Bio


Dr. Fathman is an example of a clinician scientist who has developed a clear vision for implementation of translational research. He has over 300 publications, many of them in the top peer-reviewed journals including Science, Nature, Cell, Journal of Experimental Medicine, JCI, Immunity, Nature Medicine and Nature Immunology. Among Dr. Fathman’s seminal contributions to his field, is the initial cloning of CD4 T lymphocytes while he was a member of the Basel Institute for Immunology. The use of soft agar seeding of activated cells had allowed the cloning of what are now called hybridomas and drove the field monoclonal antibody production. Using this same technology, Dr. Fathman was able to clone allo-reactive T lymphocytes. Dr. Fathman left Basel and became an Associate Professor of Immunology at Mayo Medical School in 1977. There, along with one of his postdoctoral fellows, he adapted the soft agar cloning technology to clone antigen specific CD4 T cells for the first time. The ability to study single T cell specificities allowed rapid advancement in understanding the components of the ternary complex for T cell activation and led Dr. Fathman to identify trans-complementing MHC Class 2 products used in antigen presentation before the biochemical two chain nature of MHC Class 2 products was described. Shortly thereafter, he was the first to identify “idiotypic structures“ on cloned CD4 T cells predating the identification of the T cell receptor for antigen by molecular biological techniques. Dr. Fathman moved from Mayo to Stanford in 1981 and continued his studies on T cell clones, initially identifying the “shared epitope” on HLA Class 2 molecules in RA patients. As a new faculty member at Stanford, he expanded his studies to examine animal models of autoimmunity. The initial observation that led to his studies on the use of monoclonal antibodies to treat animal models of autoimmunity came from the observation that immune unresponsiveness could be induced in mice by the use of anti-CD4 antibodies at the time of antigen immunization. Subsequently he was the first to use anti-CD4 antibodies to block allograft transplant rejection and was the first to use peptides of an autoantigen (myelin basic peptide), to induce a state of “anergy” in mice to ameliorate disease. Initially, anti-CD4 antibody was used to block progression to diabetes in NOD mice. Many subsequent publications were linked to his NOD colony including several seminal observations on pathophysiology, immunotherapy, and gene expression. One major finding was the identification of a gene, DEAF-1, expressed in pancreatic lymph nodes whose non-canonical splice variant was involved in defective non-thymic mechanisms for inducing or maintaining peripheral tolerance in NOD and in human T1D. More recently he has used gene expression studies of peripheral blood cells from type one diabetes (T1D) patients and relatives to demonstrate a gene expression signature of risk of disease and of disease progression in T1D. He is currently developing a novel therapeutic approach to the treatment of autoimmune and allergic diseases by targeting the endogenous regulatory T cell to "turn up" its activity to prevent or treat these inflammatory diseases.

Academic Appointments


Administrative Appointments


  • President, Federation of Clinical Immunology Societies (FOCIS) (2002 - Present)
  • Associate Director, ITI Institute Stanford (2008 - Present)
  • President, Clinical Immunology Society (2000 - 2001)
  • Director, Center for Clinical Immunology at Stanford (CCIS) (1993 - Present)
  • Division Chief, Division of Immunology and Rheumatology (1997 - 2014)
  • Associate Editor, Annual Review of Immunology (1981 - 2005)
  • Council, American Society for Clinical Investigation (1984 - 1987)
  • Council, Midwinter Conference of Immunologists (1981 - 1986)

Honors & Awards


  • Member and elected Council member, American Society of Clinical Investigation (1984-1987)
  • Member, American Association of Physicians (1990-present)
  • Naomi M. Kanof Award for Distinguished Achievement in Clinical Investigation, Society for Investigative Dermatology (1997)
  • Alumni Achievement Award, Washington University Medical School (1999)
  • President's Award, Clinical Immunology Society (2006)
  • Master, American College of Rheumatology (2007)
  • Member, Council member, Henry Kunkel Society (2007-2010)
  • Founder's Award, Federation of Clinical immunology Societies (2010)
  • Division Teaching Award, Stanford University School of Medicine, Department of Medicine (2011)
  • Mayo Clinic Distinguished Alumnus, Mayo Clinic (2015)
  • Commencement Address, Washington University Medical School (2016)

Boards, Advisory Committees, Professional Organizations


  • Editorial Board, Transplantation (1979 - 1984)
  • Editorial Board, Journal of Molecular and Cellular Immunology (1984 - 1988)
  • Editorial Board, Annales de l’Institut Pasteur Immunologie (1985 - 1989)
  • Editorial Board, Journal of Clinical Investigation (1985 - 1990)
  • Section Chief, Clinical Immunology, Journal of Immunology (1986 - 1990)
  • Member, AAI (1987 - 2014)
  • Member, Council, President, Clinical Immunology Society (1990 - Present)
  • member, ACR (1990 - Present)
  • Director, Center for Clinical Immunology,Stanford (1995 - Present)
  • Editorial Board, The Immunologist (1996 - 1999)
  • Associate Editor, Clinical Immunology and Immunopathology (1998 - 2003)
  • Editorial Board, Journal of Clinical Immunology (1998 - 2003)
  • Associate Editor, Annual Review of Immunology (1998 - 2005)
  • Member, ADA (2002 - Present)
  • Advisory Board Member, Nature Clinical Practice Rheumatology (2005 - Present)
  • Associate Director, Institute for Immunology, Transplantation and Infection, Stanford (2007 - Present)
  • Scientific Advisory Committee, Lupus Research Alliance (2016 - Present)

Professional Education


  • B.A., Univ. Kentucky, Lexington, Pre-Med (1964)
  • M.D., Washington Univ., St. Louis, Medicine (1969)

Current Research and Scholarly Interests


My laboratory of molecular and cellular immunology is interested in mechanisms of T cell anergy and the pathophysiology and immunotherapy of preclinical animal models of autoimmune disease.
I. T Cell Anergy: We have identified a ubiquitin E3 ligase (GRAIL) that seems to be central to the control of regulatory T cell (Treg) function. This regulation is controlled by inhibition of the desensitization of the Treg IL-2 receptor allowing prolonged pStat5 transcription of Treg centric genes. Two deubiquiting enzymes, USP8 and OTUB1, play contrasting roles in maintaining GRAIL stability and thus inhibition of IL-2R desensitization.
II. Gene Therapy: We have demonstrated that the local delivery of anti-inflammatory proteins via adoptive cellular gene therapy using syngeneic dendritic cells (DCs) transduced to express immunoregulatory proteins, in three murine models of autoimmunity (RA, MS and T1D), provide therapeutic effect both in the prevention of disease onset and in therapy of established disease.
III. Gene expression studies in autoimmunity: The major emphasis placed on disease associated genetic mutations or polymorphisms to understand the genetics of T1D has failed to advance either understanding of T1D pathogenesis or to identify therapeutic targets. Recent studies from my lab have demonstrated that tissue- and disease-specific changes in mRNA expression, rather than DNA variants, may underlie the progression of T1D. By combining the expertise of the lab in T1D research with established preclinical models and patient samples/tissues from the Network for Pancreatic Organ Donors with Diabetes, nPOD (http://www.jdrfnpod.org/), as well as from TrialNet, my lab has both demonstrated a potential defect in peripheral tolerance in NOD mice that has homologies in T1D patients and has identified a signature of predisposition to developing T1D (risk) as well as a signature of T1D disease progression.
iv. Development of new therapeutics to treat autoimmune and allergic diseases. Using the knowledge that Treg IL-2R desensitization is important in Treg function, we have developed a screening system to identify lead candidates that can inhibit Il-2R desensitization to be used in concert with low dose IL-2 therapy to treat or prevent autoimmune and allergic diseases.

2023-24 Courses


Graduate and Fellowship Programs


All Publications


  • Identification of Novel Disease-Relevant Genes and Pathways in the Pathogenesis of Type 1 Diabetes: A Potential Defect in Pancreatic Iron Homeostasis DIABETES Yip, L., Alkhataybeh, R., Taylor, C., Fuhlbrigge, R., Fathman, C. 2022; 71 (7): 1490-1507

    Abstract

    Multiple pathways contribute to the pathophysiological development of type 1 diabetes (T1D); however, the exact mechanisms involved are unclear. We performed differential gene expression analysis in pancreatic islets of NOD mice versus age-matched congenic NOD.B10 controls to identify genes that may contribute to disease pathogenesis. Novel genes related to extracellular matrix development and glucagon and insulin signaling/secretion were changed in NOD mice during early inflammation. During "respective" insulitis, the expression of genes encoding multiple chemosensory olfactory receptors were upregulated, and during "destructive" insulitis, the expression of genes involved in antimicrobial defense and iron homeostasis were downregulated. Islet inflammation reduced the expression of Hamp that encodes hepcidin. Hepcidin is expressed in β-cells and serves as the key regulator of iron homeostasis. We showed that Hamp and hepcidin levels were lower, while iron levels were higher in the pancreas of 12-week-old NOD versus NOD.B10 mice, suggesting that a loss of iron homeostasis may occur in the islets during the onset of "destructive" insulitis. Interestingly, we showed that the severity of NOD disease correlates with dietary iron intake. NOD mice maintained on low-iron diets had a lower incidence of hyperglycemia, while those maintained on high-iron diets had an earlier onset and higher incidence of disease, suggesting that high iron exposure combined with a loss of pancreatic iron homeostasis may exacerbate NOD disease. This mechanism may explain the link seen between high iron exposure and the increased risk for T1D in humans.

    View details for DOI 10.2337/db21-0948

    View details for Web of Science ID 000905183700009

    View details for PubMedID 35499603

    View details for PubMedCentralID PMC9233262

  • How GRAIL controls Treg function to maintain self-tolerance. Frontiers in immunology Fathman, C. G., Yip, L., Gomez-Martin, D., Yu, M., Seroogy, C. M., Hurt, C. R., Lin, J. T., Jenks, J. A., Nadeau, K. C., Soares, L. 2022; 13: 1046631

    Abstract

    Regulatory T cells (Tregs) normally maintain self-tolerance. Tregs recognize "self" such that when they are not working properly, such as in autoimmunity, the immune system can attack and destroy one's own tissues. Current therapies for autoimmunity rely on relatively ineffective and too often toxic therapies to "treat" the destructive inflammation. Restoring defective endogenous immune regulation (self-tolerance) would represent a paradigm shift in the therapy of these diseases. One recent approach to restore self-tolerance is to use "low dose IL-2" as a therapy to increase the number of circulating Tregs. However, studies to-date have not demonstrated that low-dose IL-2 therapy can restore concomitant Treg function, and phase 2 studies in low dose IL-2 treated patients with autoimmune diseases have failed to demonstrate significant clinical benefit. We hypothesize that the defect in self-tolerance seen in autoimmunity is not due to an insufficient number of available Tregs, but rather, due to defects in second messengers downstream of the IL-2R that normally control Treg function and stability. Previous studies from our lab and others have demonstrated that GRAIL (a ubiquitin E3 ligase) is important in Treg function. GRAIL expression is markedly diminished in Tregs from patients with autoimmune diseases and allergic asthma and is also diminished in Tregs of mice that are considered autoimmune prone. In the relevant pathway in Tregs, GRAIL normally blocks cullin ring ligase activity, which inhibits IL-2R desensitization in Tregs and consequently promotes Treg function. As a result of this defect in GRAIL expression, the Tregs of patients with autoimmune diseases and allergic asthma degrade IL-2R-associated pJAK1 following activation with low dose IL-2, and thus cannot maintain pSTAT5 expression. pSTAT5 controls the transcription of genes required for Treg function. Additionally, the GRAIL-mediated defect may also allow the degradation of the mTOR inhibitor, DEP domain-containing mTOR interacting protein (Deptor). This can lead to IL-2R activation of mTOR and loss of Treg stability in autoimmune patients. Using a monoclonal antibody to the remnant di-glycine tag on ubiquitinated proteins after trypsin digestion, we identified a protein that was ubiquitinated by GRAIL that is important in Treg function, cullin5. Our data demonstrate that GRAIL acts a negative regulator of IL-2R desensitization by ubiquitinating a lysine on cullin5 that must be neddylated to allow cullin5 cullin ring ligase activity. We hypothesize that a neddylation inhibitor in combination with low dose IL-2 activation could be used to substitute for GRAIL and restore Treg function and stability in the Tregs of autoimmune and allergic asthma patients. However, the neddylation activating enzyme inhibitors (NAEi) are toxic when given systemically. By generating a protein drug conjugate (PDC) consisting of a NAEi bound, via cleavable linkers, to a fusion protein of murine IL-2 (to target the drug to Tregs), we were able to use 1000-fold less of the neddylation inhibitor drug than the amount required for therapeutically effective systemic delivery. The PDC was effective in blocking the onset or the progression of disease in several mouse models of autoimmunity (type 1 diabetes, systemic lupus erythematosus, and multiple sclerosis) and a mouse model of allergic asthma in the absence of detectable toxicity. This PDC strategy represents targeted drug delivery at its best where the defect causing the disease was identified, a drug was designed and developed to correct the defect, and the drug was targeted and delivered only to cells that needed it, maximizing safety and efficacy.

    View details for DOI 10.3389/fimmu.2022.1046631

    View details for PubMedID 36569931

  • Gene Expression Analysis of the Pre-Diabetic Pancreas to Identify Pathogenic Mechanisms and Biomarkers of Type 1 Diabetes. Frontiers in endocrinology Yip, L., Fuhlbrigge, R., Alkhataybeh, R., Fathman, C. G. 2020; 11: 609271

    Abstract

    Type 1 Diabetes (T1D) occurs as a result of the autoimmune destruction of pancreatic beta-cells by self-reactive T cells. The etiology of this disease is complex and difficult to study due to a lack of disease-relevant tissues from pre-diabetic individuals. In this study, we performed gene expression analysis on human pancreas tissues obtained from the Network of Pancreatic Organ Donors with Diabetes (nPOD), and showed that 155 genes were differentially expressed by ≥2-fold in the pancreata of autoantibody-positive (AA+) at-risk individuals compared to healthy controls. Only 48 of these genes remained changed by ≥2-fold in the pancreata of established T1D patients. Pathway analysis of these genes showed a significant association with various immune pathways. We were able to validate the differential expression of eight disease-relevant genes by QPCR analysis: A significant upregulation of CADM2, and downregulation of TRPM5, CRH, PDK4, ANGPL4, CLEC4D, RSG16, and FCGR2B was confirmed in the pancreata of AA+ individuals versus controls. Studies have already implicated FCGR2B in the pathogenesis of disease in non-obese diabetic (NOD) mice. Here we showed that CADM2, TRPM5, PDK4, and ANGPL4 were similarly changed in the pancreata of pre-diabetic 12-week-old NOD mice compared to NOD.B10 controls, suggesting a possible role for these genes in the pathogenesis of both T1D and NOD disease. The loss of the leukocyte-specific gene, FCGR2B, in the pancreata of AA+ individuals, is particularly interesting, as it may serve as a potential whole blood biomarker of disease progression. To test this, we quantified FCGR2B expression in peripheral blood samples of T1D patients, and AA+ and AA- first-degree relatives of T1D patients enrolled in the TrialNet Pathway to Prevention study. We showed that FCGR2B was significantly reduced in the peripheral blood of AA+ individuals compared to AA- controls. Together, these findings demonstrate that gene expression analysis of pancreatic tissue and peripheral blood samples can be used to identify disease-relevant genes and pathways and potential biomarkers of disease progression in T1D.

    View details for DOI 10.3389/fendo.2020.609271

    View details for PubMedID 33424774

  • Identical and Nonidentical Twins: Risk and Factors Involved in Development of Islet Autoimmunity and Type 1 Diabetes DIABETES CARE Triolo, T. M., Fouts, A., Pyle, L., Yu, L., Gottlieb, P. A., Steck, A. K., Greenbaum, C. J., Atkinson, M., Baidal, D., Battaglia, M., Becker, D., Bingley, P., Bosi, E., Buckner, J., Clements, M., Colman, P., DiMeglio, L., Gitelman, S., Goland, R., Gottlieb, P., Goland, R., Gottlieb, P., Herold, K., Knip, M., Krischer, J., Lernmark, A., Moore, W., Moran, A., Muir, A., Palmer, J., Peakman, M., Philipson, L., Raskin, P., Redondo, M., Rodriguez, H., Russell, W., Spain, L., Schatz, D. A., Sosenko, J., Wentworth, J., Wherrett, D., Wilson, D., Winter, W., Ziegler, A., Anderson, M., Antinozzi, P., Benoist, C., Blum, J., Bourcier, K., Chase, P., Clare-Salzler, M., Clynes, R., Eisenbarth, G., Fathman, C. G., Grave, G., Hering, B., Insel, R., Kaufman, F., Kay, T., Leschek, E., Mahon, J., Marks, J. B., Nanto-Salonen, K., Nepom, G., Orban, T., Parkman, R., Pescovitz, M., Peyman, J., Pugliese, A., Roep, B., Roncarolo, M., Savage, P., Simell, O., Sherwin, R., Siegelman, M., Skyler, J. S., Steck, A., Thomas, J., Trucco, M., Wagner, J., Krischer, J. P., Leschek, E., Rafkin, L., Bourcier, K., Cowie, C., Foulkes, M., Insel, R., Krause-Steinrauf, H., Lachin, J. M., Malozowski, S., Peyman, J., Ridge, J., Savage, P., Skyler, J. S., Zafonte, S. J., Rafkin, L., Sosenko, J. M., Kenyon, N. S., Santiago, Krischer, J. P., Bundy, B., Abbondondolo, M., Adams, T., Amado, D., Asif, Boonstra, M., Burroughs, C., Cuthbertson, D., Deemer, M., Eberhard, C., Fiske, S., Ford, J., Garmeson, J., Guillette, H., Geyer, S., Hays, B., Henderson, C., Henry, M., Heyman, K., Hsiao, B., Karges, C., Keaton, N., Kinderman, A., Law, P., Leinbach, A., Liu, S., Lloyd, J., Malloy, J., Maddox, K., Martin, J., Miller, J., Milliot, E., Moore, M., Muller, S., Nguyen, T., O'Donnell, R., Roberts, A., Sadler, K., Stavros, T., Tamura, R., Wood, K., Xu, P., Young, K., Alies, P., Badias, F., Baker, A., Bassi, M., Beam, C., Boulware, D., Bounmananh, L., Bream, S., Freeman, D., Gough, J., Ginem, J., Granger, M., Kieffer, M., Lane, P., Linton, C., Nallamshetty, L., Oduah, Parrimon, Y., Paulus, K., Pilger, J., Ramiro, J., Ritzie, A., Sharma, A., Shor, A., Song, X., Terry, A., Weinberger, J., Wootten, M., Lachin, J. M., Foulkes, M., Harding, P., Krause-Steinrauf, H., McDonough, S., McGee, P. F., Hess, K., Phoebus, D., Quinlan, S., Raiden, E., Batts, E., Buddy, C., Kirpatrick, K., Ramey, M., Shultz, A., Webb, C., Romesco, M., Fradkin, J., Leschek, E., Spain, L., Savage, P., Aas, S., Blumberg, E., Beck, G., Brillon, D., Gubitosi-Klug, R., Laffel, L., Vigersky, R., Wallace, D., Braun, J., Lernmark, A., Lo, B., Mitchell, H., Naji, A., Nerup, J., Orchard, T., Steffes, M., Tsiatis, A., Veatch, R., Zinman, B., Loechelt, B., Baden, L., Green, M., Weinberg, A., Marcovina, S., Palmer, J. P., Weinberg, A., Yu, L., Babu, S., Winter, W., Eisenbarth, G. S., Bingley, P., Clynes, R., DiMeglio, L., Eisenbarth, G., Hays, B., Leschek, E., Marks, J., Matheson, D., Rafkin, L., Rodriguez, H., Spain, L., Wilson, D., Redondo, M., Gomez, D., McDonald, A., Pena, S., Pietropaolo, M., Shippy, K., Batts, E., Brown, T., Buckner, J., Dove, A., Hammond, M., Hefty, D., Klein, J., Kuhns, K., Letlau, M., Lord, S., McCulloch-Olson, M., Miller, L., Nepom, G., Odegard, J., Ramey, M., Sachter, E., St Marie, M., Stickney, K., VanBuecken, D., Vellek, B., Webber, C., Allen, L., Bollyk, J., Hilderman, N., Ismail, H., Lamola, S., Sanda, S., Vendettuoli, H., Tridgell, D., Monzavi, R., Bock, M., Fisher, L., Halvorson, M., Jeandron, D., Kim, M., Wood, J., Geffner, M., Kaufman, F., Parkman, R., Salazar, C., Goland, R., Clynes, R., Cook, S., Freeby, M., Gallagher, P. M., Gandica, R., Greenberg, E., Kurland, A., Pollak, S., Wolk, A., Chan, M., Koplimae, L., Levine, E., Smith, K., Trast, J., DiMeglio, L., Blum, J., Evans-Molina, C., Hufferd, R., Jagielo, B., Kruse, C., Patrick, Rigby, M., Spall, M., Swinney, K., Terrell, J., Christner, L., Ford, L., Lynch, S., Menendez, M., Merrill, P., Pescovitz, M., Rodriguez, H., Alleyn, C., Baidal, D., Fay, S., Gaglia, J., Resnick, B., Szubowicz, S., Weir, G., Benjamin, R., Conboy, D., deManbey, A., Jackson, R., Jalahej, H., Orban, T., Ricker, A., Wolfsdorf, J., Zhang, H. H., Wilson, D., Aye, T., Baker, B., Barahona, K., Buckingham, B., Esrey, K., Esrey, T., Fathman, G., Snyder, R., Aneja, B., Chatav, M., Espinoza, O., Frank, E., Liu, J., Perry, J., Pyle, R., Rigby, A., Riley, K., Soto, A., Gitelman, S., Adi, S., Anderson, M., Berhel, A., Breen, K., Fraser, K., Gerard-Gonzalez, A., Jossan, P., Lustig, R., Moassesfar, S., Mugg, A., Ng, D., Prahalod, P., Rangel-Lugo, M., Sanda, S., Tarkoff, J., Torok, C., Wesch, R., Aslan, Buchanan, J., Cordier, J., Hamilton, C., Hawkins, L., Ho, T., Jain, A., Ko, K., Lee, T., Phelps, S., Rosenthal, S., Sahakitrungruang, T., Stehl, L., Taylor, L., Wertz, M., Wong, J., Philipson, L., Briars, R., Devine, N., Littlejohn, E., Grant, T., Gottlieb, P., Klingensmith, G., Steck, A., Alkanani, A., Bautista, K., Bedoy, R., Blau, A., Burke, B., Cory, L., Dang, M., Fitzgerald-Miller, L., Fouts, A., Gage, Garg, S., Gesauldo, P., Gutin, R., Hayes, C., Hoffman, M., Ketchum, K., Logsden-Sackett, N., Maahs, D., Messer, L., Meyers, L., Michels, A., Peacock, S., Rewers, M., Rodriguez, P., Sepulbeda, F., Sippl, R., Steck, A., Taki, Tran, B. K., Tran, T., Wadwa, R. P., Zeitler, P., Barker, J., Barry, S., Birks, L., Bomsburger, L., Bookert, T., Briggs, L., Burdick, P., Cabrera, R., Chase, P., Cobry, E., Conley, A., Cook, G., Daniels, J., DiDomenico, D., Eckert, J., Ehler, A., Eisenbarth, G., Fain, P., Fiallo-Scharer, R., Frank, N., Goettle, H., Haarhues, M., Harris, S., Horton, L., Hutton, J., Jeffrrey, J., Jenison, R., Jones, K., Kastelic, W., King, M. A., Lehr, D., Lungaro, J., Mason, K., Maurer, H., Nguyen, L., Proto, A., Realsen, J., Schmitt, K., Schwartz, M., Skovgaard, S., Smith, J., Vanderwel, B., Voelmle, M., Wagner, R., Wallace, A., Walravens, P., Weiner, L., Westerhoff, B., Westfall, E., Widmer, K., Wright, H., Schatz, D., Abraham, A., Atkinson, M., Cintron, M., Clare-Salzler, M., Ferguson, J., Haller, M., Hosford, J., Mancini, D., Rohrs, H., Silverstein, J., Thomas, J., Winter, W., Cole, G., Cook, R., Coy, R., Hicks, E., Lewis, N., Marks, J., Pugliese, A., Blaschke, C., Matheson, D., Pugliese, A., Sanders-Branca, N., Arce, R. A., Cisneros, M., Sabbag, S., Moran, A., Gibson, C., Fife, B., Hering, B., Kwong, C., Leschyshyn, J., Nathan, B., Pappenfus, B., Street, A., Boes, M. A., Eck, P. S., Finney, L., Fischer, A. T., Martin, A., Muzamhindo, J. C., Rhodes, M., Smith, J., Wagner, J., Wood, B., Becker, D., Delallo, K., Diaz, A., Elnyczky, B., Libman, Pasek, B., Riley, K., Trucco, M., Copemen, B., Gwynn, D., Toledo, F., Rodriguez, H., Bollepalli, S., Diamond, F., Eyth, E., Henson, D., Lenz, A., Shulman, D., Raskin, P., Adhikari, S., Dickson, B., Dunnigan, E., Lingvay, Pruneda, L., Ramos-Roman, M., Raskin, P., Rhee, C., Richard, J., Siegelman, M., Sturges, D., Sumpter, K., White, P., Alford, M., Arthur, J., Aviles-Santa, M. L., Cordova, E., Davis, R., Fernandez, S., Fordan, S., Hardin, T., Jacobs, A., Kaloyanova, P., Lukacova-Zib, Mirfakhraee, S., Mohan, A., Noto, H., Smith, O., Torres, N., Wherrett, D., Balmer, D., Eisel, L., Kovalakovska, R., Mehan, M., Sultan, F., Ahenkorah, B., Cevallos, J., Razack, N., Ricci, J. M., Rhode, A., Srikandarajah, M., Steger, R., Russell, W. E., Black, M., Brendle, F., Brown, A., Moore, D., Pittel, E., Robertson, A., Shannon, A., Thomas, J. W., Herold, K., Feldman, L., Sherwin, R., Tamborlane, W., Weinzimer, S., Toppari, J., Kallio, T., Karkkainen, M., Mantymaki, E., Niininen, T., Nurmi, B., Rajala, P., Romo, M., Suomenrinne, S., Nanto-Salonen, K., Simell, O., Simell, T., Bosi, E., Battaglia, M., Bianconi, E., Bonfanti, R., Grogan, P., Laurenzi, A., Martinenghi, S., Meschi, F., Pastore, M., Falqui, L., Muscato, T. M., Viscardi, M., Bingley, P., Castleden, H., Farthing, N., Loud, S., Matthews, C., McGhee, J., Morgan, A., Pollitt, J., Elliot-Jones, R., Wheaton, C., Knip, M., Siljander, H., Suomalainen, H., Colman, P., Healy, F., Mesfin, S., Redl, L., Wentworth, J., Willis, J., Farley, M., Harrison, L., Perry, C., Williams, F., Mayo, A., Paxton, J., Thompson, Volin, L., Fenton, C., Carr, L., Lemon, E., Swank, M., Luidens, M. K., Salgam, M., Sharma, Schade, D., King, C., Carano, R., Heiden, J., Means, N. D., Holman, L., Thomas, Madrigal, D., Muth, T., Martin, C. L., Plunkett, C., Ramm, C., Auchus, R. J., Lane, W., Avots, E., Buford, M., Hale, C., Hoyle, J., Lane, B., Muir, A., Shuler, S., Raviele, N., Ivie, E., Jenkins, M., Lindsley, K., Hansen, Fadoju, D. O., Felner, E., Bode, B., Hosey, R., Sax, J., Jefferies, C., Mannering, S., Prentis, R., She, J. X., Stachura, M., Hopkins, D., Williams, J., Steed, L., Asatapova, E., Nunez, S., Knight, S., Dixon, P., Ching, J., Donner, T., Longnecker, S., Abel, K., Arcara, K., Blackman, S., Clark, L., Cooke, D., Plotnick, L., Levin, P. A., Bromberger, L., Klein, K., Sadurska, K., Allen, C., Michaud, D., Snodgrass, H., Burghen, G., Chatha, S., Clark, C., Silverberg, J., Wittmer, C., Gardner, J., LeBoeuf, C., Bell, P., McGlore, O., Tennet, H., Alba, N., Carroll, M., Baert, L., Beaton, H., Cordell, E., Haynes, A., Reed, C., Lichter, K., McCarthy, P., McCarthy, S., Monchamp, T., Roach, J., Manies, S., Gunville, F., Marosok, L., Nelson, T., Ackerman, K., Rudolph, J., Stewart, M., McCormick, K., May, S., Falls, T., Barrett, T., Dale, K., Makusha, L., McTernana, C., K, P., Sullivan, K., Narendran, P., Robbie, J., Smith, D., Christensen, R., Koehler, B., Royal, C., Arthur, T., Houser, H., Renaldi, J., Watsen, S., Wu, P., Lyons, L., House, B., Yu, J., Holt, H., Nation, M., Vickers, C., Watling, R., Heptulla, R., Trast, J., Agarwal, C., Newell, D. J., Katikaneni, R., Gardner, C., A, D., Rio, A., Logan, H., Collier, C., Rishton, G., Whalley, A., Ali, S., Ramtoola, T., Quattrin, L., Mastrandea, A. J., House, M., Ecker, C., Huang, C., Gougeon, J., D, H., Pacuad, D., Dunger, J., May, C., O'Brien, C., Acerini, B., Salgin, A., Thankamony, R., Williams, J., Buse, G., Fuller, M., Duclos, J., Tricome, H., Brown, D., Pittard, D., Bowlby, A., Blue, T., Headley, S., Bendre, K., Lewis, K., Sutphin, C., Soloranzo, J., Puskaric, H., Madison, M., Rincon, M., Carlucci, R., B, S., Rusk, E., Tessman, D. M., Huffman, H., Abrams, B., Biederman, M. D., Jones, Leathers, W., Brickman, P., Petrie, D., Zimmerman, J., Howard, L., Miller, R., Alemzadeh, D., Mihailescu, R., Melgozza-Walker, N., Abdulla, C., Boucher-Berry, D., Ize-Ludlow, R., Levy, C., Swenson, B. N., Crimmins, D., Edler, T., Weis, C., Schultz, D., Rogers, D., Latham, C., Mawhorter, C., Switzer, W., Spencer, P., Konstantnopoulus, S., Broder, J., Klein, L., Knight, L., Szadek, G., Welnick, B., Thompson, R., Hoffman, A., Revell, J., Cherko, K., Carter, E., Gilson, J., Haines, G., Arthur, B., Bowen, W. B., Zipf, P., Graves, R. A., Lozano, D., Seiple, K., Spicer, A., Chang, J., Fregosi, J., Harbinson, C., Paulson, S., Stalters, P., Wright, D., Zlock, A. E., Freeth, J., Victory, H., Maheshwari, A., Maheshwari, T., Holmstrom, J., Bueno, R., Arguello, J., Ahern, L., Noreika, Watson, S., Hourse, P., Breyer, C., Kissel, Y., Nicholson, M., Pfeifer, S., Almazan, J., Bajaj, M., Quinn, K., Funk, J., McCance, E., Moreno, R., Veintimilla, A., Wells, J., Cook, S., Trunnel, J., Henske, S., Desai, K., Frizelis, F., Khan, R., Sjoberg, K., Allen, P. P., Manning, G., Hendry, B., Taylor, S., Jones, W., Strader, M. E., Bencomo, T., Bailey, L., Bedolla, C., Roldan, C., Moudiotis, B., Vaidya, C., Anning, S., Bunce, S., Estcourt, E., Folland, E., Gordon, C., Harrill, J., Ireland, J., Piper, L., Scaife, K., Sutton, S., Wilkins, M., Costelloe, J., Palmer, L., Casas, C., Miller, M., Burgard, C., Erickson, J., Hallanger-Johnson, P., Clark, W., Taylor, A., Lafferty, S., Gillett, C., Nolan, M., Pathak, L., Sondrol, T., Hjelle, S., Hafner, J., Kotrba, R., Hendrickson, A. P., Cemeroglu, T., Symington, M., Daniel, Y., Appiagyei-Dankah, D. C., Postellon, M. S., Racine, L., Kleis, K., Barnes, S. E., Godwin, H., McCullough, K., Shaheen, G., Buck, L., Noel, M. L., Warren, S., Weber, S. M., Parker, Gillespie, B. A., Nelson, C., Frost, J., Amrhein, E. C., Moreland, A., Hayes, J., Peggram, J., Aisenberg, M. E., Riordan, J., Zasa, E., Cummings, K., Scott, T., Pinto, A., Mokashi, K., McAssey, E., Helden, P., Hammond, L., Dinning, S., Rahman, S., Ray, C., Dimicri, S., Guppy, H., Nielsen, C. K., Vogel, C., Ariza, L., Morales, Y. T., Chang, R. A., Gabbay, L., Ambrocio, L., Manley, R., Nemery, W., Charlton, P., Smith, L., Kerr, B., Steindel-Kopp, M., Alamaguer, D., Liljenquist, G., Browning, T., Coughenour, M. B., Sulk, E., Tsalikan, M., Tansey, J., Cabbage, N., Dixit, S., Pasha, M., King, K., Adcock, H., Atterberry, L., Fox, K., Englert, N., Mauras, J., Permuy, K., Sikes, T., Berhe, B., Guendling, L., McLennan, L., Paganessi, C., Murphy, M. B., Draznin, M., Kamboj, S., Sheppard, Lewis, L., Coates, W., Moore, G., Babar, J., Bedard, D., Brenson-Hughes, J., Cernich, M., Clements, R., Duprau, S., Goodman, L., Hester, L., Huerta-Saenz, A., Karmazin, T., Letjen, S., Raman, D., Morin, W., Bestermann, E. J., Morawski, J. L., White, A., Brockmyer, R., Bays, S., Campbell, A., Stapleton, N., Stone, A., Donoho, H., Everett, H., Hensley, M., Johnson, C., Marshall, N., Skirvin, P., R, T., Williams, L., Ray, C., Wolverton, D. A., Nickels, C., Dothard, P. W., Speiser, M., Pellizzari, L., Bokor, K., Izuora, S., Abdelnour, P., Cummings, S., Paynor, M., Leahy, M., Riedl, S., Shockley, R., Saad, T., Briones, S., Casella, C., Herz, K., Walsh, J., Greening, F., Hay, S., Hunt, N., Sikotra, L., Simons, D. G., Karounos, R., Oremus, L., Dye, L., Myers, D., Ballard, W., Miers, R., Sparks, K. M., Thraikill, K., Edwards, J., Fowlkes, S., Kemp, A., Morales, L., Holland, L., Johnson, P., Paul, A., Ghatak, K., Phelen, H., Leyland, T., Henderson, D., Brenner, E., Oppenheimer, Mamkin, C., Moniz, C., Clarson, M., Lovell, A., Peters, Ruelas, D., Borut, D., Burt, M., Jordan, S., Castilla, P., Flores, M., Ruiz, L., Hanson, J., Green-Blair, R. J., Sheridan, K. A., Wintergerst, G., Pierce, A., Omoruyi, M., Foster, S., Kingery, A., Lunsford, Cervantes, T., Parker, P., Price, J., Urben, Doughty, H., Haydock, Parker, P., Bergman, S., Duncum, C., Rodda, A., Perelman, R., Calendo, C., Barrera, E., Arce-Nunez, Y., Martinez, M., De la Portilla, Cardenas, L., Garrido, M., Villar, R., Lorini, E., Calandra, G., D'Annuzio, K., Perri, N., Minuto, C., Rebora, R., Callegari, O., Ali, J., Kramer, B., Auble, S., Cabrera, P., Donohoue, R., Fiallo-Scharer, M., Hessner, P., Wolfgram, A., Kansra, N., Bettin, R., McCuller, A., Miller, S., Accacha, J., Corrigan, E., Fiore, R. L., Levine, T. A., Mahoney, C., Polychronakos, Gagne, H., Starkman, M., Fox, D., Chin, F., Melchionne, L. A., Silverman, Marshall, L., Cerracchio, J., Cruz, A., Viswanathan, J., Wilson, S., Chalew, S., Valley, S., Layburn, A., Lala, P., Clesi, M., Genet, G., Uwaifo, A., Charron, T., Allerton, W., Cefalu, L., Melendez-Ramirez, R., Richards, C., Alleyn, E., Gustafson, M., Lizanna, J., Wahlen, S., Aleiwe, M., Hansen, H., Wahlen, C. J., Levy, A., Bonaccorso, R., Rapaport, Y., Tomer, D., Chia, M., Goldis, L., Iazzetti, M., Klein, C., Levister, L., Waldman, E., Wallach, M. O., Regelmann, Z., Antal, M., Aranda, C., Reynholds, A., Vinik, P., Barlow, M., Bourcier, M. L., Nevoret, J., Couper, S., Beresford, N., Thalagne, H., Roper, J., Gibbons, J., Hill, S., Balleaut, C., Brennan, J., Ellis-Gage, L., Fear, T., Gray, L., Jones, C., McNerney, L., Pointer, N., Price, K., Few, D., Tomlinson, N., Leech, D., Wake, C., Owens, M., Burns, J., Wotherspoon, A., Murray, K., Short, G., Curry, S., Kelsey, J., Lawson, J., Porter, S., Stevens, E., Thomson, S., Winship, L., Wynn, E., Wiltshire, J., Krebs, P., Cresswell, H., Faherty, C., Ross, L., Denvir, J., Drew, T., Randell, P., Mansell, S. A., Bell, S., Butler, Y., Hooton, H., Navarra, A., Roper, G., Babington, L., Crate, H., Cripps, A., Ledlie, C., Moulds, R., Norton, B., Petrova, O., Silkstone, C., Smith, K., Ghai, M., Murray, Viswanathan, M., Henegan, O., Kawadry, J. A., Olson, L., Patterson, T., Ahmad, B., Flores, D., Domek, S., Domek, K., Copeland, M., George, J., Less, T., Davis, M., Short, A., Dwarakanathan, P., O'Donnell, B., Boerner, L., Larson, M., Phillips, M., Rendell, K., Larson, C., Smith, K., Zebrowski, L., Kuechenmeister, M., Thevarayapillai, M., Daniels, H., Speer, N., Forghani, R., Quintana, C., Reh, A., Bhangoo, P., Desrosiers, L., Ireland, T., Misla, C., Torres, S., Wells, J., Villar, M., Yu, D., Berry, D., Cook, J., Soder, A., Powell, M., Ng, M., Morrison, Z., Haslam, M., Lawson, B., Bradley, J., Courtney, C., Richardson, C., Watson, E., Keely, D., DeCurtis, M., Vaccarcello-Cruz, Z., Torres, K., Sandberg, H., Hsiang, B., Joy, D., McCormick, A., Powell, H., Jones, J., Bell, S., Hargadon, S., Hudson, M., Kummer, S., Sauder, E., Sutton, K., Gensel, R., Aguirre-Castaneda, Benavides, L. D., Hemp, S., Allen, J., Stear, E., Davis, T., Jones, A., Roberts, J. A., Dart, N., Paramalingam, L. E., Katz, L. N., Chaudhary, K. M., Murphy, S. M., Willi, B., Schwartzman, C., Kapadia, D., Larson, D., McClellan, G., Shaibai, L. A., Kelley, G., Villa, C., Kelley, R., Diamond, M., Kabbani, T., Dajani, F., Hoekstra, M., Magorno, J., Holst, Chauhan, N., Wilson, P., Bononi, M., Sperl, A., Millward, M., Eaton, L., Dean, J., Olshan, H., Renna, C., Milliard, B. L., Bacon, J. B., Quintos, L. S., Topor, S., Bialo, B., Bancroft, A. G., Soto, W., Lagarde, H., Lockemer, T., Vanderploeg, M. A., Ibrahim, M., Huie, Sanchez, R., Edelen, R., Marchiando, J., Palmer, T., Repas, M., Wasson, P., Auker, J., Culbertson, T., Kieffer, D., Voorhees, T., Borgwardt, L., DeRaad, K., Eckert, E., Isaacson, H., Kuhn, A., Carroll, M., Schubert, G., Francis, S., Hagan, T., Le, M., Penn, E., Wickham, C., Leyva, K., Rivera, J., Padilla, Rodriguez, N., Jospe, J., Czyzyk, B., Johnson, U., Nadgir, N., Marlen, G., Prakasam, C., Rieger, N., Glaser, E. C., Heiser, B., Harris, C., Foster, H., Slater, K., Wheeler, D. L., Donaldson, M., Murray, D. E., Hale, R., Tragus, D. R., Word, J., Lynch, L., Pankratz, W., Rogers, R., Newfield, S., Holland, M., Hashiguchi, M., Gottschalk, A., Philis-Tsimikas, R., Rosal, S., Franklin, S. M., Guardado, N., Bohannon, M., Garcia, T., Aguinaldo, J., Phan, Barraza, D., Cohen, J., Pinsker, U., Khan, J., Wiley, L., Jovanovic, P., Misra, M., Wright, D., Cohen, K., Huang, M., Skiles, S., Maxcy, C., Pihoker, K., Cochrane, J., Fosse, S., Kearns, M., Klingsheim, N., Wright, L., Viles, H., Smith, S., Heller, M., Cunningham, A., Daniels, L., Zeiden, J., Field, R., Walker, K. J., Griffin, L., Bartholow, C., Erickson, J., Howard, B., Krabbenhoft, C., Sandman, A., Vanveldhuizen, J., Wurlger, A., Zimmerman, K., Hanisch, L., Davis-Keppen, A., Cotterill, J., Kirby, M., Harris, A., Schmidt, C., Kishiyama, C., Flores, J., Milton, W., Martin, C., Whysham, A., Yerka, T., Freels, J. M., Hassing, J., Webster, R., Green, P., Carter, J., Galloway, D., Hoelzer, S., Roberts, S., Said, P., Sullivan, H. F., Allen, E., Reiter, E., Feinberg, C., Johnson, L. A., Newhook, D., Hagerty, N. H., White, L., Levandoski, J., Kyllo, M., Johnson, C., Benoit, P., Iyer, F., Diamond, H., Hosono, S., Jackman, L., Barette, P., Jones, Sills, S., Bzdick, J., Bulger, R., Weinstock, Douek, R., Andrews, G., Modgill, G., Gyorffy, L., Robin, N., Vaidya, S., Crouch, K., O'Brien, C., Thompson, N., Thorne, J., Blumer, J., Kalic, L., Klepek, J., Paulett, B., Rosolowski, J., Horner, M., Watkins, J. L., Casey, K., Carpenter, C., Burns, J., Horton, C., Pritchard, D., Soetaert, A. G., Wynne, C., Chin, O. Y., Molina, C., Patel, R., Senguttuvan, M., Wheeler, O., Furet, C., Steuhm, D. H., Jelley, S., Goudeau, L., Chalmers, D., Greer, C., Panagiotopoulos, D. L., Metzger, D., Nguyen, M., Horowitz, M. P., Christiansen, E., Glades, C., Morimoto, M., Macarewich, R., Norman, K., Patin, C., Vargas, A., Barbanica, A., Yu, P., Vaidyanathan, W., Osborne, R., Mehra, S., Kaster, S., Neace, J., Horner, G., Reeves, C., Cordrey, L., Marrs, T., Miller, S., Dowshen, D., Doyle, S., Walker, D., Catte, H., Dean, M., Drury-Brown, B., Hackman, M. C., Lee, S., Malkani, K., Cullen, K., Johnson, P., Hampton, M., McCarrell, C., Curtis, E., Paul, Z. Y., Type 1 Diabet TrialNet Study Grp 2019; 42 (2): 192–99

    Abstract

    There are variable reports of risk of concordance for progression to islet autoantibodies and type 1 diabetes in identical twins after one twin is diagnosed. We examined development of positive autoantibodies and type 1 diabetes and the effects of genetic factors and common environment on autoantibody positivity in identical twins, nonidentical twins, and full siblings.Subjects from the TrialNet Pathway to Prevention Study (N = 48,026) were screened from 2004 to 2015 for islet autoantibodies (GAD antibody [GADA], insulinoma-associated antigen 2 [IA-2A], and autoantibodies against insulin [IAA]). Of these subjects, 17,226 (157 identical twins, 283 nonidentical twins, and 16,786 full siblings) were followed for autoantibody positivity or type 1 diabetes for a median of 2.1 years.At screening, identical twins were more likely to have positive GADA, IA-2A, and IAA than nonidentical twins or full siblings (all P < 0.0001). Younger age, male sex, and genetic factors were significant factors for expression of IA-2A, IAA, one or more positive autoantibodies, and two or more positive autoantibodies (all P ≤ 0.03). Initially autoantibody-positive identical twins had a 69% risk of diabetes by 3 years compared with 1.5% for initially autoantibody-negative identical twins. In nonidentical twins, type 1 diabetes risk by 3 years was 72% for initially multiple autoantibody-positive, 13% for single autoantibody-positive, and 0% for initially autoantibody-negative nonidentical twins. Full siblings had a 3-year type 1 diabetes risk of 47% for multiple autoantibody-positive, 12% for single autoantibody-positive, and 0.5% for initially autoantibody-negative subjects.Risk of type 1 diabetes at 3 years is high for initially multiple and single autoantibody-positive identical twins and multiple autoantibody-positive nonidentical twins. Genetic predisposition, age, and male sex are significant risk factors for development of positive autoantibodies in twins.

    View details for DOI 10.2337/dc18-0288

    View details for Web of Science ID 000457193000013

    View details for PubMedID 30061316

    View details for PubMedCentralID PMC6341285

  • Low-Dose Anti-Thymocyte Globulin (ATG) Preserves -Cell Function and Improves HbA(1c) in New-Onset Type 1 Diabetes DIABETES CARE Haller, M. J., Schatz, D. A., Skyler, J. S., Krischer, J. P., Bundy, B. N., Miller, J. L., Atkinson, M. A., Becker, D. J., Baidal, D., DiMeglio, L. A., Gitelman, S. E., Goland, R., Gottlieb, P. A., Herold, K. C., Marks, J. B., Moran, A., Rodriguez, H., Russell, W., Wilson, D. M., Greenbaum, C. J., Type 1 Diabet TrialNet ATG-GCSF St 2018; 41 (9): 1917–25

    Abstract

    A pilot study suggested that combination therapy with low-dose anti-thymocyte globulin (ATG) and pegylated granulocyte colony-stimulating factor (GCSF) preserves C-peptide in established type 1 diabetes (T1D) (duration 4 months to 2 years). We hypothesized that 1) low-dose ATG/GCSF or 2) low-dose ATG alone would slow the decline of β-cell function in patients with new-onset T1D (duration <100 days).A three-arm, randomized, double-masked, placebo-controlled trial was performed by the Type 1 Diabetes TrialNet Study Group in 89 subjects: 29 subjects randomized to ATG (2.5 mg/kg intravenously) followed by pegylated GCSF (6 mg subcutaneously every 2 weeks for 6 doses), 29 to ATG alone (2.5 mg/kg), and 31 to placebo. The primary end point was mean area under the curve (AUC) C-peptide during a 2-h mixed-meal tolerance test 1 year after initiation of therapy. Significance was defined as one-sided P value < 0.025.The 1-year mean AUC C-peptide was significantly higher in subjects treated with ATG (0.646 nmol/L) versus placebo (0.406 nmol/L) (P = 0.0003) but not in those treated with ATG/GCSF (0.528 nmol/L) versus placebo (P = 0.031). HbA1c was significantly reduced at 1 year in subjects treated with ATG and ATG/GCSF, P = 0.002 and 0.011, respectively.Low-dose ATG slowed decline of C-peptide and reduced HbA1c in new-onset T1D. Addition of GCSF did not enhance C-peptide preservation afforded by low-dose ATG. Future studies should be considered to determine whether low-dose ATG alone or in combination with other agents may prevent or delay the onset of the disease.

    View details for DOI 10.2337/dc18-0494

    View details for Web of Science ID 000442337700017

    View details for PubMedID 30012675

    View details for PubMedCentralID PMC6105329

  • Identification of a common immune regulatory pathway induced by small heat shock proteins, amyloid fibrils, and nicotine PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Rothbard, J. B., Rothbard, J. J., Soares, L., Fathman, C., Steinman, L. 2018; 115 (27): 7081–86
  • Identification of a common immune regulatory pathway induced by small heat shock proteins, amyloid fibrils, and nicotine. Proceedings of the National Academy of Sciences of the United States of America Rothbard, J. B., Rothbard, J. J., Soares, L., Fathman, C. G., Steinman, L. 2018

    Abstract

    Although certain dogma portrays amyloid fibrils as drivers of neurodegenerative disease and neuroinflammation, we have found, paradoxically, that amyloid fibrils and small heat shock proteins (sHsps) are therapeutic in experimental autoimmune encephalomyelitis (EAE). They reduce clinical paralysis and induce immunosuppressive pathways, diminishing inflammation. A key question was the identification of the target for these molecules. When sHsps and amyloid fibrils were chemically cross-linked to immune cells, a limited number of proteins were precipitated, including the alpha7 nicotinic acetylcholine receptor (alpha7 NAChR). The alpha7 NAChR is noteworthy among the over 20 known receptors for amyloid fibrils, because it plays a central role in a well-defined immune-suppressive pathway. Competitive binding between amyloid fibrils and alpha-bungarotoxin to peritoneal macrophages (MPhis) confirmed the involvement of alpha7 NAChR. The mechanism of immune suppression was explored, and, similar to nicotine, amyloid fibrils inhibited LPS induction of a common set of inflammatory cytokines while inducing Stat3 signaling and autophagy. Consistent with this, previous studies have established that nicotine, sHsps, and amyloid fibrils all were effective therapeutics in EAE. Interestingly, B lymphocytes were needed for the therapeutic effect. These results suggest that agonists of alpha7 NAChR might have therapeutic benefit for a variety of inflammatory diseases.

    View details for PubMedID 29915045

  • Effect of Oral Insulin on Prevention of Diabetes in Relatives of Patients With Type 1 Diabetes A Randomized Clinical Trial JAMA-JOURNAL OF THE AMERICAN MEDICAL ASSOCIATION Greenbaum, C., Atkinson, M., Baidal, D., Battaglia, M., Bingley, P., Bosi, E., Buckner, J., Clements, M., Colman, P., DiMeglio, L., Evans-Molina, C., Gitelman, S., Goland, R., Gottlieb, P., Herold, K., Knip, M., Krischer, J., Lernmark, A., Moore, W., Moran, A., Muir, A., Palmer, J., Peakman, M., Philipson, L., Raskin, P., Redondo, M., Rodriguez, H., Russell, W., Spain, L., Schatz, D. A., Sosenko, J., Wherrett, D., Wilson, D., Winter, W., Ziegler, A., Anderson, M., Antinozzi, P., Benoist, C., Blum, J., Bourcier, K., Chase, P., Clare-Salzler, M., Clynes, R., Cowie, C., Eisenbarth, G., Fathman, C. G., Grave, G., Harrison, L., Hering, B., Insel, R., Jordan, S., Kaufman, F., Kay, T., Kenyon, N., Klines, R., Lachin, J., Leschek, E., Mahon, J., Marks, J. B., Monzavi, R., Nanto-Salonen, K., Nepom, G., Orban, T., Parkman, R., Pescovitz, M., Peyman, J., Pugliese, A., Ridge, J., Roep, B., Roncarolo, M., Savage, P., Simell, O., Sherwin, R., Siegelman, M., Skyler, J. S., Thomas, J., Trucco, M., Wagner, J., Bourcier, K., Greenbaum, C. J., Krischer, J. P., Leschek, E., Rafkin, L., Spain, L., Cowie, C., Foulkes, M., Insel, R., Krause-Steinrauf, H., Lachin, J. M., Malozowski, S., Peyman, J., Ridge, J., Savage, P., Skyler, J. S., Zafonte, S. J., Greenbaum, C. J., Rafkin, L., Sosenko, J., Skyler, J. S., Kenyon, N. S., Santiago, I., Krischer, J. P., Bundy, B., Abbondondolo, M., Adams, T., Asif, D., Boonstra, M., Boulware, D., Bundy, B., Burroughs, C., Cuthbertson, D., Eberhard, C., Fiske, S., Ford, J., Garmeson, J., Guillette, H., Geyer, S., Hays, B., Henderson, C., Henry, M., Heyman, K., Hsiao, B., Karges, C., Kinderman, A., Lane, L., Leinbach, A., Liu, S., Lloyd, J., Malloy, J., Maddox, K., Martin, J., Miller, J., Moore, M., Muller, S., Nguyen, T., O'Donnell, R., Parker, M., Pereyra, M. J., Reed, N., Roberts, A., Sadler, K., Stavros, T., Tamura, R., Wood, K., Xu, P., Young, K., Alies, P., Badias, F., Baker, A., Bassi, M., Beam, C., Boulware, D., Bounmananh, L., Bream, S., Deemer, M., Freeman, D., Gough, J., Ginem, J., Granger, M., Holloway, M., Kieffer, M., Lane, P., Law, P., Linton, C., Nallamshetty, L., Oduah, V., Parrimon, Y., Paulus, K., Pilger, J., Ramiro, J., Ritzie, A., Sharma, A., Shor, A., Song, X., Terry, A., Weinberger, J., Wootten, M., Lachin, J. M., Harding, M., Krause-Steinrauf, H., McDonough, S., Mcgee, P. F., Hess, K., Phoebus, D., Quinlan, S., Raiden, E., Fradkin, J., Leschek, E., Spain, L., Cowie, C., Malozowski, S., Savage, P., Beck, G., Blumberg, E., Gubitosi-Klug, R., Laffel, L., Veatch, R., Wallace, D., Braun, J., Brillon, D., Lernmark, A., Lo, B., Mitchell, H., Naji, A., Nerup, J., Orchard, T., Steffes, M., Tsiatis, A., Zinman, B., Loechelt, B., Baden, L., Green, M., Weinberg, A., Marcovina, S., Palmer, J. P., Weinberg, A., Yu, L., Winter, W., Shultz, A., Batts, E., Fitzpatrick, K., Ramey, M., Guerra, R., Webb, C., Caffey, F., Carr, L., Ergun-Longmire, B., Fenton, C., Giebner, D., Johnson, J., Maglionico, D., Marinelli, M., Martin, K., Minnozzi, E., Riley, W., Wilson, M., Gougeon, C., Ho, J., Huang, C., Pacaud, D., Virtanen, H., Craig, C., Ghatak, A., Henderson, T., Leyland, H., Padmore, K., Paul, P., Brickman, W., Halsey-Lyda, M., Petrie, P., Rizzo, D., Steuer, R., Suchyta, K., Torchen, L., Zimmerman, D., Bode, B., Dial, M., Gazaway, K., Hosey, R., Alkanani, A., Barker, J., Barr, M., Blau, A., Burdick, P., Burke, B., Chase, H., Drye, M., Eisenbarth, G., Escobar, E., Fitzgerald-Miller, L., Fouts, A., Gage, V., Gall, E., Goettle, H., Gottlieb, P., Harris, S., Ketchum, K., King, M., Klingensmith, G., Lehr, D., Lehr, J., Lewis, L., Logsden-Sackett, N., Lykens, J., Maahs, D., Michels, A., Pelletier, S., Rihanek, M., Rodriguez, P., Schauwecker, A., Simmons, K., Smith, J., Steck, A., Tran, B., Tran, T., Wadwa, P., Wagner, R., Wright, H., Betancourt, J., Bui, V., DeSalvo, D., Gomez, D., Jake, K., Lynds, J., McCartney, T., McDonald, A., Pena, S., Pietropaolo, M., Redondo, M., Shippy, K., Zheng, X., Allen, L., Batts, E., Brown, T., Buckner, J., Dove, A., Graziano, E., Greenbaum, C., Hao, W., Harrington, R., Hefty, D., Kang, D., Klein, J., Kuhns, K., Lamola, S., Lettau, M., Lord, S., Machmer, H., McCulloch-Olson, M., Miller, L., Odegard, J., Ramey, M., Romasco, M., Russell, B., Sachter, E., Sanda, S., Scheuffele, T., Shultz, A., Snavely, J., St Marie, M., Tobin, M., Tordillos, C., Tridgell, D., VanBuecken, D., Varner, K., Vellek, B., Vendettuoli, H., Vizzutti, M., Webber, C., Wickstrom, N., Ackerman, K., Gunville, F., Nelson, T., Aston, K., Barrett, T., Dale, K., Gray, Z., Kershaw, M., Makusha, L., McTernan, C., Okwu, F., Penny-Thomas, K., Surplice, I., Frattaroli, P., Gihawi, A., Kanumakala, S., Laycock, C., Ramsay, R., Symes, L., Wlazly, D., Healy, F., Bowden, K., Doughty, I., Haydock, H., Kennedy, K., Parker, V., Plimmer, N., Swart, L., Wood, C., Leyva, C., Padilla, J., Rodriguez, I., Ahmad, T., Bhatia, S., Conrad, S., Egli, C., Flores, B., Higa, A., Leong, K., Ng, V., Oakes, S., Olson, J., Blackmore, A., Bradley, B., Cooper, T., Courtney, J., Lawson, M., Richardson, C., Watson, C., Blind, J., Bowden, S., Bowen, B., Carter, K., Cecrle, M., Chaudhari, M., Cherko, J., Dyer, J., Ellis, K., Haines, J., Hapanowicz, C., Hardin, D., Henwood, M., Hoffman, R., Lamberjack, K., Leary, J., Lewis, S., Mhaskar, R., Rowe, S., Schoeginger, L., Stiltner, T., Bock, M., Castaneda, N., Fisher, L., Geffner, M., Gonzalez, M., Halvorson, M., Hisakado, M., Jeandron, D., Kaufman, F., Kwan, K., Marks, L., Medina, B., Miller, D., Monzavi, R., Ng, E., Parkman, R., Salazar, C., Tjauw, A., Wood, J., Woods, A., Xu, Y., Chalew, S., Daniels, J., Gomez, R., Lala, A., Layburn, S., Meyers, M., Sonnier, S., Valley, S., Ayala, N., Bhangoo, A., Bowen, S., Cervisi, J., Chase, Y., Clark, S., Daniels, M., Flannery, T., Forghani, N., Humphrey, L., Krause, G., Less, J., Lester, S., Magedman, G., Montgomery, K., Preasmyer, S., Quintana, R., Randhawa, R., Reh, C., Speer, H., Stockton, W., Sutton, F., Tran, A., Trihn, L., Tu, K., Varni, N., Ackermann, A., Capella, C., Clark, C., Gralewski, K., Hawkes, C., Kim, R., Katz, L., Liilii, R., McKenzie, O., Murphy, K., Norris, M., Orellana, J., Schwartzman, B., Sheth, S., Volpe, R., Willi, S., Healy, F., Heenan, H., Hodgman, S., Kendall, D., Logan, F., Lunt, H., Willis, J., Crimmins, N., Elder, D., Lagory, D., Schultz, C., Stamper, M., Weis, T., Armbruster, D., Klein, J., Konstantinopoulus, P., Latham, D., Markle, T., Mawhorter, C., Rizk, M., Rogers, D., Schmidt, N., Switzer, C., Bokor, L., DeMers, C., Pellizzari, M., Speiser, P., Clynes, R., Cook, S., Dinapoli, G., Eng, C., Engelman, H., Freeby, M., Gallagher, M., Gandica, R., Goland, R., Greenberg, E., Jezioro, J., Kurland, A., Leibel, N., Levine, E., Maher, C., Nieva, D., Parra, Z., Pollak, S., Pope, K., Ridder, R., Scotto, M., Softness, B., Uche, E., Williams, K., Wolk, A., Zhang, H., Burns, C., Casey, J., Doty, B., Horton, J., Moore, H., Pritchard, C., Wynne, A., Brown, L., Cordrey, C., Dowshen, S., Doyle, D., Kidd, G., Marrs, L., Miller, T., Reeves, G., Brousell, C., Healy, F., Hendry, G., Manning, P., Willis, J., Ali, A., Collier, H., Del Rio, A., Gardner, C., Logan, A., Patel, I., Ramtoola, S., Rishton, C., Robinson, D., Whalley, G., Childs, E., Dothard, C., Jordan, K., Batajoo, R., Kerrigan, J., Nickels, D., Tapiador, C., Van Audenhove, J., Wirthwein, E., Allen, C., Anderson, K., Michaud, D., Sadurska, K., Snodgrass, H., Antich, A., Brown, M., Clark-Stuart, T., Cossen, K., Fadoju, D., Felner, E., Greber, G., Ivie, E., Jenkins, M., Kwapil, L., Lindsley, K., Muir, A., Panagiotakopoulos, L., Raviele, N., Sanchez, W., Shuler, S., Davis, A., Gillespie, I., Kane, V., Koval, T., Manchester, T., Parker, S., Rulevitch, N., Weber, S., Healy, F., Amrhein, J., Frost, C., Hannah, D., Looper, L., Moreland, E., Nelson, B., Owens, C., Peggram, J., Phillips, J., Reifeis, E., Andel, A., Arrieta, P., Bena, P., Couch, R., Deol, S., Gordon, M., Lieberman, D., Martin, J., Neiley, R., Qureshi, S., Sadiasa, G., Baldwin, J., Helden, E., McAssey, K., Brunskill, C., Clarke, A., Collins, A., Dinning, L., Hammond, P., Idrees, T., Jones, H., Meredith, S., Moull, A., Rahman, S., Ray, S., Annillo, S., Briones, T., Saad, R., Adamsson, A., Iljamo, M., Jokipuu, S., Kallio, T., Karlsson, L., Kero, J., Leppanen, M., Mantymaki, E., Nanto-Salonen, K., Nurmi, B., Rajala, P., Romo, M., Rouhiainen, J., Ruohonen, E., Simell, O., Simell, S., Simell, T., Suomenrinne, S., Toppari, J., Torma, M., Wahlback, J., Cummings, E., Mokashi, A., Murphy, W., Pinto, T., Scott, K., Tiller, M., Allard, M., Anders, J., Blum, J., Book, B., Cox, D., Davies, V., DeYampert, L., DiMeglio, L., Dykstra, E., Effinger, J., Eugster, E., Evans-Molina, C., Ford, L., Fuqua, J., Haddad, N., Hannon, T., Hildinger, M., Hufferd, R., Imel, E., Jagielo, B., Johnson, A., Johnson, N., Kost, B., Kruse, C., Laskowski, P., Leung, E., Mantravadi, M., Melvin, E., Mirmira, R., Mott, L., Mullen, M., Murphy, R., Nabhan, Z., Nebesio, T., Newnum, A., Nicholson-Spall, M., Patrick, V., Pfeiffer, J., Purtlebaugh, D., Rigby, M., Sanchez, J., Sims, E., Swan, E., Acharya, S., Garvey, L., Stormer, J., Bzdick, S., Conboy, P., Doolittle, S., Izquierdo, R., Sills, I., Weinstock, R., Alleyn, C., Baidal, D., Bryant, N., Conboy, D., Demanbey, A., Fay, S., Gaglia, J., Jackson, R., Jalahej, H., Koshy, N., Krishfield, S., Migre, M., Montero, M., Orban, T., Resnick, B., Ricker, A., Szubowicz, S., Turley, J., Weir, G., Wolfsdorf, J., Zhang, H., Balapatebendi, M., Barradell, A., Brunskill, C., Cheney, S., Greening, J., Hay, F., Hunt, S., Punniyakodi, S., Sikotra, N., Sundaram, P., Thanawala, N., Voce, R., Biggs, J., Faherty, A., Healy, F., Jefferies, C., Mannering, S., Prentis, R., Willis, J., Baskerville, T., Cotterill, A., Harris, M., Healy, F., Kirby, J., Healy, F., Cheetham, T., Clark, Y., Cullen, K., Curry, G., Dixon, J., Leech, N., Logan, M., Owens, C., Porter, J., Richardson, V., Short, K., Thomson, E., Wake, D., Wotherspoon, J., Wynn, L., Brennan, C., Ellis-Gage, J., Fear, L., Few, K., Gibbons, J., Gray, T., Hill, J., Jones, L., McNerney, C., Oakley, S., Price, N., Rajan, S., Roper, H., Thalange, N., Tomlinson, D., Babington, G., Coupe, A., Crate, L., Denvir, L., Gazis, T., Hooton, Y., Mansell, P., Petrova, B., Saxton, M., Silkstone, O., Spick, D., Sudera, S., Alleyn, C., Cooper, J., Hall, S., Lizanna, M., Pandya, P., Ramsey, K., Ratti, P., Sanders, C., Stewart, S., Uwaifo, G., Ahmann, A., Bogan, C., Cain, N., Chambers, V., DeFrang, D., Fitch, R., Guttman-Bauman, I., Hanavan, K., Jahnke, K., Klopfenstein, B., Lester, L., Muhly, R., Nicol, L., Raymond, J., Wollam, B., Cooper, T., Cortes, L., Dingle, A., Duff, L., Elio, L., Fedorchenko, L., Goulet-Ingram, R., Gutierrez, J., Hollis, S., Hyderkhan, C., Hyderkhan, M., Kellogg, S., Klugh, S., Langarica, D., Leslie, C., Luna, J., Maceda, D., Miller, A., Nakhle, D., Nakhle, S., Neylon, E., O'Sullivan, M., Palal, B., Pedraza, S., Poynor, S., Ramiro, J., Ramos, M., Rosario, A., Sandry, S., Stricklin, K., Sushenko, N., Torres, P., Valle, M., Vernetti, N., Westerman, K., Alvi, S., Clapham, M., English, P., Greatrex, J., Khan, H., Millward, A., Storr, E., Dart, J., Healy, F., Jones, T., Baldwin, D., Bruckner, J., Conroy, C., Culbertson, J., Edelen, R., Ferkovich, M., Palmer, J., Voorhees, D., Wasson, M., Williams, A., Barbieri, K., Browning, G., Coughenour, T., Heagy, N., Judge, H., Liljenquist, D., Sulik, M., Connon, M., Abid, N., Beckett, R., Bell, P., Harvey, C., McFarland, M., McGlone, O., Tennet, H., Wells, B., Beamish, N., Cavan, D., Cooke, D., Graja, A., Nation, M., Sanders, P., Vickers, C., Walker, J., Watling, R., Healy, F., Estcourt, S., Foden, A., Gordon, E., Harrill, C., Ireland, J., Jones, N., Moudiotis, C., Piper, J., Scaife, L., Vaidya, B., Walton-Salih, E., Whitmore, H., Wilkins, S., Wilkinson, L., Battaglia, M., Belloni, C., Bianconi, E., Bolla, A., Bonfanti, R., Bonopane, M., Bosi, E., Corti, M., Costa, S., Falqui, L., Fontana, B., Galluccio, E., Grogan, P., Laurenzi, A., Lombardoni, C., Martinenghi, S., Meschi, F., Molinari, C., Molteni, L., Monti, L., Pastore, M., Privitera, D., Ragogna, F., Spadoni, S., Stabilini, A., Vecchione, F., Viscardi, M., Al Nofal, A., Albers, D., Austad, S., Austin, S., Bartholow, L., Brantz-Miller, A., Broadbent, M., Brosnahan, J., Cortes, G., Coyne, C., Davis-Keppen, L., Griffin, K., Hahn, D., Hanisch, K., Hanson, D., Hauge, C., Hein, T., Howard, J., Huber, C., Johnson, J., Karmazin, A., Keller, L., Kirschbaum, S., Klinghagen, R., Krabbenhoft, B., Krell, E., Meier, J., Olson, C., Prenger, S., Sandman, C., Shelso, J., Springman, C., Thompson, M., Vandermark, J., Vanveldhuizen, A., Wurgler, J., Zimmerman, A., Alving, E., Benitez, S., Bryant, S., Cochrane, K., DiBlasi, C., Fechner, P., Gama, K., Harry, J., Jacob, S., Kearns, S., Klingsheim, M., Knutzen, S., Kong, A., Koves, I., Loots, B., Malik, F., Mano, E., Martinez, O., Nandi-Munshi, D., Ness, K., O'Connor, R., Pihoker, C., Roth, C., Salehi, P., Semana, S., Sexton, A., Taplin, C., Yaptangco, M., Bull, J., Gormley, S., Jones, K., Redfearn, K., Shackleton, J., Smith, H., Strong, L., Thomas, L., Viles, L., Wright, N., Kordel, J., Agardh, C., Ahlkvist, L., Ask, M., Berggren, S., Borg, H., Gerardsson, J., Gustavsson, B., Hakansson, R., Hansen, M., Hansson, G., Jarvirova, M., Jonsdottir, B., Katsarou, A., Kulinski, M., Larsson, H., Lernmark, A., Lind, A., Lindstrom, M., Lundgren, M., Massadakis, T., Melin, J., Mestan, Z., Mulder, H., Nilsson, C., Rosengren, A., Salami, F., Skarstrand, H., Tekum-Amboh, E., Torn, C., Ulvenhag, U., Wimar, A., Arthur, T., Buchanan, M., Cardoni, C., Christensen, R., Filicetti, M., Gerrard, X., Haven, K., Ioli, M., Jackson, J., Jones, E., Kauk, K., Koehler, B., Nihill, K., Parra, B., Russell, N., Schott, S., Tawney, L., Taylor, L., Waldren, C., Watsen, S., Whitham, L., Atkins, M., Aye, T., Bachrach, L., Baker, B., Barahona, K., Berry, B., Buckingham, B., Chau, C., Crossen, S., DeSalvo, D., Espinoza, O., Esrey, T., Kumar, R., Ly, T., Nally, L., Patel, P., Seeley, H., Shah, A., Shah, S., Soto, A., Stenerson, M., Wilson, D., Kioroglo, Y., Mann, C., Marlen, N., Nadgir, U., Olsen-Wilson, K., Prakasam, G., Bunk, M., Chmiel, R., Fischer, F., Gavrisan, A., Haupt, F., Heinrich, M., Herbst, M., Hivner, S., Hofelich, A., Holzmaier, M., Kriesen, Y., Lagoda, N., Loebner, S., Maison, N., Mau, E., Peplow, C., Puff, R., Ramminger, C., Sebelefsky, C., Walter, M., Warncke, K., Ziegler, A., Zillmer, S., Babar, G., Bedard, J., Bloom, K., Broussard, J., Bruce, C., Cernich, J., Clements, M., Clifton, T., Craig, E., Drees, A., Duprau, R., Feldt, M., Fridlington, A., Goodman, S., Hess, K., Hester, L., Huseman, C., Karmazin, A., Kim, E., King, A., Kover, K., Luetjen, T., Martin, K., McDonough, R., Moore, W., Musick, T., Newman, K., Nichols, C., Peterson, K., Raman, S., Reddig, N., Swiderski, S., Tong, P., Turpin, A., Turpin, A., Ugrasbul, F., Watkins, D., Weigel, S., Whisenhunt, M., Wierson, J., Wilcox, R., Wolfe, D., Zacharko, P., Zebley, J., Albini, C., Bethin, K., Borowski, R., Buchlis, J., Ecker, M., Elsinghorst, H., Fourtner, S., Gartner, L., Gorman, E., House, A., Kraengel, K., Krolczyk, A., Majumdar, I., Marrone, A., Mastrandrea, L., Michalovic, S., Musial, W., Quattrin, T., Russell, M., Rychlicki, L., Shelat, T., Shine, B., Sickau, J., Van der Kloet, E., Young, B., Ahenkorah, B., Balmer, D., Bedford, M., Cevallos, J., Chapman, K., De Lima, S., Duong, T., Eisel, L., Fiset, J., Harrington, J., Kovalakovska, R., Mehan, M., Nguyen, H., Perro, B., Ricci, M., Ricci, M., Roode, A., Sriskandarajah, M., Steger, R., Sultan, F., Wherrett, D., Aslanov, R., Crummell, C., Hagerty, D., Newhook, L., Penney, S., Stokes, J., Beck, J., Copeland, K., George, M., Larson, S., Less, J., Lopez, C., Roof, A., Schanuel, J., Sparling, D., Tryggestad, J., Lee, M., Shaw, B., Bobik, C., Bollepalli, S., Brownstein, R., Diamond, F., Eyth, E., Henson, D., Iyer, P., Jorgensen, V., Martin, J., Norman, J., O'Brian, J., Rodriguez, H., Shulman, D., Smith, L., Steinbrueck, J., Terry, A., Tindell, S., Garza, A., Grohman, C., Hale, D., Kral, J., Tragus, R., Word, D., Barrett, T., Holloway, S., Lighton, B., Morgan, R., Narendran, P., Smith, D., Ambrose, M., Chin, C., Durazo, G., Gonzalez-Garcia, Z., Gordon, M., Hollis, M., Senguttuvan, R., Stuehm, C., Wheeler, M., Aitken, R., Bingley, P., Castleden, H., Farthing, N., Hughes, T., Loud, S., Matthews, C., Mcgee, J., Morgan, A., Munoz, P., Pollitt, J., Pope, C., Rouquette, C., Thorne, B., Baynham, S., Gardiner, S., Genereaux, D., Jantzen, C., Lai, J., Lutley, P., Mammon, B., Membreve, J., Metzger, D., Morrison, K., Nguyen, D., Panagiotopoulos, C., Ronsley, C., Roston, A., Suen, J., Abalos, M., Adi, S., Anderson, M., Auerback, G., Berhel, A., Bomberg, E., Breen, K., Buchanan, J., Cook, A., Cakmak, A., Ferrara, C., Fields, S., Finney, Z., Fraser, K., Gonzalez, A., Ghods, S., Gitelman, S., Hamid, L., Hamilton, C., Hawkins, L., Honrada, R., Huang, A., Jain, A., Jossan, P., Ko, K., Larocque, N., Lilley, B., Long, R., Lustig, D. R., Ly, E., Malik, A., Melaku, A., Moassesfar, S., Mugg, A., Ng, D., Ng, D., O'Brien, C., Perez, E., Phelps, S., Prahalod, P., Ramos, E., Lugo, M., Rodriguez, T., Arao, A., Demeterco-Berggren, C., Duong, J., Gottschalk, M., Hashiguchi, M., Kelly, T., Marinkovic, M., Marois, N., Newfield, R., Phillips, S., Rosenblum, D., Abdullah, N., Dunger, D., Gilbert, A., Guy, C., Hendricks, E., May, J., O'Brien, C., Salgin, B., Thankamony, A., Vyse, N., Watts, A., Whitehead, K., Whitehead, L., Willemsen, R., Williams, R., Wingate, D., Devine, N., Gannon, G., Grant, T., Letourneau, L., Littlejohn, E., Norstrom, M., O'Malley, T., Philipson, L., Abraham, A., Agustin, E., Albanese-O'Neill, A., Beltz, S., Clare-Salzler, M., Cole, G., Cook, R., Coy, R., Ferguson, J., Ferguson, R., Haller, M., Hicks, E., Hosford, J., Jacobsen, L., Johnson, M., Kahler, D., Kerr, N., Kimsey, R., Lucas, A., Meehan, C., Paguio, G., Rohrs, H., Schatz, D., Smith, M., Thomas, J., Towe, P., White, D., Winter, W., Zimmerman, C., Hamalainen, J., Harkonen, T., Helander, S., Hero, M., Hirvasniemi, M., Isoaho, K., Jaminki, S., Joutsjoki, L., Kalliola, P., Kararic, M., Knip, M., Koski, K., Koski, M., Koski, M. L., Koskinen, M., Kytola, J., Laamanen, T., Latva-Koivisto, M., Laurinen, S., Mustila, T., Nyblom, M., Ollila, I., Pekkola, M., Salonen, K., Selvenius, J., Siljamaki, S., Siljander, H., Snygg, S., Suomalainen, H., Suomi, A., Tuomaala, A., Cabbage, J., Coffey, J., Hobbs, T., Johnson, K., Martin, M., Rosazza, S., Tansey, M., Tsalikian, E., Deuser, A., Foster, M., Pierce, G., Rayborn, L., Rodriguez-Luna, M., Rush, H., Wintergerst, K., Bloomfield, E., Catte, D., Dean, H., Ferens, H., Kerr, L., Kozak, B., Maharaj, R., Marks, S., Minuk, L., Rossum, K., Sneesby, K., Stierman, T., Sucharov-Benarroch, A., Taback, S., Woo, V., Yakimoski, A., Allende, G., Arazo, L., Arce, R., Baidal, D., Blaschke, C., Marks, J., Matheson, D., Pugliese, A., Sanders-Branca, N., Snowhite, I., Burant, C., Chen, M., Haddad, A., Herman, W., Hooks, H., Martin, C., Menon, R., Pietropaolo, M., Pietropaolo, S., Plunkett, C., Pop-Busui, R., Soleimanpour, A., Surhigh, J., Thomas, I., Wood, M., Bartyzal, A., Christianson, T., Flaherty, N., Forlenza, G., Gibson, C., Halper, A., Halvorsen, T., Hamdoun, E., Helms, H., Kwong, C., Lee, C., Leschyshyn, J., Luke, D., McVean, J., Moran, T., Nathan, B., Nelson, B., Omann, T., Pappenfus, B., Parchem, B., Storo, K., Street, A., Sunni, M., Tafuri, M., Vang, N., Weingartner, D., Becker, D., DeLallo, K., Diaz, A., Elnyczky, B., Groscost, D., Baldauff, N., Hoffmann, P., Ismail, H., Klein, M., Lamm, V., Libman, I., McDowell, K., Minshall, V., Pasek, B., Riley, K., Shelleby, C., Sigmund, L., Smith, M., Tas, E., Trucco, M., Yates, C., Artman, H., Johnson, B., Jospe, N., Miller, A., Orlowski, C., Jackson, M., Johnson, B., Knight, L., Szadek, L., Thompson, B., Welnick, G., Al-Zubeidi, H., Bansal, S., Bissler, M., Carroll, L., Cockroft, J., Dourisseau, D., Ferry, R., Foster, C., Johnson, T., Kassim, N., Lee, K., Logan, B., Mazhar, G., McCommon, D., Moisan, A., Parish, M., Sands, C., Sinha, S., Smith, L., Thomas, A., Thompson, L., Trzil, J., Wilson, N., Green, L., Harden, T., Kreymer, R., Mohan, A., Pruneda, M., Raskin, P., Richard, J., Schnurr-Breen, L., Smith, O., Sturges, D., Torres, N., Ziemian, L., Allred, M., Baker, S., Calder, T., Dansie, P., Donaldson, D., Foster, C., Garcia, E., Jarrett, K., Langvardt, J., Lener, M., Lusted, K., Murray, M., Reynolds, L., Slater, H., Thompson, D., Underlin, K., Vickers, L., Wheeler, K., Bere, L., Clarson, C., Gallego, P., Lovell, M., Mahon, J., McCallum, J., Stein, R., Babington, B., Barnes, K., Black, M., Bremer, A., Brendle, F., Brown, A., Dixon, B., Frazier, E., Gregg, A., Moore, D., Mountz, G., Olayinka, K., Pittel, E., Robertson, A., Russell, W., Shah, K., Shannon, A., Thomas, J., Yoder, S., Anderson, T., Bailey, D., Basnet, D., Branch, M., Bruce, G., Francis, G., Hagan, S., Henderson, G., Khandan-Barani, M., King, T., Le, T., Lemmons, J., Miller, M., Nesgoda, L., Penn, M., Schmid, J., Shankar, R., Usry, M., Wickham, E., Banks, W., Brown, H., Constantino, M., Hutson, J., Kellum, G., Lagarde, W., Lewis, M., Lockemer, H., McLaughlin, T., Piszczak, M., Reif, S., Vanderploeg, T., Andaloro, E., Breen, C., Colman, P., Dalgleish, N., Fourlanos, S., Gellert, S., Harrison, L., Healy, F., Hong, E., Hsieh, C., Mesfin, S., Mohammed, E., Redl, L., Watson, K., Wentworth, J., Cresswell, P., Faherty, H., Gould, A., Healy, F., Krebs, J., Maister, C., Ross, C., Wiltshire, E., Beresford, S., Campbell, S., Cortis, L., Couper, J., Cranwell, A., Fairchild, J., Healy, F., Richichi, K., Abdelghany, O., Feldman, L., Forbes, N., Herold, K., Huang, Y., Kunze, K., Rink, L., Sherr, J., Sherwin, R., Tamborlane, W., Weinzimer, S., Wurtz, A., Yama, N., Young, L., Writing Comm Type Diabet 2017; 318 (19): 1891–1902

    Abstract

    Type 1 diabetes requires major lifestyle changes and carries increased morbidity and mortality. Prevention or delay of diabetes would have major clinical effect.To determine whether oral insulin delays onset of type 1 diabetes in autoantibody-positive relatives of patients with type 1 diabetes.Between March 2, 2007, and December 21, 2015, relatives with at least 2 autoantibodies, including insulin autoantibodies and normal glucose tolerance, were enrolled in Canada, the United States, Australia, New Zealand, the United Kingdom, Italy, Sweden, Finland, and Germany. The main study group (n = 389) had first-phase insulin release on an intravenous glucose tolerance test that was higher than the threshold. The 55 patients in the secondary stratum 1 had an identical antibody profile as the main study group except they had first-phase insulin release that was lower than the threshold. Secondary strata 2 (n = 114) and strata 3 (n = 3) had different autoantibody profiles and first-phase insulin release threshold combinations. Follow-up continued through December 31, 2016.Randomization to receive 7.5 mg/d of oral insulin (n = 283) or placebo (n = 277), including participants in the main study group who received oral insulin (n = 203) or placebo (n = 186).The primary outcome was time to diabetes in the main study group. Significance was based on a 1-sided threshold of .05, and 1-sided 95% CIs are reported.Of a total of 560 randomized participants (median enrollment age, 8.2 years; interquartile range [IQR], 5.7-12.1 years; 170 boys [60%]; 90.7% white non-Hispanic; 57.6% with a sibling with type 1 diabetes), 550 completed the trial including 389 participants (median age, 8.4 years; 245 boys [63%]), 382 (96%) in the main study group. During a median follow-up of 2.7 years (IQR, 1.5-4.6 years) in the main study group, diabetes was diagnosed in 58 participants (28.5%) in the oral insulin group and 62 (33%) in the placebo group. Time to diabetes was not significantly different between the 2 groups (hazard ratio [HR], 0.87; 95% CI, 0-1.2; P = .21). In secondary stratum 1 (n = 55), diabetes was diagnosed in 13 participants (48.1%) in the oral insulin group and in 19 participants (70.3%) in the placebo group. The time to diabetes was significantly longer with oral insulin (HR, 0.45; 95% CI, 0-0.82; P = .006). The HR for time to diabetes for the between-group comparisons for the 116 participants in the other secondary stratum was 1.03 (95% CI, 0-2.11; P = .53) and for the entire cohort of 560 participants was 0.83 (95% CI, 0-1.07; P = .11), which were not significantly different. The most common adverse event was infection (n = 254), with 134 events in the oral insulin group and 120 events in the placebo group, but no significant study-related adverse events occurred.Among autoantibody-positive relatives of patients with type 1 diabetes, oral insulin at a dose of 7.5 mg/d, compared with placebo, did not delay or prevent the development of type 1 diabetes over 2.7 years. These findings do not support oral insulin as used in this study for diabetes prevention.clinicaltrials.gov Identifier: NCT00419562.

    View details for DOI 10.1001/jama.2017.17070

    View details for Web of Science ID 000415870300019

    View details for PubMedID 29164254

    View details for PubMedCentralID PMC5798455

  • Impact of blood collection and processing on peripheral blood gene expression profiling in type 1 diabetes BMC GENOMICS Yip, L., Fuhlbrigge, R., Atkinson, M. A., Fathman, C. 2017; 18: 636

    Abstract

    The natural history of type 1 diabetes (T1D) is challenging to investigate, especially as pre-diabetic individuals are difficult to identify. Numerous T1D consortia have been established to collect whole blood for gene expression analysis from individuals with or at risk to develop T1D. However, with no universally accepted protocol for their collection, differences in sample processing may lead to variances in the results. Here, we examined whether the choice of blood collection tube and RNA extraction kit leads to differences in the expression of genes that are changed during the progression of T1D, and if these differences could be minimized by measuring gene expression directly from the lysate of whole blood.Microarray analysis showed that the expression of 901 genes is highly influenced by sample processing using the PAXgene versus the Tempus system. These included a significant number of lymphocyte-specific genes and genes whose expression has been reported to differ in the peripheral blood of at-risk and T1D patients compared to controls. We showed that artificial changes in gene expression occur when control and T1D samples were processed differently. The sample processing-dependent differences in gene expression were largely due to loss of transcripts during the RNA extraction step using the PAXgene system. The majority of differences were not observed when gene expression was measured in whole blood lysates prepared from blood collected in PAXgene and Tempus tubes.We showed that the gene expression profile of samples processed using the Tempus system is more accurate than that of samples processed using the PAXgene system. Variation in sample processing can result in misleading changes in gene expression. However, these differences can be minimized by measuring gene expression directly in whole blood lysates.

    View details for PubMedID 28821222

  • Selective expansion of human regulatory T cells in nasal polyps, and not adjacent tissue microenvironments, in individual patients exposed to steroids. Clinical immunology Edward, J. A., Sanyal, M., Le, W., Soudry, E., Ramakrishnan, V. R., Bravo, D. T., Nguyen, A. L., Zarabanda, D., Kingdom, T. T., Hwang, P. H., Garrison Fathman, C., Nayak, J. V. 2017; 179: 66-76

    Abstract

    Severe forms of chronic rhinosinusitis (CRS), a common upper airway inflammatory disorder, are associated with nasal polyps (NPs). NP disease is ameliorated by glucocorticoid (GC) treatment, whose cellular effects are poorly understood. We therefore assessed the influence of GC therapy on NPs in CRS patients, focusing on regulatory T (Treg) cells. Treg cell populations were analyzed by flow cytometry in NPs and control tissues from GC-treated CRS patients and controls. After GC exposure, selective expansion of Treg cells was seen within NPs, and not blood or adjacent ethmoid tissues. To confirm direct GC effects, NPs from the same patients were biopsied prior to, and following, 1week of oral GC exposure. Direct expansion of Tregs into the same NP bed was detected in 4/4 CRS patients following GC exposure. Treg cell spikes into NPs were secondary to cellular recruitment given limited Ki67 expression within these regulatory cells. Chemokine gene expression profiling identified several chemokines, notably CCL4, induced within NPs upon GC treatment. Neutralization of chemokine receptor/ligand interactions using CCR4 small molecule antagonists reduced Treg migration towards GC-treated NPs in an ex vivo migration assay. Our findings suggest that the common use of GCs in the treatment of NP disease leads to recruitment of Treg cells from peripheral sites into NP tissues, which may be critical to the anti-inflammatory effect of GCs. Mechanistically Treg expansion appears to be conferred, in part, by chemokine receptor/ligand interactions induced following corticosteroid therapy.

    View details for DOI 10.1016/j.clim.2017.02.002

    View details for PubMedID 28279811

  • Autoantibody-Positive Healthy Individuals Display Unique Immune Profiles That May Regulate Autoimmunity. Arthritis & rheumatology Slight-Webb, S., Lu, R., Ritterhouse, L. L., Munroe, M. E., Maecker, H. T., Fathman, C. G., Utz, P. J., Merrill, J. T., Guthridge, J. M., James, J. A. 2016; 68 (10): 2492-2502

    Abstract

    Antinuclear antibodies (ANAs) are detected in ∼18% of females, yet autoimmune disease develops in only 5-8%. Immunologic differences between ANA-positive healthy individuals and patients with systemic lupus erythematosus (SLE) may elucidate the regulatory mechanisms by which ANA-positive individuals avoid transition to clinical autoimmune disease.Healthy individuals (n = 790) were screened for autoantibodies specific for 11 antigens associated with lupus, systemic sclerosis, and Sjögren's syndrome. From this screening, 31 European American ANA-positive healthy individuals were selected and demographically matched to ANA-negative controls and SLE patients. Serum cytokine profiles, leukocyte subset frequency, and reactivity were analyzed by multiplex assays, immunophenotyping, and phosphospecific flow cytometry.Of 790 individuals screened, 57 (7%) were ANA-positive. The majority of proinflammatory cytokines, including interferon-γ (IFNγ), tumor necrosis factor, interleukin-17 (IL-17), and granulocyte colony-stimulating factor, exhibited a stepwise increase in serum levels from ANA-negative controls to ANA-positive healthy individuals to SLE patients (P < 0.0001). IFNα, IFNβ, IL-12p40, and stem cell factor/c-Kit ligand were increased in SLE patients only (P < 0.05). B lymphocyte stimulator (BlyS) was elevated in SLE patients but decreased in ANA-positive individuals (P < 0.001). Further, IL-1 receptor antagonist (IL-1Ra) was down-regulated in SLE patients only (P < 0.0001). ANA-positive individuals had increased frequencies of monocytes, memory B cells, and plasmablasts and increased levels of pSTAT-1 and pSTAT-3 following IFNα stimulation compared with ANA-negative controls (P < 0.05).ANA-positive healthy individuals exhibit dysregulation in multiple immune pathways yet differ from SLE patients by the absence of elevated IFNs, BLyS, IL-12p40, and stem cell factor/c-Kit ligand. Further, severely decreased levels of IL-1Ra in SLE patients compared with ANA-positive individuals may contribute to disease development. These results highlight the importance of IFN-related pathways and regulatory elements in SLE pathogenesis.

    View details for DOI 10.1002/art.39706

    View details for PubMedID 27059145

    View details for PubMedCentralID PMC5042816

  • Expression-Based Genome-Wide Association Study Links Vitamin D-Binding Protein With Autoantigenicity in Type 1 Diabetes DIABETES Kodama, K., Zhao, Z., Toda, k., Yip, L., Fuhlbrigge, R., Miao, D., Fathman, C. G., Yamada, S., Butte, A. J., Yu, L. 2016; 65 (5): 1341-1349

    Abstract

    Type 1 diabetes (T1D) is caused by autoreactive T cells that recognize pancreatic islet antigens and destroy insulin-producing β-cells. This attack results from a breakdown in tolerance for self-antigens, which is controlled by ectopic antigen expression in the thymus and pancreatic lymph nodes (PLNs). The autoantigens known to be involved include a set of islet proteins, such as insulin, GAD65, IA-2, and ZnT8. In an attempt to identify additional antigenic proteins, we performed an expression-based genome-wide association study using microarray data from 118 arrays of the thymus and PLNs of T1D mice. We ranked all 16,089 protein-coding genes by the likelihood of finding repeated differential expression and the degree of tissue specificity for pancreatic islets. The top autoantigen candidate was vitamin D-binding protein (VDBP). T-cell proliferation assays showed stronger T-cell reactivity to VDBP compared with control stimulations. Higher levels and frequencies of serum anti-VDBP autoantibodies (VDBP-Abs) were identified in patients with T1D (n = 331) than in healthy control subjects (n = 77). Serum vitamin D levels were negatively correlated with VDBP-Ab levels in patients in whom T1D developed during the winter. Immunohistochemical localization revealed that VDBP was specifically expressed in α-cells of pancreatic islets. We propose that VDBP could be an autoantigen in T1D.

    View details for DOI 10.2337/db15-1308

    View details for Web of Science ID 000375028000023

    View details for PubMedID 26983959

    View details for PubMedCentralID PMC4839207

  • Concise Review: Cell-Based Therapies and Other Non-Traditional Approaches for Type 1 Diabetes STEM CELLS Creusot, R. J., Battaglia, M., Roncarolo, M., Fathman, C. G. 2016; 34 (4): 809-819

    Abstract

    The evolution of Type 1 diabetes (T1D) therapy has been marked by consecutive shifts, from insulin replacement to immunosuppressive drugs and targeted biologics (following the understanding that T1D is an autoimmune disease), and to more disease-specific or patient-oriented approaches such as antigen-specific and cell-based therapies, with a goal to provide efficacy, safety, and long-term protection. At the same time, another important paradigm shift from treatment of new onset T1D patients to prevention in high-risk individuals has taken place, based on the hypothesis that therapeutic approaches deemed sufficiently safe may show better efficacy if applied early enough to maintain endogenous β cell function, a concept supported by many preclinical studies. This new strategy has been made possible by capitalizing on a variety of biomarkers that can more reliably estimate the risk and rate of progression of the disease. More advanced ("omic"-based) biomarkers that also shed light on the underlying contributors of disease for each individual will be helpful to guide the choice of the most appropriate therapies, or combinations thereof. In this review, we present current efforts to stratify patients according to biomarkers and current alternatives to conventional drug-based therapies for T1D, with a special emphasis on cell-based therapies, their status in the clinic and potential for treatment and/or prevention.

    View details for DOI 10.1002/stem.2290

    View details for Web of Science ID 000374697700002

    View details for PubMedID 26840009

  • Amyloid fibrils activate B-1a lymphocytes to ameliorate inflammatory brain disease PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Kurnellas, M. P., Ghosn, E. E., Schartner, J. M., Baker, J., Rothbard, J. J., Negrin, R. S., Herzenberg, L. A., Fathman, C. G., Steinman, L., Rothbard, J. B. 2015; 112 (49): 15016-15023

    Abstract

    Amyloid fibrils composed of peptides as short as six amino acids are therapeutic in experimental autoimmune encephalomyelitis (EAE), reducing paralysis and inflammation, while inducing several pathways of immune suppression. Intraperitoneal injection of fibrils selectively activates B-1a lymphocytes and two populations of resident macrophages (MΦs), increasing IL-10 production, and triggering their exodus from the peritoneum. The importance of IL-10-producing B-1a cells in this effective therapy was established in loss-of-function experiments where neither B-cell-deficient (μMT) nor IL10(-/-) mice with EAE responded to the fibrils. In gain-of-function experiments, B-1a cells, adoptively transferred to μMT mice with EAE, restored their therapeutic efficacy when Amylin 28-33 was administered. Stimulation of adoptively transferred bioluminescent MΦs and B-1a cells by amyloid fibrils resulted in rapid (within 60 min of injection) trafficking of both cell types to draining lymph nodes. Analysis of gene expression indicated that the fibrils activated the CD40/B-cell receptor pathway in B-1a cells and induced a set of immune-suppressive cell-surface proteins, including BTLA, IRF4, and Siglec G. Collectively, these data indicate that the fibrils activate B-1a cells and F4/80(+) MΦs, resulting in their migration to the lymph nodes, where IL-10 and cell-surface receptors associated with immune-suppression limit antigen presentation and T-cell activation. These mechanisms culminate in reduction of paralytic signs of EAE.

    View details for DOI 10.1073/pnas.1521206112

    View details for PubMedID 26621719

  • 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

  • Virtual Time-Course Analysis of Genome-Wide Gene-Expression Profile in Pancreatic Lymph Nodes from Human Prediabetic Subjects Kodama, K., Yip, L., Fuhlbrigge, R., Butte, A. J., Fathman, C. AMER DIABETES ASSOC. 2015: A61
  • Mass cytometry as a platform for the discovery of cellular biomarkers to guide effective rheumatic disease therapy ARTHRITIS RESEARCH & THERAPY Nair, N., Mei, H. E., Chen, S., Hale, M., Nolan, G. P., Maecker, H. T., Genovese, M., Fathman, C. G., Whiting, C. C. 2015; 17

    Abstract

    The development of biomarkers for autoimmune diseases has been hampered by a lack of understanding of disease etiopathogenesis and of the mechanisms underlying the induction and maintenance of inflammation, which involves complex activation dynamics of diverse cell types. The heterogeneous nature and suboptimal clinical response to treatment observed in many autoimmune syndromes highlight the need to develop improved strategies to predict patient outcome to therapy and personalize patient care. Mass cytometry, using CyTOF®, is an advanced technology that facilitates multiparametric, phenotypic analysis of immune cells at single-cell resolution. In this review, we outline the capabilities of mass cytometry and illustrate the potential of this technology to enhance the discovery of cellular biomarkers for rheumatoid arthritis, a prototypical autoimmune disease.

    View details for DOI 10.1186/s13075-015-0644-z

    View details for Web of Science ID 000354850500001

    View details for PubMedID 25981462

    View details for PubMedCentralID PMC4436107

  • Autoantibody-positive healthy individuals display unique immune profiles that regulate autoimmunity Slight-Webb, S., Lu, R., Ritterhouse, L., Maecker, H., Fathman, C., Merrill, J., Guthridge, J., James, J. AMER ASSOC IMMUNOLOGISTS. 2015
  • A Novel Transcription Factor, T-bet, Directs Th1 Lineage Commitment JOURNAL OF IMMUNOLOGY Szabo, S. J., Kim, S. T., Costa, G. L., Zhang, X., Fathman, C. G., Glimcher, L. H. 2015; 194 (7): 2961-2975
  • Inflammation and Hyperglycemia Mediate Deaf1 Splicing in the Pancreatic Lymph Nodes via Distinct Pathways During Type 1 Diabetes. Diabetes Yip, L., Fuhlbrigge, R., Taylor, C., Creusot, R. J., Nishikawa-Matsumura, T., Whiting, C. C., Schartner, J. M., Akter, R., von Herrath, M., Fathman, C. G. 2015; 64 (2): 604-617

    Abstract

    Peripheral tolerance is partially controlled by the expression of peripheral tissue antigens (PTAs) in lymph node stromal cells (LNSCs). We previously identified a transcriptional regulator, deformed epidermal autoregulatory factor 1 (Deaf1), that can regulate PTA expression in LNSCs of the pancreatic lymph nodes (PLNs). During the pathogenesis of type 1 diabetes (T1D), Deaf1 is spliced to form the dominant-negative isoform Deaf1-Var1. Here we show that Deaf1-Var1 expression correlates with the severity of disease in NOD mice and is reduced in the PLNs of mice that do not develop hyperglycemia. Inflammation and hyperglycemia independently drive Deaf1 splicing through activation of the splicing factors Srsf10 and Ptbp2, respectively. Inflammation induced by injection of activated splenocytes increased Deaf1-Var1 and Srsf10, but not Ptbp2, in the PLNs of NOD.SCID mice. Hyperglycemia induced by treatment with the insulin receptor agonist S961 increased Deaf1-Var1 and Ptbp2, but not Srsf10, in the PLNs of NOD.B10 and NOD mice. Overexpression of PTBP2 and/or SRSF10 also increased human DEAF1-VAR1 and reduced PTA expression in HEK293T cells. These data suggest that during the progression of T1D, inflammation and hyperglycemia mediate the splicing of DEAF1 and loss of PTA expression in LNSCs by regulating the expression of SRSF10 and PTBP2.

    View details for DOI 10.2337/db14-0803

    View details for PubMedID 25187368

    View details for PubMedCentralID PMC4303971

  • 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

  • Mass cytometry as a platform for the discovery of cellular biomarkers to guide effective rheumatic disease therapy. Arthritis research & therapy Nair, N., Mei, H. E., Chen, S., Hale, M., Nolan, G. P., Maecker, H. T., Genovese, M., Fathman, C. G., Whiting, C. C. 2015; 17: 127-?

    Abstract

    The development of biomarkers for autoimmune diseases has been hampered by a lack of understanding of disease etiopathogenesis and of the mechanisms underlying the induction and maintenance of inflammation, which involves complex activation dynamics of diverse cell types. The heterogeneous nature and suboptimal clinical response to treatment observed in many autoimmune syndromes highlight the need to develop improved strategies to predict patient outcome to therapy and personalize patient care. Mass cytometry, using CyTOF®, is an advanced technology that facilitates multiparametric, phenotypic analysis of immune cells at single-cell resolution. In this review, we outline the capabilities of mass cytometry and illustrate the potential of this technology to enhance the discovery of cellular biomarkers for rheumatoid arthritis, a prototypical autoimmune disease.

    View details for DOI 10.1186/s13075-015-0644-z

    View details for PubMedID 25981462

  • Mechanisms of action of therapeutic amyloidogenic hexapeptides in amelioration of inflammatory brain disease. journal of experimental medicine Kurnellas, M. P., Schartner, J. M., Fathman, C. G., Jagger, A., Steinman, L., Rothbard, J. B. 2014; 211 (9): 1847-1856

    Abstract

    Amyloid fibrils composed of peptides as short as six amino acids are effective therapeutics for experimental autoimmune encephalomyelitis (EAE). Immunosuppression arises from at least two pathways: (1) expression of type 1 IFN by pDCs, which were induced by neutrophil extracellular traps arising from the endocytosis of the fibrils; and (2) the reduced expression of IFN-γ, TNF, and IL-6. The two independent pathways stimulated by the fibrils can act in concert to be immunosuppressive in Th1 indications, or in opposition, resulting in inflammation when Th17 T lymphocytes are predominant. The generation of type 1 IFN can be minimized by using polar, nonionizable, amyloidogenic peptides, which are effective in both Th1 and Th17 polarized EAE.

    View details for DOI 10.1084/jem.20140107

    View details for PubMedID 25073790

    View details for PubMedCentralID PMC4144739

  • Poly-l-Arginine Topical Lotion Tested in a Mouse Model for Frostbite Injury. Wilderness & environmental medicine Auerbach, L. J., DeClerk, B. K., Garrison Fathman, C., Gurtner, G. C., Auerbach, P. S. 2014; 25 (2): 160-165

    Abstract

    Frostbite injury occurs when exposure to cold results in frozen tissue. We recently reported a novel mouse model for frostbite injury to be used in screening potentially therapeutic drugs and other modalities.We used the mouse skin frostbite model to evaluate the effect of poly-l-arginine contained in lotion (PAL) applied topically to involved skin.Sixty mice were studied in a randomized, double-blind method. Standardized 2.9-cm-diameter circles were tattooed on the mouse dorsum. Magnets snap frozen in dry ice (-78.5°C) were used to create a frostbite injury on skin within the circle as a continuous 5-minute freeze. Mice were treated with prefreeze placebo, postthaw placebo, combined prefreeze and postthaw placebo, prefreeze with PAL, postthaw with PAL, or combined prefreeze and postthaw with PAL. Appearance, healing rate, tissue loss, and histology were recorded until the wounds were healed.Application of PAL before inducing frostbite injury resulted in decreased tissue loss as compared with other treatment conditions.Applying PAL topically to frostbitten mouse skin caused decreased tissue loss. Poly-l-arginine should be studied further to determine whether it is a beneficial therapeutic modality for frostbite injury.

    View details for DOI 10.1016/j.wem.2014.01.006

    View details for PubMedID 24631228

  • Type 1 diabetes in mice and men: gene expression profiling to investigate disease pathogenesis. Immunologic research Yip, L., Fathman, C. G. 2014; 58 (2-3): 340-350

    Abstract

    Type 1 diabetes (T1D) is a complex polygenic disease that is triggered by various environmental factors in genetically susceptible individuals. The emphasis placed on genome-wide association studies to explain the genetics of T1D has failed to advance our understanding of T1D pathogenesis or identify biomarkers of disease progression or therapeutic targets. Using the nonobese diabetic (NOD) mouse model of T1D and the non-disease prone congenic NOD.B10 mice, our laboratory demonstrated striking tissue-specific and age-dependent changes in gene expression during disease progression. We established a "roadmap" of differential gene expression and used this to identify candidate genes in mice (and human orthologs) that play a role in disease pathology. Here, we describe two genes, Deformed epidermal autoregulatory factor 1 (Deaf1) and Adenosine A1 receptor (Adora1), that are differentially expressed and alternatively spliced in the pancreatic lymph nodes or islets of NOD mice and T1D patients to form dominant-negative non-functional isoforms. Loss of Deaf1 function leads to reduced peripheral tissue antigen expression in lymph node stromal cells and may contribute to a breakdown in peripheral tolerance, while reduced Adora1 function results in an early intrinsic alpha cell defect that may explain the hyperglucagonemia and resulting beta cell stress observed prior to the onset of diabetes. Remarkably, both genes were also alternatively spliced in the same tissues of auto-antibody positive prediabetic patients, and these splicing events resulted in similar downstream effects as those seen in NOD mice. These findings demonstrate the value of gene expression profiling in studying disease pathogenesis in T1D.

    View details for DOI 10.1007/s12026-014-8501-8

    View details for PubMedID 24682832

  • Vitamin D Deficiency in a Multiethnic Healthy Control Cohort and Altered Immune Response in Vitamin D Deficient European-American Healthy Controls PLOS ONE Ritterhouse, L. L., Lu, R., Shah, H. B., Robertson, J. M., Fife, D. A., Maecker, H. T., Du, H., Fathman, C. G., Chakravarty, E. F., Scofield, R. H., Kamen, D. L., Guthridge, J. M., James, J. A. 2014; 9 (4)

    Abstract

    In recent years, vitamin D has been shown to possess a wide range of immunomodulatory effects. Although there is extensive amount of research on vitamin D, we lack a comprehensive understanding of the prevalence of vitamin D deficiency or the mechanism by which vitamin D regulates the human immune system. This study examined the prevalence and correlates of vitamin D deficiency and the relationship between vitamin D and the immune system in healthy individuals.Healthy individuals (n = 774) comprised of European-Americans (EA, n = 470), African-Americans (AA, n = 125), and Native Americans (NA, n = 179) were screened for 25-hydroxyvitamin D [25(OH)D] levels by ELISA. To identify the most noticeable effects of vitamin D on the immune system, 20 EA individuals with severely deficient (<11.3 ng/mL) and sufficient (>24.8 ng/mL) vitamin D levels were matched and selected for further analysis. Serum cytokine level measurement, immune cell phenotyping, and phosphoflow cytometry were performed.Vitamin D sufficiency was observed in 37.5% of the study cohort. By multivariate analysis, AA, NA, and females with a high body mass index (BMI, >30) demonstrate higher rates of vitamin D deficiency (p<0.05). Individuals with vitamin D deficiency had significantly higher levels of serum GM-CSF (p = 0.04), decreased circulating activated CD4+ (p = 0.04) and CD8+ T (p = 0.04) cell frequencies than individuals with sufficient vitamin D levels.A large portion of healthy individuals have vitamin D deficiency. These individuals have altered T and B cell responses, indicating that the absence of sufficient vitamin D levels could result in undesirable cellular and molecular alterations ultimately contributing to immune dysregulation.

    View details for DOI 10.1371/journal.pone.0094500

    View details for Web of Science ID 000336736200083

    View details for PubMedID 24727903

    View details for PubMedCentralID PMC3984168

  • Vitamin d deficiency in a multiethnic healthy control cohort and altered immune response in vitamin D deficient European-American healthy controls. PloS one Ritterhouse, L. L., Lu, R., Shah, H. B., Robertson, J. M., Fife, D. A., Maecker, H. T., Du, H., Fathman, C. G., Chakravarty, E. F., Scofield, R. H., Kamen, D. L., Guthridge, J. M., James, J. A. 2014; 9 (4)

    Abstract

    In recent years, vitamin D has been shown to possess a wide range of immunomodulatory effects. Although there is extensive amount of research on vitamin D, we lack a comprehensive understanding of the prevalence of vitamin D deficiency or the mechanism by which vitamin D regulates the human immune system. This study examined the prevalence and correlates of vitamin D deficiency and the relationship between vitamin D and the immune system in healthy individuals.Healthy individuals (n = 774) comprised of European-Americans (EA, n = 470), African-Americans (AA, n = 125), and Native Americans (NA, n = 179) were screened for 25-hydroxyvitamin D [25(OH)D] levels by ELISA. To identify the most noticeable effects of vitamin D on the immune system, 20 EA individuals with severely deficient (<11.3 ng/mL) and sufficient (>24.8 ng/mL) vitamin D levels were matched and selected for further analysis. Serum cytokine level measurement, immune cell phenotyping, and phosphoflow cytometry were performed.Vitamin D sufficiency was observed in 37.5% of the study cohort. By multivariate analysis, AA, NA, and females with a high body mass index (BMI, >30) demonstrate higher rates of vitamin D deficiency (p<0.05). Individuals with vitamin D deficiency had significantly higher levels of serum GM-CSF (p = 0.04), decreased circulating activated CD4+ (p = 0.04) and CD8+ T (p = 0.04) cell frequencies than individuals with sufficient vitamin D levels.A large portion of healthy individuals have vitamin D deficiency. These individuals have altered T and B cell responses, indicating that the absence of sufficient vitamin D levels could result in undesirable cellular and molecular alterations ultimately contributing to immune dysregulation.

    View details for DOI 10.1371/journal.pone.0094500

    View details for PubMedID 24727903

    View details for PubMedCentralID PMC3984168

  • It's Time to Bring Dendritic Cell Therapy to Type 1 Diabetes DIABETES Creusot, R. J., Giannoukakis, N., Trucco, M., Clare-Salzler, M. J., Fathman, C. 2014; 63 (1): 20–30

    View details for DOI 10.2337/db13-0886

    View details for Web of Science ID 000328680400003

    View details for PubMedID 24357690

    View details for PubMedCentralID PMC3968436

  • Diminished Adenosine A1 Receptor Expression in Pancreatic a-Cells May Contribute to the Pathology of Type 1 Diabetes. Diabetes Yip, L., Taylor, C., Whiting, C. C., Fathman, C. G. 2013; 62 (12): 4208-4219

    Abstract

    Prediabetic NOD mice exhibit hyperglucagonemia, possibly due to an intrinsic α-cell defect. Here, we show that the expression of a potential glucagon inhibitor, the adenosine A1 receptor (Adora1), is gradually diminished in α-cells of NOD mice, autoantibody-positive (AA(+)) and overtly type 1 diabetic (T1D) patients during the progression of disease. We demonstrated that islet inflammation was associated with loss of Adora1 expression through the alternative splicing of Adora1. Expression of the spliced variant (Adora1-Var) was upregulated in the pancreas of 12-week-old NOD versus age-matched NOD.B10 (non-diabetes-susceptible) control mice and was detected in the pancreas of AA(+) patients but not in control subjects or overtly diabetic patients, suggesting that inflammation drives the splicing of Adora1. We subsequently demonstrated that Adora1-Var expression was upregulated in the islets of NOD.B10 mice after exposure to inflammatory cytokines and in the pancreas of NOD.SCID mice after adoptive transfer of activated autologous splenocytes. Adora1-Var encodes a dominant-negative N-terminal truncated isoform of Adora1. The splicing of Adora1 and loss of Adora1 expression on α-cells may explain the hyperglucagonemia observed in prediabetic NOD mice and may contribute to the pathogenesis of human T1D and NOD disease.

    View details for DOI 10.2337/db13-0614

    View details for PubMedID 24264405

    View details for PubMedCentralID PMC3837064

  • Effectiveness of Early Intensive Therapy on beta-Cell Preservation in Type 1 Diabetes DIABETES CARE Buckingham, B., Beck, R. W., Ruedy, K. J., Cheng, P., Kollman, C., Weinzimer, S. A., Dimeglio, L. A., Bremer, A. A., Slover, R., Tamborlane, W. V. 2013; 36 (12): 4030-4035

    Abstract

    To assess effectiveness of inpatient hybrid closed-loop control (HCLC) followed by outpatient sensor-augmented pump (SAP) therapy initiated within 7 days of diagnosis of type 1 diabetes on the preservation of β-cell function at 1 year.Sixty-eight individuals (mean age 13.3 ± 5.7 years; 35% female, 92% Caucasian) were randomized to HCLC followed by SAP therapy (intensive group; N = 48) or to the usual-care group treated with multiple daily injections or insulin pump therapy (N = 20). Primary outcome was C-peptide concentrations during mixed-meal tolerance tests at 12 months.Intensive-group participants initiated HCLC a median of 6 days after diagnosis for a median duration of 71.3 h, during which median participant mean glucose concentration was 140 mg/dL (interquartile range 134-153 mg/dL). During outpatient SAP, continuous glucose monitor (CGM) use decreased over time, and at 12 months, only 33% of intensive participants averaged sensor use ≥6 days/week. In the usual-care group, insulin pump and CGM use were initiated prior to 12 months by 15 and 5 participants, respectively. Mean HbA1c levels were similar in both groups throughout the study. At 12 months, the geometric mean (95% CI) of C-peptide area under the curve was 0.43 (0.34-0.52) pmol/mL in the intensive group and 0.52 (0.32-0.75) pmol/mL in the usual-care group (P = 0.49). Thirty-seven (79%) intensive and 16 (80%) usual-care participants had a peak C-peptide concentration ≥0.2 pmol/mL (P = 0.30).In new-onset type 1 diabetes, HCLC followed by SAP therapy did not provide benefit in preserving β-cell function compared with current standards of care.

    View details for DOI 10.2337/dc13-1074

    View details for Web of Science ID 000327211500053

    View details for PubMedID 24130350

  • Interleukin-1 antagonism in type 1 diabetes of recent onset: two multicentre, randomised, double-blind, placebo-controlled trials LANCET Moran, A., Bundy, B., Becker, D. J., Dimeglio, L. A., Gitelman, S. E., Goland, R., Greenbaum, C. J., Herold, K. C., Marks, J. B., Raskin, P., Sanda, S., Schatz, D., Wherrett, D. K., Wilson, D. M., Krischer, J. P., Skyler, J. S., Pickersgill, L., de Koning, E., Ziegler, A., Boeehm, B., Badenhoop, K., Schloot, N., Bak, J. F., Pozzilli, P., Mauricio, D., Donath, M. Y., Castano, L., Waegner, A., Lervang, H. H., Perrild, H., Mandrup-Poulsen, T. 2013; 381 (9881): 1905-1915

    Abstract

    Innate immunity contributes to the pathogenesis of autoimmune diseases, such as type 1 diabetes, but until now no randomised, controlled trials of blockade of the key innate immune mediator interleukin-1 have been done. We aimed to assess whether canakinumab, a human monoclonal anti-interleukin-1 antibody, or anakinra, a human interleukin-1 receptor antagonist, improved β-cell function in recent-onset type 1 diabetes.We did two randomised, placebo-controlled trials in two groups of patients with recent-onset type 1 diabetes and mixed-meal-tolerance-test-stimulated C peptide of at least 0·2 nM. Patients in the canakinumab trial were aged 6-45 years and those in the anakinra trial were aged 18-35 years. Patients in the canakinumab trial were enrolled at 12 sites in the USA and Canada and those in the anakinra trial were enrolled at 14 sites across Europe. Participants were randomly assigned by computer-generated blocked randomisation to subcutaneous injection of either 2 mg/kg (maximum 300 mg) canakinumab or placebo monthly for 12 months or 100 mg anakinra or placebo daily for 9 months. Participants and carers were masked to treatment assignment. The primary endpoint was baseline-adjusted 2-h area under curve C-peptide response to the mixed meal tolerance test at 12 months (canakinumab trial) and 9 months (anakinra trial). Analyses were by intention to treat. These studies are registered with ClinicalTrials.gov, numbers NCT00947427 and NCT00711503, and EudraCT number 2007-007146-34.Patients were enrolled in the canakinumab trial between Nov 12, 2010, and April 11, 2011, and in the anakinra trial between Jan 26, 2009, and May 25, 2011. 69 patients were randomly assigned to canakinumab (n=47) or placebo (n=22) monthly for 12 months and 69 were randomly assigned to anakinra (n=35) or placebo (n=34) daily for 9 months. No interim analyses were done. 45 canakinumab-treated and 21 placebo-treated patients in the canakinumab trial and 25 anakinra-treated and 26 placebo-treated patients in the anakinra trial were included in the primary analyses. The difference in C peptide area under curve between the canakinumab and placebo groups at 12 months was 0·01 nmol/L (95% CI -0·11 to 0·14; p=0·86), and between the anakinra and the placebo groups at 9 months was 0·02 nmol/L (-0·09 to 0·15; p=0·71). The number and severity of adverse events did not differ between groups in the canakinumab trial. In the anakinra trial, patients in the anakinra group had significantly higher grades of adverse events than the placebo group (p=0·018), which was mainly because of a higher number of injection site reactions in the anakinra group.Canakinumab and anakinra were safe but were not effective as single immunomodulatory drugs in recent-onset type 1 diabetes. Interleukin-1 blockade might be more effective in combination with treatments that target adaptive immunity in organ-specific autoimmune disorders.National Institutes of Health and Juvenile Diabetes Research Foundation.

    View details for DOI 10.1016/S0140-6736(13)60023-9

    View details for Web of Science ID 000320319500031

  • The Effects of Inpatient Hybrid Closed-Loop Therapy Initiated Within 1 Week of Type 1 Diabetes Diagnosis DIABETES TECHNOLOGY & THERAPEUTICS Buckingham, B. A., Beck, R. W., Ruedy, K. J., Cheng, P., Kollman, C., Weinzimer, S. A., Dimeglio, L. A., Bremer, A. A., Slover, R., Cantwell, M., Tsalikian, E., Tansey, M. J., Coffey, J., Cabbage, J., Salamati, S., Mauras, N., Fox, L. A., Englert, K., Permuy, J., Sikes, K., Buckingham, B. A., Wilson, D. M., Clinton, P., Caswell, K., Weinzimer, S. A., Tamborlane, W. V., Sherr, J., Steffen, A., Weyman, K., Zgorski, M., Tichy, E., White, N. H., Arbelaez, A. M., Levandoski, L., Starnes, A., Skyler, J. S., Greenbaum, C. J., Kenyon, N. S., Rafkin, L., Santiago, I., Sosenko, J. M. 2013; 15 (5): 401-408
  • The gene related to anergy in lymphocytes regulates stat-mediated differentiation of CD4 T cells via ubiquitination and degradation of the kinase chaperone CDC37 (P1128) Schartner, J., Su, L., Fathman, C., Whiting, C. AMER ASSOC IMMUNOLOGISTS. 2013
  • Inflammation-induced splicing of Deaf1 in the pancreatic lymph nodes during the progression of Type 1 diabetes Yip, L., Creusot, R., Whiting, C., Taylor, C., Akter, R., Matsumura, T., Fathman, C. AMER ASSOC IMMUNOLOGISTS. 2013
  • Reduced DEAF1 function during type 1 diabetes inhibits translation in lymph node stromal cells by suppressing Eif4g3. Journal of molecular cell biology Yip, L., Creusot, R. J., Pager, C. T., Sarnow, P., Fathman, C. G. 2013; 5 (2): 99-110

    Abstract

    The transcriptional regulator deformed epidermal autoregulatory factor 1 (DEAF1) has been suggested to play a role in maintaining peripheral tolerance by controlling the transcription of peripheral tissue antigen genes in lymph node stromal cells (LNSCs). Here, we demonstrate that DEAF1 also regulates the translation of genes in LNSCs by controlling the transcription of the poorly characterized eukaryotic translation initiation factor 4 gamma 3 (Eif4g3) that encodes eIF4GII. Eif4g3 gene expression was reduced in the pancreatic lymph nodes of Deaf1-KO mice, non-obese diabetic mice, and type 1 diabetes patients, where functional Deaf1 is absent or diminished. Silencing of Deaf1 reduced Eif4g3 expression, but increased the expression of Caspase 3, a serine protease that degrades eIF4GII. Polysome profiling showed that reduced Eif4g3 expression in LNSCs resulted in the diminished translation of various genes, including Anpep, the gene for aminopeptidase N, an enzyme involved in fine-tuning antigen presentation on major histocompatibility complex (MHC) class II. Together these findings suggest that reduced DEAF1 function, and subsequent loss of Eif4g3 transcription may affect peripheral tissue antigen (PTA) expression in LNSCs and contribute to the pathology of T1D.

    View details for DOI 10.1093/jmcb/mjs052

    View details for PubMedID 22923498

    View details for PubMedCentralID PMC3604916

  • Redirecting cell-type specific cytokine responses with engineered interleukin-4 superkines NATURE CHEMICAL BIOLOGY Junttila, I. S., Creusot, R. J., Moraga, I., Bates, D. L., Wong, M. T., Alonso, M. N., Suhoski, M. M., Lupardus, P., Meier-Schellersheim, M., Engleman, E. G., Utz, P. J., Fathman, C. G., Paul, W. E., Garcia, K. C. 2012; 8 (12): 990-998

    Abstract

    Cytokines dimerize their receptors, with the binding of the 'second chain' triggering signaling. In the interleukin (IL)-4 and IL-13 system, different cell types express varying numbers of alternative second receptor chains (γc or IL-13Rα1), forming functionally distinct type I or type II complexes. We manipulated the affinity and specificity of second chain recruitment by human IL-4. A type I receptor-selective IL-4 'superkine' with 3,700-fold higher affinity for γc was three- to ten-fold more potent than wild-type IL-4. Conversely, a variant with high affinity for IL-13Rα1 more potently activated cells expressing the type II receptor and induced differentiation of dendritic cells from monocytes, implicating the type II receptor in this process. Superkines showed signaling advantages on cells with lower second chain numbers. Comparative transcriptional analysis reveals that the superkines induce largely redundant gene expression profiles. Variable second chain numbers can be exploited to redirect cytokines toward distinct cell subsets and elicit new actions, potentially improving the selectivity of cytokine therapy.

    View details for DOI 10.1038/NCHEMBIO.1096

    View details for Web of Science ID 000311491200014

    View details for PubMedID 23103943

    View details for PubMedCentralID PMC3508151

  • New tools for classification and monitoring of autoimmune diseases NATURE REVIEWS RHEUMATOLOGY Maecker, H. T., Lindstrom, T. M., Robinson, W. H., Utz, P. J., Hale, M., Boyd, S. D., Shen-Orr, S. S., Fathman, C. G. 2012; 8 (6): 317-328

    Abstract

    Rheumatologists see patients with a range of autoimmune diseases. Phenotyping these diseases for diagnosis, prognosis and selection of therapies is an ever increasing problem. Advances in multiplexed assay technology at the gene, protein, and cellular level have enabled the identification of 'actionable biomarkers'; that is, biological metrics that can inform clinical practice. Not only will such biomarkers yield insight into the development, remission, and exacerbation of a disease, they will undoubtedly improve diagnostic sensitivity and accuracy of classification, and ultimately guide treatment. This Review provides an introduction to these powerful technologies that could promote the identification of actionable biomarkers, including mass cytometry, protein arrays, and immunoglobulin and T-cell receptor high-throughput sequencing. In our opinion, these technologies should become part of routine clinical practice for the management of autoimmune diseases. The use of analytical tools to deconvolve the data obtained from use of these technologies is also presented here. These analyses are revealing a more comprehensive and interconnected view of the immune system than ever before and should have an important role in directing future treatment approaches for autoimmune diseases.

    View details for DOI 10.1038/nrrheum.2012.66

    View details for PubMedID 22647780

  • Ectopic expression and presentation of diabetogenic antigens in the lymph nodes of NOD mice Creusot, R., Yip, L., Fathman, C. AMER ASSOC IMMUNOLOGISTS. 2012
  • Exploiting a natural conformational switch to engineer an interleukin-2 'superkine' NATURE Levin, A. M., Bates, D. L., Ring, A. M., Krieg, C., Lin, J. T., Su, L., Moraga, I., Raeber, M. E., Bowman, G. R., Novick, P., Pande, V. S., Fathman, C. G., Boyman, O., Garcia, K. C. 2012; 484 (7395): 529-U159

    Abstract

    The immunostimulatory cytokine interleukin-2 (IL-2) is a growth factor for a wide range of leukocytes, including T cells and natural killer (NK) cells. Considerable effort has been invested in using IL-2 as a therapeutic agent for a variety of immune disorders ranging from AIDS to cancer. However, adverse effects have limited its use in the clinic. On activated T cells, IL-2 signals through a quaternary 'high affinity' receptor complex consisting of IL-2, IL-2Rα (termed CD25), IL-2Rβ and IL-2Rγ. Naive T cells express only a low density of IL-2Rβ and IL-2Rγ, and are therefore relatively insensitive to IL-2, but acquire sensitivity after CD25 expression, which captures the cytokine and presents it to IL-2Rβ and IL-2Rγ. Here, using in vitro evolution, we eliminated the functional requirement of IL-2 for CD25 expression by engineering an IL-2 'superkine' (also called super-2) with increased binding affinity for IL-2Rβ. Crystal structures of the IL-2 superkine in free and receptor-bound forms showed that the evolved mutations are principally in the core of the cytokine, and molecular dynamics simulations indicated that the evolved mutations stabilized IL-2, reducing the flexibility of a helix in the IL-2Rβ binding site, into an optimized receptor-binding conformation resembling that when bound to CD25. The evolved mutations in the IL-2 superkine recapitulated the functional role of CD25 by eliciting potent phosphorylation of STAT5 and vigorous proliferation of T cells irrespective of CD25 expression. Compared to IL-2, the IL-2 superkine induced superior expansion of cytotoxic T cells, leading to improved antitumour responses in vivo, and elicited proportionally less expansion of T regulatory cells and reduced pulmonary oedema. Collectively, we show that in vitro evolution has mimicked the functional role of CD25 in enhancing IL-2 potency and regulating target cell specificity, which has implications for immunotherapy.

    View details for DOI 10.1038/nature10975

    View details for Web of Science ID 000303200400054

    View details for PubMedID 22446627

    View details for PubMedCentralID PMC3338870

  • Therapeutic Effects of Systemic Administration of Chaperone alpha B-Crystallin Associated with Binding Proinflammatory Plasma Proteins JOURNAL OF BIOLOGICAL CHEMISTRY Rothbard, J. B., Kurnellas, M. P., Brownell, S., Adams, C. M., Su, L., Axtell, R. C., Chen, R., Fathman, C. G., Robinson, W. H., Steinman, L. 2012; 287 (13): 9708-9721

    Abstract

    The therapeutic benefit of the small heat shock protein αB-crystallin (HspB5) in animal models of multiple sclerosis and ischemia is proposed to arise from its increased capacity to bind proinflammatory proteins at the elevated temperatures within inflammatory foci. By mass spectral analysis, a common set of ∼70 ligands was precipitated by HspB5 from plasma from patients with multiple sclerosis, rheumatoid arthritis, and amyloidosis and mice with experimental allergic encephalomyelitis. These proteins were distinguished from other precipitated molecules because they were enriched in the precipitate as compared with their plasma concentrations, and they exhibited temperature-dependent binding. More than half of these ligands were acute phase proteins or members of the complement or coagulation cascades. Consistent with this proposal, plasma levels of HspB5 were increased in patients with multiple sclerosis as compared with normal individuals. The combination of the thermal sensitivity of the HspB5 combined with the high local concentration of these ligands at the site of inflammation is proposed to explain the paradox of how a protein believed to exhibit nonspecific binding can bind with some relative apparent selectivity to proinflammatory proteins and thereby modulate inflammation.

    View details for DOI 10.1074/jbc.M111.337691

    View details for PubMedID 22308023

  • Differential mTOR and ERK pathway utilization by effector CD4 T cells suggests combinatorial drug therapy of arthritis CLINICAL IMMUNOLOGY Lin, J. T., Stein, E. A., Wong, M. T., Kalpathy, K. J., Su, L. L., Utz, P. J., Robinson, W. H., Fathnnan, C. G. 2012; 142 (2): 127-138

    Abstract

    The signaling pathways utilized by naïve and experienced effector CD4 T cells during activation and proliferation were evaluated. While inhibition of either mTOR or MAPK alone was able to inhibit naïve T cell proliferation, both mTOR and MAPK (ERK) pathway inhibition was required to efficiently block experienced, effector CD4 T cell proliferation. This was demonstrated both in vitro, and in vivo by treating mice with collagen-induced arthritis using mTOR and/or ERK inhibitors. The combination of mTOR and ERK inhibition prevented or treated disease more efficiently than either agent alone. These data illustrate the different requirements of naïve and experienced effector CD4 T cells in the use of the mTOR and MAPK pathways in proliferation, and suggest that therapies targeting both the mTOR and MAPK pathways may be more effective than targeting either pathway alone in the treatment of CD4 T cell-mediated autoimmunity.

    View details for DOI 10.1016/j.clim.2011.09.008

    View details for PubMedID 22075384

  • SLE patients and autoantibody-positive healthy individuals display unique cytokine profiles: shared features of inflammation as well as select features of immunosuppression in autoantibody-positive healthy individuals Ritterhouse, L. L., Maecker, H. T., Fathman, C. G., Merrill, J. T., Guthridge, J. M., James, J. A. BIOMED CENTRAL LTD. 2012
  • Vitamin D Deficient Healthy Individuals Have Decreased Activated T Cells and Altered Lymphocyte Responses to Cytokine Stimulation 75th Annual Scientific Meeting of the American-College-of-Rheumatology/46th Annual Scientific Meeting of the Association-of-Rheumatology-Health-Professionals (ARHP) Ritterhouse, L. L., Maecker, H. T., Du, H., Fathman, C. G., Guthridge, J., James, J. A. WILEY-BLACKWELL. 2011: S19–S19
  • What Keeps An Autoantibody-Positive Healthy Individual Healthy? 75th Annual Scientific Meeting of the American-College-of-Rheumatology/46th Annual Scientific Meeting of the Association-of-Rheumatology-Health-Professionals (ARHP) Ritterhouse, L. L., Maecker, H. T., Du, H., Fathman, C. G., Merrill, J. T., Guthridge, J., James, J. A. WILEY-BLACKWELL. 2011: S1008–S1009
  • Antigen-based therapy with glutamic acid decarboxylase (GAD) vaccine in patients with recent-onset type 1 diabetes: a randomised double-blind trial LANCET Wherrett, D. K., Bundy, B., Becker, D. J., Dimeglio, L. A., Gitelman, S. E., Goland, R., Gottlieb, P. A., Greenbaum, C. J., Herold, K. C., Marks, J. B., Monzavi, R., Moran, A., Orban, T., Palmer, J. P., Raskin, P., Rodriguez, H., Schatz, D., Wilson, D. M., Krischer, J. P., Skyler, J. S. 2011; 378 (9788): 319-327

    Abstract

    Glutamic acid decarboxylase (GAD) is a major target of the autoimmune response that occurs in type 1 diabetes mellitus. In animal models of autoimmunity, treatment with a target antigen can modulate aggressive autoimmunity. We aimed to assess whether immunisation with GAD formulated with aluminum hydroxide (GAD-alum) would preserve insulin production in recent-onset type 1 diabetes.Patients aged 3-45 years who had been diagnosed with type 1 diabetes for less than 100 days were enrolled from 15 sites in the USA and Canada, and randomly assigned to receive one of three treatments: three injections of 20 μg GAD-alum, two injections of 20 μg GAD-alum and one of alum, or 3 injections of alum. Injections were given subcutaneously at baseline, 4 weeks later, and 8 weeks after the second injection. The randomisation sequence was computer generated at the TrialNet coordinating centre. Patients and study personnel were masked to treatment assignment. The primary outcome was the baseline-adjusted geometric mean area under the curve (AUC) of serum C-peptide during the first 2 h of a 4-h mixed meal tolerance test at 1 year. Secondary outcomes included changes in glycated haemoglobin A(1c) (HbA(1c)) and insulin dose, and safety. Analysis included all randomised patients with known measurements. This trial is registered with ClinicalTrials.gov, number NCT00529399.145 patients were enrolled and treated with GAD-alum (n=48), GAD-alum plus alum (n=49), or alum (n=48). At 1 year, the 2-h AUC of C-peptide, adjusted for age, sex, and baseline C-peptide value, was 0·412 nmol/L (95% CI 0·349-0·478) in the GAD-alum group, 0·382 nmol/L (0·322-0·446) in the GAD-alum plus alum group, and 0·413 nmol/L (0·351-0·477) in the alum group. The ratio of the population mean of the adjusted geometric mean 2-h AUC of C-peptide was 0·998 (95% CI 0·779-1·22; p=0·98) for GAD-alum versus alum, and 0·926 (0·720-1·13; p=0·50) for GAD-alum plus alum versus alum. HbA(1c), insulin use, and the occurrence and severity of adverse events did not differ between groups.Antigen-based immunotherapy therapy with two or three doses of subcutaneous GAD-alum across 4-12 weeks does not alter the course of loss of insulin secretion during 1 year in patients with recently diagnosed type 1 diabetes. Although antigen-based therapy is a highly desirable treatment and is effective in animal models, translation to human autoimmune disease remains a challenge.US National Institutes of Health.

    View details for DOI 10.1016/S0140-6736(11)60895-7

    View details for Web of Science ID 000293615800029

    View details for PubMedID 21714999

  • Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial LANCET Orban, T., Bundy, B., Becker, D. J., Dimeglio, L. A., Gitelman, S. E., Goland, R., Gottlieb, P. A., Greenbaum, C. J., Marks, J. B., Monzavi, R., Moran, A., Raskin, P., Rodriguez, H., Russell, W. E., Schatz, D., Wherrett, D., Wilson, D. M., Krischer, J. P., Skyler, J. S. 2011; 378 (9789): 412-419

    Abstract

    The immunopathogenesis of type 1 diabetes mellitus is associated with T-cell autoimmunity. To be fully active, immune T cells need a co-stimulatory signal in addition to the main antigen-driven signal. Abatacept modulates co-stimulation and prevents full T-cell activation. We evaluated the effect of abatacept in recent-onset type 1 diabetes.In this multicentre, double-blind, randomised controlled trial, patients aged 6-45 years recently diagnosed with type 1 diabetes were randomly assigned (2:1) to receive abatacept (10 mg/kg, maximum 1000 mg per dose) or placebo infusions intravenously on days 1, 14, 28, and monthly for a total of 27 infusions over 2 years. Computer-generated permuted block randomisation was used, with a block size of 3 and stratified by participating site. Neither patients nor research personnel were aware of treatment assignments. The primary outcome was baseline-adjusted geometric mean 2-h area-under-the-curve (AUC) serum C-peptide concentration after a mixed-meal tolerance test at 2 years' follow-up. Analysis was by intention to treat for all patients for whom data were available. This trial is registered at ClinicalTrials.gov, NCT00505375.112 patients were assigned to treatment groups (77 abatacept, 35 placebo). Adjusted C-peptide AUC was 59% (95% CI 6·1-112) higher at 2 years with abatacept (n=73, 0·378 nmol/L) than with placebo (n=30, 0·238 nmol/L; p=0·0029). The difference between groups was present throughout the trial, with an estimated 9·6 months' delay (95% CI 3·47-15·6) in C-peptide reduction with abatacept. There were few infusion-related adverse events (36 reactions occurred in 17 [22%] patients on abatacept and 11 reactions in six [17%] on placebo). There was no increase in infections (32 [42%] patients on abatacept vs 15 [43%] on placebo) or neutropenia (seven [9%] vs five [14%]).Co-stimulation modulation with abatacept slowed reduction in β-cell function over 2 years. The beneficial effect suggests that T-cell activation still occurs around the time of clinical diagnosis of type 1 diabetes. Yet, despite continued administration of abatacept over 24 months, the decrease in β-cell function with abatacept was parallel to that with placebo after 6 months of treatment, causing us to speculate that T-cell activation lessens with time. Further observation will establish whether the beneficial effect continues after cessation of abatacept infusions.US National Institutes of Health.

    View details for DOI 10.1016/S0140-6736(11)60886-6

    View details for Web of Science ID 000293615900032

    View details for PubMedID 21719096

  • Engineering cell-type selective immune responses using mechanism-based designer IL-4 cytokines. Creusot, R., Junttila, I., Bates, D., Moraga, I., Lupardus, P., Fathman, C., Paul, W., Garcia, K. AMER ASSOC IMMUNOLOGISTS. 2011
  • GRAIL targets CDC37 to maintain CD4 T cell unresponsiveness Whiting, C., Su, L., Lin, J., Lineberry, N., Fathman, C. AMER ASSOC IMMUNOLOGISTS. 2011
  • Dengue-2 Structural Proteins Associate with Human Proteins to Produce a Coagulation and Innate Immune Response Biased Interactome BMC INFECTIOUS DISEASES Folly, B. B., Weffort-Santos, A. M., Fathman, C. G., Soares, L. R. 2011; 11

    Abstract

    Dengue virus infection is a public health threat to hundreds of millions of individuals in the tropical regions of the globe. Although Dengue infection usually manifests itself in its mildest, though often debilitating clinical form, dengue fever, life-threatening complications commonly arise in the form of hemorrhagic shock and encephalitis. The etiological basis for the virus-induced pathology in general, and the different clinical manifestations in particular, are not well understood. We reasoned that a detailed knowledge of the global biological processes affected by virus entry into a cell might help shed new light on this long-standing problem.A bacterial two-hybrid screen using DENV2 structural proteins as bait was performed, and the results were used to feed a manually curated, global dengue-human protein interaction network. Gene ontology and pathway enrichment, along with network topology and microarray meta-analysis, were used to generate hypothesis regarding dengue disease biology.Combining bioinformatic tools with two-hybrid technology, we screened human cDNA libraries to catalogue proteins physically interacting with the DENV2 virus structural proteins, Env, cap and PrM. We identified 31 interacting human proteins representing distinct biological processes that are closely related to the major clinical diagnostic feature of dengue infection: haemostatic imbalance. In addition, we found dengue-binding human proteins involved with additional key aspects, previously described as fundamental for virus entry into cells and the innate immune response to infection. Construction of a DENV2-human global protein interaction network revealed interesting biological properties suggested by simple network topology analysis.Our experimental strategy revealed that dengue structural proteins interact with human protein targets involved in the maintenance of blood coagulation and innate anti-viral response processes, and predicts that the interaction of dengue proteins with a proposed human protein interaction network produces a modified biological outcome that may be behind the hallmark pathologies of dengue infection.

    View details for DOI 10.1186/1471-2334-11-34

    View details for Web of Science ID 000287239500001

    View details for PubMedID 21281507

    View details for PubMedCentralID PMC3037883

  • GRAIL: a unique mediator of CD4 T-lymphocyte unresponsiveness FEBS JOURNAL Whiting, C. C., Su, L. L., Lin, J. T., Fathman, C. G. 2011; 278 (1): 47-58

    Abstract

    GRAIL (gene related to anergy in lymphocytes, also known as RNF128), an ubiquitin-protein ligase (E3), utilizes a unique single transmembrane protein with a split-function motif, and is an important gatekeeper of T-cell unresponsiveness. Although it may play a role in other CD4 T-cell functions including activation, survival and differentiation, GRAIL is most well characterized as a negative regulator of T-cell receptor responsiveness and cytokine production. Here, we review the recent literature on this remarkable E3 in the regulation of human and mouse CD4 T-cell unresponsiveness.

    View details for DOI 10.1111/j.1742-4658.2010.07922.x

    View details for Web of Science ID 000285249100006

    View details for PubMedID 21078124

    View details for PubMedCentralID PMC3058357

  • A Short Pulse of IL-4 Delivered by DCs Electroporated With Modified mRNA Can Both Prevent and Treat Autoimmune Diabetes in NOD Mice MOLECULAR THERAPY Creusot, R. J., Chang, P., Healey, D. G., Tcherepanova, I. Y., Nicolette, C. A., Fathman, C. G. 2010; 18 (12): 2112-2120

    Abstract

    Bone marrow-derived dendritic cells (DCs) are cells of the immune system that have been used as a tool to boost, modulate, or dampen immune responses. In the context of autoimmunity, DCs can be modified to express immunoregulatory products encoded by transgenes, and used therapeutically in adoptive cellular therapy. DCs that were lentivirally transduced (lt) to express interleukin 4 (IL-4) can significantly delay or prevent the onset of autoimmune diabetes in nonobese diabetic (NOD) mice. However, modifying cells using viral vectors carries the dual risk of oncogenicity or immunogenicity. This study demonstrates that NOD DCs, electroporated with "translationally enhanced" IL-4 mRNA (eDC/IL-4), can be equally efficient therapeutically, despite the reduced amount and shorter duration of IL-4 secretion. Moreover, a single injection of eDC/IL-4 in NOD mice shortly after the onset of hyperglycemia was able to maintain stable glycemia for up to several months in a significant fraction of treated mice. Treatment with eDC/IL-4 boosted regulatory T (Tregs) cell functions and modulated T helper responses to reduce pathogenicity. Thus, treatment with DCs, electroporated with modified IL-4 mRNA to express IL-4 for up to 24 hours, constitutes a viable cellular therapy approach for the regulation of autoimmune diabetes, as a preferred alternative to the use of viral vectors.

    View details for DOI 10.1038/mt.2010.146

    View details for Web of Science ID 000284828000012

    View details for PubMedID 20628358

    View details for PubMedCentralID PMC2997578

  • A model for harmonizing flow cytometry in clinical trials. Nature immunology Maecker, H. T., McCoy, J. P., Amos, M., Elliott, J., Gaigalas, A., Wang, L., Aranda, R., Banchereau, J., Boshoff, C., Braun, J., Korin, Y., Reed, E., Cho, J., Hafler, D., Davis, M., Fathman, C. G., Robinson, W., Denny, T., Weinhold, K., Desai, B., Diamond, B., Gregersen, P., Di Meglio, P., Nestle, F. O., Peakman, M., Villanova, F., Ferbas, J., Field, E., Kantor, A., Kawabata, T., Komocsar, W., Lotze, M., Nepom, J., Ochs, H., O'Lone, R., Phippard, D., Plevy, S., Rich, S., Roederer, M., Rotrosen, D., Yeh, J. 2010; 11 (11): 975-978

    Abstract

    Complexities in sample handling, instrument setup and data analysis are barriers to the effective use of flow cytometry to monitor immunological parameters in clinical trials. The novel use of a central laboratory may help mitigate these issues.

    View details for DOI 10.1038/ni1110-975

    View details for PubMedID 20959798

    View details for PubMedCentralID PMC3400260

  • A model for harmonizing flow cytometry in clinical trials NATURE IMMUNOLOGY Maecker, H. T., McCoy, J. P. 2010; 11 (11): 975-978

    Abstract

    Complexities in sample handling, instrument setup and data analysis are barriers to the effective use of flow cytometry to monitor immunological parameters in clinical trials. The novel use of a central laboratory may help mitigate these issues.

    View details for DOI 10.1038/ni1110-975

    View details for Web of Science ID 000283127800003

    View details for PubMedCentralID PMC3400260

  • New technologies for autoimmune disease monitoring CURRENT OPINION IN ENDOCRINOLOGY DIABETES AND OBESITY Maecker, H. T., Nolan, G. P., Fathman, C. G. 2010; 17 (4): 322-328

    Abstract

    This article will review new technologies used to characterize the immune phenotype of cells and serum for potential use in studies of autoimmunity.One area of recent development in studies of immune phenotyping is the contrast between cells of the immune system at rest and following activation. This simply involves comparing these cells at rest and following ligand-induced activation and measuring signaling system activation (phosphoepitope identification) or intracellular cytokine production or activation-induced gene expression. Preliminary data using these techniques have begun to identify signatures of disease (biomarkers) that are only seen when using these activation-induced assays. One of the most exciting new technologies, cytometry by time-of-flight mass spectrometry, couples a flow cytometer to a mass spectrometer, allowing many more parameters to be analyzed per cell, and without spillover between assay reagents, compared to conventional optical flow cytometry (heavy ions, mass, replaces fluorophore readout). Another new technology to analyze soluble proteins, bead-based immunoassays, can simultaneously measure up to 75 soluble analytes in a multiplexed array. Other technologies provide similar innovations in terms of analytical content, throughput, and miniaturization.We believe that new cellular genomic and protein-based technologies can provide key insights into autoimmune disease pathogenesis, progression, and therapy, and that these assays need to be applied in a systematic way to samples from patients with autoimmune diseases.

    View details for DOI 10.1097/MED.0b013e32833ada91

    View details for Web of Science ID 000285063800003

    View details for PubMedID 20531181

  • Inflammation-induced Changes in Deaf1 Splicing Alter Peripheral Tissue Antigen Gene Expression in the Pancreatic Lymph Node during the Pathogenesis of Type I Diabetes 10th Annual Meeting of the Federation-of-Clinical-Immunology-Societies Yip, L., Creusot, R., Su, L., Fathman, C. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2010: S72–S72
  • Targeting mTOR and MAPK Pathways to Inhibit Naive and Experienced Effector CD4 T Cells in Autoimmunity 10th Annual Meeting of the Federation-of-Clinical-Immunology-Societies Lin, J., Stein, E., Wong, M., Kalpathy, K., Su, L., Utz, P., Robinson, W., Fathman, C. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2010: S69–S69
  • Deaf1 isoforms control the expression of genes encoding peripheral tissue antigens in the pancreatic lymph nodes during type 1 diabetes NATURE IMMUNOLOGY Yip, L., Su, L., Sheng, D., Chang, P., Atkinson, M., Czesak, M., Albert, P. R., Collier, A., Turley, S. J., Fathman, C. G., Creusot, R. J. 2009; 10 (9): 1026-U107

    Abstract

    Type 1 diabetes may result from a breakdown in peripheral tolerance that is partially controlled by the expression of peripheral tissue antigens (PTAs) in lymph nodes. Here we show that the transcriptional regulator Deaf1 controls the expression of genes encoding PTAs in the pancreatic lymph nodes (PLNs). The expression of canonical Deaf1 was lower, whereas that of an alternatively spliced variant was higher, during the onset of destructive insulitis in the PLNs of nonobese diabetic (NOD) mice. We identified an equivalent variant Deaf1 isoform in the PLNs of patients with type 1 diabetes. Both the NOD mouse and human Deaf1 variant isoforms suppressed PTA expression by inhibiting the transcriptional activity of canonical Deaf1. Lower PTA expression resulting from the alternative splicing of DEAF1 may contribute to the pathogenesis of type 1 diabetes.

    View details for DOI 10.1038/ni.1773

    View details for Web of Science ID 000269120900017

    View details for PubMedID 19668219

    View details for PubMedCentralID PMC2752139

  • The Transmembrane E3 Ligase GRAIL Ubiquitinates and Degrades CD83 on CD4 T Cells JOURNAL OF IMMUNOLOGY Su, L. L., Iwai, H., Lin, J. T., Fathman, C. G. 2009; 183 (1): 438-444

    Abstract

    Ubiquitination of eukaryotic proteins regulates a broad range of cellular processes, including T cell activation and tolerance. We have previously demonstrated that GRAIL (gene related to anergy in lymphocytes), a transmembrane RING finger ubiquitin E3 ligase, initially described as induced during the induction of CD4 T cell anergy, is also expressed in resting CD4 T cells. In this study, we show that GRAIL can down-modulate the expression of CD83 (previously described as a cell surface marker for mature dendritic cells) on CD4 T cells. GRAIL-mediated down-modulation of CD83 is dependent on an intact GRAIL extracellular protease-associated domain and an enzymatically active cytosolic RING domain, and proceeds via the ubiquitin-dependent 26S proteosome pathway. Ubiquitin modification of lysine residues K168 and K183, but not K192, in the cytoplasmic domain of CD83 was shown to be necessary for GRAIL-mediated degradation of CD83. Reduced CD83 surface expression levels were seen both on anergized CD4 T cells and following GRAIL expression by retroviral transduction, whereas GRAIL knock-down by RNA interference in CD4 T cells resulted in elevated CD83 levels. Furthermore, CD83 expression on CD4 T cells contributes to T cell activation as a costimulatory molecule. This study supports the novel mechanism of ubiquitination by GRAIL, identifies CD83 as a substrate of GRAIL, and ascribes a role for CD83 in CD4 T cell activation.

    View details for DOI 10.4049/jimmunol.0900204

    View details for Web of Science ID 000275119400048

    View details for PubMedID 19542455

  • Lymphoid tissue-specific homing of bone marrow-derived dendritic cells BLOOD Creusot, R. J., Yaghoubi, S. S., Chang, P., Chia, J., Contag, C. H., Gambhir, S. S., Fathman, C. G. 2009; 113 (26): 6638-6647

    Abstract

    Because of their potent immunoregulatory capacity, dendritic cells (DCs) have been exploited as therapeutic tools to boost immune responses against tumors or pathogens, or dampen autoimmune or allergic responses. Murine bone marrow-derived DCs (BM-DCs) are the closest known equivalent of the blood monocyte-derived DCs that have been used for human therapy. Current imaging methods have proven unable to properly address the migration of injected DCs to small and deep tissues in mice and humans. This study presents the first extensive analysis of BM-DC homing to lymph nodes (and other selected tissues) after intravenous and intraperitoneal inoculation. After intravenous delivery, DCs accumulated in the spleen, and preferentially in the pancreatic and lung-draining lymph nodes. In contrast, DCs injected intraperitoneally were found predominantly in peritoneal lymph nodes (pancreatic in particular), and in omentum-associated lymphoid tissue. This uneven distribution of BM-DCs, independent of the mouse strain and also observed within pancreatic lymph nodes, resulted in the uneven induction of immune response in different lymphoid tissues. These data have important implications for the design of systemic cellular therapy with DCs, and in particular underlie a previously unsuspected potential for specific treatment of diseases such as autoimmune diabetes and pancreatic cancer.

    View details for DOI 10.1182/blood-2009-02-204321

    View details for Web of Science ID 000267789600024

    View details for PubMedID 19363220

    View details for PubMedCentralID PMC2710920

  • Naive CD4 T Cell Proliferation Is Controlled by Mammalian Target of Rapainaycin Regulation of GRAIL Expression JOURNAL OF IMMUNOLOGY Lin, J. T., Lineberry, N. B., Kattah, M. G., Su, L. L., Utz, P. J., Fathman, C. G., Wu, L. 2009; 182 (10): 5919-5928

    Abstract

    In this study, we demonstrate that the E3 ubiquitin ligase gene related to anergy in lymphocytes (GRAIL) is expressed in quiescent naive mouse and human CD4 T cells and has a functional role in inhibiting naive T cell proliferation. Following TCR engagement, CD28 costimulation results in the expression of IL-2 whose signaling through its receptor activates the Akt-mammalian target of rapamycin (mTOR) pathway. Activation of mTOR allows selective mRNA translation, including the epistatic regulator of GRAIL, Otubain-1 (Otub1), whose expression results in the degradation of GRAIL and allows T cell proliferation. The activation of mTOR appears to be the critical component of IL-2R signaling regulating GRAIL expression. CTLA4-Ig treatment blocks CD28 costimulation and resultant IL-2 expression, whereas rapamycin and anti-IL-2 treatment block mTOR activation downstream of IL-2R signaling. Thus, all three of these biotherapeutics inhibit mTOR-dependent translation of mRNA transcripts, resulting in blockade of Otub1 expression, maintenance of GRAIL, and inhibition of CD4 T cell proliferation. These observations provide a mechanistic pathway sequentially linking CD28 costimulation, IL-2R signaling, and mTOR activation as important requirements for naive CD4 T cell proliferation through the regulation of Otub1 and GRAIL expression. Our findings also extend the role of GRAIL beyond anergy induction and maintenance, suggesting that endogenous GRAIL regulates general cell cycle and proliferation of primary naive CD4 T cells.

    View details for DOI 10.4049/jimmunol.0803986

    View details for Web of Science ID 000265899800008

    View details for PubMedID 19414743

    View details for PubMedCentralID PMC2853371

  • Adoptive Immunotherapy of Experimental Autoimmune Hearing Loss via T Cell Delivery of the IL-12 p40 Subunit 65th Annual Meeting of the American-Academy-of-Allergy-Asthma-and-Immunology Zhou, B., Kermany, M. H., Zhou, Y., Cai, C., Cai, Q., Tamer, I. H., Fathman, C. G., Kim, P., Kim, J., Liu, W., Yoo, T. MOSBY-ELSEVIER. 2009: S208–S208
  • A Short Pulse of IL-4 Delivered Locally by mRNA Electroporated DCs is Sufficient to Prevent or Treat Autoimmune Diabetes in NOD Mice Creusot, R., Chang, P., Yaghoubi, S., Healey, D., Tcherepanova, I., Nicolette, C., Fathman, C. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2009: S53
  • CTLA-4 Ig Serves as a Surrogate for Tregs by Engaging Activation Induced CD80/86 on Murine T Effector Cells Acharya, S., Fathman, C. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2009: S127
  • The TransmembraneE3 Ligase, GRAIL Ubiquitinates and Degrades CD83 on CD4+T cells 9th Annual Meeting of the Federation-of-Clinical-Immunology-Societies Su, L., Iwai, H., Lin, J., Fathman, C. G. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2009: S115–S115
  • Tissue- and age-specific changes in gene expression during disease induction and progression in NOD mice CLINICAL IMMUNOLOGY Kodama, K., Butte, A. J., Creusot, R. J., Su, L., Sheng, D., Hartnett, M., Iwai, H., Soares, L. R., Fathman, C. G. 2008; 129 (2): 195-201

    Abstract

    Whole genome oligo-microarrays were used to characterize age-dependent and tissue-specific changes in gene expression in pancreatic lymph nodes, spleen, and peripheral blood cells, obtained from up to 8 individual NOD mice at 6 different time points (1.5 to 20 weeks of age), compared to NOD.B10 tissue controls. "Milestone Genes" are genes whose expression was significantly changed (approximately 3 fold) as the result of splicing or changes in transcript level. Milestone Genes were identified among genes within type one diabetes (T1D) susceptibility regions (Idd). Milestone Genes showing uniform patterns of changes in expression at various time points were identified, but the patterns of distribution and kinetics of expression were unique to each tissue. Potential T1D candidate genes were identified among Milestone Genes within Idd regions and/or hierarchical clusters. These studies identified tissue- and age-specific changes in gene expression that may play an important role in the inductive or destructive events of T1D.

    View details for DOI 10.1016/j.clim.2008.07.028

    View details for Web of Science ID 000260553600003

    View details for PubMedID 18801706

    View details for PubMedCentralID PMC2592195

  • The single subunit transmembrane E3 ligase gene related to anergy in lymphocytes (GRAIL) captures and then ubiquitinates transmembrane proteins across the cell membrane JOURNAL OF BIOLOGICAL CHEMISTRY Lineberry, N., Su, L., Soares, L., Fathman, C. G. 2008; 283 (42): 28497-28505

    Abstract

    The ubiquitin E3 ligase gene related to anergy in lymphocytes (GRAIL) (Rnf128) is a type 1 transmembrane protein that induces T cell anergy through the ubiquitination activity of its cytosolic RING finger. GRAIL also contains an equally large luminal region consisting primarily of an uncharacterized protease-associated (PA) domain. Using two-hybrid technology to screen for proteins that bound the PA domain we identified CD151, a member of the tetraspanin family of membrane proteins. GRAIL bound to the luminal/extracellular portion of both CD151 and the related tetraspanin CD81 using its PA domain, which promoted ubiquitination of cytosolic lysine residues. GRAIL exhibited specificity for lysines only within the tetraspanin amino terminus even in the presence of other cytosolic lysine residues in the substrate. GRAIL-mediated ubiquitination promoted proteasomal degradation and cell surface down-regulation of tetraspanins via Lys-48 linkages. As a result, the juxtaposition of PA and RING finger domains across a lipid bilayer facilitates the capture of transmembrane substrates for subsequent ubiquitination. These findings identify for the first time a single subunit E3 ligase containing a substrate-binding domain spatially restricted by a membrane from its E2 recruitment domain as well as an E3 ligase for members of the tetraspanin family.

    View details for DOI 10.1074/jbc.M805092200

    View details for Web of Science ID 000259969300052

    View details for PubMedID 18713730

    View details for PubMedCentralID PMC2568916

  • Cutting edge: The transmembrane E3 ligase GRAIL ubiquitinates the costimulatory molecule CD40 ligand during the induction of T cell anergy JOURNAL OF IMMUNOLOGY Lineberry, N. B., Su, L. L., Lin, J. T., Coffey, G. P., Seroogy, C. M., Fathman, C. G. 2008; 181 (3): 1622-1626

    Abstract

    Activation of naive T lymphocytes is regulated through a series of discrete checkpoints that maintain unresponsiveness to self. During this multistep process, costimulatory interactions act as inducible signals that allow APCs to selectively mobilize T cells against foreign Ags. In this study, we provide evidence that the anergy-associated E3 ubiquitin ligase GRAIL (gene related to anergy in lymphocytes) regulates expression of the costimulatory molecule CD40L on CD4 T cells. Using its luminal protease-associated domain, GRAIL binds to the luminal/extracellular portion of CD40L and facilitates transfer of ubiquitin molecules from the intracellular GRAIL RING (really interesting new gene) finger to the small cytosolic portion of CD40L. Down-regulation of CD40L occurred following ectopic expression of GRAIL in naive T cells from CD40(-/-) mice, and expression of GRAIL in bone marrow chimeric mice was associated with diminished lymphoid follicle formation. These data provide a model for intrinsic T cell regulation of costimulatory molecules and a molecular framework for the initiation of clonal T cell anergy.

    View details for Web of Science ID 000257898100006

    View details for PubMedID 18641297

  • Tissue-targeted therapy of autoimmune diabetes using dendritic cells transduced to express IL-4 in NOD mice CLINICAL IMMUNOLOGY Creusot, R. J., Yaghoubi, S. S., Kodama, K., Dang, D. N., Dang, V. H., Breckpot, K., Thielemans, K., Gambhir, S. S., Fathman, C. G. 2008; 127 (2): 176-187

    Abstract

    A deficit in IL-4 production has been previously reported in both diabetic human patients and non-obese diabetic (NOD) mice. In addition, re-introducing IL-4 into NOD mice systemically, or as a transgene, led to a beneficial outcome in most studies. Here, we show that prediabetic, 12-week old female NOD mice have a deficit in IL-4 expression in the pancreatic lymph nodes (PLN) compared to age-matched diabetes-resistant NOD.B10 mice. By bioluminescence imaging, we demonstrated that the PLN was preferentially targeted by bone marrow-derived dendritic cells (DCs) following intravenous (IV) administration. Following IV injection of DCs transduced to express IL-4 (DC/IL-4) into 12-week old NOD mice, it was possible to significantly delay or prevent the onset of hyperglycemia. We then focused on the PLN to monitor, by microarray analysis, changes in gene expression induced by DC/IL-4 and observed a rapid normalization of the expression of many genes, that were otherwise under-expressed compared to NOD.B10 PLN. The protective effect of DC/IL-4 required both MHC and IL-4 expression by the DCs. Thus, adoptive cellular therapy, using DCs modified to express IL-4, offers an effective, tissue-targeted cellular therapy to prevent diabetes in NOD mice at an advanced stage of pre-diabetes, and may offer a safe approach to consider for treatment of high risk human pre-diabetic patients.

    View details for DOI 10.1016/j.clim.2007.12.009

    View details for Web of Science ID 000255231100010

    View details for PubMedID 18337172

    View details for PubMedCentralID PMC2453076

  • The earliest islet-infiltrating T cells in NOD mice Creusot, R., Chang, P., Fathman, C. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2008: S108–S109
  • ShRNA mediated downregulation of genes identified as 'Core Transcriptome' in murine T regulatory cells 8th Annual Meeting of the Federation-of-Clinical-Immunology-Societies Acharya, S., Hacohen, N., Soares, L., Fathman, C. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2008: S16–S16
  • Time-dependent and tissue-specific changes in gene expression during disease induction and progression in NOD mice 8th Annual Meeting of the Federation-of-Clinical-Immunology-Societies Yip, L., Kodama, K., Butte, A., Creusot, R., Su, L., Fathman, C. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2008: S40–S40
  • High cell surface expression of CD4 allows distinction of CD4(+)CD25(+) antigen-specific effector T cells from CD4(+)CD25(+) regulatoiy T cells in murine experimental autoimmune encephalomyelitis JOURNAL OF NEUROIMMUNOLOGY Li, J., Ridgway, W., Fathman, C. G., Tse, H. Y., Shaw, M. K. 2007; 192 (1-2): 57-67

    Abstract

    Analysis of T regulatory cells (Treg) and T effector cells (Teff) in experimental autoimmune encephalomyelitis is complicated by the fact that both cell types express CD4 and CD25. We demonstrate that encephalitogenic T cells, following antigen recognition, up-regulate cell surface expression of CD4. The CD4(high) sub-population contains all of the antigen response as shown by proliferation and cytokine secretion, and only these cells are capable of transferring EAE to naive animals. On the other hand, a FACS separable CD25(+) sub-population of cells displayed consistent levels of CD4 prior to and after antigen stimulation. These cells displayed characteristics of Treg, such as expressing high levels of the Foxp3 gene and the ability to suppress mitogenic T cell responses.

    View details for DOI 10.1016/j.jneuroim.2007.09.004

    View details for Web of Science ID 000252163200008

    View details for PubMedID 17920698

    View details for PubMedCentralID PMC2212588

  • Naive and memory T cells induce different types of graft-versus-host disease JOURNAL OF IMMUNOLOGY Dutt, S., Tseng, D., Ermann, J., George, T. I., Liu, Y. P., Davis, C. R., Fathman, C. G., Strober, S. 2007; 179 (10): 6547-6554

    Abstract

    The goal of this study was to compare the ability of donor naive and alloantigen-primed effector memory T cells to induce graft-vs-host disease after bone marrow transplantation in MHC-mismatched irradiated host mice. Purified CD4(+) naive (CD62L(high)CD44(low)) T cells and CD4(+) effector memory (CD62L(low)CD44(high)) T cells obtained from unprimed donors and donors primed to host alloantigens, respectively, were injected into host mice, and the rapidity, severity, and pattern of tissue injury of graft-vs-host disease was assessed. Unexpectedly, the naive T cells induced a more acute and severe colitis than the primed memory cells. Whereas the naive T cells expressing CD62L and CCR7 lymph node homing receptors vigorously expanded in mesenteric lymph nodes and colon by day 6 after transplantation, the primed memory T cells without these receptors had 20- to 100-fold lower accumulation at this early time point. These differences were reflected in the significantly more rapid decline in survival and weight loss induced by naive T cells. The primed memory T cells had a greater capacity to induce chronic colitis and liver injury and secrete IL-2 and IFN-gamma in response to alloantigenic stimulation compared with memory T cells from unprimed donors. Nevertheless, the expected increase in potency as compared with naive T cells was not observed due to differences in the pattern and kinetics of tissue injury.

    View details for PubMedID 17982043

  • Multimodality imaging of T-cell hybridoma trafficking in collagen-induced arthritic mice: image-based estimation of the number of cells accumulating in mouse paws JOURNAL OF BIOMEDICAL OPTICS Yaghoubi, S. S., Creusot, R. J., Ray, P., Fathman, C. G., Gambhir, S. S. 2007; 12 (6)

    Abstract

    Appropriate targeting of therapeutic cells is essential in adoptive cellular gene therapy (ACGT). Imaging cell trafficking in animal models and patients will guide development of ACGT protocols. Collagen type II (C-II)-specific T cell hybridomas are transduced with a lentivirus carrying a triple fusion reporter gene (TFR) construct consisting of a fluorescent reporter gene (RG), a bioluminescent RG (hRluc), and a positron emission tomography (PET) RG. Collagen-induced arthritic (CIA) mice are scanned with a bioluminescence imaging camera before and after implantation of various known cell quantities in their paws. Linear regression analysis yields equations relating two parameters of image signal intensity in mice paws to the quantity of hRluc expressing cells in the paws. Afterward, trafficking of intravenously injected cells is studied by quantitative analysis of bioluminescence images. Comparison of the average cell numbers does not demonstrate consistently higher accumulation of T-cell hybridomas in the paws with higher inflammation scores, and injecting more cells does not cause increased accumulation. MicroPET images illustrate above background signal in the inflamed paws and chest areas of CIA mice. The procedures described in this study can be used to derive equations for cells expressing other bioluminescent RGs and in other animal models.

    View details for DOI 10.1117/1.2821415

    View details for Web of Science ID 000252851100046

    View details for PubMedID 18163841

  • Molecular mechanisms of CD4(+) T-cell anergy NATURE REVIEWS IMMUNOLOGY Fathman, C. G., Lineberry, N. B. 2007; 7 (8): 599-609

    Abstract

    Directing both innate and adaptive immune responses against foreign pathogens with correct timing, location and specificity is a fundamental objective for the immune system. Full activation of CD4+ T cells requires the binding of peptide-MHC complexes coupled with accessory signals provided by the antigen-presenting cell. However, aberrant activation of the T-cell receptor alone in mature T cells can produce a long-lived state of functional unresponsiveness, known as anergy. Recent studies probing both immune signalling pathways and the ubiquitin-proteasome system have helped to refine and elaborate current models for the molecular mechanisms underlying T-cell anergy. Controlling anergy induction and maintenance will be a key component in the future to mitigate unwanted T-cell activation that leads to autoimmune disease.

    View details for DOI 10.1038/nri2131

    View details for Web of Science ID 000248394100012

    View details for PubMedID 17612584

  • Preservation of self: An overview of E3 ubiquitin ligases and T cell tolerance SEMINARS IN IMMUNOLOGY Schartner, J. M., Fathman, C. G., Seroogy, C. M. 2007; 19 (3): 188-196

    Abstract

    Until recently ubiquitination of a protein was thought to simply serve the mundane task of targeting a protein for proteasomal degradation. Accumulating evidence over the past decade has demonstrated the importance of ubiquitination in non-degradative functions including regulating cellular signaling, that highlight its role in human disease and thus potential development of novel therapeutics. Much has been written about ubiquitination in the immune system, in this review we will outline our current knowledge of ubiquitination with respect to T cell tolerance. Specifically, we will provide on overview of E3 ubiquitin ligases and their role in various states of CD4+ T cell tolerance: central and peripheral.

    View details for DOI 10.1016/j.smim.2007.02.010

    View details for Web of Science ID 000247466200007

    View details for PubMedID 17403607

  • GRAIL is up-regulated in CD4(+) CD25(+) T regulatory cells and is sufficient for conversion of T cells to a regulatory phenotype JOURNAL OF BIOLOGICAL CHEMISTRY MacKenzie, D. A., Schartner, J., Lin, J., Timmel, A., Jennens-Clough, M., Fathman, C. G., Seroogy, C. M. 2007; 282 (13): 9696-9702

    Abstract

    GRAIL (gene related to anergy in lymphocytes) is an ubiquitin-protein isopeptide ligase (E3) ubiquitin ligase necessary for the induction of CD4(+) T cell anergy in vivo. We have extended our previous studies to characterize the expression pattern of GRAIL in other murine CD4(+) T cell types with a described anergic phenotype. These studies revealed that GRAIL expression is increased in naturally occurring (thymically derived) CD4(+) CD25(+) T regulatory cells (mRNA levels 10-fold higher than naive CD25(-) T cells). Further investigation demonstrated that CD25(+) Foxp3(+) antigen-specific T cells were induced after a "tolerizing-administration" of antigen and that GRAIL expression correlated with the CD25(+) Foxp3(+) antigen-specific subset. Lastly, using retroviral transduction, we demonstrated that forced expression of GRAIL in a T cell line was sufficient for conversion of these cells to a regulatory phenotype in the absence of detectable Foxp3. These data demonstrate that GRAIL is differentially expressed in naturally occurring and peripherally induced CD25(+) T regulatory cells and that the expression of GRAIL is linked to their functional regulatory activity.

    View details for DOI 10.1074/jbc.M604192200

    View details for Web of Science ID 000245421700043

    View details for PubMedID 17259178

  • Identification of candidate IDDM disease susceptibility genes in the idd regions of NOD mice by temporal microarray gene expression data analysis 7th Annual Meeting of the Federation-of-Clinical-Immunology-Societies Kodama, K., Dang, D., Hotness, C., Iwai, H., Hartnett, M., Butte, A., Fathman, C. G. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2007: S19–S19
  • Gene expression analysis by microarray following anti-CD3 antibody therapy of NOD mice Iwai, H., Kodama, K., Dang, D., Fathman, C., Bluestone, J. A. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2007: S9
  • A novel E3 ubiquitin ligase substrate screen identifies Rho guanine dissociation inhibitor as a substrate of gene related to anergy in lymphocytes JOURNAL OF IMMUNOLOGY Su, L., Lineberry, N., Huh, Y., Soares, L., Fathman, C. G. 2006; 177 (11): 7559-7566

    Abstract

    Ubiquitination of eukaryotic proteins regulates a broad range of cellular processes, including regulation of T cell activation and tolerance. We have previously demonstrated that gene related to anergy in lymphocytes (GRAIL), a ring finger ubiquitin E3 ligase, is required for the induction of T cell anergy; however, the substrate(s) for GRAIL E3 ligase activity is/are unknown. In this study, we report a novel prokaryotic system developed to screen for substrates of E3 ligases. Using this screen, Rho guanine dissociation inhibitor (RhoGDI) was identified as a potential substrate of GRAIL. GRAIL was subsequently demonstrated to bind and ubiquitinate RhoGDI, although GRAIL-mediated ubiquitination of RhoGDI did not result in proteosomal degradation. Expression of GRAIL in T cells resulted in specific inhibition of RhoA GTPase activation; activation of Rac1, cdc42, and Ras GTPases were not affected. Interestingly, stable T cell lines expressing dominant-negative RhoA mimicked the GRAIL-mediated IL-2 inhibition phenotype, and T cells expressing constitutively active RhoA were able to overcome GRAIL-mediated inhibition of IL-2 expression. These findings validate our prokaryotic screen as a method of identifying substrates for ubiquitin E3 ligases and suggest a role for Rho effector molecules in T cell anergy.

    View details for Web of Science ID 000242261800012

    View details for PubMedID 17114425

  • Allosensitized memory CD4 T cells induce chronic graft versus host disease. 48th Annual Meeting of the American-Society-of-Hematology Dutt, S., Tseng, D., George, T. I., Ermann, J., Liu, Y., Fathman, C. G., Strober, S. AMER SOC HEMATOLOGY. 2006: 137A–137A
  • Aberrant regulation of Wnt/beta-catenin pathway mediators in chronic myelogenous leukemia stem cells 48th Annual Meeting of the American-Society-of-Hematology Abrahamsson, A., Geron, I., Gotlib, J., Durocher, J., Creusot, R., Kavalerchik, E., Goff, D., Fathman, C. G., Lilleberg, S. L., Giles, F., Weissman, I., Jamieson, C. AMER SOC HEMATOLOGY. 2006: 605A–605A
  • Does our current understanding of the molecular basis of immune tolerance predict new therapies for autoimmune disease? NATURE CLINICAL PRACTICE RHEUMATOLOGY Tarner, I. H., Fathman, C. G. 2006; 2 (9): 491-499

    Abstract

    The creation of specific immune tolerance has often been referred to as the ultimate goal of immunotherapy, because it would allow autoimmune disease to be reversed without the need for nonspecific and potentially harmful immunosuppressive therapy. Studies performed during the past decade have been immensely fruitful in terms of advances in our understanding of the cellular and molecular mechanisms of immune tolerance, and have paved the way for successful exploitation of these mechanisms for therapeutic purposes. Important developments include an increased understanding of central and peripheral tolerance, and treatment strategies that mimic the mechanisms behind deletion of self-reactive cells, the identification of crucial gene products that are involved in the induction of anergy, and the characterization of regulatory T cells and protocols for their induction and expansion for therapeutic applications. These landmarks of immune-tolerance research are summarized and their potential use in the immunotherapy of autoimmune disease discussed.

    View details for DOI 10.1038/ncprheum0272

    View details for Web of Science ID 000240200400010

    View details for PubMedID 16951704

  • T cell anergy: Where it's LAT (vol 24, pg 501, 2006) IMMUNITY Lineberry, N., Fathman, C. 2006; 25 (1): 175
  • T cell anergy: Where it's LAT IMMUNITY Lineberry, N., Fathman, C. G. 2006; 24 (5): 501-503

    Abstract

    T cell receptor engagement activates selective signaling pathways in T lymphocytes under different conditions. In this issue of Immunity, demonstrate that anergic T cells are selectively defective in LAT activation.

    View details for DOI 10.1016/j.immuni.2006.05.002

    View details for Web of Science ID 000237970100004

    View details for PubMedID 16713965

  • CYLD: deubiquitination-induced TCR signaling NATURE IMMUNOLOGY Lineberry, N., Fathman, C. G. 2006; 7 (4): 369-370

    View details for DOI 10.1038/ni0406-369

    View details for Web of Science ID 000236171000009

    View details for PubMedID 16550202

  • Developing the concept of adoptive cellular gene therapy of rheumatoid arthritis CIS Spring School on Systemic Autoimmune Diseases Tarner, I. H., Neumann, E., GAY, S., Fathman, C. G., Mulller-Ladner, U. ELSEVIER SCIENCE BV. 2006: 148–52

    Abstract

    Progressive destruction of articular cartilage and bone is the pivotal problem of rheumatoid arthritis (RA). Joint destruction is the cause of severe disability and determines the long-term outcome of disease. Conventional therapy does not control this destructive process sufficiently and the anti-rheumatic drugs available today can cause severe systemic adverse effects. Local application of chondroprotective and osteoprotective agents by means of gene therapy would be an attractive alternative to conventional therapy of RA and could provide long-term expression of the therapeutic agents and minimize systemic adverse effects. For this purpose, we have developed the concept of adoptive cellular gene therapy. This treatment strategy is based on using genetically engineered cells that home specifically to sites of autoimmune inflammation and thus allow local delivery of therapeutic gene products. Ex vivo transduction of these cells avoids systemic exposure of the host to the transgene-encoding vector and thus adds to the safety of this approach. In this article of the CIS Spring School in Autoimmune Diseases 2005 proceedings, we review our work on developing the strategy of adoptive cellular gene therapy and summarize recent advances in the evaluation of therapeutic effects and the identification of novel therapeutic targets.

    View details for DOI 10.1016/j.autrev.2005.09.009

    View details for Web of Science ID 000235349400018

    View details for PubMedID 16431349

  • CD4(+)CD25(+) regulatory T cells and their therapeutic potential ANNUAL REVIEW OF MEDICINE Randolph, D. A., Fathman, C. G. 2006; 57: 381-402

    Abstract

    The human immune system mounts specific responses to a vast array of antigens. Although this is clearly beneficial in fighting off harmful infections and cancerous cells, the system must be carefully controlled to ensure that normal self-antigens are not targeted. A recently characterized subset of T cells, identified by their cell surface expression of CD4 and CD25, is critical in regulating the function of other immune cells and preventing potentially harmful autoimmune responses. This article reviews what is currently known about these so-called regulatory T cells and discusses the therapeutic potential of these cells to modulate human immune-based diseases.

    View details for DOI 10.1146/annurev.med.57.121304.131337

    View details for Web of Science ID 000235981900024

    View details for PubMedID 16409156

  • Immature dendritic cells as vehicles for application in adoptive cellular gene therapy. Purath, U., Neumann, E., Creusot, R., Yaghoubi, S., Haerle, P., Straub, R., Fathman, C., Mueller-Ladner, U., Tarner, I. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2006: S34
  • GRAIL, an E3 ligase, is differentially regulated by TCR-dependent and CD28-dependent IL-2 signals. 6th Annual Meeting of the Federation-of-Clinical-Immunology-Societies Wu, L., Fathman, C. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2006: S80–S80
  • L-selectin and beta(7) integrin on donor CD4 T cells are required for the early migration to host mesenteric lymph nodes and acute colitis of graft-versus-host disease BLOOD Dutt, S., Ermann, J., Tseng, D., Liu, Y. P., George, T. I., Fathman, C. G., Strober, S. 2005; 106 (12): 4009-4015

    Abstract

    The homing receptors L-selectin and alpha4beta7 integrin facilitate entry of T cells into the gut-associated organized lymphoid tissues such as the mesenteric lymph nodes and Peyer patches. We studied the impact of inactivation of genes encoding these receptors on the ability of purified donor CD4+ T cells to induce acute lethal graft-versus-host disease (GVHD) associated with severe colitis in irradiated major histocompatibility complex (MHC)-mismatched mice. Whereas lack of expression of a single receptor had no significant impact on the severity of colitis and GVHD, the lack of expression of both receptors markedly ameliorated colitis and early deaths observed with wild-type (WT) T cells. The changes in colitis and GVHD were reflected in a marked reduction in the early accumulation of donor T cells in the mesenteric lymph nodes and subsequently in the colon. The purified WT donor CD4+ T cells did not accumulate early in the Peyer patches and failed to induce acute injury to the small intestine. In conclusion, the combination of CD62L and beta7 integrin is required to induce acute colitis and facilitate entry of CD4+ donor T cells in the mesenteric nodes associated with lethal GVHD in allogeneic hosts.

    View details for DOI 10.1182/blood-2005-06-2339

    View details for PubMedID 16105972

  • Memory CD4 T cells induce graft versus host disease. 47th Annual Meeting of the American-Society-of-Hematology Dutt, S., Tseng, D., Ermann, J., Liu, Y. P., George, T. I., Fathman, C. G., Strober, S. AMER SOC HEMATOLOGY. 2005: 380A–380A
  • Bioluminescent imaging of human leukemic stem cell engraftment. 47th Annual Meeting of the American-Society-of-Hematology Jamieson, C., Karimi, M., Creusot, R., Negrin, R., Gotlib, J., Chao, M., Jones, C., Keating, A., Fathman, C. G., Zehnder, J., Weissman, I. L. AMER SOC HEMATOLOGY. 2005: 205A–205A
  • Targeted gene therapy of autoimmune diseases: advances and prospects. Expert review of clinical immunology Creusot, R. J., Fathman, C. G., Müller-Ladner, U., Tarner, I. H. 2005; 1 (3): 385-404

    Abstract

    Idealized gene therapy of autoimmune diseases would mean getting the right drug to the right place at the right time to affect the right mechanism of action. In other words, a specific gene therapy strategy needs to have functional, spatial and temporal specificity. Functional specificity implies targeting the cellular, molecular and/or genetic mechanisms relevant to the disease, without affecting nondiseased organs or tissues through mechanisms that cause adverse effects. Spatial specificity means the delivery of the therapeutic agent exclusively to sites and cells that are relevant to the disease. Temporal specificity is, in principle, synonymous with controlled on-demand expression of the therapeutic gene and thus represents a major safety feature. This article reviews recent advances in strategies to use gene therapy in the treatment of autoimmune diseases.

    View details for DOI 10.1586/1744666X.1.3.385

    View details for PubMedID 20476990

  • Molecular imaging using labeled donor tissues reveals patterns of engraftment, rejection, and survival in transplantation TRANSPLANTATION Cao, Y. A., Bachmann, M. H., Beilhack, A., Yang, Y., Tanaka, M., Swijnenburg, R. J., Reeves, R., Taylor-Edwards, C., Schulz, S., Doyle, T. C., Fathman, C. G., Robbins, R. C., Herzenberg, L. A., Negrin, R. S., Contag, C. H. 2005; 80 (1): 134-139

    Abstract

    Tissue regeneration and transplantation of solid organs involve complex processes that can only be studied in the context of the living organism, and methods of analyzing these processes in vivo are essential for development of effective transplantation and regeneration procedures. We utilized in vivo bioluminescence imaging (BLI) to noninvasively visualize engraftment, survival, and rejection of transplanted tissues from a transgenic donor mouse that constitutively expresses luciferase. Dynamic early events of hematopoietic reconstitution were accessible and engraftment from as few as 200 transplanted whole bone marrow (BM) cells resulted in bioluminescent foci in lethally irradiated, syngeneic recipients. The transplantation of autologous pancreatic Langerhans islets and of allogeneic heart revealed the tempo of transplant degeneration or immune rejection over time. This imaging approach is sensitive and reproducible, permits study of the dynamic range of the entire process of transplantation, and will greatly enhance studies across various disciplines involving transplantation.

    View details for DOI 10.1097/01.TP.0000164347.50559.A3

    View details for Web of Science ID 000230473800023

    View details for PubMedID 16003245

  • An array of possibilities for the study of autoimmunity NATURE Fathman, C. G., Soares, L., Chan, S. M., Utz, P. J. 2005; 435 (7042): 605-611

    Abstract

    Since the completion of the sequencing of the human genome, scientific focus has shifted from studying genes to analysing the much larger number of proteins encoded by them. Several proteins can be generated from a single gene depending on how the genetic information is read (transcribed) and how the resultant protein is modified following translation (post-translational modification). Genomic and proteomic technologies are already providing useful information about autoimmune disease, and they are likely to lead to important discoveries within the next decade.

    View details for DOI 10.1038/nature03726

    View details for Web of Science ID 000229476200038

    View details for PubMedID 15931213

  • Only the CD62L(+) subpopulation of CD4(+)CD25(+) regulatory T cells protects from lethal acute GVHD BLOOD Ermann, J., Hoffmann, P., Edinger, M., Dutt, S., Blankenberg, F. G., Higgins, J. P., Negrin, R. S., Fathman, C. G., Strober, S. 2005; 105 (5): 2220-2226

    Abstract

    CD4+CD25+ regulatory T (Treg) cells are potent modulators of alloimmune responses. In murine models of allogeneic bone marrow transplantation, adoptive transfer of donor CD4+CD25+ Treg cells protects recipient mice from lethal acute graft-versus-host disease (aGVHD) induced by donor CD4+CD25- T cells. Here we examined the differential effect of CD62L+ and CD62L- subsets of CD4+CD25+ Treg cells on aGVHD-related mortality. Both subpopulations showed the characteristic features of CD4+CD25+ Treg cells in vitro and did not induce aGVHD in vivo. However, in cotransfer with donor CD4+CD25- T cells, only the CD62L+ subset of CD4+CD25+ Treg cells prevented severe tissue damage to the colon and protected recipients from lethal aGVHD. Early after transplantation, a higher number of donor-type Treg cells accumulated in host mesenteric lymph node (LN) and spleen when CD4+CD25+CD62L+ Treg cells were transferred compared with the CD62L- subset. Subsequently, CD4+CD25+CD62L+ Treg cells showed a significantly higher capacity than their CD62L- counterpart to inhibit the expansion of donor CD4+CD25- T cells. The ability of Treg cells to efficiently enter the priming sites of pathogenic allo-reactive T cells appears to be a prerequisite for their protective function in aGVHD.

    View details for DOI 10.1182/blood-2004-05-2044

    View details for PubMedID 15546950

  • Bioluminescent tracking of candidate leukemic stem cell engraftment in immunocompromised mice Joint Meeting of the American-Society-for-Blood-and-Marrow-Transplantation/Center-for-International-Blood-and-Marrow-Transplant-Research Jamieson, C. H., Karimi, M., Creusot, R., Fathman, C. G., Negrin, R., Weissman, I. L. ELSEVIER SCIENCE INC. 2005: 86–86
  • Protein microarrays for multiplex analysis of signal transduction pathways Nature Medicine Chan, Steven M., Ermann, Joerg, Su, Leon, Fathman, C. Garrison, Utz, Paul J. 2005; 10: 1390-1396
  • Studies on signal transduction pathways downstream of CD28/IL-2 that regulate the E3 ligase, GRAIL. 5th Annual Meeting of the Federation-of-Clinical-Immunology-Societies Wu, L. X., Soares, L., Fathman, C. G. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2005: S211–S211
  • Fostering interdisciplinary immunology Clinical Immunology Huston, David P., Utz Paul J., Fathman C. Garrison 2005; 115: 1-2
  • Protein microarrays for multiplex analysis of signal transduction pathways NATURE MEDICINE Chan, S. M., Ermann, J., Su, L., Fathman, C. G., Utz, P. J. 2004; 10 (12): 1390-1396

    Abstract

    We have developed a multiplexed reverse phase protein (RPP) microarray platform for simultaneous monitoring of site-specific phosphorylation of numerous signaling proteins using nanogram amounts of lysates derived from stimulated living cells. We first show the application of RPP microarrays to the study of signaling kinetics and pathway delineation in Jurkat T lymphocytes. RPP microarrays were used to profile the phosphorylation state of 62 signaling components in Jurkat T cells stimulated through their membrane CD3 and CD28 receptors, identifying a previously unrecognized link between CD3 crosslinking and dephosphorylation of Raf-1 at Ser259. Finally, the potential of this technology to analyze rare primary cell populations is shown in a study of differential STAT protein phosphorylation in interleukin (IL)-2-stimulated CD4(+)CD25(+) regulatory T cells. RPP microarrays, prepared using simple procedures and standard microarray equipment, represent a powerful new tool for the study of signal transduction in both health and disease.

    View details for DOI 10.1038/nm1139

    View details for Web of Science ID 000225500900035

    View details for PubMedID 15558056

  • Murine CD4(+) CD25(+) regulatory T cells fail to undergo chromatin remodeling across the proximal promoter region of the IL-2 gene JOURNAL OF IMMUNOLOGY Su, L., Creusot, R. J., Gallo, E. M., Chan, S. M., Utz, P. J., Fathman, C. G., Ermann, J. 2004; 173 (8): 4994-5001

    Abstract

    CD4+CD25+ regulatory T cells (Treg) acquire unique immunosuppressive properties while maintaining an anergy phenotype when activated in vitro under conditions that induce IL-2 production and proliferation in conventional CD4+ T cells. We investigated the mechanism underlying one component of this naturally anergic phenotype, the inability of the Treg cells to produce IL-2 following activation. Analysis of freshly isolated murine CD4+CD25+ Treg and conventional CD4+CD25- T cells following PMA/ionomycin stimulation demonstrated no differences in inducible AP-1 formation, an important transcriptional complex in regulating IL-2 gene expression. Although p38 MAPK and ERK1/2 protein kinases were phosphorylated with similar kinetics, we observed diminished activation of JNK in the CD4+CD25+ Treg cells. However, lentiviral-mediated reconstitution of the JNK pathway using a constitutively active construct did not overcome the block in IL-2 synthesis. Using a PCR-based chromatin accessibility assay we found that the minimal IL-2 promoter region of CD4+CD25+ Treg cells, unlike conventional CD4 T cells, did not undergo chromatin remodeling following stimulation, suggesting that the inability of CD4+CD25+ Treg cells to secrete IL-2 following activation is controlled at the chromatin level.

    View details for Web of Science ID 000224392200028

    View details for PubMedID 15470042

  • Gene therapy for type 1 diabetes: a novel approach for targeted treatment of autoimmunity JOURNAL OF CLINICAL INVESTIGATION Creusot, R. J., Fathman, C. G. 2004; 114 (7): 892-894

    Abstract

    It has been difficult to develop therapies that target those T cells initiating and mediating the pathogenesis of autoimmune disease. Indeed, most current treatments indiscriminately affect both the autoreactive T cells and the "good" T cells, putting the patient at risk of compromised immune function. A new approach raises the possibility of targeted therapy for autoimmunity. Transplantation of hematopoietic stem cells modified to express a protective form of MHC class II corrects a defect in central tolerance. This method contrasts with other targeted therapies that attempt to modify peripheral tolerance, which is also defective in type 1 diabetes mellitus.

    View details for DOI 10.1172/JC1200423168

    View details for Web of Science ID 000224223800008

    View details for PubMedID 15467826

    View details for PubMedCentralID PMC518674

  • Essential role of the E3 ubiquitin ligase Cbl-b in T cell anergy induction IMMUNITY Jeon, M. S., Atfield, A., Venuprasad, K., Krawczyk, C., Sarao, R., Elly, C., Yang, C., Arya, S., Bachmaier, K., Su, L., Bouchard, D., Jones, R., Gronski, M., Ohashi, P., Wada, T., Bloom, D., Fathman, C. G., Liu, Y. C., Penninger, J. M. 2004; 21 (2): 167-177

    Abstract

    Antigen-specific immunotolerance limits the expansion of self-reactive T cells involved in autoimmune diseases. Here, we show that the E3 ubiquitin ligase Cbl-b is upregulated in T cells after tolerizing signals. Loss of Cbl-b in mice results in impaired induction of T cell tolerance both in vitro and in vivo. Importantly, rechallenge of Cbl-b mutant mice with the tolerizing antigen results in massive lethality. Moreover, ablation of Cbl-b resulted in exacerbated autoimmunity. Mechanistically, loss of Cbl-b rescues reduced calcium mobilization of anergic T cells, which was attributed to Cbl-b-mediated regulation of PLCgamma-1 phosphorylation. Our results show a critical role for Cbl-b in the regulation of peripheral tolerance and anergy of T cells.

    View details for Web of Science ID 000223441900006

    View details for PubMedID 15308098

  • The gene related to anergy in lymphocytes, an E3 ubiquitin ligase, is necessary for anergy induction in CD4 T cells JOURNAL OF IMMUNOLOGY Seroogy, C. M., Soares, L., Ranheim, E. A., Su, L., Holness, C., Bloom, D., Fathman, C. G. 2004; 173 (1): 79-85

    Abstract

    Acquisition of the anergy phenotype in T cells is blocked by inhibitors of protein synthesis and calcineurin activity, suggesting that anergic T cells may have a unique genetic program. Retroviral transduction of hemopoietic stem cells from TCR transgenic mice and subsequent reconstitution of syngeneic mice to express the E3 ubiquitin ligase, gene related to anergy in lymphocytes (GRAIL), or an enzymatically inactive form, H2N2 GRAIL, allowed analysis of the role of GRAIL in T cell anergy in vivo. Constitutive expression of GRAIL was sufficient to render naive CD4 T cells anergic, however, when the enzymatically inactive form H2N2 GRAIL was expressed, it functioned as a dominant negative of endogenous GRAIL and blocked the development of anergy. These data provide direct evidence that a biochemical pathway composed of GRAIL and/or GRAIL-interacting proteins is important in the development of the CD4 T cell anergic phenotype in vivo.

    View details for Web of Science ID 000222170900014

    View details for PubMedID 15210761

  • Targeted gene therapy: frontiers in the development of 'smart drugs' TRENDS IN BIOTECHNOLOGY Tarner, I. H., Muller-Ladner, U., Fathman, C. G. 2004; 22 (6): 304-310

    Abstract

    Chronic diseases, particularly malignancies and immune-mediated inflammatory diseases (IMIDs), are a challenging frontier for clinical diagnosis and treatment, as well as for biomedical research. Current treatment regimens are frequently insufficient and thus new treatment strategies are needed. Novel therapies for disabling such diseases should provide improvements with respect to safety, efficacy and cost. To fulfill these three key criteria, recent research efforts have focused on the development of 'smart drugs'. This review highlights some examples of the rapidly expanding possibilities that current biotechnology has to offer in the development of novel therapeutic strategies for complex diseases such as IMIDs. Special attention is given to advances in, and limitations of, controlled and targeted gene product application in inflammatory diseases.

    View details for Web of Science ID 000222301000011

    View details for PubMedID 15158060

  • Two isoforms of otubain 1 regulate T cell anergy via GRAIL NATURE IMMUNOLOGY Soares, L., Seroogy, C., Skrenta, H., Anandasabapathy, N., Lovelace, P., Chung, C. D., Engleman, E., Fathman, C. G. 2004; 5 (1): 45-54

    Abstract

    The active ubiquitin E3 ligase GRAIL is crucial in the induction of CD4 T cell anergy. Here we show that GRAIL is associated with and regulated by two isoforms of the ubiquitin-specific protease otubain 1. In lethally irradiated mice reconstituted with bone marrow cells from T cell receptor-transgenic mice retrovirally transduced to express the genes encoding these proteases, otubain 1-expressing cells contained negligible amounts of endogenous GRAIL, proliferated well and produced large amounts of interleukin 2 after antigenic stimulation. In contrast, cells expressing the alternatively spliced isoform, otubain 1 alternative reading frame 1, contained large amounts of endogenous GRAIL and were functionally anergic, and they proliferated poorly and produced undetectable interleukin 2 when stimulated in a similar way. Thus, these two proteins have opposing epistatic functions in controlling the stability of GRAIL expression and the resultant anergy phenotype in T cells.

    View details for DOI 10.1038/ni1017

    View details for Web of Science ID 000187635800013

    View details for PubMedID 14661020

  • Scratching the (T cell) surface GENOME BIOLOGY Ermann, J., Chung, C. D., Fathman, C. G. 2004; 5 (1)
  • Costimulatory signals controlling regulatory T cells PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Ermann, J., Fathman, C. G. 2003; 100 (26): 15292-15293

    View details for DOI 10.1073/pnas.0307001100

    View details for Web of Science ID 000187554600003

    View details for PubMedID 14676329

    View details for PubMedCentralID PMC307559

  • Survival and homing of ex vivo expanded donor derived dendritic cells after allogeneic BMT. 45th Annual Meeting and Exhibition of the American-Society-of-Hematology Schimmelpfennig, C. H., Schulz, S., Arber, C., Baker, J., Tarner, I. H., Mcbride, J. M., Fathman, C. G., Contag, C. H., Negrin, R. S. AMER SOC HEMATOLOGY. 2003: 695A–695A
  • Only the CD62L+ subpopulation of CD4+CD25+ regulatory T cells protects against lethal acute graft versus host disease. Dutt, S., Ermann, J., Hoffmann, P., Edinger, M., Negrin, R. S., Fathman, C. G., Strober, S. AMER SOC HEMATOLOGY. 2003: 950A
  • CD4(+)CD25(+) regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation NATURE MEDICINE Edinger, M., Hoffmann, P., Ermann, J., Drago, K., Fathman, C. G., Strober, S., Negrin, R. S. 2003; 9 (9): 1144-1150

    Abstract

    Mature donor T cells cause graft-versus-host disease (GVHD), but they are also the main mediators of the beneficial graft-versus-tumor (GVT) activity of allogeneic bone marrow transplantation. Suppression of GVHD with maintenance of GVT activity is a desirable outcome for clinical transplantation. We have previously shown that donor-derived CD4+CD25+ regulatory T cells inhibit lethal GVHD after allogeneic bone marrow transplantation across major histocompatibility complex (MHC) class I and II barriers in mice. Here we demonstrate that in host mice with leukemia and lymphoma, CD4+CD25+ regulatory T cells suppress the early expansion of alloreactive donor T cells, their interleukin-2-receptor (IL-2R) alpha-chain expression and their capacity to induce GVHD without abrogating their GVT effector function, mediated primarily by the perforin lysis pathway. Thus, CD4+CD25+ T cells are potent regulatory cells that can separate GVHD from GVT activity mediated by conventional donor T cells.

    View details for DOI 10.1038/nm915

    View details for PubMedID 12925844

  • Localized expression of an anti-TNF single-chain antibody prevents development of collagen-induced arthritis GENE THERAPY Smith, R., Tarner, I. H., Hollenhorst, M., Lin, C., Levicnik, A. U., Fathman, C. G., Nolan, G. P. 2003; 10 (15): 1248-1257

    Abstract

    Although systemic administration of neutralizing anti-TNF antibodies has been used successfully in treating rheumatoid arthritis, there is a potential for side effects. We transduced a collagen reactive T-cell hybridoma with tissue-specific homing properties to assess therapeutic effects of local delivery to inflamed joints of anti-TNF single-chain antibodies (scFv) by adoptive cellular gene therapy. Cell culture medium conditioned with 1 x 10(6) scFv producer cells/ml had TNF neutralizing capacity in vitro equivalent to 50 ng/ml anti-TNF monoclonal antibody. Adding a kappa chain constant domain to the basic scFv (construct TN3-Ckappa) gave increased in vitro stability and in vivo therapeutic effect. TN3-Ckappa blocked development of collagen-induced arthritis in DBA/1LacJ mice for >60 days. Transgene expression was detected in the paws but not the spleen of treated animals for up to 55 days postinjection. No significant variations in cell proliferation or cytokine secretion were found in splenocytes or peripheral lymphocytes. IL-6 expression was blocked in the diseased paws of mice in the scFv treatment groups compared to controls. In conclusion, we have shown that local expression of an anti-inflammatory agent blocks disease development without causing demonstrable systemic immune function changes. This is encouraging for the potential development of safe adoptive cellular therapies to treat autoimmunity.

    View details for DOI 10.1038/sj.gt.3301980

    View details for Web of Science ID 000184207000007

    View details for PubMedID 12858190

  • The role of GRAIL, an e3 ligase, in CD4+T cells and anergy 90th Annual Meeting of the American-Association-for-Immunologists Seroogy, C. M., Ranheim, E. A., Fathman, C. G., Soares, L. FEDERATION AMER SOC EXP BIOL. 2003: C212–C212
  • GRAIL: An E3 ubiquitin ligase that inhibits cytokine gene transcription is expressed in anergic CD4(+) T cells IMMUNITY Anandasabapathy, N., Ford, G. S., Bloom, D., Holness, C., Paragas, V., Seroogy, C., Skrenta, H., Hollenhorst, M., Fathman, C. G., Soares, L. 2003; 18 (4): 535-547

    Abstract

    T cell anergy may serve to limit autoreactive T cell responses. We examined early changes in gene expression after antigen-TCR signaling in the presence (activation) or absence (anergy) of B7 costimulation. Induced expression of GRAIL (gene related to anergy in lymphocytes) was observed in anergic CD4(+) T cells. GRAIL is a type I transmembrane protein that localizes to the endocytic pathway and bears homology to RING zinc-finger proteins. Ubiquitination studies in vitro support GRAIL function as an E3 ubiquitin ligase. Expression of GRAIL in retrovirally transduced T cell hybridomas dramatically limits activation-induced IL-2 and IL-4 production. Additional studies suggest that GRAIL E3 ubiquitin ligase activity and intact endocytic trafficking are critical for cytokine transcriptional regulation. Expression of GRAIL after an anergizing stimulus may result in ubiquitin-mediated regulation of proteins essential for mitogenic cytokine expression, thus positioning GRAIL as a key player in the induction of the anergic phenotype.

    View details for Web of Science ID 000182353900009

    View details for PubMedID 12705856

  • Treatment of autoimmune disease by adoptive cellular gene therapy 10th International Conference on Myasthenia Gravis and Related Disorders Tarner, I. H., Slavin, A. J., McBride, J., Levicnik, A., Smith, R., Nolan, G. P., Contag, C. H., Fathman, C. G. NEW YORK ACAD SCIENCES. 2003: 512–519

    Abstract

    Autoimmune disorders represent inappropriate immune responses directed at self-tissue. Antigen-specific CD4+ T cells and antigen-presenting dendritic cells (DCs) are important mediators in the pathogenesis of auto-immune disease and thus are ideal candidates for adoptive cellular gene therapy, an ex vivo approach to therapeutic gene transfer. Using retrovirally transduced cells and luciferase bioluminescence, we have demonstrated that primary T cells, T cell hybridomas, and DCs rapidly and preferentially home to the sites of inflammation in animal models of multiple sclerosis, arthritis, and diabetes. These cells, transduced with retroviral vectors to drive expression of various "regulatory proteins" such as IL-4, IL-10, IL-12p40, and anti-TNF scFv, deliver these immunoregulatory proteins to the inflamed lesions, providing therapy for experimental autoimmune encephalitis (EAE), collagen-induced arthritis (CIA), and nonobese diabetic mice (NOD).

    View details for Web of Science ID 000186107400066

    View details for PubMedID 14592922

  • T-cell anergy - From phenotype to genotype and back IMMUNOLOGIC RESEARCH Seroogy, C. M., Fathman, C. G. 2003; 28 (3): 255-264

    Abstract

    For many decades, anergy has been used as a descriptive term to describe a state of antigen-specific unresponsiveness. A better understanding of this phenotype was revealed in the 1980s using in vitro model systems. These model systems demonstrated that protein synthesis and mobilization of Ca2+ was required leading to the pursuit of a novel gene(s) that would be unique to the anergy phenotype. Several putative "anergy factors" have been suggested. In this review, we provide an overview of the anergy phenotype and proposed anergy-related genes. To date, no single gene has been described that would completely fulfill the criteria of an "anergy factor." We review work from our laboratory describing a novel gene that we have termed Gene Related to Anergy In Lymphocytes (GRAIL) that is upregulated in T cells anergized in vitro and in vivo and, following transduction into T cells, reiterates the anergy phenotype.

    View details for Web of Science ID 000187357900007

    View details for PubMedID 14713718

  • Scratching the (T cell) surface. Genome biology Ermann, J., Chung, C. D., Fathman, C. G. 2003; 5 (1): 202-?

    Abstract

    Using a genome-scale approach to study transcription levels in a human CD8+ T-cell clone, a recent study has suggested that the repertoire of molecules on the surface of T cells is close to being completely characterized.

    View details for PubMedID 14709166

  • Short polymers of arginine rapidly translocate into vascular cells - Effects on nitric oxide synthesis CIRCULATION JOURNAL Uemura, S., Rothbard, J. B., Matsushita, H., Tsao, P. S., Fathman, C. G., Cooke, J. P. 2002; 66 (12): 1155-1160

    Abstract

    The present study was designed to determine the efficiency of translocation of short polymers of arginine into vascular smooth muscle cells (VSMC) and to determine their effect on nitric oxide (NO) synthesis. Immunostaining revealed that heptamers of L-arginine (R7) rapidly translocated into the VSMC. This rapid transport was not observed with shorter polymers of L-arginine (R5) nor heptamers of lysine (K7). Translocation of R7 was not inhibited by the addition of free L-arginine into the media. When cells were transiently pretreated with R7 or a nonamer of arginine (R9), NO(2) production from cytokine stimulated VSMC was significantly increased, whereas incubation with R5 and K7 had no effect. Short polymers of arginine not only have a unique ability of rapid VSMC translocation but once internalized enhance NO production. Heptamers (or larger polypeptides) of arginine may be useful in therapy to enhance NO production in the vascular system.

    View details for Web of Science ID 000179496000015

    View details for PubMedID 12499624

  • Retroviral gene therapy of collagen-induced arthritis by local delivery of IL-4 CLINICAL IMMUNOLOGY Tarner, I. H., Nakajima, A., Seroogy, C. M., Ermann, J., Levicnik, A., Contag, C. H., Fathman, C. G. 2002; 105 (3): 304-314

    Abstract

    Rheumatoid arthritis (RA) is an autoimmune arthritis, for which treatment options remain limited. This study investigated the potential role of adoptive cellular gene therapy as a novel means for treating the RA animal model collagen-induced arthritis (CIA). Adoptive transfer of antigen-specific T-cell hybridomas retrovirally transduced to express IL-4 1 day before booster immunization significantly reduced the number of inflamed joints. Cell transfer after clinical onset of disease had no therapeutic effect. Bioluminescence imaging showed that the hybridomas migrated to the inflamed joints, thus delivering the regulatory protein locally at the site of inflammation. The homing was, at least in part, due to chemotaxis in response to proinflammatory chemokines that are expressed in inflamed joints. There were no significant changes in the cytokine milieu of the draining lymph nodes, nor in the systemic levels of anti-collagen antibodies in treated mice. We conclude that the beneficial clinical effects observed in our model were most likely based on the local action(s) of IL-4 in the inflamed joints and that the local delivery (and effects) of regulatory cytokines, like IL-4, constitutes a novel and effective method of preventing organ-specific autoimmune disease and of minimizing systemic adverse effects.

    View details for DOI 10.1006/clim.2002.5299

    View details for Web of Science ID 000180187600008

    View details for PubMedID 12498812

  • Trafficking of ex vivo expanded donor dendritic cells after allogeneic BMT. 44th Annual Meeting of the American-Society-of-Hematology Schimmelpfennig, C. H., Mcbride, J. M., Tarner, I. H., Baker, J., McGuire, J., Fathman, C. G., Contag, C. H., Negrin, R. S. AMER SOC HEMATOLOGY. 2002: 408A–408A
  • The CD8 alpha(+) dendritic cell is responsible for inducing peripheral self-tolerance to tissue-associated antigens JOURNAL OF EXPERIMENTAL MEDICINE Belz, G. T., Behrens, G. M., Smith, C. M., MILLER, J. F., Jones, C., Lejon, K., Fathman, C. G., Mueller, S. N., Shortman, K., Carbone, F. R., Heath, W. R. 2002; 196 (8): 1099-1104

    Abstract

    We previously described a mechanism for the maintenance of peripheral self-tolerance. This involves the cross-presentation of tissue-associated antigens by a bone marrow-derived cell type that stimulates the proliferation and ultimate deletion of self-reactive CD8 T cells. This process has been referred to as cross-tolerance. Here, we characterize the elusive cell type responsible for inducing cross-tolerance as a CD8alpha(+) dendritic cell (DC). To achieve this aim, transgenic mice were generated expressing yellow fluorescent protein (YFP) linked to CTL epitopes for ovalbumin and glycoprotein B (gB) of herpes simplex virus under the rat insulin promoter (RIP). Although tracking of YFP was inconclusive, the use of a highly sensitive gB-specific hybridoma that produced beta-galactosidase on encounter with antigen, enabled detection of antigen presentation by cells isolated from the pancreatic lymph node. This showed that a CD11c(+)CD8alpha(+) cell was responsible for cross-tolerance, the same DC subset as previously implicated in cross-priming. These data indicate that CD8alpha(+) DCs play a critical role in both tolerance and immunity to cell-associated antigens, providing a potential mechanism by which cytotoxic T lymphocyte can be immunized to viral antigens while maintaining tolerance to self.

    View details for DOI 10.1084/jem.20020861

    View details for Web of Science ID 000178893100011

    View details for PubMedID 12391021

    View details for PubMedCentralID PMC2194045

  • CD4 T-helper cells engineered to produce IL-10 prevent allergen-induced airway hyperreactivity and inflammation JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY Oh, J. W., Seroogy, C. M., Meyer, E. H., Akbari, O., Berry, G., Fathman, C. G., DeKruyff, R. H., Umetsu, D. T. 2002; 110 (3): 460-468

    Abstract

    T(H)2 cells play a critical role in the pathogenesis of asthma, but the precise immunologic mechanisms that inhibit T(H)2 cell function in vivo are not well understood.The purpose of our studies was to determine whether T cells producing IL-10 regulate the development of asthma.We used gene therapy to generate ovalbumin-specific CD4 T-helper cells to express IL-10, and we examined their capacity to regulate allergen-induced airway hyperreactivity.We demonstrated that the CD4 T-helper cells engineered to express IL-10 abolished airway hyperreactivity and airway eosinophilia in BALB/c mice sensitized and challenged with ovalbumin and in SCID mice reconstituted with ovalbumin-specific T(H)2 effector cells. The inhibitory effect of the IL-10-secreting T-helper cells was accompanied by the presence of increased quantities of IL-10 in the bronchoalveolar lavage fluid, was antigen-specific, and was reversed by neutralization of IL-10. Moreover, neutralization of IL-10 by administration of anti-IL-10 mAb in mice sensitized and challenged with ovalbumin seriously exacerbated airway hyperreactivity and airway inflammation.Our results demonstrate that T cells secreting IL-10 in the respiratory mucosa can indeed regulate T(H)2-induced airway hyperreactivity and inflammation, and they strongly suggest that IL-10 plays an important inhibitory role in allergic asthma.

    View details for DOI 10.1067/mai.2002.127512

    View details for Web of Science ID 000177936900018

    View details for PubMedID 12209095

  • The potential for gene therapy in the treatment of autoimmune disease CLINICAL IMMUNOLOGY Tarner, I. H., Fathman, C. G. 2002; 104 (3): 204-216

    View details for DOI 10.1006/clim.2002.5235

    View details for Web of Science ID 000178549100002

    View details for PubMedID 12217329

  • The Subpopulation of CD4(+) CD25(+) splenocytes that delays adoptive transfer of diabetes expresses L-selectin and high levels of CCR7 JOURNAL OF IMMUNOLOGY Szanya, V., Ermann, J., TAYLOR, C., Holness, C., Fathman, C. G. 2002; 169 (5): 2461-2465

    Abstract

    Recently, CD4(+)CD25(+) T cells have been implicated in the control of diabetes, suggesting that the inflamed islets of Langerhans in prediabetic NOD mice are under peripheral immune surveillance. Here we show that CD4(+)CD25(+) splenocytes inhibit diabetes in cotransfer with islet-infiltrating cells. Furthermore, CD62L expression is necessary for this disease-delaying effect of CD4(+)CD25(+) cells in vivo, but not for their suppressor function in vitro. We demonstrate that the CD4(+)CD25(+)CD62L(+) splenocytes express CCR7 at high levels and migrate toward secondary lymphoid tissue chemokine and ELC (macrophage-inflammatory protein-3beta), lymphoid chemokines, whereas CD4(+)CD25(+)CD62L(-) splenocytes preferentially express CCR2, CCR4, and CXCR3 and migrate toward the corresponding inflammatory chemokines. These data demonstrate that CD4(+)CD25(+)CD62L(+), but not CD4(+)CD25(+)CD62L(-), splenocytes delay diabetes transfer, and that CD4(+)CD25(+) suppressor T cells are comprised of at least two subpopulations that behave differently in cotransfer in vivo and express distinct chemokine receptor and chemotactic response profiles despite demonstrating equivalent suppressor functions in vitro.

    View details for Web of Science ID 000177594100030

    View details for PubMedID 12193715

  • Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation JOURNAL OF EXPERIMENTAL MEDICINE Hoffmann, P., Ermann, J., Edinger, M., Fathman, C. G., Strober, S. 2002; 196 (3): 389-399

    Abstract

    Acute graft-versus-host disease (aGVHD) is still a major obstacle in clinical allogeneic bone marrow (BM) transplantation. CD4(+)CD25(+) regulatory T (T(reg)) cells have recently been shown to suppress proliferative responses of CD4(+)CD25(-) T cells to alloantigenic stimulation in vitro and are required for ex vivo tolerization of donor T cells, which results in their reduced potential to induce aGVHD. Here we show that CD4(+)CD25(+) T cells isolated from the spleen or BM of donor C57BL/6 (H-2(b)) mice that have not been tolerized are still potent inhibitors of the alloresponse in vitro and of lethal aGVHD induced by C57BL/6 CD4(+)CD25(-) T cells in irradiated BALB/c (H-2(d)) hosts in vivo. The addition of the CD4(+)CD25(+) T(reg) cells at a 1:1 ratio with responder/inducer CD4(+)CD25(-) T cells resulted in a >90% inhibition of the mixed leukocyte reaction and marked protection from lethal GVHD. This protective effect depended in part on the ability of the transferred CD4(+)CD25(+) T cells to secrete interleukin 10 and occurred if the T(reg) cells were of donor, but not host, origin. Our results demonstrate that the balance of donor-type CD4(+)CD25(+) T(reg) and conventional CD4(+)CD25(-) T cells can determine the outcome of aGVHD.

    View details for DOI 10.1084/jem.20020399

    View details for Web of Science ID 000177658200011

    View details for PubMedID 12163567

    View details for PubMedCentralID PMC2193938

  • Adoptive cellular gene therapy of autoimmune disease. Autoimmunity reviews Slavin, A. J., Tarner, I. H., Nakajima, A., Urbanek-Ruiz, I., McBride, J., Contag, C. H., Fathman, C. G. 2002; 1 (4): 213-219

    Abstract

    Autoimmune disorders represent inappropriate immune responses directed at self-tissue. Because CD4+ T cells are important mediators in the pathogenesis of autoimmune disease, they are ideal candidates for cell-based gene therapy. Using retrovirally-transduced cells and luciferase bioluminescence, we have demonstrated that primary T cells and hybridomas, rapidly and preferentially home to the sites of inflammation in organ-specific autoimmune disease. These cells, transduced with retroviral vectors to drive expression of various 'regulatory proteins', such as IL-4, IL-10 and IL-12p40, deliver these immunoregulatory proteins to the inflamed lesions, providing therapy for experimental models of autoimmune disease such as EAE, CIA and NOD mice. This technique was originally developed in our lab in the murine model of multiple sclerosis, EAE, where T cell hybridomas reactive with myelin basic protein (MBP) were transduced to express and used to deliver the modulatory cytokine, IL-4. Recently we have observed that the cytokine receptor antagonist, IL-12p40 transduced anti-myelin basic protein (MBP) TCR-transgenic T cells (but not CII-reactive T cells) were effective in preventing EAE whereas the CII-reactive, but not MBP-reactive T cells, transduced to express IL-12p40, would treat CIA.

    View details for PubMedID 12848998

  • Immunomodulatory vaccination in autoimmune disease ENDOCRINOLOGY AND METABOLISM CLINICS OF NORTH AMERICA Urbanek-Ruiz, I., Ruiz, P. J., Steinman, L., Fathman, C. G. 2002; 31 (2): 441-?

    Abstract

    The development of vaccines is arguably the most significant achievement in medicine to date. The practice of innoculation with the fluid from a sore to protect from a disease actually dates back to ancient China; however, with the introduction of Jenner's smallpox vaccine, and greater understanding of the immune system, vaccines have become specific and systematic. Traditional vaccines have used killed pathogens (hepatitis A and the Salk polio vaccines), immunogenic subunits of a given pathogen (hepatitis B subunit vaccine), or live attenuated pathogens (measles, mumps, rubella, Sabin polio vaccines) to generate protective immunity. Currently, a new generation of vaccines that use the genetic material of a pathogen to elicit protective immunity are being developed. Although the most widespread and successful use of vaccines today remains in the arena of infectious diseases, manipulations of immune responses to protect against cancers, neurologic diseases, and autoimmunity are being explored rigorously.

    View details for Web of Science ID 000176324600012

    View details for PubMedID 12092460

  • IL-2 and the homeostasis of marine CD4+CD25+ regulatory T cells. Ermann, J., Su, L., Fathman, C. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2002: S68–S68
  • Gene therapy to treat autoimmune diseases Fathman, C. BLACKWELL PUBLISHING INC. 2002: 738
  • A complicated relationship: fulfilling the interactive needs of the T lymphocyte and the dendritic cell. pharmacogenomics journal Mcbride, J. M., Fathman, C. G. 2002; 2 (6): 367-376

    Abstract

    T cells recognize antigenic peptides displayed on the surface of MHC-bearing antigen-presenting cells (APCs), and with sufficient costimulation become activated. However, the ability of an APC (even bearing the correct peptide) to initiate and fulfill the requirements for T cell activation is not easily achieved. Naive T cells use multiple copies of a single receptor to survey the vast array of peptides presented on an APC, and require multiple receptor engagements to initiate T cell activation. Dendritic cells (DCs) are specialized cells with optimal capabilities for priming naive CD4+ T cells. Activation occurs, after initial antigen recognition by T cells, followed by a rapid dialogue between the T cells and the DCs. The resulting changes in both the cytoskeleton and the expression or regulation of cell-surface molecules on both cell types act to further strengthen engagement. In this report, we review the fundamentals of CD4+ T helper cell : DC interactions and discuss recent data concerning the molecular characteristics of this engagement.

    View details for PubMedID 12629502

  • Bioluminescence imaging of lymphocyte trafficking in vivo EXPERIMENTAL HEMATOLOGY Hardy, J., Edinger, M., Bachmann, M. H., Negrin, R. S., Fathman, C. G., Contag, C. H. 2001; 29 (12): 1353-1360

    Abstract

    Lymphocytes are highly mobile cells that travel throughout the body in response to a tremendous variety of stimuli. Revealing lymphocyte trafficking patterns in vivo is necessary for a complete understanding of immune function, as well as cell-cell and cell-tissue interactions in immune development and in response to insult. Although the location of cell populations in various tissues at any given point in time may be revealed by techniques such as flow cytometry and immunofluorescence, these methods are not readily amenable to the assessment of dynamic cell migration patterns in vivo. In the past 5 years, technologies for imaging molecular and cellular changes in living animals have advanced to a point where it is possible to reveal the migratory paths of these vitally important cells. Here, we review one advancement in cellular imaging, in vivo bioluminescence imaging, which addresses the problem of lymphocyte tracking. This imaging strategy has the potential to elucidate the temporal patterns of immune responses and the spatial distribution of lymphocytes within the body.

    View details for PubMedID 11750093

  • Gene therapy in autoimmune disease CURRENT OPINION IN IMMUNOLOGY Tarner, I. H., Fathman, C. G. 2001; 13 (6): 676-682

    Abstract

    Recent work on gene therapies for autoimmune disease has continued to provide insight into the pathogenesis of autoimmunity. Reliable, effective and targeted gene therapy applications have been achieved by using transduced dendritic cells and antigen-specific T cells as delivery vehicles. Bioluminescence imaging has been implemented to visualize cell trafficking and homing in vivo. As a first step into human gene therapy, a phase I clinical trial for assessing the feasibility and safety of gene transfer has been completed in a group of rheumatoid arthritis patients.

    View details for Web of Science ID 000171797600008

    View details for PubMedID 11677089

  • CD4(+)CD25(+) T cells facilitate the induction of T cell anergy JOURNAL OF IMMUNOLOGY Ermann, J., Szanya, V., Ford, G. S., Paragas, V., Fathman, C. G., Lejon, K. 2001; 167 (8): 4271-4275

    Abstract

    T cell anergy is characterized by the inability of the T cell to produce IL-2 and proliferate. It is reversible by the addition of exogenous IL-2. A similar state of unresponsiveness is observed when the proliferative response of murine CD4(+)CD25(-) T cells is suppressed in vitro by coactivated CD4(+)CD25(+) T cells. We have developed a suppression system that uses beads coated with anti-CD3 and anti-CD28 Abs as surrogate APCs to study the interaction of CD4(+)CD25(+) and CD4(+)CD25(-) T cells in vitro. CD4(+)CD25(+) T cell-induced suppression, in this model, was not abrogated by blocking the B7-CTLA-4 pathway. When the CD4(+)CD25(-) T cells were separated from the CD4(+)CD25(+) suppressor cells after 24 h of coactivation by the Ab-coated beads, the CD4(+)CD25(-) T cells were unable to proliferate or to produce IL-2 upon restimulation. The induction of this anergic phenotype in the CD4(+)CD25(-) T cells correlated with the up-regulated expression of the gene related to anergy in lymphocytes (GRAIL), a novel anergy-related gene that acts as a negative regulator of IL-2 transcription. This system constitutes a novel mechanism of anergy induction in the presence of costimulation.

    View details for Web of Science ID 000171858300018

    View details for PubMedID 11591749

  • Autoimmune diseases: genes, bugs and failed regulation NATURE IMMUNOLOGY Ermann, J., Fathman, C. G. 2001; 2 (9): 759-761

    View details for Web of Science ID 000170781200003

    View details for PubMedID 11526377

  • Adoptive immunotherapy of experimental autoimmune encephalomyelitis via T cell delivery of the IL-12 p40 subunit JOURNAL OF IMMUNOLOGY Costa, G. L., Sandora, M. R., Nakajima, A., Nguyen, E. V., Taylor-Edwards, C., Slavin, A. J., Contag, C. H., Fathman, C. G., Benson, J. M. 2001; 167 (4): 2379-2387

    Abstract

    CD4+ T cells are believed to play a central role in the initiation and perpetuation of autoimmune diseases such as multiple sclerosis. In the murine model for multiple sclerosis, experimental autoimmune encephalomyelitis, pathogenic T cells exhibit a Th1-like phenotype characterized by heightened expression of proinflammatory cytokines. Systemic administration of "regulatory" cytokines, which serve to counter Th1 effects, has been shown to ameliorate autoimmune responses. However, the inherent problems of nonspecific toxicity limit the usefulness of systemic cytokine delivery as a potential therapy. Therefore, we used the site-specific trafficking properties of autoantigen-reactive CD4+ T cells to develop an adoptive immunotherapy protocol that provided local delivery of a Th1 cytokine antagonist, the p40 subunit of IL-12. In vitro analysis demonstrated that IL-12 p40 suppressed IFN-gamma production in developing and effector Th1 populations, indicating its potential to modulate Th1-promoted inflammation. We have previously demonstrated that transduction of myelin basic protein-specific CD4+ T cells with pGC retroviral vectors can result in efficient and stable transgene expression. Therefore, we adoptively transferred myelin basic protein-specific CD4+ T cells transduced to express IL-12 p40 into mice immunized to develop experimental autoimmune encephalomyelitis and demonstrated a significant reduction in clinical disease. In vivo tracking of bioluminescent lymphocytes, transduced to express luciferase, using low-light imaging cameras demonstrated that transduced CD4+ T cells trafficked to the central nervous system, where histological analysis confirmed long-term transgene expression. These studies have demonstrated that retrovirally transduced autoantigen-specific CD4+ T cells inhibited inflammation and promoted immunotherapy of autoimmune disorders.

    View details for Web of Science ID 000170949600070

    View details for PubMedID 11490028

  • Immunization with DNA encoding an immunodominant peptide of insulin prevents diabetes in NOD mice CLINICAL IMMUNOLOGY Urbanek-Ruiz, I., Ruiz, P. J., Paragas, V., Garren, H., Steinman, L., Fathman, C. G. 2001; 100 (2): 164-171

    Abstract

    DNA vaccination is an effective means of protecting experimental animals against infectious pathogens and cancer and has more recently been used to prevent autoimmune disease. Insulin-dependent diabetes mellitus (IDDM) is an autoimmune disease characterized by T-cell-mediated destruction of the insulin-secreting beta cells in the pancreas. The NOD mouse is an animal model of IDDM in which several autoantigens, including insulin, have been identified. In this study we demonstrate that vaccination of NOD mice with DNA encoding an immunodominant peptide of insulin (residues 9-23 of the B chain) protects the animals from developing diabetes. Animals injected intramuscularly with a bacterial plasmid encoding the insulin B chain peptide show significantly lower disease incidence and delayed onset of disease when compared to controls. Protection appears to be mediated by insulin B (9-23)-specific down-regulation of IFN-gamma. Our results confirm that DNA vaccination has a protective effect on autoimmunity, the understanding of which will reveal new insights into the immune system and open doors for novel therapies.

    View details for DOI 10.1006/clim.2001.5055

    View details for Web of Science ID 000170304800006

    View details for PubMedID 11465945

  • Antigen-specific T cell-mediated gene therapy in collagen-induced arthritis JOURNAL OF CLINICAL INVESTIGATION Nakajima, A., Seroogy, C. M., Sandora, M. R., Tarner, I. H., Costa, G. L., Taylor-Edwards, C., Bachmann, M. H., Contag, C. H., Fathman, C. G. 2001; 107 (10): 1293-1301

    Abstract

    Autoantigen-specific T cells have tissue-specific homing properties, suggesting that these cells may be ideal vehicles for the local delivery of immunoregulatory molecules. We tested this hypothesis by using type II collagen-specific (CII-specific) CD4(+) T hybridomas or primary CD4(+) T cells after gene transfer, as vehicles to deliver an immunoregulatory protein for the treatment of collagen-induced arthritis (CIA), a mouse model of rheumatoid arthritis (RA). CII-specific T cells or hybridomas were transduced using retroviral vectors to constitutively express the IL-12 antagonist, IL-12 p40. Transfer of engineered CD4(+) T cells after immunization significantly inhibited the development of CIA, while cells transduced with vector control had no effect. The beneficial effect on CIA of IL-12 p40-transduced T cells required TCR specificity against CII, since transfer of T cells specific for another antigen producing equivalent amounts of IL-12 p40 had no effect. In vivo cell detection using bioluminescent labels and RT-PCR showed that transferred CII-reactive T-cell hybridomas accumulated in inflamed joints in mice with CIA. These results indicate that the local delivery of IL-12 p40 by T cells inhibited CIA by suppressing autoimmune responses at the site of inflammation. Modifying antigen-specific T cells by retroviral transduction for local expression of immunoregulatory proteins thus offers a promising strategy for treating RA.

    View details for Web of Science ID 000168867400014

    View details for PubMedID 11375419

  • Understanding the interaction of genetics and cellular responses in nonobese diabetic mice. Current directions in autoimmunity Ridgway, W. M., Fathman, C. G. 2001; 4: 218-238

    View details for PubMedID 11569404

  • Retroviral gene therapy of collagen-induced arthritis by local delivery of IL-4 Tarner, I. H., Nakajima, A., Seroogy, C. M., Ermann, J., Sandora, M. R., Contag, C. H., Fathman, C. G. BIOMED CENTRAL LTD. 2001
  • T cell receptor (TCR)-mediated repertoire selection and loss of TCR V beta diversity during the initiation of a CD4(+) T cell response in vivo JOURNAL OF EXPERIMENTAL MEDICINE Fasso, M., Anandasabapathy, N., Crawford, F., Kappler, J., Fathman, C. G., Ridgway, W. M. 2000; 192 (12): 1719-1730

    Abstract

    We recently described a novel way to isolate populations of antigen-reactive CD4(+) T cells with a wide range of reactivity to a specific antigen, using immunization with a fixed dose of nominal antigen and FACS((R)) sorting by CD4(high) expression. Phenotypic, FACS((R)), functional, antibody inhibition, and major histocompatibility complex-peptide tetramer analyses, as well as T cell receptor Vbeta sequence analyses, of the antigen-specific CD4(high) T cell populations demonstrated that a diverse sperm whale myoglobin 110-121-reactive CD4(+) T cell repertoire was activated at the beginning (day 3 after immunization) of the immune response. Within 6 d of immunization, lower affinity clones were lost from the responding population, leaving an expanded population of oligoclonal, intermediate affinity (and residual high affinity) T cells. This T cell subset persisted for at least 4 wk after immunization and dominated the secondary immune response. These data provide evidence that CD4(+) T cell repertoire selection occurs early in the immune response in vivo and suggest that persistence and expansion of a population of oligoclonal, intermediate affinity T cells is involved in CD4(+) T cell memory.

    View details for Web of Science ID 000166013100005

    View details for PubMedID 11120769

  • Rapid and efficient vascular transport of arginine polymers inhibits myointimal hyperplasia CIRCULATION Uemura, S., Fathman, C. G., Rothbard, J. B., Cooke, J. P. 2000; 102 (21): 2629-2635

    Abstract

    We recently discovered that short polymers of arginine efficiently translocate across the cytoplasmic membrane independent of the basic amino acid transporter. We evaluated the kinetics and biological effects of heptamers of L-arginine and D-arginine (L-R7 and D-R7, respectively) in vascular cells. We assessed the effects of these peptides on the NO synthesis pathway and vascular cell proliferation.Human umbilical vein endothelial cell and rabbit vascular segments were incubated in medium containing biotin-labeled L-R7 or D-R7. Both polymers rapidly translocated through the vessel wall and into the vascular cells in a dose- and time-dependent fashion. At a dose of 10 micromol/L for 30 minutes, 100% of the endothelial cells showed evidence of cytoplasmic and nuclear localization of the peptides. To evaluate the biological effects of the polymer translocation on myointimal formation, rabbit jugular vein segments were incubated with polymers (10 micromol/L, 30 minutes) or vehicle before arterial interposition grafting. Planimetric measurement 28 days after surgery revealed that L-R7 and D-R7 substantially reduced myointimal formation compared with the control condition (intima/media ratio: control 1. 50.5, L-R7 0.40.2, and D-R7 0.80.2; P:<0.05). Furthermore, basal nitrate and nitrite production from L-R7-treated grafts was significantly higher than that from both control and D-R7-treated veins. Studies in vitro of cultured vascular smooth muscle cells revealed that both polymers also exhibit an NO-independent inhibition of vascular smooth muscle cell proliferation.Short polymers of arginine have the unique ability of vascular cell translocation, and they also have direct biological effects. These attributes are potentially useful in treating myointimal hyperplasia.

    View details for Web of Science ID 000165405800013

    View details for PubMedID 11085967

  • Polyarginine enters cells more efficiently than other polycationic homopolymers JOURNAL OF PEPTIDE RESEARCH Mitchell, D. J., Kim, D. T., Steinman, L., Fathman, C. G., Rothbard, J. B. 2000; 56 (5): 318-325

    Abstract

    Homopolymers or peptides containing a high percentage of cationic amino acids have been shown to have a unique ability to cross the plasma membrane of cells, and consequently have been used to facilitate the uptake of a variety of biopolymers and small molecules. To investigate whether the polycationic character of these molecules, or some other structural feature, was the molecular basis for the effect, the ability of a variety of homopolymers to enter cells was assayed by confocal microscopy and flow cytometry. Polymers of L- or D-arginine containing six or more amino acids entered cells far more effectively than polymers of equal length composed of lysine, ornithine and histidine. Peptides of fewer than six amino acids were ineffective. The length of the arginine side-chain could be varied without significant loss of activity. These data combined with the inability of polymers of citrulline to enter cells demonstrated that the guanidine headgroup of arginine was the critical structural component responsible for the biological activity. Cellular uptake could be inhibited by preincubation of the cells with sodium azide, but not by low temperature (3 degrees C), indicating that the process was energy dependent, but did not involve endocytosis.

    View details for Web of Science ID 000165173900006

    View details for PubMedID 11095185

  • CD4+CD25+T cell suppressor activity does not require antigen presenting cells Ermann, J., Fathman, C. G. FEDERATION AMER SOC EXP BIOL. 2000: A1217
  • Targeting rare populations of murine antigen-specific T cells using retroviral transduction for application of gene therapy in experimental autoimmune encephalomyelitis (EAE) Costa, G. L., Sandora, M. R., Fathman, C. G., Benson, J. M. FEDERATION AMER SOC EXP BIOL. 2000: A1079
  • Real time in vivo imaging of T cell trafficking in autoimmune disease Sandora, M. R., Costa, G. L., Benson, J. M., Lejon, K., Slavin, A. J., Contag, C. H., Fathman, C. G. FEDERATION AMER SOC EXP BIOL. 2000: A1148
  • Identification and functional characterization of a novel gene which is upregulated in anergic T cells Ford, G. S., Ermann, J., Paragas, Bloom, D., Fathman, C. G. FEDERATION AMER SOC EXP BIOL. 2000: A1216
  • Targeting rare populations of murine antigen-specific T lymphocytes by retroviral transduction for potential application in gene therapy for autoimmune disease JOURNAL OF IMMUNOLOGY Costa, G. L., Benson, J. M., Seroogy, C. M., Achacoso, P., Fathman, C. G., Nolan, G. P. 2000; 164 (7): 3581-3590

    Abstract

    CD4+ T cells are important mediators in the pathogenesis of autoimmunity and would therefore provide ideal candidates for lymphocyte-based gene therapy. However, the number of Ag-specific T cells in any single lesion of autoimmunity may be quite low. Successful gene transfer into autoantigen-specific CD4+ T cells would serve as an ideal vehicle for site-targeted gene therapy if it were possible to transduce preferentially the small number of autoantigen-specific T cells. In this study we have demonstrated that retroviral infection of CD4+ lymphocytes from either autoantigen-stimulated TCR transgenic mice, or Ag-activated immunized nontransgenic mice, with a retroviral vector (pGCIRES), resulted in the transduction of only the limited number of Ag-reactive CD4+ T cells. In contrast, polyclonal activation of the same cultures resulted in transduction of non-antigen-specific lymphocytes. Transduction of Ag-reactive CD4+ T cells with pGCIRES retrovirus encoding the regulatory genes IL-4 (IL4) and soluble TNF receptor (STNFR) resulted in stable integration and long-term expression of recombinant gene products. Moreover, expression of the pGCIRES marker protein, GFP, directly correlated with the expression of the upstream regulatory gene. Retroviral transduction of CD4+ T cells targeted specifically Ag-reactive cells and was cell cycle-dependent and evident only during the mitosis phase. These studies suggest that retroviral transduction of autoantigen-specific murine CD4+ T cells, using the pGCIRES retroviral vector, may provide a potential method to target and isolate the low frequency of autoantigen-specific murine CD4+ T cells, and provides a rational approach to gene therapy in animal models of autoimmunity.

    View details for Web of Science ID 000086020700019

    View details for PubMedID 10725713

  • Ligand-independent down-regulation of IFN-gamma receptor 1 following TCR engagement JOURNAL OF IMMUNOLOGY Skrenta, H., Yang, Y., Pestka, S., Fathman, C. G. 2000; 164 (7): 3506-3511

    Abstract

    Activated T lymphocytes modulate the level of many molecules on their cell surface, including cytokine receptors. This regulation of cytokine receptor expression affects the ability of T cells to respond to cytokines and thus influences the outcome of an immune response. The receptor for IFN-gamma, a proinflammatory cytokine, consists of two copies of a ligand binding chain (IFN-gammaR1) as well as two copies of a second chain (IFN-gammaR2) required for signal transduction. The expression of IFN-gammaR2 is down-regulated at the mRNA level on CD4+ T cells when they differentiate into the Th1, but not the Th2, phenotype. This down-regulation has been demonstrated to depend on the ligand, IFN-gamma, which is produced by Th1 but not Th2 T cells. The regulation of the cell-surface expression of IFN-gamma receptors during primary T cell activation has not been reported. Naive and differentiated T lymphocytes express IFN-gammaR1 at the mRNA level and as a cell-surface protein. In this study, we present evidence that cell-surface expression of IFN-gammaR1 is transiently down-regulated on the surface of naive CD4+ T cells shortly after TCR engagement. Furthermore, this down-regulation is not mediated by the ligand, IFN-gamma, but results from TCR engagement and can be inhibited by cyclosporin A.

    View details for Web of Science ID 000086020700010

    View details for PubMedID 10725704

  • Gene therapy for autoimmune disease 1st Annual Forum on Immunologic Science - A View of the Cutting Edge Fathman, C. G., Costa, G. L., Seroogy, C. M. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2000: S39–S43

    Abstract

    Autoantigen-specific CD4(+) T lymphocytes have been implicated in the pathogenesis of autoimmune diseases. Tissue-specific homing properties of autoantigen-specific CD4(+) T cells suggested that these cells might be ideal vehicles for delivery of retroviral-encoded regulatory proteins in a site-specific manner as a therapy for autoimmune diseases. Application of retroviral transduction of autoantigen-reactive CD4(+) T cells in gene therapy of autoimmunity must include systems capable of targeting these rare populations of antigen-activated T cells. Studies discussed below suggest that retroviral transduction of autoantigen-specific murine CD4(+) T cells may provide a method to target and isolate nontransformed autoantigen-specific murine CD4(+) T cells and provide a rational approach to gene therapy in animal models of autoimmunity.

    View details for Web of Science ID 000086408700007

    View details for PubMedID 10729236

  • A novel transcription factor, T-bet, directs Th1 lineage commitment CELL Szabo, S. J., Kim, S. T., Costa, G. L., Zhang, X. K., Fathman, C. G., Glimcher, L. H. 2000; 100 (6): 655-669

    Abstract

    Naive T helper cells differentiate into two subsets, Th1 and Th2, each with distinct functions and cytokine profiles. Here, we report the isolation of T-bet, a Th1-specific T box transcription factor that controls the expression of the hallmark Th1 cytokine, IFNgamma. T-bet expression correlates with IFNgamma expression in Th1 and NK cells. Ectopic expression of T-bet both transactivates the IFNgamma gene and induces endogenous IFNgamma production. Remarkably, retroviral gene transduction of T-bet into polarized Th2 and Tc2 primary T cells redirects them into Th1 and Tc1 cells, respectively, as evidenced by the simultaneous induction of IFNgamma and repression of IL-4 and IL-5. Thus, T-bet initiates Th1 lineage development from naive Thp cells both by activating Th1 genetic programs and by repressing the opposing Th2 programs.

    View details for Web of Science ID 000085983800008

    View details for PubMedID 10761931

  • Identification and characterization of the antigen-specific subpopulation of alloreactive CD4+T cells in vitro and in vivo TRANSPLANTATION Krieger, N. R., Fathman, C. G., Shaw, M. K., Ridgway, W. M. 2000; 69 (4): 605-609

    Abstract

    We report the identification and characterization of the small subpopulation of alloantigen-specific T cells in vitro and in vivo. This subpopulation of T cells was distinguished by up-regulation of cell surface CD4 expression. These CD4high T cells were alloantigen specific in proliferation assays in vitro, and they expressed memory/activation markers, including CD44high and CD69high. Further studies demonstrated that these allospecific CD4high cells were also present (< or = 1% of CD4+ T cells) in vivo in BALB/c (H-2d) recipients of C57BL/6 (H-2b) skin allografts. CD4high T cells isolated from regional draining lymph nodes in these skin graft recipients reacted in a donor-specific fashion to C57BL/6 splenocyte stimulator cells in mixed lymphocyte culture. Adoptive transfer of CD4high, but not CD4normal T cells, just before skin engraftment in CD4 knockout mice, reconstituted rejection. The discovery that a small subpopulation of CD4high lymph node cells contained all of the alloantigen-specific T cells may allow study of tissue-specificity and subsequent alloantigen identification in transplantation.

    View details for Web of Science ID 000085611500023

    View details for PubMedID 10708118

  • Short polymers of arginine inhibit myointimal hyperplasia: Efficient intracellular translocation and activation of nitric oxide synthesis Uemura, S., Fathman, C. G., Rothbard, J. B., Cooke, J. P. ELSEVIER SCIENCE INC. 2000: 548A–549A
  • Application of gene therapy in autoimmune disease. Current directions in autoimmunity Fathman, C. G., Seroogy, C. M. 2000; 2: 189-202

    View details for PubMedID 11791456

  • The application of gene therapy in autoimmune diseases GENE THERAPY Seroogy, C. M., Fathman, C. G. 2000; 7 (1): 9-13

    Abstract

    The application of gene therapy in autoimmune disease represents a novel use of this technology. The goal of gene therapy in the treatment of autoimmune disease is to restore 'immune homeostasis' by countering the pro-inflammatory effects of the CD4+ T cells in the lesions of autoimmunity. This can be accomplished by adoptive therapy with transduced T cells which can specifically home to the site of inflammation and secrete 'regulatory' protein(s) to ameliorate the inflammation or by direct targeting of the retroviral vector to activated T cells in the sites of inflammation. Transduction of autoantigen recognizing CD4+ T cells, to secrete anti-inflammatory products, may become the 'magic bullet' to combat the ravages of autoimmune inflammation and tissue destruction. Gene Therapy (2000) 7, 9-13.

    View details for Web of Science ID 000084878300002

    View details for PubMedID 10680009

  • MHC structure and autoimmune T cell repertoire development CURRENT OPINION IN IMMUNOLOGY Ridgway, W. M., Fathman, C. G. 1999; 11 (6): 638-642

    Abstract

    Recent work has continued to clarify the relationship between MHC structure and thymic selection that leads to peripheral T cell repertoire development in the pathogenesis of autoimmune diseases. Particular attention has been focused on the nonobese diabetic model of autoimmune diabetes, in which a unique MHC class II molecule (I-Ag7) plays a central role. In the past year, reports on the biochemistry of I-Ag7-combined with analysis of the role of I-Ag7 in T cell repertoire selection--support a model of defective thymic selection as the basis of the association between particular MHC molecules and autoimmune diseases. Analogous work has been done on the structure of the human MHC disease-susceptible and -resistant alleles, DQA1*0301 DQB1*0302 and DQA1*0102 DQB1*0602, and their effect on autoimmune repertoire selection. Comparison of these results (in naturally occurring, spontaneous autoimmune human and murine diabetes), with results in a variety of transgenic and knockout models, has produced an integrated view of how avidity considerations in repertoire selection in the thymus could affect predisposition towards autoimmunity.

    View details for Web of Science ID 000083930100008

    View details for PubMedID 10631548

  • Isolation of self antigen-reactive cells from inflamed islets of nonobese diabetic mice using CD4(high) expression as a marker JOURNAL OF IMMUNOLOGY Lejon, K., Fathman, C. G. 1999; 163 (10): 5708-5714

    Abstract

    The low precursor frequency of Ag-reactive CD4+ T cells has been a barrier to the study of CD4+ T cell responses to conventional Ags as well as CD4+ T cell responses to autoantigens recognized during the course of an autoimmune disease. We have recently reported that all "conventional Ag" reactive CD4+ T cells are contained within the subpopulation expressing high levels of the CD4 molecule, termed CD4high. We have identified a CD4high population in the islets of Langerhans of prediabetic nonobese diabetic (NOD) mice that is extremely potent in transferring disease. As few as 500 CD4high islet-infiltrating CD4+ T cells transferred insulin-dependent diabetes mellitus to CD8 reconstituted NOD-SCID mice within 30 days of transfer. In contrast, CD4high T cells isolated from either NOD spleen or salivary glands did not transfer insulin-dependent diabetes mellitus into similar CD8-reconstituted NOD-SCID recipients. These data indicate that the precursor frequency of NOD islet-reactive, pathogenic CD4+ T cells is much higher in the prediabetic NOD pancreas than in these other organs. The islet-infiltrating CD4high T cells displayed selected memory markers, by cell surface analysis, and displayed a Th 1 phenotype by RNase protection assay, but had a marked decrease in IL-4 mRNA determined by quantitative real time PCR when compared with the less pathogenic CD4normal islet-infiltrating T cells. Use of the CD4high marker to select Ag activated T cells represents a tool to isolate and study pathogenic CD4+ T cells from autoimmune lesions in which the Ag has not been previously defined.

    View details for Web of Science ID 000083638400067

    View details for PubMedID 10553102

  • A new look at MHC and autoimmune disease. Science Ridgway, W. M., Fassò, M., Fathman, C. G. 1999; 284 (5415): 749-?

    View details for PubMedID 10336398

  • Suppressive immunization with DNA encoding a self-peptide prevents autoimmune disease: Modulation of T cell costimulation JOURNAL OF IMMUNOLOGY Ruiz, P. J., Garren, H., Ruiz, I. U., Hirschberg, D. L., Nguyen, L. V., Karpuj, M. V., Cooper, M. T., Mitchell, D. J., Fathman, C. G., Steinman, L. 1999; 162 (6): 3336-3341

    Abstract

    Usually we rely on vaccination to promote an immune response to a pathogenic microbe. In this study, we demonstrate a suppressive from of vaccination, with DNA encoding a minigene for residues 139-151 of myelin proteolipid protein (PLP139-151), a pathogenic self-Ag. This suppressive vaccination attenuates a prototypic autoimmune disease, experimental autoimmune encephalomyelitis, which presents clinically with paralysis. Proliferative responses and production of the Th1 cytokines, IL-2 and IFN-gamma, were reduced in T cells responsive to PLP139-151. In the brains of mice that were successfully vaccinated, mRNA for IL-2, IL-15, and IFN-gamma were reduced. A mechanism underlying the reduction in severity and incidence of paralytic autoimmune disease and the reduction in Th1 cytokines involves altered costimulation of T cells; loading of APCs with DNA encoding PLP139-151 reduced the capacity of a T cell line reactive to PLP139-151 to proliferate even in the presence of exogenous CD28 costimulation. DNA immunization with the myelin minigene for PLP-altered expression of B7.1 (CD80), and B7.2 (CD86) on APCs in the spleen. Suppressive immunization against self-Ags encoded by DNA may be exploited to treat autoimmune diseases.

    View details for Web of Science ID 000079105000029

    View details for PubMedID 10092787

  • Local delivery of TNF by retrovirus-transduced T lymphocytes exacerbates experimental autoimmune encephalomyelitis CLINICAL IMMUNOLOGY Dal Canto, R. A., Shaw, M. K., Nolan, G. P., Steinman, L., Fathman, C. G. 1999; 90 (1): 10-14

    Abstract

    Experimental autoimmune encephalomyelitis (EAE) is an inflammatory autoimmune disease of the central nervous system that serves as a model for the human disease multiple sclerosis. Paralysis is "induced" by CD4+ T cells of the Th1 phenotype. Tumor necrosis factor (TNF), a Th1 type cytokine, has been shown to be upregulated in the CNS during the onset of EAE, and systemic manipulations of TNF have had substantial effects on disease progression. However, the precise role of TNF in EAE has been called into question by recent experiments utilizing TNF and lymphotoxin knockout mice. We demonstrate here that the local delivery of TNF by myelin basic protein (MBP)-specific T cells, retrovirally transduced to express TNF, exacerbated MBP-induced disease following adoptive transfer into syngeneic mice.

    View details for Web of Science ID 000079158600002

    View details for PubMedID 9884347

  • Analysis of the role of variation of major histocompatibility complex class II expression on nonobese diabetic (NOD) peripheral T cell response JOURNAL OF EXPERIMENTAL MEDICINE Ridgway, W. M., Ito, H., Fasso, M., Yu, C., Fathman, C. G. 1998; 188 (12): 2267-2275

    Abstract

    The current paradigm of major histocompatibility complex (MHC) and disease association suggests that efficient binding of autoantigens by disease-associated MHC molecules leads to a T cell-mediated immune response and resultant autoimmune sequelae. The data presented below offer a different model for this association of MHC with autoimmune diabetes. We used several mouse lines expressing different levels of I-Ag7 and I-Ak on the nonobese diabetic (NOD) background to evaluate the role of MHC class II in the previously described NOD T cell autoproliferation. The ratio of I-Ag7 to I-Ak expression correlated with the peripheral T cell autoproliferative phenotype in the mice studied. T cells from the NOD, [NOD x NOD. I-Anull]F1, and NOD I-Ak transgenic mice demonstrated autoproliferative responses (after priming with self-peptides), whereas the NOD.H2(h4) (containing I-Ak) congenic and [NOD x NOD. H2(h4) congenic]F1 mice did not. Analysis of CD4(+) NOD I-Ak transgenic primed lymph node cells showed that autoreactive CD4(+) T cells in the NOD I-Ak transgenic mice were restricted exclusively by I-Ag7. Considered in the context of the avidity theory of T cell activation and selection, the reported poor peptide binding capacity of NOD I-Ag7 suggested a new hypothesis to explain the effects of MHC class II expression on the peripheral autoimmune repertoire in NOD mice. This new explanation suggests that the association of MHC with diabetes results from "altered" thymic selection in which high affinity self-reactive (potentially autoreactive) T cells escape negative selection. This model offers an explanation for the requirement of homozygous MHC class II expression in NOD mice (and in humans) in susceptibility to insulin-dependent diabetes mellitus.

    View details for Web of Science ID 000077713600008

    View details for PubMedID 9858513

  • Local delivery of cytokines by retrovirally transduced antigen-specific TCR+ hybridoma cells in experimental autoimmune encephalomyelitis Vth International Workshop on Cytokines at the Joint Meeting of the Ares-Sereno Foundation / European-Cytokine-Society Dal Canto, R. A., Costa, G., Shaw, M. D., Seroogy, C., Nolan, G. P., Fathman, C. G. JOHN LIBBEY EUROTEXT LTD. 1998: 83–91

    Abstract

    Autoimmune diseases in humans represent an immune attack on self tissue. Current therapies for almost all autoimmune diseases utilize potent and nonspecific immunosuppressive regimens. These therapies are complicated by their side effects and also place the patient at increased risk for opportunistic infections and malignancies. Our current understanding of immune mechanisms underlying autoimmune diseases remains limited. Ongoing studies include identifying genes that predispose an individual to developing autoimmunity, identification of autoantigens that trigger or perpetuate autoimmunity, and studies of immune cell interactions that lead to immune response. Although it may be many years before a full understanding of autoimmunity is obtained, treatment in animal models of autoimmune disease and some human clinical trials have begun to study alternative treatment approaches to therapy of autoimmune disease. Future therapies for autoimmune diseases should target the inappropriate autoimmune response. This article will describe the use of gene therapy in the treatment of autoimmune disease. We believe that autoimmunity can be ameliorated by delivering trans-acting immunoregulatory molecules by retrovirally transduced autoantigen specific T cells that home to lesions of autoimmunity. Until recently, there has not been a practical alternative to systemic delivery of immunoregulatory molecules, however systemic delivery suffers from toxic side effects and dangerous global immunosuppression. In order to study immune regulation using retroviral transduction for local delivery of immunoregulatory products, we used myelin basic protein (MBP) reactive T cell hybridomas in the murine model of multiple sclerosis (MS), experimental allergic encephalomyelitis (EAE). In this report, we show that MBP reactive T cell hybridomas transduced to express IL-4 or TNF, ameliorated or exacerbated disease, respectively. Additionally, the effects of these cells were dependent on T cell receptor (TCR) expression, indicating that the effects were due to homing of the T cells and the local delivery of cytokines. We believe that gene therapy, allowing local delivery of immunoregulatory proteins by autoantigen specific T cells, represents an interesting potential therapy for autoimmune disease.

    View details for Web of Science ID 000076885900015

    View details for PubMedID 9831193

  • T cell receptor (TCR) engagement leads to activation-induced splicing of tumor necrosis factor (TNF) nuclear pre-mRNA JOURNAL OF EXPERIMENTAL MEDICINE Yang, Y., Chang, J. F., Parnes, J. R., Fathman, C. G. 1998; 188 (2): 247-254

    Abstract

    Inducible gene expression is primarily regulated at the level of transcription. Additional steps of "processing" pre-mRNA, involved in the regulation of induced gene expression, have not been previously reported. Here we report a novel mechanism of "activation-induced splicing" of preexisting tumor necrosis factor (TNF) message (pre-mRNA) in naive T lymphocytes after engagement of the T cell receptor (TCR), which still occurs after inhibition of transcription. Expression of TNF has been previously demonstrated to be regulated at both the transcriptional and translational levels. However, neither the large pool of TNF mRNA observed in activated T cells nor TNF protein production, which peaks very shortly after activation, can be solely attributed to increased transcription. Evidence is presented that activation-induced splicing of TNF pre-mRNA plays a significant role in the rapid production of TNF seen in activated T cells. Activation triggers processing of TNF pre-mRNA that has accumulated in naive T cells (before activation-induced transcription), and the mature TNF mRNA is translocated to the cytoplasm for rapid translation and protein production. This novel form of activation-induced splicing of TNF may allow T cells to mount an immediate response to activation stimuli under physiological conditions.

    View details for Web of Science ID 000075236900003

    View details for PubMedID 9670037

  • Following antigen challenge, T cells up-regulate cell surface expression of CD4 in vitro and in vivo JOURNAL OF IMMUNOLOGY Ridgway, W., Fasso, M., Fathman, C. G. 1998; 161 (2): 714-720

    Abstract

    The low precursor frequency of Ag-specific T cells has raised significant barriers to studying the T cell response in vivo. We demonstrate that T cells up-regulate the cell surface expression of CD4 following Ag recognition, which identifies Ag-specific T cells in vitro and in vivo and allows their characterization. The CD4high cell subpopulation contains the Ag-specific population as indicated by Ag-induced proliferation and limiting dilution analyses. The use of the CD4high marker will allow analysis of the dynamics of the T cell immune response in vivo, the study of the suboptimal T cell response to Ag, and the identification of T cells which are reactive to known and unknown autoantigens.

    View details for Web of Science ID 000074728400024

    View details for PubMedID 9670947

  • Anti-CD4 therapy in combined heart-kidney, heart-liver, and heart small bowel allotransplants in high-responder rats TRANSPLANTATION Yin, D. P., Sankary, H. N., Talor-Edwards, C., Chong, A. S., Foster, P., Shen, J. K., Ma, L. L., Williams, J. W., Fathman, C. G. 1998; 66 (1): 1-5

    Abstract

    In these experiments, we studied the role of anti-CD4 (Ox38) monoclonal antibody in the induction of allograft unresponsiveness in high-responder Lewis rats in the single liver, kidney, small bowel, and heart versus the combined heart-kidney, heart-liver, and heart-small bowel transplantation models.ACI heart, kidney, liver, and small bowel allografts were transplanted into untreated and anti-CD4 treated Lewis rats. In selected animals bearing long-surviving ACI liver or kidney allografts for over 3 months, donor-matched second heart or third-party (Brown Norway) heart allografts were transplanted. Simultaneously, heart-liver, heart-kidney, and heart-small bowel transplants were performed on the day of operation. Rejected allografts were verified by autopsy and pathology.ACI liver allografts were permanently accepted by Lewis recipients treated with either regular-dose (5 mg/kg for 4 days) or low-dose (5 mg/kg for 2 days) of anti-CD4 monoclonal antibody. Pretransplant anti-CD4 therapy (5 mg/kg for 4 days but not 5 mg/kg for 2 days) resulted in a long-term survival of kidney allografts (mean survival time [MST] > 100.0 days, n=5). Pretransplant anti-CD4 treatment (5 mg/kg for 4 days) could not induce tolerance when single ACI hearts were transplanted; however, long-term survival of ACI heart allografts could be induced when heart transplants were combined with liver (n=7) or kidney (n=8) transplants. The survival of both ACI heart allografts (MST=25.0 days, n=4) and small bowel allografts (MST=28.0 days, n=4) was also prolonged when simultaneous heart and small bowel transplantation was performed in anti-CD4-treated recipients. The second ACI heart allograft was permanently accepted by tolerant Lewis recipients of ACI liver or kidney allografts induced by anti-CD4 treatment, and third-party heart grafts were acutely rejected without affecting survival of the primary allografts.Our current results show that: (1) there is a vigorous rejection of heart > or = small bowel > kidney > liver in high-responder Lewis rats after pretransplant anti-CD4 therapy; and (2) simultaneous or metachronous combined liver-heart and kidney-heart transplants may protect heart allografts from rejection.

    View details for Web of Science ID 000074889300001

    View details for PubMedID 9679814

  • Prolongation of cardiac graft survival with anti-CD4Ig plus hCTLA4Ig in primates JOURNAL OF SURGICAL RESEARCH Krieger, N. R., Yuh, D., McIntyre, W. B., Flavin, T. F., Yin, D. P., Robbins, R., Fathman, C. G. 1998; 76 (2): 174-178

    Abstract

    The aim of this study was to determine whether the use of combined immunotherapy with a brief course of humanized anti-CD4Ig and hCTLA4Ig would prolong heterotopic cardiac allograft survival in primates (rhesus monkeys). This model was based on work in "high responder" rats where a brief course of depletive anti-CD4mAb plus hCTLA4Ig was successful in inducing transplantation tolerance.Heterotopic cardiac transplants were performed in rhesus recipients. Donor/recipient pairs between groups were confirmed to be reactive prior to transplantation by MLR matching. Humanized anti-CD4Ig, a recently developed anti-CD4mAb, was given at a dose of 20 mg/kg i.v. on days -3, -2, -1, and 0. hCTLA4Ig was administered at 6 mg/kg/dose i.v. on days 0 and 2 for the first recipient and days 0, 2, 4, and 6 for the second recipient. No further immunosuppression was administered. The treated (n = 2) or untreated (n = 5) recipients were followed for graft function by daily palpitation.Treatment with anti-CD4Ig plus hCTLA4Ig resulted in a significant prolongation of heart graft survival (42 days for the first recipient and 52 days for the second recipient) compared to untreated recipients (7 days x 4, 11 days x 1). FACS analysis demonstrated CD4 depletion of anti-CD4 treated animals to <2% on posttransplant day 1. The CD4+ T cells gradually repopulated to 50-70% pretransplant levels just prior to rejection. No adverse responses (fever, tachypnea, tachycardia, infections) were observed.These are the first results demonstrating that a brief course of combined specific induction immunotherapy with humanized anti-CD4Ig plus hCTLA4Ig, in the absence of adjuvant posttransplant immunosuppression, was well tolerated and resulted in marked prolongation of cardiac allograft survival in primates.

    View details for Web of Science ID 000075343200012

    View details for PubMedID 9698519

  • Th1 unresponsiveness can be infectious for unrelated antigens IMMUNOLOGY AND CELL BIOLOGY Charlton, B., Fathman, C. G., Slattery, R. M. 1998; 76 (2): 173-178

    Abstract

    CD4+ T cells may be assigned a functional status (Th1 or Th2) according to the cytokines they produce including IL-2, IFN-gamma and IL-4. Th1 and Th2 CD4+ T cells deliver different isotype-switching signals to antigen-specific B cells which bias the serum Ig isotypes. The stimulation of Th1 or Th2 responses is influenced by adjuvants and administration of antigen in IFA results in Th1 unresponsiveness as evidenced by: (i) reduced T cell proliferation to antigen; (ii) reduced IFN-gamma production in response to antigen; and (iii) reduced IgG2a isotype antigen-specific antibodies following antigen/CFA challenge. The impact of established human gamma globulin (HGG) specific Th1 unresponsiveness on subsequent immunization with an unrelated antigen, human serum albumin (HSA) in Th1-inducing CFA was then examined. When subsequently challenged with a mixture of HSA and HGG in CFA the HGG-specific Th1 unresponsiveness was infectious and dominant, preventing the induction of a Th1 response to HSA. Reduced T cell proliferation, IFN-gamma production and IgG2a antibody were consequently observed in response to HSA. The HGG-specific Th1 unresponsiveness was not infectious when HGG/CFA and HSA/CFA were administered at separate sites. This demonstrates that antigen-specific Th1 unresponsiveness can be infectious for new, molecularly unrelated antigens and supports studies showing that Th1-mediated autoimmune diseases such as experimental allergic encephalomyelitis (EAE) and diabetes can be ameliorated using antigens molecularly distinct from the disease-inducing immunogen.

    View details for Web of Science ID 000073494100008

    View details for PubMedID 9619488

  • Following antigen challenge, T cells upregulate cell surface expression of CD4 in vitro and in vivo Ridgway, W., Fasso, M., Fathman, C. G. FEDERATION AMER SOC EXP BIOL. 1998: A936
  • Prolongation of cardiac graft survival with anti-CD4Ig plus hCTLA4Ig in primates Krieger, N. R., Yuh, D., Robbins, R. C., Fathman, C. G. FEDERATION AMER SOC EXP BIOL. 1998: A898
  • Effects of locally delivered cytokines on EAE Dal Canto, R. A., Shaw, M. K., Steinman, L., Nolan, G. P., Fathman, C. G. FEDERATION AMER SOC EXP BIOL. 1998: A308–A308
  • Definition of a cytotoxic domain of HIV-1 tat Kim, D., Mitchell, D., Brockstedt, D., Engleman, E., Rothbard, J., Fathman, C. G. FEDERATION AMER SOC EXP BIOL. 1998: A294
  • Regulation of programmed cell death following T cell activation in vivo INTERNATIONAL IMMUNOLOGY Yang, Y., Kim, D., Fathman, C. G. 1998; 10 (2): 175-183

    Abstract

    Activation of T cell hybridomas in vitro induces rapid Fas-Fas ligand (FasL)-mediated programmed cell death (apoptosis). In contrast, T cells activated by antigen or superantigen in vivo undergo a population expansion and then decline due to Fas-FasL-mediated activation-induced apoptosis (AIA). We asked how T cells activated by antigen in vivo proliferated before undergoing apoptosis. Two possibilities were analyzed: either (i) the apoptosis program was not 'turned on' or (ii) was 'blocked' during the period of cellular proliferation in vivo. Data presented in this manuscript support the second of these possibilities. CD4+ T cells activated in vivo were resistant to anti-fas-mediated apoptosis until 48 h following staphylococcal enterotoxin B (SEB) administration, despite the fact that activated proliferating T cells expressed high levels of Fas (CD95) antigen and many 'apoptosis genes' were induced within 24 h of SEB administration. The analysis of the expression patterns of 'apoptosis genes' during the T cell activation further suggested that temporal blockade of AIA may be due to the induction of apoptosis-preventing genes, such as bag-1.

    View details for Web of Science ID 000072440200009

    View details for PubMedID 9533445

  • A gene therapy approach to treatment of autoimmune disease IMMUNOLOGIC RESEARCH Seroogy, C. M., Fathman, C. G. 1998; 18 (1): 15-26

    Abstract

    New insights into the underlying mechanisms for the development of autoimmune diseases in humans and various animal models continue to increase with our understanding of factors that drive polarization of T helper (Th) responses and tolerance. This information has led to the development of new treatment strategies, including oral tolerance clinical trails and the use of altered peptide ligands in animal models. These approaches have shown some promise and provided additional insight into the disease processes. The use of gene therapy in many disease states continues to increase. We are starting to see the application of gene therapy in chronic diseases in humans. Gene therapy has been used in several animal models of autoimmune disease with promising preliminary results. In this article, an overview will be provided for the use of gene therapy in autoimmune disease.

    View details for Web of Science ID 000075425800002

    View details for PubMedID 9724846

  • The association of MHC with autoimmune diseases: Understanding the pathogenesis of autoimmune diabetes CLINICAL IMMUNOLOGY AND IMMUNOPATHOLOGY Ridgway, W. M., Fathman, C. G. 1998; 86 (1): 3-10

    Abstract

    The current paradigm of MHC and disease association is efficient binding of autoantigens by disease-associated MHC molecules leading to a T cell-mediated immune response and resultant autoimmune sequelae. Data presented here offer a different model for this association of MHC with autoimmune diabetes. This new explanation suggests that the association of MHC with autoimmunity results from "altered" thymic selection in which high-affinity self-reactive (potentially autoreactive) T cells escape negative selection. This model offers an explanation for the requirement of homozygous MHC class II expression in NOD mice (and in man) in susceptibility to IDDM.

    View details for Web of Science ID 000071367200002

    View details for PubMedID 9434791

  • CD45RB(high) CD4(+) T cells from IFN-gamma knockout mice do not induce wasting disease JOURNAL OF AUTOIMMUNITY Ito, H., Fathman, C. G. 1997; 10 (5): 455-459

    Abstract

    Transfer of CD45RBhigh CD4+ T cells from normal mice to congenic SCID mice induces wasting disease, a murine model of inflammatory bowel disease. In this model, colonic inflammation is considered to be caused by a disregulated Th1 response, and Th1 cytokines, especially IFN-gamma, have been suggested to play an important role in the pathogenesis of wasting disease. In order to elucidate the potential role of IFN-gamma in the pathogenesis of wasting disease, we transferred CD45RBhigh CD4+ T cells from IFN-gamma knockout (GKO) mice to congenic SCID mice. The recipient mice were absolutely free of symptoms and clinical signs of disease and showed body-weight gain similar to that seen in normal mice. These data demonstrate the essential and non-redundant role of IFN-gamma in the pathogenesis of wasting disease.

    View details for Web of Science ID A1997YA16000005

    View details for PubMedID 9376073

  • Introduction of soluble proteins into the MHC class I pathway by conjugation to an HIV tat peptide JOURNAL OF IMMUNOLOGY Kim, D. T., Mitchell, D. J., Brockstedt, D. G., Fong, L., Nolan, G. P., Fathman, C. G., Engleman, E. G., Rothbard, J. B. 1997; 159 (4): 1666-1668

    Abstract

    Protection against most intracellular pathogens requires T cells that recognize pathogen-derived peptides in association with MHC class I molecules on the surface of infected cells. However, because exogenous proteins do not ordinarily enter the cytosol and access the MHC class I-processing pathway, protein-based vaccines that induce class I-restricted CTL responses have proved difficult to design. We have addressed this problem by conjugating proteins, such as OVA, to a short cationic peptide derived from HIV-1 tat (residues 49-57). When APC were exposed in vitro to such protein conjugates, they processed and presented the peptides in association with MHC class I molecules and stimulated CD8+ Ag-specific T cells. Moreover, Ag-specific CTLs were generated in vivo by immunizing mice with histocompatible dendritic cells that had been exposed to protein-tat conjugates.

    View details for Web of Science ID A1997XP43800012

    View details for PubMedID 9257826

  • Rat pancreatic islet and skin xenograft survival in CD4 and CD8 knockout mice 2nd Congress of the Immunology-of-Diabetes-Society Krieger, N. R., Ito, H., Fathman, C. G. ACADEMIC PRESS LTD ELSEVIER SCIENCE LTD. 1997: 309–15

    Abstract

    The relative contributions of the CD4+ and CD8+ T cell subpopulations in xenotransplant rejection were studied using CD4 and CD8 knockout (KO) mice. Wistar Furth (WF, RT1a) rat pancreatic islet or skin xenografts were transplanted into either CD4 or CD8 KO recipients and compared to wild-type controls. Long-term survival of WF islet xenografts was observed in the CD4 KO mice (MST, >66+/-8 days) whereas CD8 KO mice rejected their islet xenografts within 8 days, similar to controls (MST, 7+/-0.2 days). In contrast, WF skin xenografts were rejected in both CD4 and CD8 KO recipients within 8 days. CD4 KO recipients which maintained xenoislets >90 days posttransplant rejected WF skin grafts within 9 days, without rejecting their original islet xenografts. These results suggest that CD4+ cells are essential for mediating islet xenograft rejection. These data also suggest that the absence of either CD4+ or CD8+ T cells is not sufficient to prevent rejection of skin xenografts.

    View details for Web of Science ID A1997XG03300013

    View details for PubMedID 9218759

  • Local delivery of interleukin 4 by retrovirus-transduced T lymphocytes ameliorates experimental autoimmune encephalomyelitis JOURNAL OF EXPERIMENTAL MEDICINE Shaw, M. K., Lorens, J. B., Dhawan, A., DalCanto, R., Tse, H. Y., Tran, A. B., Bonpane, C., Eswaran, S. L., Brocke, S., Sarvetnick, N., Steinman, L., Nolan, G. P., Fathman, C. G. 1997; 185 (9): 1711-1714

    Abstract

    Experimental autoimmune encephalomyelitis (EAE) is an inflammatory autoimmune disease of the central nervous system which serves as a model for the human disease multiple sclerosis. We demonstrate here that encephalitogenic T cells, transduced with a retroviral gene, construct to express interleukin 4, and can delay the onset and reduce the severity of EAE when adoptively transferred to myelin basic protein-immunized mice. Thus, T lymphocytes transduced with retroviral vectors can deliver "regulatory cytokines" in a site-specific manner and may represent a viable therapeutic strategy for the treatment of autoimmune disease.

    View details for Web of Science ID A1997WY11700020

    View details for PubMedID 9151908

  • The use of CD4 and CD8 knockout mice to study the role of T-cell subsets in allotransplant rejection JOURNAL OF HEART AND LUNG TRANSPLANTATION Krieger, N. R., Fathman, C. G. 1997; 16 (3): 263-267

    View details for Web of Science ID A1997WQ62800001

    View details for PubMedID 9087868

  • Local delivery of interleukin-4 by retrovirus-transduced lymphocytes ameliorates experimental autoimmune encephalomyelitis. Shaw, M. K., Lorens, J. B., Dhawan, A., DalCanto, R., Tse, H. Y., Tran, A. B., Bonpane, C., Eswaran, S. L., Brocke, S., Sarvetnick, N., Steinman, L., Nolan, G. P., Fathman, C. G. MOSBY-ELSEVIER. 1997: 1976–76
  • Transport of immunogens into the MHC class I and II pathways by a peptide from HIV tat 40th Symposium of the Alfred-Benzon-Foundation on HLA and Disease - the Molecular Basis Rothbard, J., Kim, D., Mitchell, D., Bockstedt, D., Fong, L., Nolan, G., Fathman, C. G., Engleman, E. MUNKSGAARD. 1997: 161–175
  • Induction of tolerance to small bowel allografts in high-responder rats by combining anti-CD4 with CTLA4Ig TRANSPLANTATION Yin, D. P., Sankary, H. N., Williams, J., Krieger, N., Fathman, C. G. 1996; 62 (11): 1537-1539

    Abstract

    This study was designed to investigate the effectiveness of combined perioperative anti-CD4 and human (h)CTLA4Ig therapy in preventing allorejection of small bowel transplantation in high-responder Lewis rat recipients of ACI grafts. Anti-CD4 (5 mg/kg x 4 days) or hCTLA4Ig (0.5 mg/rat x 2 days) therapy alone delayed, but did not prevent, allograft rejection after small bowel transplantation of ACI into Lewis rats. All grafts were rejected in 18 and 10 days, respectively. However, a regimen of anti-CD4 (5 mg/kg x 4 days) combined with hCTLA4Ig (0.5 mg/rat x 2 days) allowed indefinite survival of ACI small bowel allografts. Second donor-matched heart grafts were permanently accepted, whereas third-party (Sprague-Dawley) heart allografts were rejected by the tolerant recipients. These data suggest that these two reagents produced a synergistic effect in preventing allorejection of small bowel transplantation.

    View details for Web of Science ID A1996VZ23500001

    View details for PubMedID 8970603

  • CD4(+) but not CD8(+) cells are essential for allorejection JOURNAL OF EXPERIMENTAL MEDICINE Krieger, N. R., Yin, D. P., Fathman, C. G. 1996; 184 (5): 2013-2018

    Abstract

    The generation of knockout mice with targeted gene disruption has provided a valuable tool for studying the immune response. Here we describe the use of CD4 and CD8 knockout mice to examine the role of CD4+ and CD8+ cells in initiating allotransplantation rejection. Pretreatment with a brief course of depletive anti-CD4 monoclonal antibody therapy allowed permanent survival of heart, but not skin, allografts transplanted across a major histocompatibility barrier. However, skin as well as heart grafts were permanently accepted in the CD4 knockout mice. Transfer of CD4+ cells into CD4 knockout recipient mice 1 d before skin engraftment reconstituted rejection, demonstrating that CD4+ cells are necessary for initiating rejection of allogeneic transplants. Major histocompatibility complex disparate heart and skin allografts transplanted into CD8 knockout recipients were rejected within 10 d. This study demonstrates that CD4+ but not CD8+ T cells are absolutely required to initiate allograft rejection.

    View details for Web of Science ID A1996VT69400040

    View details for PubMedID 8920888

  • beta-cell destruction may be a late consequence of the autoimmune process in nonobese diabetic mice DIABETES Shimada, A., Charlton, B., TAYLOREDWARDS, C., Fathman, C. G. 1996; 45 (8): 1063-1067

    Abstract

    The NOD mouse is an animal model of IDDM that shows many of the characteristics of human IDDM. It has been proposed that beta-cell destruction in IDDM progresses over time in a linear manner. Recently, we and others have demonstrated that T helper type 1 (Th1) cells have pathogenic roles in the NOD model and proposed that cytokine balances change as the disease progresses. However, it has not been demonstrated how or when the cytokine balances change or how the beta-cell destruction progresses. We have recently demonstrated that the cytokine profiles of CD45RB(low) CD4+ cells correlate either with their pathogenic or with their protective roles in the NOD mouse. To further analyze this apparent correlation between the shift in cytokine level and IDDM, we examined the anti-CD3-induced cytokine profiles of this subset from NOD mice of various ages compared with that from age-matched I-Ak transgenic NOD and BALB/c mice as controls. A significantly higher ratio of anti-CD3-induced interferon-gamma/interleukin-4 was found in diabetic NOD mice (P < 0.0001) but not in age-matched nondiabetic NOD mice. This cytokine ratio did not change significantly until the onset of diabetes in NOD mice. Based upon these results, we propose that IDDM in the NOD mouse progresses as a predominant inflammatory beta-cell dysfunction without actual beta-cell destruction until late in the disease process. This supports the possibility that late-stage immunotherapy may preserve islet beta-cell mass.

    View details for Web of Science ID A1996VA07300010

    View details for PubMedID 8690153

  • Quantitative analysis of T cell activation - Role of TCR ligand density and TCR affinity JOURNAL OF IMMUNOLOGY Kim, D. T., Rothbard, J. B., Bloom, D. D., Fathman, C. G. 1996; 156 (8): 2737-2742

    Abstract

    (B6 X A)F1 mice were immunized with sperm whale myoglobin, and T cell clones and hybridomas were generated. Hybridoma 74a.e9 was specific for the sperm whale myoglobin 67-79 peptide and could be partially activated by a peptide analogue, equine myoglobin with a natural 74G substitution. Using this hybridoma in T cell activation assays, we studied the effects of varying the avidity of the TCR for its ligand, the concentration of MHC:peptide complex on the APC, and the density of TCR on the surface. Varying ligand concentration on the surface of the APC, the TCR avidity, or the density of TCR on the T cell were equally important parameters in driving T cell activation. The mouse myoglobin (74T) analogue, however, acted as an antagonist to the T cell response. Its effectiveness was also partially determined by its ability to bind to MHC. By independently altering each of these variables and following T cell activation, we describe the interrelationships among these three components (MHC:peptide:TCR) that control the activation of the T cell.

    View details for Web of Science ID A1996UE19200013

    View details for PubMedID 8609391

  • Breaking self-tolerance in nonobese diabetic mice JOURNAL OF EXPERIMENTAL MEDICINE Ridgway, W. M., Fasso, M., LANCTOT, A., Garvey, C., Fathman, C. G. 1996; 183 (4): 1657-1662

    Abstract

    Unresponsiveness to self is maintained through two mechanisms of immune regulation: thymic-negative selection and peripheral tolerance. Although thymic-negative selection is a major mechanism to eliminate self-reactive T cells, normal mice have readily detectable populations of T cells reactive to self-proteins but do not exhibit autoimmune responses. It has been postulated that autoimmune disease results from breakdown or loss of peripheral tolerance. We present data that demonstrate that peripheral tolerance or unresponsiveness to self can be broken in nonobese diabetic (NOD) mice. Immunization of NOD mice (but not of conventional mice) with self-peptides caused an immune response to self-peptide with resultant autoproliferation of peripheral lymphocytes. Autoproliferation of self-reactive T cells in NOD mice resulted from the recognition and proliferation of the activated T cells to endogenously processed and presented self-antigens. This loss of self-tolerance demonstrated in vitro may well be the basis of NOD autoimmune disease in vivo.

    View details for Web of Science ID A1996UH14400039

    View details for PubMedID 8666923

  • Immune regulation in type 1 diabetes Symposium on Physiology and Disease Shimada, A., Charlton, B., Rohane, P., TAYLOREDWARDS, C., Fathman, C. G. ACADEMIC PRESS LTD- ELSEVIER SCIENCE LTD. 1996: 263–69

    Abstract

    The non-obese diabetic (NOD) mouse is an animal model of insulin-dependent diabetes mellitus (IDDM) that shows many of the characteristics of human IDDM. In the NOD model, there exists a discrepancy between the onset of insulitis and diabetes suggesting the potential existence of some form of immune regulation that delays beta cell destruction. Our transfer system using NOD-scid/scid (NOD-scid) mice as recipients of donor NOD cells suggested that immune regulatory cells exist in the periphery of NOD mice, not in the islets. These regulatory cells are considered to be memory CD4+ cells which show a Th2 (or Th zero) type cytokine profile following activation in vitro. The function of the memory CD4+ cells seems to change from protective to pathogenic as the disease progresses. Moreover, cytokine profiles of this CD4+ CD45RBlow (memory) population shifted from a Th2 (or Th zero) to a Th1 type response coincident with the onset of hyperglycaemia. These data suggest that the progression of NOD disease from insulitis to frank hyperglycaemia is under the control of CD4+ CD45RBlow immune 'regulatory' cells.

    View details for Web of Science ID A1996UL12400019

    View details for PubMedID 8738972

  • Monoclonal T cells identified in early NOD islet infiltrates IMMUNITY Yang, Y., Charlton, B., Shimada, A., DalCanto, R., Fathman, C. G. 1996; 4 (2): 189-194

    Abstract

    To examine the hypothesis that a single initiating antigen was recognized by a monoclonal T cell population leading to subsequent inflammatory insulitis in non-obese (NOD) mouse islets, we examined the T cell receptor TCR V beta repertoire of islet-infiltrating T cells in very young (2-week-old) NOD mice. In independent experiments, we repeatedly identified one monoclonal TCR V-beta 8.2 gene product expressed by T lymphocytes infiltrating the islets of NOD mice at 2 weeks of age. The resultant inflammatory response quickly obscures the monoclonal nature of the initiating event. These data suggest that autoimmune diabetes in NOD mice may be initiated by recognition of a single autoantigen.

    View details for Web of Science ID A1996TX22500009

    View details for PubMedID 8624809

  • Treatment of experimental encephalomyelitis with a peptide analogue of myelin basic protein NATURE Brocke, S., Gijbels, K., Allegretta, M., Ferber, I., Piercy, C., Blankenstein, T., Martin, R., Utz, U., Karin, N., Mitchell, D., VEROMAA, T., Waisman, A., Gaur, A., Conlon, P., Ling, N., Fairchild, P. J., Wraith, D. C., OGARRA, A., Fathman, C. G., Steinman, L. 1996; 379 (6563): 343-346

    Abstract

    Following induction of experimental encephalomyelitis with a T-cell clone, L10C1, that is specific for the myelin basic protein epitope p87-99, the inflammatory infiltrate in the central nervous system contains a diverse collection of T cells with heterogeneous receptors. We show here that when clone L10C1 is tolerized in vivo with an analogue of p87-99, established paralysis is reversed, inflammatory infiltrates regress, and the heterogeneous T-cell infiltrate disappears from the brain, with only the T-cell clones that incited disease remaining in the original lesions. We found that antibody raised against interleukin-4 reversed the tolerance induced by the altered peptide ligand. Treatment with this altered peptide ligand selectively silences pathogenic T cells and actively signals for the efflux of other T cells recruited to the site of disease as a result of the production of interleukin-4 and the reduction of tumour-necrosis factor-alpha in the lesion.

    View details for Web of Science ID A1996TR32300058

    View details for PubMedID 8552189

  • Pathogenic and protective roles of CD45RB(low) CD4(+) cells correlate with cytokine profiles in the spontaneously autoimmune diabetic mouse DIABETES Shimada, A., Rohane, P., Fathman, C. G., Charlton, B. 1996; 45 (1): 71-78

    Abstract

    The adoptive transfer of splenocytes from diabetic NOD mice to NOD-scid/scid (NOD-scid) recipients results in diabetes. This model was used to test the effect of cotransfer of splenocyte subsets from young nondiabetic NOD mice. As shown previously in other NOD models, the CD4+ subset from young nondiabetic mice significantly delayed the onset of diabetes in splenocyte cotransfers (P < 0.001). The data presented here showed that the development of diabetes in NOD-scid recipients correlated with a rapid increase in peripheral CD45RB(low) CD4+ cells. However, the CD45RB(low) subset of CD4+ cells from young nondiabetic mice protected from diabetes transfer in this model. We therefore examined whether CD45RB(low) CD4+ cells from diabetic mice were pathogenic rather than protective. CD45RB(low) CD4+ splenocytes from diabetic NOD mice were transferred along with CD8+ splenocytes from diabetic mice into NOD-scid recipients, and all of the recipients became diabetic within 5 weeks posttransfer. In contrast, no recipients (0 of 10) of CD45RB(high) CD4+ cells along with CD8+ splenocytes from diabetic mice became diabetic within 5 weeks posttransfer (P < 0.001). A correlate for the difference between CD45RB(low) CD4+ cells from diabetic NOD mice and CD45RB(low) CD4+ cells from nondiabetic mice, which showed protective effect in splenocyte cotransfers, was found in cytokine production after stimulation with anti-CD3 antibodies in vitro. CD45RB(low) CD4+ cells from diabetic mice showed a significantly higher ratio (approximately fivefold) of gamma-interferon (IFN-gamma) to interleukin (IL)-4 when compared with CD45RB(low) CD4+ cells from nondiabetic mice (P < 0.001). In conclusion, the function of the CD45RB(low) population of CD4+ cells changes from a protective to a pathogenic one during the development of disease in the NOD mouse. This change in function correlates with cytokine production in vitro; increased IFN-gamma-to-IL-4 ratio is associated with pathogenic potential and occurs coincident with (or after) the onset of diabetes.

    View details for Web of Science ID A1996TM67700011

    View details for PubMedID 8522063

  • Mice with a disrupted IFN-gamma gene are susceptible to the induction of experimental autoimmune encephalolmyelitis (EAE) JOURNAL OF IMMUNOLOGY Ferber, I. A., Brocke, S., TAYLOREDWARDS, C., Ridgway, W., Dinisco, C., Steinman, L., Dalton, D., Fathman, C. G. 1996; 156 (1): 5-7

    Abstract

    Experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, is an autoimmune disorder seen in mice and rats following immunization with myelin basic protein (MBP) or MBP-derived peptides. IFN-gamma, a cytokine produced by a variety of cells, is involved in many inflammatory and immune regulatory events. Contradictory results concerning exacerbations and the disease course were seen comparing injections of IFN-gamma in humans suffering from multiple sclerosis to studies using anti-IFN-gamma Abs in mice with EAE. To study the role of IFN-gamma and IFN-gamma-producing cells in EAE, we crossed IFN-gamma knockout mice (H-2b) (unable to produce IFN-gamma due to the disruption of the IFN-gamma gene) with an EAE-susceptible mouse strain, B10.PL (H-2u). EAE was seen in IFN-gamma knockout mice, heterozygotic (IFN-gamma +/-) mice, as well as wild-type littermates following immunization with MBP. Histologic analyses of the central nervous system of IFN-gamma knockout mice with EAE revealed massive infiltrates composed of lymphocytes, macrophages, and granulocytes. We conclude that the presence of IFN-gamma is not crucial to the induction or the clinical course of EAE.

    View details for Web of Science ID A1996TM17100002

    View details for PubMedID 8598493

  • Tissue-specific effects of anti-CD4 therapy in induction of allograft unresponsiveness in high and low responder rats. Transplant immunology Yin, D., Fathman, C. G. 1995; 3 (3): 258-264

    Abstract

    In these experiments, we studied the role of anti-CD4 (Ox38) monoclonal antibody in the prevention of heart and/or kidney allograft rejection in low (ACI) and high (Lewis) responder rats. In low responder ACI rats, donor-specific tolerance for heart and kidney allografts (individually or in combination) was achieved by pretransplant anti-CD4 therapy. In high responder Lewis rats, anti-CD4 therapy alone (or combined with anti-CD8 (Ox8), thymectomy or total lymphoid irradiation) did not prevent first-set rejection of heart allografts. This difference was correlated with a more profound and longer lasting CD4+ cell depletion in the low responder strain. Anti-CD4 treatment, however, produced tolerance of kidney transplants in high responder rats. Additionally, anti-CD4 treatment induced tolerance to heart (as well as kidney) allografts in Lewis recipients of combined kidney and heart allografts from ACI. The effects of anti-CD4 treatment thus depend upon the recipient responder status as well as the organs transplanted and the order of transplantation.

    View details for PubMedID 8581415

  • TERNARY COMPLEX THERAPY FOR AUTOIMMUNE-DISEASE HOSPITAL PRACTICE Fathman, C. G. 1995; 30 (8): 57-?

    Abstract

    Induction of an immune response is remarkably simple, involving the interaction of three components: processed antigen, an MHC class II molecule, and a CD4 T-cell receptor. Each part of this ternary complex represents a novel target for interventions that may block subsequent effector phenomena of autoimmune disease. Such specific treatments will be less toxic than those currently available.

    View details for Web of Science ID A1995RN91300018

    View details for PubMedID 7635912

  • INDUCTION OF TOLERANCE TO HEART ALLOGRAFTS IN HIGH RESPONDER RATS BY COMBINING ANTI-CD4 WITH CTLA4IG JOURNAL OF IMMUNOLOGY Yin, D. P., Fathman, C. G. 1995; 155 (4): 1655-1659

    Abstract

    It has been difficult to induce donor-specific transplantation tolerance in high responder Lewis rats. Results presented below demonstrate that amounts of pretransplant anti-CD4 sufficient to allow allograft tolerance in low responder strains (5 mg/kg x 4 days) did not prevent the acute rejection of ACI heart allografts in high responder Lewis recipients. Higher doses of pretransplant anti-CD4 (10 mg/kg, 15 mg/kg, and 20 mg/kg) given alone could delay but not prevent allograft rejection. Pretransplant anti-CD4 combined with anti-CD8, thymectomy, and total lymphoid irradiation all failed to produce tolerance to ACI heart allografts. However, a regimen of anti-CD4 combined with CTLA4Ig allowed indefinite survival of ACI heart allografts (mean survival time, > 100 day). Second-donor matched heart grafts were permanently accepted, and third-party heart grafts were permanently accepted, and third-party heart allografts were rejected by the tolerant recipients. These results suggest a new combination therapeutic strategy for clinical transplantation.

    View details for Web of Science ID A1995RN46400003

    View details for PubMedID 7636225

  • CD4-POSITIVE SUPPRESSOR CELLS BLOCK ALLOTRANSPLANT REJECTION JOURNAL OF IMMUNOLOGY Yin, D. P., Fathman, C. G. 1995; 154 (12): 6339-6345

    Abstract

    Mechanisms of maintenance of transplantation tolerance induced in adult rats by depletive regimens of anti-CD4 before transplantation of vascularized heart allografts were studied. Despite the fact that there has been little evidence that tolerant lymphocytes could prevent allograft rejection after adoptive transfer, we demonstrated a suppressive role for lymphocytes from tolerant animals in vivo. These experiments analyzed the ability of lymphocytes from tolerant rats to protect passenger leukocyte-depleted Lewis heart grafts that had been "parked" in ACI rats (treated with pretransplant anti-CD4 and maintained for > 100 days) compared with their ability to protect transplantation of fresh Lewis heart grafts in naive ACI rats. Although parked Lewis heart grafts were rejected in unmanipulated ACI recipients, parked hearts (but not naive Lewis heart allografts), were permanently accepted by naive ACI rats when syngeneic tolerant spleen cells were adoptively transferred at the time of transplantation. Further, we demonstrated that the suppressor cells in the tolerant spleen cells were CD4+. These results suggest that CD4+ spleen cells from tolerant rats inhibit allograft recognition and may maintain allograft tolerance by blocking the indirect pathway of allorecognition.

    View details for Web of Science ID A1995RC61000014

    View details for PubMedID 7759872

  • ISLET-INFILTRATING LYMPHOCYTES FROM PREDIABETIC NOD MICE RAPIDLY TRANSFER DIABETES TO NOD-SCID/SCID MICE DIABETES Rohane, P. W., Shimada, A., Kim, D. T., Edwards, C. T., Charlton, B., Shultz, L. D., Fathman, C. G. 1995; 44 (5): 550-554

    Abstract

    In an effort to study the development of diabetes in NOD mice, our laboratory developed a novel adoptive transfer model using NOD-scid/scid (NOD-scid) mice as recipients of islet-infiltrating lymphocytes from donor prediabetic female NOD mice. We first confirmed previous results that demonstrated that splenocytes of diabetic and prediabetic female NOD mice could transfer diabetes to NOD-scid mice. We demonstrated that the kinetics of disease transfer were dependent on the age of transferred lymphocytes and reiterated the kinetics of diabetes in conventional female NOD mice. We then demonstrated that islet-infiltrating lymphocytes from prediabetic female NOD mice could transfer diabetes. In contrast with the age-dependent transfer of diabetes seen using splenocytes, islet-infiltrating lymphocytes obtained from prediabetic female NOD mice aged > or = 40 days rapidly transferred diabetes to NOD-scid recipients. The time required to transfer insulin-dependent diabetes mellitus (IDDM) using islet-infiltrating lymphocytes from young prediabetic mice (25 +/- 9 days) was not statistically different from the time required to transfer IDDM using splenocytes from overtly diabetic mice (32 +/- 5 days). Cotransfer of splenocyte cells or CD4+, but not CD8+ spleen cells, from 60- to 80-day-old prediabetic female NOD mice together with either splenocytes from diabetic mice or islet-infiltrating lymphocytes from prediabetic NOD mice delayed the rapid transfer of IDDM, suggesting that CD4+ cells mediated immunoregulation. Use of the NOD-scid islet-infiltrating lymphocyte-adoptive transfer model should help elucidate the pathophysiology of the early inflammatory events leading to insulitis and subsequent beta-cell destruction.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1995QZ82000012

    View details for PubMedID 7729614

  • REGULATION OF AUTOIMMUNE-RESPONSE CURRENT OPINION IN IMMUNOLOGY Ridgway, W. M., Weiner, H. L., Fathman, C. G. 1994; 6 (6): 946-955

    Abstract

    Recent work on such apparently disparate fields as T-cell receptor peptide-induced regulation, superantigens, antigen-induced tolerance, models of peripheral tolerance, apoptosis, and T-cell receptor antagonists demonstrates a similarity in immune response from a regulatory perspective. In many systems, a 'tolerance' pathway is observed, characterized broadly as an initial disturbance in the immune system, with a resulting predominance of effector cells, followed by a homeostatic response (often requiring CD8+ cells) which leads the effector population into T-cell receptor downregulation, T-cell inactivation, anergy and, often, eventual apoptotic death. In the regulated immune response, mixed populations of anergized and apoptosing T cells can be found. In some cases, anergy appears to lead to death while, in other instances, cells revert to a functional state. This review focuses on recent papers examining each of these topics in an attempt to obtain a preliminary integrated picture of immune regulation in autoimmune diseases.

    View details for Web of Science ID A1994PZ42600018

    View details for PubMedID 7710719

  • PREVENTION OF DIABETES AND INSULITIS BY NEONATAL INTRATHYMIC ISLET ADMINISTRATION IN NOD MICE JOURNAL OF AUTOIMMUNITY Charlton, B., TAYLOREDWARDS, C., Tisch, R., Fathman, C. G. 1994; 7 (5): 549-560

    Abstract

    The murine model of human insulin dependent diabetes mellitus (IDDM), the non-obese diabetic (NOD) mouse, develops a T cell-dependent destruction of pancreatic islets. While the target antigens are unknown, there is clearly a lack of tolerance to them. Neonatal intrathymic (i.t.) antigen injection has been successfully employed to prevent insulitis in BB rats but previous i.t. islet antigen studies in NOD mice were done on older mice. We have injected syngeneic islets into the thymus of NOD mice at birth and found that diabetes and insulitis can be completely prevented by this procedure. The effect is islet antigen-specific since other T cell responses, including autoimmune salivary infiltration, ard unaffected. Furthermore, contrary to previous studies, cyclophosphamide administration was unable to induce diabetes in treated mice which suggests that deletion or anergy might be the mechanism by which neonatal intrathymic islet injection protects from disease. However, anti-islet antigen antibodies were still present in these mice which suggests that the mechanism of disease protection may be more complex.

    View details for Web of Science ID A1994PL69600001

    View details for PubMedID 7840850

  • CD4-POSITIVE HEAT-STABLE ANTIGEN-POSITIVE THYMOCYTES CAUSE GRAFT-VERSUS-HOST DISEASE ACROSS NONMAJOR HISTOCOMPATIBILITY COMPLEX INCOMPATIBILITIES EUROPEAN JOURNAL OF IMMUNOLOGY Charlton, B., Meltzer, J., Fathman, C. G. 1994; 24 (7): 1706-1709

    Abstract

    Single-positive thymocytes are the immediate precursors of peripheral recent thymic emigrants (RTE) which develop into mature peripheral T cells. The functional ability of RTE is unclear but their state of differentiation may be relevant to the development of tolerance to peripheral "self" antigens. Since RTE are difficult to analyze, precursor CD4+/8- thymocytes were assessed in a model in vivo to determine their functional capability and their susceptibility to tolerance induction. The ability of both heat-stable antigen-positive (HSA+) (immature) and HSA- (mature) single-positive thymocytes to cause graft-versus-host disease (GVHD) across non-major histocompatibility complex differences were examined. Both HSA- and HSA+ CD4+/8- thymocytes from C3H mice caused lethal GVHD in AKR recipients as did CD4+ peripheral T cells in controls. Further, neonatal C3H thymocytes also caused lethal GVHD in AKR recipients. Since CD4+/8- thymocytes are the precursors of RTE, these results suggest that RTE are not susceptible to tolerance induced to "minor" antigens and may have a normal immune function in vivo. This would suggest that peripheral tolerance may be dependent upon the manner of antigen presentation rather than T cell maturity.

    View details for Web of Science ID A1994NX26100036

    View details for PubMedID 7913040

  • T-CELLS IN THE PATHOGENESIS OF NOD IDDM Fathman, C. G., Rohane, P., Yang, Y., Charlton, B. WILEY-BLACKWELL. 1994: 131–131
  • IMMUNOTHERAPEUTIC STRATEGIES DIRECTED AT THE TRIMOLECULAR COMPLEX ADVANCES IN IMMUNOLOGY, VOLUME 56 Gaur, A., Fathman, C. G. 1994; 56: 219-265

    View details for Web of Science ID A1994BB09D00005

    View details for PubMedID 7521115

  • ISOLATION OF NONOBESE DIABETIC MOUSE T-CELLS THAT RECOGNIZE NOVEL AUTOANTIGENS INVOLVED IN THE EARLY EVENTS OF DIABETES DIABETES Gelber, C., PABORSKY, L., Singer, S., McAteer, D., Tisch, R., Jolicoeur, C., Buelow, R., McDevitt, H., Fathman, C. G. 1994; 43 (1): 33-39

    Abstract

    Insulin-dependent diabetes mellitus (IDDM) is thought to result from chronic, cell-mediated, autoimmune islet damage. Our aim was to identify the earliest T-cell autoantigen in IDDM, reasoning that this antigen could be causally involved in the initiation of the disease. Identification of the earliest beta-cell-specific autoantigen is extremely important in allowing advances in prevention and treatment of initial events in the development of inflammatory insulitis that precedes beta-cell destruction and overt diabetes. Therefore, we analyzed the proliferative responses of peripheral T-cells from young, female nonobese diabetic (NOD) mice to extracts of pancreatic beta-cell lines. We were able to demonstrate that T-cells responsive to beta-cell antigens exist in the peripheral lymphoid tissue of these mice in the absence of deliberate priming before the manifestation of histologically detectable insulitis. T-cell lines and clones isolated from the peripheral lymphatic tissues of young, unimmunized, female NOD mice were also shown to react with extracts of beta-cells. Fractionation of the beta-cell extracts showed that these T-cell clones recognized multiple beta-cell-specific autoantigens but none of the previously reported putative autoantigens (glutamic acid decarboxylase [GAD]65, GAD67, Hsp65, insulin, ICA 69, carboxypeptidase-H, and peripherin). Thus, we can conclude that these responses are specific for novel beta-cell autoantigens. Finally, NOD T-cell proliferative responses were also seen to an extract of human islets suggesting potential shared antigenic determinants between human and mouse beta-cells.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1994MP92000005

    View details for PubMedID 8262314

  • MECHANISMS OF TRANSPLANTATION TOLERANCE ANNUAL REVIEW OF IMMUNOLOGY Charlton, B., Auchincloss, H., Fathman, C. G. 1994; 12: 707-734

    Abstract

    Transplantation tolerance, the long-term acceptance of grafted tissue in the absence of continuous immunosuppression, remains an elusive goal in humans, but it has been achieved in animal models using numerous approaches. The mechanisms behind graft acceptance vary according to the means used to create the state of acceptance. Several major mechanisms can now be recognized. While thymic deletion of T cells appears to be a mainstay of self-tolerance, its role in transplantation tolerance now seems to be less significant. In contrast, extrathymic mechanisms of transplantation tolerance seem to be major factors in long-term graft acceptance. If donor antigens are presented in a nonimmunogenic manner on the graft, e.g. due to modification of graft tissue by culture, peripheral T cells of the recipient may ignore the graft. Alternatively, nonstimulatory presentation of donor antigens on graft tissue can induce a state of unresponsiveness in recipient T cells, i.e. anergy, rather than activating them to destroy the graft. Suppression mechanisms also operate to control graft rejection and may be specific or nonspecific in nature. Specific suppression mechanisms might act in an idiotype or antigen-specific fashion, and evidence is accumulating that this may be mediated through the elaboration of cytokines. Donor antigen-specific T cells may be activated to produce "protective" cytokines which then regulate the generation of destructive T cells. Future therapies will be aimed at affecting graft acceptance through these peripheral mechanisms.

    View details for Web of Science ID A1994NF48700022

    View details for PubMedID 8011295

  • SELECTION FOR AMINO-ACID-SEQUENCE AND J-BETA-ELEMENT USAGE IN THE BETA-CHAIN OF DBA 2V BETA(B)-DERIVED AND DBA 2V BETA(A)-DERIVED MYOGLOBIN-SPECIFIC T-CELL CLONES JOURNAL OF IMMUNOLOGY Ruberti, G., Paragas, V., Kim, D., Fathman, C. G. 1993; 151 (11): 6185-6194

    Abstract

    Previous experiments in our laboratory have demonstrated that there is a marked restriction on the TCR beta-chain usage in DBA/2 mice in response to the sperm whale myoglobin (SpWMb) determinant 110-121, predominated by the use of the V beta 8.2 gene element. We analyzed the response of mice that had been genetically depleted of V beta 8+ T cells by generating a DBA/2 line that carries the V beta a TCR haplotype. Despite the very limited TCR repertoire expressed by DBA/2V beta a mice, they made an excellent response after immunization with the SpWMb 110-121 peptide. Data presented in this manuscript demonstrate that there is an equally restricted TCR V beta-chain utilization in the T-cell response to the determinant SpWMb 110-121 in DBA/2V beta a mice. Unexpectedly, there was a shift of MHC restriction of this determinant to T cells in the V beta a strain when compared with the V beta b strain of DBA/2 mice. We had previously demonstrated that DBA/2 mice utilized both the hybrid E alpha dA beta d MHC molecule as well as the conventional A alpha dA beta d molecule as presenting elements in response to SpWMb 110-121. Data presented in this manuscript demonstrate that the T-cell response in DBA/2V beta a mice is entirely restricted by the A alpha dA beta d MHC class II molecule. By analyzing a panel of SpWMb 110-121-specific T-cell clones from DBA/2V beta a mice, we were able to study the TCR repertoire expressed on T cells from mice that lack the V beta 8.2 gene. The V beta usage by the panel of clones analyzed was remarkably homogeneous. Thirteen of the 17 clones analyzed used the V beta 1 gene segment. Perhaps more striking was the junctional region nucleotide and amino acid sequences that were shared among these clones and that were similar to the V beta 8.2 clones analyzed previously. All clones assayed used the J beta 2.6 element, as did the great majority of the V beta 8.2 clones analyzed from DBA/2 (and B10.D2) V beta b mice. Importantly, in each strain of mice, irrespective of the V beta utilized, each TCR appeared to have selected an acidic amino acid in the beta-chain at position 100.(ABSTRACT TRUNCATED AT 400 WORDS)

    View details for Web of Science ID A1993MH75600031

    View details for PubMedID 8245460

  • PEPTIDES AS THERAPY OF AUTOIMMUNE-DISEASE IDIG Symposium, on the Occasion of the EASD Conference Fathman, C. G. JOHN WILEY & SONS LTD. 1993: 239–44

    View details for Web of Science ID A1993NN66400002

    View details for PubMedID 7924820

  • INDUCTION OF RELAPSING PARALYSIS IN EXPERIMENTAL AUTOIMMUNE ENCEPHALOMYELITIS BY BACTERIAL SUPERANTIGEN NATURE Brocke, S., Gaur, A., Piercy, C., Gautam, A., Gijbels, K., Fathman, C. G., Steinman, L. 1993; 365 (6447): 642-644

    Abstract

    The role of infection in the pathogenesis of clinical relapses that occur in most autoimmune diseases, including multiple sclerosis, remains to be established. Experimental autoimmune encephalomyelitis (EAE) serves as a model for multiple sclerosis, with episodes of relapsing paralysis. In certain strains of mice, T-lymphocytes expressing the V beta 8 T-cell receptor (TCR) engage the amino-terminal epitope Ac1-11 of myelin basic protein, leading to EAE. The bacterial superantigen staphylococcal enterotoxin B (SEB) activates V beta 8-expressing T cells. Here we show that after immunization with Ac1-11, or after transfer of encephalitogenic T-cell lines or clones reactive to Ac1-11, SEB induces exacerbation or relapses of paralytic disease in mice that are in clinical remission following an initial episode of paralysis, and triggers paralysis in mice with subclinical disease. Tumour necrosis factor has a critical role in the mechanism underlying SEB-induced exacerbation of disease, because anti-tumour necrosis factor antibody given in vivo delays the onset of paralysis triggered by SEB. On reactivation of autoaggressive cells through their T-cell receptor, superantigens may induce clinical relapses of autoimmune disease.

    View details for Web of Science ID A1993MB84600057

    View details for PubMedID 7692305

  • EVIDENCE THAT CLONAL ANERGY IS INDUCED IN THYMIC MIGRANT CELLS AFTER ANTI-CD4-MEDIATED TRANSPLANTATION TOLERANCE TRANSPLANTATION Alters, S. E., Song, H. K., Fathman, C. G. 1993; 56 (3): 633-638

    Abstract

    Diabetic (B6) (IE-) mice treated with a depleting regimen of anti-CD4 monoclonal antibody at the time of transplantation with A/J (IEK) islets of Langerhans showed indefinite acceptance of their islet allograft, as evidenced by persistent normoglycemia. To address the mechanisms involved in such anti-CD4 induced transplantation tolerance we studied potentially IE-reactive V beta 11+ T cells from the tolerant allografted mice. Following complete repopulation of the CD4+ cells, both the CD4+V beta 11+ and CD8+V beta 11+ T cell subsets of the transplanted mice were unresponsive to anti-V beta 11 specific crosslinking. In contrast, lymphocytes tested within the first ten days following transplant were responsive to anti-V beta 11 specific crosslinking; this response decreased as a function of time and reached background levels by day 120 posttransplant. Sorting experiments indicated that the response of lymphocytes to anti-V beta 11 specific crosslinking seen during the initial 120 days posttransplant was confined to the peripheral CD8+ cells; the repopulating CD4+V beta 11+ T cells were unresponsive. In addition, administration of r-IL-2 at the time of transplantation induced rejection in anti-CD4-treated animals, again indicating that the peripheral CD8+ cells could respond shortly after transplant if provided with appropriate help. The decreasing response of CD8+ T cells from transplanted animals to anti-V beta 11 stimulation was inversely correlated with the rate of migration of cells from the thymus to the periphery, implying that new thymic migrant V beta 11+ cells, both CD4+ and CD8+, were rendered anergic upon encountering peripheral alloantigen. These data suggest the possibility that recent thymic migrants are rendered anergic upon encountering antigen in the periphery, a simple model to serve as a "fail-safe" mechanism to prevent autoreactivity.

    View details for Web of Science ID A1993LZ87000027

    View details for PubMedID 8212161

  • Stimulating the lymphocytes. Current biology Garrison Fathman, C. 1993; 3 (8): 558-559

    View details for PubMedID 15335703

  • TUMOR IMMUNOLOGY - STIMULATING THE LYMPHOCYTES CURRENT BIOLOGY Fathman, C. G. 1993; 3 (8): 558-559
  • REQUIREMENT FOR CD8+ CELLS IN T-CELL RECEPTOR PEPTIDE-INDUCED CLONAL UNRESPONSIVENESS (VOL 259, PG 91, 1993) SCIENCE FATHMAN, C. G. 1993; 260 (5106): 280

    Abstract

    Through an inadvertent error on my part, I neglected to include the name of one author-Giovina Ruberti-of the report "Requirement for CD8(+) cells in T cell receptor peptide-induced clonal unresponsiveness" (1 Jan., p. 91). The correct order of the authors is as follows: Amitabh Gaur, Giovina Ruberti, Richard Haspel, John P. Mayer, and myself.

    View details for DOI 10.1126/science.260.5106.280-a

    View details for Web of Science ID A1993KX80000010

    View details for PubMedID 17838233

  • SEB INDUCED ANERGY - MODULATION OF IMMUNE-RESPONSE TO T-CELL DETERMINANTS OF MYOGLOBIN AND MYELIN BASIC-PROTEIN JOURNAL OF IMMUNOLOGY Gaur, A., Fathman, C. G., Steinman, L., Brocke, S. 1993; 150 (7): 3062-3069

    Abstract

    Superantigens have the ability to stimulate a subset of T cells based upon their expressed TCR beta-chain. It has been demonstrated that the administration of staphylococcal enterotoxin B (SEB) in mice leads to unresponsiveness in V beta 8+ T cells in vivo which are the same T cells that could be stimulated in vitro by this enterotoxin. We present here data on the effect of SEB administration in DBA/2 and (PL/J x SJL)F1 mice on their T cell response to two different T cell determinants, the responses against which are dominated by the use of V beta 8+ T cells. Treatment of mice with SEB not only diminished their primary T cell proliferative response to these determinants, but also was able to effectively reduce the memory T cell response. SEB treatment, however, showed only a modest effect in preventing Ac 1-11-induced experimental autoimmune encephalomyelitis in H-2u mice.

    View details for Web of Science ID A1993KT80500051

    View details for PubMedID 7681087

  • THE USE OF GRANZYME A AS A MARKER OF HEART-TRANSPLANT REJECTION IN CYCLOSPORINE OR ANTI-CD4 MONOCLONAL ANTIBODY-TREATED RATS TRANSPLANTATION CHEN, R. H., IVENS, K. W., Alpert, S., Billingham, M. E., Fathman, C. G., Flavin, T. F., Shizuru, J. A., Starnes, V. A., Weissman, I. L., Griffiths, G. M. 1993; 55 (1): 146-153

    Abstract

    Granzyme A is a serine protease expressed by populations of human and mouse natural killer cells and activated CD4+ and CD8+ cytotoxic lymphocytes; its expression marks a subset of inflammatory cells in allografts, autoimmune diabetes, and a number of other inflammatory lesions. In order to describe more completely the correlation between granzyme A expression and the presence of in vivo cytolytic effects, we grafted allogeneic rat hearts with vascular anastomoses in a heterotopic location, and treated the hosts with either cyclosporine, anti-CD4 monoclonal antibody (MRC OX38), or no therapy. The grafts were evaluated by palpation for cardiac functions, by immunohistochemistry for CD4/CD8 expression, by hematoxylin-and-eosin staining for inflammatory infiltration, and by in situ hybridization for granzyme A expression. The appearance of granzyme A+ cells in untreated allografts preceded both functional and standard histopathological and immunohistochemical evidence of graft rejection by two days. In donor-recipient combinations where cyclosporine and anti-CD4 treatments allowed indefinite allograft survival, the allografts showed minimal numbers of granzyme A+ cells, whether cellular infiltrates developed or not. The number of granzyme A+ cells present in the cardiac allografts in treated and untreated animals correlated with either current or impending episodes of rejection. The early time course of granzyme A expression suggests that it can be used as an early and reliable marker of graft rejection.

    View details for Web of Science ID A1993KH66000027

    View details for PubMedID 8420039

  • REQUIREMENT FOR CD8+ CELLS IN T-CELL RECEPTOR PEPTIDE-INDUCED CLONAL UNRESPONSIVENESS SCIENCE Gaur, A., Haspel, R., Mayer, J. P., Fathman, C. G. 1993; 259 (5091): 91-94

    Abstract

    T cell receptor (TCR) vaccination in rats prevents the development of experimental allergic encephalomyelitis (EAE), an animal model of multiple sclerosis. The mechanism of this potential immunotherapy was examined by vaccinating mice with an immunogenic peptide fragment of the variable region of the TCR V beta 8.2 gene. Another immunogen that usually induces an immune response mediated by V beta 8.2+ T cells was subsequently inhibited because specific clonal unresponsiveness (anergy) had been induced. Depletion of CD8+ cells before TCR peptide vaccination blocked such inhibition. Thus, the clonal anergy was dependent on CD8+ T cells, and such immunoregulatory T cells may participate in the normal course of EAE.

    View details for Web of Science ID A1993KE60100036

    View details for PubMedID 8418501

  • EVIDENCE THAT ANTI-CD8 ABROGATES ANTI-CD4 MEDIATED CLONAL ANERGY BUT ALLOWS ALLOGRAFT SURVIVAL IN MICE TRANSPLANTATION Song, H. K., Alters, S. E., Fathman, C. G. 1993; 55 (1): 133-139

    Abstract

    Monoclonal antibodies directed against different T cell subpopulations have been used in several rodent models of transplantation to induce long-term unresponsiveness to allografts by a variety of mechanisms. To investigate whether different mechanisms may be operative when different regimens of mAb therapy are used, we studied the effects of various combinations of anti-T-cell antibody treatment on the induction of tolerance in a mouse islet allograft model. Anti-CD4 mAb alone, anti-CD8 mAb alone, anti-CD4 mAb plus anti-CD8 mAb, and anti-Thy1.2 mAb alone were given at the time of engraftment. Only the anti-CD4 mAb and the anti-CD4 mAb plus anti-CD8 mAb regimens were successful in inducing permanent unresponsiveness to islet allografts. We have previously shown that anti-CD4 mAb alone induces permanent unresponsiveness to islet allografts by a mechanism of clonal anergy, as demonstrated by unresponsiveness of potentially alloreactive T cells to anti-T-cell receptor-specific cross-linking. Interestingly, the potentially alloreactive T cell subsets of recipient mice (V beta 5+ and V beta 11+) made unresponsive to islet allografts by anti-CD4 mAb plus anti-CD8 mAb therapy were not found to be anergic using the same assay. Differences between the repopulation kinetics of CD8+ T cells of anti-CD4 mAb plus anti-CD8 mAb treated recipient mice, which accepted islet allografts, and anti-Thy1.2 treated recipient mice, which rejected islet allografts despite similar levels of initial T cell depletion, suggest that unresponsiveness to alloantigen may have been induced in anti-CD4 mAb plus anti-CD8 mAb treated recipients by clearance of donor passenger leukocytes during prolonged CD8+ T cell depletion.

    View details for Web of Science ID A1993KH66000025

    View details for PubMedID 8420037

  • Anti-CD4 antibodies in diabetes. Immunology series Shizuru, J. A., Fathman, C. G. 1993; 59: 237-252

    View details for PubMedID 8096401

  • AMELIORATION OF AUTOIMMUNE ENCEPHALOMYELITIS BY MYELIN BASIC-PROTEIN SYNTHETIC PEPTIDE INDUCED ANERGY SCIENCE Gaur, A., WIERS, B., Liu, A., Rothbard, J., Fathman, C. G. 1992; 258 (5087): 1491-1494

    Abstract

    Experimental autoimmune encephalomyelitis (EAE), a demyelinating disease of the central nervous system that can be induced in susceptible strains of mice by immunization with myelin basic protein (MBP) or its immunodominant T cell determinants, serves as a model of human multiple sclerosis. Tolerance to MBP in adult mice was induced by intraperitoneal injection of synthetic peptides of immunodominant determinants of MBP and prevented MBP-induced EAE. Furthermore, tolerance-inducing regimens of peptides administered to mice after the disease had begun (10 days after induction with MBP) blocked the progression and decreased the severity of EAE. Peptide-induced tolerance resulted from the induction of anergy in proliferative, antigen-specific T cells.

    View details for Web of Science ID A1992JZ62500031

    View details for PubMedID 1279812

  • IMMUNOTHERAPY OF RHEUMATIC DISEASES BASED ON UNDERSTANDING GENETIC PREDISPOSITION TO THE DEVELOPMENT OF THESE DISEASES RHEUMATIC DISEASE CLINICS OF NORTH AMERICA Fathman, C. G. 1992; 18 (4): 915-926

    Abstract

    During the past two decades, investigators have made great inroads in understanding markers of genetic predisposition to the development of rheumatic diseases. An important question that must be addressed by investigators and clinicians is whether this knowledge will ultimately benefit patients, either through genetic counseling or predictions for beneficial therapeutic intervention. This article discusses various disease mechanisms and modes of immunotherapy such as anti-CD4, major histocompatibility complex blockade, and T-cell receptor-based and determinant-induced responsiveness.

    View details for Web of Science ID A1992LA28000012

    View details for PubMedID 1280848

  • ANTI-CD4 MONOCLONAL-ANTIBODIES IN THERAPY - CREATION OF NONCLASSICAL TOLERANCE IN THE ADULT IMMUNOLOGICAL REVIEWS Shizuru, J. A., Alters, S. E., Fathman, C. G. 1992; 129: 105-130

    View details for Web of Science ID A1992JZ99700005

    View details for PubMedID 1361179

  • INHIBITION OF IMMUNE INDUCTION - THE TARGET OF CURRENT TRANSPLANTATION TOLERANCE STRATEGIES CURRENT OPINION IN IMMUNOLOGY FATHMAN, C. G. 1992; 4 (5): 545–47

    View details for Web of Science ID A1992KN23800001

    View details for PubMedID 1418717

  • LIMITED T-CELL RECEPTOR BETA-CHAIN USAGE IN THE SPERM WHALE MYOGLOBIN 110-121/E-ALPHA(D)A-BETA(D) RESPONSE BY H-2(D) CONGENIC MOUSE STRAINS JOURNAL OF IMMUNOLOGY Sellins, K. S., Danska, J. S., Paragas, V., Fathman, C. G. 1992; 149 (7): 2323-2327

    Abstract

    The specificity and TCR gene usage of a panel of sperm whale myoglobin (SpWMb)-reactive T cell clones from DBA/2 mice have previously been characterized, to study structure-function relationships between components of the ternary complex consisting of Ag, TCR, and MHC class II molecules, whose interaction leads to Th cell activation. These DBA/2 clones were specific for epitopes within the residue 110 to 121 region of SpWMb, in the context of the mixed isotype molecule E alpha dA beta d, and expressed the TCR V beta 8.2 gene element. SpWMb-specific T cell hybridomas from the H-2d-congenic B10.D2 mouse strain, which differs from the DBA/2 strain only in the non-MHC background, were generated and compared with the T cell hybridomas from DBA/2 mice, in order to investigate the influence of non-MHC genes on the specificity of the T cell response to the 110-121 epitope. V beta usage by these hybridomas was very homogeneous; three of three DBA/2 and eight of nine B10.D2 hybridomas specific for the 110-121 epitope, in the context of the mixed isotype molecule E alpha dA beta d, expressed the V beta 8.2 gene product. Nucleotide and amino acid sequences of D beta, J beta, and N regions were also similar. One 110-121/E alpha dA beta d-specific B10.D2 hybridoma used V beta 7, a V beta that is clonally deleted in DBA/2 mice. These experiments suggest that a similar set of TCR beta genes are used to respond to a given epitope, regardless of non-MHC background, and they support the hypothesis that, despite great variability between individuals in their non-MHC background genes, human HLA-associated diseases might result from the formation of a particular ternary complex consisting of a shared MHC molecule, a common "disease-associated" epitope, and a shared TCR.

    View details for Web of Science ID A1992JP18900012

    View details for PubMedID 1382096

  • CYCLOSPORINE THERAPY FOR AUTOIMMUNE-DISEASE NEW ENGLAND JOURNAL OF MEDICINE FATHMAN, C. G., MYERS, B. D. 1992; 326 (25): 1693–95

    View details for DOI 10.1056/NEJM199206183262509

    View details for Web of Science ID A1992HY62800009

    View details for PubMedID 1588984

  • What a rheumatologist needs to know about T cell receptor structure and function. journal of rheumatology. Supplement Fathman, C. G. 1992; 32: 12-15

    Abstract

    By understanding normal immune response, it has been possible to develop therapeutic strategies toward the treatment of autoimmune disease. The association of autoimmune disease with the major histocompatibility complex (MHC) class II gene products suggests that the inductive events in which the putative autoantigen is presented on the surface of antigen presenting cells in the context of the MHC class II gene products and is recognized by CD4(+) helper or inducer T cells form an interesting target for immunotherapeutic intervention. By understanding the structure/function relationships of T cell receptors for antigen, it might be possible to develop novel immunotherapeutic strategies for the treatment of seropositive rheumatoid arthritis. Studies described below review recent progress in understanding the components of the ternary complex and suggest possible areas of immunotherapeutic intervention.

    View details for PubMedID 1613729

  • WHAT A RHEUMATOLOGIST NEEDS TO KNOW ABOUT T-CELL RECEPTOR STRUCTURE AND FUNCTION 4TH INTERNATIONAL SYMP ON RHEUMATOID ARTHRITIS : CURRENT CONCEPTS AND FUTURE DIRECTIONS Fathman, C. G. J RHEUMATOL PUBL CO. 1992: 12–17

    Abstract

    By understanding normal immune response, it has been possible to develop therapeutic strategies toward the treatment of autoimmune disease. The association of autoimmune disease with the major histocompatibility complex (MHC) class II gene products suggests that the inductive events in which the putative autoantigen is presented on the surface of antigen presenting cells in the context of the MHC class II gene products and is recognized by CD4(+) helper or inducer T cells form an interesting target for immunotherapeutic intervention. By understanding the structure/function relationships of T cell receptors for antigen, it might be possible to develop novel immunotherapeutic strategies for the treatment of seropositive rheumatoid arthritis. Studies described below review recent progress in understanding the components of the ternary complex and suggest possible areas of immunotherapeutic intervention.

    View details for Web of Science ID A1992HG40700005

  • PRESENTATION OF ANTIGEN BY MIXED ISOTYPE CLASS-II MOLECULES IN NORMAL H-2D MICE JOURNAL OF EXPERIMENTAL MEDICINE Ruberti, G., Sellins, K. S., Hill, C. M., Germain, R. N., Fathman, C. G., Livingstone, A. 1992; 175 (1): 157-162

    Abstract

    A panel of DBA/2 T cell hybridomas specific for the sperm whale myoglobin epitope 110-121 was found to recognize antigen presented by the mixed isotype class II molecule E alpha dA beta d. The response was blocked by monoclonal antibodies specific for E alpha and A beta d chains; in addition, the hybridomas responded to antigen presented by L cells expressing E alpha A beta d molecules, and made no response with L cells expressing I-Ad or I-Ed molecules. Two more groups of hybridomas isolated from DBA/2 and B10.D2 mice immunized with myoglobin also recognized peptide 110-121 presented by E alpha d A beta d. Thus, although it is expressed at biochemically undetectable levels on spleen cells, the E alpha d A beta d molecule is an important presenting element in normal H-2d mice making a conventional immune response to a protein antigen. These results suggest that high levels of class II expression are not a prerequisite for T cell activation.

    View details for Web of Science ID A1992GY43600019

    View details for PubMedID 1730914

  • ANTI-CD8 ABROGATES EFFECT OF ANTI-CD4-MEDIATED ISLET ALLOGRAFT SURVIVAL IN RAT MODEL DIABETES SEYDEL, K., Shizuru, J., Grossman, D., Wu, A., Alters, S., Fathman, C. G. 1991; 40 (11): 1430-1434

    Abstract

    We studied the effects of anti-CD4 treatment of diabetic ACI rats on the induction of tolerance to allogeneic (Lewis) islet allografts. When given as a 4-day treatment regimen, OX38, a mouse anti-rat CD4 antibody, caused depletion of greater than 80% of CD4+ cells from the peripheral blood of treated rats. After induction of diabetes (a single high-dose bolus of streptozocin) and 3 days after the initiation of anti-CD4 immunotherapy, recipient ACI rats were transplanted with fully allogeneic (Lewis) islets of Langerhans via the portal circulation. These transplanted islets were capable of returning the anti-CD4-treated ACI recipients to normoglycemia, which was maintained indefinitely in the absence of further immunosuppression. In contrast, treatment of recipient rats with OX8, an anti-CD8 monoclonal antibody (MoAb), induced only a slight prolongation of graft survival (less than or equal to 30 days). Further characterization of the cellular requirements for the induction of long-term transplantation survival revealed that successful pretransplantation anti-CD4 therapy could be ablated by the coincident treatment of recipient rats with depleting levels of anti-CD8 MoAb. These data point to the necessity of a regulator CD8+ cell in the induction of anti-CD4-mediated transplantation survival in this rat model of islet transplantation.

    View details for Web of Science ID A1991GN93200010

    View details for PubMedID 1834499

  • GENETIC DISSECTION OF T-CELL RECEPTOR-V-BETA GENE REQUIREMENTS FOR SPONTANEOUS MURINE DIABETES JOURNAL OF EXPERIMENTAL MEDICINE Shizuru, J. A., TAYLOREDWARDS, C., Livingstone, A., Fathman, C. G. 1991; 174 (3): 633-638

    Abstract

    It has been demonstrated, in certain autoimmune disease models, that pathogenic T cells express antigen receptors of limited diversity. It has been suggested that the T cells responsible for the pathogenesis of type I diabetes mellitus might similarly demonstrate restricted T cell receptor (TCR) usage. Recently, attempts have been made to identify the V beta subset(s) that initiates and/or perpetuates the antiislet response in a mouse model of spontaneous autoimmune diabetes (non-obese diabetic [NOD] mice). In studies reported here, we have bred NOD mice to a mouse strain that congenitally lacks approximately one-half of the conventional TCR V beta alleles. Included in this deletion are TCR V beta gene products previously implicated as being involved in the pathogenesis of NOD disease. By studying second backcross-intercross animals, we were able to demonstrate that this deletion of TCR V beta gene segments did not prevent the development of insulitis or diabetes.

    View details for Web of Science ID A1991GC96000014

    View details for PubMedID 1831491

  • THE T-CELL RECEPTOR REPERTOIRE INFLUENCES V-BETA ELEMENT USAGE IN RESPONSE TO MYOGLOBIN JOURNAL OF EXPERIMENTAL MEDICINE Ruberti, G., Gaur, A., Fathman, C. G., Livingstone, A. M. 1991; 174 (1): 83-92

    Abstract

    T cell clones recognizing the sperm whale myoglobin (SpWMb) epitope 110-121 in association with H-2d major histocompatibility complex class II molecules display a very limited heterogeneity of T cell receptor (TCR) V beta usage in DBA/2 mice. All clones previously tested used the same V beta 8.2 gene segment and very restricted junctional regions. To investigate the significance of this observation in vivo, we immunized DBA/2 mice with the intact SpW Mb protein or peptide 110-121. Only the V beta 8+ T cells showed any significant response to the 110-121 epitope. The response to peptide 110-121 was then analyzed in mice which, either as a consequence of antibody depletion or through genetic deletion of TCR V beta genes, lacked V beta 8+ peripheral T cells. DBA/2 mice depleted of V beta 8+ T cells by antibody treatment responded poorly to the 110-121 peptide, and only at high antigen concentrations. In contrast, DBA/2V beta a mice (homozygous for a deletion of multiple V beta gene segments including the V beta 8 family) made a response at least as great as that made by DBA/2 mice, even though the DBA/2V beta a mice had a very restricted TCR V beta repertoire compared with DBA/2 mice. Mechanisms which might determine differences in the 110-121 specific response of DBA/2, DBA/2V beta a and F23.1-treated DBA/2 mice are discussed.

    View details for Web of Science ID A1991FU89700011

    View details for PubMedID 2056283

  • ANALYSIS OF THE TERNARY COMPLEX OF ANTIGEN, MHC AND T-CELL RECEPTOR - THE INFLUENCE OF THE T-CELL RECEPTOR V-BETA REPERTOIRE ON THE V-BETA GENE ELEMENT USAGE INTERNATIONAL SYMP ON IR GENES : FROM BIOLOGY TO MEDICINE Ruberti, G., Livingstone, A., Danska, J. S., Gaur, A., Fathman, C. G. EDITIONS SCIENTIFIQUES ELSEVIER. 1991: 491–93

    View details for Web of Science ID A1991GL67400020

    View details for PubMedID 1721721

  • MOLECULAR ANALYSIS OF THE ROLE OF THE HLA CLASS-II GENES DRB1, DQA1, DQB1, AND DPB1 IN SUSCEPTIBILITY TO LYME ARTHRITIS HUMAN IMMUNOLOGY Ruberti, G., Begovich, A. B., STEERE, A. C., Klitz, W., Erlich, H. A., Fathman, C. G. 1991; 31 (1): 20-27

    Abstract

    Using the polymerase chain reaction and sequence-specific oligonucleotide probes, we have determined the distribution of the alleles at the polymorphic class II loci, DRB1, DQA1, DQB1, and DPB1 in 25 patients with Lyme arthritis. Comparison of the frequencies of the DRB1 and DPB1 alleles in Lyme arthritis patients to the Centre d'Etude du Polymorphisme Humaine control panel revealed statistically significant differences between the two groups. An increase in DRB1*1301 is noted along with a unique distribution of the DR4 subtypes within the 9 DR4+ patients. In addition, an increase in the rate DPB1*1001 and the more common DPB1*0201 alleles is reported.

    View details for Web of Science ID A1991FH13300004

    View details for PubMedID 1679052

  • ANTI-CD4 MEDIATES CLONAL ANERGY DURING TRANSPLANTATION TOLERANCE INDUCTION JOURNAL OF EXPERIMENTAL MEDICINE Alters, S. E., Shizuru, J. A., Ackerman, J., Grossman, D., Seydel, K. B., Fathman, C. G. 1991; 173 (2): 491-494

    Abstract

    Depletion of CD4+ cells using anti-CD4 monoclonal antibodies leads to allograft tolerance. Here we show that anti-CD4-mediated tolerance to pancreatic islets of Langerhans transplanted from an A/J (IEk) donor to a diabetic C57B1/6 (B6) (IE-) recipient occurs in the absence of clonal deletion of the potentially IE-reactive V beta 11+ T cells. Instead, a state of clonal anergy is induced in both the CD4+V beta 11+ and CD8+V beta 11+ T cell subsets. This clonal anergy can be partially overcome in vitro by the addition of recombinant interleukin 2.

    View details for Web of Science ID A1991EV17500025

    View details for PubMedID 1899105

  • GENETIC-ANALYSIS OF DIABETES IN THE NONOBESE DIABETIC MOUSE .1. MHC AND T-CELL RECEPTOR BETA-GENE EXPRESSION JOURNAL OF IMMUNOLOGY Livingstone, A., Edwards, C. T., Shizuru, J. A., Fathman, C. G. 1991; 146 (2): 529-534

    Abstract

    Backcross nonobese diabetic (NOD) ((NOD x SWr)F1 x NOD) mice (108 females and 105 males) were typed for MHC, TCR V beta, and monitored for 350 days for the onset of diabetes. The presence of "antipolar" antibodies in the sera and the occurrence of insulitis was examined in a proportion of these backcross mice. There was no difference in the incidence of diabetes in mice heterozygous for TCR V beta b/a vs those homozygous for TCR V beta b/b. Among the 17 diabetics (all female) detected in this backcross, 14/17 were H-2nod/nod but 3/17 were H-2nod/q. This supports a previous observation suggesting that the MHC-linked diabetogenic gene originally thought to be recessive may rather be dominant but have a low penetrance in the heterozygous state. Antipolar autoantibodies were found in both female and male backcross mice, and were similarly distributed in diabetic and nondiabetic mice. There appeared to be no correlation between the level of these auto-antibodies and development of diabetes. The incidence and severity of insulitis was linked to MHC but no influence of TCR genes on insulitis nor an association between insulitis and antipolar antibodies could be demonstrated in this study. Further analyses of H-2nod/nod intercross mice homozygous for TCR V beta a or TCR V beta b are currently underway.

    View details for Web of Science ID A1991EU50500019

    View details for PubMedID 1824775

  • INDUCTION OF DONOR-SPECIFIC UNRESPONSIVENESS TO CARDIAC ALLOGRAFTS IN RATS BY PRETRANSPLANT ANTI-CD4 MONOCLONAL-ANTIBODY THERAPY TRANSPLANTATION Shizuru, J. A., Seydel, K. B., Flavin, T. F., Wu, A. P., Kong, C. C., Hoyt, E. G., Fujimoto, N., Billingham, M. E., Starnes, V. A., Fathman, C. G. 1990; 50 (3): 366-373

    Abstract

    In the present report a monoclonal antibody designated OX-38 directed against the rat CD4 molecule was tested for its ability to prolong the survival of heterotopic vascularized rat heart allografts transplanted across major histocompatibility barriers. Fluorescence-activated cell-sorter analysis showed that administration of OX-38 selectively depleted 80-95% of CD4+ cells from peripheral blood of treated rats. The immunosuppressive effects of OX-38 in vivo were verified by suppression of an antibody response against OX-38 itself as a heterologous protein immunogen. Recipient rats received OX-38 antibody as a single agent given in pretransplant regimens. Nine of 12 treated rats have maintained heterotopic abdominal heart allografts for greater than 175 days. Control rats that did not receive antibody therapy rejected their grafts within 14 days. Rats that maintained heart allografts for greater than 100 days accepted second donor strain hearts but rejected third-party heart grafts transplanted into the femoral space. Anti-CD4-induced allograft unresponsiveness persisted for at least 90 days following surgical removal of donor tissue and retransplantation of a second donor-matched heart. These results indicated that transient, pretransplant therapy with monoclonal antibodies directed against the CD4+ lymphocyte induced specific, long-lasting unresponsiveness to fully MHC-mismatched cardiac allografts in rats without additional immunosuppression.

    View details for Web of Science ID A1990DY85500002

    View details for PubMedID 1976282

  • SELECTIVE T-CELL DEPLETION WITH OX-38 ANTI-CD4 MONOCLONAL-ANTIBODY PREVENTS CARDIAC ALLOGRAFT-REJECTION IN RATS 9TH ANNUAL MEETING AND SCIENTIFIC SESSIONS OF THE INTERNATIONAL SOC FOR HEART TRANSPLANTATION Flavin, T., Shizuru, J., SEYDEL, K., Wu, A., Fujimoto, N., Hoyt, E. G., Ivens, K., Billingham, M., Fathman, C. G., Starnes, V. A. MOSBY-YEAR BOOK INC. 1990: 482–88

    Abstract

    New monoclonal antibodies directed to membrane molecules unique to lymphocyte subsets have provided the means to alter the immune response to alloantigens in a more selective fashion. This investigation demonstrates that monoclonal antibody-induced depletion of CD4 helper/inducer T lymphocytes before transplantation of a fully mismatched heart allograft allows permanent engraftment in rats without further immunosuppression. Five adult male ACI (RT1a) rats received cell-depleting doses of the mouse anti-rat CD4 monoclonal antibody, MRC Ox-38, for 1 month before undergoing heterotopic abdominal heart transplantation. No other immunosuppression was administered, and immunotherapy was discontinued the day of transplantation. After all five Lewis rat (RT1(1)) hearts were maintained free of rejection for more than 3 months, a second heterotopic transplant was performed, this time to the femoral vessels, using either fresh Lewis heart allografts (n = 3) or third-party, Brown-Norway (RT1n) hearts (n = 2). Histologic evaluation was performed 3 weeks later and revealed severe rejection of the femoral Brown-Norway grafts with no evidence of rejection in any of the femoral or original abdominal Lewis grafts. These results suggested that limited, pretransplant anti-CD4 immunotherapy allowed permanent engraftment of fully mismatched cardiac allografts in rats and conferred donor-specific unresponsiveness.

    View details for Web of Science ID A1990EC26800005

    View details for PubMedID 1977898

  • DRB1-STAR-LY10 - A NEW DRB1 ALLELE AND ITS HAPLOTYPIC ASSOCIATION IMMUNOGENETICS MCCLURE, G. R., Ruberti, G., Fathman, C. G., Erlich, H. A., Begovich, A. B. 1990; 32 (3): 214-217

    View details for Web of Science ID A1990ED27600011

    View details for PubMedID 2228048

  • T-CELL RECEPTOR-MEDIATED NEGATIVE SELECTION OF AUTOREACTIVE LYMPHOCYTE-T PRECURSORS OCCURS AFTER COMMITMENT TO THE CD4 OR CD8 LINEAGES JOURNAL OF EXPERIMENTAL MEDICINE Guidos, C. J., Danska, J. S., Fathman, C. G., Weissman, I. L. 1990; 172 (3): 835-845

    Abstract

    To identify the maturational stage(s) during which T cell receptor (TCR)-mediated positive and negative selection occurs, we followed the development of CD4+8- and CD4-8+ T cells from TCRlo CD4+8+ thymic blasts in the presence of different positive and negative selecting (major histocompatibility complex or Mls) elements. We describe novel lineage-committed transitional intermediates that are TCRmed CD4+8lo or TCRmed CD4lo8+, and that show evidence of having been positively selected. Furthermore, negative selection is not evident until after cells have attained one of the TCRmed transitional phenotypes. Accordingly, we propose that negative selection in normal mice occurs only after TCRlo CD4+8+ precursors have been positively selected into either the CD4 or CD8 lineage.

    View details for Web of Science ID A1990DW49100018

    View details for PubMedID 2143774

  • THE PRESUMPTIVE CDR3 REGIONS OF BOTH T-CELL RECEPTOR ALPHA-CHAIN AND BETA-CHAIN DETERMINE T-CELL SPECIFICITY FOR MYOGLOBIN PEPTIDES JOURNAL OF EXPERIMENTAL MEDICINE Danska, J. S., Livingstone, A. M., Paragas, V., Ishihara, T., Fathman, C. G. 1990; 172 (1): 27-33

    Abstract

    The T cell receptor alpha/beta (TCR-alpha/beta) is encoded by variable (V), diversity (D), joining (J), and constant (C) segments assembled by recombination during thymocyte maturation to produce a heterodimer that imparts antigenic specificity to the T cell. Unlike immunoglobulins (Igs), which bind free antigen, the ligands of TCR-alpha/beta are cell surface complexes of intracellularly degraded antigens (i.e., peptides) bound to and presented by polymorphic products of the major histocompatibility complex (MHC). Therefore, antigen recognition by T cells is defined as MHC restricted. A model has been formulated based upon the similarity between TCR-alpha/beta V region and Ig Fab amino acid sequences, and the crystal structure of the MHC class I and Ig molecules. This model predicts that the complementarity determining regions (CDR) 1 and 2, composed of TCR V alpha and V beta segments, primarily contact residues of the MHC alpha helices, whereas V/J alpha and V/D/J beta junctional regions (the CDR3 equivalent) contact the peptide in the MHC binding groove. Because polymorphism in MHC proteins is limited relative to the enormous diversity of antigenic peptides, the TCR may have evolved to position the highly diverse junctional residues (CDR3), where they have maximal contact with antigen bound in the MHC peptide groove. Here, we demonstrate a definitive association between CDR3 sequences in both TCR alpha and beta chains, and differences in recognition of antigen fine specificity using a panel of I-Ed-restricted, myoglobin-reactive T cell clones. Acquisition of these data relied in part upon a modification of the polymerase chain reaction that uses a degenerate, consensus primer to amplify TCR alpha chains without foreknowledge of the V alpha segments they utilize.

    View details for Web of Science ID A1990DL09100005

    View details for PubMedID 1694219

  • SHARED MOLECULAR MARKERS OF GENETIC PREDISPOSITION TO SEROPOSITIVE RHEUMATOID-ARTHRITIS HUMAN IMMUNOLOGY Morel, P. A., HORN, G. T., Budd, R. C., Erlich, H. A., Fathman, C. G. 1990; 27 (2): 90-99

    Abstract

    Rheumatoid arthritis is associated with the human class II major histocompatibility complex antigens known as HLA-DR4. HLA-DR4 can be subdivided by cellular typing into five subtypes: Dw4, Dw10, Dw13, Dw14, and Dw15. By traditional serologic methods, 60-80% of rheumatoid arthritis patients type HLA-DR4 compared to approximately 20% of the general population. It has been demonstrated, using a panel of four alloreactive T-cell clones, each of which recognized HLA-DR4, Dw14 homozygous typing cells, that cells from all of a group of 23 rheumatoid arthritis patients could be recognized by one or more of these clones regardless of the patients' serologic typing. As the expressed polymorphism of the DR molecule is accounted for by the beta 1 gene, this gene was amplified, using the polymerase chain reaction, and sequenced. Seven patients whose cells were recognized by one of the DR4, DW14-specific T-cell clones, T431, were analyzed. All of these patients shared a common sequence in the third hypervariable region of the DR beta 1 chain gene. The sequence identified is the one normally associated with DR4, Dw14 and DR1. Patients and DR4-positive controls whose cells did not stimulate this clone did not share this sequence. These results suggest that this hypervariable region might be an important contribution to a restriction site for the putative causative agent(s) in rheumatoid arthritis.

    View details for Web of Science ID A1990CJ52400003

    View details for PubMedID 2298612

  • THE MOLECULAR-BASIS OF ANTIGEN-SPECIFIC RECOGNITION BY LYMPHOCYTES-T COLLOQUIUM ON DEFENSE MOLECULES Danska, J. S., Livingstone, A. M., Shizuru, J., Fathman, C. G. WILEY-LISS, INC. 1990: 241–254
  • ERROR IN DESCRIPTION OF VIRUS-ANTIGEN SYSTEM TRANSPLANTATION WEYAND, C. M., GORONZY, J. J., SWARZTRAUBER, K., FATHMAN, C. G. 1989; 48 (6): 1078
  • IMMUNOSUPPRESSION BY ANTI-CD4 TREATMENT INVIVO - CELLULAR AND HUMORAL RESPONSES TO ALLOANTIGENS TRANSPLANTATION Weyand, C. M., GORONZY, J., Swarztrauber, K., Fathman, C. G. 1989; 47 (6): 1039-1042

    Abstract

    Antibody-mediated elimination of CD4+ lymphocytes in vivo has been successfully used to suppress the humoral response to foreign antigens and to induce long-term tolerance. However, secondary humoral responses, as well as secondary cytolytic responses specific for viral antigens, could not be prevented, providing evidence for functional heterogeneity within the helper cell compartment. Data presented here support the notion that helper cell requirements for cellular responses to alloantigens are unique and do not involve CD4+ T lymphocytes. While the administration of anti-CD4 mcAb failed to suppress allospecific CTL responses, the formation of alloantibodies was initially inhibited in parallel to the deficiency in CD4+ helper cells. After regeneration of CD4+ T cells, the animals regained the ability to produce specific IgG alloantibodies. The dichotomy of helper pathways in humoral and cellular alloreactive responses challenges the concept of a single CD4+ helper cell population. Insights into the functional heterogeneity of helper cells for primary, secondary, and allospecific responses might open new avenues for selective manipulation of helper subpopulations.

    View details for Web of Science ID A1989AB98900024

    View details for PubMedID 2472024

  • IMMUNOSUPPRESSION BY ANTI-CD4 TREATMENT INVIVO - PERSISTENCE OF SECONDARY ANTIVIRAL IMMUNE-RESPONSES TRANSPLANTATION Weyand, C. M., GORONZY, J., Swarztrauber, K., Fathman, C. G. 1989; 47 (6): 1034-1038

    Abstract

    Elimination of CD4+ helper T cells by treatment with monoclonal antibodies (mcAb) in vivo has been used as a new mode of immunosuppression in organ transplantation and autoimmune diseases. To explore the potential risks of this therapeutic approach we have studied antiviral responses in mice depleted of CD4+ T cells. Depletion of CD4+ T cells in vivo completely suppressed the generation of a primary virus-specific cytotoxic response. Injection of high doses of recombinant interleukin-2 (rIL-2) given after virus immunization restored the responsiveness of helper cell-depleted mice to virus-expressing target cells, suggesting a crucial role of IL-2 in antiviral defense mechanisms. In contrast to primary responses, memory cytolytic responses to viral antigens persisted despite depletion of greater than 90% of CD4+ helper T cells. The generation of such memory cytotoxic responses was dependent upon help provided by CD4+ lymphocytes surviving the antibody therapy. After antibody treatment, frequencies of virus-specific helper cells were minimal in primed mice, excluding insufficient helper cell elimination as the reason for the persistence of memory responses. Data presented here suggest that there exist distinct helper pathways in primary and secondary cytolytic antiviral responses that might represent several subsets of helper T cells as well as differences in helper signals required by distinct effector cells.

    View details for Web of Science ID A1989AB98900023

    View details for PubMedID 2472023

  • CARDIAC ALLOGRAFT PROLONGATION IN MICE TREATED WITH COMBINED POSTTRANSPLANTATION TOTAL-LYMPHOID IRRADIATION AND ANTI-L3T4 ANTIBODY THERAPY TRANSPLANTATION TRAGER, D. K., Banks, B. A., Rosenbaum, G. E., HOLM, B. I., Shizuru, J. A., Strober, S., Fathman, C. G. 1989; 47 (4): 587-591

    Abstract

    Neonatal cardiac allograft survival was examined in mice treated with anti-L3T4 antibody, posttransplantation total lymphoid irradiation (TLI) or a combination of both therapies. Independently, both posttransplantation TLI and short-course antibody treatment allowed minimal prolongation. However, synergistic prolongation in graft survival was observed with the combination (synergistic) therapy. Fluorescence-activated cell sorter analysis of peripheral blood lymphocytes from animals treated with combined anti-L3T4 and posttransplantation TLI additionally revealed "synergy" with respect to the degree of peripheral lymphocyte depletion.

    View details for Web of Science ID A1989U208300004

    View details for PubMedID 2523098

  • PGP-1 IDENTIFIES A SUBPOPULATION OF MEMORY CD4+ LYMPHOCYTES BUDD, R. C., SCHUMACHER, J., WINSLOW, G., BUTTERFIELD, K., FATHMAN, C. G., MOSMANN, T. R. SLACK INC. 1989: A555
  • IMMUNOGENETICS OF RHEUMATOID-ARTHRITIS JOURNAL OF RHEUMATOLOGY MOREL, P. A., FATHMAN, C. G. 1989; 16 (4): 421–23

    View details for Web of Science ID A1989U606200002

    View details for PubMedID 2568488

  • TRANSIENT ANTI-CD4 MONOCLONAL-ANTIBODY THERAPY ALLOWS INDEFINITE CARDIAC ALLOGRAFT SURVIVAL IN RATS Shizuru, J. A., Seydel, K. B., Kong, C. S., Wu, A. P., Flavin, T. F., Hoyt, E. G., VENS, K., Starnes, V. A., Fathman, C. G. SLACK INC. 1989: A560–A560
  • A SUBSET OF MEMORY CD4+ HELPER LYMPHOCYTES-T IDENTIFIED BY EXPRESSION OF PGP-1 JOURNAL OF EXPERIMENTAL MEDICINE Butterfield, K., Fathman, C. G., Budd, R. C. 1989; 169 (4): 1461-1466

    Abstract

    The Pgp-1 glycoprotein (Ly-24 antigen) is acquired by mature murine T lymphocytes at the time of primary antigen stimulation Pgp-1 was previously shown to be a useful cell surface marker for distinguishing antigen-specific memory CD8+ T lymphocytes after immunization. Here we demonstrate that this observation extends to CD4+ T lymphocytes. Antigen-specific CD4+ T cells in mice immunized with sperm whale myoglobin or keyhole limpet hemocyanin were contained nearly exclusively in the minor Pgp-1+ subset.

    View details for Web of Science ID A1989T962500019

    View details for PubMedID 2564418

  • PRESENTATION OF EXOGENOUS PROTEIN ANTIGENS BY DENDRITIC CELLS TO T-CELL CLONES - INTACT PROTEIN IS PRESENTED BEST BY IMMATURE, EPIDERMAL LANGERHANS CELLS JOURNAL OF EXPERIMENTAL MEDICINE Romani, N., Koide, S., Crowley, M., WITMERPACK, M., Livingstone, A. M., Fathman, C. G., Inaba, K., Steinman, R. M. 1989; 169 (3): 1169-1178

    Abstract

    The capacity of dendritic cells to present protein antigens has been studied with two MHC class II-restricted, myoglobin-specific, T cell clones. Spleen dendritic cells and cultured epidermal Langerhans cells (LC) presented native myoglobin weakly and often not at all. These same populations were powerful stimulators of allogeneic T cells in the primary MLR. Freshly isolated LC were in contrast very active in presenting proteins to T cell clones but were weak stimulators of the MLR. Both fresh and cultured LC could present specific peptide fragments of myoglobin to the clones. These results suggest that dendritic cells in nonlymphoid tissues like skin can act as sentinels for presenting antigens in situ, their accessory function developing in two phases. First antigens are captured and appropriately presented. Further handling of antigen then is downregulated while the cells acquire strong sensitizing activity for the growth and function of resting T lymphocytes. The potent MLR stimulating activity of cultured epidermal LC and lymphoid dendritic cells probably reflects prior handling of antigens leading to the formation of allogeneic MHC-peptide complexes.

    View details for Web of Science ID A1989T533800041

    View details for PubMedID 2522497

    View details for PubMedCentralID PMC2189287

  • ENHANCED CELL-MEDIATED PROTECTION AGAINST FATAL ESCHERICHIA-COLI SEPTICEMIA INDUCED BY TREATMENT WITH RECOMBINANT IL-2 JOURNAL OF IMMUNOLOGY GORONZY, J., WEYAND, C., Quan, J., Fathman, C. G., OHANLEY, P. 1989; 142 (4): 1134-1138

    Abstract

    Administration of rIL-2 to BALB/c mice induces a rapid, cell-mediated response that is sufficient to protect mice from a lethal i.p. dose of Escherichia coli. Mice were protected from septic death if IL-2 was administered i.p. within 1 h after the bacterial challenge. Optimal protection was provided by treating the lethally challenged mice with rIL-2 at 1 and 5 h or 1, 5, and 10 h after the bacterial challenge and was dose-dependent (greater than or equal to 5.0 x 10(5) U/kg). Furthermore, treatment of mice with anti-IL-2R antibody abolished the protective effect induced by rIL-2 administration. These data suggest that the rIL-2-induced protection against septic death in mice is mediated by a cell type expressing a functional IL-2R. One potentially important therapeutic application of rIL-2 may be to modulate the course of sepsis once the host has been exposed to potentially lethal microbial pathogens.

    View details for Web of Science ID A1989T159900011

    View details for PubMedID 2644349

  • A NEW LOOK AT THE SHARED EPITOPE HYPOTHESIS AMERICAN JOURNAL OF MEDICINE Morel, P. A., Erlich, H. A., Fathman, C. G. 1988; 85 (6A): 20-22

    Abstract

    The striking correlation observed between T cell recognition and the sharing of the DR-beta-1 gene sequences (position 67-74) among patients with rheumatoid arthritis studied suggests that the third hypervariable region might be an important contribution to one restriction site for the putative causative agent(s) in rheumatoid arthritis. The fact that this sequence was found in DR1, DR4,Dw14, and DR4,Dw15 beta-1 genes lends support to the hypothesis that, in some cases, human leukocyte antigen and disease association may involve the association of discrete disease-related epitopes rather than entire human leukocyte antigen genes and that these epitopes are immunologically relevant in terms of T cell recognition. The association of these polymorphisms with susceptibility to rheumatoid arthritis would then support the hypothesis that binding and presentation of "arthritogenic peptides" by major histocompatibility complex class II molecules is one of the pathogenic events in developing rheumatoid arthritis.

    View details for Web of Science ID A1988R586200008

    View details for PubMedID 2462347

  • USE OF ANTI-L3T4 AND ANTI-IA TREATMENTS FOR PROLONGATION OF XENOGENEIC ISLET TRANSPLANTS TRANSPLANTATION Kaufman, D. S., Kong, C. S., Shizuru, J. A., Gregory, A. K., Fathman, C. G. 1988; 46 (2): 210-215

    Abstract

    The effects of T helper lymphocyte and Ia+ cell depletion were examined for their ability to independently and synergistically achieve prolongation of xenogeneic (rat-to-mouse) islet transplants. Recipient mice were depleted of T helper lymphocytes by short-term treatment with the anti-L3T4 monoclonal antibody GK1.5. Donor rat islets were treated prior to transplantation with a concentration of anti-Ia immunotoxin (13.4 x RT) that selectively depleted Ia+ cells within the islets while leaving functional insulin-secreting beta-cells unaffected. Anti-L3T4 treatment alone allowed transplants to be prolonged compared with untreated controls; however, all such treated mice rejected their xenogeneic transplant within 22 days. Although 13.4 x RT treatment of donor islets alone did not prolong engraftment, when donor rat islets were pretreated with the anti-Ia immunotoxin and grafted into L3T4-depleted mice, normoglycemia was maintained for greater than 50 days in 56% of transplants. These results suggest that neither L3T4 depletion nor anti-Ia immunotoxin treatment alone is enough to achieve indefinite survival of xenogeneic islets. However, decreasing the immunogenicity of the transplanted islets by anti-Ia immunotoxin treatment prior to transplantation into anti-L3T4 treated mice can allow greatly prolonged xenogeneic graft survival.

    View details for Web of Science ID A1988P695400005

    View details for PubMedID 2970132

  • IMMUNOTHERAPY OF THE NONOBESE DIABETIC MOUSE - TREATMENT WITH AN ANTIBODY TO T-HELPER LYMPHOCYTES SCIENCE Shizuru, J. A., TAYLOREDWARDS, C., Banks, B. A., Gregory, A. K., Fathman, C. G. 1988; 240 (4852): 659-662

    Abstract

    Spontaneous diabetes mellitus was blocked in nonobese diabetic mice by treatment with a monoclonal antibody against the L3T4 determinant present on the surface of T-helper lymphocytes. Sustained treatment with the monoclonal antibody led to cessation of the lymphocytic infiltration associated with the destruction of the insulin-producing beta cells. Moreover, the mice remained normoglycemic after the antibody therapy was stopped. These studies indicate that immunotherapy with monoclonal antibodies to the lymphocyte subset may not only halt the progression of diabetes, but may lead to long-term reversal of the disease after therapy has ended.

    View details for Web of Science ID A1988N126500035

    View details for PubMedID 2966437

  • CORRELATION OF T-CELL RECEPTOR-V-BETA GENE FAMILY WITH MHC RESTRICTION JOURNAL OF EXPERIMENTAL MEDICINE MOREL, P. A., LIVINGSTONE, A. M., FATHMAN, C. G. 1987; 166 (2): 583–88

    Abstract

    We have studied a panel of DBA/2 T cell clones specific for sperm whale myoglobin (SpW Mb) for TCR (T cell receptor) beta chain gene expression by FACS analysis using the monoclonal antibodies F23.1 and KJ16 specific for the V beta 8 family of the TCR beta chain genes. Within any given specificity group, all the clones tested came from different mice. 10 of 11 I-Ed-restricted SpW Mb-specific T cell clones were F23.1+; 8 of these were also KJ16+. Only one of the three I-Ad-restricted clones tested was F23.1+; this clone was KJ16 negative. This study has demonstrated that I-Ed-restricted T cell clones from DBA/2 mice express members of the TCR V beta 8 family irrespective of the epitopes of SpW Mb recognized. These data suggest an apparent correlation between TCR V beta expression and MHC restriction.

    View details for DOI 10.1084/jem.166.2.583

    View details for Web of Science ID A1987J401600022

    View details for PubMedID 3598467

    View details for PubMedCentralID PMC2189600

  • ISLET ALLOGRAFT SURVIVAL AFTER A SINGLE COURSE OF TREATMENT OF RECIPIENT WITH ANTIBODY TO L3T4 SCIENCE Shizuru, J. A., Gregory, A. K., CHAO, C. T., Fathman, C. G. 1987; 237 (4812): 278-280

    Abstract

    Allografts of pancreatic islets of Langerhans were induced to survive for an indefinite period in diabetic mice if, at the time of engraftment, the mice received a single course of treatment with a monoclonal antibody directed against the L3T4 determinant, a nonpolymorphic cell surface glycoprotein present on the cell surface of the murine T helper-inducer lymphocyte subset. This treatment allowed the survival of islets of Langerhans transplanted across a major histocompatibility barrier without additional immunosuppression. The results demonstrate that the lymphocyte subset defined by the expression of the L3T4 molecules is central to the induction of allograft rejection and provides a model for tolerance induction for organ allograft transplantation.

    View details for Web of Science ID A1987J154700019

    View details for PubMedID 2955518

  • ADMINISTRATION INVIVO OF RECOMBINANT INTERLEUKIN-2 PROTECTS MICE AGAINST SEPTIC DEATH JOURNAL OF CLINICAL INVESTIGATION WEYAND, C., GORONZY, J., Fathman, C. G., OHANLEY, P. 1987; 79 (6): 1756-1763

    Abstract

    Administration in vivo of recombinant interleukin 2 (rIL-2) to mice induces a polyclonal IgM response. When co-administered with a specific antigen, rIL-2 can enhance concentrations of murine IgM antibodies specific for the antigen by fivefold within 7 d of initial treatment. IgM antibodies that are induced after injection of rIL-2 include antibodies specific for J5, a cell wall core lipopolysaccharide (LPS) antigen that is shared by the different members of the Enterobactericeae family. We report here that mice pretreated with rIL-2 or immunized with J5 antigen 7 d before bacterial challenge were protected from septic death that is caused by intraperitoneal challenges with Escherichia coli. Optimal protection was provided by a combined J5 antigen and rIL-2 treatment. Acquisition of the rIL-2 and J5 antigen-induced protection against lethal bacterial infection coincided temporally with maximal serum IgM titers that also contained IgM antibodies specific for the J5 antigen. In passive immunization experiments, the affinity-purified IgM fraction in sera of rIL-2-treated animals was identified as necessary and sufficient for protection. The IgM-depleted serum had no protective effect. The nonspecific augmentation of host-defense mechanisms without the induction of endotoxin manifestations makes rIL-2 a potential candidate to any alternative LPS-containing vaccines for the prevention of bacterial infections by gram-negative organisms since the core LPS antigen is shared among gram-negative bacteria.

    View details for Web of Science ID A1987H575600029

    View details for PubMedID 3294901

    View details for PubMedCentralID PMC424518

  • HETEROGENEITY OF SIGNAL REQUIREMENTS IN T-CELL ACTIVATION WITHIN A PANEL OF HUMAN PROLIFERATIVE T-CELL CLONES JOURNAL OF IMMUNOLOGY GORONZY, J., WEYAND, C., Imboden, J., Manger, B., Fathman, C. G. 1987; 138 (10): 3087-3093

    Abstract

    Activation of T lymphocytes is initiated by receptor ligand interactions at the cell surface leading to the transduction of intracellular signals followed by the de novo synthesis and expression of T cell activation markers (including receptors for interleukin 2 (IL 2) and transferrin), production of lymphokines, and T cell proliferation. This requisite first step for activation of T lymphocytes can be mimicked in certain situations with a variety of stimuli. These include antibodies to certain integral membrane proteins, phorbol esters, and plant lectins that act as mitogens. In this paper, we report that at least two classes of human T cell clones can be distinguished based upon signal requirements necessary to induce proliferation. Although all clones analyzed expressed IL 2 receptors and secreted IL 2 after non-antigenic activation, one subset of clones did not proliferate in response to the same non-antigenic signals. In that subset, complete activation leading to proliferation required interaction of the T cell with specific antigen. The ability to subset these T cell clones into two groups did not correlate with phenotypic differences, source of the clone, nor with magnitude of intracellular calcium mobilization. By studying the stimulation requirements of these two subsets of human T cell clones through the use of specific antigen or antigen-independent stimuli, it was possible to demonstrate that different stimuli varied in their ability to induce steps of T cell activation. Analysis of reactivity of these clones to suboptimal stimulation allowed the definition of intermediate stages of T cell activation. Such intermediate stages might reflect a diversity of intracellular signaling pathways or a complexity of regulatory mechanisms distal to the events that allow intracellular calcium mobilization. Thus for the first time, it has been possible to study ordered events of T cell activation in non-transformed, antigen-dependent human T lymphocytes. The data presented in this paper suggest that T cell activation is not an all or nothing phenomenon, and there is an ordered sequence of events that can be differentiated based upon signal requirements at the T cell membrane.

    View details for Web of Science ID A1987H215900001

    View details for PubMedID 3106471

  • ANTIBODY-MEDIATED DEPLETION OF T-HELPER LYMPHOCYTES ALLOWS INDEFINITE SURVIVAL OF PANCREATIC ALLOGRAFTS Shizuru, J. A., Gregory, A. K., Fathman, C. G. SLACK INC. 1987: A465–A465
  • MODULATION OF CD4 BY ANTIGENIC ACTIVATION JOURNAL OF IMMUNOLOGY Weyand, C. M., GORONZY, J., Fathman, C. G. 1987; 138 (5): 1351-1354

    Abstract

    Cell surface expression of CD4, an invariant membrane glycoprotein, is characteristic of the MHC class II-restricted T cell helper/inducer subset. Although the specificity and restriction patterns of T lymphocytes are determined by the T cell receptor for antigen, CD4 might represent an additional "interaction" molecule that is required to strengthen the interaction between T cells, antigen, and antigen-presenting cells. In this manuscript, we have shown that the cell surface expression of CD4 is correlated with activation of T cells. Data presented in this paper have demonstrated, for the first time, that antigenic stimulation of human T cell clones caused a decrease in the expression of the CD4 marker (as well as to the CD3 marker) to about 50% of the constitutive level. As previously demonstrated, PMA caused modulation of CD4 and CD3, which suggested that phosphorylation by protein kinase C played a crucial role in the regulation of the expression of both markers. The parallel down-regulation of CD3 and CD4 after antigen stimulation suggested that both markers might be members of a multimolecular complex mediating T cell activation.

    View details for Web of Science ID A1987G268700007

    View details for PubMedID 3100638

  • CLONING OF HUMAN ALLOREACTIVE T-CELLS METHODS IN ENZYMOLOGY GORONZY, J., WEYAND, C., Fathman, C. G. 1987; 150: 333-341

    View details for Web of Science ID A1987M168400028

    View details for PubMedID 3501522

  • FACS ANALYSIS OF T-CELL RECEPTOR USAGE IN THE DBA-2 RESPONSE TO SPERM WHALE MYOGLOBIN MOREL, P., LIVINGSTONE, A., FATHMAN, C. G. WILEY-LISS. 1987: 299
  • HETEROGENEITY OF THE T-CELL RESPONSE TO SPERM WHALE MYOGLOBIN LIVINGSTONE, A. M., ROTHBARD, J. B., FATHMAN, C. G. WILEY-LISS. 1987: 262
  • MURINE T-CELL CLONES METHODS IN ENZYMOLOGY Livingstone, A., Fathman, C. G. 1987; 150: 325-333
  • PRODUCTION OF ANTIBODIES INVITRO IN CULTURES OF MURINE LYMPHOCYTES METHODS IN ENZYMOLOGY Ishihara, T., Fathman, C. G. 1987; 150: 304-309

    View details for Web of Science ID A1987M168400024

    View details for PubMedID 3323787

  • THE STRUCTURE OF T-CELL EPITOPES ANNUAL REVIEW OF IMMUNOLOGY Livingstone, A. M., Fathman, C. G. 1987; 5: 477-501

    Abstract

    We have reviewed here studies using synthetic peptides to analyze some of the properties of T-cell epitopes. Several general conclusions can be drawn. First, T-cell epitopes can usually be defined by linear sequences of about seven amino acids. However, the observation that increasing peptide length often results in increased antigenic potency has suggested that antigenicity may crucially depend upon the ability of peptides to adopt appropriate secondary structures. Two models for the prediction of T-cell epitopes on the basis of primary sequence data alone were discussed. Biophysical studies on the association of peptides with Ia molecules have shown that antigenic peptides bind directly to Ia; the evidence suggests that a binary association between Ia and peptide occurs in the absence of specific T-cells. Finally, a hypothesis to explain the observation that B-cells and T-cells generally recognize distinct epitopes on multideterminant antigens has been examined.

    View details for Web of Science ID A1987G957300020

    View details for PubMedID 2439104

  • INHIBITION OF RAT MIXED LYMPHOCYTE PANCREATIC-ISLET CULTURES WITH ANTI-IA IMMUNOTOXIN TRANSPLANTATION Shizuru, J. A., Ramakrishnan, S., Hunt, T., Merrell, R. C., Fathman, C. G. 1986; 42 (6): 660-666

    Abstract

    Studies reported here indicate that an anti-Ia immunotoxin can eliminate the allostimulatory subpopulation of cells present within the islets of Langerhans without damaging the hormone-secreting cells. Such studies made use of an in vitro correlate of transplantation rejection, the mixed lymphocyte islet cell (MLIC) reaction. Using the MLIC, it was demonstrated that an anti-Ia immunotoxin removed cells capable of stimulating the MLIC in a dose-dependent fashion without altering the hormone-secreting functions of the remaining cells when challenged with glucose and theophylline. These studies suggest the feasibility of using such anti-Ia immunotoxins in islet allograft transplantation models to circumvent problems inherent in complement-mediated cytotoxicity, a previously documented effective form of inducing islet allotransplantation tolerance.

    View details for Web of Science ID A1986F366100016

    View details for PubMedID 2947362

  • LONG-TERM HUMORAL UNRESPONSIVENESS INVIVO, INDUCED BY TREATMENT WITH MONOCLONAL-ANTIBODY AGAINST L3T4 JOURNAL OF EXPERIMENTAL MEDICINE GORONZY, J., Weyand, C. M., Fathman, C. G. 1986; 164 (3): 911-925

    Abstract

    mAbs directed against the L3T4 molecule administered in vivo caused a severe and long lasting helper cell depletion in mice. Regeneration of the L3T4+ subpopulation occurred gradually (2-3 mo) after a single antibody treatment. Experiments were designed to examine the humoral immunocompetence of such anti-L3T4-treated animals during and after regeneration of the L3T4+ T cell subset. The animals were injected with anti-L3T4, immunized with soluble antigen, and challenged with antigen every 2 wk. Antibody responses to two antigens, sperm whale myoglobin (SpWMb) and KLH, which differ with regard to their immunogenicity, were compared. The lack of humoral immune responsiveness to either of these two antigens shorty after anti-L3T4 treatment responsiveness to either of these two antigens shortly after anti-L3T4 treatment was probably due to clonal depletion. The anti-L3T4-induced immunosuppressive effect on antibody production seemed to be determined in part by the preexisting T cell repertoire, as was suggested by the recovery of responsiveness to the highly immunogenic antigen KLH and the transient inhibitory effect of anti-L3T4 treatment in primed animals. The regenerating L3T4+ T cell subpopulation was relatively incompetent in initiating B cell responses. More than 40% of the L3T4+ T cell compartment had to recover to provide help for the production of anti-KLH antibodies, whereas elimination of 90% of the L3T4+ helper cells did not inhibit a primary anti-KLH response. Evidence for a heterogeneous composition of the L3T4+ subset came from experiments using rIL-2 in vivo. The addition of rIL-2 during early helper cell depletion improved the recovery of the humoral responsiveness without apparently affecting the kinetics of the regeneration of L3T4+ T cells. Interestingly, humoral unresponsiveness to the weakly immunogenic antigen SpWMb persisted for at least 120 d. This long lasting unresponsiveness could not be explained by clonal depletion, and suggested as one possibility that the presence of antigen during regeneration of the L3T4+ helper cell population may have influenced the ultimate T cell repertoire.

    View details for Web of Science ID A1986D904900017

    View details for PubMedID 3091757

    View details for PubMedCentralID PMC2188395

  • THE EFFECTIVENESS OF ANTI-IA-IMMUNOTOXINS IN THE SUPPRESSION OF MLR TRANSPLANTATION NAKAHARA, K., KAPLAN, D., BJORN, M., FATHMAN, C. G. 1986; 42 (2): 205–11

    Abstract

    The potential use of a mouse anti-Ia monoclonal antibody conjugated with a plant toxin as an immunosuppressive agent for organ transplantation was examined by studying the mixed lymphocyte reaction (MLR). In initial observations in a murine system utilizing strains A/J and C57B1/6 (B6), it was shown that a rich-toxin-conjugated anti-I-Ek monoclonal antibody (13.4-immunotoxin) would kill Ia-bearing cells of strain A/J (I-Ek)--but not of strain B6, the cells of which lack I-E determinants. Studies reported here demonstrated that 13.4-immunotoxin effectively removed stimulatory cells from the I-E-bearing spleen cells but had no effect on spleen cells lacking I-E determinants or responder lymph node cells (I-E+ or I-E-) similarly treated (i.e., 1 microgram/10(6) cells). Taking advantage of the finding that such anti-I-E antibodies react with rat spleen cells bearing class II major histocompatibility complex molecules, we utilized these toxin-conjugated antibodies to inhibit MLR in rats. Assuming that the MLR is an in vitro representative of graft rejection, we suggest that such immunotoxins might be useful for pretreatment of organ allografts for transplantation across major histocompatibility barriers.

    View details for DOI 10.1097/00007890-198608000-00020

    View details for Web of Science ID A1986D627900020

    View details for PubMedID 2943065

  • GENE CONVERSION - A MECHANISM TO EXPLAIN HLA-D REGION AND DISEASE ASSOCIATION ANNALS OF THE NEW YORK ACADEMY OF SCIENCES Fathman, C. G., GORONZY, J., WEYAND, C. 1986; 475: 24-31

    Abstract

    In speculating about mechanisms that might give rise to T-cell epitopes appearing within different HLA-DR frameworks, we return to the hypothesis expressed above that suggests that gene-conversion-like events might be involved in shuffling the hypervariable segments of HLA-D region exons into alternative HLA-D region frameworks where they will still be recognized by the T cell (but not typed by conventional serology or mixed lymphocyte typing) as the "disease associated" HLA product. This might well explain the lack of stringent association between rheumatoid arthritis and HLA-DR4. It is possible, through the use of such alloreactive T-cell clones, that we might eventually define subgroups based upon presumed genetic susceptibility markers, which might allow therapeutic or prognostic assignment of patients with seropositive rheumatoid arthritis.

    View details for Web of Science ID A1986D835500003

    View details for PubMedID 2431644

  • ADMINISTRATION OF RECOMBINANT INTERLEUKIN-2 INVIVO INDUCES A POLYCLONAL IGM RESPONSE JOURNAL OF EXPERIMENTAL MEDICINE WEYAND, C. M., GORONZY, J., DALLMAN, M. J., FATHMAN, C. G. 1986; 163 (6): 1607–12

    Abstract

    We studied the potential immunoenhancing effects of high doses of rIL-2 on murine T and B cell functions in vivo. Injection of rIL-2 caused a threefold or more increase in the frequencies of antigen-specific proliferative T cells, suggesting that rIL-2 initiated a polyclonal T cell response. In primary and secondary humoral immune responses, administration of rIL-2 in vivo selectively enhanced the production of IgM antibodies, whereas the IgG response was unaffected. Coadministration of rIL-2 with antigen failed to induce an isotype switch from IgM to IgG in genetically low-responding mice. Interestingly, in mice treated with rIL-2 alone (in the absence of exogenous antigen), polyclonal IgM production was induced. Polyclonal IgM production of lesser magnitude was found when mice were immunized with specific antigen in the absence of exogenous rIL-2, suggesting that local IL-2 concentrations in a primary immune response might be sufficient to elicit a polyclonal IgM response.

    View details for DOI 10.1084/jem.163.6.1607

    View details for Web of Science ID A1986C563000021

    View details for PubMedID 3486940

    View details for PubMedCentralID PMC2188122

  • PROLIFERATION OF T-CELL CLONES REQUIRES SIGNALS IN ADDITION TO THE PERTUBATION OF THE T-CELL RECEPTOR - CD3 COMPLEX GORONZY, J., WEYAND, C. M., FATHMAN, C. G. FEDERATION AMER SOC EXP BIOL. 1986: 1118
  • SHARED T-CELL RECOGNITION SITES ON HUMAN HISTOCOMPATIBILITY LEUKOCYTE ANTIGEN CLASS-II MOLECULES OF PATIENTS WITH SEROPOSITIVE RHEUMATOID-ARTHRITIS JOURNAL OF CLINICAL INVESTIGATION GORONZY, J., Weyand, C. M., Fathman, C. G. 1986; 77 (3): 1042-1049

    Abstract

    Seropositive rheumatoid arthritis (RA) in adult and juvenile patients is associated with the serologic marker HLA-DR4. This association is incomplete; about one-third of the patients lack the disease-associated HLA-DR4 haplotype. The main biological function of class II molecules is to restrict the recognition of antigen by T lymphocytes. We therefore tested the hypothesis that patients with seropositive RA share T cell recognition sites for an unknown antigen and that such T cell "epitopes" are not identified by conventional serologic typing. We generated alloreactive human T cell clones by stimulating peripheral blood lymphocytes of normal donors against a lymphoblastoid cell line from a juvenile patient with seropositive RA. A panel of clones that recognized only HLA-Dw14 cells on a panel of homozygous typing cells was used to analyze class II molecules of adult patients with seropositive RA. By inhibition studies using monoclonal antibodies, the epitopes recognized by the different clones could be further characterized and assigned either to DR- or to DQ-encoded cell surface products. By using four different clones, it was possible to identify Dw14-associated T cell epitopes on all seropositive rheumatoid patients tested who typed HLA-DR4-positive and also on all eight DR4-negative patients tested. Approximately one-half of nonrheumatoid DR4-positive donors carried one or more determinants recognized by these clones; the expression of these allodeterminants in DR4-negative nonrheumatoid patients was rare (less than 10%). Thus, alloreactive human T cell clones are powerful tools to define T cell recognition sites on class II molecules that are not identified by conventional typing. Using T cell clones with specificities for determinants expressed on Dw14 homozygous typing lines, we were able to demonstrate shared epitopes on cells of all patients tested with seropositive RA irrespective of their HLA-D or HLA-DR type. These data suggest that major histocompatibility complex class II antigens of RA patients might be much more homogeneous than demonstrated by the incomplete HLA-DR4 association.

    View details for Web of Science ID A1986A542900051

    View details for PubMedID 2419361

    View details for PubMedCentralID PMC423516

  • IMMUNOENHANCING EFFECTS OF RECOMBINANT INTERLEUKIN-2 INVIVO WEYAND, C. M., GORONZY, J., FATHMAN, C. G. FEDERATION AMER SOC EXP BIOL. 1986: 716
  • HUMAN T-CELL CLONES USED TO DEFINE FUNCTIONAL EPITOPES ON HLA CLASS-II MOLECULES PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Weyand, C. M., GORONZY, J., Fathman, C. G. 1986; 83 (3): 762-766

    Abstract

    Polyclonal reagents have been used to define HLA class II molecules in conventional serologic and cellular typing. We generated human alloreactive T-cell clones to analyze the functional fine specificities of HLA class II molecules that might be important for the phenomenon of HLA and disease association. We chose to examine HLA-Dw14, an HLA-D specificity that has been associated with juvenile rheumatoid arthritis. In this paper we have presented data that suggest that conventional cellular typing does not reflect the distribution of T-cell epitopes on major histocompatibility complex class II molecules. We describe three alloreactive T-cell clones that have defined three separate Dw14-associated T-cell epitopes. Two of these epitopes were on a DR-region molecule; the third was located on a DQ-region product. In a panel of unrelated DR4-positive donors, these three DW14-associated determinants were present in a high frequency but were not linked to each other. Within the tested panel of DR4-positive cells, all possible combinatorial arrangements of these three allodeterminants were seen. The concurrent expression of any two of the three allodeterminants was equivalent to a positive typing response for Dw14. Our finding that HLA-Dw14 is not characterized by a unique allodeterminant but by the combinatorial recognition of independently distributed T-cell interaction sites suggests that analysis of HLA and disease association may be more clearly demonstrated through the use of human T-cell clones.

    View details for Web of Science ID A1986AZB0800050

    View details for PubMedID 2418442

    View details for PubMedCentralID PMC322945

  • 2 INDEPENDENT PATHWAYS OF HELPER ACTIVITY PROVIDED BY A SINGLE T-CELL CLONE JOURNAL OF IMMUNOLOGY Shigeta, M., Takahara, S., Knox, S. J., Ishihara, T., Vitetta, E. S., Fathman, C. G. 1986; 136 (1): 34-38

    Abstract

    Data presented in this paper demonstrate the existence of two separate pathways by which a single T cell clone can induce B cell differentiation. With the use of high doses of antigen, a T cell clone can induce a primary antibody response in unprimed B cells. With the use of low doses of antigen, the same T cell clone can induce an immunoglobulin (Ig)G response in primed B cells. The primary response is accompanied by T cell proliferation and lymphokine production (interleukin 2, B cell growth factor, B cell differentiation factor for immunoglobulin M, and B cell differentiation factor for immunoglobulin G). The secondary response does not require proliferation and occurs independently of detectable lymphokine production. Variants of the wild type T cell helper clone have been isolated. One variant can provide help to unprimed B cells when high doses of antigen are used. This variant cannot provide help to primed B cells when low doses of antigen are used, nor can it provide help to CBA/N "xid" B cells at any antigen concentration tested. Additional variants have been isolated that proliferate on antigen-pulsed-presenting cells, but fail to secrete detectable lymphokines and do not induce B cell differentiation. These results suggest that a single T cell helper clone has multiple functional activities that can be independently expressed.

    View details for Web of Science ID A1986AWM4600007

    View details for PubMedID 2933466

  • ONE-I REGION RESTRICTION DETERMINANT CAN ASSOCIATE WITH MULTIPLE ANTIGENIC EPITOPES MICROBIOLOGY AND IMMUNOLOGY Kimoto, M., Fathman, C. G. 1986; 30 (2): 133-142

    Abstract

    Studies presented in this paper show that T cell clones recognizing different epitopes of multideterminant antigens can be restricted by the same I-A molecule. These data further support the concept that a single I-A restriction site can present more than one antigenic epitope. This concept was supported by data on the proliferation of T cell clones reactive with either poly(L-Glu60, L-Ala30, L-Tyr10)n(GAT) or poly(Tyr, Glu)-poly D,L-Ala--poly Lys [(T,G)-A--L] which recognized different epitopes on these multideterminant antigens. Two clones recognizing different epitopes on the same multideterminant antigen can be blocked by the same monoclonal anti-I-A antibody. Additionally, the mutation in the Abm12 chain utilized in [B6.C-H-2bm12(bm12) X B10.A(4R)]F1 mice can affect the restriction determinant of clones recognizing different antigenic epitopes. These results suggest that in the strictest sense, the determinant selection theory is not tenable and would support the concept that T cell specificity is controlled by the T cell repertoire.

    View details for Web of Science ID A1986A374800005

    View details for PubMedID 2423856

  • IL-2 RESTORES MEMORY B-CELL ACTIVATION BY ANTIGEN-SPECIFIC T-CELL CLONE VARIANTS JOURNAL OF IMMUNOLOGY Ishihara, T., Takahara, S., Fathman, C. G. 1986; 136 (1): 39-43

    Abstract

    It has recently been demonstrated that there are at least two separate pathways by which a single keyhole limpet hemocyanin (KLH) reactive T cell clone can induce B cell differentiation. With the use of the high-dose antigen-driven system (10 micrograms/ml trinitrophenyl (TNP)-KLH), a KLH-specific T cell clone was able to induce a primary anti-TNP response in unprimed B cells. In the presence of aliquots of the same T cell clone, a low-dose of antigen (5 X 10(-2) micrograms/ml TNP-KLH) induced an immunoglobulin (Ig)G response in primed B cells. It has also been demonstrated that there are variant subclones of such KLH-specific helper T cell clones that are unable to provide antigen-specific help in the presence of low-dose antigen but maintain the high-dose antigen-driven helper response. This study was undertaken to investigate whether interleukin 2 (IL 2) had some activity in the low-dose, antigen-driven response induced by the T cell clone. With the use of a variant T cell clone (which lost low-dose, antigen-driven helper activity), it was demonstrated that IL 2 was capable of reconstituting the low-dose, antigen-driven helper activity. To investigate whether accessory cells were required in this system, we removed the adherent cell population from the primed spleen cells added to culture. Interestingly, removal of the G10-adherent cells eliminated the low-dose, antigen-driven response induced by IL 2. Additionally by add-back experiments, we were able to demonstrate that the necessary adherent cell population did not require major histocompatibility complex (MHC) restriction for reconstitution of the IL 2-dependent, low-dose, antigen-driven response. Furthermore, 1% concanavalin A (Con A) supernatant (Sn), but not interleukin 1 (IL 1), could replace this adherent cell function. These data suggest that in this system, IL 2 bypasses the MHC-restricted interaction between T cells and antigen-charged adherent cells; B cells can present antigen to cloned helper T cells efficiently for primary responses but need an added factor(s) to induce IgG production; and adherent cells are essential for IgG production in primed B cells, possibly through the release of soluble factor(s) included in Con A Sn.

    View details for Web of Science ID A1986AWM4600008

    View details for PubMedID 2415626

  • HUMAN T-CELL CLONES AS DIAGNOSTIC-TOOLS AND POTENTIAL THERAPEUTIC REAGENTS IMMUNOLOGICAL INVESTIGATIONS WEYAND, C., GORONZY, J., Fathman, C. G. 1986; 15 (7): 591-624

    View details for Web of Science ID A1986F292100001

    View details for PubMedID 2433217

  • An analysis of monoclonal T cell and antibody recognition sites on Ia molecules. journal of investigative dermatology Frelinger, J. G., Shigeta, M., Fathman, C. G. 1985; 85 (1): 30s-33s

    Abstract

    The advances made in understanding T cell and antibody recognition sites on Ia using monoclonal helper and alloreactive T cells are summarized. For many antibodies it has been possible to determine whether the antibody recognition site was determined by the alpha or beta chain. Such defined antibody reagents have allowed the definition of multiple functional antigen presenting sites on a given Ia molecule. Mutant antigen-presenting cells independently suggest the existence of such multiple functional sites. A detailed analysis of the I-Ab mutant bm 12 directly defines the chemical nature of one such site and suggests that it arose as a result of gene conversion. Such regions of Ia molecules may be important for both T-cell function and antibody-binding.

    View details for PubMedID 3874247

  • HUMAN T-CELL CLONES EXPRESS FUNCTIONAL HOMING RECEPTORS REQUIRED FOR NORMAL LYMPHOCYTE TRAFFICKING JOURNAL OF EXPERIMENTAL MEDICINE NAVARRO, R. F., JALKANEN, S. T., HSU, M., SOENDERSTRUPHANSEN, G., GORONZY, J., WEYAND, C., Fathman, C. G., CLAYBERGER, C., KRENSKY, A. M., BUTCHER, E. C. 1985; 162 (3): 1075-1080

    Abstract

    To function efficiently in vivo, lymphocytes must circulate from the blood into lymphoid tissues and other sites of immune reaction. Herein, we show that human cytotoxic and helper T cell clones and lines, maintained in vitro with IL-2, express the functional capacity to recognize and bind to high endothelial venules (HEV), a capacity essential for lymphocyte exit from the blood, and hence for normal lymphocyte trafficking. The expression of functional homing receptors distinguishes human T cell clones from their murine counterparts, which uniformly lack receptors for HEV and are unable to migrate normally from the blood in vivo. The results raise the possibility that human T cell clones may be more effective in mediating in vivo immune responses than is suggested by murine models.

    View details for Web of Science ID A1985AQH0300021

    View details for PubMedID 3875680

    View details for PubMedCentralID PMC2187804

  • SHARED HLA - CLASS-II ANTIGENS IN JUVENILE AND ADULT RHEUMATOID-ARTHRITIS PATIENTS DEFINED BY HUMAN T-CELL CLONES WEYAND, K., GORONZY, J., FATHMAN, C. G. FEDERATION AMER SOC EXP BIOL. 1985: 1178
  • DISSECTION OF THE DW14 SPECIFICITY BY HUMAN T-CELL CLONES GORONZY, J., WEYAND, K., HUNT, T., FATHMAN, C. G. FEDERATION AMER SOC EXP BIOL. 1985: 942
  • SUBSETTING OF ADULT RHEUMATOID-ARTHRITIS BY HUMAN T-CELL CLONES REACTIVE AGAINST HLA CLASS-II DETERMINANTS GORONZY, J., WEYAND, C. M., FATHMAN, C. G. SLACK INC. 1985: A377
  • THE EFFECTIVENESS OF ANTI-IA-IMMUNOTOXINS IN THE SUPPRESSION OF MLR TRANSPLANTATION NAKAHARA, K., KAPLAN, D., BJORN, M., FATHMAN, C. G. 1985; 40 (1): 62–67

    Abstract

    The potential use of a mouse anti-Ia monoclonal antibody conjugated with a plant toxin as an immunosuppressive agent for organ transplantation was examined by studying the mixed lymphocyte reaction (MLR). In initial observations in a murine system utilizing strains A/J and C57B1/6 (B6), it was shown that a rich-toxin-conjugated anti-I-Ek monoclonal antibody (13.4-immunotoxin) would kill Ia-bearing cells of strain A/J (I-Ek)--but not of strain B6, the cells of which lack I-E determinants. Studies reported here demonstrated that 13.4-immunotoxin effectively removed stimulatory cells from the I-E-bearing spleen cells but had no effect on spleen cells lacking I-E determinants or responder lymph node cells (I-E+ or I-E-) similarly treated (i.e., 1 micrograms/10(6) cells). Taking advantage of the finding that such anti-I-E antibodies react with rat spleen cells bearing class II major histocompatibility complex molecules, we utilized these toxin-conjugated antibodies to inhibit MLR in rats. Assuming that the MLR is an in vitro representative of graft rejection, we suggest that such immunotoxins might be useful for pretreatment of organ allografts for transplantation across major histocompatibility barriers.

    View details for DOI 10.1097/00007890-198507000-00013

    View details for Web of Science ID A1985ALX8300013

    View details for PubMedID 3160144

  • SUBSETTING OF ADULT RHEUMATOID-ARTHRITIS BY HUMAN T-CELL CLONES REACTIVE AGAINST HLA CLASS II DETERMINANTS GORONZY, J., WEYAND, C. M., FATHMAN, C. G. SLACK INC. 1985: A591
  • LONG-TERM UNRESPONSIVENESS TO A SOLUBLE-ANTIGEN AFTER INVIVO TREATMENT WITH A MONOCLONAL-ANTIBODY AGAINST L3/T4 GORONZY, J., WEYAND, C. M., FATHMAN, C. G. GUSTAV FISCHER VERLAG. 1985: 24
  • ANALYSIS OF DW14 SPECIFICITIES IN CHILDREN WITH SEROPOSITIVE JUVENILE ARTHRITIS BY HUMAN T-CELL CLONES WEYAND, C. M., GORONZY, J., FATHMAN, C. G. SLACK INC. 1985: A513
  • ANALYSIS OF DW14 SPECIFICITIES IN CHILDREN WITH SEROPOSITIVE JUVENILE ARTHRITIS BY HUMAN T-CELL CLONES WEYAND, C. M., GORONZY, J., FATHMAN, C. G. SLACK INC. 1985: A593
  • ANALYSIS OF MHC CLASS-II MOLECULES BY HUMAN T-CELL CLONES IN AN HLA-DR4 ASSOCIATED DISEASE, SEROPOSITIVE RHEUMATOID-ARTHRITIS WEYAND, C. M., GORONZY, J., FATHMAN, C. G. GUSTAV FISCHER VERLAG. 1985: 106–7
  • INVIVO TREATMENT WITH INTERLEUKIN-2 - EVIDENCE FOR POLYCLONAL B-CELL ACTIVATION WEYAND, C. M., GORONZY, J., FATHMAN, C. G. GUSTAV FISCHER VERLAG. 1985: 107
  • TYPE-II COLLAGEN-REACTIVE T-CELL CLONES FROM MICE WITH COLLAGEN-INDUCED ARTHRITIS JOURNAL OF IMMUNOLOGY Dallman, M., Fathman, C. G. 1985; 135 (2): 1113-1118

    Abstract

    Certain strains of mice develop a symmetrical polyarthritis after immunization with type II collagen. The incidence of arthritis after such immunization is variable. To study the arthritogenic potential of T cells reactive with type II collagen, we isolated draining lymph node cells from mice that had developed arthritis after immunization with bovine type II collagen. From these immune lymph node cells we were able to clone T cells reactive with type II collagen. Two separate sets of T cell clones were isolated. The first set reacted with either native bovine or native chick type II collagen, but did not react with type I collagen. The second set of T cell clones reacted with bovine type II collagen, but did not respond to either native chick type II collagen or type I collagen. These clones will be tested for their influence on the development of arthritis in vivo.

    View details for Web of Science ID A1985AQP9100041

    View details for PubMedID 2409139

  • INTERFERON-GAMMA PRODUCTION BY HERPES-SIMPLEX VIRUS ANTIGEN-SPECIFIC T-CELL CLONES FROM PATIENTS WITH RECURRENT HERPES LABIALIS JOURNAL OF GENERAL VIROLOGY Cunningham, A. L., Nelson, P. A., Fathman, C. G., Merigan, T. C. 1985; 66 (FEB): 249-258

    Abstract

    Nineteen herpes simplex virus (HSV) antigen-specific human T lymphocyte clones were established from three volunteers with recent recurrent herpes labialis. All produced interferon gamma (IFN-gamma) at titres of 200 to 700 units/ml when cultured in vitro with HSV antigen and irradiated peripheral blood mononuclear cells (PBMC) as filler cells. All 10 of those clones whose phenotype was determined were Leu 4+, Leu 2-, Leu 3+. Interleukin 2 alone failed to induce IFN-gamma in titres greater than 10 units/ml from these clones cultured at 10(4)/0.2 ml/well. However, the effect of different accessory or filler cells on IFN-gamma production by clones was quite marked. For example, high titres were produced when irradiated PBMC or plastic-adherent cells (predominantly monocytes) were added and low titres when macrophages and irradiated Epstein-Barr virus-transformed B (EBV-B) cells were added. When tested for HSV antigen-stimulated IFN production alone, the irradiated PBMC and adherent cells produced low titres, but no detectable interferon was produced by the others. However, with higher concentrations of EBV-B cells, low concentrations of IFN-alpha were occasionally produced. Irradiation strikingly reduced IFN-alpha-production by PBMC. The IFN-alpha and -gamma produced by accessory cells may contribute to total IFN production by priming the production by cloned cells, and acting in synergy with IFN-gamma produced by the cloned cells. Alternatively, the effect may be due to the presence of permissive concentrations of other lymphokines such as the interleukins. Interferon production by cloned T lymphocytes in the presence of non-producing macrophages was maximal within 24 h, much faster than with a similar polyclonal system, although attaining lower titres. EBV-B cells from only one of three patients supported antigen-specific lymphocyte activation. Almost all cells of the three cell lines expressed DR antigens, while DS/DC antigens were also expressed on nearly all cells of the antigen-presenting line and, at lower densities, on two-thirds of the cells of the other two lines.

    View details for Web of Science ID A1985ACN1300004

    View details for PubMedID 2981967

  • AN ANALYSIS OF MONOCLONAL T-CELL AND ANTIBODY RECOGNITION SITES ON IA MOLECULES JOURNAL OF INVESTIGATIVE DERMATOLOGY Frelinger, J. G., Shigeta, M., Fathman, C. G. 1985; 85 (1): S30-S33

    Abstract

    The advances made in understanding T cell and antibody recognition sites on Ia using monoclonal helper and alloreactive T cells are summarized. For many antibodies it has been possible to determine whether the antibody recognition site was determined by the alpha or beta chain. Such defined antibody reagents have allowed the definition of multiple functional antigen presenting sites on a given Ia molecule. Mutant antigen-presenting cells independently suggest the existence of such multiple functional sites. A detailed analysis of the I-Ab mutant bm 12 directly defines the chemical nature of one such site and suggests that it arose as a result of gene conversion. Such regions of Ia molecules may be important for both T-cell function and antibody-binding.

    View details for Web of Science ID A1985AME4400007

  • THE EFFECTS OF AUTACOIDS ON CLONED MURINE LYMPHOID-CELLS - MODULATION OF IL-2 SECRETION AND THE ACTIVITY OF NATURAL SUPPRESSOR CELLS JOURNAL OF IMMUNOLOGY Khan, M. M., Melmon, K. L., Fathman, C. G., HERTELWULFF, B., Strober, S. 1985; 134 (6): 4100-4106

    Abstract

    The pharmacologic effects of histamine and isoproterenol (autacoids) were studied on clones of murine T helper (TH) and natural suppressor (NS) cells. The data are consistent with receptors for the autacoids being nonrandomly distributed on phenotypically and functionally distinct clones. The effects of histamine on IL 2 secretion by TH cells could be either inhibitory or stimulatory, depending on the conditions of incubation with the autacoid. When TH cells were pretreated with histamine, their secretion of IL 2 was augmented; conversely, if histamine was added to the TH cells in the presence of antigen, IL 2 secretion was inhibited. The suppressor function of NS cells was enhanced by preincubation with histamine (10(-4) M) for 4 hr before the washed NS cells were added to responder and stimulator cells in mixed lymphocyte cultures. Two H1 receptor antagonists, mepyramine (10(-6) M) and pyrobutamine (10(-7) M), each competitively blocked the histamine activation of the NS cells, but an H2 receptor antagonist cimetidine (10(-5) M) did not alter the suppressor-enhancing function of histamine. Activation of NS cells did not occur if histamine was added with responder, stimulator, and co-cultured cells in MLR. The effects of each autacoid were additive in cytolytic T cells alone. The adenylate cyclase pools that can be stimulated by isoproterenol and histamine in cytolytic T cells may be independent of each other, but further work will be needed to prove this point.

    View details for Web of Science ID A1985AHR7300086

    View details for PubMedID 2859336

  • MULTIPLE FUNCTIONAL SITES ON A SINGLE IA MOLECULE DEFINED USING T-CELL CLONES AND ANTIBODIES WITH CHAIN-DETERMINED SPECIFICITY JOURNAL OF EXPERIMENTAL MEDICINE Frelinger, J. G., Shigeta, M., Infante, A. J., Nelson, P. A., PIERRES, M., Fathman, C. G. 1984; 159 (3): 704-715

    Abstract

    Monoclonal antibodies (mAb) were used to inhibit the proliferation of antigen-reactive (C57BL6/J X A/J)F1 restricted T cell clones. We have been able to subdivide these F1 restricted T cell clones into two groups: one of which recognizes the A alpha k A beta b molecule and the other group which recognizes the A alpha b A beta k molecule. Using clones with defined reactivities, we could assign the reactivities of monoclonals to the A alpha or A beta chains. By immunoprecipitation and two-dimensional analysis of Ia molecules from F1 spleen cells, we could independently map the reactivities of the mAb as being determined by the A alpha or A beta chain. To date, these two methods of chain localization of the antibody reactivity have agreed. Further, the differential blocking of the A alpha k A beta b restricted T cell clones suggests that there exists more than one restriction site per Ia molecule. Increasing the number of possible functional Ia restriction sites, either through combinatorial association of alpha and beta chains or by using more than one site per molecule, should increase the number of ways Ia molecules can function in antigen presentation.

    View details for Web of Science ID A1984SK46700005

    View details for PubMedID 6421980

    View details for PubMedCentralID PMC2187252

  • INTRATHYMIC PRESENTATION OF CIRCULATING NON-MAJOR HISTOCOMPATIBILITY COMPLEX ANTIGENS NATURE KYEWSKI, B. A., FATHMAN, C. G., KAPLAN, H. S. 1984; 308 (5955): 196–99

    Abstract

    Intrathymic selection of T-cell specificity has been shown to be influenced by self-major histocompatibility complex (MHC) antigens encoded by radioresistant thymic stromal cells. The role of non-MHC antigens in intrathymic T-cell differentiation, in particular induction of antigen-specific tolerance, has been unclear and the access of non-MHC antigens to the thymus is controversial. Here we present evidence that circulating protein antigens enter the thymus and are presented by thymic stromal cells. At least three distinct types of stromal cells are thought to be associated with intrathymic lymphopoiesis; after intravenous (i.v.) injection of antigen only I-A/E-positive medullary dendritic cells, but not I-A/E-negative macrophages or I-A/E-positive cortical epithelial cells co-purified with antigen-specific stimulation of cloned T-helper cells in vitro. Antigen presentation by thymic stromal cells was dependent on the dose of antigen injected and the time interval after injection.

    View details for DOI 10.1038/308196a0

    View details for Web of Science ID A1984SG11900164

    View details for PubMedID 6608055

  • THE CONUNDRUM OF IMMUNE-RESPONSE - 2 IA LOCI SPECIFICALLY CONTROL MULTIPLE IMMUNE-RESPONSES ANNALES D IMMUNOLOGIE Fathman, C. G. 1984; C135 (3): 420-423
  • GENE CONVERSION BETWEEN MURINE CLASS-II MAJOR HISTOCOMPATIBILITY COMPLEX LOCI - FUNCTIONAL AND MOLECULAR EVIDENCE FROM THE BM12 MUTANT JOURNAL OF EXPERIMENTAL MEDICINE MENGLEGAW, L., Conner, S., McDevitt, H. O., Fathman, C. G. 1984; 160 (4): 1184-1194

    Abstract

    The experiments presented in this study define the molecular basis of the bm 12 mutation. Initial characterization of an alloreactive T cell clone, 4.1.4, showed this clone to recognize an allodeterminant present on the E beta b and A beta bm12 chains, but not on the bm 12 parent A beta b chain. To define the extent of sequence shared between the I-E beta product and the mutant I-A beta product, we isolated a cDNA clone of the E beta b gene and determined its nucleotide sequence. Comparison of the nucleotide sequences of E beta b, A beta b, and A beta bm12 shows the the A beta bm12 gene to be identical to the E beta b gene in the region where it differs from its A beta b parent. We predict that the bm 12 mutation arose by gene conversion of this region, which spans 14 nucleotides between amino acid residues 67-71 of the mature A beta chain, from the E beta b locus to the corresponding position at the A beta b locus. Recognition of this region, which spans one of the previously defined E beta allelic "hypervariable" regions, by an alloreactive T cell clone provides the first direct evidence of the functional importance of these hypervariable regions in T cell stimulation. The identification of a gene conversion event involving one of these allelic variable regions implicates conversion as a mechanism that acts on class II beta genes to create sequence diversity in regions of Ia molecules that interact with foreign antigen or a T cell receptor, regions where protein sequence polymorphism would presumably be selected for by the expanded ability it affords the organism to mount effective immune responses against a wider variety of foreign antigens.

    View details for Web of Science ID A1984TM66100016

    View details for PubMedID 6434690

    View details for PubMedCentralID PMC2187482

  • CLONOTYPIC ANTIBODIES WHICH STIMULATE T-CELL CLONE PROLIFERATION IMMUNOLOGICAL REVIEWS Frelinger, J., Sing, A., INFANTE, A., Fathman, C. G. 1984; 81: 21-38

    Abstract

    We have prepared a variety of antisera which recognize the T cell receptors on both alloreactive and antigen-specific functional clones. The antisera specifically stimulate the immunizing T cell clones to proliferate without cross-stimulating a variety of other clones and are thus clonotypic. The antisera specifically precipitate an 82k molecular weight complex unique to the immunizing clone and an independent common component present on all the helper T cell clones tested. The 82k molecular weight complex consists of a heterodimer, which can be resolved into an acidic (a) chain and a more basic (beta) chain. The beta subunit differs in its isoelectric focusing pattern when isolated from two A derived clones which recognize and respond to different alloantigens. Thus, the functional specificity of the antibodies is reflected in identifiable changes in molecules at the biochemical level.

    View details for Web of Science ID A1984TT81400002

    View details for PubMedID 6334643

  • ARSONATE-SPECIFIC MURINE T-CELL CLONES .1. GENETIC-CONTROL AND ANTIGEN-SPECIFICITY JOURNAL OF EXPERIMENTAL MEDICINE HERTELWULFF, B., Goodman, J. W., Fathman, C. G., Lewis, G. K. 1983; 157 (3): 987-997

    Abstract

    The antigen-induced proliferative response of lymph node cells (LNC) from mice sensitized to the monofunctional antigen L-tyrosine-p-azobenzenearsonate (ABA-Tyr) was used to monitor genetic control. All strains tested mounted significant responses, but those that were H-2(b) at both the I-A and I-E loci [B10., B6., B10.A(18R), A.BY, and C3H.SW] gave consistently weaker responses than other haplotypes. The F(1) progeny of matings between high and low responder phenotype parents (DBA/2 and B6, respectively) were high responders, establishing the dominance of the responder trait. Proliferative responses of LNC to ABA-Tyr were blocked by the appropriate anti-Ia monoclonal reagents. For example, B10.A(4R) LNCI (I-A(k), I-E(b)) were blocked by anti-I-A(k), whereas B10.A(3R) LNC (I-A(b), I-E(k)) were blocked by anti-I-E(k). Long-term cultures of T cell lines specifically reactive to ABA-Tyr were established from LNC of A/J mice immunized with ABA-Tyr and were cloned by limiting dilution. The proliferative responses to ABA-Tyr of 14 out of 15 clones tested were I-A restricted on the basis of activation by antigen-presenting cells from appropriate recombinant strains and the blocking activity of the monoclonal anti-Ia antibodies. The response of the other clone was I-E restricted. The fine antigen specificity of the clones was studied using structural analogs of the homologous antigen to induce proliferation. The clones could be divided into three types with respect to responsiveness to ABA-histidine (ABA-His). One group responded about equally well to ABA-His and ABA-Tyr. A second set responded less strongly to ABA-His than to ABA-Tyr, while the third showed no response above background to ABA- His. In all instances, the ABA-His-responding clones discriminated exquisitely between the 2-azo and 4-azo histidine isomers, responding only to the 4-azo compound. These T cell clones provide extremely useful tools for studies of T cell specificity, antigen recognition and lymphoid cell interaction systems.

    View details for Web of Science ID A1983QH26700014

    View details for PubMedID 6187883

    View details for PubMedCentralID PMC2186958

  • LYMPHOCYTE-T CLONES ANNUAL REVIEW OF IMMUNOLOGY Fathman, C. G., Frelinger, J. G. 1983; 1: 633-655

    Abstract

    To date, the most successful uses of T-cell clones have been in the demonstration that a single type of cell can perform multiple functions. However, their potential usefulness is enormous, and the study of cell interactions using clonal populations has just begun. The development and study of more cloned populations will surely lead to a clearer analysis of cellular interactions in the immune system. The use of T-cell clones and hybridomas to analyze T-cell receptors and/or factors is well under way, and will continue to be an area of intense investigation. Molecular biologists will undoubtedly make more extensive use of T-cell clones in the future, both as a source of cloning material and as transfection recipients. The most exciting area for development, from a medical point of view, is the potential for use of these cell lines or their products in immunotherapy and in providing a mechanism for specifically modulating the immune response.

    View details for Web of Science ID A1983QP21000022

    View details for PubMedID 6242467

  • THE I-AB MUTANT B6.C-H-2BM12 ALLOWS DEFINITION OF MULTIPLE T-CELL EPITOPES ON I-A MOLECULES JOURNAL OF EXPERIMENTAL MEDICINE BECK, B. N., Nelson, P. A., Fathman, C. G. 1983; 157 (5): 1396-1404

    Abstract

    The experiments presented in this study demonstrate that there exist at least two functional epitopes on an I-A molecule that can be recognized by T cell clones. By comparing the abilities of spleen cells from C57BL/6 mice and the congenic I-A mutant line B6.C-H-2bm12 to stimulate alloreactive T cell clones specific for the I-Ab molecule, we have discriminated two sets of clones, those recognizing the I-Ab and I-Abm12 molecule equally well and those able to recognize only the I-Ab molecule. These results imply that the two sets of clones have different receptors for I-A and that they therefore recognize separate epitopes on the I-A molecule. We have similarly been able to separate T cell clones, both alloreactive and L-glutamic acid60-L-alanine30-L-tyrosine10-reactive, specific for the Ab alpha Ak beta hybrid molecule into two groups based on their ability to recognize bm 12 spleen cells. Although the recognition of bm 12 spleen cells by these clones was unexpected since none of them responds to B6 spleen cells, these data again allow us to conclude that these groups of clones have different receptors for the same I-A molecule and therefore that they recognize distinct epitopes on the molecule. Additional studies, in which monoclonal anti-I-A antibodies were used to block the stimulation of T cells by stimulator or antigen-presenting cells, have demonstrated that this blockade can be a steric effect and therefore is not necessarily indicative of direct competition between the antibody and the T cell for the same site on an I-A molecule. Although this study does not reveal the physical nature of an I region-controlled "antigen-restriction site," we can suggest that increasing the number of possible functional Ia restriction sites either through combinatorial association of alpha and beta chains or by using more than one site per molecule will increase the number of configurations the ternary complex of Ia, antigen and T cell receptor(s) can form.

    View details for Web of Science ID A1983QQ38700003

    View details for PubMedID 6189934

    View details for PubMedCentralID PMC2187021

  • ABNORMAL MIGRATION OF LYMPHOCYTE-T CLONES JOURNAL OF IMMUNOLOGY Dailey, M. O., Fathman, C. G., BUTCHER, E. C., Pillemer, E., Weissman, I. 1982; 128 (5): 2134-2136

    Abstract

    Several in vitro T cell clones were markedly deficient in their ability to home to peripheral lymphoid tissue. This was found for an alloreactive noncytolytic clone, a soluble antigen- (KLH)specific line, and cytotoxic clones specific for allogeneic cells and for Abelson virus-induced lymphoma cells. This abnormal circulation pattern was probably caused by the lack of the receptors of the lymphocytes for high endothelial venules (HEV), as implied by the lack of binding of these T cells to HEV in frozen sections of mouse lymph node and Peyer's patches. The loss of surface receptors that are necessary for normal lymphocyte migration may thereby alter the in vivo function of adoptively transferred T cells.

    View details for Web of Science ID A1982NL17700035

    View details for PubMedID 6460817

  • ARSONATE-SPECIFIC MHC-RESTRICTED MURINE T-CELL CLONES HERTELWULFF, B., FATHMAN, C. G., GOODMAN, J. W., LEWIS, G. K. GUSTAV FISCHER VERLAG. 1982: 139–40
  • MYOGLOBIN-REACTIVE T-CELL CLONES ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY Infante, A. J., Atassi, M. Z., Fathman, C. G. 1982; 150: 159-167

    View details for Web of Science ID A1982PJ77700010

    View details for PubMedID 6183942

  • ROLE OF THE MAJOR HISTOCOMPATIBILITY COMPLEX IN T-CELL ACTIVATION OF B-CELL SUB-POPULATIONS - A SINGLE MONOCLONAL T-HELPER CELL-POPULATION ACTIVATES DIFFERENT B-CELL SUB-POPULATIONS BY DISTINCT PATHWAYS JOURNAL OF EXPERIMENTAL MEDICINE Asano, Y., Shigeta, M., Fathman, C. G., Singer, A., Hodes, R. J. 1982; 156 (2): 350-360

    Abstract

    It has recently been demonstrated that the Lyb-5+ and Lyb-5- B cell subpopulations differ in their requirements for major histocompatibility complex (MHC)-restricted activation by T helper (TH) cells. To determine whether these MHC-restricted and -unrestricted pathways of B cell activation result from differences in the participating TH cell populations or reflect differences exclusively in the responding B cell subpopulations, experiments were carried out using cloned TH cells for in vitro antibody responses to trinitrophenyl-keyhole limpet hemocyanin. The same cloned T helper cells were able to activate both CBA/N (Lyb-5-) B cells and CBA/CaHN (Lyb-5+ + Lyb-5-) B cells under different experimental conditions. The activation of Lyb-5-B cells by cloned T helper cells required both MHC-restricted TH cell-B cell interaction and carrier-hapten linkage. In contrast, the activation of Lyb-5+ B cells required only MHC-restricted T helper cell interaction with accessory cells, while T-B interaction was MHC unrestricted and did not require carrier-hapten linkage. Thus, the differences in activation requirements observed for the Lyb-5- and Lyb-5+ B cell subsets do not result from differences in the TH cell populations activating these B cells, but rather reflect differences in the ability of these B cells to respond to signals from the same TH cells.

    View details for Web of Science ID A1982PB10900003

    View details for PubMedID 6980253

    View details for PubMedCentralID PMC2186748

  • T-CELL CLONES SPECIFIC FOR HYBRID I-A MOLECULES - DISCRIMINATION WITH MONOCLONAL ANTI-I-AK ANTIBODIES JOURNAL OF EXPERIMENTAL MEDICINE BECK, B. N., Frelinger, J. G., Shigeta, M., Infante, A. J., Cummings, D., Hammerling, G., Fathman, C. G. 1982; 156 (4): 1186-1194

    Abstract

    Alloreactive and soluble antigen-reactive, I-A-restricted T cell clones were examined for their ability to recognize hybrid I-A antigens. Several clones that recognized hybrid I-A(b)/I-A(k) molecules on (C57BL/6 x A/J)F(1) [(B6A)F(1)] spleen cells were studied. We were able to distinguish clones that recognized hybrid I-A molecules of the A(b)(a)A(k)(beta) type from those that recognized A(k)(a)A(b)(beta) molecules. We reached this conclusion by considering data from three independent types of experiments. (a) Monoclonal antibodies were used to inhibit T cell stimulation. Antibodies 10.2.16 and H116.32 distinguished two mutually exclusive "families" of T cell clones. One group of clones was inhibited by 10-2.16 and not H116.32, the other group exhibited reciprocal inhibition. (b) T cell proliferation was assayed using antigen-presenting cells from B6.C-H-2(bml2) (bml2) and [bml2 x B10.A(4R)]F(1) mice. Because the bml2 strain has a mutation that results in an altered A(b)(beta) polypeptide chain (A(bm12)(beta)), we reasoned that clones that could recognize the [bm12 x B 10.A(4R)]F(1) cells were recognizing A(b)(a)A(k)(beta) molecules. Alternatively, clones not recognizing [bml2 x B10.A(4R)]F(1) cells had specificity for A(k)(a)A(b)(beta) molecules. (c) I-A molecules immunoprecipitated from radiolabeled (B6A)F(1) splenocyte extracts were analyzed by two-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis. These experiments confirmed an earlier report that antibody 10.2.16 recognized determinants on the A(k)(beta) chain (12). Antibody H116.32 immunoprecipitated products consistent with recognition of A(k)(a) determinants. Taken together, these three types of results offer conclusive evidence that T cell clones recognizing "hybrid" I-A molecules use either A(b(k)A(k)(beta) or A(k)(a)A(b)(beta) molecules as recognition or restriction sites. Clones whose proliferation was supported by [bm 12 x B10.A(4R)]F(1) cells and blocked by anti-I-A(k) antibody 10-2.16 recognized A(b)(a)A(k)(beta) B molecules. Clones that were blocked by antibody H116.32 and did not recognize [bml2 X B10.A(4R)]F(1) cells use a recognition site(s) on A(b)(a)A(k)(beta) molecules. Thus, we can demonstrate both functionally and biochemically that hybrid F(1) I-A molecules of the structure A(k)(a)A(b)(beta) and A(b)(a)A(k)(beta) both exist on (B6A)F(1) splenocytes and that both configurations are used in immune recognition phenomena.

    View details for Web of Science ID A1982PP46100019

    View details for PubMedID 6185607

    View details for PubMedCentralID PMC2186826

  • DEFINITION OF T-CELL IDIOTYPES USING ANTI-IDIOTYPIC ANTISERA PRODUCED BY IMMUNIZATION WITH T-CELL CLONES JOURNAL OF EXPERIMENTAL MEDICINE Infante, A. J., Infante, P. D., Gillis, S., Fathman, C. G. 1982; 155 (4): 1100-1107

    Abstract

    Alloreactive T cell clones with distinct specificities were used to raise anti-idiotypic antisera via an F1 anti-(parent anti-F1) protocol. Antisera were raised that could stimulate the proliferation of the appropriate T cell clone, but not other clones. The active fraction of the antisera for T cell proliferation was immunoglobulin. In addition to proliferation, an anti-idiotypic antiserum could induce the appropriate T cell clone to secrete substantial amounts of interleukin 2 (IL-2). Production of IL-2 appeared independent of the involvement of accessory cells. These accessory cells may be unnecessary for IL-2 production in our assay, or their effect may be produced by anti-idiotype. Thus, anti-idiotype may provide two or more specific T cell signals.

    View details for Web of Science ID A1982NH42800013

    View details for PubMedID 6174671

    View details for PubMedCentralID PMC2186651

  • FUNCTIONAL-STUDIES OF IMMUNE-RESPONSE UTILIZING MURINE T-CELL CLONES UCLA SYMPOSIA ON MOLECULAR AND CELLULAR BIOLOGY Fathman, C. G., Asano, Y., Infante, A. J., Shigeta, M., Nelson, P., Frelinger, J., Kimoto, M., Singer, A., Hodes, R. 1982; 24: 97-106
  • FUNCTIONAL SIMILARITIES OF AEE-ALPHA IA MOLECULES AS DETERMINED BY ANALYSIS WITH T-CELL CLONES IMMUNOGENETICS BECK, B. N., Infante, A. J., Fathman, C. G. 1982; 15 (6): 565-571

    Abstract

    Recognition of AeE alpha Ia antigens at the functional level was investigated using T-cell clones. The reactivities of an alloreactive and an antigen-reactive clone, both of which recognized AeE alpha Ia molecules, were compared on a panel of stimulator/antigen-presenting cells of various genotypes. The two clones recognized all tested AebE alpha x Ia molecules, where x is a haplotype capable of expressing an Ia.7-bearing E alpha polypeptide. Ia antigen recognition by either clone could be inhibited by the monoclonal antibody Y-17, which recognizes a combinatorial serologic determinant on certain AeE alpha molecules. There were no differences in the recognition of Ia by the alloreactive versus the antigen-reactive clone, suggesting that Ia antigens are recognized by the two clones in a fundamentally similar way. The recognition of these various Ia molecules by the two cloned T-cell lines provides evidence that the E alpha polypeptides from H-2 haplotypes k, d, r, and u are functionally indistinguishable.

    View details for Web of Science ID A1982NW25200003

    View details for PubMedID 6179865

  • DISTINCT B-CELL SUB-POPULATIONS DIFFER IN THEIR GENETIC REQUIREMENTS FOR ACTIVATION BY T-HELPER CELLS IMMUNOLOGICAL REVIEWS Singer, A., Asano, Y., Shigeta, M., Hathcock, K. S., Ahmed, A., Fathman, C. G., Hodes, R. J. 1982; 64: 137-160

    View details for Web of Science ID A1982NM58100007

    View details for PubMedID 6806171

  • ROLE OF THE MAJOR HISTOCOMPATIBILITY COMPLEX IN T-CELL ACTIVATION OF B-CELL SUB-POPULATIONS - ANTIGEN-SPECIFIC AND H-2-RESTRICTED MONOCLONAL TH-CELLS ACTIVATE LYB-5+ B-CELLS THROUGH AN ANTIGEN-NONSPECIFIC AND H-2-UNRESTRICTED EFFECTOR PATHWAY JOURNAL OF IMMUNOLOGY Hodes, R. J., Shigeta, M., Hathcock, K. S., Fathman, C. G., Singer, A. 1982; 129 (1): 267-271

    Abstract

    Monoclonal T helper (TH) cell populations were employed to study the mechanism of activation of the Lyb-5+ B cell subpopulation in T cell-dependent antibody responses in vitro. It was demonstrated that monoclonal T cell populations were sufficient to help rigorously T-depleted unprimed (B + accessory) cells for direct plaque-forming cell responses to trinitrophenyl- (TNP) conjugated keyhole limpet hemocyanin (KLH). The activation of several lines of cloned (H-2b X H-2k)F1 TH cells was antigen (KLH) specific and H-2 restricted. Individual clones were restricted to H -2b, H-2k, or unique (H-2b X H-2k)F1 encoded determinants. Under the experimental conditions employed, responses mediated by cloned TH cells were found to result in the activation of the Lyb-5+ B cell subpopulation. The activation of Lyb-5+ B cells by cloned TH cells did not require covalent linkage of carrier and hapten, and responses could be stimulated in the presence of free KLH plus TNP conjugated to an irrelevant carrier. The H-2 restriction of TH cell function was shown to reflect a requirement for T cell recognition of determinants expressed by accessory cells, whereas no requirement existed for restricted T cell recognition of B cells. These findings suggest that the help provided by monoclonal TH cells, once activated, was both antigen nonspecific and H-2 unrestricted. Consistent with this interpretation, it was found that the supernatant of antigen-stimulated TH cells provided antigen-nonspecific help to T-depleted spleen cells. Thus, these results demonstrate that the activation of Lyb-5+ B cells by antigen-specific and H-2-restricted monoclonal TH cell populations is itself antigen nonspecific and H-2 unrestricted.

    View details for Web of Science ID A1982NU21100050

    View details for PubMedID 6177752

  • MAJOR HISTOCOMPATIBILITY COMPLEX-RESTRICTED ANTIGEN PRESENTATION TO ANTIGEN-REACTIVE T-CELLS BY LYMPHOCYTE-B TUMOR-CELLS JOURNAL OF EXPERIMENTAL MEDICINE McKean, D. J., Infante, A. J., Nilson, A., Kimoto, M., Fathman, C. G., Walker, E., Warner, N. 1981; 154 (5): 1419-1431

    Abstract

    Previous reports have demonstrated that accessory cells function to present soluble protein antigens in association with gene products encoded within the I region of the major histocompatibility complex (MHC) to antigen-reactive T helper cells. The biochemical events that occur during antigen presentation are, however, not well-documented primarily because of the difficulties involved in purifying sufficient numbers of homogeneous antigen-presenting cells. In this paper, a number of Ia-positive B lymphocyte tumor lines are shown to be capable of presenting soluble protein antigens to antigen-reactive continuous T cell lines in an MHC-restricted fashion. The characterization of the antigen presentation function of these tumor cells indicates that the tumor cells have many of the functional antigen-presenting characteristics previously thought to be limited to macrophages. These tumor cells should provide a useful model system for determining the biochemical events that occur in antigen uptake and processing as well as for determining the potential interactions between processed antigen and Ia molecules on the plasma membrane of these antigen-presenting cells.

    View details for Web of Science ID A1981MP25500013

    View details for PubMedID 6170720

  • ANTIGEN-REACTIVE T-CELL CLONES .2. UNIQUE HOMOZYGOUS AND (HIGH RESPONDER X LOW RESPONDER)F-1 HYBRID ANTIGEN-PRESENTING DETERMINANTS DETECTED USING POLY(TYR, GLU)-POLY D, L-ALA--POLY LYS-REACTIVE T-CELL CLONES JOURNAL OF EXPERIMENTAL MEDICINE Kimoto, M., Fathman, C. G. 1981; 153 (2): 375-385

    Abstract

    Using murine (T,G)-A--L-reactive T cell clones, we have demonstrated the existence of unique homozygous antigen-presenting determinants expressed on C57bl/6 mice, controlled by the I-A subregion of the murine major histocompatibility complex (MHC), which are not expressed on semisyngeneic (C57Bl/6 x A/J)F1 [(B6A)F1] cells. Additionally, we were able to demonstrate that there exist (T,G)-A--L-reactive clones in F1 mice derived between low responder and high responder parents [(B6A)F1] that recognize antigen in association with transcomplementing hybrid I-A subregion determinants expressed uniquely on (B6A)F1 cells not expressed on cells of either of the parental strains. These data suggest that phenotypic high responsiveness exhibited by (higher responder x low responder)F1 mice was not simply controlled by the high responder parental genome, but was controlled at the phenotypic level of expression of antigen-presenting determinants. Such antigen-presenting determinants can be created by complementation using products of the low responder as well as high responder genome. The significance of the existence of such F1 specific hybrid antigen-presenting determinants for T cell specificity and recognition of self was discussed.

    View details for Web of Science ID A1981LB84500013

    View details for PubMedID 6165797

  • STUDIES UTILIZING MURINE T-CELL CLONES - IR GENES, IA ANTIGENS AND MLR STIMULATING DETERMINANTS IMMUNOLOGICAL REVIEWS Fathman, C. G., Kimoto, M. 1981; 54: 57-79

    View details for Web of Science ID A1981LB03500003

    View details for PubMedID 6166538

  • FUNCTIONAL HELPER ACTIVITY OF MONOCLONAL T-CELL POPULATIONS - ANTIGEN-SPECIFIC AND H-2 RESTRICTED CLONED T-CELLS PROVIDE HELP FOR INVITRO ANTIBODY-RESPONSES TO TRINITROPHENYL-POLY(LTYR,GLU)-POLY(DLALA)--POLY(LLYS) PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA-BIOLOGICAL SCIENCES Hodes, R. J., Kimoto, M., Hathcock, K. S., Fathman, C. G., Singer, A. 1981; 78 (10): 6431-6435

    Abstract

    The ability of long-term cultured and monoclonal T cell populations to provide antigen-specific help was assessed in a system of Ir gene-controlled in vitro antibody responses to soluble antigens. T-cell colonies and monoclonal T-cell lines were generated which proliferated specifically in response to poly(LTyr,Glu)-poly(DLAla)--poly(LLys) [(T,G)-A--L] and were I-A restricted in these proliferative responses. These (T,G)-A--L-specific T-cell populations were evaluated for their ability to help unprimed and T-cell depleted spleen cell populations in the generation of antibody responses to trinitrophenyl (TNP)-(T,G)-A--L in vitro. It was found that long-term T-cell lines, including monoclonal T-cell populations derived by limiting dilution, were highly efficient helper cells for IgM responses to TNP-(T,G)-A--L. These helper T cells were both antigen-specific and I-A restricted in their ability to be activated and to cooperate with T-cell depleted spleen cell populations. Once specifically activated, however, these clones provided help that was antigen nonspecific. These studies have thus demonstrated the ability of antigen-specific and H-2-restricted monoclonal T-cell populations to provide help for responses to soluble antigens in vitro.

    View details for Web of Science ID A1981MM06900099

    View details for PubMedID 6975940

  • I-REGION GENETIC RESTRICTIONS IMPOSED UPON THE RECOGNITION OF KLH BY MURINE T-CELL CLONES IMMUNOGENETICS Shigeta, M., Fathman, C. G. 1981; 14 (5): 415-422

    Abstract

    Data presented in this paper show that the recognition of keyhole limpet hemocyanin by murine T-cell clones is restricted by products of the I region. These data have been obtained by genetic mapping studies as well as by the use of monoclonal Ia-specific antibodies which inhibit the ability of antigen-presenting cells to effectively present antigen to such T- cell clones. Use of heterozygous antigen-presenting cells derived from crosses between B6.C-H-2bm12 and B10.A(4R) mice have allowed us to show that both trans-complementing I-A products are used for restriction of recognition of KLH. These data were confirmed using monoclonal Ia antibodies to inhibition recognition of KLH by the same T -cell clones. Thus, we have shown that there exist hybrid molecules formed by free combinatorial association of products encoded within the I-A subregion which restrict the recognition of soluble antigen. Additionally, we have shown that the molecule formed by complementation between the alpha chain encoded within the I-E region and a beta chain encoded within the A region (Ae) can fuction effectively in presenting KLH to certain murine T -cell clones. These results support the hypothesis that the recognition of individual antigenic epitopes within large multideterminant antigens is under the control of Ir genes.

    View details for Web of Science ID A1981MR12800006

    View details for PubMedID 6800947

  • T-CELL CLONES REACTIVE WITH SPERM WHALE MYOGLOBIN - ISOLATION OF CLONES WITH SPECIFICITY FOR INDIVIDUAL DETERMINANTS ON MYOGLOBIN JOURNAL OF EXPERIMENTAL MEDICINE Infante, A. J., Atassi, M. Z., Fathman, C. G. 1981; 154 (5): 1342-1356

    Abstract

    We have been able to isolate clones of sperm whale muscle myoglobin (Mb)-reactive T cells from (C57BL/6 x A/J)F1 [(B6A)F1] mice. Four types of clones were isolated, distinguished by their patterns of recognition of Mb cyanogen bromide (CNBr) fragments and antigen presenting cell (APC) requirements. Individual T cell clones proliferated in response to one of three CNBr fragments of Mb. Dose-response curves of all clones were identical for native Mb and the appropriate fragment. T cell clones reactive to fragment 1-55 did not proliferate in response to peptide 15-22 (a peptide that binds to serum antibody directed against 1-55). These data support previous findings suggesting differences between antigen recognition by T and B cells, i.e., T cells may not recognize antigen in its native conformation and/or T and B cells may recognize distinct epitopes on the same antigen. Using T cell clones to analyze genetic control of responsiveness to Mb, we found that certain (B6A)F1 T cells recognize Mb presented by low responder strain APC. Thus, genetically determined low responsiveness in this case is probably not due to failure of APC function. We also found that responsiveness to certain Mb epitopes mapped to the I-A subregion whereas others mapped, via gene complementation, to the I-A and I-E subregions. We found no examples of responsiveness mapping to the I-C subregion and suggest an alternative explanation for previous reports mapping genetic control of responsiveness to certain Mb determinants to I-C.

    View details for Web of Science ID A1981MP25500007

    View details for PubMedID 6170716

    View details for PubMedCentralID PMC2186505

  • RECONSTITUTION OF IR GENES, IA-ANTIGENS, AND MIXED LYMPHOCYTE-REACTION DETERMINANTS BY GENE COMPLEMENTATION PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA-BIOLOGICAL SCIENCES Fathman, C. G., Kimoto, M., MELVOLD, R., David, C. S. 1981; 78 (3): 1853-1857

    Abstract

    By using clones of murine T cells reactive with alloantigens as well as soluble antigens, it has been possible to demonstrate that Ia antigens, Ir gene phenomena (defined as the ability of immune T cells to recognize antigen), and mixed lymphocyte reaction (MLR)-stimulating determinants encoded within the I-A subregion are different manifestations associated with same product. These studies utilized the I-Ab mutant mouse B6.C-H-2bm (bm12), whose defect in the normal expression of the cell surface products of the I-Ab subregion can be partially circumvented through trans-complementation by deriving hybrids between bm12 and mice expressing normal I-A subregion products. Such mice [e.g., (bm12 X B10.A)F1] express several serologically normal I-Ab products but have defects in certain I-A subregion gene products normally expressed on cells of H-2a X H-2b heterozygote mice that are used as restricting elements for antigen recognition by antigen-reactive T cell clones or recognized as alloantigens by alloreactive T cell clones. This defect in cell surface expression of certain normal I-Ab products on the mutant bm12 cells has allowed us to suggest that there exist trans-complementing products on H-2a X H-2b heterozygote mice consisting of Ab alpha Ak beta and Ak alpha Ab beta that restrict antigen recognition by cloned T cells, stimulate MLR responses by cloned T cells, and react with certain anti-Ia antisera.

    View details for Web of Science ID A1981LJ93300110

    View details for PubMedID 6165020

  • ANTIGEN-REACTIVE T-CELL CLONES .3. LOW RESPONDER ANTIGEN-PRESENTING CELLS FUNCTION EFFECTIVELY TO PRESENT ANTIGEN TO SELECTED T-CELL CLONES DERIVED FROM (HIGH RESPONDER X LOW RESPONDER)F1 MICE JOURNAL OF EXPERIMENTAL MEDICINE Kimoto, M., KRENZ, T. J., Fathman, C. G. 1981; 154 (3): 883-891

    Abstract

    Long-term-cultured poly(Tyr, Glu)-poly-D,L,-Ala-poly-Lys [(T,G)-A--L]-reactive T cells and clones derived from (high responder x low responder)F1 [(C57BL/6 x A/J)F1] mice were shown to recognize (T,G)-A--L presented by cells from low responder strain A/J mice. The antigen-presenting determinant(s) that allowed recognition of (T,G)-A--L by such T cell clones was controlled by the I-A subregion of the major histocompatibility complex. These results suggest that there is no functional defect in the ability of low responder Ir gene products (I-A antigens) to associate with (T,G)-A--L for effective recognition by T cells. Although these results might tentatively be interpreted to suggest that Ir gene-controlled low responsiveness is due to the inability of the T cell to recognize the association between (T,G)-A--L and low responder I-A gene products, it is similarly possible that there might be a defect in the functional capabilities of low responder antigen-presenting cells to effectively process (T,G)-A--L into immunodominant epitopes.

    View details for Web of Science ID A1981MG02000025

    View details for PubMedID 6456323

  • PRODUCTION OF ALLOREACTIVE T-CELL LYMPHOMAS NATURE Fathman, C. G., Weissman, I. L. 1980; 283 (5745): 404-406

    View details for Web of Science ID A1980JC08100058

    View details for PubMedID 6965424

  • ANTIGEN-REACTIVE T-CELL CLONES .1. TRANS-COMPLEMENTING HYBRID I-A-REGION GENE-PRODUCTS FUNCTION EFFECTIVELY IN ANTIGEN PRESENTATION JOURNAL OF EXPERIMENTAL MEDICINE Kimoto, M., Fathman, C. G. 1980; 152 (4): 759-770

    Abstract

    Studies in our laboratory and elsewhere have shown that it is possible to propagate antigen-specific murine T cells in vitro with resultant specific stepwise enrichment of antigen-induced proliferative cells. The proliferative responses of these T cells are antigen specific and dependent upon the presence of antigen-presenting cells (spleen cells) that share the I-A subregion with the proliferating T cell. Using techniques of soft-agar cloning, it has been further possible to isolate clones of antigen-reactive T lymphocytes from such long-term cultures. Data suggesting that these were clones of antigen-reactive T cells were obtained by studying the recognition of antigen in association with antigen-presenting cells with a panel of such clones of antigen-reactive T cells. Proof of clonality was obtained by subcloning. Clones derived from F1-immune mice can be divided into three separate categories: one clone recognizes antigen in association with antigen-presenting determinants of parent A and the F1; the second type recognizes antigen in association with antigen-presenting determinants of parent B and the F1; and the third type recognizes antigen only in association with antigen-presenting determinants of the F1 mouse. Genetic studies on the major histocompatibility complex requirements for antigen presentation to such F1-reactive T cell clones suggests that the hybrid antigen-presenting determinant in this system results from transcomplementation of products of the I-A region of haplotypes a and b. These studies support the concept developed in our laboratory that there exist unique F1 hybrid determinants on (A/J X C57BL/6) F1 cells and suggest that these determinants can be utilized physiologically by hybrid mice in immunocompetent cellular interactions.

    View details for Web of Science ID A1980KL44100001

    View details for PubMedID 6158548

  • DIFFERENTIATION OF THYMUS-CELLS FEDERATION PROCEEDINGS Weissman, I. L., Small, M., Fathman, C. G., Herzenberg, L. A. 1975; 34 (2): 141-144

    View details for Web of Science ID A1975V451000006

    View details for PubMedID 1090451

  • THYMUS-CELL MATURATION .2. DIFFERENTIATION OF 3 MATURE SUBCLASSES INVIVO CELLULAR IMMUNOLOGY Fathman, C. G., Small, M., Herzenberg, L. A., Weissman, I. L. 1975; 15 (1): 109-128

    View details for Web of Science ID A1975V156100011

    View details for PubMedID 1109155

  • CHARACTERIZATION OF SUBPOPULATIONS OF T-LYMPHOCYTES .1. SEPARATION AND FUNCTIONAL STUDIES OF PERIPHERAL T-CELLS BINDING DIFFERENT AMOUNTS OF FLUORESCENT ANTI-THY 1.2 (THETA) ANTIBODY USING A FLUORESCENCE-ACTIVATED CELL SORTER (FACS) CELLULAR IMMUNOLOGY Cantor, H., Simpson, E., Sato, V. L., Garrison, C., Herzenberg, L. A. 1975; 15 (1): 180-196

    View details for Web of Science ID A1975V156100016

    View details for PubMedID 1088903