Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
J Immunol ; 212(11): 1658-1669, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38587315

RESUMEN

Chronic destruction of insulin-producing pancreatic ß cells by T cells results in autoimmune diabetes. Similar to other chronic T cell-mediated pathologies, a role for T cell exhaustion has been identified in diabetes in humans and NOD mice. The development and differentiation of exhausted T cells depends on exposure to Ag. In this study, we manipulated ß cell Ag presentation to target exhausted autoreactive T cells by inhibiting IFN-γ-mediated MHC class I upregulation or by ectopically expressing the ß cell Ag IGRP under the MHC class II promotor in the NOD8.3 model. Islet PD-1+TIM3+CD8+ (terminally exhausted [TEX]) cells were primary producers of islet granzyme B and CD107a, suggestive of cells that have entered the exhaustion program yet maintained cytotoxic capacity. Loss of IFN-γ-mediated ß cell MHC class I upregulation correlated with a significant reduction in islet TEX cells and diabetes protection in NOD8.3 mice. In NOD.TII/8.3 mice with IGRP expression induced in APCs, IGRP-reactive T cells remained exposed to high levels of IGRP in the islets and periphery. Consequently, functionally exhausted TEX cells, with reduced granzyme B expression, were significantly increased in these mice and this correlated with diabetes protection. These results indicate that intermediate Ag exposure in wild-type NOD8.3 islets allows T cells to enter the exhaustion program without becoming functionally exhausted. Moreover, Ag exposure can be manipulated to target this key cytotoxic population either by limiting the generation of cytotoxic TIM3+ cells or by driving their functional exhaustion, with both resulting in diabetes protection.


Asunto(s)
Linfocitos T CD8-positivos , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Ratones Endogámicos NOD , Animales , Ratones , Linfocitos T CD8-positivos/inmunología , Células Secretoras de Insulina/inmunología , Diabetes Mellitus Tipo 1/inmunología , Granzimas/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Presentación de Antígeno/inmunología , Femenino
2.
J Autoimmun ; 140: 103090, 2023 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-37572540

RESUMEN

CXCL10 is an IFNγ-inducible chemokine implicated in the pathogenesis of type 1 diabetes. T-cells attracted to pancreatic islets produce IFNγ, but it is unclear what attracts the first IFNγ -producing T-cells in islets. Gut dysbiosis following administration of pathobionts induced CXCL10 expression in pancreatic islets of healthy non-diabetes-prone (C57BL/6) mice and depended on TLR4-signaling, and in non-obese diabetic (NOD) mice, gut dysbiosis induced also CXCR3 chemokine receptor in IGRP-reactive islet-specific T-cells in pancreatic lymph node. In amounts typical to low-grade endotoxemia, bacterial lipopolysaccharide induced CXCL10 production in isolated islets of wild type and RAG1 or IFNG-receptor-deficient but not type-I-IFN-receptor-deficient NOD mice, dissociating lipopolysaccharide-induced CXCL10 production from T-cells and IFNγ. Although mostly myeloid-cell dependent, also ß-cells showed activation of innate immune signaling pathways and Cxcl10 expression in response to lipopolysaccharide indicating their independent sensitivity to dysbiosis. Thus, CXCL10 induction in response to low levels of lipopolysaccharide may allow islet-specific T-cells imprinted in pancreatic lymph node to enter in healthy islets independently of IFN-g, and thus link gut dysbiosis to early islet-autoimmunity via dysbiosis-associated low-grade endotoxemia.

3.
Cell Rep ; 39(4): 110747, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35476975

RESUMEN

Interferon gamma (IFNγ) is a proinflammatory cytokine implicated in autoimmune diseases. However, deficiency or neutralization of IFNγ is ineffective in reducing disease. We characterize islet antigen-specific T cells in non-obese diabetic (NOD) mice lacking all three IFN receptor genes. Diabetes is minimally affected, but at 125 days of age, antigen-specific CD8+ T cells, quantified using major histocompatibility complex class I tetramers, are present in 10-fold greater numbers in Ifngr-mutant NOD mice. T cells from Ifngr-mutant mice have increased proliferative responses to interleukin-2 (IL-2). They also have reduced phosphorylated STAT1 and its target gene, suppressor of cytokine signaling 1 (SOCS-1). IFNγ controls the expansion of antigen-specific CD8+ T cells by mechanisms which include increased SOCS-1 expression that regulates IL-2 signaling. The expanded CD8+ T cells are likely to contribute to normal diabetes progression despite reduced inflammation in Ifngr-mutant mice.


Asunto(s)
Diabetes Mellitus , Interleucina-2 , Animales , Autoantígenos , Linfocitos T CD8-positivos , Citocinas/metabolismo , Interferón gamma/metabolismo , Interferones/metabolismo , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos NOD , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
4.
Adv Exp Med Biol ; 1363: 97-118, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35220567

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease in which immune cells mediate the specific destruction of the insulin-producing ß cells in the pancreatic islets. Genetic and transcriptome studies for T1D indicate that a relatively large number of long noncoding RNAs (lncRNAs), detected in both immune cells and ß cells, contribute to the underlying inflammation and autoimmune pathology. Although lncRNAs do not encode proteins, their biochemical versatility as RNA molecules enables them to interact with proteins, DNA or RNA to exert regulatory effects on various cellular processes. Recent studies have begun to determine these effects for a small number of lncRNAs in modulating specific immune cell and ß-cell responses to elevated glucose levels and pro-inflammatory cytokines that are present within the islets during T1D pathogenesis. These findings are reviewed here and highlight the potential for different lncRNAs to act in concert to inhibit or exacerbate inflammatory and autoimmune responses. Despite this progress to date, additional investigations are required for a more in-depth understanding of their individual functional roles in this interplay, as well as identifying which lncRNAs are likely diagnostic biomarkers or therapeutic targets for autoimmune diseases such as T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Islotes Pancreáticos , ARN Largo no Codificante , Autoinmunidad/genética , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
5.
Diabetologia ; 64(4): 878-889, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33483762

RESUMEN

AIMS/HYPOTHESIS: Stimulator of IFN genes (STING) is a central hub for cytosolic nucleic acid sensing and its activation results in upregulation of type I IFN production in innate immune cells. A type I IFN gene signature seen before the onset of type 1 diabetes has been suggested as a driver of disease initiation both in humans and in the NOD mouse model. A possible source of type I IFN is through activation of the STING pathway. Recent studies suggest that STING also has antiproliferative and proapoptotic functions in T cells that are independent of IFN. To investigate whether STING is involved in autoimmune diabetes, we examined the impact of genetic deletion of STING in NOD mice. METHODS: CRISPR/Cas9 gene editing was used to generate STING-deficient NOD mice. Quantitative real-time PCR was used to assess the level of type I IFN-regulated genes in islets from wild-type and STING-deficient NOD mice. The number of islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)206-214-specific CD8+ T cells was determined by magnetic bead-based MHC tetramer enrichment and flow cytometry. The incidence of spontaneous diabetes and diabetes after adoptive transfer of T cells was determined. RESULTS: STING deficiency partially attenuated the type I IFN gene signature in islets but did not suppress insulitis. STING-deficient NOD mice accumulated an increased number of IGRP206-214-specific CD8+ T cells (2878 ± 642 cells in NOD.STING-/- mice and 728.8 ± 196 cells in wild-type NOD mice) in peripheral lymphoid tissue, associated with a higher incidence of spontaneous diabetes (95.5% in NOD.STING-/- mice and 86.2% in wild-type NOD mice). Splenocytes from STING-deficient mice rapidly induced diabetes after adoptive transfer into irradiated NOD recipients (median survival 75 days for NOD recipients of NOD.STING-/- mouse splenocytes and 121 days for NOD recipients of NOD mouse splenocytes). CONCLUSIONS/INTERPRETATION: Data suggest that sensing of endogenous nucleic acids through the STING pathway may be partially responsible for the type I IFN gene signature but not autoimmunity in NOD mice. Our results show that the STING pathway may play an unexpected intrinsic role in suppressing the number of diabetogenic T cells.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Diabetes Mellitus Tipo 1/metabolismo , Islotes Pancreáticos/metabolismo , Activación de Linfocitos , Proteínas de la Membrana/metabolismo , Traslado Adoptivo , Animales , Autoinmunidad , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Islotes Pancreáticos/inmunología , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos NOD , Ratones Noqueados , Transducción de Señal
6.
Diabetes ; 68(6): 1251-1266, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30936146

RESUMEN

Genome-wide association studies have identified PTPN2 as an important non-MHC gene for autoimmunity. Single nucleotide polymorphisms that reduce PTPN2 expression have been linked with the development of various autoimmune disorders, including type 1 diabetes. The tyrosine phosphatase PTPN2 attenuates T-cell receptor and cytokine signaling in T cells to maintain peripheral tolerance, but the extent to which PTPN2 deficiency in T cells might influence type 1 diabetes onset remains unclear. NOD mice develop spontaneous autoimmune type 1 diabetes similar to that seen in humans. In this study, T-cell PTPN2 deficiency in NOD mice markedly accelerated the onset and increased the incidence of type 1 diabetes as well as that of other disorders, including colitis and Sjögren syndrome. Although PTPN2 deficiency in CD8+ T cells alone was able to drive the destruction of pancreatic ß-cells and the onset of diabetes, T-cell-specific PTPN2 deficiency was also accompanied by increased CD4+ T-helper type 1 differentiation and T-follicular-helper cell polarization and increased the abundance of B cells in pancreatic islets as seen in human type 1 diabetes. These findings causally link PTPN2 deficiency in T cells with the development of type 1 diabetes and associated autoimmune comorbidities.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Diabetes Mellitus Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Linfocitos T/metabolismo , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Colitis/genética , Colitis/inmunología , Diabetes Mellitus Tipo 1/inmunología , Técnicas de Inactivación de Genes , Ratones , Ratones Endogámicos NOD , Proteína Tirosina Fosfatasa no Receptora Tipo 2/inmunología , Síndrome de Sjögren/genética , Síndrome de Sjögren/inmunología , Linfocitos T/inmunología , Células TH1/inmunología , Células TH1/metabolismo
7.
Int J Mol Sci ; 19(10)2018 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-30347820

RESUMEN

Nonobese diabetic (NOD) mice spontaneously develop lacrimal and salivary gland autoimmunity similar to human Sjögren syndrome. In both humans and NOD mice, the early immune response that drives T-cell infiltration into lacrimal and salivary glands is poorly understood. In NOD mice, lacrimal gland autoimmunity spontaneously occurs only in males with testosterone playing a role in promoting lacrimal gland inflammation, while female lacrimal glands are protected by regulatory T cells (Tregs). The mechanisms of this male-specific lacrimal gland autoimmunity are not known. Here, we studied the effects of Treg depletion in hormone-manipulated NOD mice and lacrimal gland gene expression to determine early signals required for lacrimal gland inflammation. While Treg-depletion was not sufficient to drive dacryoadenitis in castrated male NOD mice, chemokines (Cxcl9, Ccl19) and other potentially disease-relevant genes (Epsti1, Ubd) were upregulated in male lacrimal glands. Expression of Cxcl9 and Ccl19, in particular, remained significantly upregulated in the lacrimal glands of lymphocyte-deficient NOD-severe combined immunodeficiency (SCID) mice and their expression was modulated by type I interferon signaling. Notably, Ifnar1-deficient NOD mice did not develop dacryoadenitis. Together these data identify disease-relevant genes upregulated in the context of male-specific dacryoadenitis and demonstrate a requisite role for type I interferon signaling in lacrimal gland autoimmunity in NOD mice.


Asunto(s)
Dacriocistitis/metabolismo , Interferón Tipo I/metabolismo , Síndrome de Sjögren/metabolismo , Animales , Células Cultivadas , Quimiocina CCL19/metabolismo , Quimiocina CXCL9/metabolismo , Femenino , Aparato Lagrimal/metabolismo , Aparato Lagrimal/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Transducción de Señal , Linfocitos T Reguladores/metabolismo
8.
Stem Cell Res ; 31: 5-10, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29979973

RESUMEN

The nonobese diabetic (NOD) mouse strain is a predominant animal model of type 1 diabetes. However, this mouse strain is considered to be non-permissive for embryonic stem cell (ESC) derivation using conventional methods. We examined small molecule inhibition of glycogen synthase kinase 3 (GSK3) to block spontaneous cell differentiation and promote pluripotency persistence. Here we show a single pharmacological GSK3 inhibitor, 6-bromoindirubin-3'-oxime (BIO), in combination with leukemia inhibition factor (LIF), promoted generation of stable NOD ESC lines at >80% efficiency. Significantly, expansion of the established NOD ESC lines no longer required treatment with BIO. These NOD ESC lines contributed to chimeric mice and transmitted to germline progeny that spontaneously developed diabetes. By contrast, 5-aza-2'-deoxycytidine (AZA), a small molecule inhibitor of DNA methylation, and trichostatin A (TSA) and valproic acid (VPA), small molecule inhibitors of histone deacetylase, could not promote generation of NOD ESCs by epigenetic remodeling. These combined findings provide strategic insights for imposing pluripotency in cells isolated from a non-permissive strain.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Germinativas/metabolismo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Animales , Diferenciación Celular , Línea Celular , Células Cultivadas , Ratones , Ratones Endogámicos NOD
9.
Infect Immun ; 86(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29263105

RESUMEN

Mycoplasmas are bacterial pathogens of a range of animals, including humans, and are a common cause of respiratory disease. However, the host genetic factors that affect resistance to infection or regulate the resulting pulmonary inflammation are not well defined. We and others have previously demonstrated that nonobese diabetic (NOD) mice can be used to investigate disease loci that affect bacterial infection and autoimmune diabetes. Here we show that NOD mice are more susceptible than C57BL/6 (B6) mice to infection with Mycoplasma pulmonis, a natural model of pulmonary mycoplasmosis. The lungs of infected NOD mice had higher loads of M. pulmonis and more severe inflammatory lesions. Moreover, congenic NOD mice that harbored different B6-derived chromosomal intervals enabled identification and localization of a new mycoplasmosis locus, termed Mpr2, on chromosome 13. These congenic NOD mice demonstrated that the B6 allele for Mpr2 reduced the severity of pulmonary inflammation caused by infection with M. pulmonis and that this was associated with altered cytokine and chemokine concentrations in the infected lungs. Mpr2 also colocalizes to the same genomic interval as Listr2 and Idd14, genetic loci linked to listeriosis resistance and autoimmune diabetes susceptibility, respectively, suggesting that allelic variation within these loci may affect the development of both infectious and autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/genética , Predisposición Genética a la Enfermedad , Infecciones por Mycoplasma/genética , Mycoplasma pulmonis/fisiología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/microbiología , Femenino , Sitios Genéticos , Humanos , Pulmón/inmunología , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Infecciones por Mycoplasma/inmunología , Infecciones por Mycoplasma/microbiología , Mycoplasma pulmonis/genética
10.
Diabetes ; 66(12): 3041-3050, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28733313

RESUMEN

Granzyme A is a protease implicated in the degradation of intracellular DNA. Nucleotide complexes are known triggers of systemic autoimmunity, but a role in organ-specific autoimmune disease has not been demonstrated. To investigate whether such a mechanism could be an endogenous trigger for autoimmunity, we examined the impact of granzyme A deficiency in the NOD mouse model of autoimmune diabetes. Granzyme A deficiency resulted in an increased incidence in diabetes associated with accumulation of ssDNA in immune cells and induction of an interferon response in pancreatic islets. Central tolerance to proinsulin in transgenic NOD mice was broken on a granzyme A-deficient background. We have identified a novel endogenous trigger for autoimmune diabetes and an in vivo role for granzyme A in maintaining immune tolerance.


Asunto(s)
Diabetes Mellitus Tipo 1/etiología , Granzimas/fisiología , Tolerancia Inmunológica , Interferón Tipo I/fisiología , Animales , ADN de Cadena Simple/metabolismo , Femenino , Granzimas/deficiencia , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA