2023-24 Courses


Stanford Advisees


All Publications


  • FOXP3 deficiency, from the mechanisms of the disease to curative strategies. Immunological reviews Borna, S., Meffre, E., Bacchetta, R. 2023

    Abstract

    FOXP3 gene is a key transcription factor driving immune tolerance and its deficiency causes immune dysregulation, polyendocrinopathy, enteropathy X-linked syndrome (IPEX), a prototypic primary immune regulatory disorder (PIRD) with defective regulatory T (Treg) cells. Although life-threatening, the increased awareness and early diagnosis have contributed to improved control of the disease. IPEX currently comprises a broad spectrum of clinical autoimmune manifestations from severe early onset organ involvement to moderate, recurrent manifestations. This review focuses on the mechanistic advancements that, since the IPEX discovery in early 2000, have informed the role of the human FOXP3+ Treg cells in controlling peripheral tolerance and shaping the overall immune landscape of IPEX patients and carrier mothers, contributing to defining new treatments.

    View details for DOI 10.1111/imr.13289

    View details for PubMedID 37994657

  • TLR9 ligand sequestration by chemokine CXCL4 negatively affects central B cell tolerance JOURNAL OF EXPERIMENTAL MEDICINE Cakan, E., Kioon, M., Garcia-Carmona, Y., Glauzy, S., Oliver, D., Yamakawa, N., Loza, A., Du, Y., Schickel, J., Boeckers, J. M., Yang, C., Baldo, A., Ivashkiv, L. B., Young, R. M., Staudt, L. M., Moody, K. L., Nundel, K., Marshak-Rothstein, A., van der Made, C. I., Hoischen, A., Hayward, A., Rossato, M., Radstake, T. J., Cunningham-Rundles, C., Ryu, C., Herzog, E. L., Barrat, F. J., Meffre, E. 2023; 220 (12)

    Abstract

    Central B cell tolerance is believed to be regulated by B cell receptor signaling induced by the recognition of self-antigens in immature B cells. Using humanized mice with defective MyD88, TLR7, or TLR9 expression, we demonstrate that TLR9/MYD88 are required for central B cell tolerance and the removal of developing autoreactive clones. We also show that CXCL4, a chemokine involved in systemic sclerosis (SSc), abrogates TLR9 function in B cells by sequestering TLR9 ligands away from the endosomal compartments where this receptor resides. The in vivo production of CXCL4 thereby impedes both TLR9 responses in B cells and the establishment of central B cell tolerance. We conclude that TLR9 plays an essential early tolerogenic function required for the establishment of central B cell tolerance and that correcting defective TLR9 function in B cells from SSc patients may represent a novel therapeutic strategy to restore B cell tolerance.

    View details for DOI 10.1084/jem.20230944

    View details for Web of Science ID 001090346300001

    View details for PubMedID 37773045

    View details for PubMedCentralID PMC10541333

  • Isoform-specific knockdown of long and intermediate prolactin receptors interferes with evolution of B-cell neoplasms. Communications biology Taghi Khani, A., Kumar, A., Sanchez Ortiz, A., Radecki, K. C., Aramburo, S., Lee, S. J., Hu, Z., Damirchi, B., Lorenson, M. Y., Wu, X., Gu, Z., Stohl, W., Sanz, I., Meffre, E., Müschen, M., Forman, S. J., Koff, J. L., Walker, A. M., Swaminathan, S. 2023; 6 (1): 295

    Abstract

    Prolactin (PRL) is elevated in B-cell-mediated lymphoproliferative diseases and promotes B-cell survival. Whether PRL or PRL receptors drive the evolution of B-cell malignancies is unknown. We measure changes in B cells after knocking down the pro-proliferative, anti-apoptotic long isoform of the PRL receptor (LFPRLR) in vivo in systemic lupus erythematosus (SLE)- and B-cell lymphoma-prone mouse models, and the long plus intermediate isoforms (LF/IFPRLR) in human B-cell malignancies. To knockdown LF/IFPRLRs without suppressing expression of the counteractive short PRLR isoforms (SFPRLRs), we employ splice-modulating DNA oligomers. In SLE-prone mice, LFPRLR knockdown reduces numbers and proliferation of pathogenic B-cell subsets and lowers the risk of B-cell transformation by downregulating expression of activation-induced cytidine deaminase. LFPRLR knockdown in lymphoma-prone mice reduces B-cell numbers and their expression of BCL2 and TCL1. In overt human B-cell malignancies, LF/IFPRLR knockdown reduces B-cell viability and their MYC and BCL2 expression. Unlike normal B cells, human B-cell malignancies secrete autocrine PRL and often express no SFPRLRs. Neutralization of secreted PRL reduces the viability of B-cell malignancies. Knockdown of LF/IFPRLR reduces the growth of human B-cell malignancies in vitro and in vivo. Thus, LF/IFPRLR knockdown is a highly specific approach to block the evolution of B-cell neoplasms.

    View details for DOI 10.1038/s42003-023-04667-8

    View details for PubMedID 36941341

    View details for PubMedCentralID 5050045

  • Autoantibodies are highly prevalent in non-SARS-CoV-2 respiratory infections and critical illness. JCI insight Feng, A., Yang, E. Y., Moore, A. R., Dhingra, S., Chang, S. E., Yin, X., Pi, R., Mack, E. K., Völkel, S., Geßner, R., Gündisch, M., Neubauer, A., Renz, H., Tsiodras, S., Fragkou, P. C., Asuni, A. A., Levitt, J. E., Wilson, J. G., Leong, M., Lumb, J. H., Mao, R., Pinedo, K., Roque, J., Richards, C. M., Stabile, M., Swaminathan, G., Salagianni, M. L., Triantafyllia, V., Bertrams, W., Blish, C. A., Carette, J. E., Frankovich, J., Meffre, E., Nadeau, K. C., Singh, U., Wang, T. T., Luning Prak, E. T., Herold, S., Andreakos, E., Schmeck, B., Skevaki, C., Rogers, A. J., Utz, P. J. 2023; 8 (3)

    Abstract

    The widespread presence of autoantibodies in acute infection with SARS-CoV-2 is increasingly recognized, but the prevalence of autoantibodies in non-SARS-CoV-2 infections and critical illness has not yet been reported. We profiled IgG autoantibodies in 267 patients from 5 independent cohorts with non-SARS-CoV-2 viral, bacterial, and noninfectious critical illness. Serum samples were screened using Luminex arrays that included 58 cytokines and 55 autoantigens, many of which are associated with connective tissue diseases (CTDs). Samples positive for anti-cytokine antibodies were tested for receptor blocking activity using cell-based functional assays. Anti-cytokine antibodies were identified in > 50% of patients across all 5 acutely ill cohorts. In critically ill patients, anti-cytokine antibodies were far more common in infected versus uninfected patients. In cell-based functional assays, 11 of 39 samples positive for select anti-cytokine antibodies displayed receptor blocking activity against surface receptors for Type I IFN, GM-CSF, and IL-6. Autoantibodies against CTD-associated autoantigens were also commonly observed, including newly detected antibodies that emerged in longitudinal samples. These findings demonstrate that anti-cytokine and autoantibodies are common across different viral and nonviral infections and range in severity of illness.

    View details for DOI 10.1172/jci.insight.163150

    View details for PubMedID 36752204

  • Partial RAG deficiency in humans induces dysregulated peripheral lymphocyte development and humoral tolerance defect with accumulation of T-bet+ B cells. Nature immunology Csomos, K., Ujhazi, B., Blazso, P., Herrera, J. L., Tipton, C. M., Kawai, T., Gordon, S., Ellison, M., Wu, K., Stowell, M., Haynes, L., Cruz, R., Zakota, B., Nguyen, J., Altrich, M., Geier, C. B., Sharapova, S., Dasso, J. F., Leiding, J. W., Smith, G., Al-Herz, W., de Barros Dorna, M., Fadugba, O., Fronkova, E., Kanderova, V., Svaton, M., Henrickson, S. E., Hernandez, J. D., Kuijpers, T., Kandilarova, S. M., Naumova, E., Milota, T., Sediva, A., Moshous, D., Neven, B., Saco, T., Sargur, R., Savic, S., Sleasman, J., Sunkersett, G., Ward, B. R., Komatsu, M., Pittaluga, S., Kumanovics, A., Butte, M. J., Cancro, M. P., Pillai, S., Meffre, E., Notarangelo, L. D., Walter, J. E. 2022

    Abstract

    The recombination-activating genes (RAG) 1 and 2 are indispensable for diversifying the primary B cell receptor repertoire and pruning self-reactive clones via receptor editing in the bone marrow; however, the impact of RAG1/RAG2 on peripheral tolerance is unknown. Partial RAG deficiency (pRD) manifesting with late-onset immune dysregulation represents an 'experiment of nature' to explore this conundrum. By studying B cell development and subset-specific repertoires in pRD, we demonstrate that reduced RAG activity impinges on peripheral tolerance through the generation of a restricted primary B cell repertoire, persistent antigenic stimulation and an inflammatory milieu with elevated B cell-activating factor. This unique environment gradually provokes profound B cell dysregulation with widespread activation, remarkable extrafollicular maturation and persistence, expansion and somatic diversification of self-reactive clones. Through the model of pRD, we reveal a RAG-dependent 'domino effect' that impacts stringency of tolerance and B cell fate in the periphery.

    View details for DOI 10.1038/s41590-022-01271-6

    View details for PubMedID 35902638

  • Inflammasome activation in infected macrophages drives COVID-19 pathology. Nature Sefik, E., Qu, R., Junqueira, C., Kaffe, E., Mirza, H., Zhao, J., Brewer, J. R., Han, A., Steach, H. R., Israelow, B., Blackburn, H. N., Velazquez, S. E., Chen, Y. G., Halene, S., Iwasaki, A., Meffre, E., Nussenzweig, M., Lieberman, J., Wilen, C. B., Kluger, Y., Flavell, R. A. 2022; 606 (7914): 585-593

    Abstract

    Severe COVID-19 is characterized by persistent lung inflammation, inflammatory cytokine production, viral RNA and a sustained interferon (IFN) response, all of which are recapitulated and required for pathology in the SARS-CoV-2-infected MISTRG6-hACE2 humanized mouse model of COVID-19, which has a human immune system1-20. Blocking either viral replication with remdesivir21-23 or the downstream IFN-stimulated cascade with anti-IFNAR2 antibodies in vivo in the chronic stages of disease attenuates the overactive immune inflammatory response, especially inflammatory macrophages. Here we show that SARS-CoV-2 infection and replication in lung-resident human macrophages is a critical driver of disease. In response to infection mediated by CD16 and ACE2 receptors, human macrophages activate inflammasomes, release interleukin 1 (IL-1) and IL-18, and undergo pyroptosis, thereby contributing to the hyperinflammatory state of the lungs. Inflammasome activation and the accompanying inflammatory response are necessary for lung inflammation, as inhibition of the NLRP3 inflammasome pathway reverses chronic lung pathology. Notably, this blockade of inflammasome activation leads to the release of infectious virus by the infected macrophages. Thus, inflammasomes oppose host infection by SARS-CoV-2 through the production of inflammatory cytokines and suicide by pyroptosis to prevent a productive viral cycle.

    View details for DOI 10.1038/s41586-022-04802-1

    View details for PubMedID 35483404

    View details for PubMedCentralID PMC9288243

  • A humanized mouse model of chronic COVID-19. Nature biotechnology Sefik, E., Israelow, B., Mirza, H., Zhao, J., Qu, R., Kaffe, E., Song, E., Halene, S., Meffre, E., Kluger, Y., Nussenzweig, M., Wilen, C. B., Iwasaki, A., Flavell, R. A. 2022; 40 (6): 906-920

    Abstract

    Coronavirus disease 2019 (COVID-19) is an infectious disease that can present as an uncontrolled, hyperactive immune response, causing severe immunological injury. Existing rodent models do not recapitulate the sustained immunopathology of patients with severe disease. Here we describe a humanized mouse model of COVID-19 that uses adeno-associated virus to deliver human ACE2 to the lungs of humanized MISTRG6 mice. This model recapitulates innate and adaptive human immune responses to severe acute respiratory syndrome coronavirus 2 infection up to 28 days after infection, with key features of chronic COVID-19, including weight loss, persistent viral RNA, lung pathology with fibrosis, a human inflammatory macrophage response, a persistent interferon-stimulated gene signature and T cell lymphopenia. We used this model to study two therapeutics on immunopathology, patient-derived antibodies and steroids and found that the same inflammatory macrophages crucial to containing early infection later drove immunopathology. This model will enable evaluation of COVID-19 disease mechanisms and treatments.

    View details for DOI 10.1038/s41587-021-01155-4

    View details for PubMedID 34921308

    View details for PubMedCentralID PMC9203605

  • A Novel AICDA Splice-Site Mutation in Two Siblings with HIGM2 Permits Somatic Hypermutation but Abrogates Mutational Targeting. Journal of clinical immunology Dirks, J., Haase, G., Cantaert, T., Frey, L., Klaas, M., Rickert, C. H., Girschick, H., Meffre, E., Morbach, H. 2022; 42 (4): 771-782

    Abstract

    Hyper-IgM syndrome type 2 (HIGM2) is a B cell intrinsic primary immunodeficiency caused by mutations in AICDA encoding activation-induced cytidine deaminase (AID) which impair immunoglobulin class switch recombination (CSR) and somatic hypermutation (SHM). Whereas autosomal-recessive AID-deficiency (AR-AID) affects both CSR and SHM, the autosomal-dominant form (AD-AID) due to C-terminal heterozygous variants completely abolishes CSR but only partially affects SHM. AR-AID patients display enhanced germinal center (GC) reactions and autoimmune manifestations, which are not present in AD-AID, suggesting that SHM but not CSR regulates GC reactions and peripheral B cell tolerance. Herein, we describe two siblings with HIGM2 due to a novel homozygous AICDA mutation (c.428-1G > T) which disrupts the splice acceptor site of exon 4 and results in the sole expression of a truncated AID variant that lacks 10 highly conserved amino acids encoded by exon 4 (AID-ΔE4a). AID-ΔE4a patients suffered from defective CSR and enhanced GC reactions and were therefore indistinguishable from other AR-AID patients. However, the AID-ΔE4a variant only partially affected SHM as observed in AD-AID patients. In addition, AID-ΔE4a but not AD-AID patients revealed impaired targeting of mutational hotspot motives and distorted mutational patterns. Hence, qualitative defects in AID function and altered SHM rather than global decreased SHM activity may account for the disease phenotype in these patients.

    View details for DOI 10.1007/s10875-022-01233-5

    View details for PubMedID 35246784

    View details for PubMedCentralID PMC9166864

  • Loss of Treg identity in IPEX patients is associated with increase autoreactivity in Teff cells Borna, S., Lee, E., Lakshmanan, U., Narula, M., Schulze, J., Ramachandran, A., Bertaina, A., Meffre, E., Maria, R., Bacchetta, R. SPRINGER/PLENUM PUBLISHERS. 2022: S78-S79
  • High-throughput identification of autoantibodies that target the human exoproteome. Cell reports methods Wang, E. Y., Dai, Y., Rosen, C. E., Schmitt, M. M., Dong, M. X., Ferré, E. M., Liu, F., Yang, Y., González-Hernández, J. A., Meffre, E., Hinchcliff, M., Koumpouras, F., Lionakis, M. S., Ring, A. M. 2022; 2 (2)

    Abstract

    Autoantibodies that recognize extracellular proteins (the exoproteome) exert potent biological effects but are challenging to detect. Here, we developed rapid extracellular antigen profiling (REAP), a high-throughput technique for the comprehensive discovery of exoproteome-targeting autoantibodies. Patient samples are applied to a genetically barcoded yeast surface display library containing 2,688 human extracellular proteins. Antibody-coated yeast are isolated, and sequencing of barcodes is used to identify displayed antigens. To benchmark REAP's performance, we screened 77 patients with autoimmune polyglandular syndrome type 1 (APS-1). REAP sensitively and specifically detected both known and previously unidentified autoantibodies in APS-1. We further screened 106 patients with systemic lupus erythematosus (SLE) and identified numerous autoantibodies, several of which were associated with disease severity or specific clinical manifestations and exerted functional effects on cell signaling ex vivo. These findings demonstrate the utility of REAP to atlas the expansive landscape of exoproteome-targeting autoantibodies and their impacts on patient health outcomes.

    View details for DOI 10.1016/j.crmeth.2022.100172

    View details for PubMedID 35360706

    View details for PubMedCentralID PMC8967185

  • Defective Early B Cell Tolerance Checkpoints in Patients With Systemic Sclerosis Allow the Production of Self Antigen-Specific Clones. Arthritis & rheumatology (Hoboken, N.J.) Glauzy, S., Olson, B., May, C. K., Parisi, D., Massad, C., Hansen, J. E., Ryu, C., Herzog, E. L., Meffre, E. 2022; 74 (2): 307-317

    Abstract

    Early selection steps preventing autoreactive naive B cell production are often impaired in patients with autoimmune diseases, but central and peripheral B cell tolerance checkpoints have not been assessed in patients with systemic sclerosis (SSc). This study was undertaken to characterize early B cell tolerance checkpoints in patients with SSc.Using an in vitro polymerase chain reaction-based approach that allows the expression of recombinant antibodies cloned from single B cells, we tested the reactivity of antibodies expressed by 212 CD19+CD21low CD10+IgMhigh CD27- new emigrant/transitional B cells and 190 CD19+CD21+CD10-IgM+CD27- mature naive B cells from 10 patients with SSc.Compared to serum from healthy donors, serum from patients with SSc displayed elevated proportions of polyreactive and antinuclear-reactive new emigrant/transitional B cells that recognize topoisomerase I, suggesting that defective central B cell tolerance contributes to the production of serum autoantibodies characteristic of the disease. Frequencies of autoreactive mature naive B cells were also significantly increased in SSc patients compared to healthy donors, thus indicating that a peripheral B cell tolerance checkpoint may be impaired in SSc.Defective counterselection of developing autoreactive naive B cells in SSc leads to the production of self antigen-specific B cells that may secrete autoantibodies and allow the formation of immune complexes, which promote fibrosis in SSc.

    View details for DOI 10.1002/art.41927

    View details for PubMedID 34279059

    View details for PubMedCentralID PMC8766600

  • Autoantibodies targeting cytokines and connective tissue disease autoantigens are common in acute non-SARS-CoV-2 infections. Research square Feng, A., Yang, E., Moore, A., Dhingra, S., Chang, S., Yin, X., Pi, R., Mack, E., Völkel, S., Geßner, R., Gundisch, M., Neubauer, A., Renz, H., Tsiodras, S., Fragkou, P., Asuni, A., Levitt, J., Wilson, J., Leong, M., Lumb, J., Mao, R., Pinedo, K., Roque, J., Richards, C., Stabile, M., Swaminathan, G., Salagianni, M., Triantafyllia, V., Bertrams, W., Blish, C., Carette, J., Frankovich, J., Meffre, E., Nadeau, K. C., Singh, U., Wang, T., Prak, E. L., Herold, S., Andreakos, E., Schmeck, B., Skevaki, C., Rogers, A., Utz, P. 2022

    Abstract

    The widespread presence of autoantibodies in acute infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is increasingly recognized, but the prevalence of autoantibodies in infections with organisms other than SARS-CoV-2 has not yet been reported. We used protein arrays to profile IgG autoantibodies from 317 samples from 268 patients across a spectrum of non-SARS-CoV-2 infections, many of whom were critically ill with pneumonia. Anti-cytokine antibodies (ACA) were identified in > 50% of patients infected with non-SARS-CoV-2 viruses and other pathogens, including patients with pneumonia attributed to bacterial causes. In cell-based functional assays, some ACA blocked binding to surface receptors for type I interferons (Type I IFN), granulocyte-macrophage colony-stimulating factor (GM-CSF), and interleukin-6 (IL-6). Autoantibodies against traditional autoantigens associated with connective tissue diseases (CTDs) were also commonly observed in these cohorts, including newly-detected antibodies that emerged in longitudinal samples from patients infected with influenza. We conclude that autoantibodies, some of which are functionally active, may be much more prevalent than previously appreciated in patients who are symptomatically infected with diverse pathogens.

    View details for DOI 10.21203/rs.3.rs-1233038/v1

    View details for PubMedID 35075455

    View details for PubMedCentralID PMC8786233

  • Positive and negative selection shape the human naive B cell repertoire. The Journal of clinical investigation Chen, J. W., Schickel, J. N., Tsakiris, N., Sng, J., Arbogast, F., Bouis, D., Parisi, D., Gera, R., Boeckers, J. M., Delmotte, F. R., Veselits, M., Schuetz, C., Jacobsen, E. M., Posovszky, C., Schulz, A. S., Schwarz, K., Clark, M. R., Menard, L., Meffre, E. 2022; 132 (2)

    Abstract

    Although negative selection of developing B cells in the periphery is well described, yet poorly understood, evidence of naive B cell positive selection remains elusive. Using 2 humanized mouse models, we demonstrate that there was strong skewing of the expressed immunoglobulin repertoire upon transit into the peripheral naive B cell pool. This positive selection of expanded naive B cells in humanized mice resembled that observed in healthy human donors and was independent of autologous thymic tissue. In contrast, negative selection of autoreactive B cells required thymus-derived Tregs and MHC class II-restricted self-antigen presentation by B cells. Indeed, both defective MHC class II expression on B cells of patients with rare bare lymphocyte syndrome and prevention of self-antigen presentation via HLA-DM inhibition in humanized mice resulted in the production of autoreactive naive B cells. These latter observations suggest that Tregs repressed autoreactive naive B cells continuously produced by the bone marrow. Thus, a model emerged, in which both positive and negative selection shaped the human naive B cell repertoire and that each process was mediated by fundamentally different molecular and cellular mechanisms.

    View details for DOI 10.1172/JCI150985

    View details for PubMedID 34813502

    View details for PubMedCentralID PMC8759783

  • Developmental partitioning of SYK and ZAP70 prevents autoimmunity and cancer. Molecular cell Sadras, T., Martin, M., Kume, K., Robinson, M. E., Saravanakumar, S., Lenz, G., Chen, Z., Song, J. Y., Siddiqi, T., Oksa, L., Knapp, A. M., Cutler, J., Cosgun, K. N., Klemm, L., Ecker, V., Winchester, J., Ghergus, D., Soulas-Sprauel, P., Kiefer, F., Heisterkamp, N., Pandey, A., Ngo, V., Wang, L., Jumaa, H., Buchner, M., Ruland, J., Chan, W. C., Meffre, E., Martin, T., Müschen, M. 2021; 81 (10): 2094-2111.e9

    Abstract

    Even though SYK and ZAP70 kinases share high sequence homology and serve analogous functions, their expression in B and T cells is strictly segregated throughout evolution. Here, we identified aberrant ZAP70 expression as a common feature in a broad range of B cell malignancies. We validated SYK as the kinase that sets the thresholds for negative selection of autoreactive and premalignant clones. When aberrantly expressed in B cells, ZAP70 competes with SYK at the BCR signalosome and redirects SYK from negative selection to tonic PI3K signaling, thereby promoting B cell survival. In genetic mouse models for B-ALL and B-CLL, conditional expression of Zap70 accelerated disease onset, while genetic deletion impaired malignant transformation. Inducible activation of Zap70 during B cell development compromised negative selection of autoreactive B cells, resulting in pervasive autoantibody production. Strict segregation of the two kinases is critical for normal B cell selection and represents a central safeguard against the development of autoimmune disease and B cell malignancies.

    View details for DOI 10.1016/j.molcel.2021.03.043

    View details for PubMedID 33878293

    View details for PubMedCentralID PMC8239336

  • Disease-associated CTNNBL1 mutation impairs somatic hypermutation by decreasing nuclear AID. The Journal of clinical investigation Kuhny, M., Forbes, L. R., Çakan, E., Vega-Loza, A., Kostiuk, V., Dinesh, R. K., Glauzy, S., Stray-Pedersen, A., Pezzi, A. E., Hanson, I. C., Vargas-Hernandez, A., Xu, M. L., Coban-Akdemir, Z. H., Jhangiani, S. N., Muzny, D. M., Gibbs, R. A., Lupski, J. R., Chinn, I. K., Schatz, D. G., Orange, J. S., Meffre, E. 2020; 130 (8): 4411-4422

    Abstract

    Patients with common variable immunodeficiency associated with autoimmune cytopenia (CVID+AIC) generate few isotype-switched B cells with severely decreased frequencies of somatic hypermutations (SHMs), but their underlying molecular defects remain poorly characterized. We identified a CVID+AIC patient who displays a rare homozygous missense M466V mutation in β-catenin-like protein 1 (CTNNBL1). Because CTNNBL1 binds activation-induced cytidine deaminase (AID) that catalyzes SHM, we tested AID interactions with the CTNNBL1 M466V variant. We found that the M466V mutation interfered with the association of CTNNBL1 with AID, resulting in decreased AID in the nuclei of patient EBV-transformed B cell lines and of CTNNBL1 466V/V Ramos B cells engineered to express only CTNNBL1 M466V using CRISPR/Cas9 technology. As a consequence, the scarce IgG+ memory B cells from the CTNNBL1 466V/V patient showed a low SHM frequency that averaged 6.7 mutations compared with about 18 mutations per clone in healthy-donor counterparts. In addition, CTNNBL1 466V/V Ramos B cells displayed a decreased incidence of SHM that was reduced by half compared with parental WT Ramos B cells, demonstrating that the CTNNBL1 M466V mutation is responsible for defective SHM induction. We conclude that CTNNBL1 plays an important role in regulating AID-dependent antibody diversification in humans.

    View details for DOI 10.1172/JCI131297

    View details for PubMedID 32484799

    View details for PubMedCentralID PMC7410074

  • Autoreactivity in naïve human fetal B cells is associated with commensal bacteria recognition. Science (New York, N.Y.) Chen, J. W., Rice, T. A., Bannock, J. M., Bielecka, A. A., Strauss, J. D., Catanzaro, J. R., Wang, H., Menard, L. C., Anolik, J. H., Palm, N. W., Meffre, E. 2020; 369 (6501): 320-325

    Abstract

    Restricted V(D)J recombination during fetal development was postulated to limit antibody repertoire breadth and prevent autoimmunity. However, newborn serum contains abundant autoantibodies, suggesting that B cell tolerance during gestation is not yet fully established. To investigate this apparent paradox, we evaluated the reactivities of more than 450 antibodies cloned from single B cells from human fetal liver, bone marrow, and spleen. We found that incomplete B cell tolerance in early human fetal life favored the accumulation of polyreactive B cells that bound both apoptotic cells and commensal bacteria from healthy adults. Thus, the restricted fetal preimmune repertoire contains potentially beneficial self-reactive innate-like B cell specificities that may facilitate the removal of apoptotic cells during development and shape gut microbiota assembly after birth.

    View details for DOI 10.1126/science.aay9733

    View details for PubMedID 32675374

  • Impaired ATM activation in B cells is associated with bone resorption in rheumatoid arthritis. Science translational medicine Mensah, K. A., Chen, J. W., Schickel, J. N., Isnardi, I., Yamakawa, N., Vega-Loza, A., Anolik, J. H., Gatti, R. A., Gelfand, E. W., Montgomery, R. R., Horowitz, M. C., Craft, J. E., Meffre, E. 2019; 11 (519)

    Abstract

    Patients with rheumatoid arthritis (RA) may display atypical CD21-/lo B cells in their blood, but the implication of this observation remains unclear. We report here that the group of patients with RA and elevated frequencies of CD21-/lo B cells shows decreased ataxia telangiectasia-mutated (ATM) expression and activation in B cells compared with other patients with RA and healthy donor controls. In agreement with ATM involvement in the regulation of V(D)J recombination, patients with RA who show defective ATM function displayed a skewed B cell receptor (BCR) Igκ repertoire, which resembled that of patients with ataxia telangiectasia (AT). This repertoire was characterized by increased Jκ1 and decreased upstream Vκ gene segment usage, suggesting improper secondary recombination processes and selection. In addition, altered ATM function in B cells was associated with decreased osteoprotegerin and increased receptor activator of nuclear factor κB ligand (RANKL) production. These changes favor bone loss and correlated with a higher prevalence of erosive disease in patients with RA who show impaired ATM function. Using a humanized mouse model, we also show that ATM inhibition in vivo induces an altered Igκ repertoire and RANKL production by immature B cells in the bone marrow, leading to decreased bone density. We conclude that dysregulated ATM function in B cells promotes bone erosion and the emergence of circulating CD21-/lo B cells, thereby contributing to RA pathophysiology.

    View details for DOI 10.1126/scitranslmed.aaw4626

    View details for PubMedID 31748230

    View details for PubMedCentralID PMC7167286

  • Transitional B cells in quiescent SLE: An early checkpoint imprinted by IFN. Journal of autoimmunity Dieudonné, Y., Gies, V., Guffroy, A., Keime, C., Bird, A. K., Liesveld, J., Barnas, J. L., Poindron, V., Douiri, N., Soulas-Sprauel, P., Martin, T., Meffre, E., Anolik, J. H., Korganow, A. S. 2019; 102: 150-158

    Abstract

    Systemic lupus (SLE) is characterized by a break of B cell tolerance that plays a central role in disease pathophysiology. An early checkpoint defect occurs at the transitional stage leading to the survival of autoreactive B cells and consequently the production of pathogenic autoantibodies. The main purpose of our work was to determine whether transitional B cells, as the most immature naïve B cell subset upstream of pathogenic B cells, display specific features compared to healthy non SLE subjects. Through extensive analysis of transitional B cells from untreated or low treated, mostly Caucasian, SLE patients, we demonstrated that transitional (T1 and T2) B cell frequencies were increased in SLE and positively correlated with disease activity. SLE transitional B cells displayed defects in two closely inter-related molecules (i.e. TLR9 defective responses and CD19 downregulation). RNA sequencing of sorted transitional B cells from untreated patients revealed a predominant overexpression of interferon stimulated genes (ISGs) even out of flares. In addition, early transitional B cells from the bone marrow displayed the highest interferon score, reflecting a B cell interferon burden of central origin. Hence, the IFN signature in transitional B cells is not confined to African American SLE patients and exists in quiescent disease since the medullary stage. These results suggest that in SLE these 3 factors (i.e. IFN imprintment, CD19 downregulation and TLR9 responses impairment) could take part at the early transitional B cell stage in B cell tolerance by-pass, ultimately leading in periphery to the expansion of autoantibodies-secreting cells.

    View details for DOI 10.1016/j.jaut.2019.05.002

    View details for PubMedID 31085070

    View details for PubMedCentralID PMC6642027

  • Human DEF6 deficiency underlies an immunodeficiency syndrome with systemic autoimmunity and aberrant CTLA-4 homeostasis. Nature communications Serwas, N. K., Hoeger, B., Ardy, R. C., Stulz, S. V., Sui, Z., Memaran, N., Meeths, M., Krolo, A., Yüce Petronczki, Ö., Pfajfer, L., Hou, T. Z., Halliday, N., Santos-Valente, E., Kalinichenko, A., Kennedy, A., Mace, E. M., Mukherjee, M., Tesi, B., Schrempf, A., Pickl, W. F., Loizou, J. I., Kain, R., Bidmon-Fliegenschnee, B., Schickel, J. N., Glauzy, S., Huemer, J., Garncarz, W., Salzer, E., Pierides, I., Bilic, I., Thiel, J., Priftakis, P., Banerjee, P. P., Förster-Waldl, E., Medgyesi, D., Huber, W. D., Orange, J. S., Meffre, E., Sansom, D. M., Bryceson, Y. T., Altman, A., Boztug, K. 2019; 10 (1): 3106

    Abstract

    Immune responses need to be controlled tightly to prevent autoimmune diseases, yet underlying molecular mechanisms remain partially understood. Here, we identify biallelic mutations in three patients from two unrelated families in differentially expressed in FDCP6 homolog (DEF6) as the molecular cause of an inborn error of immunity with systemic autoimmunity. Patient T cells exhibit impaired regulation of CTLA-4 surface trafficking associated with reduced functional CTLA-4 availability, which is replicated in DEF6-knockout Jurkat cells. Mechanistically, we identify the small GTPase RAB11 as an interactor of the guanine nucleotide exchange factor DEF6, and find disrupted binding of mutant DEF6 to RAB11 as well as reduced RAB11+CTLA-4+ vesicles in DEF6-mutated cells. One of the patients has been treated with CTLA-4-Ig and achieved sustained remission. Collectively, we uncover DEF6 as player in immune homeostasis ensuring availability of the checkpoint protein CTLA-4 at T-cell surface, identifying a potential target for autoimmune and/or cancer therapy.

    View details for DOI 10.1038/s41467-019-10812-x

    View details for PubMedID 31308374

    View details for PubMedCentralID PMC6629652

  • B cell depletion or absence does not impede anti-tumor activity of PD-1 inhibitors. Journal for immunotherapy of cancer Damsky, W., Jilaveanu, L., Turner, N., Perry, C., Zito, C., Tomayko, M., Leventhal, J., Herold, K., Meffre, E., Bosenberg, M., Kluger, H. M. 2019; 7 (1): 153

    Abstract

    PD-1 inhibitors are approved for multiple malignancies and function by stimulating T cells. However, the role of B cells in the anti-tumor activity of these drugs is unknown, as is their activity in patients who have received B cell depleting drugs or with immunoglobulin deficiencies.We studied B cell content in 40 melanomas from patients treated with pembrolizumab or nivolumab and assessed the association with response to therapy. Murine MC38 colon cancer and YUMMER1.7 melanoma models were used to determine whether concomitant anti-CD20 antibody injections diminish the anti-tumor effects of anti-PD-1. Results were validated in muMT mice, which lack B cells.B cells were sparse in most melanomas and B cell content was not associated with response to anti-PD-1 or overall survival. Employing MC38 and YUMMER1.7 models, we demonstrated that anti-CD20 antibodies reduce tumor-infiltrating B cells yet had no effect on tumor growth, response to PD-1 inhibition, or survival. In muMT mice, T-cell dependent tumor rejection and anti-PD-1 responses were no different than in wildtype C57BL/6 J mice.The degree of tumor infiltrating B cell content is not associated with response to anti-PD-1 inhibitors in melanoma. PD-1 inhibitors cause tumor shrinkage in murine cancer models even when B cells are absent or are depleted. PD-1 inhibitors are likely to be active in patients with impaired B cell function, such as patients undergoing B cell depletion with drugs including rituximab for conditions such as B cell malignancies or autoimmune disorders.

    View details for DOI 10.1186/s40425-019-0613-1

    View details for PubMedID 31200747

    View details for PubMedCentralID PMC6567557

  • Early B cell tolerance defects in neuromyelitis optica favour anti-AQP4 autoantibody production. Brain : a journal of neurology Cotzomi, E., Stathopoulos, P., Lee, C. S., Ritchie, A. M., Soltys, J. N., Delmotte, F. R., Oe, T., Sng, J., Jiang, R., Ma, A. K., Vander Heiden, J. A., Kleinstein, S. H., Levy, M., Bennett, J. L., Meffre, E., O'Connor, K. C. 2019; 142 (6): 1598-1615

    Abstract

    Neuromyelitis optica spectrum disorders (NMOSD) constitute rare autoimmune disorders of the CNS that are primarily characterized by severe inflammation of the spinal cord and optic nerve. Approximately 75% of NMOSD patients harbour circulating pathogenic autoantibodies targeting the aquaporin-4 water channel (AQP4). The source of these autoantibodies remains unclear, but parallels between NMOSD and other autoantibody-mediated diseases posit compromised B cell tolerance checkpoints as common underlying and contributing factors. Using a well established assay, we assessed tolerance fidelity by creating recombinant antibodies from B cell populations directly downstream of each checkpoint and testing them for polyreactivity and autoreactivity. We examined a total of 863 recombinant antibodies. Those derived from three anti-AQP4-IgG seropositive NMOSD patients (n = 130) were compared to 733 antibodies from 15 healthy donors. We found significantly higher frequencies of poly- and autoreactive new emigrant/transitional and mature naïve B cells in NMOSD patients compared to healthy donors (P-values < 0.003), thereby identifying defects in both central and peripheral B cell tolerance checkpoints in these patients. We next explored whether pathogenic NMOSD anti-AQP4 autoantibodies can originate from the pool of poly- and autoreactive clones that populate the naïve B cell compartment of NMOSD patients. Six human anti-AQP4 autoantibodies that acquired somatic mutations were reverted back to their unmutated germline precursors, which were tested for both binding to AQP4 and poly- or autoreactivity. While the affinity of mature autoantibodies against AQP4 ranged from modest to strong (Kd 15.2-559 nM), none of the germline revertants displayed any detectable binding to AQP4, revealing that somatic hypermutation is required for the generation of anti-AQP4 autoantibodies. However, two (33.3%) germline autoantibody revertants were polyreactive and four (66.7%) were autoreactive, suggesting that pathogenic anti-AQP4 autoantibodies can originate from the pool of autoreactive naïve B cells, which develops as a consequence of impaired early B cell tolerance checkpoints in NMOSD patients.

    View details for DOI 10.1093/brain/awz106

    View details for PubMedID 31056665

    View details for PubMedCentralID PMC6536857

  • AIRE expression controls the peripheral selection of autoreactive B cells. Science immunology Sng, J., Ayoglu, B., Chen, J. W., Schickel, J., Ferre, E. M., Glauzy, S., Romberg, N., Hoenig, M., Cunningham-Rundles, C., Utz, P. J., Lionakis, M. S., Meffre, E. 2019; 4 (34)

    Abstract

    Autoimmune regulator (AIRE) mutations result in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) syndrome characterized by defective central T cell tolerance and the production of many autoantibodies targeting tissue-specific antigens and cytokines. By studying CD3- and AIRE-deficient patients, we found that lack of either T cells or AIRE function resulted in the peripheral accumulation of autoreactive mature naive B cells. Proteomic arrays and Biacore affinity measurements revealed that unmutated antibodies expressed by these autoreactive naive B cells recognized soluble molecules and cytokines including insulin, IL-17A, and IL-17F, which are AIRE-dependent thymic peripheral tissue antigens targeted by autoimmune responses in APECED. AIRE-deficient patients also displayed decreased frequencies of regulatory T cells (Tregs) that lacked common TCRbeta clones found instead in their conventional T cell compartment, thereby suggesting holes in the Treg TCR repertoire of these patients. Hence, AIRE-mediated T cell/Treg selection normally prevents the expansion of autoreactive naive B cells recognizing peripheral self-antigens.

    View details for PubMedID 30979797

  • A novel ATM mutation associated with elevated atypical lymphocyte populations, hyper-IgM, and cutaneous granulomas. Clinical immunology (Orlando, Fla.) Minto, H., Mensah, K. A., Reynolds, P. R., Meffre, E., Rubtsova, K., Gelfand, E. W. 2019; 200: 55-63

    Abstract

    Ataxia-Telangiectasia (AT) is an immunodeficiency most often associated with T cell abnormalities. We describe a patient with a hyper-IgM phenotype and immune cell abnormalities that suggest a distinct clinical phenotype. Significant B cell abnormalities with increased unswitched memory B cells, decreased naive transitional B cells, and an elevated frequency of CD19+CD38loCD27-CD10-CD21-/low B cells expressing high levels of T-bet and Fas were demonstrated. The B cells were hyporesponsive to in vitro stimulation through the B cell receptor, Toll like receptors (TLR) 7 and 9, and CD40. T cell homeostasis was also disturbed with a significant increase in γδ T cells, circulating T follicular helper cells (Tfh), and decreased numbers of T regulatory cells. The ATM mutations in this patient are posited to have resulted in the perturbations in the frequencies and distributions of B and T cell subsets, resulting in the phenotype in this patient. KEY MESSAGES: A novel mutation creating a premature stop codon and a nonsense mutation in the ATM gene are postulated to have resulted in the unique clinical picture characterized by abnormal B and T cell populations, lymphocyte subset dysfunction, granuloma formation, and a hyper-IgM phenotype. CAPSULE SUMMARY: A patient presented with ataxia-telangiectasia, cutaneous granulomas, and a hyper-IgM phenotype; a novel combination of mutations in the ATM gene was associated with abnormal distributions, frequencies, and function of T and B lymphocyte subsets.

    View details for DOI 10.1016/j.clim.2019.01.002

    View details for PubMedID 30639167

    View details for PubMedCentralID PMC7027322

  • Patients with common variable immunodeficiency with autoimmune cytopenias exhibit hyperplastic yet inefficient germinal center responses. The Journal of allergy and clinical immunology Romberg, N., Le Coz, C., Glauzy, S., Schickel, J. N., Trofa, M., Nolan, B. E., Paessler, M., Xu, M. L., Lambert, M. P., Lakhani, S. A., Khokha, M. K., Jyonouchi, S., Heimall, J., Takach, P., Maglione, P. J., Catanzaro, J., Hsu, F. I., Sullivan, K. E., Cunningham-Rundles, C., Meffre, E. 2019; 143 (1): 258-265

    Abstract

    The lack of pathogen-protective, isotype-switched antibodies in patients with common variable immunodeficiency (CVID) suggests germinal center (GC) hypoplasia, yet a subset of patients with CVID is paradoxically affected by autoantibody-mediated autoimmune cytopenias (AICs) and lymphadenopathy.We sought to compare the physical characteristics and immunologic output of GC responses in patients with CVID with AIC (CVID+AIC) and without AIC (CVID-AIC).We analyzed GC size and shape in excisional lymph node biopsy specimens from 14 patients with CVID+AIC and 4 patients with CVID-AIC. Using paired peripheral blood samples, we determined how AICs specifically affected B-and T-cell compartments and antibody responses in patients with CVID.We found that patients with CVID+AIC displayed irregularly shaped hyperplastic GCs, whereas GCs were scarce and small in patients with CVID-AIC. GC hyperplasia was also evidenced by an increase in numbers of circulating follicular helper T cells, which correlated with decreased regulatory T-cell frequencies and function. In addition, patients with CVID+AIC had serum endotoxemia associated with a dearth of isotype-switched memory B cells that displayed significantly lower somatic hypermutation frequencies than their counterparts with CVID-AIC. Moreover, IgG+ B cells from patients with CVID+AIC expressed VH4-34-encoded antibodies with unmutated Ala-Val-Tyr and Asn-His-Ser motifs, which recognize both erythrocyte I/i self-antigens and commensal bacteria.Patients with CVID+AIC do not contain mucosal microbiota and exhibit hyperplastic yet inefficient GC responses that favor the production of untolerized IgG+ B-cell clones that recognize both commensal bacteria and hematopoietic I/i self-antigens.

    View details for DOI 10.1016/j.jaci.2018.06.012

    View details for PubMedID 29935219

    View details for PubMedCentralID PMC6400323

  • TACI Isoforms Regulate Ligand Binding and Receptor Function. Frontiers in immunology Garcia-Carmona, Y., Ting, A. T., Radigan, L., Athuluri Divakar, S. K., Chavez, J., Meffre, E., Cerutti, A., Cunningham-Rundles, C. 2018; 9: 2125

    Abstract

    TACI signals activate B cell proliferation, isotype switch and antibody production in both normal immunity and autoimmune states. In contrast to murine TACI, the human TACI gene undergoes alternative splicing to produce short and long isoforms (TACI-S and TACI-L). In previous studies, we showed that transduction of the short, but not long isoform, into murine B cells or human pre-B cells lacking TACI, caused them to become transcriptional and morphologically identical to plasma cells. These data suggest that the expression of different isoforms in humans provides unique controls on B cell maturation. In these studies we show that TACI-S and TACI-L form complexes in a ligand-independent manner, not dependent on a single extracellular domain. Both TACI isoforms are detectable in the endosomal cellular compartment where they co-localize with MyD88, TRAF6, and the activated 65 kDa form of TLR9, depending on a conserved intracellular TACI sequence. In contrast to TACI-L expressing cells, or cells bearing both isoforms, TACI-S binds ligands BAFF and APRIL with substantially greater affinity and promotes enhanced NF-kB activation. Using isoform-specific monoclonal antibodies, we show that while TACI-L is predominant as a surface receptor surface on human B cells, significantly more TACI-S is noted in the intracellular compartment and also in marginal zone, isotype switched and plasmablast in resting B cells. TACI-S is increased in tonsillar B cells and also in the intracellular compartment of activated peripheral B cells. These data shows that alternative splicing of the human TACI gene leads to two isoforms both of which intersect with MyD88 and TRAF6 and form complexes with TLR9, but the two isoforms have different ligand binding capacities, subcellular locations and activation capabilities.

    View details for DOI 10.3389/fimmu.2018.02125

    View details for PubMedID 30333819

    View details for PubMedCentralID PMC6176016

  • Reprogramming human T cell function and specificity with non-viral genome targeting NATURE Roth, T. L., Puig-Saus, C., Yu, R., Shifrut, E., Carnevale, J., Li, P., Hiatt, J., Saco, J., Krystofinski, P., Li, H., Tobin, V., Nguyen, D. N., Lee, M. R., Putnam, A. L., Ferris, A. L., Chen, J. W., Schickel, J., Pellerin, L., Carmody, D., Alkorta-Aranburu, G., del Gaudio, D., Matsumoto, H., Morell, M., Mao, Y., Cho, M., Quadros, R. M., Gurumurthy, C. B., Smith, B., Haugwitz, M., Hughes, S. H., Weissman, J. S., Schumann, K., Esensten, J. H., May, A. P., Ashworth, A., Kupfer, G. M., Greeley, S. W., Bacchetta, R., Meffre, E., Roncarolo, M., Romberg, N., Herold, K. C., Ribas, A., Leonetti, M. D., Marson, A. 2018; 559 (7714): 405-+

    Abstract

    Decades of work have aimed to genetically reprogram T cells for therapeutic purposes1,2 using recombinant viral vectors, which do not target transgenes to specific genomic sites3,4. The need for viral vectors has slowed down research and clinical use as their manufacturing and testing is lengthy and expensive. Genome editing brought the promise of specific and efficient insertion of large transgenes into target cells using homology-directed repair5,6. Here we developed a CRISPR-Cas9 genome-targeting system that does not require viral vectors, allowing rapid and efficient insertion of large DNA sequences (greater than one kilobase) at specific sites in the genomes of primary human T cells, while preserving cell viability and function. This permits individual or multiplexed modification of endogenous genes. First, we applied this strategy to correct a pathogenic IL2RA mutation in cells from patients with monogenic autoimmune disease, and demonstrate improved signalling function. Second, we replaced the endogenous T cell receptor (TCR) locus with a new TCR that redirected T cells to a cancer antigen. The resulting TCR-engineered T cells specifically recognized tumour antigens and mounted productive anti-tumour cell responses in vitro and in vivo. Together, these studies provide preclinical evidence that non-viral genome targeting can enable rapid and flexible experimental manipulation and therapeutic engineering of primary human immune cells.

    View details for PubMedID 29995861

  • Antigen-mediated regulation in monoclonal gammopathies and myeloma. JCI insight Nair, S., Sng, J., Boddupalli, C. S., Seckinger, A., Chesi, M., Fulciniti, M., Zhang, L., Rauniyar, N., Lopez, M., Neparidze, N., Parker, T., Munshi, N. C., Sexton, R., Barlogie, B., Orlowski, R., Bergsagel, L., Hose, D., Flavell, R. A., Mistry, P. K., Meffre, E., Dhodapkar, M. V. 2018; 3 (8)

    Abstract

    A role for antigen-driven stimulation has been proposed in the pathogenesis of monoclonal gammopathy of undetermined significance (MGUS) and multiple myeloma (MM) based largely on the binding properties of monoclonal Ig. However, insights into antigen binding to clonal B cell receptors and in vivo responsiveness of the malignant clone to antigen-mediated stimulation are needed to understand the role of antigenic stimulation in tumor growth. Lysolipid-reactive clonal Ig were detected in Gaucher disease (GD) and some sporadic gammopathies. Here, we show that recombinant Ig (rIg) cloned from sort-purified single tumor cells from lipid-reactive sporadic and GD-associated gammopathy specifically bound lysolipids. Liposome sedimentation and binding assays confirmed specific interaction of lipid-reactive monoclonal Ig with lysolipids. The clonal nature of lysolipid-binding Ig was validated by protein sequencing. Gene expression profiling and cytogenetic analyses from 2 patient cohorts showed enrichment of nonhyperdiploid tumors in lipid-reactive patients. In vivo antigen-mediated stimulation led to an increase in clonal Ig and plasma cells (PCs) in GD gammopathy and also reactivated previously suppressed antigenically related nonclonal PCs. These data support a model wherein antigenic stimulation mediates an initial polyclonal phase, followed by evolution of monoclonal tumors enriched in nonhyperdiploid genomes, responsive to underlying antigen. Targeting underlying antigens may therefore prevent clinical MM.

    View details for DOI 10.1172/jci.insight.98259

    View details for PubMedID 29669929

    View details for PubMedCentralID PMC5931125

  • Alternations in repertoire of T and B cell subsets in patients with partial recombination activating gene (RAG) deficiency with autoimmunity and history of viral infections Csomos, K., Ujhazi, B., Butte, M. J., Hernandez, J., Geha, R., Al-Herz, W., Notarangelo, L. D., Meffre, E., Walter, J. E. SPRINGER/PLENUM PUBLISHERS. 2018: 420–21
  • Impaired TLR9 responses in B cells from patients with systemic lupus erythematosus. JCI insight Gies, V., Schickel, J. N., Jung, S., Joublin, A., Glauzy, S., Knapp, A. M., Soley, A., Poindron, V., Guffroy, A., Choi, J. Y., Gottenberg, J. E., Anolik, J. H., Martin, T., Soulas-Sprauel, P., Meffre, E., Korganow, A. S. 2018; 3 (5)

    Abstract

    B cells play a central role in systemic lupus erythematosus (SLE) pathophysiology but dysregulated pathways leading to a break in B cell tolerance remain unclear. Since Toll-like receptor 9 (TLR9) favors the elimination of autoreactive B cells in the periphery, we assessed TLR9 function in SLE by analyzing the responses of B cells and plasmacytoid dendritic cells (pDCs) isolated from healthy donors and patients after stimulation with CpG, a TLR9 agonist. We found that SLE B cells from patients without hydroxychloroquine treatment displayed defective in vitro TLR9 responses, as illustrated by the impaired upregulation of B cell activation molecules and the diminished production of various cytokines including antiinflammatory IL-10. In agreement with CD19 controlling TLR9 responses in B cells, decreased expression of the CD19/CD21 complex on SLE B cells was detected as early as the transitional B cell stage. In contrast, TLR7 function was preserved in SLE B cells, whereas pDCs from SLE patients properly responded to TLR9 stimulation, thereby revealing that impaired TLR9 function in SLE was restricted to B cells. We conclude that abnormal CD19 expression and TLR9 tolerogenic function in SLE B cells may contribute to the break of B cell tolerance in these patients.

    View details for DOI 10.1172/jci.insight.96795

    View details for PubMedID 29515028

    View details for PubMedCentralID PMC5922279

  • Naive B cells are prone to develop into polyreactive autoantibody secreting cells from adult RAG2-deficient patient with combined immunodeficiency Stowell, M. T., Walter, J. E., Pennix, T. C., Ujhazi, B., Mueller, G., Butte, M., Hoyte, E. G., Hernandez, J. D., Meffre, E. F., Csomos, K. MOSBY-ELSEVIER. 2018: AB22
  • Accumulation of Antigen-Driven Lymphoproliferations in Complement Receptor 2/CD21-/low B Cells From Patients With Sjögren's Syndrome. Arthritis & rheumatology (Hoboken, N.J.) Glauzy, S., Boccitto, M., Bannock, J. M., Delmotte, F. R., Saadoun, D., Cacoub, P., Ice, J. A., Sivils, K. L., James, J. A., Wolin, S. L., Meffre, E. 2018; 70 (2): 298-307

    Abstract

    Patients with Sjögren's syndrome (SS) are prone to develop malignant lymphomas, and a correlation has been established between the lymphoproliferations occurring in these disorders and the presence in patients' blood of an unusual B cell population that down-regulates complement receptor 2/CD21. This study was undertaken to identify the B cell compartment from which these lymphoproliferations emerge and determine the mechanisms that promote clonal B cell expansion in patients with SS.The reactivity of antibodies expressed by CD19+CD10-CD27-IgM+CD21-/low cells isolated from the blood of patients with SS was tested using a polymerase chain reaction-based approach that allows us to clone and express, in vitro, recombinant antibodies produced by single B cells.Clonal expansions were identified in CD21-/low B cells isolated from the peripheral blood of 3 patients with SS. These lymphoproliferations expressed B cell receptors (BCRs) that displayed somatic hypermutation lineage trees characteristic of a strong selection by antigens; one of these antigens was identified as a ribosomal self antigen. When the mutated BCR sequences expressed by the expanded CD21-/low B cell clones from patients with SS were reverted in vitro to their germline counterparts, one clone remained autoreactive.Clonal lymphoproliferations in patients with SS preferentially accumulate in the autoreactive CD21-/low B cell compartment often expanded in these subjects, and recognition of self antigens may drive the clonal B cell expansion while further refining BCR self-reactivity.

    View details for DOI 10.1002/art.40352

    View details for PubMedID 29073352

    View details for PubMedCentralID PMC5788702

  • mTOR intersects antibody-inducing signals from TACI in marginal zone B cells. Nature communications Sintes, J., Gentile, M., Zhang, S., Garcia-Carmona, Y., Magri, G., Cassis, L., Segura-Garzón, D., Ciociola, A., Grasset, E. K., Bascones, S., Comerma, L., Pybus, M., Lligé, D., Puga, I., Gutzeit, C., He, B., DuBois, W., Crespo, M., Pascual, J., Mensa, A., Aróstegui, J. I., Juan, M., Yagüe, J., Serrano, S., Lloreta, J., Meffre, E., Hahne, M., Cunningham-Rundles, C., Mock, B. A., Cerutti, A. 2017; 8 (1): 1462

    Abstract

    Mechanistic target of rapamycin (mTOR) enhances immunity in addition to orchestrating metabolism. Here we show that mTOR coordinates immunometabolic reconfiguration of marginal zone (MZ) B cells, a pre-activated lymphocyte subset that mounts antibody responses to T-cell-independent antigens through a Toll-like receptor (TLR)-amplified pathway involving transmembrane activator and CAML interactor (TACI). This receptor interacts with mTOR via the TLR adapter MyD88. The resulting mTOR activation instigates MZ B-cell proliferation, immunoglobulin G (IgG) class switching, and plasmablast differentiation through a rapamycin-sensitive pathway that integrates metabolic and antibody-inducing transcription programs, including NF-κB. Disruption of TACI-mTOR interaction by rapamycin, truncation of the MyD88-binding domain of TACI, or B-cell-conditional mTOR deficiency interrupts TACI signaling via NF-κB and cooperation with TLRs, thereby hampering IgG production to T-cell-independent antigens but not B-cell survival. Thus, mTOR drives innate-like antibody responses by linking proximal TACI signaling events with distal immunometabolic transcription programs.

    View details for DOI 10.1038/s41467-017-01602-4

    View details for PubMedID 29133782

    View details for PubMedCentralID PMC5684130

  • Defective Early B Cell Tolerance Checkpoints in Sjögren's Syndrome Patients. Arthritis & rheumatology (Hoboken, N.J.) Glauzy, S., Sng, J., Bannock, J. M., Gottenberg, J. E., Korganow, A. S., Cacoub, P., Saadoun, D., Meffre, E. 2017; 69 (11): 2203-2208

    Abstract

    Central and peripheral B cell tolerance checkpoints are defective in many patients with autoimmune diseases, but the functionality of each discrete checkpoint has not been assessed in patients with Sjögren's syndrome (SS). We undertook this study to assess this functionality in SS patients.Using a polymerase chain reaction-based approach that allows us to clone and express, in vitro, recombinant antibodies produced by single B cells, we tested the reactivity of recombinant antibodies cloned from single CD19+CD21low CD10+IgMhigh CD27- newly emigrant/transitional B cells and CD19+CD21+CD10-IgM+CD27- mature naive B cells from 5 SS patients.We found that the frequencies of newly emigrant/transitional B cells expressing polyreactive antibodies were significantly increased in SS patients compared to those in healthy donors, revealing defective central B cell tolerance in SS patients. Frequencies of mature naive B cells expressing autoreactive antibodies were also significantly increased in SS patients, thereby illustrating an impaired peripheral B cell tolerance checkpoint in these patients.Defective counterselection of developing autoreactive B cells observed in SS patients is a feature common to many other autoimmune diseases and may favor the development of autoimmunity by allowing autoreactive B cells to present self antigens to T cells.

    View details for DOI 10.1002/art.40215

    View details for PubMedID 28704602

    View details for PubMedCentralID PMC6062007

  • HSC extrinsic sex-related and intrinsic autoimmune disease-related human B-cell variation is recapitulated in humanized mice. Blood advances Borsotti, C., Danzl, N. M., Nauman, G., Hölzl, M. A., French, C., Chavez, E., Khosravi-Maharlooei, M., Glauzy, S., Delmotte, F. R., Meffre, E., Savage, D. G., Campbell, S. R., Goland, R., Greenberg, E., Bi, J., Satwani, P., Yang, S., Bathon, J., Winchester, R., Sykes, M. 2017; 1 (23): 2007-2018

    Abstract

    B cells play a major role in antigen presentation and antibody production in the development of autoimmune diseases, and some of these diseases disproportionally occur in females. Moreover, immune responses tend to be stronger in female vs male humans and mice. Because it is challenging to distinguish intrinsic from extrinsic influences on human immune responses, we used a personalized immune (PI) humanized mouse model, in which immune systems were generated de novo from adult human hematopoietic stem cells (HSCs) in immunodeficient mice. We assessed the effect of recipient sex and of donor autoimmune diseases (type 1 diabetes [T1D] and rheumatoid arthritis [RA]) on human B-cell development in PI mice. We observed that human B-cell levels were increased in female recipients regardless of the source of human HSCs or the strain of immunodeficient recipient mice. Moreover, mice injected with T1D- or RA-derived HSCs displayed B-cell abnormalities compared with healthy control HSC-derived mice, including altered B-cell levels, increased proportions of mature B cells and reduced CD19 expression. Our study revealed an HSC-extrinsic effect of recipient sex on human B-cell reconstitution. Moreover, the PI humanized mouse model revealed HSC-intrinsic defects in central B-cell tolerance that recapitulated those in patients with autoimmune diseases. These results demonstrate the utility of humanized mouse models as a tool to better understand human immune cell development and regulation.

    View details for DOI 10.1182/bloodadvances.2017006932

    View details for PubMedID 29296847

    View details for PubMedCentralID PMC5728279

  • Smith-Magenis Syndrome Patients Often Display Antibody Deficiency but Not Other Immune Pathologies JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE Perkins, T., Rosenberg, J. M., Le Coz, C., Alaimo, J. T., Trofa, M., Mullegama, S. V., Antaya, R. J., Jyonouchi, S., Elsea, S. H., Utz, P. J., Meffre, E., Romberg, N. 2017; 5 (5): 1344-+

    Abstract

    Smith-Magenis syndrome (SMS) is a complex neurobehavioral disorder associated with recurrent otitis. Most SMS cases result from heterozygous interstitial chromosome 17p11.2 deletions that encompass not only the intellectual disability gene retinoic acid-induced 1 but also other genes associated with immunodeficiency, autoimmunity, and/or malignancy.The goals of this study were to describe the immunological consequence of 17p11.2 deletions by determining the prevalence of immunological diseases in subjects with SMS and by assessing their immune systems via laboratory methods.We assessed clinical histories of 76 subjects with SMS with heterozygous 17p11.2 deletions and performed in-depth immunological testing on 25 representative cohort members. Laboratory testing included determination of serum antibody concentrations, vaccine titers, and lymphocyte subset frequencies. Detailed reactivity profiles of SMS serum antibodies were performed using custom-made antigen microarrays.Of 76 subjects with SMS, 74 reported recurrent infections including otitis (88%), pneumonia (47%), sinusitis (42%), and gastroenteritis (34%). Infections were associated with worsening SMS-related neurobehavioral symptoms. The prevalence of autoimmune and atopic diseases was not increased. Malignancy was not reported. Laboratory evaluation revealed most subjects with SMS to be deficient of isotype-switched memory B cells and many to lack protective antipneumococcal antibodies. SMS antibodies were not more reactive than control antibodies to self-antigens.Patients with SMS with heterozygous 17p.11.2 deletions display an increased susceptibility to sinopulmonary infections, but not to autoimmune, allergic, or malignant diseases. SMS sera display an antibody reactivity profile favoring neither recognition of pathogen-associated antigens nor self-antigens. Prophylactic strategies to prevent infections may also provide neurobehavioral benefits to selected patients with SMS.

    View details for PubMedID 28286158

    View details for PubMedCentralID PMC5591748

  • Novel in vitro booster vaccination to rapidly generate antigen-specific human monoclonal antibodies. The Journal of experimental medicine Sanjuan Nandin, I., Fong, C., Deantonio, C., Torreno-Pina, J. A., Pecetta, S., Maldonado, P., Gasparrini, F., Ordovas-Montanes, J., Kazer, S. W., Kjaer, S., Borley, D. W., Nair, U., Coleman, J. A., Lingwood, D., Shalek, A. K., Meffre, E., Poignard, P., Burton, D. R., Batista, F. D. 2017; 214 (8): 2471-2490

    Abstract

    Vaccines remain the most effective tool to prevent infectious diseases. Here, we introduce an in vitro booster vaccination approach that relies on antigen-dependent activation of human memory B cells in culture. This stimulation induces antigen-specific B cell proliferation, differentiation of B cells into plasma cells, and robust antibody secretion after a few days of culture. We validated this strategy using cells from healthy donors to retrieve human antibodies against tetanus toxoid and influenza hemagglutinin (HA) from H1N1 and newly emergent subtypes such as H5N1 and H7N9. Anti-HA antibodies were cross-reactive against multiple subtypes, and some showed neutralizing activity. Although these antibodies may have arisen as a result of previous influenza infection, we also obtained gp120-reactive antibodies from non-HIV-infected donors, indicating that we can generate antibodies without prior antigenic exposure. Overall, our novel approach can be used to rapidly produce therapeutic antibodies and has the potential to assess the immunogenicity of candidate antigens, which could be exploited in future vaccine development.

    View details for DOI 10.1084/jem.20170633

    View details for PubMedID 28739603

    View details for PubMedCentralID PMC5551578

  • Germline hypomorphic CARD11 mutations in severe atopic disease. Nature genetics Ma, C. A., Stinson, J. R., Zhang, Y., Abbott, J. K., Weinreich, M. A., Hauk, P. J., Reynolds, P. R., Lyons, J. J., Nelson, C. G., Ruffo, E., Dorjbal, B., Glauzy, S., Yamakawa, N., Arjunaraja, S., Voss, K., Stoddard, J., Niemela, J., Zhang, Y., Rosenzweig, S. D., McElwee, J. J., DiMaggio, T., Matthews, H. F., Jones, N., Stone, K. D., Palma, A., Oleastro, M., Prieto, E., Bernasconi, A. R., Dubra, G., Danielian, S., Zaiat, J., Marti, M. A., Kim, B., Cooper, M. A., Romberg, N., Meffre, E., Gelfand, E. W., Snow, A. L., Milner, J. D. 2017; 49 (8): 1192-1201

    Abstract

    Few monogenic causes for severe manifestations of common allergic diseases have been identified. Through next-generation sequencing on a cohort of patients with severe atopic dermatitis with and without comorbid infections, we found eight individuals, from four families, with novel heterozygous mutations in CARD11, which encodes a scaffolding protein involved in lymphocyte receptor signaling. Disease improved over time in most patients. Transfection of mutant CARD11 expression constructs into T cell lines demonstrated both loss-of-function and dominant-interfering activity upon antigen receptor-induced activation of nuclear factor-κB and mammalian target of rapamycin complex 1 (mTORC1). Patient T cells had similar defects, as well as low production of the cytokine interferon-γ (IFN-γ). The mTORC1 and IFN-γ production defects were partially rescued by supplementation with glutamine, which requires CARD11 for import into T cells. Our findings indicate that a single hypomorphic mutation in CARD11 can cause potentially correctable cellular defects that lead to atopic dermatitis.

    View details for DOI 10.1038/ng.3898

    View details for PubMedID 28628108

    View details for PubMedCentralID PMC5664152

  • Self-reactive VH4-34-expressing IgG B cells recognize commensal bacteria. The Journal of experimental medicine Schickel, J. N., Glauzy, S., Ng, Y. S., Chamberlain, N., Massad, C., Isnardi, I., Katz, N., Uzel, G., Holland, S. M., Picard, C., Puel, A., Casanova, J. L., Meffre, E. 2017; 214 (7): 1991-2003

    Abstract

    The germline immunoglobulin (Ig) variable heavy chain 4-34 (VH4-34) gene segment encodes in humans intrinsically self-reactive antibodies that recognize I/i carbohydrates expressed by erythrocytes with a specific motif in their framework region 1 (FWR1). VH4-34-expressing clones are common in the naive B cell repertoire but are rarely found in IgG memory B cells from healthy individuals. In contrast, CD27+IgG+ B cells from patients genetically deficient for IRAK4 or MYD88, which mediate the function of Toll-like receptors (TLRs) except TLR3, contained VH4-34-expressing clones and showed decreased somatic hypermutation frequencies. In addition, VH4-34-encoded IgGs from IRAK4- and MYD88-deficient patients often displayed an unmutated FWR1 motif, revealing that these antibodies still recognize I/i antigens, whereas their healthy donor counterparts harbored FWR1 mutations abolishing self-reactivity. However, this paradoxical self-reactivity correlated with these VH4-34-encoded IgG clones binding commensal bacteria antigens. Hence, B cells expressing germline-encoded self-reactive VH4-34 antibodies may represent an innate-like B cell population specialized in the containment of commensal bacteria when gut barriers are breached.

    View details for DOI 10.1084/jem.20160201

    View details for PubMedID 28500047

    View details for PubMedCentralID PMC5502416

  • A novel humanized mouse model with significant improvement of class-switched, antigen-specific antibody production. Blood Yu, H., Borsotti, C., Schickel, J. N., Zhu, S., Strowig, T., Eynon, E. E., Frleta, D., Gurer, C., Murphy, A. J., Yancopoulos, G. D., Meffre, E., Manz, M. G., Flavell, R. A. 2017; 129 (8): 959-969

    Abstract

    Humanized mice are a powerful tool for the study of human hematopoiesis and immune function in vivo. However, the existing models cannot support robust adaptive immune responses, especially the generation of class-switched, antigen-specific antibody responses. Here we describe a new mouse strain, in which human interleukin 6 (IL-6) gene encoding the cytokine that is important for B- and T-cell differentiation was knocked into its respective mouse locus. The provision of human IL-6 not only enhanced thymopoiesis and periphery T-cell engraftment, but also significantly increased class switched memory B cells and serum immunoglobulin G (IgG). In addition, immunization with ovalbumin (OVA) induced OVA-specific B cells only in human IL-6 knock-in mice. These OVA-specific antibodies displayed the highest frequency of somatic mutation, further suggesting that human IL-6 is important for efficient B-cell activation and selection. We conclude that human IL-6 knock-in mice represent a novel and improved model for human adaptive immunity without relying on complex surgery to transplant human fetal thymus and liver. These mice can therefore be used to exploit or evaluate immunization regimes that would be unethical or untenable in humans.

    View details for DOI 10.1182/blood-2016-04-709584

    View details for PubMedID 28077418

    View details for PubMedCentralID PMC5324713

  • Deficiency of base excision repair enzyme NEIL3 drives increased predisposition to autoimmunity. The Journal of clinical investigation Massaad, M. J., Zhou, J., Tsuchimoto, D., Chou, J., Jabara, H., Janssen, E., Glauzy, S., Olson, B. G., Morbach, H., Ohsumi, T. K., Schmitz, K., Kyriacos, M., Kane, J., Torisu, K., Nakabeppu, Y., Notarangelo, L. D., Chouery, E., Megarbane, A., Kang, P. B., Al-Idrissi, E., Aldhekri, H., Meffre, E., Mizui, M., Tsokos, G. C., Manis, J. P., Al-Herz, W., Wallace, S. S., Geha, R. S. 2016; 126 (11): 4219-4236

    Abstract

    Alterations in the apoptosis of immune cells have been associated with autoimmunity. Here, we have identified a homozygous missense mutation in the gene encoding the base excision repair enzyme Nei endonuclease VIII-like 3 (NEIL3) that abolished enzymatic activity in 3 siblings from a consanguineous family. The NEIL3 mutation was associated with fatal recurrent infections, severe autoimmunity, hypogammaglobulinemia, and impaired B cell function in these individuals. The same homozygous NEIL3 mutation was also identified in an asymptomatic individual who exhibited elevated levels of serum autoantibodies and defective peripheral B cell tolerance, but normal B cell function. Further analysis of the patients revealed an absence of LPS-responsive beige-like anchor (LRBA) protein expression, a known cause of immunodeficiency. We next examined the contribution of NEIL3 to the maintenance of self-tolerance in Neil3-/- mice. Although Neil3-/- mice displayed normal B cell function, they exhibited elevated serum levels of autoantibodies and developed nephritis following treatment with poly(I:C) to mimic microbial stimulation. In Neil3-/- mice, splenic T and B cells as well as germinal center B cells from Peyer's patches showed marked increases in apoptosis and cell death, indicating the potential release of self-antigens that favor autoimmunity. These findings demonstrate that deficiency in NEIL3 is associated with increased lymphocyte apoptosis, autoantibodies, and predisposition to autoimmunity.

    View details for DOI 10.1172/JCI85647

    View details for PubMedID 27760045

    View details for PubMedCentralID PMC5096910

  • Decreased somatic hypermutation induces an impaired peripheral B cell tolerance checkpoint. The Journal of clinical investigation Cantaert, T., Schickel, J. N., Bannock, J. M., Ng, Y. S., Massad, C., Delmotte, F. R., Yamakawa, N., Glauzy, S., Chamberlain, N., Kinnunen, T., Menard, L., Lavoie, A., Walter, J. E., Notarangelo, L. D., Bruneau, J., Al-Herz, W., Kilic, S. S., Ochs, H. D., Cunningham-Rundles, C., van der Burg, M., Kuijpers, T. W., Kracker, S., Kaneko, H., Sekinaka, Y., Nonoyama, S., Durandy, A., Meffre, E. 2016; 126 (11): 4289-4302

    Abstract

    Patients with mutations in AICDA, which encodes activation-induced cytidine deaminase (AID), display an impaired peripheral B cell tolerance. AID mediates class-switch recombination (CSR) and somatic hypermutation (SHM) in B cells, but the mechanism by which AID prevents the accumulation of autoreactive B cells in blood is unclear. Here, we analyzed B cell tolerance in AID-deficient patients, patients with autosomal dominant AID mutations (AD-AID), asymptomatic AICDA heterozygotes (AID+/-), and patients with uracil N-glycosylase (UNG) deficiency, which impairs CSR but not SHM. The low frequency of autoreactive mature naive B cells in UNG-deficient patients resembled that of healthy subjects, revealing that impaired CSR does not interfere with the peripheral B cell tolerance checkpoint. In contrast, we observed decreased frequencies of SHM in memory B cells from AD-AID patients and AID+/- subjects, who were unable to prevent the accumulation of autoreactive mature naive B cells. In addition, the individuals with AICDA mutations, but not UNG-deficient patients, displayed Tregs with defective suppressive capacity that correlated with increases in circulating T follicular helper cells and enhanced cytokine production. We conclude that SHM, but not CSR, regulates peripheral B cell tolerance through the production of mutated antibodies that clear antigens and prevent sustained interleukin secretions that interfere with Treg function.

    View details for DOI 10.1172/JCI84645

    View details for PubMedID 27701145

    View details for PubMedCentralID PMC5096912

  • PTPN22 inhibition resets defective human central B cell tolerance. Science immunology Schickel, J. N., Kuhny, M., Baldo, A., Bannock, J. M., Massad, C., Wang, H., Katz, N., Oe, T., Menard, L., Soulas-Sprauel, P., Strowig, T., Flavell, R., Meffre, E. 2016; 1 (1)

    Abstract

    The 1858T protein tyrosine phosphatase nonreceptor type 22 (PTPN22 T) allele is one of the main risk factors associated with many autoimmune diseases and correlates with a defective removal of developing autoreactive B cells in humans. To determine whether inhibiting PTPN22 favors the elimination of autoreactive B cells, we first demonstrated that the PTPN22 T allele interfered with the establishment of central B cell tolerance using NOD-scid-common γ chain knockout (NSG) mice engrafted with human hematopoietic stem cells expressing this allele. In contrast, the inhibition of either PTPN22 enzymatic activity or its expression by RNA interference restored defective central B cell tolerance in this model. Thus, PTPN22 blockade may represent a therapeutic strategy for the prevention or treatment of autoimmunity.

    View details for DOI 10.1126/sciimmunol.aaf7153

    View details for PubMedID 27917411

    View details for PubMedCentralID PMC5127630

  • Rituximab does not reset defective early B cell tolerance checkpoints. The Journal of clinical investigation Chamberlain, N., Massad, C., Oe, T., Cantaert, T., Herold, K. C., Meffre, E. 2016; 126 (1): 282-7

    Abstract

    Type 1 diabetes (T1D) patients show abnormalities in early B cell tolerance checkpoints, resulting in the accumulation of large numbers of autoreactive B cells in their blood. Treatment with rituximab, an anti-CD20 mAb that depletes B cells, has been shown to preserve β cell function in T1D patients and improve other autoimmune diseases, including rheumatoid arthritis and multiple sclerosis. However, it remains largely unknown how anti-B cell therapy thwarts autoimmunity in these pathologies. Here, we analyzed the reactivity of Abs expressed by single, mature naive B cells from 4 patients with T1D before and 52 weeks after treatment to determine whether rituximab resets early B cell tolerance checkpoints. We found that anti-B cell therapy did not alter the frequencies of autoreactive and polyreactive B cells, which remained elevated in the blood of all patients after rituximab treatment. Moreover, the limited proliferative history of autoreactive B cells after treatment revealed that these clones were newly generated B cells and not self-reactive B cells that had escaped depletion and repopulated the periphery through homeostatic expansion. We conclude that anti-B cell therapy may provide a temporary dampening of autoimmune processes through B cell depletion. However, repletion with autoreactive B cells may explain the relapse that occurs in many autoimmune patients after anti-B cell therapy.

    View details for DOI 10.1172/JCI83840

    View details for PubMedID 26642366

    View details for PubMedCentralID PMC4701568

  • Activation-Induced Cytidine Deaminase Expression in Human B Cell Precursors Is Essential for Central B Cell Tolerance. Immunity Cantaert, T., Schickel, J. N., Bannock, J. M., Ng, Y. S., Massad, C., Oe, T., Wu, R., Lavoie, A., Walter, J. E., Notarangelo, L. D., Al-Herz, W., Kilic, S. S., Ochs, H. D., Nonoyama, S., Durandy, A., Meffre, E. 2015; 43 (5): 884-95

    Abstract

    Activation-induced cytidine deaminase (AID), the enzyme-mediating class-switch recombination (CSR) and somatic hypermutation (SHM) of immunoglobulin genes, is essential for the removal of developing autoreactive B cells. How AID mediates central B cell tolerance remains unknown. We report that AID enzymes were produced in a discrete population of immature B cells that expressed recombination-activating gene 2 (RAG2), suggesting that they undergo secondary recombination to edit autoreactive antibodies. However, most AID+ immature B cells lacked anti-apoptotic MCL-1 and were deleted by apoptosis. AID inhibition using lentiviral-encoded short hairpin (sh)RNA in B cells developing in humanized mice resulted in a failure to remove autoreactive clones. Hence, B cell intrinsic AID expression mediates central B cell tolerance potentially through its RAG-coupled genotoxic activity in self-reactive immature B cells.

    View details for DOI 10.1016/j.immuni.2015.10.002

    View details for PubMedID 26546282

    View details for PubMedCentralID PMC4654975

  • The Genotype and Phenotype (GAP) Registry: A National Resource for Studying Genetic Regulation of Immune Function in Normal Populations Gregersen, P., Keogh, M., Meffre, E., Nolan, G., Kimberly, R., Diamond, B. ACADEMIC PRESS INC ELSEVIER SCIENCE. 2010: S136