Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Immunol ; 205(12): 3263-3276, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33199538

RESUMO

Signaling lymphocytic activation molecule-associated protein (SAP), a critical intracellular signaling molecule for T-B lymphocyte interactions, drives T follicular helper (Tfh) cell development in germinal centers (GCs). High-affinity islet autoantibodies predict type 1 diabetes (T1D) but do not cause ß cell destruction. This paradox intimates Tfh cells as key pathologic effectors, consistent with an observed Tfh signature in T1D. To understand how fully developed Tfh (GC Tfh) contribute to different autoimmune processes, we investigated the role of SAP in T1D and autoantibody-mediated arthritis. Whereas spontaneous arthritis depended on SAP in the autoantibody-mediated K/BxN model, organized insulitis and diabetes onset were unabated, despite a blocked anti-insulin vaccine response in SAP-deficient NOD mice. GC Tfh and GC B cell development were blocked by loss of SAP in K/BxN mice. In contrast, although GC B cell formation was markedly reduced in SAP-deficient NOD mice, T cells with a GC Tfh phenotype were found at disease sites. CXCR3+ CCR6- (Tfh1) subset bias was observed among GC Tfh cells infiltrating the pancreas of NOD mice, which was enhanced by loss of SAP NOD T cells override SAP requirement to undergo activation and proliferation in response to Ag presentation, demonstrating the potential for productive cognate T-B lymphocyte interactions in T1D-prone mice. We find that SAP is essential when autoantibody-driven immune complexes promote inflammation but is not required for effective organ-specific autoimmune attack. Thus, Tfh induced in classic GC reactions are dispensable for T1D, but the autoimmune process in the NOD model retains pathogenic Tfh without SAP.


Assuntos
Linfócitos B/imunologia , Comunicação Celular/imunologia , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/imunologia , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária/imunologia , Células Th1/imunologia , Animais , Autoanticorpos/genética , Autoanticorpos/imunologia , Linfócitos B/patologia , Comunicação Celular/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária/genética , Células Th1/patologia
2.
J Immunol ; 201(3): 861-873, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29950508

RESUMO

Early breaches in B cell tolerance are central to type 1 diabetes progression in mouse and man. Conventional BCR transgenic mouse models (VH125.Tg NOD) reveal the power of B cell specificity to drive disease as APCs. However, in conventional fixed IgM models, comprehensive assessment of B cell development is limited. To provide more accurate insight into the developmental and functional fates of anti-insulin B cells, we generated a new NOD model (VH125SDNOD) in which anti-insulin VDJH125 is targeted to the IgH chain locus to generate a small (1-2%) population of class switch-competent insulin-binding B cells. Tracking of this rare population in a polyclonal repertoire reveals that anti-insulin B cells are preferentially skewed into marginal zone and late transitional subsets known to have increased sensitivity to proinflammatory signals. Additionally, IL-10 production, characteristic of regulatory B cell subsets, is increased. In contrast to conventional models, class switch-competent anti-insulin B cells proliferate normally in response to mitogenic stimuli but remain functionally silent for insulin autoantibody production. Diabetes development is accelerated, which demonstrates the power of anti-insulin B cells to exacerbate disease without differentiation into Ab-forming or plasma cells. Autoreactive T cell responses in VH125SDNOD mice are not restricted to insulin autoantigens, as evidenced by increased IFN-γ production to a broad array of diabetes-associated epitopes. Together, these results independently validate the pathogenic role of anti-insulin B cells in type 1 diabetes, underscore their diverse developmental fates, and demonstrate the pathologic potential of coupling a critical ß cell specificity to predominantly proinflammatory Ag-presenting B cell subsets.


Assuntos
Apresentação de Antígeno/imunologia , Subpopulações de Linfócitos B/imunologia , Diabetes Mellitus Tipo 1/imunologia , Anticorpos Anti-Insulina/imunologia , Insulina/imunologia , Animais , Autoanticorpos/imunologia , Autoantígenos/imunologia , Feminino , Tolerância Imunológica/imunologia , Inflamação/imunologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos B/imunologia
3.
J Immunol ; 195(3): 853-64, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26109644

RESUMO

Autoreactive B lymphocytes that escape central tolerance and mature in the periphery are a liability for developing autoimmunity. IgG insulin autoantibodies that predict type 1 diabetes and complicate insulin therapies indicate that mechanisms for tolerance to insulin are flawed. To examine peripheral tolerance in anti-insulin B cells, we generated C57BL/6 mice that harbor anti-insulin VDJH-125 site directed to the native IgH locus (VH125(SD)). Class switch-competent anti-insulin B cells fail to produce IgG Abs following T cell-dependent immunization of VH125(SD) mice with heterologous insulin, and they exhibit markedly impaired proliferation to anti-CD40 plus insulin in vitro. In contrast, costimulation with LPS plus insulin drives robust anti-insulin B cell proliferation. Furthermore, VH125(SD) mice produce both IgM and IgG2a anti-insulin Abs following immunization with insulin conjugated to type 1 T cell-independent Brucella abortus ring test Ag (BRT). Anti-insulin B cells undergo clonal expansion in vivo and emerge as IgM(+) and IgM(-) GL7(+)Fas(+) germinal center (GC) B cells following immunization with insulin-BRT, but not BRT alone. Analysis of Igκ genes in VH125(SD) mice immunized with insulin-BRT reveals that anti-insulin Vκ from the preimmune repertoire is selected into GCs. These data demonstrate that class switch-competent anti-insulin B cells remain functionally silent in T cell-dependent immune responses, yet these B cells are vulnerable to reversal of anergy following combined BCR/TLR engagement that promotes Ag-specific GC responses and Ab production. Environmental factors that lead to infection and inflammation could play a critical yet underappreciated role in driving loss of tolerance and promoting autoimmune disease.


Assuntos
Autoanticorpos/imunologia , Linfócitos B/imunologia , Diabetes Mellitus Tipo 1/imunologia , Anticorpos Anti-Insulina/imunologia , Insulina/imunologia , Animais , Autoanticorpos/biossíntese , Autoimunidade/imunologia , Antígenos CD40/imunologia , Diabetes Mellitus Tipo 1/genética , Tolerância Imunológica/imunologia , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Imunoglobulina M/biossíntese , Imunoglobulina M/imunologia , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Éxons VDJ/imunologia
4.
J Immunol ; 189(2): 711-20, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22675201

RESUMO

The ability to induce Ab responses to pathogens while maintaining the quiescence of autoreactive cells is an important aspect of immune tolerance. During activation of TLR4, dendritic cells (DCs) and macrophages (MFs) repress autoantibody production through their secretion of IL-6 and soluble CD40L (sCD40L). These soluble mediators selectively repress B cells chronically exposed to Ag, but not naive cells, suggesting a means to maintain tolerance during TLR4 stimulation, yet allow immunity. In this study, we identify TNF-α as a third repressive factor, which together with IL-6 and CD40L account for nearly all the repression conferred by DCs and MFs. Similar to IL-6 and sCD40L, TNF-α did not alter B cell proliferation or survival. Instead, it reduced the number of Ab-secreting cells. To address whether the soluble mediators secreted by DCs and MFs functioned in vivo, we generated mice lacking IL-6, CD40L, and TNF-α. Compared to wild-type mice, these mice showed prolonged anti-nuclear Ab responses following TLR4 stimulation. Furthermore, adoptive transfer of autoreactive B cells into chimeric IL-6(-/-) × CD40L(-/-) × TNF-α(-/-) mice showed that preplasma cells secreted autoantibodies independent of germinal center formation or extrafollicular foci. These data indicate that in the absence of genetic predisposition to autoimmunity, loss of endogenous IL-6, CD40L, and TNF-α promotes autoantibody secretion during TLR4 stimulation.


Assuntos
Autoanticorpos/biossíntese , Células Dendríticas/imunologia , Tolerância Imunológica , Macrófagos/imunologia , Plasmócitos/imunologia , Células-Tronco/imunologia , Transferência Adotiva , Animais , Antígenos Nucleares/genética , Antígenos Nucleares/imunologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Ligante de CD40/deficiência , Células Cultivadas , Células Dendríticas/metabolismo , Tolerância Imunológica/genética , Interleucina-6/deficiência , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Camundongos Knockout , Camundongos Transgênicos , Plasmócitos/metabolismo , Plasmócitos/transplante , Quimera por Radiação/imunologia , Células-Tronco/metabolismo , Receptor 4 Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/deficiência
5.
J Immunol ; 183(10): 6403-12, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19841184

RESUMO

Type 1 diabetes results from T cell-mediated destruction of insulin-producing beta cells. Although elimination of B lymphocytes has proven successful at preventing disease, modulation of B cell function as a means to prevent type 1 diabetes has not been investigated. The development, fate, and function of B lymphocytes depend upon BCR signaling, which is mediated in part by Bruton's tyrosine kinase (BTK). When introduced into NOD mice, btk deficiency only modestly reduces B cell numbers, but dramatically protects against diabetes. In NOD, btk deficiency mirrors changes in B cell subsets seen in other strains, but also improves B cell-related tolerance, as indicated by failure to generate insulin autoantibodies. Introduction of an anti-insulin BCR H chain transgene restores diabetes in btk-deficient NOD mice, indicating that btk-deficient B cells are functionally capable of promoting autoimmune diabetes if they have a critical autoimmune specificity. This suggests that the disease-protective effect of btk deficiency may reflect a lack of autoreactive specificities in the B cell repertoire. Thus, signaling via BTK can be modulated to improve B cell tolerance, and prevent T cell-mediated autoimmune diabetes.


Assuntos
Autoanticorpos/imunologia , Subpopulações de Linfócitos B/imunologia , Anticorpos Anti-Insulina/imunologia , Insulina/imunologia , Proteínas Tirosina Quinases/imunologia , Subpopulações de Linfócitos T/imunologia , Tirosina Quinase da Agamaglobulinemia , Animais , Autoanticorpos/metabolismo , Subpopulações de Linfócitos B/metabolismo , Diabetes Mellitus Tipo 1 , Tolerância Imunológica/genética , Tolerância Imunológica/imunologia , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/metabolismo , Insulina/metabolismo , Anticorpos Anti-Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Mutação/genética , Mutação/imunologia , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Receptores de Antígenos de Linfócitos B/genética , Receptores de Antígenos de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/metabolismo , Transgenes
6.
Immunogenetics ; 62(8): 507-20, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20556377

RESUMO

The diversity of immunoglobulin (Ig) and T cell receptor (TCR) genes available to form the lymphocyte repertoire has the capacity to produce a broad array of both protective and harmful specificities. In type 1 diabetes (T1D), the presence of antibodies to insulin and other islet antigens predicts disease development in both mice and humans, and demonstrate that immune tolerance is lost early in the disease process. Anti-insulin T cells isolated from T1D-prone non-obese diabetic (NOD) mice use polymorphic TCRalpha chains, suggesting that the available T cell repertoire is altered in these autoimmune mice. To probe whether insulin-binding B cells also possess polymorphic V genes, Ig light chains were isolated and sequenced from NOD mice that harbor an Ig heavy chain transgene. Three insulin-binding Vkappa genes were identified, all of which were polymorphic to the closest germline sequence matches present in the GenBank database. Additional analysis of over 300 light chain sequences from multiple sources, including germline DNA, shows that polymorphisms are spread throughout the entire NOD Igkappa locus, as these polymorphic sequences represent 43 distinct Vkappa genes which belong to 14 Vkappa families. Database searches reveal that a majority of polymorphic Vkappa genes identified in NOD are identical to Vkappa genes isolated from SLE-prone NZBxNZW F1 or MRL strains of mice, suggesting that a shared Igkappa haplotype may be present. Predicted amino acid changes preferentially occur in CDR, and thus could alter antigen recognition by the germline B cell repertoire of autoimmune versus non-autoimmune mouse strains.


Assuntos
Autoimunidade/genética , Genes de Cadeia Leve de Imunoglobulina , Cadeias kappa de Imunoglobulina/genética , Polimorfismo Genético , Substituição de Aminoácidos , Animais , Linfócitos B/imunologia , Sequência de Bases , Primers do DNA/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Genes de Cadeia Pesada de Imunoglobulina , Região Variável de Imunoglobulina/genética , Insulina/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Dados de Sequência Molecular , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie , Linfócitos T/imunologia
7.
ACS Appl Bio Mater ; 3(9): 6319-6330, 2020 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-35021762

RESUMO

Adaptive immunity plays a central role in the pathogenesis of type 1 diabetes. Among past treatment approaches, B cell ablation has yielded unmistakable therapeutic potency; however, global immunosuppression imposes unacceptable risks to a patient population consisting of children. Multivalent antigen arrays represent a compelling strategy for targeted immunosuppression by selectively engaging and inactivating autoreactive B cells. Here, we report the design and characterization of 4-arm polyethylene glycol-insulin (PEG-Ins) conjugates as multivalent arrays for autoreactive B cell engagement. First, we selectively modified human insulin at the B29 residue to retain antigenicity. Next, we conjugated the modified proteins to 20 kDa, 4-arm polyethylene glycol backbones to produce multivalent PEG-Ins constructs. Mass spectrometry, circular dichroism, and dynamic light scattering indicated that the structure of insulin was maintained in the much larger, multivalent construct. PEG-Ins conjugates demonstrated an ex vivo immunological effect in splenocytes harboring an anti-insulin B cell receptor (VH125SD) by inactivating B cells and promoting an anergic phenotype that was downregulated in B cell receptor expression (CD79b), and PEG-Ins conjugates did not mobilize calcium upon B cell receptor stimulation. These data support the further study of PEG-Ins conjugates in animal models of type 1 diabetes.

8.
J Immunol ; 174(2): 827-33, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15634904

RESUMO

Loss of tolerance is considered to be an early event that is essential for the development of autoimmune disease. In contrast to this expectation, autoimmune (type 1) diabetes develops in NOD mice that harbor an anti-insulin Ig transgene (125Tg), even though anti-insulin B cells are tolerant. Tolerance is maintained in a similar manner in both normal C57BL/6 and autoimmune NOD mice, as evidenced by B cell anergy to stimulation through their Ag receptor (anti-IgM), TLR4 (LPS), and CD40 (anti-CD40). Unlike B cells in other models of tolerance, anergic 125Tg B cells are not arrested in development, and they enter mature subsets of follicular and marginal zone B cells. In addition, 125Tg B cells remain competent to increase CD86 expression in response to both T cell-dependent (anti-CD40) and T cell-independent (anti-IgM or LPS) signals. Thus, for anti-insulin B cells, tolerance is characterized by defective B cell proliferation uncoupled from signals that promote maturation and costimulator function. In diabetes-prone NOD mice, anti-insulin B cells in this novel state of tolerance provide the essential B cell contribution required for autoimmune beta cell destruction. These findings suggest that the degree of functional impairment, rather than an overt breach of tolerance, is a critical feature that governs B cell contribution to T cell-mediated autoimmune disease.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Diferenciação Celular/imunologia , Anergia Clonal/imunologia , Diabetes Mellitus Tipo 1/imunologia , Anticorpos Anti-Insulina/biossíntese , Animais , Antígenos CD/biossíntese , Subpopulações de Linfócitos B/citologia , Antígeno B7-2 , Diferenciação Celular/genética , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Anergia Clonal/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Humanos , Imunoglobulina M/biossíntese , Imunoglobulina M/genética , Anticorpos Anti-Insulina/genética , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Baço/imunologia , Baço/patologia , Linfócitos T/imunologia
9.
Eur J Immunol ; 34(9): 2387-95, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15307171

RESUMO

Type 1 diabetes mellitus (T1DM) results from autoimmune destruction of insulin-producing beta cells in the pancreatic islets. Although T1DM is mediated by T lymphocytes, B lymphocytes are essential for insulitis and disease progression in the non-obese diabetic mouse model. We find that B cells invading the pancreas phenotypically resemble B1a B cells in the peritoneal cavity, including the presence of CD5+. To investigate the link between the peritoneal cavity and lymphocytes invading the pancreas, we used intraperitoneal hypotonic lysis to target these cells. B1a cells were eliminated from the peritoneal compartment by this treatment and were quickly replaced by B2 cells. Both B1a and B2 B cells were concordantly redistributed away from insulitis lesions, while pancreatic T cells showed little change. As a consequence of these events, the onset of diabetes was significantly delayed. These findings indicate that simple perturbations of the B cell-enriched peritoneal compartment can affect the disease process in the pancreas even after islet invasion has begun.


Assuntos
Linfócitos B/fisiologia , Diabetes Mellitus Tipo 1/etiologia , Peritônio/imunologia , Animais , Diabetes Mellitus Tipo 1/imunologia , Feminino , Soluções Hipotônicas , Tolerância Imunológica , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos NOD , Linfócitos T/fisiologia , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA