Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Diabetes Care ; 38(1): 43-50, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25414154

RESUMO

OBJECTIVE: Disease-associated T-cell autoreactivities are seen in most type 1 diabetic patients and are thought to emerge before islet autoantibodies, but host factors that impact autoimmune elements remain uncertain. We assessed if adiposity and measures of insulin sensitivity impact T- and B-cell autoimmunity in children with insulin-requiring diabetes. RESEARCH DESIGN AND METHODS: Insulin-requiring children and adolescents diagnosed between January 2004 and June 2008 were studied (n = 261): age 9.7 ± 4 years, 92% white, and 60% male. T-cell responses to 10 diabetes-associated antigens, ß-cell autoantibodies (GADA, IA-2A, IAA, and ICA), BMI z score (BMIz), and waist percentile were measured at onset and 3 months later. RESULTS: All but one subject had either T- or B-cell autoimmunity. Diabetes-associated T-cell autoreactivities were found in 92% of subjects. Higher amplitude T-cell autoreactivities to neuronal diabetes-associated autoantigens were seen in those with the highest BMIz quintile, BMI ≥85th percentile (P < 0.05), and waist circumference ≥85th percentile (P < 0.05). There were no relationships between the number of T-cell reactivities or T-cell diversity with adiposity measures or autoantibody number or type. Patients with positive T-cell reactivities but without autoantibodies had the highest BMIz (P = 0.006). CONCLUSIONS: Our observations link obesity and diabetes-related autoimmunity, suggesting an amplification of neuronal T-cell autoimmunity associated with adiposity and/or insulin resistance, with obesity-related inflammation possibly enhancing islet autoimmunity.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Insulina/uso terapêutico , Obesidade Infantil/imunologia , Linfócitos T/imunologia , Adolescente , Alelos , Autoanticorpos/imunologia , Autoantígenos/imunologia , Autoimunidade/imunologia , Linfócitos B/imunologia , Índice de Massa Corporal , Peptídeo C/sangue , Proliferação de Células , Criança , Estudos Transversais , Diabetes Mellitus Tipo 1/genética , Feminino , Seguimentos , Humanos , Células Secretoras de Insulina/imunologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Masculino , Circunferência da Cintura
2.
Mol Med ; 19: 149-59, 2013 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-23689362

RESUMO

Multiple sclerosis (MS) is a chronic progressive, demyelinating condition whose therapeutic needs are unmet, and whose pathoetiology is elusive. We report that transient receptor potential vanilloid-1 (TRPV1) expressed in a major sensory neuron subset, controls severity and progression of experimental autoimmune encephalomyelitis (EAE) in mice and likely in primary progressive MS. TRPV1-/- B6 congenics are protected from EAE. Increased survival reflects reduced central nervous systems (CNS) infiltration, despite indistinguishable T cell autoreactivity and pathogenicity in the periphery of TRPV1-sufficient and -deficient mice. The TRPV1+ neurovascular complex defining the blood-CNS barriers promoted invasion of pathogenic lymphocytes without the contribution of TRPV1-dependent neuropeptides such as substance P. In MS patients, we found a selective risk-association of the missense rs877610 TRPV1 single nucleotide polymorphism (SNP) in primary progressive disease. Our findings indicate that TRPV1 is a critical disease modifier in EAE, and we identify a predictor of severe disease course and a novel target for MS therapy.


Assuntos
Encefalomielite Autoimune Experimental/fisiopatologia , Esclerose Múltipla/genética , Canais de Cátion TRPV/fisiologia , Transferência Adotiva , Adulto , Animais , Encéfalo/patologia , Encéfalo/fisiologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/terapia , Feminino , Humanos , Linfonodos/citologia , Masculino , Camundongos , Camundongos Transgênicos , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos , Toxina Pertussis , Polimorfismo de Nucleotídeo Único , Medula Espinal/patologia , Medula Espinal/fisiologia , Baço/citologia
3.
Diabetes Metab Res Rev ; 27(8): 913-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22069284

RESUMO

Obesity-associated insulin resistance is a core element of metabolic syndrome and type 2 diabetes (T2D). Notably, insulin resistance is also a feature of type 1 diabetes (T1D), where findings in the non-obese diabetic mouse model have implicated transient receptor potential vanilloid-1 (TRPV1+) sensory neurons in local islet inflammation and glucose metabolism. Here, we briefly review the role of TRPV1 in non-obese diabetic (NOD) T1D pathogenesis, highlighting commonalities that suggest TRPV1 may contribute to obesity and T2D as well. With the recently discovered importance of adipose infiltrating lymphocytes in the metabolic disturbances of obesity and T2D, sensory innervation of fat may thus play an analogous role to sensory neurons in the islet--modulating neuroendocrine homeostasis and inflammation. In such a scenario, TRPV1+ sensory nerves would provide the pathoaetiological link connecting the shared metabolic and immunologic features of type 1 diabetes and T2D.


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Células Receptoras Sensoriais/fisiologia , Canais de Cátion TRPV/fisiologia , Tecido Adiposo/inervação , Animais , Resistência à Insulina/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Obesidade/complicações , Obesidade/fisiopatologia
4.
J Immunol ; 187(4): 1998-2005, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21775681

RESUMO

Type 1 diabetes mellitus is believed to be due to the autoimmune destruction of ß-cells by T lymphocytes, but a single course of rituximab, a monoclonal anti-CD20 B lymphocyte Ab, can attenuate C-peptide loss over the first year of disease. The effects of B cell depletion on disease-associated T cell responses have not been studied. We compare changes in lymphocyte subsets, T cell proliferative responses to disease-associated target Ags, and C-peptide levels of participants who did (responders) or did not (nonresponders) show signs of ß-cell preservation 1 y after rituximab therapy in a placebo-controlled TrialNet trial. Rituximab decreased B lymphocyte levels after four weekly doses of mAb. T cell proliferative responses to diabetes-associated Ags were present at baseline in 75% of anti-CD20- and 82% of placebo-treated subjects and were not different over time. However, in rituximab-treated subjects with significant C-peptide preservation at 6 mo (58%), the proliferative responses to diabetes-associated total (p = 0.032), islet-specific (p = 0.048), and neuronal autoantigens (p = 0.005) increased over the 12-mo observation period. This relationship was not seen in placebo-treated patients. We conclude that in patients with type 1 diabetes mellitus, anti-B cell mAb causes increased proliferative responses to diabetes Ags and attenuated ß-cell loss. The way in which these responses affect the disease course remains unknown.


Assuntos
Anticorpos Monoclonais Murinos/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Tipo 1/tratamento farmacológico , Fatores Imunológicos/administração & dosagem , Células Secretoras de Insulina/imunologia , Subpopulações de Linfócitos T/imunologia , Adolescente , Adulto , Anticorpos Monoclonais Murinos/efeitos adversos , Anticorpos Monoclonais Murinos/imunologia , Autoantígenos/imunologia , Peptídeo C/imunologia , Criança , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Feminino , Humanos , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/imunologia , Células Secretoras de Insulina/patologia , Masculino , Rituximab , Subpopulações de Linfócitos T/patologia , Fatores de Tempo
5.
Nat Med ; 17(5): 610-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21499269

RESUMO

Chronic inflammation characterized by T cell and macrophage infiltration of visceral adipose tissue (VAT) is a hallmark of obesity-associated insulin resistance and glucose intolerance. Here we show a fundamental pathogenic role for B cells in the development of these metabolic abnormalities. B cells accumulate in VAT in diet-induced obese (DIO) mice, and DIO mice lacking B cells are protected from disease despite weight gain. B cell effects on glucose metabolism are mechanistically linked to the activation of proinflammatory macrophages and T cells and to the production of pathogenic IgG antibodies. Treatment with a B cell-depleting CD20 antibody attenuates disease, whereas transfer of IgG from DIO mice rapidly induces insulin resistance and glucose intolerance. Moreover, insulin resistance in obese humans is associated with a unique profile of IgG autoantibodies. These results establish the importance of B cells and adaptive immunity in insulin resistance and suggest new diagnostic and therapeutic modalities for managing the disease.


Assuntos
Linfócitos B/imunologia , Imunoglobulina G/biossíntese , Resistência à Insulina/imunologia , Linfócitos T/imunologia , Animais , Autoanticorpos/biossíntese , Autoantígenos/imunologia , Autoimunidade , Gorduras na Dieta/efeitos adversos , Glucose/metabolismo , Humanos , Cadeias mu de Imunoglobulina/genética , Inflamação/imunologia , Gordura Intra-Abdominal/imunologia , Ativação Linfocitária , Depleção Linfocítica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/etiologia , Obesidade/imunologia , Obesidade/metabolismo
6.
Diabetes ; 59(10): 2588-96, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20522597

RESUMO

OBJECTIVE: Autoimmune target tissues in type 1 diabetes include pancreatic ß-cells and peri-islet Schwann cells (pSC)--the latter active participants or passive bystanders in pre-diabetic autoimmune progression. To distinguish between these alternatives, we sought to suppress pSC autoimmunity by transgenic expression of the negative costimulatory molecule B7-H1 in NOD pSC. RESEARCH DESIGN AND METHODS: A B7-H1 transgene was placed under control of the glial fibrillary acidic protein (GFAP) promoter. Transgenic and wild-type NOD mice were compared for transgene PD-1 affinities, diabetes development, insulitis, and pSC survival. Mechanistic studies included adoptive type 1 diabetes transfer, B7-H1 blockade, and T-cell autoreactivity and sublineage distribution. RESULTS: Transgenic and endogenous B7-H1 bound PD-1 with equal affinities. Unexpectedly, the transgene generated islet-selective CD8(+) bias with accelerated rather than suppressed diabetes progression. T-cells of diabetic transgenics transferred type 1 diabetes faster. There were no earlier pSC losses due to conceivable transgene toxicity, but transgenic pSC loss was enhanced by 8 weeks, preceded by elevated GFAP autoreactivity, with high-affinity T-cells targeting the major NOD K(d)-GFAP epitope, p253-261. FoxP3(+) regulatory T- and CD11c(+) dendritic cell pools were unaffected. CONCLUSIONS: In contrast with transgenic B7-H1 in NOD mouse ß-cells, transgenic B7-H1 in pSC promotes rather than protects from type 1 diabetes. Here, ectopic B7-H1 enhanced the pathogenicity of effector T-cells, demonstrating that pSC can actively impact diabetes progression-likely through modification of intraislet T-cell selection. Although pSC cells emerge as a new candidate for therapeutic targets, caution is warranted with regard to the B7-H1-PD1 axis, where B7-H1 overexpression can lead to accelerated autoimmune disease.


Assuntos
Antígenos CD/genética , Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/fisiologia , Animais , Antígenos CD/análise , Antígenos CD/metabolismo , Antígeno B7-1/genética , Antígeno B7-H1 , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/patologia , Expressão Gênica , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Histocitoquímica , Humanos , Ilhotas Pancreáticas/patologia , Ativação Linfocitária , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Peptídeos/genética , Regiões Promotoras Genéticas , Ressonância de Plasmônio de Superfície , Linfócitos T/imunologia , Linfócitos T/fisiologia
7.
Diabetes Care ; 32(12): 2251-7, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19808924

RESUMO

OBJECTIVE: In patients with long-standing type 1 diabetes, we investigated whether improved beta-cell function can be achieved by combining intensive insulin therapy with agents that may 1) promote beta-cell growth and/or limit beta-cell apoptosis and 2) weaken the anti-beta-cell autoimmunity. RESEARCH DESIGN AND METHODS: For this study, 20 individuals (mean age 39.5 +/- 11.1 years) with long-standing type 1 diabetes (21.3 +/- 10.7 years) were enrolled in this prospective open-label crossover trial. After achieving optimal blood glucose control, 16 subjects were randomized to exenatide with or without daclizumab. Endogenous insulin production was determined by repeatedly measuring serum C-peptide. RESULTS: In 85% of individuals with long-standing type 1 diabetes who were screened for participation in this trial, C-peptide levels >or=0.05 ng/ml (0.02 nmol/l) were found. Residual beta-cells responded to physiological (mixed-meal) and pharmacological (arginine) stimuli. During exenatide treatment, patients lost 4.1 +/- 2.9 kg body wt and insulin requirements declined significantly (total daily dose on exenatide 0.48 +/- 0.11 vs. 0.55 +/- 0.13 units x kg(-1) x day(-1) without exenatide; P = 0.0062). No signs of further activation of the underlying autoimmune disease were observed. Exenatide delayed gastric emptying, suppressed endogenous incretin levels, but did not increase C-peptide secretion. CONCLUSIONS: In long-standing type 1 diabetes, which remains an active autoimmune disease even decades after its onset, surviving beta-cells secrete insulin in a physiologically regulated manner. However, the combination of intensified insulin therapy, exenatide, and daclizumab did not induce improved function of these remaining beta-cells.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Imunoglobulina G/uso terapêutico , Imunossupressores/uso terapêutico , Células Secretoras de Insulina/metabolismo , Peptídeos/uso terapêutico , Peçonhas/uso terapêutico , Adulto , Idade de Início , Anticorpos Monoclonais Humanizados , Autoimunidade/efeitos dos fármacos , Estudos Cross-Over , Daclizumabe , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/imunologia , Quimioterapia Combinada , Exenatida , Feminino , Hemoglobinas Glicadas/metabolismo , Antígenos HLA-DR/análise , Cadeias HLA-DRB1 , Humanos , Insulina/uso terapêutico , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/imunologia , Masculino , Projetos de Pesquisa , Inquéritos e Questionários , Estados Unidos , Adulto Jovem
8.
Diabetes ; 58(11): 2588-95, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19675135

RESUMO

OBJECTIVE: Type 1 diabetes results from an immunemediated destruction of beta-cells, likely to be mediated by T lymphocytes, but the sensitivity, specificity, and other measures of validity of existing assays for islet autoreactive T-cells are not well established. Such assays are vital for monitoring responses to interventions that may modulate disease progression. RESEARCH DESIGN AND METHODS: We studied the ability of cellular assays to discriminate responses in patients with type 1 diabetes and normal control subjects in a randomized blinded study in the U.S. and U.K. We evaluated the reproducibility of these measurements overall and to individual analytes from repeat collections. RESULTS: Responses in the cellular immunoblot, U.K.-ELISPOT, and T-cell proliferation assays could differentiate patients from control subjects with odds ratios of 21.7, 3.44, and 3.36, respectively, with sensitivity and specificity as high as 74 and 88%. The class II tetramer and U.S. ELISPOT assays performed less well. Despite the significant association of the responses with type 1 diabetes, the reproducibility of the measured responses, both overall and individual analytes, was relatively low. Positive samples from normal control subjects (i.e., false positives) were generally isolated to single assays. CONCLUSIONS: The cellular immunoblot, U.K.-ELISPOT, and T-cell proliferation assays can distinguish responses from patients with type 1 diabetes and healthy control subjects. The limited reproducibility of the measurements overall and of responses to individual analytes may reflect the difficulty in detection of low frequency of antigen-specific T-cells or variability in their appearance in peripheral blood.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Ilhotas Pancreáticas/imunologia , Reprodutibilidade dos Testes , Linfócitos T/imunologia , Adolescente , Adulto , Autoanticorpos/análise , Criança , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Feminino , Vetores Genéticos , Antígenos HLA-DR/genética , Teste de Histocompatibilidade , Humanos , Imunidade Celular , Immunoblotting , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Ativação Linfocitária , Masculino , Valores de Referência , Adulto Jovem
9.
Eur J Immunol ; 39(9): 2629-35, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19662632

RESUMO

Obesity is associated with numerous inflammatory conditions including atherosclerosis, autoimmune disease and cancer. Although the precise mechanisms are unknown, obesity-associated rises in TNF-alpha, IL-6 and TGF-beta are believed to contribute. Here we demonstrate that obesity selectively promotes an expansion of the Th17 T-cell sublineage, a subset with prominent pro-inflammatory roles. T-cells from diet-induced obese mice expand Th17 cell pools and produce progressively more IL-17 than lean littermates in an IL-6-dependent process. The increased Th17 bias was associated with more pronounced autoimmune disease as confirmed in two disease models, EAE and trinitrobenzene sulfonic acid colitis. In both, diet-induced obese mice developed more severe early disease and histopathology with increased IL-17(+) T-cell pools in target tissues. The well-described association of obesity with inflammatory and autoimmune disease is mechanistically linked to a Th17 bias.


Assuntos
Colite/imunologia , Encefalomielite Autoimune Experimental/imunologia , Interleucina-17/imunologia , Obesidade/complicações , Obesidade/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Colite/etiologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/etiologia , Glicoproteínas/farmacologia , Interleucina-6/imunologia , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos/farmacologia , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Ácido Trinitrobenzenossulfônico/farmacologia
10.
Nat Med ; 15(8): 921-9, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19633657

RESUMO

Obesity and its associated metabolic syndromes represent a growing global challenge, yet mechanistic understanding of this pathology and current therapeutics are unsatisfactory. We discovered that CD4(+) T lymphocytes, resident in visceral adipose tissue (VAT), control insulin resistance in mice with diet-induced obesity (DIO). Analyses of human tissue suggest that a similar process may also occur in humans. DIO VAT-associated T cells show severely biased T cell receptor V(alpha) repertoires, suggesting antigen-specific expansion. CD4(+) T lymphocyte control of glucose homeostasis is compromised in DIO progression, when VAT accumulates pathogenic interferon-gamma (IFN-gamma)-secreting T helper type 1 (T(H)1) cells, overwhelming static numbers of T(H)2 (CD4(+)GATA-binding protein-3 (GATA-3)(+)) and regulatory forkhead box P3 (Foxp3)(+) T cells. CD4(+) (but not CD8(+)) T cell transfer into lymphocyte-free Rag1-null DIO mice reversed weight gain and insulin resistance, predominantly through T(H)2 cells. In obese WT and ob/ob (leptin-deficient) mice, brief treatment with CD3-specific antibody or its F(ab')(2) fragment, reduces the predominance of T(H)1 cells over Foxp3(+) cells, reversing insulin resistance for months, despite continuation of a high-fat diet. Our data suggest that the progression of obesity-associated metabolic abnormalities is under the pathophysiological control of CD4(+) T cells. The eventual failure of this control, with expanding adiposity and pathogenic VAT T cells, can successfully be reversed by immunotherapy.


Assuntos
Resistência à Insulina/imunologia , Obesidade/complicações , Obesidade/terapia , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/fisiologia , Separação Celular , Glucose/metabolismo , Proteínas de Homeodomínio/genética , Homeostase/imunologia , Imunoterapia/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/imunologia , Obesidade/patologia
11.
Trends Mol Med ; 13(10): 405-13, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17900987

RESUMO

Type 1 diabetes (T1D) results from autoimmune-mediated loss of insulin-producing beta-cells. Recent findings suggest that the events controlling T1D development are not only immunological, but also neuronal in nature. In the non-obese diabetic (NOD) mouse model of T1D, a mutant sensory neuron channel, TRPV1, initiates chronic, progressive beta-cell stress, inducing islet cell inflammation. This novel mechanism of organ-specific damage requires a permissive, autoimmune-prone host, but ascribes tissue specificity to the local secretory dysfunction of sensory afferent neurons. In NOD mice, normalizing this neuronal function by administration of the neurotransmitter substance P clears islet cell inflammation, reduces insulin resistance and restores normoglycemia. Here, we discuss this neuro-immuno-endocrine model, its implications and the involvement of sensory neurons in other autoimmune disorders. These developments might provide novel neuronal-based therapeutic interventions, particularly in diabetes.


Assuntos
Autoimunidade/imunologia , Diabetes Mellitus Tipo 1/imunologia , Animais , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Modelos Biológicos , Neurônios Aferentes/imunologia , Neurônios Aferentes/metabolismo , Neurotransmissores/metabolismo
12.
Cell ; 127(6): 1123-35, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17174891

RESUMO

In type 1 diabetes, T cell-mediated death of pancreatic beta cells produces insulin deficiency. However, what attracts or restricts broadly autoreactive lymphocyte pools to the pancreas remains unclear. We report that TRPV1(+) pancreatic sensory neurons control islet inflammation and insulin resistance. Eliminating these neurons in diabetes-prone NOD mice prevents insulitis and diabetes, despite systemic persistence of pathogenic T cell pools. Insulin resistance and beta cell stress of prediabetic NOD mice are prevented when TRPV1(+) neurons are eliminated. TRPV1(NOD), localized to the Idd4.1 diabetes-risk locus, is a hypofunctional mutant, mediating depressed neurogenic inflammation. Delivering the neuropeptide substance P by intra-arterial injection into the NOD pancreas reverses abnormal insulin resistance, insulitis, and diabetes for weeks. Concordantly, insulin sensitivity is enhanced in trpv1(-/-) mice, whereas insulitis/diabetes-resistant NODxB6Idd4-congenic mice, carrying wild-type TRPV1, show restored TRPV1 function and insulin sensitivity. Our data uncover a fundamental role for insulin-responsive TRPV1(+) sensory neurons in beta cell function and diabetes pathoetiology.


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Inflamação , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/inervação , Neurônios Aferentes/fisiologia , Canais de Cátion TRPV/fisiologia , Animais , Autoimunidade , Capsaicina/farmacologia , Diabetes Mellitus Tipo 1/patologia , Feminino , Resistência à Insulina , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Neurônios Aferentes/efeitos dos fármacos , Substância P/farmacologia , Linfócitos T/imunologia , Canais de Cátion TRPV/genética
13.
Diabetes ; 55(9): 2588-94, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16936208

RESUMO

Type 1 diabetes is a chronic autoimmune disease mediated by autoreactive T-cells. Several experimental therapies targeting T-cells are in clinical trials. To understand how these therapies affect T-cell responses in vivo, assays that directly measure human T-cell function are needed. In a blinded, multicenter, case-controlled study conducted by the Immune Tolerance Network, we tested responses in an immunoblot and T-cell proliferative assay to distinguish type 1 diabetic patients from healthy control subjects. Peripheral blood cells from 39 healthy control subjects selected for DR4 and 23 subjects with recently diagnosed type 1 diabetes were studied. Autoantibody responses were measured in serum samples. Positive responses in both assays were more common in peripheral blood mononuclear cells from new-onset type 1 diabetic patients compared with control subjects. The proliferative, immunoblot, and autoantibody assays had sensitivities of 58, 91, and 78% with specificities of 94, 83, and 85%, respectively. When cellular assays were combined with autoantibody measurements, the sensitivity of the measurements was 75% with 100% specificity. We conclude that cellular assays performed on peripheral blood have a high degree of accuracy in discriminating responses in subjects with type 1 diabetes from healthy control subjects. They may be useful for assessment of cellular autoimmune responses involved in type 1 diabetes.


Assuntos
Autoimunidade/imunologia , Diabetes Mellitus Tipo 1/imunologia , Linfócitos T/imunologia , Adolescente , Adulto , Autoanticorpos/análise , Proliferação de Células , Criança , Feminino , Glutamato Descarboxilase/análise , Humanos , Immunoblotting , Isoenzimas/análise , Masculino , Sensibilidade e Especificidade
15.
Eur J Immunol ; 33(2): 546-55, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12645954

RESUMO

To better understand loss of self-tolerance in diabetes-prone NOD mice, we are generating ICA69 transgenes under control of the tetracycline-regulated tet07 minimal promoter. In vitro pilot studies showed leaky transgene expression, but addition of beta-globin genomic insulator flanks prevented leakage and dramatically enhanced transgene expression even in transient transfection, with excellent suppression by Doxycycline. In vivo, the accidental loss of insulator flanks during transgene insertion in one transgenic NOD founder, tet1, re-established leakiness with high level, exclusive ICA69-transgene expression in stromal elements of thymus and spleen. This led to persistent deletion of T cells targeting the immunodominant ICA69 epitope, Tep69, but emergence of T cell pools targeting cryptic ICA69 epitopes not normally generated in sufficient density to select and maintain ICA69-autoreactive T cells. This subtle modification of T cell repertoires reduced insulitis, and protected from diabetes in transgenics and in wild-type mice carrying irradiated tet1 thymus grafts. The low pathogenicity of T cells targeting cryptic epitopes likely reflects the fact that the major ICA69 determinant presented in the islet milieu remains Tep69, while cryptic epitopes are under-represented. Deviation of T cell autoreactivity from major to cryptic target epitopes in tet1 mice provides a fortuitous model to explain previously observed diabetes protection by immunotherapy or autoantigen transgenes despite apparent failure to achieve tolerance to the full length islet antigens.


Assuntos
Autoantígenos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Epitopos/imunologia , Ilhotas Pancreáticas/imunologia , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Animais , Autoantígenos/genética , Deleção Clonal , DNA Complementar/genética , Modelos Animais de Doenças , Feminino , Vetores Genéticos/química , Vetores Genéticos/genética , Epitopos Imunodominantes/imunologia , Inflamação , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Quimera por Radiação , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Tolerância a Antígenos Próprios/imunologia , Baço/imunologia , Baço/patologia , Baço/transplante , Células Estromais/metabolismo , Timo/imunologia , Timo/patologia , Timo/transplante , Transgenes
16.
Nat Med ; 9(2): 198-205, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12539039

RESUMO

Pancreatic islets of Langerhans are enveloped by peri-islet Schwann cells (pSC), which express glial fibrillary acidic protein (GFAP) and S100beta. pSC-autoreactive T- and B-cell responses arise in 3- to 4-week-old diabetes-prone non-obese diabetic (NOD) mice, followed by progressive pSC destruction before detectable beta-cell death. Humans with probable prediabetes generate similar autoreactivities, and autoantibodies in islet-cell autoantibody (lCA) -positive sera co-localize to pSC. Moreover, GFAP-specific NOD T-cell lines transferred pathogenic peri-insulitis to NOD/severe combined immunodeficient (NOD/SCID) mice, and immunotherapy with GFAP or S100beta prevented diabetes. pSC survived in rat insulin promoter Iymphocytic choriomeningitis virus (rip-LCMV) glycoprotein/CD8+ T-cell receptor(gp) double-transgenic mice with virus-induced diabetes, suggesting that pSC death is not an obligate consequence of local inflammation and beta-cell destruction. However, pSC were deleted in spontaneously diabetic NOD mice carrying the CD8+/8.3 T-cell receptor transgene, a T cell receptor commonly expressed in earliest islet infiltrates. Autoimmune targeting of pancreatic nervous system tissue elements seems to be an integral, early part of natural type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Ilhotas Pancreáticas/imunologia , Animais , Autoanticorpos/análise , Autoanticorpos/imunologia , Sequência de Bases , Primers do DNA , Feminino , Imunofluorescência , Proteína Glial Fibrilar Ácida/metabolismo , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Fatores de Crescimento Neural/metabolismo , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/metabolismo , Células de Schwann/imunologia , Células de Schwann/metabolismo , Especificidade da Espécie
17.
Lancet ; 360(9339): 1063-9, 2002 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-12383988

RESUMO

BACKGROUND: Sjögren's syndrome is a common (about 1% of the population) autoimmune disease of salivary and lacrimal glands. Its cause and pathogenesis are poorly understood, and treatments are mostly for symptoms of the disease. ICA69 is a self-antigen expressed in brain, pancreas, salivary, and lacrimal glands. NOD-strain mice are an animal model of spontaneous Sjögren's syndrome. We aimed to assess the role of ICA69 in autoimmunity against Sjögren's syndrome. METHODS: We inactivated the genomic ICA69 locus, generated NOD congenic mice that were deficient in ICA69, and assessed development of Sjögren's syndrome. ICA69 autoimmunity was investigated in controls and in patients with primary Sjögren's syndrome or systemic lupus erythematosus, and in various NOD mice, some of which were given an ICA69-directed prototype peptide vaccine. FINDINGS: Disruption of the ICA69 locus prevented lacrimal gland disease and greatly reduced salivary gland disease in NOD mice. In healthy NOD mice, ICA69-specific T cells accumulated in lymph nodes that drain salivary tissue. T-cell and B-cell autoreactivity against ICA69 was much the same in patients with primary Sjögren's syndrome, but not in those with systemic lupus erythematosus or in healthy controls. Immunotherapy with a high-affinity mimicry peptide targeting ICA69-specific T-cells reduced established Sjögren's syndrome in wild-type NOD mice in the long term. INTERPRETATION: ICA69 is a new autoantigen in primary Sjögren's syndrome that has an important role in progression of disease and could be of diagnostic value. Immunotherapy of primary Sjögren's syndrome is promising, since autoimmunity in NOD mice with Sjögren's syndrome seems to be uniquely susceptible to such treatment even late in disease.


Assuntos
Autoantígenos/imunologia , Modelos Animais de Doenças , Síndrome de Sjogren/imunologia , Adulto , Fatores Etários , Animais , Autoantígenos/genética , Linfócitos B/imunologia , Estudos de Casos e Controles , Progressão da Doença , Feminino , Humanos , Imunoterapia/métodos , Lúpus Eritematoso Sistêmico/diagnóstico , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/terapia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos NOD , Síndrome de Sjogren/diagnóstico , Síndrome de Sjogren/genética , Síndrome de Sjogren/terapia , Glândula Submandibular/imunologia , Glândula Submandibular/patologia , Linfócitos T/imunologia
19.
J Immunol ; 168(2): 680-8, 2002 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11777961

RESUMO

EBV is a human tumor virus that infects and establishes latency in the majority of humans worldwide. In vitro, EBV growth transforms primary B lymphocytes into lymphoblastoid cell lines with high efficiency. We have used cDNA subtraction cloning to identify cellular target genes required for growth transformation and identified a new C(2)H(2) (Krüppel-type) zinc finger gene, ZNF(EB), that is trans-activated early following EBV infection. In this study, we characterize ZNF(EB), including its intronless locus, and human and mouse protein variants. The gene is transiently expressed during normal lymphocyte activation, and its expression is sustained in EBV-positive but not EBV-negative B cell lines. There is limited expression in nonhemopoietic tissues. Its critical role in the growth transformation of B lineage cells is indicated by the abrogation of transformation with antisense strategies. ZNF(EB) maps to chromosome 18q12, a region with mutations in numerous, predominantly hemopoietic malignancies.


Assuntos
Linfócitos B/imunologia , Linfócitos B/patologia , Proteínas de Ciclo Celular/genética , Transformação Celular Neoplásica/genética , Transformação Celular Viral/genética , Proteínas de Ligação a DNA/genética , Regulação Viral da Expressão Gênica/imunologia , Herpesvirus Humano 4/imunologia , Ativação Linfocitária/genética , Dedos de Zinco/genética , Sequência de Aminoácidos , Animais , Linfócitos B/metabolismo , Sequência de Bases , Proteínas de Ciclo Celular/isolamento & purificação , Linhagem Celular Transformada , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/patologia , Transformação Celular Viral/imunologia , Células Cultivadas , Cromossomos Humanos Par 18/genética , Clonagem Molecular , DNA Complementar/isolamento & purificação , Proteínas de Ligação a DNA/isolamento & purificação , Biblioteca Gênica , Células HL-60 , Células HeLa , Humanos , Células Jurkat , Células K562 , Camundongos , Dados de Sequência Molecular , Família Multigênica/imunologia , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Isoformas de Proteínas/genética , Análise de Sequência de DNA , Células U937 , Dedos de Zinco/imunologia
20.
J Immunol ; 168(1): 475-82, 2002 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11751995

RESUMO

ICA69 (islet cell Ag 69 kDa) is a diabetes-associated autoantigen with high expression levels in beta cells and brain. Its function is unknown, but knockout of its Caenorhabditis elegans homologue, ric-19, compromised neurotransmission. We disrupted the murine gene, ica-1, in 129-strain mice. These animals aged normally, but speed-congenic ICA69(null) nonobese diabetic (NOD) mice developed mid-life lethality, reminiscent of NOD-specific, late lethal seizures in glutamic acid decarboxylase 65-deficient mice. In contrast to wild-type and heterozygous animals, ICA69(null) NOD congenics fail to generate, even after immunization, cross-reactive T cells that recognize the dominant Tep69 epitope in ICA69, and its environmental mimicry Ag, the ABBOS epitope in BSA. This antigenic mimicry is thus driven by the endogenous self Ag, and not initiated by the environmental mimic. Insulitis, spontaneous, and adoptively transferred diabetes develop normally in ICA69(null) NOD congenics. Like glutamic acid decarboxylase 65, ICA69 is not an obligate autoantigen in diabetes. Unexpectedly, ICA69(null) NOD mice were resistant to cyclophosphamide (CY)-accelerated diabetes. Transplantation experiments with hemopoietic and islet tissue linked CY resistance to ICA69 deficiency in islets. CY-accelerated diabetes involves not only ablation of lymphoid cells, but ICA69-dependent drug toxicity in beta cells that boosts autoreactivity in the regenerating lymphoid system.


Assuntos
Autoantígenos/genética , Autoantígenos/fisiologia , Proteínas de Caenorhabditis elegans , Ciclofosfamida/farmacologia , Diabetes Mellitus Tipo 1/etiologia , Transferência Adotiva , Animais , Autoantígenos/imunologia , Autoimunidade , Células Cultivadas , Diabetes Mellitus Tipo 1/patologia , Progressão da Doença , Epitopos/imunologia , Feminino , Marcação de Genes , Transplante das Ilhotas Pancreáticas , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Mimetismo Molecular , Linfócitos T/imunologia , Linfócitos T/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...