2002; 161 (1): 115-123

    Abstract

    Arterial wall damage in giant cell arteritis (GCA) is mediated by several different macrophage effector functions, including the production of metalloproteinases and lipid peroxidation. Tissue-invading macrophages also express nitric oxide synthase (NOS)-2, but it is not known whether nitric oxide-related mechanisms contribute to the disease process. Nitric oxide can form nitrating agents, including peroxynitrite, a nitric oxide congener formed in the presence of reactive oxygen intermediates. Protein nitration selectively targets tyrosine residues and can result in a gain, as well as a loss, of protein function. Nitrated tyrosine residues in GCA arteries were detected almost exclusively on endothelial cells of newly formed microcapillaries in the media, whereas microvessels in the adventitia and the intima were spared. Nitration correlated with endothelial NOS-3 expression and not with NOS-2-producing macrophages, which preferentially homed to the hyperplastic intima. The restriction of nitration to the media coincided with the production of reactive oxygen intermediates as demonstrated by the presence of the toxic aldehyde, 4-hydroxynonenal. Depletion of tissue-infiltrating macrophages in human temporal artery-SCID mouse chimeras disrupted nitrotyrosine generation, demonstrating a critical role of macrophages in the nitration process that targeted medial microvessels. Thus, protein nitration in GCA is highly compartmentalized, reflecting the production of reactive oxygen and reactive nitrogen intermediates in the inflamed arterial wall. Heterogeneity of microvessels in NOS-3 regulation may be an additional determinant contributing to this compartmentalization and could explain the preferential targeting of newly generated capillary beds.

    View details for Web of Science ID 000176718300015

    View details for PubMedID 12107096

    View details for PubMedCentralID PMC1850706

  • The Ogryzlo Day Lecture - Premature immunosenescence in rheumatoid arthritis - Presented at the University of Toronto Rheumatic Disease Unit Annual Ogryzlo Research Day, June 26, 2001 Annual Ogryzlo Research Day Weyand, C. M., Goronzy, J. J. J RHEUMATOL PUBL CO. 2002: 1141–46
  • CD8 T cells are required for the formation of ectopic germinal centers in rheumatoid synovitis JOURNAL OF EXPERIMENTAL MEDICINE Kang, Y. M., Zhang, X. Y., Wagner, U. G., Yang, H. Y., Beckenbaugh, R. D., Kurtin, P. J., Goronzy, J. J., Weyand, C. M. 2002; 195 (10): 1325-1336

    Abstract

    The assembly of inflammatory lesions in rheumatoid arthritis is highly regulated and typically leads to the formation of lymphoid follicles with germinal center (GC) reactions. We used microdissection of such extranodal follicles to analyze the colonizing T cells. Although the repertoire of follicular T cells was diverse, a subset of T cell receptor (TCR) sequences was detected in multiple independent follicles and not in interfollicular zones, suggesting recognition of a common antigen. Unexpectedly, the majority of shared TCR sequences were from CD8 T cells that were highly enriched in the synovium and present in low numbers in the periphery. To examine their role in extranodal GC reactions, CD8 T cells were depleted in human synovium-SCID mouse chimeras. Depletion of synovial CD8 T cells caused disintegration of the GC-containing follicles. In the absence of CD8 T cells, follicular dendritic cells disappeared, production of lymphotoxin-alpha1beta2 markedly decreased, and immunoglobulin (Ig) secretion ceased. Immunohistochemical studies demonstrated that these CD8 T cells accumulated at the edge of the mantle zone. Besides their unique localization, they were characterized by the production of interferon (IFN)-gamma, lack of the pore-forming enzyme perforin, and expression of CD40 ligand. Perifollicular IFN-gamma+ CD8 T cells were rare in secondary lymphoid tissues but accounted for the majority of IFN-gamma+ cells in synovial infiltrates. We propose that CD8+ T cells regulate the structural integrity and functional activity of GCs in ectopic lymphoid follicles.

    View details for DOI 10.1084/jem.20011565

    View details for Web of Science ID 000176110900009

    View details for PubMedID 12021312

    View details for PubMedCentralID PMC2193749

  • Human leukocyte antigen class II immune response genes, female gender, and cigarette smoking as risk and modulating factors in abdominal aortic aneurysms JOURNAL OF VASCULAR SURGERY Rasmussen, T. E., Hallett, J. W., Tazelaar, H. D., Miller, V. M., Schulte, S., O'Fallon, W. M., Weyand, C. M. 2002; 35 (5): 988-993

    Abstract

    Aortic inflammation and the genes that regulate the immune response play an important role in abdominal aortic aneurysm pathogenesis. However, the modulating effects of such genetic and other environmental factors on the severity on aneurysm inflammation is not known. The objective of this study was to determine the influence of the human leukocyte antigen (HLA) class II genes, gender, and environmental factors on degree of abdominal aortic aneurysm tissue inflammation.Aneurysm specimens were obtained at the time of operation from 96 consecutive patients who underwent abdominal aortic aneurysm repair and were graded for degree of histologic inflammation. Multivariate analysis was used to determine the association of genetic and environmental factors with degree of inflammation and to determine the HLA-associated disease risk for aneurysm.Active cigarette smoking and female gender were independently associated with high-grade tissue inflammation identified histologically (odds ratio [OR], confidence interval [CI]: 5.6, 1.6 to 19.3; and 6.0, 1.4 to 26.2, respectively), and a specific HLA allele (DR B1(*)01) was inversely associated with inflammation (OR, CI: 0.2, 0.04 to 0.7). Overall, the HLA-DR B1(*)02 and B1(*)04 alleles were significantly associated with disease risk, more than doubling risk for abdominal aortic aneurysm (OR, CI: 2.5, 1.4 to 4.3; and 2.1, 1.2 to 3.7, respectively).Active cigarette smoking and female gender are significant disease-modulating factors associated with increased abdominal aortic aneurysm inflammation. In addition, the HLA class II immune response genes possess both disease modulating and disease risk properties, which may be useful in early aneurysm detection and surveillance.

    View details for DOI 10.1067/mva.2002.121753

    View details for Web of Science ID 000175919100034

    View details for PubMedID 12021716

  • Cytokines in giant-cell arteritis 10th International Vasculitis and ANCA Workshop Goronzy, J. J., Weyand, C. M. CLEVELAND CLINIC. 2002: 91–94

    Abstract

    Cytokines are small proteins that serve as chemical messengers between cells, regulating cell growth and differentiation, tissue repair and remodeling, and many aspects of the immune response. Cytokines are instrumental in determining the nature, magnitude, and duration of inflammatory reactions and, as such, represent ideal targets for interfering with pathogenic processes. In OCA and PMR, cytokines are encountered in two locations, the inflammatory infiltrates accumulating in the arterial wall and in the circulation. IL-6, a cytokine involved in stimulating acute-phase responses, is located upstream of many of the laboratory abnormalities considered helpful in diagnosing and managing GCA/PMR, including elevated ESR and CRP. IL-6 has the potential to be helpful in predicting disease severity and may allow for a tailoring of immunosuppressive therapy. There is evidence suggesting that IL-6 outperforms other chemical markers in detecting disease activity and could, therefore, have a role in monitoring treatment. Interesting pathogenic clues have been derived from studies of cytokines produced in the vascular lesions. IFN-gamma has emerged as a key regulator in determining the nature and direction of the inflammatory response. IFN-gamma appears to be critically involved in modulating the process of intimal hyperplasia, the most destructive consequence of vasculitis, and, as such, emerges as a prime target for novel therapeutic approaches.

    View details for Web of Science ID 000177952200019

  • Formation of the killer Ig-like receptor repertoire on CD4(+)CD28(null) T cells JOURNAL OF IMMUNOLOGY Snyder, M. R., Muegge, L. O., Offord, C., O'Fallon, W. M., Bajzer, Z., Weyand, C. M., Goronzy, J. J. 2002; 168 (8): 3839-3846

    Abstract

    Killer Ig-like receptors (KIRs) are expressed on CD4(+)CD28(null) T cells, a highly oligoclonal subset of T cells that is expanded in patients with rheumatoid arthritis. It is unclear at what stage of development these T cells acquire KIR expression. To determine whether KIR expression is a consequence of clonal expansion and replicative senescence, multiple CD4(+)CD28(null) T cell clones expressing the in vivo dominant TCR beta-chain sequences were identified in three patients and analyzed for their KIR gene expression pattern. Based on sharing of TCR sequences, the clones were grouped into five clone families. The repertoire of KIRs was diverse, even within each clone family; however, the gene expression was not random. Three particular receptors, KIR2DS2, KIR2DL2, and KIR3DL2, had significant differences in gene expression frequencies between the clone families. These data suggest that KIRs are successively acquired after TCR rearrangement, with each clone family developing a dominant expression pattern. The patterns did not segregate with the individual from whom the clones were derived, indicating that peripheral selection in the host environment was not a major shaping force. Several models were examined using a computer algorithm that was designed to simulate the expression of KIRs at various times during T cell proliferation. The computer simulations favored a model in which KIR gene expression is inducible for a limited time during the initial stages of clonal expansion.

    View details for Web of Science ID 000174913300022

    View details for PubMedID 11937537

  • T-cell-mediated lysis of endothelial cells in acute coronary syndromes CIRCULATION Nakajima, T., Schulte, S., Warrington, K. J., Kopecky, S. L., Frye, R. L., Goronzy, J. J., Weyand, C. M. 2002; 105 (5): 570-575

    Abstract

    CD4 T lymphocytes accumulate in unstable plaque. The direct and indirect involvement of these T cells in tissue injury and plaque instability is not understood.Gene profiling identified perforin, CD161, and members of the killer-cell immunoglobulin-like receptors as being differentially expressed in CD4(+)CD28(null) T cells, a T-cell subset that preferentially infiltrates unstable plaque. Frequencies of CD161(+) and perforin-expressing CD4 T cells in peripheral blood were significantly increased in patients with unstable angina (UA). CD161 appeared on CD4(+)CD28(null) T cells after stimulation, suggesting spontaneous activation of circulating CD4 T cells in UA. Perforin-expressing CD4(+) T-cell clones from patients with UA exhibited cytotoxic activity against human umbilical vein endothelial cells (HUVECs) in redirected cytotoxicity assays after T-cell receptor triggering and also after stimulation of major histocompatibility complex class I-recognizing killer-cell immunoglobulin-like receptors. HUVEC cytolysis was dependent on granule exocytosis, as demonstrated by the paralyzing effect of pretreating CD4(+)CD28(null) T cells with strontium. Incubation of HUVECs with C-reactive protein (CRP) increased HUVEC lysis in a dose-dependent fashion.In patients with UA, CD4 T cells undergo a change in functional profile and acquire cytotoxic capability. Cytotoxic CD4 T cells effectively kill endothelial cells; CRP sensitizes endothelial cells to the cytotoxic process. We propose that T-cell-mediated endothelial cell injury is a novel pathway of tissue damage that contributes to plaque destabilization. The sensitizing effect of CRP suggests synergy between dysregulated T-cell function and acute phase proteins in acute coronary syndromes.

    View details for Web of Science ID 000173735400022

    View details for PubMedID 11827921

  • Therapeutic effects of acetylsalicylic acid in giant cell arteritis ARTHRITIS AND RHEUMATISM Weyand, C. M., Kaiser, M., Yang, H. Y., Younge, B., Goronzy, J. J. 2002; 46 (2): 457-466

    Abstract

    In giant cell arteritis (GCA), inflammatory lesions typically produce interferon-gamma(IFNgamma)-- and nuclear factor kappaB (NF-kappaB)-dependent monokines. Corticosteroids influence disease activity by repressing NF-kappaB-dependent genes but have only marginal effects on IFNgamma. The current study explored whether acetylsalicylic acid (ASA) had cytokine-repressing activity in GCA and could function as a steroid-sparing agent.Temporal artery-severe combined immunodeficiency (SCID) mouse chimeras were created by engrafting inflamed temporal arteries into SCID mice. Chimeras were treated with ASA, indomethacin, or dexamethasone for 3 weeks. Temporal artery grafts were harvested and cytokine message was semiquantified by polymerase chain reaction-enzyme-linked immunosorbent assay. The ability of dexamethasone and ASA to suppress IFNgamma and interleukin-1beta (IL-1beta) messenger RNA and protein production was also tested in vitro using T cell clones and monocytes derived from patients with GCA. Drug-induced effects on the transcription factors NF-kappaB and activator protein 1 (AP-1) were assessed by electrophoretic mobility shift assays (EMSAs).At clinically relevant doses, 20-100 mg/kg, ASA was a highly effective inhibitor of cytokine transcription in temporal arteries. While dexamethasone preferentially targeted NF-kappaB-regulated monokines, ASA acted predominantly by suppressing IFNgamma. Indomethacin failed to reduce tissue IFNgamma transcription, which therefore excluded the inhibition of cyclooxygenases as a critical mechanism. IFNgamma production by T cell clones was highly sensitive to ASA-mediated suppression, whereas IL-1beta production by lipopolysaccharide-stimulated monocytes responded primarily to dexamethasone. The combination of ASA and dexamethasone had synergistic effects. EMSAs demonstrated that ASA interfered with the formation of AP-1, whereas dexamethasone suppressed the nuclear translocation of NF-kappaB.The results of this study provide evidence of the complementary action of ASA and corticosteroids in suppressing proinflammatory cytokines in the vascular lesions of GCA.

    View details for Web of Science ID 000173785800021

    View details for PubMedID 11840449

  • Pathogenic mechanisms in giant cell arteritis. Cleveland Clinic journal of medicine Weyand, C. M., Goronzy, J. J. 2002; 69: SII28-32

    Abstract

    T lymphocytes, encountering stimulatory signals in the adventitia of medium-size arteries, emerge as the key players in inflammation-associated injury pathways. In GCA, all injury mechanisms have been related to effector macrophages. Regulated by IFN-gamma-producing T cells, macrophages commit to distinct avenues of differentiation and acquire a spectrum of potentially harmful capabilities (Figure 1). Macrophages in the adventitia focus on production of pro-inflammatory cytokines. Macrophages in the media specialize in oxidative damage with lipid peroxidation attacking smooth muscle cells and matrix components. These macrophages also supply reactive oxygen intermediates that, in combination with nitrogen intermediates, cause protein nitration of endothelial cells. Production of oxygen radicals is complemented by the production of metalloproteinases, likely essential in the breakdown of elastic membranes. With the fragmentation of the internal elastic lamina, the intimal layer becomes accessible to migratory myofibroblasts that, driven by PDGF, form a hyperplastic intimal layer and cause occlusion of the vessel lumen. Expansion of the hyperplastic intima is accompanied by intense neoangiogenesis, supported by angiogenesis factors that again derive from specialized macrophages. Similarities in injury pathways between GCA and another arterial disease, atherosclerosis, are beginning to be recognized. Specifically, activated T cells and macrophages are increasingly appreciated as key players in the process of instability and rupture of atherosclerotic plaque. A specialized subset of CD4 T cells, CD4+ CD28- T cells, are suspected to participate in tissue injury in the plaque. These T cells are equipped with cytolytic capabilities and release large amounts of IFN-gamma. Comparative studies between patients with GCA and those with acute coronary syndromes should enhance our ability to define the principles of arterial wall inflammation, the specifics of injury in that microenvironment, and help in the identification of the eliciting signals.

    View details for PubMedID 12086261

  • The pathogenesis of giant cell arteritis BULLETIN ON THE RHEUMATIC DISEASES Weyand, C. M., Goronzy, J. J. 2002; 51 (8): 1-8
  • Cytokines in giant-cell arteritis. Cleveland Clinic journal of medicine Goronzy, J. J., Weyand, C. M. 2002; 69: SII91-4

    Abstract

    Cytokines are small proteins that serve as chemical messengers between cells, regulating cell growth and differentiation, tissue repair and remodeling, and many aspects of the immune response. Cytokines are instrumental in determining the nature, magnitude, and duration of inflammatory reactions and, as such, represent ideal targets for interfering with pathogenic processes. In OCA and PMR, cytokines are encountered in two locations, the inflammatory infiltrates accumulating in the arterial wall and in the circulation. IL-6, a cytokine involved in stimulating acute-phase responses, is located upstream of many of the laboratory abnormalities considered helpful in diagnosing and managing GCA/PMR, including elevated ESR and CRP. IL-6 has the potential to be helpful in predicting disease severity and may allow for a tailoring of immunosuppressive therapy. There is evidence suggesting that IL-6 outperforms other chemical markers in detecting disease activity and could, therefore, have a role in monitoring treatment. Interesting pathogenic clues have been derived from studies of cytokines produced in the vascular lesions. IFN-gamma has emerged as a key regulator in determining the nature and direction of the inflammatory response. IFN-gamma appears to be critically involved in modulating the process of intimal hyperplasia, the most destructive consequence of vasculitis, and, as such, emerges as a prime target for novel therapeutic approaches.

    View details for PubMedID 12086274

  • Bi-directional modulation of T cell-dependent antibody production by prostaglandin E-2 INTERNATIONAL IMMUNOLOGY He, X. W., Weyand, C. M., Goronzy, J. J., Zhong, W. Y., Stuart, J. M. 2002; 14 (1): 69-77

    Abstract

    T cell-dependent Ig production involves interaction between T cells and B cells. This study evaluated the effects of prostaglandin (PG) E(2) on Ig production in a system in which B cells were co-cultured with autologous CD4(+) T cell clones non-specifically activated by anti-CD3. The effects of PGE(2) on T cell-dependent Ig production differed substantially, depending on the T cells employed. We selected six T cell clones that were able to enhance Ig production (resistant T cell clones) and six T cell clones that inhibited Ig production in the presence of PGE(2) (sensitive T cell clones) for comparison. The resistant T cells produced high levels (>1000 pg/ml) of IL-2 and/or IL-4, and expressed high CD40L, OX40 and CD45RA, and low CD45RO. In contrast, sensitive T cells secreted low IL-2 (<500 pg/ml) and IL-4 (<200 pg/ml), and expressed low CD40, OX40 and CD45RA, and high CD45RO. Adding supernatant derived from resistant T cell clones restored Ig production inhibited by PGE(2), while removing IL-2, IL-4 or IL-10 using specific antibodies inhibited Ig production. In addition, we demonstrated a direct effect of PGE(2) on B cells to enhance Ig production. Consistently, in the presence of resistant T cells, PGE(2) increased B cell proliferation and differentiation. In conclusion, the effects of PGE(2) on Ig production consist of its indirect effects through T cells and its direct effects on B cells. The outcome of the effects can be up-regulatory or down-regulatory, depending whether resistant or sensitive T cells are involved.

    View details for Web of Science ID 000173387200009

    View details for PubMedID 11751754

  • Value of immunological markers in predicting responsiveness to influenza vaccination in elderly individuals JOURNAL OF VIROLOGY Goronzy, J. J., Fulbright, J. W., Crowson, C. S., Poland, G. A., O'Fallon, W. M., Weyand, C. M. 2001; 75 (24): 12182-12187

    Abstract

    Elderly individuals are at high risk for morbidity and mortality when infected with influenza virus. Vaccinations with inactivated virus are less effective in the elderly due to the declining competency of the aging immune system. We have explored whether immunological parameters predict poor anti-influenza virus vaccine responses and can be used as biological markers of immunosenescence. One hundred fifty-three residents of community-based retirement facilities aged 65 to 98 years received a trivalent influenza vaccine. Vaccine-induced antibody responses were determined by comparing hemagglutination inhibition titers before and 28 days after immunization. The composition of the T-cell compartment was analyzed by flow cytometry and the sizes of three T-cell subsets, CD4(+) CD45RO(+) cells, CD4(+) CD28(null) cells, and CD8(+) CD28(null) cells, were determined. Only 17% of the vaccine recipients were able to generate an increase in titers of antibody to all three vaccine components, and 46% of the immunized individuals failed to respond to any of the three hemagglutinins. The likelihood of successful vaccination declined with age and was independently correlated with the expansion of a particular T-cell subset, CD8(+) CD28(null) T cells. The sizes of the CD4(+) CD45RO(+) memory T-cell and CD4(+) CD28(null) T-cell subsets had no effect on the ability to mount anti-influenza virus antibody responses. Frequencies of CD8(+) CD28(null) T cells are useful biological markers of compromised immunocompetence, identifying individuals at risk for insufficient antibody responses.

    View details for Web of Science ID 000172355300026

    View details for PubMedID 11711609

    View details for PubMedCentralID PMC116115

  • T cell activation in rheumatoid synovium is B cell dependent JOURNAL OF IMMUNOLOGY Takemura, S., Klimiuk, P. A., Braun, A., Goronzy, J. J., Weyand, C. M. 2001; 167 (8): 4710-4718

    Abstract

    Rheumatoid arthritis results from a T cell-driven inflammation in the synovial membrane that is frequently associated with the formation of tertiary lymphoid structures. The significance of this extranodal lymphoid neogenesis is unknown. Microdissection was used to isolate CD4 T cells residing in synovial tissue T cell/B cell follicles. CD4 T cells with identical TCR sequences were represented in independent, nonadjacent follicles, suggesting recognition of the same Ag in different germinal centers. When adoptively transferred into rheumatoid arthritis synovium-SCID mouse chimeras, these CD4 T cell clones enhanced the production of IFN-gamma, IL-1beta, and TNF-alpha. In vivo activity of adoptively transferred CD4 T cells required matching of HLA-DRB1 alleles and also the presence of T cell/B cell follicles. HLA-DRB1-matched synovial tissues that were infiltrated by T cells, macrophages, and dendritic cells, but that lacked B cells, did not support the activation of adoptively transferred CD4 T cell clones, raising the possibility that B cells provided a critical function in T cell activation or harbored the relevant Ag. Dependence of T cell activation on B cells was confirmed in B cell depletion studies. Treatment of chimeric mice with anti-CD20 mAb inhibited the production of IFN-gamma and IL-1beta, indicating that APCs other than B cells could not substitute in maintaining T cell activation. The central role of B cells in synovial inflammation identifies them as excellent targets for immunosuppressive therapy.

    View details for Web of Science ID 000171858300071

    View details for PubMedID 11591802

  • T-cell immunity in acute coronary syndromes MAYO CLINIC PROCEEDINGS Weyand, C. M., Goronzy, J. J., Liuzzo, G., Kopecky, S. L., Holmes, D. R., Frye, R. L. 2001; 76 (10): 1011-1020

    Abstract

    Acute coronary syndromes (ACS) are complications of atherosclerotic vascular disease that are triggered by the sudden rupture of an atheroma. Atherosclerotic plaque stability is determined by multiple factors, of which immune and inflammatory pathways are critical. Unstable plaque is characterized by an infiltrate of T cells and macrophages, thereby resembling a delayed hypersensitivity reaction. On activation, T cells secrete cytokines that regulate the activity of macrophages, or the T cells may differentiate into effector cells with tissue-damaging potential. Constitutive stimulation of T cells and macrophages in ACS is not limited to the vascular lesion but also involves peripheral immune cells, suggesting fundamental abnormalities in homeostatic mechanisms that control the assembly, turnover, and diversity of the immune system as a whole. This review gives particular attention to the emergence of a specialized T-cell subset, natural killer T cells, in patients with ACS. Natural killer T cells have proinflammatory properties and the capability of directly contributing to vascular injury.

    View details for Web of Science ID 000171341900009

    View details for PubMedID 11605685

  • Down-regulation of CD28 expression by TNF-alpha JOURNAL OF IMMUNOLOGY Bryl, E., Vallejo, A. N., Weyand, C. M., Goronzy, J. J. 2001; 167 (6): 3231-3238

    Abstract

    Aging and chronic inflammatory syndromes, such as rheumatoid arthritis, are associated with high frequencies of CD4(+)CD28(null) T cells, which are rarely seen in healthy individuals younger than 40 years. Inasmuch as rheumatoid arthritis and aging are also associated with elevated levels of TNF-alpha, we examined whether this proinflammatory cytokine influences CD28 expression. Incubation of T cell lines and clones as well as Jurkat cells with TNF-alpha induced a reduction in the levels of cell surface expression of CD28. This effect of TNF-alpha was reversible; however, continuous culture of CD4(+)CD28(+) T cell clones in TNF-alpha resulted in the appearance of a CD28(null) subset. In reporter gene bioassays, TNF-alpha was found to inhibit the activity of the CD28 minimal promoter. Inactivation of the promoter was accompanied by a marked reduction in DNA-protein complex formation by two DNA sequence motifs corresponding to the transcriptional initiator of the CD28 gene. Indeed, in vitro transcription assays showed that nuclear extracts from TNF-alpha-treated cells failed to activate transcription of DNA templates under the control of a consensus TATA box and the CD28 initiator sequences. In contrast, similar extracts from unstimulated T cells supported transcription. These results demonstrate that TNF-alpha directly influences CD28 gene transcription. We propose that the emergence of CD4(+)CD28(null) T cells in vivo is facilitated by increased production of TNF-alpha.

    View details for Web of Science ID 000172392000027

    View details for PubMedID 11544310

  • Ectopic lymphoid organogenesis - A fast track for autoimmunity AMERICAN JOURNAL OF PATHOLOGY Weyand, C. M., Kurtin, P. J., Goronzy, J. J. 2001; 159 (3): 787-793

    View details for Web of Science ID 000170872400002

    View details for PubMedID 11549568

    View details for PubMedCentralID PMC1850453

  • Lymphoid neogenesis in rheumatoid synovitis JOURNAL OF IMMUNOLOGY Takemura, S., Braun, A., Crowson, C., Kurtin, P. J., Cofield, R. H., O'Fallon, W. M., Goronzy, J. J., Weyand, C. M. 2001; 167 (2): 1072-1080

    Abstract

    In rheumatoid arthritis (RA), tissue-infiltrating lymphocytes can be arranged in sophisticated organizations that resemble microstructures usually formed in secondary lymphoid organs. Molecular pathways and host risk factors involved in this process of lymphoid neogenesis remain to be defined. In a series of 64 synovial tissue biopsies, lymphoid follicles with germinal centers (GCs) were found in 23.4% of the patients. Follicular dendritic cells (FDCs) were exclusively present in tissues with GCs, suggesting that the recruitment or in situ maturation of FDCs is a critical factor for GC formation in the synovial membrane. Primary follicles were absent, emphasizing the role of Ag recognition in the generation of inflammation-associated lymphoid organogenesis. Multivariate logistic regression analysis of tissue cytokines and chemokines identified two parameters, in situ transcription of lymphotoxin (LT)-beta and of B lymphocyte chemoattractant (BLC; BLC/CXCL13), that were predictors for FDC recruitment and synovial GC formation. LT-beta and BLC/CXCL13 were found to be independent variables that could, in part, compensate for each other to facilitate GC formation. Prediction models incorporating in situ transcription of LT-beta and BLC/CXCL13 had high negative yet moderate positive predictive values, suggesting that LT-beta and BLC/CXCL13 are necessary but not sufficient. LT-beta protein was detected on a subset of mantle zone and GC B cells, but also on T cells in follicular structures. BLC/CXCL13 was produced by FDCs in follicular centers, but was predominantly found in endothelial cells and synovial fibroblasts, suggesting heterotypic signaling between cells of the synovial membrane and infiltrating lymphocytes in regulating extranodal lymphoid neogenesis.

    View details for Web of Science ID 000170949300058

    View details for PubMedID 11441118

  • Genetic similarity in inflammatory and degenerative abdominal aortic aneurysms: A study of human leukocyte antigen class II disease risk genes 24th Annual Meeting of the Midwestern-Vascular-Surgery-Society Rasmussen, T. E., Hallett, J. W., Schulte, S., Harmsen, W. S., O'Fallon, W. M., Weyand, C. M. MOSBY-ELSEVIER. 2001: 84–88

    Abstract

    Clinically, abdominal aortic aneurysms (AAAs) display a spectrum of inflammation that extends from apparently noninflamed (degenerative) AAAs to the classic inflammatory variant. Genes encoded in the human leukocyte antigen (HLA) region are important in the development of both variants of AAA; however, their role in progression to the inflammatory variant is unknown. The purpose of this study was to compare HLA class II genes in patients with degenerative versus classic inflammatory AAAs and to quantify their impact as disease risk factors.Genotypes of the 12 major alleles of the HLA-DR B1 locus were determined in patients with degenerative (102) and inflammatory (40) AAAs who were compared with controls (118). Univariate and multivariate logistic regression analyses were used to determine allele distributions and to quantify disease risk.Distribution of the HLA-DR B1 alleles was nonrandom and similar in both degenerative and inflammatory AAA groups compared with controls. The B1*02 and B1*04 alleles were enhanced in both degenerative (39.2% vs. 25.4%, P =.03; and 35.3% vs. 24.6%, P =.08 respectively) and inflammatory (47.5% vs. 25.4%, P =.01; and 32.5% vs. 24.6%, P =.09, respectively) AAAs compared with controls. The B1*02 and B1*04 alleles were associated with risk for both degenerative (odds ratio [OR] 2.2; 95% CI, 1.2-4.0; and OR 2.0; 95% CI, 1.1-3.7, respectively) and inflammatory AAAs (OR 3.7; 95% CI, 1.8-8.6; and OR 2.5; 95% CI, 1.1-6.1).This study demonstrates that identical HLA alleles function as genetic risk factors for both inflammatory and degenerative AAAs. These results support the concept of a common, immune-mediated pathogenesis for AAAs that may be modulated by HLA-independent factors.

    View details for Web of Science ID 000169840000026

    View details for PubMedID 11436079

  • Major histocompatibility complex class I-recognizing receptors are disease risk genes in rheumatoid arthritis JOURNAL OF EXPERIMENTAL MEDICINE Yen, J. H., Moore, B. E., Nakajima, T., SCHOLL, D., Schaid, D. J., Weyand, C. M., Goronzy, J. J. 2001; 193 (10): 1159-1167

    Abstract

    Rheumatoid arthritis (RA) is a heterogeneous syndrome of which a subset of patients develops vascular inflammation. The genetic determinants that confer risk for rheumatoid vasculitis are not known, but patients with vascular complications are known to have an expansion of CD4(+)CD28(null) T cells, a cell population potentially involved in endothelial damage. CD4(+)CD28(null) T cell clones isolated from RA patients with vasculitis were found to express killer cell immunoglobulin-like receptors (KIRs) with the stimulatory KIR2DS2 often present in the absence of opposing inhibitory receptors with related specificities. To test the hypothesis that the KIR2DS2 gene is involved in the development of vasculitis, association studies were performed. The KIR2DS2 gene was significantly enriched among patients with rheumatoid vasculitis compared with normal individuals (odds ratio 5.56, P = 0.001) and patients with RA but no vasculitis (odds ratio 7.96, P = 0.001). Also, the distribution of human histocompatibility leukocyte antigen (HLA)-C, the putative ligand for KIRs, was significantly different in patients with rheumatoid vasculitis in comparison with the control populations. These data suggest that HLA class I-recognizing receptors and HLA class I genes are genetic risk determinants that modulate the pattern of RA expression. Specifically, KIR2DS2 in conjunction with the appropriate HLA-C ligand may have a role in vascular damage by regulating CD4(+)CD28(null) T cells.

    View details for Web of Science ID 000169115200006

    View details for PubMedID 11369787

    View details for PubMedCentralID PMC2193323

  • Thymic function and peripheral T-cell homeostasis in rheumatoid arthritis TRENDS IN IMMUNOLOGY Goronzy, J. J., Weyand, C. M. 2001; 22 (5): 251-255

    Abstract

    T-cell diversity is generated through the production of new thymic emigrants. Thymic function declines with age, and the T-cell pool is maintained through homeostatic proliferation of naive peripheral T cells. This article discusses the impact of thymic output and peripheral T-cell homeostasis on the development of rheumatoid arthritis (RA). It is proposed that thymic output is prematurely compromised in RA patients. A compensatory expansion of peripheral T cells results in a contracted and distorted repertoire, possibly favoring T cells with autoreactive potential. Increased risk of autoimmunity, as a consequence of abnormal T-cell population dynamics, could be a common mechanism in chronic inflammatory diseases.

    View details for Web of Science ID 000169936400011

    View details for PubMedID 11323282

  • Molecular fingerprint of interferon-gamma signaling in unstable angina CIRCULATION Liuzzo, G., Vallejo, A. N., Kopecky, S. L., Frye, R. L., Holmes, D. R., Goronzy, J. J., Weyand, C. M. 2001; 103 (11): 1509-1514

    Abstract

    Activation of circulating monocytes in patients with acute coronary syndromes may reflect exposure to bacterial products or stimulation by cytokines such as IFN-gamma. IFN-gamma induces phosphorylation and nuclear translocation of transcription factor STAT-1, which initiates a specific program of gene induction. To explore whether monocyte activation is IFN-gamma driven, patients with unstable (UA) or stable angina (SA) were compared for nuclear translocation of STAT-1 complexes and upregulation of IFN-gamma-inducible genes CD64 and IP-10.Peripheral blood mononuclear cells were stained for expression of CD64 on CD14(+) monocytes and analyzed by PCR for transcription of IP-10. Expression of CD64 was significantly increased in patients with UA. Monocytes from UA patients remained responsive to IFN-gamma in vitro, with accelerated transcriptional competency of CD64. IP-10-specific sequences were spontaneously detectable in 82% of the UA patients and 15% of SA patients (P<0.001). Most importantly, STAT-1 complexes were found in nuclear extracts prepared from freshly isolated monocytes of patients with UA, which provides compelling evidence for IFN-gamma signaling in vivo.Monocytes from UA patients exhibit a molecular fingerprint of recent IFN-gamma triggering, such as nuclear translocation of STAT-1 complexes and upregulation of IFN-gamma-inducible genes CD64 and IP-10, which suggests that monocytes are activated, at least in part, by IFN-gamma. IFN-gamma may derive from stimulated T lymphocytes, which implicates specific immune responses in the pathogenesis of acute coronary syndromes.

    View details for Web of Science ID 000167636800006

    View details for PubMedID 11257077

  • Functional disruption of the CD28 gene transcriptional initiator in senescent T cells JOURNAL OF BIOLOGICAL CHEMISTRY Vallejo, A. N., Weyand, C. M., Goronzy, J. Y. 2001; 276 (4): 2565-2570

    Abstract

    We recently reported that aging is accompanied by the emergence of CD4(+)CD28(null) T cells, a functionally aberrant lymphocyte subset rarely seen in individuals younger than 40 years. Here, we directly examined whether the lack of CD28 expression is due to a defect at the level of transcriptional initiation. Molecular studies reveal that CD28 gene transcription is controlled by two sequence motifs, sites alpha and beta. In vitro transcription assays using initiator-dependent DNA templates revealed that reversed polarity or the deletion of either motif inhibited transcription, indicating that alpha/beta sequences constitute a composite initiator. Moreover, nuclear extracts from CD28(null) cells failed to activate transcription of alphabeta-initiator DNA templates. Transcription of such templates was, however, restored with the addition of extracts from CD28(+) cells. Although previously described initiator elements have been defined by a consensus sequence, the alphabeta-initiator has no homology to such sequence. These studies demonstrate that initiators have functions other than positioning elements for the basal transcription complex. Rather, initiators can have a direct role in regulating the expression of specific genes. The gain or loss of initiator activity can be an important determinant of cell phenotypes.

    View details for Web of Science ID 000166784800039

    View details for PubMedID 11069899

  • T cell homeostasis and autoreactivity in rheumatoid arthritis. Current directions in autoimmunity Goronzy, J. J., Weyand, C. M. 2001; 3: 112-132

    View details for PubMedID 11791462

  • Lymphoid microstructures in rheumatoid synovitis. Current directions in autoimmunity Weyand, C. M., Braun, A., Takemura, S., Goronzy, J. J. 2001; 3: 168-187

    View details for PubMedID 11791464

  • CD4+,CD28-T cells in rheumatoid arthritis patients combine features of the innate and adaptive immune systems ARTHRITIS AND RHEUMATISM Warrington, K. J., Takemura, S., Goronzy, J. J., Weyand, C. M. 2001; 44 (1): 13-20

    Abstract

    To determine whether CD4+,CD28- T cells, which are expanded in patients with rheumatoid arthritis (RA), express receptors that typically regulate the function of natural killer (NK) cells.Expression of the NK cell surface molecules CD158, p70, CD94, CD161, and CD8alpha on T cell subsets was determined by multicolor flow cytometric analysis of peripheral blood mononuclear cells from 36 RA patients. Expression of CD161 on tissue-infiltrating CD4 T cells was determined by 2-color immunohistochemistry analysis of synovial tissue samples.Killer cell-inhibitory receptors (KIR) and killer cell-activating receptors (KAR) were exclusively expressed on CD4+,CD28- T cells, with the CD158b molecule being the most frequently detected isoform. A coordinated mechanism inducing KIR/KAR expression was suggested by similarities in the expression of CD158b on CD4 and CD8 T cells. CD4+,CD28- T cells were also positive for CD8-alphaalpha homodimers, another characteristic shared with NK cells. Of the C-type lectin NK cell receptors (NK receptors), CD94 was consistently absent, but CD161 was found on a CD4 T cell population that is significantly expanded in RA patients (P = 0.01). Involvement in disease of NK receptor-expressing CD4 T cells was suggested by the presence of CD4+,CD161+ T cells in follicular microstructures typical of rheumatoid synovitis.Patients with RA have an expanded and unusual subset of CD4 T cells that infiltrates the tissue lesions and is characterized by a deficiency of CD28, the expression of CD8-alphaalpha homodimers, and the expression of several types of HLA class I-recognizing NK receptors. CD4 T cells bearing NK receptors can bridge functions of the innate and adaptive immune systems, such as responsiveness to specific antigen, rapid release of interferon-gamma, cytotoxicity, independence from classic costimulatory pathways, and integration of multiple activating and inhibitory signals to control effector functions.

    View details for Web of Science ID 000166659100003

    View details for PubMedID 11212151

  • Clonality and longevity of CD4(+)CD28(null) T cells are associated with defects in apoptotic pathways JOURNAL OF IMMUNOLOGY Vallejo, A. N., Schirmer, M., Weyand, C. M., Goronzy, J. J. 2000; 165 (11): 6301-6307

    Abstract

    CD4(+)CD28(null) T cells are oligoclonal lymphocytes rarely found in healthy individuals younger than 40 yr, but are found in high frequencies in elderly individuals and in patients with chronic inflammatory diseases. Contrary to paradigm, they are functionally active and persist over many years. Such clonogenic potential and longevity suggest altered responses to apoptosis-inducing signals. In this study, we show that CD4(+)CD28(null) T cells are protected from undergoing activation-induced cell death. Whereas CD28(+) T cells underwent Fas-mediated apoptosis upon cross-linking of CD3, CD28(null) T cells were highly resistant. CD28(null) T cells were found to progress through the cell cycle, and cells at all stages of the cell cycle were resistant to apoptosis, unlike their CD28(+) counterparts. Neither the activation-induced up-regulation of the IL-2R alpha-chain (CD25) nor the addition of exogenous IL-2 renders them susceptible to Fas-mediated apoptosis. These properties of CD28(null) T cells were related to high levels of Fas-associated death domain-like IL-1-converting enzyme-like inhibitory protein, an inhibitor of Fas signaling that is normally degraded in T cells following activation in the presence of IL-2. Consistent with previous data showing protection of CD28(null) cells from spontaneous cell death, the present studies unequivocally show dysregulation of apoptotic pathways in CD4(+)CD28(null) T cells that favor their clonal outgrowth and maintenance in vivo.

    View details for Web of Science ID 000165467100036

    View details for PubMedID 11086066

  • Pathogenic principles in giant cell arteritis International Conference on Takayasu Arteritis and Buerger Disease Weyand, C. M., Goronzy, J. J. ELSEVIER IRELAND LTD. 2000: S9–S15

    Abstract

    In giant cell arteritis, an immune insult in the vascular wall initiates a reaction in the artery that leads to structural changes, intimal hyperplasia, and luminal occlusion. The mechanisms triggering the immune stimulation are unknown; however, the process is strictly dependent on T cells that are found in the vicinity of the vasa vasorum in the adventitia and that produce interferon-gamma. The major effector cells in the artery are macrophages and giant cells that are ultimately under T-cell control but assume different functions depending on their location in the arterial wall. The response of the artery to the injury is maladaptive and includes mobilization and proliferation of smooth muscle cells in conjunction with matrix production and neoangionesis, resulting in the formation of a lumen-obstructive neointima. Heterogeneity in the immune insult and the resulting arterial response patterns correlate with variations in clinical disease.

    View details for Web of Science ID 000089537800002

    View details for PubMedID 10980331

  • T cell homeostasis in patients with rheumatoid arthritis PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Koetz, K., Bryl, E., Spickschen, K., O'Fallon, W. M., Goronzy, J. J., Weyand, C. M. 2000; 97 (16): 9203-?

    Abstract

    The immune system is equipped with an extremely large spectrum of structurally diverse receptors to recognize all potential antigens. This fundamental principle of receptor diversity is no longer upheld in patients with rheumatoid arthritis (RA), who have a marked contraction of the T cell receptor repertoire. In this study, the ability of RA patients to produce T cells and to maintain T cell homeostasis was examined. CD4 T cells containing T cell receptor rearrangement excision circles (TREC) were substantially reduced in RA patients; TREC levels in young adult patients matched those of controls 20 years older. Increased self-replication of T cells in RA was indicated by age-inappropriate erosion of telomeres in circulating T cells with almost complete attrition of telomeric reserves in patients 20-30 yr of age. The degree of telomere loss was not related to disease duration or the use of disease-modifying medication and was most pronounced in CD4(+)CD45RO(null) (naive) T cells. The loss of TREC-positive T cells could be a consequence of a primary defect in peripheral T cell homeostasis. Alternatively, RA patients may have impaired thymic function with the increased turnover of peripheral T cells being a secondary compensatory event.

    View details for Web of Science ID 000088608000080

    View details for PubMedID 10922071

    View details for PubMedCentralID PMC16846

  • Killer cell activating receptors function as costimulatory molecules on CD4(+)CD28(null) T cells clonally expanded in rheumatoid arthritis JOURNAL OF IMMUNOLOGY NAMEKAWA, T., Snyder, M. R., Yen, J. H., Goehring, B. E., Leibson, P. J., Weyand, C. M., Goronzy, J. J. 2000; 165 (2): 1138-1145

    Abstract

    Expansion of CD4+CD28null T cells is a characteristic finding in patients with rheumatoid arthritis. Despite lacking CD28 molecules, these unusual CD4 T cells undergo clonal proliferation and form large and long-lived clonal populations. They produce high levels of IFN-gamma, exhibit autoreactivity, and have cytolytic function. The mechanisms facilitating the expansion and longevity of CD4+CD28null T cell clones in vivo are unknown. Here, we report that CD4+CD28null, but not CD4+CD28+, T cells express MHC class I-recognizing receptors normally found on NK cells. CD4+CD28null T cells preferentially expressed killer cell activating receptors (KAR), often in the absence of killer cell inhibitory receptors. Cross-linking of KAR molecules enhanced the proliferative response to TCR-mediated stimulation, but not the cytolytic function of CD4+CD28null T cells, suggesting different signaling pathways in CD4 T cells and NK cells. Triggering of KAR signaling led to the phosphorylation of several cellular targets, although the pattern of phosphorylation differed from that induced by the TCR. Aberrant expression of KAR molecules in the absence of inhibitory receptors and in the appropriate HLA setting may lead to the clonal outgrowth of autoreactive CD4+CD28null T cells commonly seen in rheumatoid arthritis.

    View details for Web of Science ID 000088085600064

    View details for PubMedID 10878393

  • Monoclonal T-cell proliferation and plaque instability in acute coronary syndromes CIRCULATION Liuzzo, G., Goronzy, J. J., Yang, H. Y., Kopecky, S. L., Holmes, D. R., Frye, R. L., Weyand, C. M. 2000; 101 (25): 2883-2888

    Abstract

    Unstable angina (UA) is associated with systemic inflammation and with expansion of interferon-gamma-producing T lymphocytes. The cause of T-cell activation and the precise role of activated T cells in plaque instability are not understood.Peripheral blood T cells from 34 patients with stable angina and 34 patients with UA were compared for the distribution of functional T-cell subsets by flow cytometric analysis. Clonality within the T-cell compartment was identified by T-cell receptor spectrotyping and subsequent sequencing. Tissue-infiltrating T cells were examined in extracts from coronary arteries containing stable or unstable plaque. The subset of CD4(+)CD28(null) T cells was expanded in patients with UA and infrequent in patients with stable angina (median frequencies: 10.8% versus 1.5%, P<0.001). CD4(+)CD28(null) T cells included a large monoclonal population, with 59 clonotypes isolated from 20 UA patients. T-cell clonotypes from different UA patients used antigen receptors with similar sequences. T-cell receptor sequences derived from monoclonal T-cell populations were detected in the culprit but not in the nonculprit lesion of a patient with fatal myocardial infarction.UA is associated with the emergence of monoclonal T-cell populations, analogous to monoclonal gammopathy of unknown significance. Shared T-cell receptor sequences in clonotypes of different patients implicate chronic stimulation by a common antigen, for example, persistent infection. The unstable plaque but not the stable plaque is invaded by clonally expanded T cells, suggesting a direct involvement of these lymphocytes in plaque disruption.

    View details for Web of Science ID 000087810100011

    View details for PubMedID 10869258

  • New insights into the pathogenesis of rheumatoid arthritis International Conference on Challenges and Advances in Rheumatoid Arthritis Weyand, C. M. OXFORD UNIV PRESS. 2000: 3–8

    Abstract

    T lymphocytes play a critical role in the inflammatory process of rheumatoid arthritis (RA). Studies in a new animal model of RA, created by implanting human inflamed synovium into SCID mice, have confirmed that the production of matrix-degrading enzymes and pro-inflammatory cytokines is ultimately under T-cell control. T-cell dysfunction in RA patients also alters T-cell dynamics, resulting in profound abnormalities in T-cell pool composition. The cause and consequences of altered T-cell dynamics in RA are not yet understood, but factors determining T-cell homeostasis include the generation of new T cells, loss of T cells during immune responses and self-renewal of T cells within the system. Understanding the mechanisms that govern the formation of the T-cell pool in RA emphasizes the dynamic and quantitative aspects of lymphocyte behaviour in RA and has profound therapeutic implications when devising strategies to counteract T-cell dysfunction.

    View details for Web of Science ID 000087832200002

    View details for PubMedID 11001373

  • Treatment of giant cell arteritis - Interleukin-6 as a biologic marker of disease activity ARTHRITIS AND RHEUMATISM Weyand, C. M., Fulbright, J. W., Hunder, G. G., Evans, J. M., Goronzy, J. J. 2000; 43 (5): 1041-1048

    Abstract

    To determine the value of the erythrocyte sedimentation rate (ESR) and plasma interleukin-6 (IL-6) as biologic markers for monitoring disease activity in giant cell arteritis (GCA).Twenty-five patients with biopsy-proven GCA were enrolled into a prospective treatment study. Therapy was initiated with prednisone, 60 mg/day, followed by a predetermined tapering schedule. Patients were monitored monthly for clinical signs of active vasculitis and laboratory parameters indicative of inflammation, including elevated ESR (>30 mm/hour) and elevated plasma IL-6 concentrations (>6.1 pg/ml).Upon initiation of corticosteroid treatment, clinical signs of GCA disappeared in all patients; however, 60% of the patients developed symptoms of recurrent disease, on 1 or more occasions, while the prednisone dosage was being reduced. These 31 disease flares diagnosed over 550 days were associated with symptoms of systemic inflammation but did not result in vascular complications. The ESR was elevated in 76% of the patients prior to initiation of treatment (median 65 mm/hour) and normalized by day 28 of therapy (median 6 mm/hour). The median ESR remained in the normal range during the followup period. Plasma IL-6 levels, which were abnormal in 92% of untreated patients (median 16 pg/ml), were partially responsive to the initial high doses of corticosteroids by day 28 (median 6 pg/ml), but levels did not completely normalize with continued therapy. Elevation of the ESR was seen during only 58% of all disease flares, but 89% of disease recurrences were associated with increased plasma IL-6 levels (P = 0.03).Plasma IL-6 is more sensitive than ESR for indicating disease activity in untreated and treated GCA patients. Standard corticosteroid regimens only partially suppress vascular inflammation. Smoldering disease activity may expose GCA patients to the risk of progressive vascular disease (e.g., formation of aortic aneurysms) and chronic systemic complications such as IL-6-mediated osteopenia.

    View details for Web of Science ID 000086969000012

    View details for PubMedID 10817557

  • Anti-psoriatic drug anthralin activates JNK via lipid peroxidation: Mononuclear cells are more sensitive than keratinocytes JOURNAL OF INVESTIGATIVE DERMATOLOGY Peus, D., Beyerle, A., Rittner, H. L., Pott, M., Meves, A., WEYAND, C., PITTELKOW, M. R. 2000; 114 (4): 688-692

    Abstract

    Anthralin is a widely used, topical therapy for psoriasis. Anti-proliferative and anti-inflammatory properties of anthralin have been identified. Little is known, however, about differential sensitivities of targeted cell types and specific mechanisms of signaling pathway activation. We demonstrate that anthralin exerts potent effects on keratinocytes and mononuclear cells through strong induction of lipid peroxidation and JNK activation, a stress-induced signal transduction pathway. Lipid peroxidation was observed rapidly and half-maximal levels of lipid peroxidation were reached at a 10-fold lower concentration of anthralin for peripheral blood mononuclear cells vs normal keratinocytes. JNK activation was detected in peripheral blood mononuclear cells at a 40-fold lower anthralin dose compared with keratinocytes. For both cell types, selected inhibitors of lipid peroxidation prevented JNK activation. This study demonstrates that mononuclear leukocytes are markedly more sensitive than keratinocytes to anthralin-induced lipid peroxidation and JNK activation. We identify anthralin as a novel and potent inducer of JNK activation and demonstrate that this process is mediated, at least in part, by lipid peroxidation which is among the earliest and most proximate, membrane-related responses to anthralin yet described.

    View details for Web of Science ID 000086346300014

    View details for PubMedID 10733674

  • Central role of thrombospondin-1 in the activation and clonal expansion of inflammatory T cells JOURNAL OF IMMUNOLOGY Vallejo, A. N., Mugge, L. O., Klimiuk, P. A., Weyand, C. M., Goronzy, J. J. 2000; 164 (6): 2947-2954

    Abstract

    Thrombospondin-1 (TSP) is a transiently expressed matricellular protein known to promote chemotaxis of leukocytes to inflammatory sites. However, TSP and its receptor CD36 are abundantly expressed in chronically inflamed tissues such as the rheumatoid synovium. Here, we show that TSP provides the costimulatory signal that is necessary for the activation of autoreactive T cells. Data presented reveal that TSP-mediated costimulation is achieved through its independent interaction with CD36 on APCs and with CD47 on T cells. We propose that a CD47-TSP-CD36 trimolecular complex is a novel costimulatory pathway that significantly decreases the threshold of T cell activation. Consistent with the paradigm that lesions in rheumatoid synovitis are sites of antigenic recognition, the characteristic focal expression of TSP on APCs such as macrophages and fibroblast-like synoviocytes suggest a central role of TSP in the expansion of tissue-infiltrating T cells.

    View details for Web of Science ID 000085766800014

    View details for PubMedID 10706681

  • [Clinical and genetic heterogeneity of rheumatoid arthritis]. Postepy higieny i medycyny doswiadczalnej Klimiuk, P. A., Weyand, C. M., Sierakowski, S., Goronzy, J. J. 2000; 54 (6): 845-853

    Abstract

    The diagnostic category of rheumatoid arthritis, a syndrome of chronic inflammatory disease of the synovial membrane and of extraarticular tissues, covers a broad spectrum of clinical phenotypes. Here we propose that distinct combinations of disease risk genes produce heterogeneity of rheumatoid disease. Recognition of this genetic and clinical heterogeneity has immediate implications as it provides the opportunity to develop selective therapies for the different variants of disease.

    View details for PubMedID 11227380

  • Cell-cell interactions in synovitis - Interactions between T cells and B cells in rheumatoid arthritis ARTHRITIS RESEARCH Weyand, C. M., Goronzy, J. J., Takemura, S., Kurtin, P. J. 2000; 2 (6): 457-463

    Abstract

    In rheumatoid arthritis, T cells and B cells participate in the immune responses evolving in the synovial lesions. Interaction between T cells and B cells is probably antigen specific because complex microstructures typical of secondary lymphoid organs are generated. Differences between patients in forming follicles with germinal centers, T-cell-B-cell aggregates without germinal center reactions, or loosely organized T-cell-B-cell infiltrates might reflect the presence of different antigens or a heterogeneity in host response patterns to immune injury. Tertiary lymphoid microstructures in the rheumatoid lesions can enhance the sensitivity of antigen recognition, optimize the collaboration of immunoregulatory and effector cells, and support the interaction between the tissue site and the aberrant immune response. The molecular basis of lymphoid organogenesis studied in gene-targeted mice will provide clues to why the synovium is a preferred site for tertiary lymphoid tissue. B cells have a critical role in lymphoid organogenesis. Their contribution to synovial inflammation extends beyond antibody secretion and includes the activation and regulation of effector T cells.

    View details for Web of Science ID 000167432900007

    View details for PubMedID 11094459

    View details for PubMedCentralID PMC128875

  • The role of T cells in rheumatoid arthritis. Archivum immunologiae et therapiae experimentalis Weyand, C. M., Bryl, E., Goronzy, J. J. 2000; 48 (5): 429-435

    Abstract

    In rheumatoid arthritis (RA), T cells infiltrate into the synovial membrane where they initiate and maintain activation of macrophages and synovial fibroblasts, transforming them into tissue-destructive effector cells. The diversity of the disease process and the formation of complex lymphoid microstructures indicate that multiple T cell activation pathways are involved. This model is supported by the association of distinct disease patterns with different variants and combinations of HLA class II molecules. T cell pathology in RA, however, is not limited to the joint. Affected patients have major abnormalities in the T cell pool, with a marked contraction in T cell receptor diversity and an outgrowth of large clonal populations. Clonally expanded CD4+ T cells lose expression of the CD28 molecule and gain expression of perforin and granzyme. Consequently, the functional profile of expanded CD4(+)CD28null T cells is fundamentally changed and is shifted towards tissue-injurious capabilities. CD4(+)CD28null T cells are particularly important in patients with extra-articular manifestations of RA, where they may have a direct role in vascular injury. Understanding the mechanisms underlying the loss of T cell diversity and the emergence of pro-inflammatory CD4(+)CD28null T cell clonotypes may have implications for other autoimmune syndromes.

    View details for PubMedID 11140470

  • Association of MHC and rheumatoid arthritis HLA polymorphisms in phenotypic variants of rheumatoid arthritis ARTHRITIS RESEARCH Weyand, C. M., Goronzy, J. J. 2000; 2 (3): 212-216

    Abstract

    Genes in the human leukocyte antigen (HLA) region remain the most powerful disease risk genes in rheumatoid arthritis (RA). Several allelic variants of HLA-DRB1 genes have been associated with RA, supporting a role for T-cell receptor-HLA-antigen interactions in the pathologic process. Disease-associated HLA-DRB1 alleles are similar but not identical and certain allelic variants are preferentially enriched in patient populations with defined clinical characteristics. Also, a gene dosing effect of HLA-DRB1 alleles has been suggested by the accumulation of patients with two RA-associated alleles, especially in patient subsets with a severe disease course. Therefore, polymorphisms in HLA genes are being explored as tools to dissect the clinical heterogeneity of the rheumatoid syndrome. Besides HLA polymorphisms, other risk genes will be helpful in defining genotypic profiles correlating with disease phenotypes. One such phenotype is the type of synovial lesion generated by the patient. HLA genes in conjunction with other genetic determinants may predispose patients to a certain pathway of synovial inflammation. Also, patients may or may not develop extraarticular manifestations, which are critical in determining morbidity and mortality. HLA genes, complemented by other RA risk genes, are likely involved in shaping the T-cell repertoire, including the emergence of an unusual T-cell population characterized by the potential of vascular injury, such as seen in extraarticular RA.

    View details for Web of Science ID 000167357700010

    View details for PubMedID 11094432

    View details for PubMedCentralID PMC130005

  • Perturbation of the T-cell repertoire in patients with unstable angina CIRCULATION Liuzzo, G., Kopecky, S. L., Frye, R. L., O'Fallon, W. M., Maseri, A., Goronzy, J. J., Weyand, C. M. 1999; 100 (21): 2135-2139

    Abstract

    Monocytes are constitutively activated in unstable angina (UA), resulting in the production of IL-6 and the upregulation of acute phase proteins. Underlying mechanisms are not understood. To explore whether the production of the potent monocyte activator IFN-gamma is altered in UA, we compared cytokine production by T lymphocytes in patients with UA (Braunwald's class IIIB) and with stable angina (SA).Peripheral blood lymphocytes were collected at the time of hospitalization and after 2 and 12 weeks. Cytokine-producing CD4(+) and CD8(+) T cells were quantified by 3-color flow cytometry after stimulation with phorbol myristate acetate and ionomycin. UA was associated with an increased number of CD4(+) and CD8(+) T cells producing IFN-gamma, whereas patients with SA had higher frequencies of IL-2(+) and IL-4(+) CD4(+) T cells. Expansion of the IFN-gamma( +) T-cell population in UA persisted for at least 3 months. Increased production of IFN-gamma in UA could be attributed to the expansion of an unusual subset of T cells, CD4(+)CD28(null) T cells.Patients with UA are characterized by a perturbation of the functional T-cell repertoire with a bias toward IFN-gamma production, suggesting that monocyte activation and acute phase responses are consequences of T-cell activation. IFN-gamma is produced by CD4(+)CD28(null) T cells, which are expanded in UA and distinctly low in SA and controls. The emergence of CD4(+)CD28(null) T cells may result from persistent antigenic stimulation.

    View details for Web of Science ID 000083945000007

    View details for PubMedID 10571971

  • Formation of new vasa vasorum in vasculitis - Production of angiogenic cytokines by multinucleated giant cells AMERICAN JOURNAL OF PATHOLOGY Kaiser, M., Younge, B., Bjornsson, J., Goronzy, J. J., Weyand, C. M. 1999; 155 (3): 765-774

    Abstract

    Inflammation of the arterial wall in giant cell arteritis induces a series of structural changes, including the formation of new vasa vasorum. To study the regulation of neoangiogenesis in giant cell arteritis, temporal arteries were examined for the extent and localization of microvessel generation and for the production of angiogenic factors. In normal arteries, vasa vasorum were restricted to the adventitia, but in inflamed arteries, capillaries emerged in the media and the intima. These capillaries displayed a distinct topography with a circumferential arrangement in the external one-third of the intima. Neovascularization was closely correlated with the formation of lumen-obstructing intima, the fragmentation of the internal elastic lamina, and the presence of multinucleated giant cells. Comparison of tissue cytokine transcription in temporal arteries of giant cell arteritis patients with and without up-regulated neoangiogenesis identified interferon-gamma and vascular endothelial growth factor but not fibroblast growth factor-2 as mediators associated with vasa vasorum proliferation. Giant cells and CD68-positive macrophages at the media-intima junction were found to be the major cellular sources of vascular endothelial growth factor. These data demonstrate that formation of new vasa vasorum in vasculitis is regulated by inflammatory cells and not by arterial wall cells, raising the possibility that it represents a primary disease mechanism and not a secondary hypoxia-induced event. Increased neovascularization in interferon-gamma-rich arteries suggests that the formation of new vasa vasorum is determined by the nature of the immune response in the arterial wall, possibly resulting from the generation and functional activity of multinucleated giant cells.

    View details for Web of Science ID 000082537800013

    View details for PubMedID 10487834

    View details for PubMedCentralID PMC1866901

  • Modulation of CD28 expression: Distinct regulatory pathways during activation and replicative senescence JOURNAL OF IMMUNOLOGY Vallejo, A. N., Brandes, J. C., Weyand, C. M., Goronzy, J. J. 1999; 162 (11): 6572-6579

    Abstract

    The costimulatory molecule CD28 has a restricted tissue distribution and is expressed on T cells and some plasmacytoma cells. Although CD28 is constitutively expressed, its expression is transiently down-regulated following T cell activation and declines progressively with in vitro senescence. In vivo, CD8+ T cells and, less frequently, CD4+ T cells may completely lose CD28 surface expression during chronic infections and with aging. This correlates with changes of nuclear protein-binding activities to two motifs, site alpha and beta, within the CD28 minimal promoter. Both alpha- and beta-bound complexes are found only in lymphoid tissues, in CD28+ T cells, and in some transformed B cells. These complexes are coordinately expressed except during replicative senescence, which is characterized by the down-modulation of site beta- but not site alpha-binding activities. In contrast, T cell activation induces a parallel decline in both site alpha- and beta-binding activities. CD4+ and CD8+ T cells differ in their beta-binding profiles, which may explain the more pronounced down-regulation of CD28 in senescent CD8+ T cells. In vivo expanded CD4+CD28null and CD8+CD28null T cells uniformly lack alpha- and beta-bound complexes, resembling the pattern seen in chronically activated cells and not of senescent cells.

    View details for Web of Science ID 000080444100035

    View details for PubMedID 10352273

  • Tissue-destructive macrophages in giant cell arteritis CIRCULATION RESEARCH Rittner, H. L., Kaiser, M., Brack, A., Szweda, L. I., Goronzy, J. J., Weyand, C. M. 1999; 84 (9): 1050-1058

    Abstract

    Giant cell arteritis (GCA) is an inflammatory vasculopathy in which T cells and macrophages infiltrate the wall of medium and large arteries. Clinical consequences such as blindness and stroke are related to arterial occlusion. Formation of aortic aneurysms may result from necrosis of smooth muscle cells and fragmentation of elastic membranes. The molecular mechanisms of arterial wall injury in GCA are not understood. To identify mechanisms of arterial damage, gene expression in inflamed and unaffected temporal artery specimens was compared by differential display polymerase chain reaction. Genes differentially expressed in arterial lesions included 3 products encoded by the mitochondrial genome. Immunohistochemistry with antibodies specific for a 65-kDa mitochondrial antigen revealed that increased expression of mitochondrial products was characteristic of multinucleated giant cells and of CD68+ macrophages that cluster in the media and at the media-intima junction. 4-Hydroxy-2-nonenal adducts, products of lipid peroxidation, were detected on smooth muscle cells and on tissue infiltrating cells, in close proximity to multinucleated giant cells and CD68+ macrophages. Also, giant cells and macrophages with overexpression of mitochondrial products were able to synthesize metalloproteinase-2. Our data suggest that in the vascular lesions characteristic for GCA, a subset of macrophages has the potential to support several pathways of arterial injury, including the release of reactive oxygen species and the production of metalloproteinase-2. This macrophage subset is topographically defined and is also identified by overexpression of mitochondrial genes. Because these macrophages have a high potential to promote several mechanisms of arterial wall damage, they should be therapeutically targeted to prevent blood vessel destruction.

    View details for Web of Science ID 000080506800009

    View details for PubMedID 10325242

  • T-cell responses in rheumatoid arthritis: systemic abnormalities-local disease. Current opinion in rheumatology Weyand, C. M., Goronzy, J. J. 1999; 11 (3): 210-217

    Abstract

    One manifestation of rheumatoid arthritis (RA) is a destructive inflammation of the joint, but many other organs can be targeted by this disease, classifying it as a truly systemic disorder. Accordingly, pathogenic models have to account for the multiorgan character of RA. This article proposes that the primary abnormalities in RA lie in the assembly of the T-cell pool and in the maintenance of T-cell homeostasis. Evidence has accumulated that the repertoire of CD4 T cells in RA patients is distinct and includes a high frequency of disease-relevant T cells. Emergence of T cells with self-aggressive potential could indicate a failure of negative selection in the thymus. Also, the turnover of mature T cells in the periphery is altered in RA patients with a sharp contraction in diversity. Loss of diversity results from the replacement of rare T-cell specificities by multiplying T-cell clones. Large clonal T-cell populations in RA patients acquire a distinct phenotype (CD4+CD28null) and functional profile (overproduction of interferon-gamma, cytotoxicity), giving them the ability to function as proinflammatory cells. Optimal conditions for T-cell stimulation are encountered in the synovium, where ectopic lymphoid tissue with germinal centers is formed. Considering the systemic nature of RA, therapeutic strategies suppressing synovial inflammation while ignoring systemic abnormalities could lack the potential of a curative intervention.

    View details for PubMedID 10328581

  • Arterial wall injury in giant cell arteritis ARTHRITIS AND RHEUMATISM Weyand, C. M., Goronzy, J. J. 1999; 42 (5): 844-853

    View details for Web of Science ID 000080140400002

    View details for PubMedID 10323439

  • Aberrations in the primary T-cell receptor repertoire as a predisposition for synovial inflammation in rheumatoid arthritis JOURNAL OF INVESTIGATIVE MEDICINE Yang, H. Y., Rittner, H., Weyand, C. M., Goronzy, J. J. 1999; 47 (5): 236-245

    Abstract

    Rheumatoid arthritis (RA) is an HLA-DR associated disease with a pivotal role of T cells in the pathogenesis. The mechanisms underlying the HLA association and the generation of a synovial T-cell response are unclear. We have hypothesized that the selection of the primary T-cell repertoire is a predisposing factor for rheumatoid synovitis.The repertoire of T-cell receptors (TCR) expressed by circulating naive CD4+ CD45RO- T cells was compared in 10 patients with RA, 11 HLA-DR matched normal donors and 10 mismatched normal donors by determining the frequencies of TCR BV-BJ combinations in 3 different BV gene segment families. Clonally expanded synovium-specific CD4 T cells were identified in 8 patients by TCR BV-BJ-specific PCR of purified T-cell subsets followed by size fractionation and sequencing of the PCR product. The TCR BV-BJ repertoires of naive peripheral T cells and of synovial clones were compared.The repertoires of naive circulating CD4+ CD45RO- T cells were different in RA patients and in HLA-DR matched and unmatched controls, suggesting HLA-DR as well as disease-specific features of T-cell selection. To test the disease relevance of the shifts in the naive repertoire, CD4 T cells undergoing joint-specific clonal expansion were identified. The usage of BV-BJ gene combinations in these synovium-specific clones was biased and significantly different from the expected distribution with a preference for combinations favored in the naive TCR repertoire of RA patients.These data suggest that primary T-cell selection in RA patients is of functional importance for the generation of synovium-specific T-cell responses. The synovial repertoire is influenced by aberrations in the naive T-cell repertoire that might indicate a defect in thymic education with the selection of high-affinity self-reactive T cells in RA.

    View details for Web of Science ID 000080491900009

    View details for PubMedID 10361384

  • IL-16 as an anti-inflammatory cytokine in rheumatoid synovitis JOURNAL OF IMMUNOLOGY Klimiuk, P. A., Goronzy, J. J., Weyand, C. M. 1999; 162 (7): 4293-4299

    Abstract

    T lymphocytes are a major component of the inflammatory infiltrate in rheumatoid synovitis, but their exact role in the disease process is not understood. Functional activities of synovial T cells were examined by adoptive transfer experiments in human synovium-SCID mouse chimeras. Adoptive transfer of tissue-derived autologous CD8+ T cells induced a marked reduction in the activity of lesional T cells and macrophages. Injection of CD8+, but not CD4+, T cells decreased the production of tissue IFN-gamma, IL-1beta, and TNF-alpha by >90%. The down-regulatory effect of adoptively transferred CD8+ T cells was not associated with depletion of synovial CD3+ T cells or synovial CD68+ macrophages, and it could be blocked by Abs against IL-16, a CD8+ T cell-derived cytokine. In the synovial tissue, CD8+ T cells were the major source of IL-16, a natural ligand of the CD4 molecule that can anergize CD4-expressing cells. The anti-inflammatory activity of IL-16 in rheumatoid synovitis was confirmed by treating synovium-SCID mouse chimeras with IL-16. Therapy for 14 days with recombinant human IL-16 significantly inhibited the production of IFN-gamma, IL-1beta, and TNF-alpha in the synovium. We propose that tissue-infiltrating CD8+ T cells in rheumatoid synovitis have anti-inflammatory activity that is at least partially mediated by the release of IL-16. Spontaneous production of IL-16 in synovial lesions impairs the functional activity of CD4+ T cells but is insufficient to completely abrogate their stimulation. Supplemental therapy with IL-16 may be a novel and effective treatment for rheumatoid arthritis.

    View details for Web of Science ID 000079278000075

    View details for PubMedID 10201961

  • Aldose reductase functions as a detoxification system for lipid peroxidation products in vasculitis JOURNAL OF CLINICAL INVESTIGATION Rittner, H. L., Hafner, V., Klimiuk, P. A., Szweda, L. I., Goronzy, J. J., Weyand, C. M. 1999; 103 (7): 1007-1013

    Abstract

    Giant cell arteritis (GCA) is a systemic vasculitis preferentially affecting large and medium-sized arteries. Inflammatory infiltrates in the arterial wall induce luminal occlusion with subsequent ischemia and degradation of the elastic membranes, allowing aneurysm formation. To identify pathways relevant to the disease process, differential display-PCR was used. The enzyme aldose reductase (AR), which is implicated in the regulation of tissue osmolarity, was found to be upregulated in the arteritic lesions. Upregulated AR expression was limited to areas of tissue destruction in inflamed arteries, where it was detected in T cells, macrophages, and smooth muscle cells. The production of AR was highly correlated with the presence of 4-hydroxynonenal (HNE), a toxic aldehyde and downstream product of lipid peroxidation. In vitro exposure of mononuclear cells to HNE was sufficient to induce AR production. The in vivo relationship of AR and HNE was explored by treating human GCA temporal artery-severe combined immunodeficiency (SCID) mouse chimeras with the AR inhibitors Sorbinil and Zopolrestat. Inhibition of AR increased HNE adducts twofold and the number of apoptotic cells in the arterial wall threefold. These data demonstrate that AR has a tissue-protective function by preventing damage from lipid peroxidation. We propose that AR is an oxidative defense mechanism able to neutralize the toxic effects of lipid peroxidation and has a role in limiting the arterial wall injury mediated by reactive oxygen species.

    View details for Web of Science ID 000079633400010

    View details for PubMedID 10194473

    View details for PubMedCentralID PMC408253

  • Corticosteroid requirements in polymyalgia rheumatica ARCHIVES OF INTERNAL MEDICINE Weyand, C. M., Fulbright, J. W., Evans, J. M., Hunder, G. G., Goronzy, J. J. 1999; 159 (6): 577-584

    Abstract

    Polymyalgia rheumatica (PMR) is a systemic inflammatory disease of unknown cause that affects older individuals. Clinical symptoms respond promptly to corticosteroids, but treatment is often required for several years, frequently resulting in adverse drug effects. Guidelines for the optimal use of corticosteroids that maximize relief of symptoms but minimize adverse effects of the therapy are needed.To determine whether clinical or laboratory parameters in PMR could be identified that allow for stratifying patients into subsets with differences in corticosteroid requirements.We studied 27 patients with PMR treated with a standardized schedule of prednisone. Patients were reevaluated at monthly intervals for pain scores and physician and patient assessments. Plasma interleukin 6 level and the erythrocyte sedimentation rate were measured at each visit. The duration of steroid therapy and the cumulative steroid dose were calculated.Based on the initial response to therapy and the duration of disease, the 27 patients could be subdivided into 3 distinct groups. Eight with low erythrocyte sedimentation rates responded rapidly and required corticosteroids for less than 1 year with rare disease flares on tapering of prednisone. Twelve others responded well initially but did not tolerate reduction to lower doses and had remitting disease of more than 1 year. Seven patients had only a partial response to the initial steroid regimen. After 4 weeks of therapy, the erythrocyte sedimentation rates improved, but levels of interleukin 6 remained elevated. Pretreatment pain scores were also higher in these partial responder patients (P = .05).Polymyalgia rheumatica is a heterogeneous disease with variations in the treatment duration and dose of corticosteroids required for suppression of symptoms. Pretreatment erythrocyte sedimentation rate and nonresponsiveness of interleukin 6 to steroid therapy are helpful in dividing patients into subsets with different treatment requirements.

    View details for Web of Science ID 000079234200007

    View details for PubMedID 10090114

  • Disease pattern in cranial and large-vessel giant cell arteritis ARTHRITIS AND RHEUMATISM Brack, A., Martinez-Taboada, V., Stanson, A., Goronzy, J. J., Weyand, C. M. 1999; 42 (2): 311-317

    Abstract

    To identify variables that distinguish large-vessel giant cell arteritis (GCA) with subclavian/axillary/brachial artery involvement from cranial GCA.Seventy-four case patients with subclavian/axillary GCA diagnosed by angiography and 74 control patients with temporal artery biopsy-proven GCA without large vessel involvement matched for the date of first diagnosis were identified. Pertinent initial symptoms, time delay until diagnosis, and clinical symptoms, as well as clinical and laboratory findings at the time of diagnosis, were recorded by retrospective chart review. Expression of cytokine messenger RNA in temporal artery tissue from patients with large-vessel and cranial GCA was determined by semiquantitative polymerase chain reaction analysis. Distribution of disease-associated HLA-DRB1 alleles in patients with aortic arch syndrome and cranial GCA was assessed.The clinical presentation distinguished patients with large-vessel GCA from those with classic cranial GCA. Upper extremity vascular insufficiency dominated the clinical presentation of patients with large-vessel GCA, whereas symptoms related to impaired cranial blood flow were infrequent. Temporal artery biopsy findings were negative in 42% of patients with large-vessel GCA. Polymyalgia rheumatica occurred with similar frequency in both patient groups. Large-vessel GCA was associated with higher concentrations of interleukin-2 gene transcripts in arterial tissue and overrepresentation of the HLA-DRB1*0404 allele, indicating differences in pathogenetic mechanisms.GCA is not a single entity but includes several variants of disease. Large-vessel GCA produces a distinct spectrum of clinical manifestations and often occurs without involvement of the cranial arteries. Large-vessel GCA requires a different approach to the diagnosis and probably also to treatment.

    View details for Web of Science ID 000078469000014

    View details for PubMedID 10025926

  • HLA polymorphisms and T cells in rheumatoid arthritis. International reviews of immunology Weyand, C. M., Goronzy, J. J. 1999; 18 (1-2): 37-59

    Abstract

    A dense infiltrate of activated T cells, macrophages, and B cells in the synovial membrane is the cardinal pathological feature of rheumatoid arthritis (RA). Frequently, tissue infiltrating cells acquire a morphological organization reminiscent of secondary lymphoid tissue. The composition of the inflammatory lesions, the production of autoantibodies, and the association of disease risk with genes related to the HLA-D region have all been cited as evidence for a critical role of T cells in disease pathogenesis. Investigations on the precise role of HLA genes in RA have confirmed the importance of this genetic risk factor and have identified a consensus sequence within the HLA-DRBI genes. The observation that HLA polymorphisms are mostly associated with disease progression and severity and that a gene dose effect for HLA-DR genes is operational has challenged the simple model that HLA molecules select and present an arthritogenic antigen. Studies analyzing the repertoire of tissue infiltrating T cells have not been able to identify a dominant and common disease relevant T cell. The infiltrate is diverse in terms of T cell receptor gene usage but consistently includes clonally expanded populations. Recent evidence indicates that RA patients carry expanded CD4 clonotypes which are characterized by deficient CD28 expression and autoreactivity. These autoreactive CD4 T cells are not restricted to the joint, raising the possibility that rheumatoid synovitis is a manifestation of a systemic autoimmune disease. Support for this model has come from studies in T cell receptor (TCR) transgenic animals which develop inflammation of the synovial membrane stimulated by a T cell response to ubiquitously expressed self-MHC molecules. Antigens driving the chronic persistent immune response in RA may not be restricted to the joint but rather may be widely distributed, providing an explanation for the difficulties in identifying arthritogenic antigens directly or indirectly through the selection of joint infiltrating T cells.

    View details for PubMedID 10614738

  • Results of a phase II rheumatoid arthritis clinical trial using T-cell receptor peptides International Symposium on Advances in Osteoarthritis Brostoff, S. W., Carlo, D. J., Diveley, J. P., Morgan, E. E., Nardo, C. J., Richieri, S. P., Adamson, T. C., Fronek, Z., Calabrese, L. H., Cash, J. M., Markenson, J. A., Bathon, J., Matsumoto, A. K., Matteson, E. L., Uramoto, K. M., Weyand, C. M., Strand, V., Heck, L. W., Koopman, W. J., Moreland, L. W. SPRINGER-VERLAG TOKYO. 1999: 153–162
  • Production of cytokines and metalloproteinases in rheumatoid synovitis is T cell dependent CLINICAL IMMUNOLOGY Klimiuk, P. A., Yang, H. Y., Goronzy, J. J., Weyand, C. M. 1999; 90 (1): 65-78

    Abstract

    In rheumatoid arthritis, T lymphocytes have been proposed to play a pivotal role in the disease process, but they have also been considered to simply represent an epiphenomenon in a primarily synoviocyte/macrophage-driven disease. To directly examine the contribution of CD4 T cells in synovitis, T cells were either depleted from or adoptively transferred into NOD-SCID mice engrafted with rheumatoid synovial tissue. Injection of anti-CD2 antibody resulted in the elimination of 80-90% of tissue-infiltrating T cells in the synovial grafts and was followed by a marked decline in the production of IL-1beta (loss of 70%), TNF-alpha (loss of 86%), and IL-15 (loss of 84%) mRNA. Also, transcription of MMP-1 and MMP-2 was reduced by 72% in anti-CD2-treated chimeras. Immunohistochemistry demonstrated that the cytokines and proteases derived mostly from CD68(+) synovial cells, which disappeared from the tissue upon T cell depletion. Adoptive transfer of autologous tissue-derived T cell lines and T cell clones into synovium-SCID mouse chimeras augmented the production of IFN-gamma as well as TNF-alpha in the synovial infiltrates. Administration of IFN-gamma in small doses to anti-CD2-treated chimeras restored the survival and the functional activity of CD68(+) synovial cells. In vitro studies confirmed the critical role of synovial T cells and IFN-gamma in the survival of synovial CD68(+) cells. These data demonstrate that the production of proinflammatory cytokines and of tissue-degrading enzymes in rheumatoid synovitis is T cell dependent and that CD4 T cells are primary regulatory cells in this disease.

    View details for Web of Science ID 000079158600009

    View details for PubMedID 9884354

  • The role of CD8(+) CD40L(+) T cells in the formation of germinal centers in rheumatoid synovitis JOURNAL OF IMMUNOLOGY Wagner, U. G., Kurtin, P. J., Wahner, A., Brackertz, M., Berry, D. J., Goronzy, J. J., Weyand, C. M. 1998; 161 (11): 6390-6397

    Abstract

    In rheumatoid synovitis, lymphocytes can be arranged in follicular structures resembling secondary lymphoid follicles. To understand the organizing principles of this ectopic lymphoid tissue, the cellular components contributing to synovial follicles were examined. In 9 of 24 synovial tissue biopsies, lymphoid aggregates were found consisting of CD4+ T cells and CD20+ B cells. In four of the nine patients, the follicular centers were occupied by CD23+ CD21+ cellular networks representing follicular dendritic cells involved in germinal center reactions. In five patients, CD23+ cells were absent from the centers of the aggregates, suggesting that fully developed germinal centers are generated in only a subset of patients. To identify factors involved in the regulation of the synovial microarchitecture, cell populations contributing to the follicles were quantified by digital image analysis of immunostained tissue and by flow cytometry of tissue-derived lymphocytes. Proportions of CD4+, CD20+, and CD68+ cell subsets were surprisingly invariant, irrespective of the presence or absence of CD23+ follicular dendritic cells. Instead, tissue biopsies with CD23+ germinal center-like regions could be distinguished from those with CD23- T cell-B cell aggregates by a fourfold increase in the frequency of tissue-infiltrating CD8+ T cells, a fraction of which expressed CD40 ligand (CD40L). The data suggest a previously unsuspected role of CD8+ lymphocytes in modulating germinal center formation and raise the possibility that CD8+ CD40L+ T cells are involved in aggravating pathologic immune responses in rheumatoid synovitis.

    View details for Web of Science ID 000077206900079

    View details for PubMedID 9834130

  • Functional subsets of CD4 T cells in rheumatoid synovitis ARTHRITIS AND RHEUMATISM NAMEKAWA, T., Wagner, U. G., Goronzy, J. J., Weyand, C. M. 1998; 41 (12): 2108-2116

    Abstract

    To identify the functional properties of CD4+ CD28- T cells, which accumulate and clonally expand in patients with rheumatoid arthritis (RA).The gene expression of molecules involved in T cell effector functions was compared in CD4+ CD28- and CD4+ CD28+ T cell clones. The expression of differentially up-regulated genes was confirmed by flow cytometry of T cells and by 2-color immunohistochemistry of rheumatoid synovial tissue. Cytotoxicity of CD4+ CD28- T cells was tested by anti-CD3 redirected lysis of Fc receptor-positive target cells.CD4+ CD28- T cell clones lacked messenger RNA for the CD40 ligand (CD40L) but transcribed the perforin gene. Perforin was also found in freshly isolated CD4+ CD28- peripheral blood lymphocytes from RA patients. CD4+ CD28-, but not CD4+ CD28+, T cell clones lysed Fc receptor-bearing target cells. CD4+ perforin-positive T cells were present in the synovial tissue, where their frequency correlated with the expansion of the CD4+ CD28- compartment in the periphery. Among tissue-infiltrating CD4+ T cells, only the CD40L-negative subset expressed perforin transcripts.Clonally expanded CD4+ CD28- T cells are functionally specialized for killing, while they lack the ability to provide B cell help. Tissue-infiltrating CD4+ T cells can be subdivided phenotypically and functionally into at least 2 distinct subsets based on their expression of perforin and CD40L. Because the expansion of CD4+ CD28- T cells is associated with extraarticular RA, T cell-mediated cytotoxicity may be particularly important in these most severe complications of RA.

    View details for Web of Science ID 000077616700004

    View details for PubMedID 9870867

  • Perturbation of the T cell repertoire in rheumatoid arthritis PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Wagner, U. G., Koetz, K., Weyand, C. M., Goronzy, J. J. 1998; 95 (24): 14447-14452

    Abstract

    Aberrations in the T cell repertoire with the emergence of oligoclonal populations have been described in patients with rheumatoid arthritis (RA). However, the extent of the repertoire perturbations as well as the underlying mechanisms are not known. We now have examined the diversity of the peripheral CD4 T cell repertoire by determining the frequencies of arbitrarily selected T cell receptor (TCR) beta-chain sequences. Healthy individuals displayed a highly diverse repertoire, with a median frequency of individual TCR beta-chain sequences of 1 in 2.4 x 10(7) CD4 T cells. In RA patients, the median TCR beta-chain frequency was increased 10-fold, indicating marked contraction of the repertoire (P < 0.001). The loss in TCR diversity was not limited to CD4 memory T cells but also involved the compartment of naive T cells, suggesting that it reflected an abnormality in T cell repertoire formation and not a consequence of antigen recognition in the synovium. Also, control patients with chronic inflammatory disease such as hepatitis C expressed a diverse repertoire indistinguishable from that of normals. Telomere length studies indicated an increased replicative history of peripheral CD4 T cells in RA patients, suggesting an enhanced turnover within the CD4 compartment. Compared with age-matched controls, terminal restriction fragment sizes were 1.7 kilobases shorter (P < 0.001). These data demonstrate an altered CD4 T cell homeostasis in RA that may contribute to the autoimmune response as well as to the immunodeficiency in these patients.

    View details for Web of Science ID 000077191200082

    View details for PubMedID 9826720

    View details for PubMedCentralID PMC24393

  • T cell receptor peptide vaccination in rheumatoid arthritis - A placebo-controlled trial using a combination of V(beta)3, V(beta)14, and V(beta)17 peptides ARTHRITIS AND RHEUMATISM Moreland, L. W., Morgan, E. E., Adamson, T. C., Fronek, Z., Calabrese, L. H., Cash, J. M., Markenson, J. A., Matsumoto, A. K., Bathon, J., Matteson, E. L., Uramoto, K. M., Weyand, C. M., Koopman, W. J., Heck, L. W., Strand, V., Diveley, J. P., Carlo, D. J., Nardo, C. J., Richieri, S. P., Brostoff, S. W. 1998; 41 (11): 1919-1929

    Abstract

    Restricted T cell receptor (TCR) gene usage has been demonstrated in animal models of autoimmune disease and has resulted in the successful use of TCR peptide therapy in animal studies. This clinical trial was undertaken to determine the safety and efficacy of a combination of Vbeta3, Vbeta14, and Vbeta17 TCR peptides in Freund's incomplete adjuvant (IFA) in patients with rheumatoid arthritis (RA).A double-blind, placebo-controlled, multicenter, phase II clinical trial was undertaken using IR501 therapeutic vaccine, which consists of a combination of 3 peptides derived from TCRs (Vbeta3, Vbeta14, and Vbeta17) in IFA. A total of 99 patients with active RA received either 90 microg (n = 31) or 300 microg (n = 35) of IR501 or IFA alone (n = 33) as a control. The study medication and placebo were administered as a single intramuscular injection (1 ml) at weeks 0, 4, 8, and 20.Treatment with IR501 was safe and well tolerated. None of the patients discontinued the trial because of treatment-related adverse events. Efficacy was measured according to the American College of Rheumatology 20% improvement criteria. Using these criteria, patients in both IR501 dosage groups showed improvement in disease activity. In the most conservative analysis used to evaluate efficacy, an intent-to-treat analysis including all patients who enrolled, the 90-microg dosage group showed a statistically significant improvement compared with control patients at the 20-week time point after the third injection. Trends toward improvement were shown in both the 90-microg and the 300-microg dosage groups at week 24 after the fourth injection.IR501 therapeutic vaccine therapy was safe and well tolerated, immunogenic, and demonstrated clinical improvement in RA patients. Additional clinical trials are planned to confirm and extend these observations.

    View details for Web of Science ID 000076781400004

    View details for PubMedID 9811045

  • Giant cell arteritis - a molecular approach to the multiple facets of the syndrome ANNALES DE MEDECINE INTERNE Weyand, C. M., Hafner, V., Kaiser, M., Goronzy, J. J. 1998; 149 (7): 420-424

    Abstract

    Molecular studies of giant cell arteritis indicate that T cells are recruited to the wall of medium-sized and large arteries, are activated locally, produce IL-2 and IFN-gamma, and regulate the activity of tissue-infiltrating macrophages. Downstream effects of T cell activation include the production of proinflammatory cytokines, metalloproteinases, and growth factors. Growth factors are instrumental in the process of intimal hyperplasia, leading to luminal occlusion and tissue ischemia. The amounts of IL-2, IFN-gamma, and the growth factor PDGF in the vascular lesions varies among patients and are correlated with differences in patterns of clinical manifestations. Giant cell arteritis complicated by cranial ischemia, such as anterior optic neuropathy or stroke, is characterized by high levels of IFN-gamma and PDGF. If the IFN-gamma-PDGF loop is less developed, fever and wasting can dominate the disease. Dominant production of IL-2 is associated with polymyalgia rheumatica. The finding of different inflammatory pathways translating into different clinical phenotypes may reflect differences in the contribution of the arterial wall. Alternative hypotheses include a role of multiple disease-inducing antigens with different tissue distributions or tropisms.

    View details for Web of Science ID 000077942300005

    View details for PubMedID 9921395

  • Interferon-alpha treatment of Behcet's disease JOURNAL OF RHEUMATOLOGY O'Duffy, J. D., Calamia, K., Cohen, S., Goronzy, J. J., Herman, D., Jorizzo, J., WEYAND, C., Matteson, E. 1998; 25 (10): 1938-1944

    Abstract

    To determine if interferon-alpha (IFN-alpha) is effective in Behçet's disease (BD).Ten patients having active BD were entered into a 6 month open label trial of IFN-alpha therapy given by self-administered dose of 3 million units subcutaneously daily. Provision was made for downward or upward dosing depending on predetermined responses. Four study centers followed the same protocol. Patients having ocular or neurologic inflammation at onset of the study were excluded.Seven patients completed the trial, while 3 were removed at 3 and 4 months for side effects or lack of efficacy. There was a substantial reduction in the number of oral and genital ulcers, cutaneous lesions, and articular signs and symptoms by the end of the study. Pain scores, global assessments, and prednisone dose also declined. The IFN-alpha dose at end of study averaged 1.5 million units. Significant side effects, besides the anticipated flu-like syndrome experienced by all the patients, included single instances of seizure, psychosis, psoriasis, and hyperthyroidism.The results suggest that IFN-alpha is effective therapy for some manifestations of Behçet's disease.

    View details for Web of Science ID 000076212600012

    View details for PubMedID 9779847

  • Resistance to apoptosis and elevated expression of Bcl-2 in clonally expanded CD4(+)CD28(-) T cells from rheumatoid arthritis patients JOURNAL OF IMMUNOLOGY Schirmer, M., Vallejo, A. N., Weyand, C. M., Goronzy, J. J. 1998; 161 (2): 1018-1025

    Abstract

    Patients with rheumatoid arthritis have a subset of CD4+ T lymphocytes that are characterized by a defect in CD28 expression. CD4+CD28- T cells frequently undergo clonal expansion in vivo. These clonotypes include autoreactive cells and persist over many years. The clonogenic potential and longevity of these T cells could be related to an altered response to apoptosis-inducing signals. To explore this possibility, CD4+CD28- T cell lines and clones were examined for their response pattern to stimuli inducing physiologic cell death. CD4+CD28- T cells were found to be resistant to apoptosis upon withdrawal of the growth factor, IL-2. To examine whether the altered sensitivity to this apoptotic signal was correlated with the expression of proteins of the bcl-2 family, the expression of bcl-2, bcl-x, and bax proteins was determined. CD28+ and CD28-CD4+ T cells could not be distinguished by the levels of bax or bcl-xL protein; however, CD4+CD28- T cells expressed higher amounts of bcl-2 protein than did CD4+CD28+ T cells. The increased bcl-2 expression in CD4+CD28- T cells was relatively independent of signals provided by exogenous IL-2. In CD28-deficient CD4+ T cells, bcl-2 was not significantly up-regulated by the addition of exogenous IL-2 and was maintained despite IL-2 withdrawal, as opposed to CD28-expressing CD4+ T cells. We propose that CD4+CD28- T cells are characterized by a dysregulation of the survival protein, bcl-2, which may favor the clonal outgrowth of autoreactive T cells and thus contribute to the pathogenesis of rheumatoid arthritis.

    View details for Web of Science ID 000074728400060

    View details for PubMedID 9670983

  • Oligoclonal T-cell proliferation and interferon-gamma production in periprosthetic inflammation LABORATORY INVESTIGATION Weyand, C. M., Geisler, A., Brack, A., Bolander, M. E., Goronzy, J. J. 1998; 78 (6): 677-685

    Abstract

    Total joint arthroplasty has dramatically changed the treatment options for patients with destructive joint disease. The materials used to manufacture implants are regarded as biologically inert; accordingly, arthroplasty is a very successful intervention for most patients. However, a subset of patients develops an inflammatory reaction around the prosthesis, causing implant loosening and irreversible bone destruction. To identify mechanisms leading to periprosthetic inflammation, the function and composition of macrophages and T cells accumulated in the pseudosynovia were examined. Tissue-infiltrating macrophages synthesized a spectrum of proinflammatory cytokines including IL-1beta, IL-6, and TGF-beta. T cells recruited to the periprosthetic inflammatory lesions were characterized by restricted diversity of T-cell receptors and the emergence of dominant clonal populations. T cells with identical T-cell receptor sequences, and thus with identical antigen specificity, were isolated from anatomically distinct and independent regions of the tissue. Transcription of IL-2, IFN-gamma, and, in some patients, IL-4 genes in the periprosthetic membrane indicated functional activation of infiltrating T cells. Correlation of periprosthetic osteolysis with the tissue cytokine pattern demonstrated a relationship between IFN-gamma transcription and bone loss. We propose that antigen-recognition events are critically involved in the development of periprosthetic inflammation and that the functional commitment of T cells recruited to the periprosthetic region influences whether periprosthetic inflammation is complicated by bone destruction.

    View details for Web of Science ID 000074283100004

    View details for PubMedID 9645758

  • Functional properties of CD4(+)CD28(-) T cells in the aging immune system 5th EUCAMBIS Congress on the Molecular Biology of Immunosenescence Weyand, C. M., Brandes, J. C., Schmidt, D., Fulbright, J. W., Goronzy, J. J. ELSEVIER IRELAND LTD. 1998: 131–47

    Abstract

    The aging immune system is characterized by a progressive decline in the responsiveness to exogenous antigens and tumors in combination with a paradoxical increase in autoimmunity. From a clinical viewpoint, deficiencies in antibody responses to exogenous antigens, such as vaccines, have a major impact and may reflect intrinsic B cell defects or altered performance of helper T cells. Here we describe that aging is associated with the emergence of an unusual CD4 T cell subset characterized by the loss of CD28 expression. CD28 is the major costimulatory molecule required to complement signaling through the antigen receptor for complete T cell activation. CD4+ CD28- T cells are long-lived, typically undergo clonal expansion in vivo, and react to autoantigens in vitro. Despite the deficiency of CD28, these unusual T cells remain functionally active and produce high concentrations of interferon-gamma (IFN-gamma) and interleukin-2 (IL-2). The loss of CD28 expression is correlated with a lack of CD40 ligand expression rendering these CD4 T cells incapable of promoting B cell differentiation and immunoglobulin secretion. Aging-related accumulation of CD4+ CD28- T cells should result in an immune compartment skewed towards autoreactive responses and away from the generation of high-affinity B cell responses against exogenous antigens. We propose that the emergence of CD28-deficient CD4 T cells in the elderly can partially explain age-specific aberrations in immune responsiveness.

    View details for Web of Science ID 000074845900002

    View details for PubMedID 9720647

  • The influence of sex on the phenotype of rheumatoid arthritis ARTHRITIS AND RHEUMATISM Weyand, C. M., Schmidt, D., Wagner, U., Goronzy, J. J. 1998; 41 (5): 817-822

    Abstract

    To explore whether there are different disease patterns of rheumatoid arthritis (RA) in women and men.We studied 55 male case patients and 110 female control patients who developed RA between 1970 and 1985 and who resided and received medical care in Olmsted County, Minnesota, for at least 10 years after the diagnosis of RA. Case and control patients were matched for the date of first diagnosis. The pattern and extent of joint involvement, the frequency of joint surgeries, and the presence and type of extraarticular manifestations were determined by retrospective chart review.Incidence rates in women were variable and age dependent, whereas the risk in men older than 36 years was constant over their lifetime. Erosive disease was more frequent in men than in women (72% versus 55%, respectively; P < 0.05) and tended to occur earlier (47% versus 31% for erosive disease within the first 4 years of RA). Although male sex was correlated with a higher risk of bony erosions and an accelerated course of RA, structural consequences of joint destruction were more pronounced in women. Joint surgery was performed more frequently in women (50%) than in men (27%) (P = 0.01). In particular, the frequencies of arthroplasties and arthrodeses of hand and foot joints were different (34 procedures in women versus 1 procedure in men; P < 0.001). Sex influenced the risk as well as the pattern of organ involvement in RA. Nodules and rheumatoid lung disease were typical manifestations in men (P = 0.001 and P < 0.001, respectively), whereas women typically developed sicca syndrome (P = 0.05). Despite differences in disease aggressiveness and disease pattern, there was little difference in the medical therapy in the men compared with the women.RA is a heterogeneous disease with variations in phenotype. Sex-associated factors influence disease severity as well as disease pattern. Because sex-related effects influence treatment goals, treatment responses, and side effects, they should be considered in clinical study design and analysis as well as in the treatment decisions for individual patients with RA.

    View details for Web of Science ID 000073413600006

    View details for PubMedID 9588732

  • Histologic and cytokine response to immunosuppression in giant-cell myocarditis ANNALS OF INTERNAL MEDICINE Levy, N. T., Olson, L. J., WEYAND, C., Brack, A., Tazelaar, H. D., EDWARDS, W. D., Hammill, S. C. 1998; 128 (8): 648-650

    View details for Web of Science ID 000073052500005

    View details for PubMedID 9537938

  • Aging-related deficiency of CD28 expression in CD4+ T cells is associated with the loss of gene-specific nuclear factor binding activity JOURNAL OF BIOLOGICAL CHEMISTRY Vallejo, A. N., Nestel, A. R., Schirmer, M., Weyand, C. M., Goronzy, J. J. 1998; 273 (14): 8119-8129

    Abstract

    Changes in T cell populations and concomitant perturbation of T cell effector functions have been postulated to account for many aging-related immune dysfunctions. Here, we report that high frequencies of CD28(null) CD4+ T cells were found in elderly individuals. Because deviations in the function of these unusual CD4+ T cells might be directly related to CD28 deficiency, we examined the molecular basis for the loss of CD28 expression in CD4+ T cells. In reporter gene bioassays, the minimal promoter of the CD28 gene was mapped to the proximal 400 base pairs (bp) of the 5' untranslated region. CD28 deficiency was associated with the loss of two noncompeting binding activities within a 67-bp segment of the minimal promoter. These binding activities were not competed by consensus Ets, Elk, or AP3 motifs that were found within the sequence stretch. The DNA-protein complexes were also not recognized by antibodies to Ets-related transcription factors. Furthermore, introduction of mutations into the 67-bp segment at positions corresponding to the two DNA-protein interaction sites, i.e. nucleotides spanning -206 to -179 and -171 to -148, resulted in the loss of specific nuclear factor binding activities and the abrogation of promoter activity. These observations implicate at least two regulatory motifs in the constitutive expression of CD28. The loss of binding activity of trans-acting factors specific for these sequences may contribute to the accumulation CD4+CD28(null) T cells during aging.

    View details for Web of Science ID 000072876300049

    View details for PubMedID 9525915

  • Platelet-derived growth factor, intimal hyperplasia, and ischemic complications in giant cell arteritis ARTHRITIS AND RHEUMATISM Kaiser, M., Weyand, C. M., Bjornsson, J., Goronzy, J. J. 1998; 41 (4): 623-633

    Abstract

    To explore whether vasoocclusion in giant cell (temporal) arteritis (GCA) is related to intimal hyperplasia and in situ production of platelet-derived growth factor (PDGF).Temporal artery biopsy specimens from patients with GCA were analyzed for the presence of intimal hyperplasia. Expression of PDGF-A and PDGF-B was assessed by immunohistochemistry and digitized image analysis.PDGF-A and PDGF-B were widely expressed in inflamed arteries. CD68+ macrophages, smooth muscle cells, and multinucleated giant cells produced PDGF, whereas hyperplastic intimal tissue did not. Arteries with marked luminal narrowing and those with no or minimal luminal narrowing differed in the extent and distribution of PDGF expression. Concentric intimal hyperplasia was associated with the accumulation of PDGF-A- and PDGF-B-producing CD68+ macrophages at the media-intima junction. PDGF+,CD68+ macrophages in close proximity to the internal elastic lamina frequently coproduced matrix metalloproteinase 2. Intimal hyperplasia of the temporal artery correlated with ischemic complications of GCA, such as ocular involvement, jaw claudication, and aortic arch syndrome.Production of PDGF has a role in arterial occlusion in GCA. The excessive fibroproliferative response leading to luminal narrowing can be distinguished from the stenosing process in atherosclerosis and postangioplasty restenosis, suggesting that there are different response patterns to arterial injury. In GCA, macrophages at the media-intima border are the dominant source of PDGF. Since vasoocclusion is associated with a number of serious complications in GCA, inhibition of intimal proliferation should be a major goal of treatment.

    View details for Web of Science ID 000072972800008

    View details for PubMedID 9550471

  • Heterogeneity of rheumatoid arthritis: from phenotypes to genotypes SPRINGER SEMINARS IN IMMUNOPATHOLOGY Weyand, C. M., Klimiuk, P. A., Goronzy, J. J. 1998; 20 (1-2): 5-22

    Abstract

    Rheumatoid arthritis (RA) is now recognized as a multigene disorder with a number of genetic polymorphisms contributing to disease pathogenesis. Here, we propose that the diagnostic category of RA includes multiple subtypes of disease and that the different phenotypes of RA correlate to different genotypes. Support for this concept has come from a reappraisal of the clinical heterogeneity of RA and the observation that HLA-DRB1 polymorphisms are useful in describing genetic heterogeneity of RA phenotypes. A series of HLA-DRB1 genes has been identified as RA associated, and in recent years emphasis has been put on the sequence similarities of these alleles. An alternative view focuses on the amino acid variations found in RA-associated HLA-DRB1 alleles with different alleles being enriched in distinct subtypes of RA. Rheumatoid factor-positive destructive joint disease is predominantly associated with the HLA-DRB1*0401 allele, while HLA-DRB1*0404 and B1*0101 predispose for milder and often seronegative disease. Expression of disease-associated alleles on both haplotypes carries a high risk for extra-articular manifestations. In particular, patients homozygous for HLA-DRB1*0401 frequently develop rheumatoid vasculities on follow-up. Besides HLA gene polymorphisms, abnormalities in the generation and function of CD4 T cells and in inflammatory pathways established in synovial lesions can be used to dissect patient subsets with different variants of RA. Emergence of CD28-deficient CD4 T cells identifies RA patients with extra-articular manifestations. These cells undergo clonal expansion in vivo, produce high amounts of IFN-gamma, and exhibit autoreactivity. Concordance of monozygotic twins for the expression of CD4+ CD28- T cells suggests a role for genetic factors in the generation of these unusual T cells. Evidence for heterogeneity of the synovial component of RA comes from studies describing three distinct patterns of lymphoid organization in the synovium. Based upon the topography of tissue-infiltrating mononuclear cells, diffuse, follicular, and granulomatous variants of rheumatoid synovitis can be distinguished. Each pattern of lymphoid organization correlates with a unique profile of tissue cytokines, demonstrating that several pathways of immune deviation modulate disease expression in RA. A dissection of RA variants would have major implications on how the disease is studied, treated, and managed. Identifying combinations of RA risk genes that correlate with disease variants could, therefore, become an important diagnostic tool.

    View details for Web of Science ID 000077027100003

    View details for PubMedID 9836366

  • T cell receptor repertoire in rheumatoid arthritis. International reviews of immunology Goronzy, J. J., Zettl, A., Weyand, C. M. 1998; 17 (5-6): 339-363

    Abstract

    CD4+ T cells are a major component of the inflammatory infiltrate in rheumatoid synovitis. Within synovial lesions, clonal CD4+ T cell populations are detectable, supporting the notion of an antigen specific recognition even in the joint. In general, the clonal size of individual T cell clones is small and does not lead to a marked distortion of the synovial T cell receptor (TCR) repertoire. Comparison of TCR sequences derived from different patients has not provided evidence for common sequences. Either multiple antigens are recognized or the TCR repertoire is sufficiently plastic with a multitude of different TCR structures responding to the same antigen(s). However, within one individual, the repertoire of clonal T cell populations is restricted. Identical T cell clones can be identified in different joints and at different timepoints of the disease, emphasizing that the spectrum of antigens recognized is conserved over time and that the T cell response pattern is not subject to evolution. Characterization of antigens involved in the latter stages of the disease may thus provide critical information on disease-initiating events. Recent data have led to the new concept that the role of T cells in rheumatoid arthritis (RA) is not limited to synovial inflammation. Evidence has been provided that the premorbid TCR repertoires of RA patients and normal controls can be distinguished. The T cell repertoire in RA patients is prone to recognize certain microbial products and autoantigens. The selection of this response pattern can only partially be attributed to the disease associated HLA-DRB1 alleles. Additional factors common in RA patients but not in HLA-DR matched control individuals seem to be important in shaping the TCR repertoire. Furthermore, the repertoire of mature T cells in RA patients is characterized by oligoclonality which involves T cells in the peripheral blood compartment. Possibly, these clonal T cell populations react to widespread autoantigens, raising the possibility that RA patients have a defect in controlling peripheral tolerance and an anomaly of lymphoproliferation. In contrast to joint residing CD4+ T cells, expanded clonotypes isolated from the blood of different patients have been described to share TCR beta chain structures. How these characteristic features of the global TCR repertoire in RA patients translate into mechanisms of disease remains to be elucidated.

    View details for PubMedID 10036639

  • Influence of antigenic experience on BJ gene segment usage in human CD4(+) T cells INTERNATIONAL IMMUNOLOGY Walser-Kuntz, D. R., Weyand, C. M., Goronzy, J. J. 1997; 9 (12): 1785-1792

    Abstract

    HLA molecules influence the selection of naive CD4+ T cells as demonstrated by HLA-DR-dependent differences in BV8-BJ frequencies. The repertoire of mature peripheral T cells utilized in antigen responses is shaped by additional factors such as antigens encountered in the environment. To identify mechanisms underlying the formation of the memory repertoire, differences in the BV8-BJ repertoire of CD45RO- and CD45RO+ CD4+ T cells were examined in 21 normal donors. The naive and memory CD4+ compartments displayed unique BV8-BJ repertoires in all individuals, demonstrating that the recruitment of CD4+ T cells into the memory population is a non-random process. The frequencies of selected BV8-BJ combinations were increased among CD45RO+ T cells. Size fractionation of such expanded BV8-BJ populations demonstrated that most of them were polyclonal in nature. Twenty-five percent of the expanded BV-BJ combinations included a dominant TCR sequence, indicating monoclonal proliferation. Selection of BV8-BJ combinations for preferential use among memory T cells was HLA dependent. HLA-DR1/4+ individuals were characterized by an increased usage of BV8-BJ2S7+ TCR, and decreased usage of BV8-BJ2S1 + and BV8-BJ2S2+ TCR, whereas HLA-DR3/7+ individuals preferentially recruited BV8-BJ2S5+ T cells, and disfavored BV8-BJ2S3+ and BV8-BJ2S7+ T cells. HLA-imposed effects on the naive and memory repertoire were distinct. The BV-BJ frequencies of CD45RO+ T cells could not be predicted from the pattern of TCR found in naive CD4+ T cells, suggesting that the HLA-DR polymorphisms influence thymic selection processes differently than peripheral selection forces.

    View details for Web of Science ID 000071289900002

    View details for PubMedID 9466306

  • Tissue cytokine patterns distinguish variants of rheumatoid synovitis AMERICAN JOURNAL OF PATHOLOGY Klimiuk, P. A., Goronzy, J. J., Bjornsson, J., Beckenbaugh, R. D., Weyand, C. M. 1997; 151 (5): 1311-1319

    Abstract

    Rheumatoid arthritis (RA) is a chronic inflammatory disease with primary manifestations in the synovial membrane. Tissue infiltrates are composed of T cells, B cells, and macrophages, but histopathological appearances vary widely and are rarely pathognomonic. Mechanisms underlying the phenotypic heterogeneity of rheumatoid synovitis are not known. To explore whether a correlation exists between the microscopic patterns of rheumatoid synovitis and in situ production of cytokines, tissue samples from 21 consecutive patients with clinically active RA were examined. Based upon the organization of the lymphocyte infiltrate, the synovial biopsies were categorized into three distinct subsets. Ten samples were characterized by diffuse lymphoid infiltrates without further microarrangement. In seven samples, lymphoid follicles with germinal center formation were detected, and in four specimens, granuloma formation was identified. In all specimens, cytokine transcription of interferon (IFN)-gamma, interleukin (IL)-4, IL-1 beta, tumor necrosis factor (TNF)-alpha, IL-10, and transforming growth factor-beta 1 was semiquantified with polymerase chain reaction and liquid phase hybridization. Each of the morphologically defined variants of synovitis displayed a unique cytokine profile. Low-level transcription of IFN-gamma, IL-4, IL-1 beta, and TNF-alpha was typical of diffuse synovitis. In follicular synovitis, IFN-gamma was the dominant cytokine, IL-4 was virtually undetectable, and IL-10 was abundant. Granulomatous synovitis demonstrated high transcription of IFN-gamma, IL-4, IL-1 beta, and TNF-alpha and could be clearly distinguished from the other phenotypes. To investigate whether differences in the synovial lesions were related to host factors, patients were compared for clinical parameters. Diffuse synovitis was seen in most of the patients with seronegative RA, the mildest form of the disease. In contrast, extra-articular spreading of RA with nodule formation was typically associated with granulomatous synovitis. In summary, RA patients display reproducible patterns in the organization and activity of synovial infiltrates. The correlation of microanatomy with tissue cytokine production suggests that several pathomechanisms can modulate the expression of the immune response in the synovial membrane.

    View details for Web of Science ID A1997YD87800018

    View details for PubMedID 9358757

    View details for PubMedCentralID PMC1858087

  • The molecular basis of rheumatoid arthritis JOURNAL OF MOLECULAR MEDICINE-JMM Weyand, C. M., Goronzy, J. J. 1997; 75 (11-12): 772-785

    Abstract

    Rheumatoid arthritis (RA) is an inflammatory disease targeting the synovial membrane and extra-articular tissues. The most feared consequences are significant levels of pain, functional disability, and rheumatoid organ involvement. Molecular investigations of RA have markedly changed the understanding of the pathogenesis although the etiology remains unresolved. Despite the failure of intense efforts to confirm the presence of an infectious micro-organism in rheumatoid synovium, the concept that RA is infectious in origin has continued to be attractive. Theories on the autoimmune nature of RA have benefited from the enormous progress made in understanding the cellular and molecular components of normal immune responses. However, convincing experimental evidence of a joint-specific endogenous antigen in the synovial lesions is still lacking. The viewpoint that RA represents the sequelae of systemic lymphoproliferation has recently been supported by the finding of autoreactive T cells with atypical growth and differentiation behavior. Significant cross-fertilization for the understanding of RA can be expected by studies elucidating cell cycle control and the role of proto-oncogenes. The realization that RA is a genetic disease has had and will have a major impact on investigating pathological events. As in other common genetic disorders, multiple genetic determinants contribute to the risk of an individual developing chronic inflammatory rheumatoid synovitis. Individual genetic elements are seldom mutated or abnormal, but a risk threshold is reached by their accumulation and combination. Genes encoded in the HLA region are recognized as RA risk genes. Recent studies have emphasized that a gene dosing effect for RA-associated HLA alleles is functional, and that HLA polymorphisms act as progression factors rather than as initiation factors in the disease process. These data have challenged the traditional paradigm that disease-associated HLA molecules function solely through their capability to select, bind, and present an arthritogenic antigen. Current efforts focus on identifying the spectrum and nature of genes associated with various RA phenotypes. The future will likely see a broadening of biological systems involved in the pathogenesis of RA with anomalies other than immunoresponsiveness contributing to mechanisms driving articular and extra-articular RA.

    View details for Web of Science ID A1997YH42000003

    View details for PubMedID 9428608

  • Giant cell vasculitis is a T cell-dependent disease MOLECULAR MEDICINE Brack, A., Geisler, A., MARTINEZTABOADA, V. M., Younge, B. R., Goronzy, J. J., Weyand, C. M. 1997; 3 (8): 530-543

    Abstract

    Giant cell arteritis (GCA) is a systemic vasculitis that preferentially targets medium-sized and large arteries. The etiopathogenesis of the syndrome is not known, and because of the paucity of information concerning the mechanisms of blood vessel wall damage, treatment options are limited. Clues to pathogenic events in this arteritis may derive from understanding the function of tissue-infiltrating cells. Arterial injury in GCA is associated with the formation of granulomas that are composed of T cells, activated macrophages, and multinucleated giant cells. To examine the role of T cells, we implanted inflamed temporal arteries from patients with GCA into severe combined immunodeficiency (SCID) mice and studied whether the vascular lesions were T cell-dependent.Temporal artery specimens from patients with GCA were engrafted into SCID mice. The histomorphologic appearance of fresh arteries and grafts retrieved from the mice was compared by two-color immunohistochemistry, and the functional profile of tissue-infiltrating cells was analyzed by semiquantifying cytokine transcription with a polymerase chain reaction (PCR)-based assay system. The repertoire of tissue-infiltrating T cells was assessed for the presence of dominant T cell populations by using T cell receptor beta-chain-specific PCR followed by sequencing. To investigate the role of T cells in the activation of tissue-infiltrating macrophages, T cells were depleted from the arterial grafts by treating the mice with T cell-specific antibodies and the production of monokines was monitored. To demonstrate the disease relevance of T cells expanding in the implants, T cells were isolated from tissue segments and adoptively transferred into mice implanted with syngeneic arteries. The in situ production of lymphokines was then determined.The inflammatory infiltrate penetrating all layers of the arterial wall persisted in the xenotransplants, indicating that the inflammatory foci represent independent functional units. Similar quantities of T cell- and macrophage-derived cytokines were detected in fresh and engrafted tissue. However, the diversity of tissue-infiltrating T cells decreased following implantation. T cells with identical T cell receptors were expanded in different mice that had been engrafted with tissue fragments from the same patient, indicating that T cell survival in the arterial wall was a nonrandom process. To confirm the disease relevance of these T cells, T cell depletion and reconstitution experiments were performed. Antibody-mediated elimination of T cells from the xenotransplants resulted in the attenuation of the production of the monokines, IL-1 beta and IL-6. Adoptive transfer of syngeneic tissue-derived T cells, but not of peripheral blood T cells, into engrafted SCID mice enhanced the transcription of IL-2 and IFN-gamma in the implanted arteries.The vascular lesions of GCA are maintained in human artery-mouse chimeras, indicating that all cellular and noncellular components necessary for the disease are present in the temporal artery. Activation of tissue-infiltrating T cells and macrophages depends upon an infrequent subpopulation of lesional T cells that have a survival advantage in the xenotransplants. The selective proliferation of these T cells in the arteries suggests that there is recognition of a locally expressed antigen. Therefore, these T cells should be candidate targets for the development of novel therapeutic strategies in GCA.

    View details for Web of Science ID A1997XT68700005

    View details for PubMedID 9307981

    View details for PubMedCentralID PMC2230183

  • Multiple mechanisms support oligoclonal T cell expansion in rheumatoid synovitis MOLECULAR MEDICINE Rittner, H. L., Zettl, A., Jendro, M. C., BARTZBAZZANELLA, P., Goronzy, J. J., Weyand, C. M. 1997; 3 (7): 452-465

    Abstract

    The synovial T cell infiltrate in rheumatoid arthritis (RA) is diverse but contains clonally expanded CD4+ populations. Recent reports have emphasized that RA patients have a tendency to develop CD4+ T cell oligoclonality which also manifests in the peripheral blood. Clonal dominance in the tissue may thus result from antigen specific stimulation in the synovial membrane or may reflect the infiltration of expanded clonotypes present throughout the lymphoid system. We have explored to what extent clonal populations amongst tissue CD4+ T cells display joint specificity as defined by their restriction to the joint, their persistence over time, and their expression of markers indicative for local activation.Matched samples of peripheral blood and synovial fluid or synovial tissue were collected from 14 patients with active RA and CD4+ IL-2R+ and CD4+ IL-2R- T cells from both compartments were purified. Clonal populations of CD4+ T cells were detected by RT-PCR amplification of T cell receptor (TCR) transcripts with BV and BJ specific primers followed by size fractionation and direct sequencing of dominant size classes of TCR transcripts.Clonal CD4+ T cells were detected in the synovial fluid and synovial tissue of all patients. All patients carried synovial clonotypes that were undetectable in the blood but were present in independent joints or at several non-adjacent areas of the same joint. These joint restricted CD4+ clonotypes were generally small in size, were preferentially found in the IL-2R+ subpopulation, and persisted over time. A second type of clonogenic T cells in the synovial infiltrate had an unrestricted tissue distribution and was present at similar frequencies amongst activated and nonactivated T cells in the blood and affected joints. Ubiquitous clonotypes isolated from two different patients expressed sequence homologies of the TCR beta chain.Two types of expanded CD4+ clonotypes contribute to the T cell infiltrate in rheumatoid synovitis. Differences in the distribution pattern and in molecular features suggest that distinct mechanisms are supporting the clonal outgrowth of these two groups of clonotypes. Clonally expanded T cells restricted to the joint but present in several independent joints appear to respond to locally residing antigens. Clonogenic cells with an unrestricted distribution pattern and widespread activation in the blood and tissue may react to a different class of antigens which appear to be shared by multiple patients. T cell recognition in RA may be involved at several different levels and may be related to more than one pathomechanism.

    View details for Web of Science ID A1997XN95500005

    View details for PubMedID 9260157

    View details for PubMedCentralID PMC2230222

  • Glucocorticoid-mediated repression of cytokine gene transcription in human arteritis-SCID chimeras JOURNAL OF CLINICAL INVESTIGATION Brack, A., Rittner, H. L., Younge, B. R., Kaltschmidt, C., Weyand, C. M., Goronzy, J. J. 1997; 99 (12): 2842-2850

    Abstract

    Giant cell arteritis (GCA) is a vasculitic syndrome that preferentially affects medium and large-sized arteries. Glucocorticoid therapy resolves clinical symptoms within hours to days, but therapy has to be continued over several years to prevent disease relapses. It is not known whether and how glucocorticoids affect the function of the inflammatory infiltrate or why the disease persists subclinically despite chronic treatment. GCA is self-sustained in temporal arteries engrafted into SCID mice, providing a model in which the mechanisms of action and limitations of glucocorticoid therapy can be examined in vivo. Administration of dexamethasone to temporal artery-SCID chimeras for 1 wk induced a partial suppression of T cell and macrophage function as indicated by the reduced tissue concentrations of IL-2, IL-1beta, and IL-6 mRNA, and by the diminished expression of inducible NO synthase. In contrast, synthesis of IFN-gamma mRNA was only slightly decreased, and expression of TGF-beta1 was unaffected. These findings correlated with activation of the IkappaBalpha gene and blockade of the nuclear translocation of NFkappaB in the xenotransplanted tissue. Dose-response experiments suggested that steroid doses currently used in clinical medicine are suboptimal in repressing NFkappaB-mediated cytokine production in the inflammatory lesions. Chronic steroid therapy was able to deplete the T cell products IL-2 and IFN-gamma, whereas the activation of tissue-infiltrating macrophages was only partially affected. IL-1beta transcription was abrogated; in contrast, TGF-beta1 mRNA synthesis was steroid resistant. The persistence of TGF-beta1-transcribing macrophages, despite paralysis of T cell function, may provide an explanation for the chronicity of the disease, and may identify a novel therapeutic target in this inflammatory vasculopathy.

    View details for Web of Science ID A1997XH27700008

    View details for PubMedID 9185506

    View details for PubMedCentralID PMC508134

  • Expansion of unusual CD4+ T cells in severe rheumatoid arthritis ARTHRITIS AND RHEUMATISM Martens, P. B., Goronzy, J. J., Schaid, D., Weyand, C. M. 1997; 40 (6): 1106-1114

    Abstract

    The repertoire of T cells in patients with rheumatoid arthritis (RA) is characterized by clonal expansion of selected CD4+ T cells, which are autoreactive and lack the expression of the functionally important CD28 molecule. The goal of this study was to determine the contribution of these unusual lymphocytes to the disease process.RA patients (n = 108) and normal controls (n = 53) were examined for the expression of CD4+ CD28- T cells by 2-color fluorescence-activated cell sorter analysis. Clinical data were ascertained by retrospective chart review.The frequencies of CD4+ CD28- T cells displayed a bimodal distribution, defining carriers and noncarriers in normal subjects and RA patients. In longitudinal studies, the noncarrier and carrier phenotypes were stable over time. Carriers of CD4+ CD28- T cells accumulated in the RA population (64% versus 45%; P = 0.02). The expansion of CD4+ CD28- T cells correlated with extraarticular involvement, but not with disease duration, antirheumatic treatment, or severity of joint destruction. The patient subsets with nodular disease (P = 0.02) and rheumatoid organ disease (P = 0.04) had the highest proportion of CD4+ CD28- T cell carriers. The size of the CD4+ CD28- compartment correlated with extraarticular progression of RA (P = 0.001 in nodular RA, P = 0.003 in rheumatoid organ disease).The bimodality of distribution of CD4+ CD28- T cell frequencies is compatible with genetic control of the generation of these unusual T cells. In RA patients, CD4+ CD28- T cells are not an epiphenomenon of the disease process, but predispose patients to developing inflammatory lesions in extraarticular tissues.

    View details for Web of Science ID A1997XC37600014

    View details for PubMedID 9182921

  • Co-stimulatory pathways controlling activation and peripheral tolerance of human CD4(+)CD28(-) cells EUROPEAN JOURNAL OF IMMUNOLOGY Park, W., Weyand, C. M., Schmidt, D., Goronzy, J. J. 1997; 27 (5): 1082-1090

    Abstract

    Co-stimulation mediated by the CD28 molecule is considered critical in the activation of CD4+ T cells. In patients with rheumatoid arthritis and infrequently in normal individuals, CD4+ T cells lacking CD28 expression are expanded and contain clonogenic populations. To analyze whether these cells are independent of co-stimulatory requirements or whether they use co-stimulatory signals distinct from the CD28 pathway, we have compared CD4+ CD28+ and CD4+ CD28- T cell clones isolated from rheumatoid arthritis patients. Accessory cells supported the induction of CD25 expression as well as of proliferative responses after anti-CD3 cross-linking and prevented the induction of anergy in CD4+ CD28- T cell clones. In contrast to CD4+CD28+ T cells, the presence of accessory cells did not enhance the secretion of interleukin (IL)-2, interferon-gamma, or IL-4. The co-stimulatory signals did not involve CD28/CTLA-4-CD80/CD86 receptor-ligand interactions. The proliferative response of CD4+CD28- T cells could not be blocked by anti-CD2, anti-CD18, and anti-CD58 antibodies, suggesting that these receptor-ligand interactions cannot provide CD28- independent co-stimulation. Our data suggest that CD4+CD28- T cells require co-stimulatory signals for optimal induction of cell growth and CD25 expression as well as for the prevention of anergy. The co-stimulatory receptor-ligand interaction is independent of the CD28 pathway and may be involved in the oligoclonal expansion of the CD4+ CD28- T cell subset in rheumatoid arthritis.

    View details for Web of Science ID A1997WZ36900006

    View details for PubMedID 9174596

  • Genetic risk factors in inflammatory abdominal aortic aneurysms: Polymorphic residue 70 in the HLA-DR B1 gene as a key genetic element JOURNAL OF VASCULAR SURGERY Rasmussen, T. E., Hallett, J. W., Metzger, R. L., Richardson, D. M., Harmsen, W. S., Goronzy, J. J., Weyand, C. M. 1997; 25 (2): 356-364

    Abstract

    Evidence of a genetic predisposition to the development of inflammatory abdominal aortic aneurysms (AAAs) exists as a positive family history in 17% of patients. Familial clustering and other similarities between inflammatory AAAs and giant cell arteritis (GCA), which possesses a genetic risk determinant mapped to the HLA-DR molecule, suggest a role of genetic risk factors in inflammatory AAAs. The purpose of this study was to explore whether patients with inflammatory AAAs express disease-relevant genes associated with the HLA-DR region on the short arm of chromosome 6.Thirty-seven patients with histomorphologic findings of inflammatory AAA at operation were genotyped for the polymorphism of the HLA-DR B1 and HLA DQ B1 alleles and compared to ethnically matched, healthy control subjects (n = 90).Distribution of HLA-DR B1 alleles was nonrandom in patients with inflammatory AAAs versus control subjects. The HLA-DR B1 alleles B1*15 and B1*0404 were enriched in patients with inflammatory AAAs compared with control subjects (47% versus 27%, and 14% versus 3%; p < 0.05, respectively). Analysis of functionally relevant amino acid polymorphisms encoded by the HLA-DR B1 gene showed relevance at amino acid position 70. HLA-DR B1 alleles overrepresented in patients with inflammatory AAAs express a glutamine substitution at position 70, whereas alleles disfavored in the patient cohort express a negatively charged aspartic acid. Distribution of HLA-DQ B1 alleles were indistinguishable in patients and control subjects.These data indicate that a genetic risk determinant can be mapped to the HLA-DR B1 locus in patients with inflammatory AAAs. This association suggests a critical contribution of antigen binding in the pathogenesis of this disease.

    View details for Web of Science ID A1997WL46300027

    View details for PubMedID 9052571

  • Superantigens, V-H gene polymorphism, and rheumatoid factor (RF) production B LYMPHOCYTES AND AUTOIMMUNITY Goronzy, J. J., He, X. W., Weyand, C. M. 1997; 815: 357-360

    View details for Web of Science ID A1997BH93X00044

    View details for PubMedID 9186678

  • Correlation between HLA-DR sequence polymorphisms and rheumatoid factor production B LYMPHOCYTES AND AUTOIMMUNITY Weyand, C. M., Goronzy, J. J. 1997; 815: 353-356

    View details for Web of Science ID A1997BH93X00043

    View details for PubMedID 9186677

  • Disease patterns and tissue cytokine profiles in giant cell arteritis ARTHRITIS AND RHEUMATISM Weyand, C. M., TETZLAFF, N., Bjornsson, J., Brack, A., Younge, B., Goronzy, J. J. 1997; 40 (1): 19-26

    Abstract

    To determine whether clinical heterogeneity in patients with giant cell arteritis (GCA) is correlated with different patterns in the tissue-specific inflammatory response.Twenty-three patients with typical histomorphologic findings of GCA were grouped according to the presence or absence of jaw claudication and/or visual abnormalities, fever, concomitant polymyalgia rheumatica (PMR), and histologic evidence of giant cell formation. The inflammatory response in temporal artery biopsy specimens was characterized by semiquantification of cytokine messenger RNA (mRNA) transcripts using reverse transcriptase-polymerase chain reaction, followed by oligonucleotide hybridization with cytokine-specific probes. Clinical patterns were then correlated with profiles of tissue cytokines.Inflammatory cytokines were expressed in all temporal artery tissues. In situ synthesis of interleukin-2 (IL-2), interferon-gamma (IFN gamma), and IL-1 beta mRNA, but not of IL-10 and IL-12 mRNA, distinguished different patterns of inflammation, and these patterns correlated with clinical manifestations of the disease. Patients with evidence of ischemic symptoms, indicated by jaw claudication and/or visual symptoms, typically expressed higher concentrations of IFN gamma mRNA (P = 0.008) and IL-1 beta mRNA (P = 0.02). Presence of fever was correlated with lower copy numbers of IFN gamma (P = 0.02). Formation of giant cells in the granulomatous infiltrates was associated with the local synthesis of IFN gamma mRNA (P = 0.003). Tissue from GCA patients with concomitant PMR contained higher levels of IL-2 mRNA transcripts (P = 0.001).Variations in the clinical presentation of GCA were correlated with cytokine mRNA expression in the affected temporal arteries. Differences in the effector functions of tissue-infiltrating T cells distinguished disease patterns in which either local ischemic symptoms or systemic involvement was dominant, or in which there was co-occurrence of PMR. Definition of different patterns of inflammation in GCA might, therefore, facilitate the design of differentiated therapeutic approaches.

    View details for Web of Science ID A1997WD15200004

    View details for PubMedID 9008596

  • Multisystem interactions in the pathogenesis of vasculitis. Current opinion in rheumatology Weyand, C. M., Goronzy, J. J. 1997; 9 (1): 3-11

    Abstract

    The vasculitides are often considered diseases caused by an immune response to a causative antigen. However, multiple decision points are involved in guiding an inflammatory process toward healing and repair versus destruction and disease. This review focuses on determinants that have the potential to alter decision points but have only recently begun to attract attention. Vasculitic disorders often exhibit a strict age preference. Progress has been made in the understanding of immunosenescence and its implications for immunocompetence as well as for the development of chronic inflammation. A second aspect in the pathogenesis of vasculitis is the distinct tissue tropism, involving preference for vessels of defined sizes and distributions. Studies in giant cell vasculitis have begun to shed light on the interaction between the affected tissue and the inflammatory cells and suggest that the vascular microanatomy determines the organization of the infiltrate. An important role may be attributed to the tunica adventitia, which harbors regulatory cells and probably provides the port of entrance for infiltrating cells. Finally, data are accumulating on the importance of the host's immunoresponsiveness in the decision between a beneficial and a pathologic inflammation. Studies in giant cell arteritis have shown that differences in the immunomodulatory milieu that are partially accomplished through the action of cytokines are responsible for disease heterogeneity and outcome.

    View details for PubMedID 9110127

  • Pathogenesis of rheumatoid arthritis MEDICAL CLINICS OF NORTH AMERICA Weyand, C. M., Goronzy, J. J. 1997; 81 (1): 29-?

    Abstract

    Chronicity and destructive potential are characteristic features of the inflammatory response in the synovial membrane typical for RA. The dominant paradigm has proposed that an exogenous antigen, likely an infectious organism, targets the synovia and elicits a chronic immune response. Support for this disease model has come from describing the cellular components of the inflammatory lesions, which are composed of macrophages, T cells, and B cells. The observation that HLA molecules function by specifically binding antigenic peptides and presenting them to T cells has boosted the concept of an antigen-driven response. The last decade in RA research has been dominated by a shift from premolecular to molecular techniques. A major effort has been made to determine which cytokines and inflammatory mediators are produced at the site of disease. Tissue residing and infiltrating cells secrete proinflammatory cytokines in situ, which likely have a critical role in amplifying and maintaining the inflammation. We are beginning to understand that migration of inflammatory cells into the tissue is an important component of disease, specifically because adhesion molecules not only facilitate tissue infiltration, but also affect cell activation and cell-cell and cell-matrix interactions. The paradigm that RA is an antigen-driven and thus T cell-mediated disease has brought attempts to use T cell-depleting reagents as therapeutics. Although T cells could be eliminated in the peripheral blood, overall therapeutic benefits have been minimal and accompanied by major side effects. The lack of therapeutic efficacy has been demonstrated to be combined with the persistence and the selective proliferation of T cells in the joint, reemphasizing the role of tissue-infiltrating T cells in the disease. Studies of the composition of the T cell infiltrate have demonstrated heterogeneity, indicating that disease-relevant T cells are probably low in frequency. A new perspective on the role of T cells in RA has been opened by studies establishing that RA patients select a unique repertoire of T cells in the thymus and that clonal expansion of CD4 T cells is a frequent event in RA patients. Pathology of T cell function might be much more systemic than suspected so far. RFs remain the major autoantibodies in RA patients. In the last 10 years, it has become clear that they are not exclusively built under pathologic conditions but that RF-expressing B cells are an important element of normal immune responses. All immunoglobulin isotypes are represented among RF molecules. Some of them have accumulated somatic mutations, suggesting the influence of antigen recognition and T cell help. T cell control of RF production may explain the observation that RF positivity is an HLA-dependent phenomenon. Major progress in understanding pathologic events leading to RA can be expected by abandoning single hit models, which are too simplified and underestimate the complexity of the disease. In particular, taking into account that nonimmune tissues and mechanisms might be equally important in pathogenesis will open new avenues of conceptual approaches. Cross-fertilization will likely come from genetic studies aimed at detecting underlying genetic risk factors in common genetic disease. Emerging data indicate that several genetic risk determinants, each of which is nonpathologic if occurring alone, can add up to confer disease risk. One of these genetic elements in RA has been mapped to the HLA region. A sequence polymorphism in the HLA-DR B1 gene appears to be a strong genetic risk factor in several ethnic groups. Correlation of clinical presentation of RA and the inheritance of the RA risk gene suggests that the gene product is not necessary in disease initiation but functions by modulating disease pattern and severity. The next decade in RA research will be dedicated toward unraveling how genetic determinants can introduce pathology (e.g., how HLA genes can function as progre

    View details for Web of Science ID A1997WE36500003

    View details for PubMedID 9012754

  • Recognition of tissue residing antigen by T cells in vasculitic lesions of giant cell arteritis JOURNAL OF MOLECULAR MEDICINE-JMM MARTINEZTABOADA, V., HUNDER, N. N., Hunder, G. G., Weyand, C. M., Goronzy, J. J. 1996; 74 (11): 695-703

    Abstract

    The objective of this study was to explore the nature of the antigen-specific T cell response in giant cell arteritis by analyzing clonally expanded T cells in temporal artery specimens. In temporal artery tissue from eight patients, 10% of the T cell receptor beta chain repertoire was systematically screened for clonal T cells by reverse-transcriptase polymerase chain reaction with selected BV, BJ, and BC specific primers and by direct sequencing of the amplified product. In five additional patients tissue-derived T cell clones were characterized. All expanded clonotypes were analyzed for their presence at different sites of the inflamed artery. T cell lines were tested for their proliferation to autologous monocytes pulsed with temporal artery extracts from patients with giant cell arteritis, polymyalgia rheumatica, and unrelated diseases. Clonally expanded T cells were identified in 30% of the BV-J combinations of the sampled repertoire. A subset of these clones were encountered at different sites of the inflammation, but not in the peripheral blood. The T cell receptor beta chain sequences were diverse. The patients had between none and five such clonotypes in the sampled repertoire, suggesting that only few T cell specificities in each patient are involved in antigen recognition. One of these T cell clonotypes was shown to proliferate in response to an antigen selectively expressed in temporal artery specimens from giant cell arteritis and from polymyalgia rheumatica patients. Clonotypes with identical T cell receptor beta chain sequences can be found at distinct sites of the inflammation in giant cell arteritis, suggesting recognition of the same antigen at different locations. At least for some of these T cell clones the antigen is shared between different giant cell arteritis and polymyalgia rheumatica patients but not expressed in temporal arteries of patients with unrelated diseases. While different HLA-DR4+ patients utilize distinct T cell specificities, the actual number of responding T cells in individual patients is small and may be disease limiting.

    View details for Web of Science ID A1996VT12500008

    View details for PubMedID 8956156

  • Correlation of the topographical arrangement and the functional pattern of tissue-infiltrating macrophages in giant cell arteritis JOURNAL OF CLINICAL INVESTIGATION Weyand, C. M., Wagner, A. D., Bjornsson, J., Goronzy, J. J. 1996; 98 (7): 1642-1649

    Abstract

    End organ ischemia, fragmentation of elastic membranes, and aneurysm formation in patients with giant cell vasculitis results from an inflammation destroying the mural layers of large and medium sized arteries. Although the inflammatory infiltrate extends through all layers of the affected blood vessel, the most pronounced changes involve the intima and the internal elastic lamina. Analysis of the functional profile of tissue infiltrating CD68+ cells demonstrates that different subsets of macrophages can be distinguished. TGFbeta1-expressing CD68+ cells coproduce IL-1beta and IL-6, are negative for inducible nitric oxide synthase (iNOS), and exhibit a strong preference for localization in the adventitia. The adventitial homing of TGFbeta1+ CD68+ cells places them in the vicinity of IFN-gamma secreting CD4+ T cells which also accumulate in the exterior layer of the artery. Conversely, iNOS expressing CD68+ cells are negative for TGFbeta and are almost exclusively found in the intimal layer of the inflamed artery. The intimal-medial junction is the preferred site for 72-kD collagenase expressing CD68+ cells. Thus, TGFbeta1-producing macrophages colocalize with activated CD4+ T cells and home to an area of inflammation which is distant from the site of tissue damage but critical in regulating cellular influx, suggesting that TGFbeta1 functions as a proinflammatory mediator in this disease. iNOS- and 72-kD collagenase-producing macrophages accumulate at the center of pathology implying a role of these products in tissue destruction. These data indicate that the microenvironment controls the topographical arrangement as well as the functional commitment of macrophages.

    View details for Web of Science ID A1996VL11200021

    View details for PubMedID 8833914

    View details for PubMedCentralID PMC507598

  • The pathogenic role of T lymphocytes in vasculitis SARCOIDOSIS VASCULITIS AND DIFFUSE LUNG DISEASES Weyand, C. M., Goronzy, J. J. 1996; 13 (3): 217-220

    Abstract

    Several disease mechanisms have been studied in the model of GCA. GCA patients exhibit a genetic susceptibility which has been mapped to the HLA-DRB1 gene. Polymorphic amino acid residues localized at the floor of the antigen binding site are highly selected in GCA patients suggesting a role for antigen binding and presentation in the disease. This and other genetic risk factors might actually be a surrogate for the distinct geographic distribution of the disease with a marked preference for Northern Europe. Studies on functional aspects of T cells accumulating in the vasculitic foci have demonstrated a strong bias for Th1 helper T cells which locally release IL-2 and IFN-gamma. IFN-gamma appears to be a key cytokine in this vasculitis. IFN-gamma producing T cells represent a minority of the tissue infiltrating cell population suggesting that very few cells have disease relevance and the majority of T cells is recruited as bystanders. IFN-gamma secreting CD4 T cells preferentially localize to the adventitial-medial junction and are thus placed distant from the center of pathology, the intima and internal media. TCRs expressed by T cells accumulated in the affected tissue are not randomly distributed but are biased toward selected specificities. These selected T cells undergo proliferation in the tissue and can be isolated from nonadjacent and independent sites of the vasculitis. This distribution pattern indicates a common driving factor, suspected to be a tissue residing antigen [8]. Further support for an antigen driving this pathological T cell response comes from the finding that temporal artery specimens engrafted into SCID mice continue to show the typical disease process indicating that all components relevant for the disease are contained in the temporal artery wall. So far no shared TCR utilized by different patients has been identified, raising the question whether distinct antigens can elicit GCA as a common pathway of reactivity. Besides its role in investigating pathomechanisms the SCID mouse model of GCA provides the unique opportunity to study the therapeutic effects of established and novel treatments. It can be expected that some of the pathogenic rules established for GCA can be applied to other vasculitic syndromes.

    View details for Web of Science ID A1996VK18700005

    View details for PubMedID 8946585

  • Increased responsiveness of rheumatoid factor-producing B cells in seronegative and seropositiive rheumatoid arthritis ARTHRITIS AND RHEUMATISM He, X. W., Zhong, W. Y., McCarthy, T. G., Weyand, C. M., Goronzy, J. J. 1996; 39 (9): 1499-1506

    Abstract

    To compare the frequencies and responsiveness of rheumatoid factor (RF)-producing B cells in the peripheral blood of patients with seronegative and seropositive rheumatoid arthritis (RA).Frequencies of IgM+, IgG+, and RF+ B cells were determined by limiting-dilution analysis of purified peripheral blood B cells from 6 patients with seropositive RA, 8 patients with seronegative RA, and 7 normal controls. B cell help was provided by cloned T helper cells, which were stimulated by either anti-CD3 or the bacterial superantigen staphylococcal enterotoxin D (SED). IgM and IgG antibodies and RF in culture supernatants were detected by enzyme-linked immunosorbent assay.In the presence of anti-CD3-stimulated T helper cells, 2-10% of B cells from normal individuals secreted IgM and IgG antibodies. The frequency of RF+ B cells was low and ranged from 1:182 to 1:885 (RF+: IgM+) B cells. In patients with seropositive RA, the numbers of Ig-producing B cells were reduced by a factor of 2, while the fraction of RF+ B cell precursors was expanded by more than 50-fold (7-20% of IgM+ B cells; P = 0.004). Patients with seronegative RA had higher frequencies of RF-producing B cells (1.5-6% of IgM+ B cells) than normal individuals (P = 0.002), but not to the same extent as seropositive patients (P = 0.002). Stimulation of B cells using SED preferentially induced RF+ B cells in normal controls and in patients with seronegative and seropositive RA.B cell precursors with the potential to secrete RF were detectable in high frequencies in normal individuals and in patients with seropositive and seronegative RA. In all donors, these B cells could be stimulated with the bacterial superantigen SED. In normal individuals, RF+ B cells remained nonresponsive to help provided by anti-CD3-activated T cells, but were responsive in RA patients. Seronegative and seropositive RA form a continuous spectrum of disease, with a higher number of RF-secreting B cells in the seropositive patients.

    View details for Web of Science ID A1996VH52300008

    View details for PubMedID 8814061

  • The repertoire of CD4(+) CD28(-) T cells in rheumatoid arthritis MOLECULAR MEDICINE Schmidt, D., Martens, P. B., Weyand, C. M., Goronzy, J. J. 1996; 2 (5): 608-618

    Abstract

    While oligoclonality of circulating CD4- CD8 and of CD8+ T cells is not uncommon, clonal dominance within the CD4 compartment is not frequently found in healthy individuals. In contrast, the majority of patients with rheumatoid arthritis (RA) have clonally expanded CD4+ T cell populations. Previous studies have demonstrated that these clonogenic CD4+ T cells do not express the CD28 molecule. To examine the correlation between CD28 expression and clonal proliferation, we have analyzed the T cell receptor (TCR) diversity of CD4+ CD28- T cells in normal individuals and in RA patients.The size of the peripheral blood CD4+ CD28- compartment was determined in 30 healthy individuals and 30 RA patients by two-color FACS analysis. In 10 RA patients and five controls with more than 2.5% CD4+ CD28- T cells, TCR BV gene segment usage was analyzed with 19 BV-specific antibodies. Oligoclonality was assessed in sorted CD4+ CD28+ and CD28- T cells using TCR BV-BC-specific polymerase chain reaction and size fractionation. Clonal dominance was confirmed by direct sequencing.The CD4+ CD28- T cell compartment was expanded to more than 2.5% in 70% of the RA patients and 30% of the normal individuals. Compared with the CD4+ CD28+ T cells, the TCR BV gene segment usage among CD4+ CD28- cells was grossly skewed with the dominance of single BV elements. Molecular TCR analysis provided evidence for oligoclonality in 17 of 21 expanded BV elements. In two unrelated RA patients who shared both HLA-DRB1 alleles, the TCR beta-chain sequences of dominant clonotypes were highly conserved.Oligoclonality is a characteristic feature of CD4+ CD28- T cells which are expanded in some healthy individuals and in the majority of RA patients. The lack of CD28 expression is a common denominator of CD4+, CD8+, and CD4- CD8- T cells prone to develop clonal dominance. The limited TCR diversity of clonal CD4+ CD28- populations in RA patients suggests that these T cells recognize a limited spectrum of antigens. The fact that the majority of individuals with marked expansions and oligoclonality of CD4+ CD28- T cells are RA patients suggests a role for these unusual lymphocytes in the pathogenetic events leading to RA.

    View details for Web of Science ID A1996VL18900009

    View details for PubMedID 8898376

    View details for PubMedCentralID PMC2230198

  • Interferon-gamma-producing T cells in giant cell vasculitis represent a minority of tissue infiltrating cells and are located distant from the site of pathology AMERICAN JOURNAL OF PATHOLOGY Wagner, A. D., Bjornsson, J., Bartley, G. B., Goronzy, J. J., Weyand, C. M. 1996; 148 (6): 1925-1933

    Abstract

    Giant cell vasculitis is an arteritis that predominantly affects medium- and large-sized arteries. Genetic risk factors and clonal expansion of selected CD4+ T cell specificities in the vascular lesions support the model that giant cell arteritis is a T-cell-driven disease. Interferon (IFN)-gamma production in the tissue is intimately associated with the formation of the inflammatory infiltrates. Antigens inducing stimulation of T cells are unknown. To provide indirect evidence for the type and the tissue localization of the antigen, we examined CD4+ T cells in the lesions that secrete IFN-gamma. Temporal artery specimens from patients with giant cell arteritis were analyzed bu two-color immunohistochemistry applying antibodies to T cell markers. IFN-gamma, the interleukin-2 receptor alpha-chain (CD25) and talin, a cytoskeletal protein that is reorganized in T cells interacting with antigen-presenting cells. Proliferating cells in the lesions were identified through the expression of the Ki-67 nuclear antigen. More than 90% of tissue-infiltrating IFN-gamma-producing cells were CD4+ CD45RO+. They represented a minute subset (2 to 4%) of tissue-infiltrating T cells. IFN-gamma+ T cells aggregated in the adventitial layer of the inflamed artery where they were either diffusely distributed or arranged in clusters. The majority of IFN-gamma-secreting T cells expressed CD25. IFN-gamma+ T cells included a fraction of cells that had reorganized the cytoskeletal protein talin, indicating an interaction of the T cell receptor and an antigen-presenting cell. A subset of IFN-gamma-expressing T cells was undergoing proliferation in the tissue. IFN-gamma-producing T cells in vasculitic lesions of giant cell arteritis express several markers that identify them as T cells that have recently been stimulated through their antigen-specific receptor. These putatively disease-relevant T cells represent only a very minor fraction of tissue-infiltrating cells. Their preferential accumulation in the adventitia is most compatible with the model that they contact the relevant antigen primarily in this particular region of the artery. Their regulatory function appears to extend into the inner media and intima where pathological changes in giant cell arteritis are most pronounced.

    View details for Web of Science ID A1996UN46800022

    View details for PubMedID 8669478

    View details for PubMedCentralID PMC1861625

  • Clonally expanded CD8 T cells in patients with polymyalgia rheumatica and giant cell arteritis CLINICAL IMMUNOLOGY AND IMMUNOPATHOLOGY MARTINEZTABOADA, V. M., Goronzy, J. J., Weyand, C. M. 1996; 79 (3): 263-270

    Abstract

    Giant cell arteritis (GCA) is a vasculitic entity which exclusively, affects individuals older than 50 years of age. Polymyalgia rheumatica (PMR) is a closely related condition which lacks clinically significant vasculitic lesions but shares with GCA the age dependence and the HLA association. To examine whether age-related changes in the T cell receptor repertoire represent a risk factor in these two diseases, we have analyzed the diversity of peripheral blood CD8+ T cells. Untreated PMR/GCA patients carried multiple clonally expanded CD8 populations. The frequency of clonal expansion was not different from age-matched healthy controls. Molecular analysis of the CD8+ clonotypes showed a restricted repertoire in the patients with a distinct Jbeta gene segment usage compared to normal controls. Jbeta2.7+ CD8+ clonotypes were exclusively found in patients. Further evidence for selective CD8 cell expansion came from the finding that multiple clonotypes in the same patient transcribed identical Jbeta segments despite diversity of the Vbeta element. Oligoclonality in the CD8 repertoire persisted despite successful control of the disease activity, suggesting that the CD8+ clonotypes are not an epiphenomenon of the inflammation. We propose that selected CD8+ cells are of functional importance in the pathogenesis of GCA and PMR through a Jbeta-specific mechanism. Age-related changes in the composition of the CD8 T cell receptor repertoire with the emergence of such clonotypes may predispose individuals to develop PMR/GCA.

    View details for Web of Science ID A1996UM79400010

    View details for PubMedID 8635285

  • The inflammatory infiltrate in giant cell arteritis selects against B lymphocytes JOURNAL OF RHEUMATOLOGY MARTINEZTABOADA, V., Brack, A., Hunder, G. G., Goronzy, J. J., Weyand, C. M. 1996; 23 (6): 1011-1014

    Abstract

    To understand mechanisms regulating the cellular composition of the inflammatory infiltrate in giant cell arteritis (GCA) and to explore whether B lymphocytes, such as neoplastic B cells in chronic lymphocytic leukemia (CLL), can infiltrate the inflamed arterial wall of GCA.A Mayo Clinic database search was conducted to identify patients that developed GCA after the onset of CLL between 1950 and 1994. Inflammatory infiltrates were analyzed immunohistochemically for T cells, B cells, and plasma cells.Five patients with CLL were identified who subsequently developed clinical findings suggestive of GCA. Three of the 5 patients had biopsy proven disease, presented with typical clinical and histomorphological features of GCA, and responded promptly to corticosteroid treatment. Leukemic B cells known for their ability to diffusely seed organs were absent in the inflammatory lesions in the temporal artery. Plasma cells that contribute to the vasculitic infiltrate in patients without CLL were markedly reduced in the GCA lesions of patients with CLL. Two other patients had symptoms suggestive of GCA. Their temporal artery specimens were remarkable for small adventitial lymphocytic infiltrates but lacked typical panarteritis.The inflammatory infiltrate of GCA is tightly regulated, and cell accumulation in the granulomas is an active, not a passive, mechanism. The inflammatory pathway in GCA is focused on T cells and macrophages and excludes B cells.

    View details for Web of Science ID A1996UP53900012

    View details for PubMedID 8782132

  • Oligoclonal T cell proliferation in patients with rheumatoid arthritis and their unaffected siblings ARTHRITIS AND RHEUMATISM Waase, I., Kayser, C., Carlson, P. J., Goronzy, J. J., Weyand, C. M. 1996; 39 (6): 904-913

    Abstract

    To analyze whether patients with rheumatoid arthritis (RA) have an intrinsic defect in T cell proliferation and survival, possibly contributing to the infiltration of the synovial membrane with CD4+ T cells.Fifteen patients with seropositive RA, 11 patients with psoriatic arthritis, 20 normal controls, and 9 affected and 13 unaffected siblings from 7 multiplex families with RA were analyzed for clonal proliferation. To investigate this clonal T cell proliferation, CD4+ T cells were purified from peripheral blood and synovial fluid by magnetic bead separation. T cell receptor (TCR) beta-chain sequences were amplified by reverse transcriptase-polymerase chain reaction, using TCR BV and BJ gene segment-specific primer sets. Clonally expanded T cell specificities were identified by size fractionation and sequencing of the amplified product.All RA patients carried clonally expanded CD4+ T cells in the peripheral blood compartment. Such expanded CD4+ T cell clonotypes were only infrequently observed both in normal individuals (P < 0.0001) and in patients with psoriatic arthritis (P = 0.004). Lymphoproliferation of selected CD4+ T cells was shared by affected and unaffected siblings from RA multiplex families (P = 0.005 and P = 0.0003, respectively, compared with normal controls). Expanded clonotypes persisted for several years and contributed to the T cell infiltrate in the joint. Clonal T cell proliferation involved a diverse spectrum of TCR molecules.RA patients have an abnormality in the homeostasis of CD4+ T cells, characterized by the emergence of clonally proliferating populations. The presence of clonal outgrowth of selected CD4+ T cells specificities in unaffected siblings of RA patients suggests that oligoclonality of CD4+ T cells is inherited and is a risk factor for, rather than a result of, synovial inflammation.

    View details for Web of Science ID A1996UQ19200004

    View details for PubMedID 8651983

  • CD4(+) CD7(-) CD28(-) T cells are expanded in rheumatoid arthritis and are characterized by autoreactivity JOURNAL OF CLINICAL INVESTIGATION Schmidt, D., Goronzy, J. J., Weyand, C. M. 1996; 97 (9): 2027-2037

    Abstract

    Clonal expansion of CD4+ T cells is a characteristic finding in patients with RA and is only infrequently found in patients with psoriatic arthritis and healthy controls. Expanded CD4+ clonotypes are present in the blood, infiltrate into the joint, and persist over years. We have not addressed the question of whether the expanded clonotypes have unique functional and phenotypic properties which may explain the preferential in vivo expansion in RA. In contrast to most CD4+ T cells, expanded clonotypes lacked the expression of the CD28 and CD7 cell surface molecules. Accordingly, the subsets of CD4+ CD28- (9.7 vs 1.7, P = 0.00002) and CD4+ CD7- T cells (21.5 vs 12.26, P = 0.018) were increased in RA patients compared with age-matched normal individuals. Despite the lack of CD28 expression, clonally expanded CD4+ T cells were not anergic but proliferated in response to immobilized anti-CD3 and could be maintained in tissue culture. In vivo expanded CD4+ T cells were autoreactive to ubiquitously distributed autoantigens. They responded in an autologous mixed lymphocyte reaction, and T cell clones isolated from selected patients proliferated to autologous peripheral blood adherent cells. These data suggest that in RA patients selected CD4+ T cells which share the CD7- CD28- phenotype escape from peripheral tolerance.

    View details for Web of Science ID A1996UJ39900006

    View details for PubMedID 8621791

    View details for PubMedCentralID PMC507276

  • TRIGGERING OF LYMPHOCYTES-T VIA EITHER T3-TI OR T11 SURFACE-STRUCTURES OPENS A VOLTAGE-INSENSITIVE PLASMA-MEMBRANE CALCIUM-PERMEABLE CHANNEL - REQUIREMENT FOR INTERLEUKIN-2 GENE-FUNCTION JOURNAL OF BIOLOGICAL CHEMISTRY Gardner, P., Alcover, A., Kuno, M., Moingeon, P., Weyand, C. M., GORONZY, J., REINHERZ, E. L. 1989; 264 (2): 1068-1076

    Abstract

    Stimulation of human T-lymphocytes via either the surface T3-Ti antigen-major histocompatibility complex receptor complex or the T11 molecule results in clonal proliferation through a calcium-dependent mechanism. To investigate this signal transduction, plasma membrane calcium-permeable channels were characterized in T-lymphocytes by means of whole cell or single channel patch-clamp recordings. Stimulation of T-lymphocytes via either structure results in opening of an identical set of voltage-insensitive plasma membrane Ca2+-permeable channels through the action of a diffusible second messenger. Previous work with excised inside-out patches suggests that inositol 1,4,5-trisphosphate is the activating second messenger of the voltage-insensitive T-cell Ca2+-permeable channel. Since there is a significant increase in phosphoinositide turnover after stimulation via either the T3-Ti or T11 pathway, it is suggested that triggering of either structure opens a common set of channels through this mechanism. Furthermore, currents flowing through Ca2+-permeable channels are apparently autoregulated, as inward conductance is abolished by elevation of Ca2+ concentration in the bathing solution. In particular, the steady-state rise in interleukin-2 (T-cell growth factor) mRNA is dependent on the rise of [Ca2+]i resulting from ion movement across this channel.

    View details for Web of Science ID A1989R820000062

    View details for PubMedID 2562953

  • RECOGNITION OF POLYMORPHIC H-2 DOMAINS BY LYMPHOCYTES-T .1. FUNCTIONAL-ROLE OF DIFFERENT H-2 DOMAINS FOR THE GENERATION OF ALLOREACTIVE CYTO-TOXIC LYMPHOCYTES-T AND DETERMINATION OF PRECURSOR FREQUENCIES JOURNAL OF EXPERIMENTAL MEDICINE WEYAND, C., GORONZY, J., Hammerling, G. J. 1981; 154 (6): 1717-1731

    Abstract

    In the present communication, the repertoire of alloreactive cytotoxic T lymphocytes (CTL) clones was quantitatively investigated by limiting dilution analysis and by target inhibition with a panel of monoclonal antibodies (mAb). These mAb have previously been shown to define two distinct alloantigenic domains, A and B, on the H-2Kk molecule. The Poisson distribution analysis of H-2Kk-specific CTL clones generated in a limiting dilution system revealed three CTL populations with different precursor frequencies. The high frequent population is suppressed by an unknown suppressive mechanism that allows less frequent CTL populations to become visible. Target inhibition studies with a panel of Kk-specific mAb showed that these CTL populations differ not only in their precursor frequency but also in their specificity for different H-2 epitopes on the Kk molecule. Thus clones of the high frequency population are almost exclusively specific for determinants within domain A. In contrast, the low frequency population displays predominant specificity for determinants of domain B, while the population with medium frequency is blocked equally well by mAb against either domains A or B. Each mAb blocked only a fraction of clones indicating that each CTL subpopulation may consist of a large number of clonotypes with specificity for different H-2 epitopes. The data suggest that CTL recognize basically the same polymorphic domains on the H-2Kk molecule defined by antibodies, and they show that regulatory mechanisms determine the expressed repertoire in CTL populations.

    View details for Web of Science ID A1981MU61700001

    View details for PubMedID 6976406

  • RECOGNITION OF H-2 DOMAINS BY CYTO-TOXIC LYMPHOCYTES-T NATURE WEYAND, C., Hammerling, G. J., GORONZY, J. 1981; 292 (5824): 627-629

    Abstract

    The polymorphic major histocompatibility antigens (H-2) have a crucial role in the activation of antigen-specific T lymphocytes. Thus, H-2 antigens are not only recognized by allogenic lymphocytes leading to generation of cytotoxic T lymphocytes (CTLs), but it has also been demonstrated that in syngeneic systems most T cells are only able to recognize foreign antigens in conjunction with their own MHC (major histocompatibility complex) antigens. This phenomenon, termed H-2 restriction, may be the key to our understanding to the biological function of MHC antigens. It is not clear whether recognition by T cells of H-2 on a molecular level is confined to particular domains on the H-2 molecule, nor whether the same polymorphic H-2 sites, which are characterized by antibodies, are recognized by allogeneic as well as by H-2 restricted syngeneic CTLs. Previous findings indicate the existence of at least two major polymorphic domains on the H-2Kk molecule as defined by antibodies. Here we show the existence of CTLs with specifity for these polymorphic domains, and the preferential recognition of a particular domain by both alloreactive as well as H-2 restricted CTLs.

    View details for Web of Science ID A1981MB41800036

    View details for PubMedID 6166874