全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

B Lymphocytes: Development, Tolerance, and Their Role in Autoimmunity—Focus on Systemic Lupus Erythematosus

DOI: 10.1155/2013/827254

Full-Text   Cite this paper   Add to My Lib

Abstract:

B lymphocytes are the effectors of humoral immunity, providing defense against pathogens through different functions including antibody production. B cells constitute approximately 15% of peripheral blood leukocytes and arise from hemopoietic stem cells in the bone marrow. It is here that their antigen receptors (surface immunoglobulin) are assembled. In the context of autoimmune diseases defined by B and/or T cell autoreactive that upon activation lead to chronic tissue inflammation and often irreversible structural and functional damage, B lymphocytes play an essential role by not only producing autoantibodies but also functioning as antigen-presenting cells (APC) and as a source of cytokines. In this paper, we describe B lymphocyte functions in autoimmunity and autoimmune diseases with a special focus on their abnormalities in systemic lupus erythematosus. 1. Introduction Systemic lupus erythematosus (SLE) is the prototype of the systemic autoimmune diseases characterized by multiorgan involvement. This systemic compromise is mediated by a global loss of self-tolerance. The loss of tolerance is a consequence of genetic factors, in the context of specific environmental triggers, with the subsequent development of an altered immune response. Both innate and acquired immune mechanisms are implicated in the disease pathogenesis. Recently, special attention has been focused on the B cell abnormalities. In this paper, we will describe the B cell development, tolerance mechanism, and their implications in autoimmune diseases, with emphasis on SLE. 2. B Cell Development and the B Cell Receptor Formation Different populations of B cells result in preimmune pools where each cell in these quiescent populations expresses a B cell antigen receptor (BCR) with a unique specificity. When the BCRs come in contact with their specific antigen, several intracellular signals are generated leading to activation, differentiation, and formation of plasma cells and memory B cells. This last subset of B cells maintains protective antibody levels and mediates the response to subsequent antigen challenges. As the mechanisms leading to maturing and antibody production are complex, the alterations of some of these populations or critical steps have been associated with immunodeficiency and autoimmune diseases. Table 1 summarizes the most important features of each of the subpopulations (lineages) of B lymphocytes [1]. Table 1: Characteristics of primary B cell subsets and their progenitors. 2.1. B Cell Development This process begins from stem cells present in the bone marrow

References

[1]  J. F. Treml, Y. Hao, J. E. Stadanlick, and M. P. Cancro, “The BLyS family: toward a molecular understanding of B cell homeostasis,” Cell Biochemistry and Biophysics, vol. 53, no. 1, pp. 1–16, 2009.
[2]  F. A. Bonilla and H. C. Oettgen, “Adaptive immunity,” Journal of Allergy and Clinical Immunology, vol. 125, supplement 2, pp. S33–S40, 2010.
[3]  M. Fuxa and J. A. Skok, “Transcriptional regulation in early B cell development,” Current Opinion in Immunology, vol. 19, no. 2, pp. 129–136, 2007.
[4]  T. W. Lebien and T. F. Tedder, “B lymphocytes: how they develop and function,” Blood, vol. 112, no. 5, pp. 1570–1580, 2008.
[5]  Y. H. Wang and Y. J. Liu, “The IL-17 cytokine family and their role in allergic inflammation,” Current Opinion in Immunology, vol. 20, no. 6, pp. 697–702, 2008.
[6]  D. D. Chaplin, “Overview of the immune response,” Journal of Allergy and Clinical Immunology, vol. 125, supplement 2, pp. S3–S23, 2010.
[7]  D. P. Huston, “The biology of the immune system,” Journal of the American Medical Association, vol. 278, no. 22, pp. 1804–1814, 1997.
[8]  W. N. Khan, “B cell receptor and BAFF receptor signaling regulation of B cell homeostasis,” Journal of Immunology, vol. 183, no. 6, pp. 3561–3567, 2009.
[9]  T. Kurosaki and M. Hikida, “Tyrosine kinases and their substrates in B lymphocytes,” Immunological Reviews, vol. 228, no. 1, pp. 132–148, 2009.
[10]  L. Le Pottier, V. Devauchelle, J. Pers, C. Jamin, and P. Youinou, “The mosaic of B-cell subsets (with special emphasis on primary Sj?gren's syndrome),” Autoimmunity Reviews, vol. 6, no. 3, pp. 149–154, 2007.
[11]  A. Palanichamy, J. Barnard, B. Zheng et al., “Novel human transitional B cell populations revealed by B cell depletion therapy,” Journal of Immunology, vol. 182, no. 10, pp. 5982–5993, 2009.
[12]  B. Schiemann, J. L. Gommerman, K. Vora et al., “An essential role for BAFF in the normal development of B cells through a BCMA-independent pathway,” Science, vol. 293, no. 5537, pp. 2111–2114, 2001.
[13]  J. B. Chung, R. A. Sater, M. L. Fields, J. Erikson, and J. G. Monroe, “CD23 defines two distinct subsets of immature B cells which differ in their responses to T cell help signals,” International Immunology, vol. 14, no. 2, pp. 157–166, 2002.
[14]  T. Saito, S. Chiba, M. Ichikawa et al., “Notch2 is preferentially expressed in mature B cells and indispensable for marginal zone B lineage development,” Immunity, vol. 18, no. 5, pp. 675–685, 2003.
[15]  K. Willenbrock, B. Jungnickel, M. Hansmann, and R. Küppers, “Human splenic marginal zone B cells lack expression of activation-induced cytidine deaminase,” European Journal of Immunology, vol. 35, no. 10, pp. 3002–3007, 2005.
[16]  K. Reif, E. H. Ekland, L. Ohl et al., “Balanced responsiveness to chemoattractants from adjacent zones determines B-cell position,” Nature, vol. 416, no. 6876, pp. 94–99, 2002.
[17]  G. Cinamon, M. A. Zachariah, O. M. Lam, F. W. Foss Jr., and J. G. Cyster, “Follicular shuttling of marginal zone B cells facilitates antigen transport,” Nature Immunology, vol. 9, no. 1, pp. 54–62, 2008.
[18]  C. D. C. Allen and J. G. Cyster, “Follicular dendritic cell networks of primary follicles and germinal centers: phenotype and function,” Seminars in Immunology, vol. 20, no. 1, pp. 14–25, 2008.
[19]  S. Pillai and A. Cariappa, “The follicular versus marginal zone B lymphocyte cell fate decision,” Nature Reviews Immunology, vol. 9, no. 11, pp. 767–777, 2009.
[20]  R. J. Bende, F. Van Maldegem, and C. J. M. Van Noesel, “Chronic inflammatory disease, lymphoid tissue neogenesis and extranodal marginal zone B-cell lymphomas,” Haematologica, vol. 94, no. 8, pp. 1109–1123, 2009.
[21]  G. T. Hart, X. Wang, K. A. Hogquist, and S. C. Jameson, “Krüppel-like factor 2 (KLF2) regulates B-cell reactivity, subset differentiation, and trafficking molecule expression,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 2, pp. 716–721, 2011.
[22]  K. L. Hoek, L. E. Gordy, P. L. Collins et al., “Follicular B cell trafficking within the spleen actively restricts humoral immune responses,” Immunity, vol. 33, no. 2, pp. 254–265, 2010.
[23]  E. J. Goetzl, W. Wang, C. McGiffert, M. Huang, and M. H. Gr?ler, “Sphingosine 1-phosphate and its G protein-coupled receptors constitute a multifunctional immunoregulatory system,” Journal of Cellular Biochemistry, vol. 92, no. 6, pp. 1104–1114, 2004.
[24]  J. H. Wang, Q. Wu, P. Yang et al., “Type i interferon-dependent CD86high marginal zone precursor b cells are potent T cell costimulators in mice,” Arthritis and Rheumatism, vol. 63, no. 4, pp. 1054–1064, 2011.
[25]  G. Shi, K. Harrison, G. L. Wilson, C. Moratz, and J. H. Kehrl, “RGS13 regulates germinal center B lymphocytes responsiveness to CXC chemokine ligand (CXCL)12 and CXCL13,” Journal of Immunology, vol. 169, no. 5, pp. 2507–2515, 2002.
[26]  V. Pascual, Y. Liu, A. Magalski, O. De Bouteiller, J. Banchereau, and J. D. Capra, “Analysis of somatic mutation in five B cell subsets of human tonsil,” Journal of Experimental Medicine, vol. 180, no. 1, pp. 329–339, 1994.
[27]  M. Odendahl, A. Jacobi, A. Hansen et al., “Disturbed peripheral B lymphocyte homeostasis in systemic lupus erythematosus,” Journal of Immunology, vol. 165, no. 10, pp. 5970–5979, 2000.
[28]  J. Lee, S. Kuchen, R. Fischer, S. Chang, and P. E. Lipsky, “Identification and characterization of a human CD5+ pre-naive B cell population,” Journal of Immunology, vol. 182, no. 7, pp. 4116–4126, 2009.
[29]  G. P. Sims, R. Ettinger, Y. Shirota, C. H. Yarboro, G. G. Illei, and P. E. Lipsky, “Identification and characterization of circulating human transitional B cells,” Blood, vol. 105, no. 11, pp. 4390–4398, 2005.
[30]  J. O. Bohnhorst, M. B. Bj?rgan, J. E. Thoen, J. B. Natvig, and K. M. Thompson, “Bm1-Bm5 classification of peripheral blood B cells reveals circulating germinal center founder cells in healthy individuals and disturbance in the B cell subpopulations in patients with primary Sj?gren's syndrome,” Journal of Immunology, vol. 167, no. 7, pp. 3610–3618, 2001.
[31]  Y. Harada, M. M. Kawano, N. Huang et al., “Identification of early plasma cells in peripheral blood and their clinical significance,” British Journal of Haematology, vol. 92, no. 1, pp. 184–191, 1996.
[32]  A. M. Jacobi, M. Odendahl, K. Reiter et al., “Correlation between circulating CD27high plasma cells and disease activity in patients with systemic lupus erythematosus,” Arthritis and Rheumatism, vol. 48, no. 5, pp. 1332–1342, 2003.
[33]  Y. K. Parrish, I. Baez, T. Milford et al., “IL-7 dependence in human B lymphopoiesis increases during progression of ontogeny from cord blood to bone marrow,” Journal of Immunology, vol. 182, no. 7, pp. 4255–4266, 2009.
[34]  S. Giliani, L. Mori, G. De Saint Basile et al., “Interleukin-7 receptor α (IL-7Rα) deficiency: cellular and molecular bases. Analysis of clinical, immunological, and molecular features in 16 novel patients,” Immunological Reviews, vol. 203, pp. 110–126, 2005.
[35]  A. Yoshimura, T. Naka, and M. Kubo, “SOCS proteins, cytokine signalling and immune regulation,” Nature Reviews Immunology, vol. 7, no. 6, pp. 454–465, 2007.
[36]  P. Youinou, T. E. Taher, J. Pers, R. A. Mageed, and Y. Renaudineau, “B lymphocyte cytokines and rheumatic autoimmune disease,” Arthritis and Rheumatism, vol. 60, no. 7, pp. 1873–1880, 2009.
[37]  D. I. Mitsias, A. G. Tzioufas, C. Veiopoulou et al., “The Th1/Th2 cytokine balance changes with the progress of the immunopathological lesion of Sjogren's syndrome,” Clinical and Experimental Immunology, vol. 128, no. 3, pp. 562–568, 2002.
[38]  P. Szodoray, P. Alex, J. G. Brun, M. Centola, and R. Jonsson, “Circulating cytokines in primary Sj?gren's syndrome determined by a multiplex cytokine array system,” Scandinavian Journal of Immunology, vol. 59, no. 6, pp. 592–599, 2004.
[39]  M. Fuxa and M. Busslinger, “Reporter gene insertions reveal a strictly B lymphoid-specific expression pattern of Pax5 in support of its B cell identity function,” Journal of Immunology, vol. 178, no. 12, pp. 8222–8228, 2007.
[40]  S. Yurasov, H. Wardemann, J. Hammersen et al., “Defective B cell tolerance checkpoints in systemic lupus erythematosus,” Journal of Experimental Medicine, vol. 201, no. 5, pp. 703–711, 2005.
[41]  G. Meyersa, Y. S. Nga, J. M. Bannocka, et al., “Activation-induced cytidine deaminase (AID) is required for B-cell tolerance in humans,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 28, pp. 11554–11559, 2011.
[42]  E. T. Luning Prak, M. Monestier, and R. A. Eisenberg, “B cell receptor editing in tolerance and autoimmunity,” Annals of the New York Academy of Sciences, vol. 1217, no. 1, pp. 96–121, 2011.
[43]  Y. Bergman and H. Cedar, “A stepwise epigenetic process controls immunoglobulin allelic exclusion,” Nature Reviews Immunology, vol. 4, no. 10, pp. 753–761, 2004.
[44]  R. C. Lindsley, M. Thomas, B. Srivastava, and D. Allman, “Generation of peripheral B cells occurs via two spatially and temporally distinct pathways,” Blood, vol. 109, no. 6, pp. 2521–2529, 2007.
[45]  A. Br?uninger, T. Goossens, K. Rajewsky, and R. Küppers, “Regulation of immunoglobulin light chain gene rearrangements during early B cell development in the human,” European Journal of Immunology, vol. 31, no. 12, pp. 3631–3637, 2001.
[46]  K. D. Klonowski, L. L. Primiano, and M. Monestier, “Atypical V(H)-D-J(H) rearrangements in newborn autoimmune MRL mice,” Journal of Immunology, vol. 162, no. 3, pp. 1566–1572, 1999.
[47]  K. D. Klonowski and M. Monestier, “Heavy chain revision in MRL mice: a potential mechanism for the development of autoreactive B cell precursors,” Journal of Immunology, vol. 165, no. 8, pp. 4487–4493, 2000.
[48]  E. Derudder, E. J. Cadera, J. C. Vahl et al., “Development of immunoglobulin λ-chain-positive B cells, but not editing of immunoglobulin κ-chain, depends on NF-κB signals,” Nature Immunology, vol. 10, no. 6, pp. 647–654, 2009.
[49]  R. J. Benschop, D. Melamed, D. Nemazee, and J. C. Cambier, “Distinct signal thresholds for the unique antigen receptor-linked gene expression programs in mature and immature B cells,” Journal of Experimental Medicine, vol. 190, no. 6, pp. 749–756, 1999.
[50]  L. Verkoczy, B. Duong, P. Skog et al., “Basal B cell receptor-directed phosphatidylinositol 3-kinase signaling turns off RAGs and promotes B cell-positive selection,” Journal of Immunology, vol. 178, no. 10, pp. 6332–6341, 2007.
[51]  M. J. Shlomchik, A. Marshak-Rothstein, and C. B. Wolfowicz, “The role of clonal selection and somatic mutation in autoimmunity,” Nature, vol. 328, no. 6133, pp. 805–811, 1987.
[52]  T. Litzenburger, H. Bluthmann, P. Morales et al., “Development of myelin oligodendrocyte glycoprotein autoreactive transgenic B lymphocytes: receptor editing in vivo after encounter of a self-antigen distinct from myelin oligodendrocyte glycoprotein,” Journal of Immunology, vol. 165, no. 9, pp. 5360–5366, 2000.
[53]  T. Litzenburger, R. F?ssler, J. Bauer et al., “B lymphocytes producing demyelinating autoantibodies: development and function in gene-targeted transgenic mice,” Journal of Experimental Medicine, vol. 188, no. 1, pp. 169–180, 1998.
[54]  L. Menard, D. Saadoun, I. Isnardi et al., “The PTPN22 allele encoding an R620W variant interferes with the removal of developing autoreactive B cells in humans,” Journal of Clinical Investigation, vol. 121, no. 9, pp. 3635–3644, 2011.
[55]  M. Inaoki, S. Sato, B. C. Weintraub, C. C. Goodnow, and T. F. Tedder, “CD19-regulated signaling thresholds control peripheral tolerance and autoantibody production in B lymphocytes,” Journal of Experimental Medicine, vol. 186, no. 11, pp. 1923–1931, 1997.
[56]  T. Guerrier, P. Youinou, J. O. Pers, and C. Jamin, “TLR9 drives the development of transitional B cells towards the marginal zone pathway and promotes autoimmunity,” Journal of Autoimmunity, vol. 39, no. 3, pp. 173–179, 2012.
[57]  J. A. Lyons, M. San, M. P. Happ, and A. H. Cross, “B cells are critical to induction of experimental allergic encephalomyelitis by protein but not by a short encephalitogenic peptide,” European Journal of Immunology, vol. 29, no. 11, pp. 3432–3439, 1999.
[58]  E. Bettelli, D. Baeten, A. J?ger, R. A. Sobel, and V. K. Kuchroo, “Myelin oligodendrocyte glycoprotein-specific T and B cells cooperate to induce a Devic-like disease in mice,” Journal of Clinical Investigation, vol. 116, no. 9, pp. 2393–2402, 2006.
[59]  G. Krishnamoorthy, H. Lassmann, H. Wekerle, and A. Holz, “Spontaneous opticospinal encephalomyelitis in a double-transgenic mouse model of autoimmune T cell/B cell cooperation,” Journal of Clinical Investigation, vol. 116, no. 9, pp. 2385–2392, 2006.
[60]  N. L. Monson, P. Cravens, R. Hussain et al., “Rituximab therapy reduces organ-specific T cell responses and ameliorates experimental autoimmune encephalomyelitis,” PLoS ONE, vol. 6, no. 2, Article ID e17103, 2011.
[61]  O. T. Chan, L. G. Hannum, A. M. Haberman, M. P. Madaio, and M. J. Shlomchik, “A novel mouse with B cells but lacking serum antibody reveals an antibody-independent role for B cells in murine lupus,” Journal of Experimental Medicine, vol. 189, no. 10, pp. 1639–1648, 1999.
[62]  F. S. Wong, L. Wen, M. Tang et al., “Investigation of the role of B-cells in type 1 diabetes in the NOD mouse,” Diabetes, vol. 53, no. 10, pp. 2581–2587, 2004.
[63]  G. Tchernev and C. E. Orfanos, “Antigen mimicry, epitope spreading and the pathogenesis of pemphigus,” Tissue Antigens, vol. 68, no. 4, pp. 280–286, 2006.
[64]  J. Sokolove, R. Bromberg, K. D. Deane, et al., “Autoantibody epitope spreading in the pre-clinical phase predicts progression to rheumatoid arthritis,” PLoS One, vol. 7, Article ID e35296, 2012.
[65]  B. L. McRae, C. L. Vanderlugt, M. C. Dal Canto, and S. D. Miller, “Functional evidence for epitope spreading in the relapsing pathology of experimental autoimmune encephalomyelitis,” Journal of Experimental Medicine, vol. 182, no. 1, pp. 75–85, 1995.
[66]  R. Tisch, X.-D. Yang, S. M. Singer, R. S. Liblau, L. Fugger, and H. O. McDevitt, “Immune response to glutamic acid decarboxylase correlates with insulitis in non-obese diabetic mice,” Nature, vol. 366, no. 6450, pp. 72–75, 1993.
[67]  A. G. Ziegler, M. Hummel, M. Schenker, and E. Bonifacio, “Autoantibody appearance and risk for development of childhood diabetes in offspring of parents with type 1 diabetes: the 2-year analysis of the German BABYDIAB Study,” Diabetes, vol. 48, no. 3, pp. 460–468, 1999.
[68]  D. van der Woude, S. Rantap??-Dahlqvist, A. Ioan-Facsinay et al., “Epitope spreading of the anti-citrullinated protein antibody response occurs before disease onset and is associated with the disease course of early arthritis,” Annals of the Rheumatic Diseases, vol. 69, no. 8, pp. 1554–1561, 2010.
[69]  M. R. Arbuckle, M. T. McClain, M. V. Rubertone et al., “Development of autoantibodies before the clinical onset of systemic lupus erythematosus,” New England Journal of Medicine, vol. 349, no. 16, pp. 1526–1533, 2003.
[70]  P. Schneider, F. Mackay, V. Steiner et al., “BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth,” Journal of Experimental Medicine, vol. 189, no. 11, pp. 1747–1756, 1999.
[71]  F. Mackay and J. L. Browning, “BAFF: a fundamental survival factor for B cells,” Nature Reviews Immunology, vol. 2, no. 7, pp. 465–475, 2002.
[72]  F. Mackay, S. A. Woodcock, P. Lawton et al., “Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations,” Journal of Experimental Medicine, vol. 190, no. 11, pp. 1697–1710, 1999.
[73]  R. Brink, “Regulation of B cell self-tolerance by BAFF,” Seminars in Immunology, vol. 18, no. 5, pp. 276–283, 2006.
[74]  B. Schiemann, J. L. Gommerman, K. Vora et al., “An essential role for BAFF in the normal development of B cells through a BCMA-independent pathway,” Science, vol. 293, no. 5537, pp. 2111–2114, 2001.
[75]  R. Lesley, Y. Xu, S. L. Kalled et al., “Reduced competitiveness of autoantigen-engaged B cells due to increased dependence on BAFF,” Immunity, vol. 20, no. 4, pp. 441–453, 2004.
[76]  M. Thien, T. G. Phan, S. Gardam et al., “Excess BAFF rescues self-reactive B cells from peripheral deletion and allows them to enter forbidden follicular and marginal zone niches,” Immunity, vol. 20, no. 6, pp. 785–798, 2004.
[77]  J. R. Groom, C. A. Fletcher, S. N. Walters et al., “BAFF and MyD88 signals promote a lupuslike disease independent of T cells,” Journal of Experimental Medicine, vol. 204, no. 8, pp. 1959–1971, 2007.
[78]  F. Mackay, J. R. Groom, and S. G. Tangye, “An important role for B-cell activation factor and B cells in the pathogenesis of Sj?gren's syndrome,” Current Opinion in Rheumatology, vol. 19, no. 5, pp. 406–413, 2007.
[79]  P. Youinou, C. Jamin, and P. M. Lydyard, “CD5 expression in human B-cell populations,” Immunology Today, vol. 20, no. 7, pp. 312–316, 1999.
[80]  R. Berland and H. H. Wortis, “Origins and functions of B-1 cells with notes on the role of CD5,” Annual Review of Immunology, vol. 20, pp. 253–300, 2002.
[81]  K. Yanaba, J. D. Bouaziz, K. M. Haas, J. C. Poe, M. Fujimoto, and T. F. Tedder, “A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses,” Immunity, vol. 28, no. 5, pp. 639–650, 2008.
[82]  C. Mauri, D. Gray, N. Mushtaq, and M. Londei, “Prevention of arthritis by interleukin 10-producing B cells,” Journal of Experimental Medicine, vol. 197, no. 4, pp. 489–501, 2003.
[83]  S. Garaud, A. Morva, S. Lemoine et al., “CD5 promotes IL-10 production in chronic lymphocytic leukemia B cells through STAT3 and NFAT2 activation,” Journal of Immunology, vol. 186, no. 8, pp. 4835–4844, 2011.
[84]  Y. Renaudineau, S. Hillion, A. Saraux, R. A. Mageed, and P. Youinou, “An alternative exon 1 of the CD5 gene regulates CD5 expression in human B lymphocytes,” Blood, vol. 106, no. 8, pp. 2781–2789, 2005.
[85]  S. Garaud, C. Le Dantec, S. Jousse-Joulin et al., “IL-6 Modulates CD5 expression in B cells from patients with lupus by regulating DNA methylation,” Journal of Immunology, vol. 182, no. 9, pp. 5623–5632, 2009.
[86]  S. Hillion, A. Saraux, P. Youinou, and C. Jamin, “Expression of RAGs in peripheral B cells outside Germinal Centers is associated with the expression of CD5,” Journal of Immunology, vol. 174, no. 9, pp. 5553–5561, 2005.
[87]  S. Hillion, M. Dueymes, P. Youinou, and C. Jamin, “IL-6 contributes to the expression of RAGs in human mature B cells,” Journal of Immunology, vol. 179, no. 10, pp. 6790–6798, 2007.
[88]  K. L. Hippen, L. E. Tze, and T. W. Behrens, “CD5 maintains tolerance in anergic B cells,” Journal of Experimental Medicine, vol. 191, no. 5, pp. 883–889, 2000.
[89]  J. C. Poe, M. Hasegawa, and T. F. Tedder, “CD19, CD21, and CD22: multifaceted response regulators of B lymphocyte signal transduction,” International Reviews of Immunology, vol. 20, no. 6, pp. 739–762, 2001.
[90]  N. R. Pritchard and K. G. C. Smith, “B cell inhibitory receptors and autoimmunity,” Immunology, vol. 108, no. 3, pp. 263–273, 2003.
[91]  J. G. Cyster and C. C. Goodnow, “Tuning antigen receptor signaling by CD22: integrating cues from antigens and the microenvironment,” Immunity, vol. 6, no. 5, pp. 509–517, 1997.
[92]  T. F. Tedder, J. Tuscano, S. Sato, and J. H. Kehrl, “CD22, A B lymphocyte-specific adhesion molecule that regulates antigen receptor signaling,” Annual Review of Immunology, vol. 15, pp. 481–504, 1997.
[93]  S. Sato, A. S. Miller, M. Inaoki et al., “CD22 is both a positive and negative regulator of B lymphocyte antigen receptor signal transduction: altered signaling in CD22-deficient mice,” Immunity, vol. 5, no. 6, pp. 551–562, 1996.
[94]  L. Nitschke, “The role of CD22 and other inhibitory co-receptors in B-cell activation,” Current Opinion in Immunology, vol. 17, no. 3, pp. 290–297, 2005.
[95]  P. Engel, Y. Nojima, D. Rothstein et al., “The same epitope on CD22 of B lymphocytes mediates the adhesion of erythrocytes, T and B lymphocytes, neutrophils, and monocytes,” Journal of Immunology, vol. 150, no. 11, pp. 4719–4732, 1993.
[96]  T. F. Tedder, J. C. Poe, and K. M. Haas, “CD22: a multifunctional receptor that regulates B lymphocyte survival and signal transduction,” Advances in Immunology, vol. 88, pp. 1–50, 2005.
[97]  C. M. Grimaldi, R. Hicks, and B. Diamond, “B cell selection and susceptibility to autoimmunity,” Journal of Immunology, vol. 174, no. 4, pp. 1775–1781, 2005.
[98]  J. A. Walker and K. G. C. Smith, “CD22: an inhibitory enigma,” Immunology, vol. 123, no. 3, pp. 314–325, 2008.
[99]  L. Jin, P. A. McLean, B. G. Neel, and H. H. Wortis, “Sialic acid binding domains of CD22 are required for negative regulation of B cell receptor signaling,” Journal of Experimental Medicine, vol. 195, no. 9, pp. 1199–1205, 2002.
[100]  T. L. O'Keefe, G. T. Williams, S. L. Davies, and M. S. Neuberger, “Hyperresponsive B cells in CD22-deficient mice,” Science, vol. 274, no. 5288, pp. 798–801, 1996.
[101]  T. D?rner, J. Kaufmann, W. A. Wegener, N. Teoh, D. M. Goldenberg, and G. R. Burmester, “Initial clinical trial of epratuzumab (humanized anti-CD22 antibody) for immunotherapy of systemic lupus erythematosus,” Arthritis Research and Therapy, vol. 8, no. 3, p. R74, 2006.
[102]  A. M. Jacobi, D. M. Goldenberg, F. Hiepe, A. Radbruch, G. R. Burmester, and T. D?rner, “Differential effects of epratuzumab on peripheral blood B cells of patients with systemic lupus erythematosus versus normal controls,” Annals of the Rheumatic Diseases, vol. 67, no. 4, pp. 450–457, 2008.
[103]  S. D. Steinfeld, L. Tant, G. R. Burmester et al., “Epratuzumab (humanised anti-CD22 antibody) in primary Sj?gren's syndrome: an open-label phase I/II study,” Arthritis Research and Therapy, vol. 8, no. 4, p. R129, 2006.
[104]  A. Mizoguchi, E. Mizoguchi, H. Takedatsu, R. S. Blumberg, and A. K. Bhan, “Chronic intestinal inflammatory condition generates IL-10-producing regulatory B cell subset characterized by CD1d upregulation,” Immunity, vol. 16, no. 2, pp. 219–230, 2002.
[105]  S. Fillatreau, C. H. Sweenie, M. J. McGeachy, D. Gray, and S. M. Anderton, “B cells regulate autoimmunity by provision of IL-10,” Nature Immunology, vol. 3, no. 10, pp. 944–950, 2002.
[106]  C. Mauri, D. Gray, N. Mushtaq, and M. Londei, “Prevention of arthritis by interleukin 10-producing B cells,” Journal of Experimental Medicine, vol. 197, no. 4, pp. 489–501, 2003.
[107]  B. Wei, P. Velazquez, O. Turovskaya et al., “Mesenteric B cells centrally inhibit CD4+ T cell colitis through interaction with regulatory T cell subsets,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 6, pp. 2010–2015, 2005.
[108]  T. Tretter, R. K. C. Venigalla, V. Eckstein et al., “Induction of CD4+ T-cell anergy and apoptosis by activated human B cells,” Blood, vol. 112, no. 12, pp. 4555–4564, 2008.
[109]  A. Mizoguchi, E. Mizoguchi, R. N. Smith, F. I. Preffer, and A. K. Bhan, “Suppressive role of B cells in chronic colitis of T cell receptor α mutant mice,” Journal of Experimental Medicine, vol. 186, no. 10, pp. 1749–1756, 1997.
[110]  J. G. Evans, K. A. Chavez-Rueda, A. Eddaoudi et al., “Novel suppressive function of transitional 2 B cells in experimental arthritis,” Journal of Immunology, vol. 178, no. 12, pp. 7868–7878, 2007.
[111]  P. A. Blair, K. A. Chavez-Rueda, J. G. Evans et al., “Selective targeting of B cells with agonistic anti-CD40 is an efficacious strategy for the generation of induced regulatory T2-like B cells and for the suppression of lupus in MRL/lpr mice,” Journal of Immunology, vol. 182, no. 6, pp. 3492–3502, 2009.
[112]  P. A. Blair, L. Y. Nore?a, F. Flores-Borja et al., “CD19+CD24hiCD38hi B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic lupus erythematosus patients,” Immunity, vol. 32, no. 1, pp. 129–140, 2010.
[113]  C. Jamin, A. Morva, S. Lemoine, C. Daridon, A. R. De Mendoza, and P. Youinou, “Regulatory B lymphocytes in humans: a potential role in autoimmunity,” Arthritis and Rheumatism, vol. 58, no. 7, pp. 1900–1906, 2008.
[114]  M. Duddy, M. Niino, F. Adatia et al., “Distinct effector cytokine profiles of memory and naive human B cell subsets and implication in multiple sclerosis,” Journal of Immunology, vol. 178, no. 10, pp. 6092–6099, 2007.
[115]  J. Correale, M. Farez, and G. Razzitte, “Helminth infections associated with multiple sclerosis induce regulatory B cells,” Annals of Neurology, vol. 64, no. 2, pp. 187–199, 2008.
[116]  C. Mauri, “Regulation of immunity and autoimmunity by B cells,” Current Opinion in Immunology, vol. 22, no. 6, pp. 761–767, 2010.
[117]  S. Lemoine, A. Morva, P. Youinou, and C. Jamin, “Human T cells induce their own regulation through activation of B cells,” Journal of Autoimmunity, vol. 36, no. 3-4, pp. 228–238, 2011.
[118]  A. Morva, S. Lemoine, A. Achour, J. Pers, P. Youinou, and C. Jamin, “Maturation and function of human dendritic cells are regulated by B lymphocytes,” Blood, vol. 119, no. 1, pp. 106–114, 2012.
[119]  S. Lemoine, A. Morva, P. Youinou, and C. Jamin, “Regulatory B cells in autoimmune diseases: how do they work,” Annals of the New York Academy of Sciences, vol. 1173, pp. 260–267, 2009.
[120]  D. A. Einfeld, J. P. Brown, M. A. Valentine, E. A. Clark, and J. A. Ledbetter, “Molecular cloning of the human B cell CD20 receptor predicts a hydrophobic protein with multiple transmembrane domains,” EMBO Journal, vol. 7, no. 3, pp. 711–717, 1988.
[121]  M. A. Valentine, K. E. Meier, S. Rossie, and E. A. Clark, “Phosphorylation of the CD20 phosphoprotein in resting B lymphocytes. Regulation by protein kinase C,” Journal of Biological Chemistry, vol. 264, no. 19, pp. 11282–11287, 1989.
[122]  K. C. Anderson, M. P. Bates, and B. L. Slaughenhoupt, “Expression of human B cell-associated antigens on leukemias and lymphomas: a model of human B cell differentiation,” Blood, vol. 63, no. 6, pp. 1424–1433, 1984.
[123]  J. H. Anolik, J. Barnard, A. Cappione et al., “Rituximab improves peripheral B cell abnormalities in human systemic lupus erythematosus,” Arthritis and Rheumatism, vol. 50, no. 11, pp. 3580–3590, 2004.
[124]  M. J. Leandro, J. C. Edwards, G. Cambridge, M. R. Ehrenstein, and D. A. Isenberg, “An open study of B lymphocyte depletion in systemic lupus erythematosus,” Arthritis and Rheumatism, vol. 46, no. 10, pp. 2673–2677, 2002.
[125]  M. Ramos-Casals, M. J. Soto, M. J. Cuadrado, and M. A. Khamashta, “Rituximab in systemic lupus erythematosus A systematic review of off-label use in 188 cases,” Lupus, vol. 18, no. 9, pp. 767–776, 2009.
[126]  J. Merrill, J. Buyon, R. Furie et al., “Assessment of flares in lupus patients enrolled in a phase II/III study of rituximab (EXPLORER),” Lupus, vol. 20, no. 7, pp. 709–716, 2011.
[127]  B. H. Rovin, R. Furie, K. Latinis, et al., “Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study,” Arthritis & Rheumatism, vol. 64, no. 4, pp. 1215–1226, 2012.
[128]  C. Daridon, D. Blassfeld, K. Reiter et al., “Epratuzumab targeting of CD22 affects adhesion molecule expression and migration of B-cells in systemic lupus erythematosus,” Arthritis Research and Therapy, vol. 12, no. 6, p. R204, 2010.
[129]  S. V. Navarra, R. M. Guzmán, A. E. Gallacher et al., “Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial,” The Lancet, vol. 377, no. 9767, pp. 721–731, 2011.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413