Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Diabetologia ; 61(11): 2333-2343, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30094467

RESUMEN

AIMS/HYPOTHESIS: Innate immune effectors interact with the environment to contribute to the pathogenesis of the autoimmune disease, type 1 diabetes. Although recent studies have suggested that innate immune Toll-like receptors (TLRs) are involved in tissue development, little is known about the role of TLRs in tissue development, compared with autoimmunity. We aimed to fill the knowledge gap by investigating the role of TLR9 in the development and function of islet beta cells in type 1 diabetes, using NOD mice. METHODS: We generated Tlr9-/- NOD mice and examined them for type 1 diabetes development and beta cell function, including insulin secretion and glucose tolerance. We assessed islet and beta cell number and characterised CD140a expression on beta cells by flow cytometry. We also tested beta cell function in Tlr9-/- C57BL/6 mice. Finally, we used TLR9 antagonists to block TLR9 signalling in wild-type NOD mice to verify the role of TLR9 in beta cell development and function. RESULTS: TLR9 deficiency promoted pancreatic islet development and beta cell differentiation, leading to enhanced glucose tolerance, improved insulin sensitivity and enhanced first-phase insulin secretory response. This was, in part, mediated by upregulation of CD140a (also known as platelet-derived growth factor receptor-α [PDGFRα]). In the absence of TLR9, induced by either genetic targeting or treatment with TLR9 antagonists, which had similar effects on ontogenesis and function of beta cells, NOD mice were protected from diabetes. CONCLUSIONS/INTERPRETATION: Our study links TLR9 and the CD140a pathway in regulating islet beta cell development and function and indicates a potential therapeutic target for diabetes prevention and/or treatment.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Cloroquina/farmacología , Diabetes Mellitus Tipo 1/patología , Femenino , Citometría de Flujo , Prueba de Tolerancia a la Glucosa , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Mutantes , Oligodesoxirribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptor Toll-Like 9/antagonistas & inhibidores , Receptor Toll-Like 9/genética
2.
J Autoimmun ; 93: 57-65, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29960834

RESUMEN

The incidence of type 1 diabetes (T1D) is determined by both genetic and environmental factors. In recent years, the gut microbiota have been identified to be an important environmental factor that could modify diabetes susceptibility. We have previously shown that Myeloid differentiation primary response gene 88 (MyD88), a major adaptor protein downstream of most innate immune Toll-like receptor (TLR) signaling, is important for mediating diabetes susceptibility in the non-obese diabetic (NOD) mouse model of human T1D. Here we report the role of TIR-domain-containing adapter-inducing interferon-ß (TRIF) in T1D development, as TRIF is an important adaptor protein downstream of TLR3 and TLR4 signaling. We found that TRIF-deficient (TRIF-/-) NOD mice were protected from development of diabetes, but only when housed with TRIF-deficient (TRIF-/-) NOD mice. When housed with TRIF-sufficient wild type (WT, i.e., TRIF+/+) NOD mice, the mice developed diabetes. We further investigated the gut microbiota as a potential cause for the altered diabetes development. Interestingly, TRIF-/-NOD mice had a different microbiota composition compared to WT NOD mice, only if they were housed with TRIF-/-NOD mice. However, the composition of gut microbiota in the TRIF-/-NOD mice was indistinguishable from WT NOD mice, if they were housed with WT NOD mice. The difference in the gut microbiota in TRIF-/-NOD mice, due to cohousing, accorded with the diabetes development in TRIF-/-NOD mice. Comparing the gut microbiota in TRIF-/- and WT NOD mice, we identified changes in percentage of Sutterella, Rikenella and Turicibacter species. Moreover, bacteria from WT NOD mice induced significantly stronger inflammatory immune responses in vitro compared to those from TRIF-/-NOD mice. Further immunological analysis revealed impaired function of dendritic cells and reduced T cell activation and proliferation in TRIF-/-NOD mice. Our data show that TRIF-deficiency protects NOD mice from diabetes development through alteration of the gut microbiota and reduced immune cell activation; however, that protection is over-ridden upon exposure to WT NOD bacteria. Therefore exposure to different microbiota can modify disease susceptibility determined by genetic factors related to innate immunity.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/microbiología , Microbioma Gastrointestinal/inmunología , Factor 88 de Diferenciación Mieloide/genética , Linfocitos T/inmunología , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Traslado Adoptivo , Animales , Bacteroidetes/inmunología , Burkholderiales/inmunología , Proliferación Celular , Células Dendríticas/inmunología , Células Dendríticas/patología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Susceptibilidad a Enfermedades , Femenino , Firmicutes/inmunología , Regulación de la Expresión Génica , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal , Linfocitos T/patología , Linfocitos T/trasplante , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
3.
J Immunol ; 195(9): 4176-84, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26401004

RESUMEN

Type 1 diabetes (T1D) is a T cell-mediated autoimmune disease that involves the slow, progressive destruction of islet ß cells and loss of insulin production, as a result of interaction with environmental factors, in genetically susceptible individuals. The gut microbiome is established very early in life. Commensal microbiota establish mutualism with the host and form an important part of the environment to which individuals are exposed in the gut, providing nutrients and shaping immune responses. In this study, we studied the impact of targeting most Gram-negative bacteria in the gut of NOD mice at different time points in their life, using a combination of three antibiotics--neomycin, polymyxin B, and streptomycin--on diabetes development. We found that the prenatal period is a critical time for shaping the immune tolerance in the progeny, influencing development of autoimmune diabetes. Prenatal neomycin, polymyxin B, and streptomycin treatment protected NOD mice from diabetes development through alterations in the gut microbiota, as well as induction of tolerogenic APCs, which led to reduced activation of diabetogenic CD8 T cells. Most importantly, we found that the protective effect was age dependent, and the most profound protection was found when the mice were treated before birth. This indicates the importance of the prenatal environment and early exposure to commensal bacteria in shaping the host immune system and health.


Asunto(s)
Antibacterianos/farmacología , Células Presentadoras de Antígenos/fisiología , Diabetes Mellitus Tipo 1/prevención & control , Tolerancia Inmunológica , Factores de Edad , Animales , Femenino , Intestinos/microbiología , Ratones , Ratones Endogámicos NOD , Microbiota , Neomicina/farmacología , Polimixina B/farmacología , Embarazo , Estreptomicina/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
4.
J Autoimmun ; 71: 26-34, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27021275

RESUMEN

Type 1 diabetes (T1D) is an organ-specific autoimmune disease characterized by T cell-mediated destruction of the insulin-producing pancreatic ß cells. A combination of genetic and environmental factors eventually leads to the loss of functional ß cell mass and hyperglycemia. Both innate and adaptive immunity are involved in the development of T1D. In this review, we have highlighted the most recent findings on the role of innate immunity, especially the pattern recognition receptors (PRRs), in disease development. In murine models and human studies, different PRRs, such as toll-like receptors (TLRs) and nucleotide-binding domain, leucine-rich repeat-containing (or Nod-like) receptors (NLRs), have different roles in the pathogenesis of T1D. These PRRs play a critical role in defending against infection by sensing specific ligands derived from exogenous microorganisms to induce innate immune responses and shape adaptive immunity. Animal studies have shown that TLR7, TLR9, MyD88 and NLPR3 play a disease-predisposing role in T1D, while controversial results have been found with other PRRs, such as TLR2, TLR3, TLR4, TLR5 and others. Human studies also shown that TLR2, TLR3 and TLR4 are expressed in either islet ß cells or infiltrated immune cells, indicating the innate immunity plays a role in ß cell autoimmunity. Furthermore, some human genetic studies showed a possible association of TLR3, TLR7, TLR8 or NLRP3 genes, at single nucleotide polymorphism (SNP) level, with human T1D. Increasing evidence suggest that the innate immunity modulates ß cell autoimmunity. Thus, targeting pathways of innate immunity may provide novel therapeutic strategies to fight this disease.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/metabolismo , Inmunidad Innata , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Inmunidad Adaptativa , Animales , Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/patología , Ratones Endogámicos NOD , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo
5.
Hepatology ; 62(5): 1536-50, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26185095

RESUMEN

UNLABELLED: Autoimmune hepatitis (AIH) in humans is a severe inflammatory liver disease characterized by interface hepatitis, the presence of circulating autoantibodies, and hyper-gammaglobulinemia. There are two types of AIH, type 1 (AIH-1) and type 2 (AIH-2), characterized by distinct autoimmune serology. Patients with AIH-1 are positive for anti-smooth muscle and/or antinuclear autoantibodies, whereas patients with AIH-2 have anti-liver kidney microsomal type 1 and/or anti-liver cytosol type 1 autoantibodies. Cytochrome P4502D6 is the antigenic target of anti-liver kidney microsomal type 1, and formiminotransferase cyclodeaminase is the antigenic target of anti-liver cytosol type 1. It is known that AIH, both types 1 and 2, is strongly linked to the human leukocyte antigen (HLA) alleles -DR3, -DR4, and -DR7. However, direct evidence of the association of HLA with AIH is lacking. We developed a novel mouse model of AIH using the HLA-DR3 transgenic mouse on the nonobese-diabetic background by immunization of HLA-DR3- and HLA-DR3+ nonobese-diabetic mice with a DNA plasmid, coding for human cytochrome P4502D6/formiminotransferase cyclodeaminase fusion protein. Immunization with cytochrome P4502D6/formiminotransferase cyclodeaminase leads to a sustained elevation of alanine aminotransferase, development of antinuclear autoantibodies and anti-liver kidney microsomal type 1/anti-liver cytosol type 1 autoantibodies, chronic immune cell infiltration, and parenchymal fibrosis on liver histology in HLA-DR3+ mice. Immunized mice also showed an enhanced T helper 1 immune response and paucity of the frequency of regulatory T cells in the liver. Moreover, HLA-DR3+ mice with exacerbated AIH showed reduced diversity and total load of gut bacteria. CONCLUSION: Our humanized animal model has provided a novel experimental tool to further elucidate the pathogenesis of AIH and to evaluate the efficacy and safety of immunoregulatory therapeutic interventions in vivo.


Asunto(s)
Hepatitis Autoinmune/etiología , Intestinos/microbiología , Microbiota , Animales , Autoanticuerpos/inmunología , Secuencia de Bases , Citocromo P-450 CYP2D6/inmunología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Antígeno HLA-DR3/inmunología , Humanos , Inmunización , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Linfocitos T Reguladores/inmunología
6.
Rev Endocr Metab Disord ; 16(1): 55-65, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25619480

RESUMEN

Diabetes is a group of metabolic disorders characterized by persistent hyperglycemia and has become a major public health concern. Autoimmune type 1 diabetes (T1D) and insulin resistant type 2 diabetes (T2D) are the two main types. A combination of genetic and environmental factors contributes to the development of these diseases. Gut microbiota have emerged recently as an essential player in the development of T1D, T2D and obesity. Altered gut microbiota have been strongly linked to disease in both rodent models and humans. Both classic 16S rRNA sequencing and shot-gun metagenomic pyrosequencing analysis have been successfully applied to explore the gut microbiota composition and functionality. This review focuses on the association between gut microbiota and diabetes and discusses the potential mechanisms by which gut microbiota regulate disease development in T1D, T2D and obesity.


Asunto(s)
Diabetes Mellitus Tipo 1/microbiología , Diabetes Mellitus Tipo 2/microbiología , Tracto Gastrointestinal/microbiología , Microbiota/fisiología , Obesidad/microbiología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Obesidad/metabolismo
7.
J Immunol ; 191(6): 2926-37, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23956420

RESUMEN

TLR9-deficient (TLR9⁻/⁻) NOD mice develop a significantly reduced incidence of diabetes. This study was to investigate the molecular mechanisms of the protective role of TLR9 deficiency. Through gene screening and confirmation by both mRNA and protein expression, we found a significant increase in CD73-expressing immune cells from peripheral lymphoid tissues in TLR9⁻/⁻ NOD mice. The elevated frequency of CD73-expressing immune cells seemed to be specific for TLR9 deficiency and was MyD88 independent. Moreover, the increased frequency of CD73 expression was limited to the NOD background. Increased frequency of CD73 expression was also associated with lower levels of proinflammatory cytokines and more anti-inflammatory cytokine production in CD4⁺ T cells in TLR9⁻/⁻ NOD mice. Purified CD73⁺CD4⁺ T cells showed stronger immunosuppressive function in vitro and delayed diabetes development in vivo. The immunosuppression appeared to be mediated by TGF-ß. In addition, elevated frequency of CD73-expressing cells was associated with improved ß cell function. Our observations were further confirmed by protection from diabetes with similar alterations in CD73 in the NY8.3 TCR NOD mouse model crossed with TLR9⁻/⁻ mice and by the use of a TLR9 inhibitor in NOD mice. Our novel findings suggest an important immune-regulatory role of CD73 in regulation of diabetes development and may offer a new therapeutic strategy for specific intervention to prevent type 1 diabetes.


Asunto(s)
5'-Nucleotidasa/biosíntesis , Diabetes Mellitus Tipo 1/inmunología , Linfocitos T/inmunología , Receptor Toll-Like 9/inmunología , 5'-Nucleotidasa/inmunología , Traslado Adoptivo , Animales , Separación Celular , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Citometría de Flujo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/metabolismo , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética
8.
J Immunol ; 188(10): 4747-58, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22490442

RESUMEN

B cells play a critical role in the pathogenesis of autoimmune diabetes. To investigate the mechanisms by which B cell depletion therapy attenuates islet ß cell loss and particularly to examine the effect of B cells on both diabetogenic and regulatory Ag-specific T cells, we generated a transgenic BDC2.5NOD mouse expressing human CD20 on B cells. This allowed us to deplete B cells for defined time periods and investigate the effect of B cell depletion on Ag-specific BDC2.5 T cells. We depleted B cells with anti-human CD20 Ab using a multiple injection protocol. We studied two time points, before and after B cell regeneration, to examine the effect on BDC2.5 T cell phenotype and functions that included antigenic response, cytokine profile, diabetogenicity, and suppressive function of regulatory T (T(reg)) cells. We found unexpectedly that B cell depletion induced transient aggressive behavior in BDC2.5 diabetogenic T cells and reduction in T(reg) cell number and function during the depletion period. However, after B cell reconstitution, we found that more regenerated B cells, particularly in the CD1d(-) fraction, expressed immune regulatory function. Our results suggest that the regenerated B cells are likely to be responsible for the therapeutic effect after B cell depletion. Our preclinical study also provides direct evidence that B cells regulate both pathogenic and T(reg) cell function, and this knowledge could explain the increased T cell responses to islet Ag after rituximab therapy in diabetic patients in a recent report and will be useful in design of future clinical protocols.


Asunto(s)
Antígenos CD20/biosíntesis , Subgrupos de Linfocitos B/inmunología , Complicaciones de la Diabetes/inmunología , Epítopos de Linfocito T/inmunología , Linfopenia/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Antígenos CD20/genética , Antígenos CD20/inmunología , Subgrupos de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/patología , Complicaciones de la Diabetes/metabolismo , Complicaciones de la Diabetes/patología , Humanos , Recuento de Linfocitos , Linfopenia/metabolismo , Linfopenia/patología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Fenotipo , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
9.
bioRxiv ; 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38826486

RESUMEN

The risk of hypoglycemia and its serious medical sequelae restrict insulin replacement therapy for diabetes mellitus. Such adverse clinical impact has motivated development of diverse glucose-responsive technologies, including algorithm-controlled insulin pumps linked to continuous glucose monitors ("closed-loop systems") and glucose-sensing ("smart") insulins. These technologies seek to optimize glycemic control while minimizing hypoglycemic risk. Here, we describe an alternative approach that exploits an endogenous glucose-dependent switch in hepatic physiology: preferential insulin signaling (under hyperglycemic conditions) versus preferential counter-regulatory glucagon signaling (during hypoglycemia). Motivated by prior reports of glucagon-insulin co-infusion, we designed and tested an ultra-stable glucagon-insulin fusion protein whose relative hormonal activities were calibrated by respective modifications; physical stability was concurrently augmented to facilitate formulation, enhance shelf life and expand access. An N-terminal glucagon moiety was stabilized by an α-helix-compatible Lys 13 -Glu 17 lactam bridge; A C-terminal insulin moiety was stabilized as a single chain with foreshortened C domain. Studies in vitro demonstrated (a) resistance to fibrillation on prolonged agitation at 37 °C and (b) dual hormonal signaling activities with appropriate balance. Glucodynamic responses were monitored in rats relative to control fusion proteins lacking one or the other hormonal activity, and continuous intravenous infusion emulated basal subcutaneous therapy. Whereas efficacy in mitigating hyperglycemia was unaffected by the glucagon moiety, the fusion protein enhanced endogenous glucose production under hypoglycemic conditions. Together, these findings provide proof of principle toward a basal glucose-responsive insulin biotechnology of striking simplicity. The fusion protein's augmented stability promises to circumvent the costly cold chain presently constraining global insulin access. Significance Statement: The therapeutic goal of insulin replacement therapy in diabetes is normalization of blood-glucose concentration, which prevents or delays long-term complications. A critical barrier is posed by recurrent hypoglycemic events that results in short- and long-term morbidities. An innovative approach envisions co-injection of glucagon (a counter-regulatory hormone) to exploit a glycemia-dependent hepatic switch in relative hormone responsiveness. To provide an enabling technology, we describe an ultra-stable fusion protein containing insulin- and glucagon moieties. Proof of principle was obtained in rats. A single-chain insulin moiety provides glycemic control whereas a lactam-stabilized glucagon extension mitigates hypoglycemia. This dual-hormone fusion protein promises to provide a basal formulation with reduced risk of hypoglycemia. Resistance to fibrillation may circumvent the cold chain required for global access.

10.
Front Immunol ; 14: 1147925, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36911699

RESUMEN

Introduction: Gut microbiota have been linked to modulating susceptibility to Type 1 diabetes; however, there are many ways in which the microbiota interact with host cells, including through microbial ligand binding to intracellular inflammasomes (large multi-subunit proteins) to initiate immune responses. NLRP6, a microbe-recognizing inflammasome protein, is highly expressed by intestinal epithelial cells and can alter susceptibility to cancer, obesity and Crohn's disease; however, the role of NLRP6 in modulating susceptibility to autoimmune diabetes, was previously unknown. Methods: We generated NLRP6-deficient Non-obese diabetic (NOD) mice to study the effect of NLRP6-deficiency on the immune cells and susceptibility to Type 1 diabetes development. Results: NLRP6-deficient mice exhibited an expansion of CD103+ B cells and were protected from type 1 diabetes. Moreover, NLRP6-deficient CD103+ B cells express regulatory markers, secreted higher concentrations of IL-10 and TGFb1 cytokines and suppressed diabetogenic T cell proliferation, compared to NLRP6-sufficient CD103+ B cells. Microarray analysis of NLRP6-sufficient and -deficient CD103+ B cells identified 79 significantly different genes including genes regulated by lipopolysaccharide (LPS), tretinoin, IL-10 and TGFb, which was confirmed in vitro following LPS stimulation. Furthermore, microbiota from NLRP6-deficient mice induced CD103+ B cells in colonized NLRP6-sufficient germ-free mice; however, the long-term maintenance of the CD103+ B cells required the absence of NLRP6 in the hosts, or continued exposure to microbiota from NLRP6-deficient mice. Discussion: Together, our data indicate that NLRP6 deficiency promotes expansion and maintenance of a novel TGF -dependent CD103+ Breg population. Thus, targeting NLRP6 therapeutically may prove clinically useful.


Asunto(s)
Diabetes Mellitus Tipo 1 , Interleucina-10 , Animales , Ratones , Tolerancia Inmunológica , Inflamasomas/metabolismo , Lipopolisacáridos , Ratones Endogámicos NOD
11.
Am J Physiol Heart Circ Physiol ; 303(6): H732-42, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22842069

RESUMEN

Toll-like receptor (TLR)4 regulates inflammation and metabolism and has been linked to the pathogenesis of heart disease. TLR4 is upregulated in diabetic cardiomyocytes, and we examined the role of TLR4 in modulating cardiac fatty acid (FA) metabolism and the pathogenesis of diabetic heart disease in nonobese diabetic (NOD) mice. Both wild-type (WT) NOD and TLR4-deficient NOD animals had increased plasma triglyceride levels after the onset of diabetes. However, by comparison, TLR4-deficient NOD mouse hearts had lower triglyceride accumulation in the early stages of diabetes, which was associated with a reduction in myeloid differentiation primary response gene (88) (MyD88), phosphorylation of p38 MAPK (phospho-p38), lipoprotein lipase (LPL), and JNK levels but increased phospho-AMP-activated protein kinase (AMPK). Oleic acid treatment in H9C2 cardiomyocytes also led to cellular lipid accumulation, which was attenuated by TLR4 small interfering RNA. TLR4 deficiency in the cells decreased FA-induced augmentation of MyD88, phospho-p38, and LPL, suggesting that TLR4 may modulate FA-induced lipid metabolism in cardiomyocytes. In addition, although cardiac function was impaired in both diabetic WT NOD and TLR4-deficient NOD animals compared with control nondiabetic mice, this deficit was less in the diabetic TLR4-deficient NOD mice, which had greater ejection fraction, greater fractional shortening, and increased left ventricular developed pressure in the early stages after the development of diabetes compared with their diabetic WT NOD counterparts. Thus, we conclude that TLR4 plays a role in regulating lipid accumulation in cardiac muscle after the onset of type 1 diabetes, which may contribute to cardiac dysfunction.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Cardiomiopatías Diabéticas/etiología , Metabolismo de los Lípidos , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Receptor Toll-Like 4/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Glucemia/metabolismo , Línea Celular , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Cardiomiopatías Diabéticas/sangre , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/patología , Modelos Animales de Enfermedad , Ácidos Grasos no Esterificados/sangre , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipoproteína Lipasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Miocardio/patología , Miocitos Cardíacos/patología , Ácido Oléico/metabolismo , Fosforilación , Interferencia de ARN , Ratas , Factores de Tiempo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Triglicéridos/sangre , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Nat Biomed Eng ; 6(6): 683-705, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35361935

RESUMEN

Peripheral neurons that sense glucose relay signals of glucose availability to integrative clusters of neurons in the brain. However, the roles of such signalling pathways in the maintenance of glucose homoeostasis and their contribution to disease are unknown. Here we show that the selective activation of the nerve plexus of the hepatic portal system via peripheral focused ultrasound stimulation (pFUS) improves glucose homoeostasis in mice and rats with insulin-resistant diabetes and in swine subject to hyperinsulinemic-euglycaemic clamps. pFUS modulated the activity of sensory projections to the hypothalamus, altered the concentrations of metabolism-regulating neurotransmitters, and enhanced glucose tolerance and utilization in the three species, whereas physical transection or chemical blocking of the liver-brain nerve pathway abolished the effect of pFUS on glucose tolerance. Longitudinal multi-omic profiling of metabolic tissues from the treated animals confirmed pFUS-induced modifications of key metabolic functions in liver, pancreas, muscle, adipose, kidney and intestinal tissues. Non-invasive ultrasound activation of afferent autonomic nerves may represent a non-pharmacologic therapy for the restoration of glucose homoeostasis in type-2 diabetes and other metabolic diseases.


Asunto(s)
Diabetes Mellitus Experimental , Glucosa , Animales , Diabetes Mellitus Experimental/terapia , Glucosa/metabolismo , Homeostasis , Hipotálamo/metabolismo , Hígado/metabolismo , Ratones , Ratas , Porcinos
13.
Clin Immunol ; 139(3): 336-49, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21458378

RESUMEN

This study was to determine whether BMDCs cultured in the presence of IL-10 (G/10-DCs) could promote T cell tolerance and prevent autoimmune diabetes in two different animal models of T1D. Our results showed that G/10-DCs suppressed both insulitis and spontaneous diabetes in NOD and HLA-DQ8/RIP-B7.1 mice. The suppression was likely to be mediated by T cells, as we found that regulatory CD4(+)CD25(+)Foxp3(+) cells were significantly increased in G/10-DC treated animals. In vivo, the G/10-DCs inhibited diabetogenic T cell proliferation; in vitro, they had reduced expression of costimulatory molecules and produced little IL-12/23 p40 or IL-6 but a large amount of IL-10 when compared with DCs matured in the presence of IL-4 (G/4-DC). We conclude that IL-10-treated DCs are tolerogenic and induce islet-directed immune tolerance, which was likely to be mediated by T regulatory cells. This non-antigen-specific DC-based approach offers potential for a new therapeutic intervention in T1D.


Asunto(s)
Antígeno B7-1/inmunología , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Antígenos HLA-DQ/inmunología , Células Secretoras de Insulina/inmunología , Interleucina-10/farmacología , Traslado Adoptivo , Animales , Células Dendríticas/efectos de los fármacos , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Tolerancia Inmunológica/inmunología , Inmunofenotipificación , Interleucina-10/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Organismos Libres de Patógenos Específicos , Linfocitos T/inmunología
14.
Cell Mol Immunol ; 18(2): 328-338, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33432061

RESUMEN

Innate immunity mediated by Toll-like receptors (TLRs), which can recognize pathogen molecular patterns, plays a critical role in type 1 diabetes development. TLR7 is a pattern recognition receptor that senses single-stranded RNAs from viruses and host tissue cells; however, its role in type 1 diabetes development remains unclear. In our study, we discovered that Tlr7-deficient (Tlr7-/-) nonobese diabetic (NOD) mice, a model of human type 1 diabetes, exhibited a significantly delayed onset and reduced incidence of type 1 diabetes compared with Tlr7-sufficient (Tlr7+/+) NOD mice. Mechanistic investigations showed that Tlr7 deficiency significantly altered B-cell differentiation and immunoglobulin production. Moreover, Tlr7-/- NOD B cells were found to suppress diabetogenic CD4+ T-cell responses and protect immunodeficient NOD mice from developing diabetes induced by diabetogenic T cells. In addition, we found that Tlr7 deficiency suppressed the antigen-presenting functions of B cells and inhibited cytotoxic CD8+ T-cell activation by downregulating the expression of both nonclassical and classical MHC class I (MHC-I) molecules on B cells. Our data suggest that TLR7 contributes to type 1 diabetes development by regulating B-cell functions and subsequent interactions with T cells. Therefore, therapeutically targeting TLR7 may prove beneficial for disease protection.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos B/inmunología , Diferenciación Celular , Diabetes Mellitus Tipo 1/prevención & control , Inmunidad Innata , Glicoproteínas de Membrana/fisiología , Linfocitos T Citotóxicos/inmunología , Receptor Toll-Like 7/fisiología , Animales , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Femenino , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados
15.
Front Immunol ; 12: 702955, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34394099

RESUMEN

Type 1 diabetes is an autoimmune disease caused by T cell-mediated destruction of insulin-producing ß cells. BDC2.5 T cells in BDC2.5 CD4+ T cell receptor transgenic Non-Obese Diabetic (NOD) mice (BDC2.5+ NOD mice) can abruptly invade the pancreatic islets resulting in severe insulitis that progresses rapidly but rarely leads to spontaneous diabetes. This prevention of diabetes is mediated by T regulatory (Treg) cells in these mice. In this study, we investigated the role of interleukin 10 (IL-10) in the inhibition of diabetes in BDC2.5+ NOD mice by generating Il-10-deficient BDC2.5+ NOD mice (BDC2.5+Il-10-/- NOD mice). Our results showed that BDC2.5+Il-10-/- NOD mice displayed robust and accelerated diabetes development. Il-10 deficiency in BDC2.5+ NOD mice promoted the generation of neutrophils in the bone marrow and increased the proportions of neutrophils in the periphery (blood, spleen, and islets), accompanied by altered intestinal immunity and gut microbiota composition. In vitro studies showed that the gut microbiota from BDC2.5+Il-10-/- NOD mice can expand neutrophil populations. Moreover, in vivo studies demonstrated that the depletion of endogenous gut microbiota by antibiotic treatment decreased the proportion of neutrophils. Although Il-10 deficiency in BDC2.5+ NOD mice had no obvious effects on the proportion and function of Treg cells, it affected the immune response and activation of CD4+ T cells. Moreover, the pathogenicity of CD4+ T cells was much increased, and this significantly accelerated the development of diabetes when these CD4+ T cells were transferred into immune-deficient NOD mice. Our study provides novel insights into the role of IL-10 in the modulation of neutrophils and CD4+ T cells in BDC2.5+ NOD mice, and suggests important crosstalk between gut microbiota and neutrophils in type 1 diabetes development.


Asunto(s)
Inmunidad Adaptativa , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/microbiología , Microbioma Gastrointestinal/inmunología , Inmunidad Innata , Interleucina-10/deficiencia , Linfocitos T Reguladores/inmunología , Animales , Diabetes Mellitus Tipo 1/genética , Interleucina-10/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados
16.
J Autoimmun ; 32(2): 85-93, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19200691

RESUMEN

This study was designed to examine immunopathology of diabetic nephropathy in non-obese diabetic (NOD) mice and to investigate the involvement of cellular and humoral immunity at various time points after diabetes onset. We found that the glomeruli of diabetic NOD mice were infiltrated with T and B cells, as well as CD11c+ dendritic cells, which had close contact with CD4+ and CD8+ T cells in the infiltrates. We also found that IgG deposits in the glomeruli of diabetic NOD mice were accompanied by the presence of complement C3. Moreover, the serum from diabetic mice contained autoantibodies directed towards components of the glomeruli and these antibodies were not present in non-diabetic NOD mice. The immune changes in the kidney occurred together with increasing kidney weight and urinary albumin excretion along with duration of diabetes. We provide evidence that infiltrating lymphocytes and anti-kidney autoantibodies may be involved in diabetic nephropathy in autoimmune diabetes in the NOD mouse. Understanding the role that the immune system plays in the pathogenesis of diabetic nephropathy could lead to identification of new strategies and/or additional therapeutic targets for prevention and treatment of diabetic nephropathy.


Asunto(s)
Formación de Anticuerpos/inmunología , Nefropatías Diabéticas/inmunología , Nefropatías Diabéticas/patología , Albuminuria/patología , Albuminuria/orina , Animales , Autoanticuerpos/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Femenino , Regulación de la Expresión Génica , Granzimas/genética , Granzimas/metabolismo , Inmunidad Celular/inmunología , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos NOD , Microscopía Electrónica de Transmisión , Perforina/genética , Perforina/metabolismo , ARN Mensajero/genética
17.
Front Immunol ; 10: 2654, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31798584

RESUMEN

Environmental factors contribute to Type 1 diabetes (T1D) susceptibility. The gut microbiome, which includes bacteria, viruses, and fungi, contributes to this environmental influence, and can induce immunological changes. The gut viral component of the microbiome, related to T1D has mostly focused on coxsackieviruses and rotavirus. The role of norovirus, another common enteric virus, in susceptibility to T1D was hitherto unknown. Norovirus is highly infectious and encountered by many children. We studied the mouse norovirus 4 (MNV4), related to human noroviruses, in the Non-obese diabetic (NOD) mouse model, to determine its role in influencing susceptibility to T1D. We infected MNV-free NOD mice with MNV4 by exposing the mice to MNV4-positive bedding from an endemically-infected mouse colony to mimic a natural infection. Control MNV-free NOD mice were exposed to MNV-free bedding from the same colony. Interestingly, MNV4 infection protected NOD mice from the development of T1D and was associated with an expansion of Tregs and reduced proinflammatory T cells. We also found MNV4 significantly modified the gut commensal bacteria composition, promoting increased α-diversity and Firmicutes/Bacteroidetes ratio. To elucidate whether T1D protection was directly related to MNV4, or indirectly through modulating gut microbiota, we colonized germ-free (GF) NOD mice with the MNV4-containing or non-MNV4-containing viral filtrate, isolated from filtered fecal material. We found that MNV4 induced significant changes in mucosal immunity, including altered Tuft cell markers, cytokine secretion, antiviral immune signaling markers, and the concentration of mucosal antibodies. Systemically, MNV4-infection altered the immune cells including B cell subsets, macrophages and T cells, and especially induced an increase in Treg number and function. Furthermore, in vitro primary exposure of the norovirus filtrate to naïve splenocytes identified significant increases in the proportion of activated and CTLA4-expressing Tregs. Our data provide novel knowledge that norovirus can protect NOD mice from T1D development by inducing the expansion of Tregs and reducing inflammatory T cells. Our study also highlights the importance of distinguishing the mucosal immunity mediated by bacteria from that by enteric viruses.


Asunto(s)
Infecciones por Caliciviridae/inmunología , Diabetes Mellitus Tipo 1/inmunología , Susceptibilidad a Enfermedades/virología , Microbioma Gastrointestinal/inmunología , Linfocitos T/inmunología , Animales , Susceptibilidad a Enfermedades/inmunología , Ratones , Ratones Endogámicos NOD , Norovirus/inmunología
18.
Biochem Biophys Res Commun ; 369(3): 795-800, 2008 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-18045573

RESUMEN

Human dihydrofolate reductase (DHFR) is a critical target in cancer chemotherapy. Previous studies showed that an 82-nt RNA fragment within the DHFR mRNA protein-coding region functions as a DHFR cis-acting response element. In this study, we further investigated the key elements contained within this sequence that are required for the DHFR mRNA-DHFR protein interaction. Using enzymatic foot-printing assays and RNA-binding experiments, we isolated a 27-nt sequence (DHFR27, corresponding to nts 407-433), which bound with high affinity and specificity to human DHFR to form a ribonucleoprotein complex. In vivo transient transfection experiments using a luciferase reporter system revealed that DHFR27 RNA could repress the luciferase expression in a DHFR-dependent manner when placed upstream of luciferase mRNA. This work provides new insights into the essential molecular elements that mediate RNA-protein interactions.


Asunto(s)
Regulación de la Expresión Génica , Biosíntesis de Proteínas/genética , ARN Mensajero/metabolismo , Secuencias Reguladoras de Ácido Ribonucleico , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Genes Reporteros , Humanos , Luciferasas de Renilla/genética , Datos de Secuencia Molecular , ARN Mensajero/química , Tetrahidrofolato Deshidrogenasa/química
19.
JCI Insight ; 3(1)2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29321370

RESUMEN

B cells play an important role in type 1 diabetes (T1D) development. However, the role of B cell activation-induced cytidine deaminase (AID) in diabetes development is not clear. We hypothesized that AID is important in the immunopathogenesis of T1D. To test this hypothesis, we generated AID-deficient (AID-/-) NOD mice. We found that AID-/-NOD mice developed accelerated T1D, with worse insulitis and high levels of anti-insulin autoantibody in the circulation. Interestingly, neither maternal IgG transferred through placenta, nor IgA transferred through milk affected the accelerated diabetes development. AID-/-NOD mice showed increased activation and proliferation of B and T cells. We found enhanced T-B cell interactions in AID-/-NOD mice, with increased T-bet and IFN-γ expression in CD4+ T cells in the presence of AID-/- B cells. Moreover, excessive lymphoid expansion was observed in AID-/-NOD mice. Importantly, antigen-specific BDC2.5 CD4+ T cells caused more rapid onset of diabetes when cotransferred with AID-/- B cells than when cotransferred with AID+/+ B cells. Thus, our study provides insights into the role of AID in T1D. Our data also suggest that AID is a negative regulator of immune tolerance and ablation of AID can lead to exacerbated islet autoimmunity and accelerated T1D development.


Asunto(s)
Citidina Desaminasa/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Activación Enzimática , Inmunidad Adaptativa , Animales , Autoanticuerpos , Autoinmunidad , Linfocitos B/inmunología , Citidina Desaminasa/genética , Citocinas/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Tolerancia Inmunológica , Inmunoglobulina A , Inmunoglobulina G , Insulina/inmunología , Interferón gamma/metabolismo , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos NOD , Leche/inmunología , Placenta/inmunología , Embarazo , Bazo/patología , Linfocitos T/inmunología , Virulencia
20.
Nucleic Acids Res ; 30(20): 4481-8, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12384595

RESUMEN

Previous studies have shown that human dihydrofolate reductase (DHFR) acts as an RNA-binding protein, in which it binds to its own mRNA and, in so doing, results in translational repression. In this study, we used RNA gel mobility shift and nitrocellulose filter-binding assays to further investigate the specificity of the interaction between human DHFR protein and human DHFR mRNA. Site-directed mutagenesis was used to identify the critical amino acid residues on DHFR protein required for RNA recognition. Human His-Tag DHFR protein specifically binds to human DHFR mRNA, while unrelated proteins including thymidylate synthase, p53 and glutathione-S-transferase were unable to form a ribonucleoprotein complex with DHFR mRNA. The Cys6 residue is essential for RNA recognition, as mutation at this amino acid with either an alanine (C6A) or serine (C6S) residue almost completely abrogated RNA-binding activity. Neither one of the cysteine mutant proteins was able to repress the in vitro translation of human DHFR mRNA. Mutations at amino acids Ile7, Arg28 and Phe34, significantly reduced RNA-binding activity. An RNA footprinting analysis identified three different RNA sequences, bound to DHFR protein, ranging in size from 16 to 45 nt, while a UV cross-linking analysis isolated an approximately 16 nt RNA sequence bound to DHFR. These studies begin to identify the critical amino acid residues on human DHFR that mediate RNA binding either through forming direct contact points with RNA or through maintaining the protein in an optimal structure that allows for the critical RNA-binding domain to be accessible.


Asunto(s)
Aminoácidos/análisis , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/metabolismo , Sitios de Unión , Humanos , Sustancias Macromoleculares , Peso Molecular , Biosíntesis de Proteínas , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/metabolismo , Tetrahidrofolato Deshidrogenasa/genética , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA