Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 218(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34115115

RESUMO

Naturally occurring cases of monogenic type 1 diabetes (T1D) help establish direct mechanisms driving this complex autoimmune disease. A recently identified de novo germline gain-of-function (GOF) mutation in the transcriptional regulator STAT3 was found to cause neonatal T1D. We engineered a novel knock-in mouse incorporating this highly diabetogenic human STAT3 mutation (K392R) and found that these mice recapitulated the human autoimmune diabetes phenotype. Paired single-cell TCR and RNA sequencing revealed that STAT3-GOF drives proliferation and clonal expansion of effector CD8+ cells that resist terminal exhaustion. Single-cell ATAC-seq showed that these effector T cells are epigenetically distinct and have differential chromatin architecture induced by STAT3-GOF. Analysis of islet TCR clonotypes revealed a CD8+ cell reacting against known antigen IGRP, and STAT3-GOF in an IGRP-reactive TCR transgenic model demonstrated that STAT3-GOF intrinsic to CD8+ cells is sufficient to accelerate diabetes onset. Altogether, these findings reveal a diabetogenic CD8+ T cell response that is restrained in the presence of normal STAT3 activity and drives diabetes pathogenesis.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Tolerância Imunológica/genética , Mutação/genética , Fator de Transcrição STAT3/genética , Animais , Autoimunidade , Proliferação de Células , Quimiotaxia/genética , Apresentação Cruzada/imunologia , Citotoxicidade Imunológica/genética , Modelos Animais de Doenças , Epigênese Genética , Mutação com Ganho de Função , Heterozigoto , Humanos , Camundongos , Fenótipo , Regulação para Cima
2.
Cell Metab ; 31(1): 46-61, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31839487

RESUMO

Type 1 diabetes is an autoimmune disease caused by the immune-mediated destruction of pancreatic ß cells that results in lifelong absolute insulin deficiency. For nearly a century, insulin replacement has been the only therapy for most people living with this disease. Recent advances in technology and our understanding of ß cell development, glucose metabolism, and the underlying immune pathogenesis of the disease have led to innovative therapeutic and preventative approaches. A paradigm shift in immunotherapy development toward the targeting of islet-specific immune pathways involved in tolerance has driven the development of therapies that may allow for the prevention or reversal of this disease while avoiding toxicities associated with historical approaches that were broadly immunosuppressive. In this review, we discuss successes, failures, and emerging pharmacological therapies for type 1 diabetes that are changing how we approach this disease, from improving glycemic control to developing the "holy grail" of disease prevention.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Imunoterapia/métodos , Animais , Linhagem Celular , Humanos , Tolerância Imunológica , Imunoterapia/tendências
4.
Nat Med ; 23(5): 540-547, 2017 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-28475571

RESUMO

The emergence of immuno-oncology as the first broadly successful strategy for metastatic cancer will require clinicians to integrate this new pillar of medicine with chemotherapy, radiation, and targeted small-molecule compounds. Of equal importance is gaining an understanding of the limitations and toxicities of immunotherapy. Immunotherapy was initially perceived to be a relatively less toxic approach to cancer treatment than other available therapies-and surely it is, when compared to those. However, as the use of immunotherapy becomes more common, especially as first- and second-line treatments, immunotoxicity and autoimmunity are emerging as the Achilles' heel of immunotherapy. In this Perspective, we discuss evidence that the occurrence of immunotoxicity bodes well for the patient, and describe mechanisms that might be related to the induction of autoimmunity. We then explore approaches to limit immunotoxicity, and discuss the future directions of research and reporting that are needed to diminish it.


Assuntos
Transferência Adotiva/efeitos adversos , Antineoplásicos/efeitos adversos , Doenças Autoimunes/etiologia , Autoimunidade/imunologia , Fatores Imunológicos/efeitos adversos , Imunoterapia/efeitos adversos , Neoplasias/terapia , Anticorpos Monoclonais/efeitos adversos , Doenças Autoimunes/imunologia , Hipofisite Autoimune/induzido quimicamente , Hipofisite Autoimune/imunologia , Antígeno CTLA-4/antagonistas & inibidores , Humanos , Ipilimumab , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Linfócitos T/transplante , Evasão Tumoral/imunologia
5.
J Clin Endocrinol Metab ; 101(4): 1798-806, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26909799

RESUMO

CONTEXT: The mechanisms of action of incretin mimetics in patients with long-standing type 2 diabetes (T2D) and high insulin requirements have not been studied. OBJECTIVE: To evaluate changes in ß-cell function, glucagon secretion, and fat distribution after addition of liraglutide to high-dose insulin. DESIGN: A single-center, randomized, double-blind, placebo-controlled trial. SETTING: University of Texas Southwestern and Parkland Memorial Hospital clinics. PATIENTS: Seventy-one patients with long-standing (median, 17 years) T2D requiring high-dose insulin treatment (>1.5 U/kg/d; average, 2.2 ± 0.9 U/kg/d). INTERVENTION: Patients were randomized to liraglutide 1.8 mg/d or matching placebo for 6 months. MAIN OUTCOME MEASURES: We measured changes in insulin and glucagon secretion using a 4-hour mixed-meal challenge test. Magnetic resonance-based techniques were used to estimate sc and visceral fat in the abdomen and ectopic fat in the liver and pancreas. RESULTS: Glycosylated hemoglobin improved significantly with liraglutide treatment, with an end-of-trial estimated treatment difference between groups of −0.9% (95% confidence interval, −1.5, −0.4%) (P = .002). Insulin secretion improved in the liraglutide group vs placebo, as measured by the area under the curve of C-peptide (P = .002) and the area under the curves ratio of C-peptide to glucose (P = .003). Insulin sensitivity (Matsuda index) and glucagon secretion did not change significantly between groups. Liver fat and sc fat decreased in the liraglutide group vs placebo (P = .0006 and P = .01, respectively), whereas neither visceral nor pancreatic fat changed significantly. CONCLUSIONS: Treatment with liraglutide significantly improved insulin secretion, even in patients with long-standing T2D requiring high-dose insulin treatment. Liraglutide also decreased liver and sc fat, but it did not alter glucagon secretion.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Insulina/uso terapêutico , Liraglutida/uso terapêutico , Tecido Adiposo/diagnóstico por imagem , Adulto , Glicemia/análise , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/diagnóstico por imagem , Método Duplo-Cego , Feminino , Glucagon/metabolismo , Hemoglobinas Glicadas/análise , Humanos , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Insulina/farmacologia , Resistência à Insulina/fisiologia , Secreção de Insulina , Liraglutida/farmacologia , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade
6.
Diabetes Metab Res Rev ; 31(7): 734-44, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25959529

RESUMO

BACKGROUND: Metabolic syndrome (MetS) appears closely linked with ceramide accumulation, inducing insulin resistance and toxicity to multiple cell types. Animal studies demonstrate that thiazolidinediones (TZDs) reduce ceramide concentrations in plasma and skeletal muscle and support lowering of ceramide levels as a potential mediator of TZDs' mechanism of action in reducing insulin resistance; however, studies in humans have yet to be reported. This study investigated the effects of pioglitazone therapy on plasma ceramides to understand the mechanism by which TZDs improve insulin resistance in MetS. METHODS: Thirty-seven subjects with MetS were studied in a single-centre, randomized, double-blind, placebo-controlled trial comparing pioglitazone to placebo. Data were collected at baseline and after 6 months of therapy. The primary endpoint was the change from baseline in plasma ceramide concentrations. RESULTS: Treatment with pioglitazone for 6 months, compared with placebo, significantly reduced multiple plasma ceramide concentrations: C18:0 (p = 0.001), C20:0 (p = 0.0004), C24 : 1 (p = 0.009), dihydroceramide C18 :0 (p = 0.005), dihydroceramide C24:1 (p = 0.004), lactosylceramide C16:0 (p = 0.02) and the hexosylceramides C16:0 (p = 0.0003), C18 : 0 (p = 0.00001), C22:0 (p = 0.00002) and C24:1 (p = 0.0006). Additionally, significant reductions were found when ceramides were grouped by species: ceramides (p = 0.03), dihydroceramides (p = 0.02), hexosylceramides (p = 0.00001) and lactosylceramides (p = 0.02). The total of all measured ceramides was also significantly reduced (p = 0.001). Following treatment with pioglitazone, the decrease in some ceramide species correlated negatively with the change in insulin sensitivity (dihydroceramide C16:0, r = -0.54; p = 0.02) and positively with total (lactosylceramide C24:0, r = 0.53; p = 0.02) and high molecular weight (lactosylceramide C24:0, r = 0.48; p = 0.05) adiponectin measurements; however, significant associations with changes in liver fat and glycemic control reduction were not found. CONCLUSIONS: Pioglitazone in individuals with MetS induces a potent decrease in plasma ceramides, and some of the changes correlate with changes in insulin resistance and adiponectin levels.


Assuntos
Ceramidas/sangue , Hipoglicemiantes/uso terapêutico , Síndrome Metabólica/tratamento farmacológico , Tiazolidinedionas/uso terapêutico , Adiponectina/sangue , Adulto , Método Duplo-Cego , Feminino , Humanos , Resistência à Insulina , Lactosilceramidas/sangue , Masculino , Síndrome Metabólica/sangue , Pessoa de Meia-Idade , Pioglitazona
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...