Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Toxicol Appl Pharmacol ; 284(2): 254-61, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25576766

RESUMEN

Sodium meta-arsenite (SA) is an orally available arsenic compound. We investigated the effects of SA on the development of autoimmune type 1 diabetes. Female non-obese diabetic (NOD) mice were orally intubated with SA (5mg/kg/day) from 8weeks of age for 8weeks. The cumulative incidence of diabetes was monitored until 30weeks of age, islet histology was examined, and lymphocytes including T cells, B cells, CD4+ IFN-γ+ cells, CD8+ IFN-γ+ cells, CD4+ IL-4+ cells, and regulatory T cells were analyzed. We also investigated the diabetogenic ability of splenocytes using an adoptive transfer model and the effect of SA on the proliferation, activation, and expression of glucose transporter 1 (Glut1) in splenocytes treated with SA in vitro and splenocytes isolated from SA-treated mice. SA treatment decreased the incidence of diabetes and delayed disease onset. SA treatment reduced the infiltration of immunocytes in islets, and splenocytes from SA-treated mice showed a reduced ability to transfer diabetes. The number of total splenocytes and T cells and both the number and the proportion of CD4+ IFN-γ+ and CD8+ IFN-γ+ T cells in the spleen were significantly reduced in SA-treated NOD mice compared with controls. The number, but not the proportion, of regulatory T cells was decreased in SA-treated NOD mice. Treatment with SA either in vitro or in vivo inhibited proliferation of splenocytes. In addition, the expression of Glut1 and phosphorylated ERK1/2 was decreased by SA treatment. These results suggest that SA reduces proliferation and activation of T cells, thus preventing autoimmune diabetes in NOD mice.


Asunto(s)
Arsenitos/farmacología , Enfermedades Autoinmunes/prevención & control , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Compuestos de Sodio/farmacología , Animales , Enfermedades Autoinmunes/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Fosforilación/efectos de los fármacos , Bazo/efectos de los fármacos , Bazo/metabolismo
2.
Diabetologia ; 55(9): 2456-68, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22722451

RESUMEN

AIMS/HYPOTHESIS: Obesity and insulin resistance are associated with low-grade chronic inflammation. Glucagon-like peptide-1 (GLP-1) is known to reduce insulin resistance. We investigated whether GLP-1 has anti-inflammatory effects on adipose tissue, including adipocytes and adipose tissue macrophages (ATM). METHODS: We administered a recombinant adenovirus (rAd) producing GLP-1 (rAd-GLP-1) to an ob/ob mouse model of diabetes. We examined insulin sensitivity, body fat mass, the infiltration of ATM and metabolic profiles. We analysed the mRNA expression of inflammatory cytokines, lipogenic genes, and M1 and M2 macrophage-specific genes in adipose tissue by real-time quantitative PCR. We also examined the activation of nuclear factor κB (NF-κB), extracellular signal-regulated kinase 1/2 and Jun N-terminal kinase (JNK) in vivo and in vitro. RESULTS: Fat mass, adipocyte size and mRNA expression of lipogenic genes were significantly reduced in adipose tissue of rAd-GLP-1-treated ob/ob mice. Macrophage populations (F4/80(+) and F4/80(+)CD11b(+)CD11c(+) cells), as well as the expression and production of IL-6, TNF-α and monocyte chemoattractant protein-1, were significantly reduced in adipose tissue of rAd-GLP-1-treated ob/ob mice. Expression of M1-specific mRNAs was significantly reduced, but that of M2-specific mRNAs was unchanged in rAd-GLP-1-treated ob/ob mice. NF-κB and JNK activation was significantly reduced in adipose tissue of rAd-GLP-1-treated ob/ob mice. Lipopolysaccharide-induced inflammation was reduced by the GLP-1 receptor agonist, exendin-4, in 3T3-L1 adipocytes and ATM. CONCLUSIONS/INTERPRETATION: We suggest that GLP-1 reduces macrophage infiltration and directly inhibits inflammatory pathways in adipocytes and ATM, possibly contributing to the improvement of insulin sensitivity.


Asunto(s)
Tejido Adiposo/metabolismo , Antiinflamatorios/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Obesidad/metabolismo , Animales , Distribución de la Grasa Corporal , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Inflamación/tratamiento farmacológico , Resistencia a la Insulina , Macrófagos/efectos de los fármacos , Ratones , Ratones Obesos , FN-kappa B/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Glucagón/metabolismo
3.
J Exp Med ; 189(2): 347-58, 1999 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-9892617

RESUMEN

We have shown previously that the inactivation of macrophages in nonobese diabetic (NOD) mice results in the prevention of diabetes; however, the mechanisms involved remain unknown. In this study, we found that T cells in a macrophage-depleted environment lost their ability to differentiate into beta cell-cytotoxic T cells, resulting in the prevention of autoimmune diabetes, but these T cells regained their beta cell-cytotoxic potential when returned to a macrophage-containing environment. To learn why T cells in a macrophage-depleted environment lose their ability to kill beta cells, we examined the islet antigen-specific immune response and T cell activation in macrophage-depleted NOD mice. There was a shift in the immune balance, a decrease in the T helper cell type 1 (Th1) immune response, and an increase in the Th2 immune response, due to the reduced expression of the macrophage-derived cytokine IL-12. As well, there was a deficit in T cell activation, evidenced by significant decreases in the expression of Fas ligand and perforin. The administration of IL-12 substantially reversed the prevention of diabetes in NOD mice conferred by macrophage depletion. We conclude that macrophages play an essential role in the development and activation of beta cell-cytotoxic T cells that cause beta cell destruction, resulting in autoimmune diabetes in NOD mice.


Asunto(s)
Autoinmunidad/inmunología , Macrófagos/inmunología , Animales , Diferenciación Celular/inmunología , Ácido Clodrónico/farmacología , Citotoxicidad Inmunológica/inmunología , Diabetes Mellitus/genética , Diabetes Mellitus/inmunología , Proteína Ligando Fas , Femenino , Interleucina-12/farmacología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Páncreas/efectos de los fármacos , Páncreas/patología , Perforina , Proteínas Citotóxicas Formadoras de Poros , Bazo/inmunología , Linfocitos T/inmunología , Trasplante de Tejidos
4.
Science ; 284(5417): 1183-7, 1999 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-10325232

RESUMEN

Glutamic acid decarboxylase (GAD) is a pancreatic beta cell autoantigen in humans and nonobese diabetic (NOD) mice. beta Cell-specific suppression of GAD expression in two lines of antisense GAD transgenic NOD mice prevented autoimmune diabetes, whereas persistent GAD expression in the beta cells in the other four lines of antisense GAD transgenic NOD mice resulted in diabetes, similar to that seen in transgene-negative NOD mice. Complete suppression of beta cell GAD expression blocked the generation of diabetogenic T cells and protected islet grafts from autoimmune injury. Thus, beta cell-specific GAD expression is required for the development of autoimmune diabetes in NOD mice, and modulation of GAD might, therefore, have therapeutic value in type 1 diabetes.


Asunto(s)
Autoantígenos/inmunología , Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/inmunología , Glutamato Descarboxilasa/inmunología , Islotes Pancreáticos/enzimología , Traslado Adoptivo , Animales , Autoantígenos/genética , Autoantígenos/fisiología , Autoinmunidad , ADN sin Sentido , Diabetes Mellitus Tipo 1/patología , Femenino , Expresión Génica , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/fisiología , Insulina/sangre , Insulina/metabolismo , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Linfocitos T/inmunología , Transgenes
5.
J Endocrinol Invest ; 31(4): 334-40, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18475052

RESUMEN

During the development of Type 1 diabetes, inflammatory cytokines are known to induce the expression of inducible nitric oxide synthase (iNOS) in pancreatic islets, and subsequent production of nitric oxide (NO) contributes to beta cell destruction. Glucagon-like peptide-1 (GLP-1) has been shown to reduce cytokine-induced apoptosis of beta cells. In this study, we investigated whether GLP-1 affects cytokine-induced NO production, resulting in the inhibition of beta-cell apoptosis. We treated MIN6N8a mouse beta cells with interferon (IFN)-gamma in the presence or absence of GLP-1 and found that IFN-gamma treatment induced iNOS mRNA expression and NO production, which was significantly inhibited by treatment with GLP-1. Blocking of GLP-1 receptor signaling via the cyclic AMP and phosphatidylinositol 3-kinase pathway did not directly affect the suppressive effect of GLP-1 on IFN- gamma-induced iNOS mRNA expression. Further studies revealed that IFN-gamma induced the expression of TNF-alpha mRNA and protein, which synergistically induced NO production, and GLP-1 treatment inhibited this induction of TNF-alpha. To examine whether the reduction of TNF-alpha by GLP-1 treatment plays a role in suppressing NO production, we treated MIN6N8a cells with IFN-gamma in the presence of anti-TNF-alpha neutralizing antibody and found that NO production was reduced. In addition, treatment of mouse islets with GLP-1 inhibited the expression of iNOS and TNFmRNA. These results suggest that GLP-1 inhibits IFN-gamma-induced NO production by suppression of TNF-alpha production.


Asunto(s)
Péptido 1 Similar al Glucagón/fisiología , Insulina/biosíntesis , Interferón gamma/farmacología , Islotes Pancreáticos/metabolismo , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Proteínas Recombinantes/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Células Cultivadas , Insulina/metabolismo , Secreción de Insulina , Interferón gamma/antagonistas & inhibidores , Islotes Pancreáticos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis
7.
Diabetes ; 48(1): 34-42, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9892220

RESUMEN

The development of autoimmune diabetes in NOD mice results from selective destruction of beta-cells by a T-cell-dependent autoimmune process. However, the mechanisms that control the generation of beta-cell cytotoxic T-cells in vivo are poorly understood. We recently established 8.3-T-cell receptor (TCR)-beta transgenic NOD mice that show a selective acceleration of the recruitment of CD8+ T-cells into the islets of prediabetic animals, resulting in rapid beta-cell destruction and early onset of diabetes. This study was initiated to determine the role of macrophages in the development and activation of diabetogenic CD8+ T-cells in 8.3-TCR-beta transgenic NOD mice. Inactivation of macrophages in these transgenic mice resulted in the complete prevention of diabetes. When splenic T-cells from macrophage-depleted 8.3-TCR-beta transgenic NOD mice were transfused into severe combined immunodeficiency disease (NOD.scid) mice, none of the recipients developed diabetes up to 10 weeks after transfer, while most of the recipients of T-cells from age-matched control 8.3-TCR-beta transgenic NOD mice became diabetic. When intact NOD islets were transplanted under the renal capsule of macrophage-depleted 8.3-TCR-beta transgenic NOD mice, the majority of the grafted islets remained intact, while most of the islets grafted into age-matched, control 8.3-TCR-beta transgenic NOD mice were destroyed within 3 weeks after transplantation. The depletion of macrophages in these mice resulted in a decrease in the Th1 immune response along with an increase in the Th2 immune response because of significant decreases in the expression of macrophage-derived cytokines, particularly interleukin-12, and a decrease in beta-cell-specific T-cell activation, as shown by significant decreases in the expression of Fas ligand (FasL), CD40 ligand (CD40L), and perforin, as compared with control mice. We conclude that macrophages are absolutely required for the development and activation of beta-cell cytotoxic CD8+ T-cells in 8.3-TCR-beta transgenic NOD mice.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Islotes Pancreáticos/fisiopatología , Macrófagos/fisiología , Ratones Endogámicos NOD/fisiología , Ratones Transgénicos/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T Citotóxicos/fisiología , Animales , Senescencia Celular/fisiología , Islotes Pancreáticos/patología , Activación de Linfocitos/fisiología , Ratones , Ratones SCID/fisiología
8.
Diabetes ; 47(4): 576-82, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9568690

RESUMEN

The molecular mechanism by which some, but not all, variants of encephalomyocarditis (EMC) virus selectively infect pancreatic beta-cells in mice and induce IDDM has been an enigma for more than a decade. We report here that the binding site of the EMC viral capsid protein VP1 determines viral diabetogenicity. Recombinant chimeric EMC viruses containing threonine, serine, proline, aspartic acid, or valine at position 152 of the major capsid protein VP1 bind poorly to beta-cells. In contrast, recombinant chimeric EMC viruses containing alanine or glycine at position 152 of the VP1 bind efficiently to and infect beta-cells, resulting in the development of diabetes. Three-dimensional molecular modeling reveals that the van der Waals interactions are greater and the residues surrounding position 152 of the VP1 are more closely packed in recombinant chimeric viruses containing threonine, serine, proline, aspartic acid, or valine at position 152 than in recombinant chimeric viruses containing alanine or glycine at the same position. Our studies reveal that the surface areas surrounding alanine or glycine at position 152 of the VP1 are more accessible, thus increasing the availability of the binding sites for attachment to beta-cell receptors and resulting in viral infection and the development of diabetes.


Asunto(s)
Proteínas de la Cápside , Cápside/metabolismo , Diabetes Mellitus Tipo 1/virología , Virus de la Encefalomiocarditis/patogenicidad , Animales , Sitios de Unión , Cápside/química , Cápside/genética , Diabetes Mellitus Tipo 1/metabolismo , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/virología , Masculino , Ratones , Modelos Moleculares , Mutación Puntual , Conformación Proteica , Recombinación Genética
9.
Curr Gene Ther ; 5(2): 249-62, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15853732

RESUMEN

Type 1 diabetes results from insulin deficiency caused by autoimmune destruction of insulin-producing pancreatic beta cells. Islet transplantation, beta cell regeneration, and insulin gene therapy have been explored in an attempt to cure type 1 diabetes. Major progress on islet transplantation includes substantial improvements in islet isolation technology to obtain viable and functionally intact islets and less toxic immunosuppressive drug regimes to prevent islet graft failure. However, the availability of human islets from cadaveric pancreata is limited. Regeneration of pancreatic beta cells from embryonic or adult stem cells may overcome the limited source of islets and transplant rejection if beta cells are regenerated from endogenous stem cells. However, it is difficult to overcome the persisting hostile beta cell-specific autoimmune response that may destroy the regenerated beta cells. Insulin gene therapy might overcome the weakness of islet transplantation and beta cell regeneration with respect to their vulnerability to autoimmune attack. This method replaces the function of beta cells by introducing various components of the insulin synthetic and secretory machinery into non- beta cells, which are not targets of beta cell-specific autoimmune responses. However, there is no regulatory system that results in the expression and release of insulin in response to glucose with satisfactory kinetics. Although there is no perfect solution for the cure of type 1 diabetes at the present time, research on a variety of potential approaches will offer the best choices for the cure of human type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Terapia Genética , Trasplante de Células Madre , Animales , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Humanos , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos
10.
Exp Clin Endocrinol Diabetes ; 123(4): 221-6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25502579

RESUMEN

BACKGROUND: In this study, we investigated whether Smad4 signaling is involved in the regulation of beta-cell function using a high fat diet (HFD)-induced obesity mouse model. METHODS: Beta-cell-specific Smad4-knockout mice (Smad4(-/-)RIP-Cre(+); ß-Smad4KO) were generated by mating Smad4 (flox/flox) mice with rat insulin promoter (RIP)-Cre mice. Mice were fed a HFD beginning at 6 weeks of age for 16 weeks. Body weight, food intake, fasting and fed glucose levels, and glucose and insulin tolerance were measured. RESULTS: The expression of Smad4 mRNA was significantly decreased in the islets of ß-Smad4KO mice. In wild-type mice, Smad4 mRNA was significantly decreased at 18 weeks of age as compared with 8 weeks of age. On a regular chow diet, ß-Smad4KO mice showed no differences in body weight, fed and fasting blood glucose levels, and glucose tolerance compared with wild-type mice. When fed a HFD, body weight gain was significantly reduced in ß-Smad4KO mice as compared with wild-type mice, although the amount of food intake was not different. During the HFD, fed and fasting blood glucose levels, glucose stimulated insulin secretion, disposition index and glucose tolerance were significantly improved in ß-Smad4KO mice as compared with wild-type mice. However, insulin tolerance tests showed no differences between the 2 groups. CONCLUSION: Inhibition of Smad4 in beta-cells conferred mild but significant improvements in glucose levels and glucose tolerance in HFD-induced obese mice. Therefore, regulation of Smad4 expression may be one of the mechanisms regulating physiological expansion of beta-cells during development of type 2 diabetes.


Asunto(s)
Dieta Alta en Grasa , Intolerancia a la Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Obesidad/metabolismo , Proteína Smad4/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal/fisiología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/genética , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Resistencia a la Insulina , Masculino , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/genética , Proteína Smad4/genética
11.
Cancer Gene Ther ; 22(5): 271-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25857359

RESUMEN

Although H-1 parvovirus is used as an antitumor agent, not much is known about the relationship between its specific tropism and oncolytic activity. We hypothesize that VP2, a major capsid protein of H-1 virus, determines H-1-specific tropism. To assess this, we constructed chimeric H-1 viruses expressing Kilham rat virus (KRV) capsid proteins, in their complete or partial forms. Chimeric H-1 viruses (CH1, CH2 and CH3) containing the whole KRV VP2 domain could not induce cytolysis in HeLa, A549 and Panc-1 cells. However, the other chimeric H-1 viruses (CH4 and CH5) expressing a partial KRV VP2 domain induced cytolysis. Additionally, the significant cytopathic effect caused by CH4 and CH5 infection in HeLa cells resulted from preferential viral amplification via DNA replication, RNA transcription and protein synthesis. Modeling of VP2 capsid protein showed that two variable regions (VRs) (VR0 and VR2) of H-1 VP2 protein protrude outward, because of the insertion of extra amino-acid residues, as compared with those of KRV VP2 protein. This might explain the precedence of H-1 VP2 protein over KRV in determining oncolytic activity in human cancer cells. Taking these results together, we propose that the VP2 protein of oncolytic H-1 parvovirus determines its specific tropism in human cancer cells.


Asunto(s)
Proteínas de la Cápside/metabolismo , Parvovirus H-1/fisiología , Neoplasias/virología , Infecciones por Parvoviridae/virología , Animales , Proteínas de la Cápside/genética , Parvovirus H-1/genética , Parvovirus H-1/metabolismo , Células HeLa , Humanos , Neoplasias/genética , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Ratas , Transfección
12.
Autoimmunity ; 27(2): 109-22, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9583742

RESUMEN

Insulin-dependent diabetes mellitus (IDDM) is caused by the progressive autoimmune destruction of insulin-producing pancreatic beta cells. Although the pathogenesis of autoimmune IDDM has been extensively studied, the precise mechanisms involved in the initiation and progression of beta cell destruction remain unclear. Animal models used in the study of IDDM, such as the BioBreeding (BB) rat and the nonobese diabetic (NOD) mouse, have greatly enhanced our understanding of the pathogenic mechanisms involved in this disease. In these animals, macrophages and/or dendritic cells are the first cell types to infiltrate the pancreatic islets. Macrophages must be involved in the pathogenesis of IDDM early on, since inactivation of macrophages results in the near-complete prevention of insulitis and diabetes in both NOD mice and BB rats. The presentation of beta cell-specific autoantigens by macrophages and/or dendritic cells to CD4+ T helper cells, in association with MHC class II molecules, is considered the initial step in the development of autoimmune IDDM. The activated macrophages secrete IL-12, which stimulates Th1 type CD4+ T cells. The CD4+ T cells secrete IFN-gamma and IL-2. IFN-gamma activates other resting macrophages, which, in turn, release cytokines, such as IL-1beta, TNF-alpha, and free radicals, which are toxic to beta cells. During this process, IL-2 and other cytokines induce the migration of CD8+ peripheral T cells to the inflamed islets, perhaps by inducing the expression of a specific homing receptor. The precytotoxic CD8+ T cells that bear beta cell-specific autoantigen receptors differentiate into cytotoxic effector T cells upon recognition of the beta cell-specific peptide bound to MHC class I molecules in the presence of beta cell-specific CD4+ T helper cells. The cytotoxic CD8+ T cells then effect beta cell damage by releasing perforin and granzyme, and by Fas-mediated apoptosis. In this way, macrophages, CD4+ T cells, and CD8+ T cells synergistically destroy beta cells, resulting in the onset of autoimmune IDDM.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Macrófagos/inmunología , Linfocitos T/inmunología , Animales , Muerte Celular/inmunología , Citotoxicidad Inmunológica , Diabetes Mellitus Tipo 1/patología , Humanos , Ratones , Ratas
13.
Autoimmunity ; 20(1): 19-24, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7578857

RESUMEN

Non-obese diabetic (NOD) mice spontaneously develop autoimmune type 1 diabetes. Earlier studies have shown that retroviruses appear to be associated with the development of the disease in these animals. This investigation was initiated to determine whether any retroviral genes are specifically expressed in pancreatic beta cells from NOD mice, in contrast to their non-diabetic, parental strain, ICR mice. Host chromosomal DNAs from pancreatic islets, kidneys, hearts, and stomachs of NOD and ICR mice contained an equal amount of A-type retroviral genome (DNA); however, A-type retroviral gag, pol, and env mRNAs were detected in only the pancreatic islets from NOD mice. Furthermore, group-specific retroviral antigen (p73 of A-type--gag gene product) was found by immunofluorescent staining using anti-p73 antibody in only pancreatic beta cells from NOD mice. On the basis of these observations, we suggest that tissue and strain differences in transcription of the retroviral genome and beta cell-specific expression of A-type retroviral group-specific antigen p73 in NOD mice may be involved in the initiation of beta cell-specific autoimmunity leading to type 1 diabetes in these animals.


Asunto(s)
Antígenos Virales/metabolismo , Diabetes Mellitus Tipo 1/virología , Islotes Pancreáticos/virología , ARN Mensajero/análisis , Proteínas de los Retroviridae/metabolismo , Retroviridae/inmunología , Animales , Diabetes Mellitus Tipo 1/inmunología , Epítopos/genética , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos ICR , Ratones Endogámicos NOD , Retroviridae/genética , Proteínas de los Retroviridae/genética , Especificidad de la Especie
14.
Ann N Y Acad Sci ; 928: 200-11, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11795511

RESUMEN

Insulin-dependent diabetes mellitus (IDDM), also known as type 1 diabetes, is an organ-specific autoimmune disease resulting from the destruction of insulin-producing pancreatic beta cells. The hypothesis that IDDM is an autoimmune disease has been considerably strengthened by the study of animal models such as the BioBreeding (BB) rat and the nonobese diabetic (NOD) mouse, both of which spontaneously develop a diabetic syndrome similar to human IDDM. Beta cell autoantigens, macrophages, dendritic cells, B lymphocytes, and T cells have been shown to be involved in the pathogenesis of autoimmune diabetes. Among the beta cell autoantigens identified, glutamic acid decarboxylase (GAD) has been extensively studied and is the best characterized. Beta cell-specific suppression of GAD expression in NOD mice results in the prevention of IDDM. Macrophages and/or dendritic cells are the first cell types to infiltrate the pancreatic islets. Macrophages play an essential role in the development and activation of beta cell-cytotoxic T cells. B lymphocytes play a role as antigen-presenting cells, and T cells have been shown to play a critical role as final effectors that kill beta cells. Cytokines secreted by immunocytes, including macrophages and T cells, may regulate the direction of the immune response toward Th1 or Th2 as well as cytotoxic effector cell or suppressor cell dominance. Beta cells are destroyed by apoptosis through Fas-Fas ligand and TNF-TNF receptor interactions and by granzymes and perforin released from cytotoxic effector T cells. Therefore, the activated macrophages and T cells, and cytokines secreted from these immunocytes, act synergistically to destroy beta cells, resulting in the development of autoimmune IDDM.


Asunto(s)
Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Diabetes Mellitus Tipo 1/inmunología , Glutamato Descarboxilasa/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno , Apoptosis , Autoanticuerpos/inmunología , Autoantígenos/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Subgrupos de Linfocitos B/inmunología , Citocinas/fisiología , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Glutamato Descarboxilasa/metabolismo , Humanos , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/patología , Isoenzimas/inmunología , Isoenzimas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Perforina , Proteínas Citotóxicas Formadoras de Poros , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/inmunología , Proteínas Tirosina Fosfatasas/metabolismo , Ratas , Ratas Endogámicas BB , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores , Serina Endopeptidasas/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
15.
Obstet Gynecol ; 96(1): 129-131, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10928901

RESUMEN

BACKGROUND: If medical management is unsuccessful in controlling postpartum hemorrhage, conservative surgical intervention or cesarean hysterectomy is required. TECHNIQUE: Hemostatic multiple square suturing using a straight number 7 or number 8 needle and number 1 chromic catgut is a new surgical technique to approximate anterior and posterior uterine walls, especially in areas where there is heavy bleeding. It controls postpartum hemorrhage by attachment and compression of the hemorrhage site of the endometrium or myometrium. EXPERIENCE: We used this technique in 23 women with postpartum hemorrhages at cesarean who did not respond to conservative treatment. In all 23 cases, bleeding decreased markedly and hysterectomy was avoided. All resumed normal menstrual flow after surgery. In four cases, further pregnancy was achieved after this method was used. CONCLUSION: Hemostatic multiple square suturing is an easy, safe, conservative surgical alternative to hysterectomy for treating uncontrollable postpartum hemorrhage.


Asunto(s)
Cesárea/efectos adversos , Hemostasis Quirúrgica/métodos , Trastornos Puerperales/cirugía , Técnicas de Sutura , Hemorragia Uterina/cirugía , Femenino , Humanos , Embarazo , Hemorragia Uterina/etiología
16.
Adv Exp Med Biol ; 347: 207-20, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-7976732

RESUMEN

Insulin-dependent diabetes mellitus (IDDM) is believed to be an autoimmune disease, characterized by lymphocytic infiltration of the islets and the presence of islet cell autoantibodies. Autoimmunity may result from an intrinsically abnormal immune system, primary alterations of the target beta cell or both. However, the initial event that causes the beta cell-specific autoimmunity remains unknown. Our recent experimental results showed that islet grafts from neonatal BB rats remained intact without insulitis when transplanted into the renal subcapsular space of acutely diabetic BB rats. In contrast, islet grafts from adult BB rats (which had been treated with silica for the prevention of insulitis) revealed severe insulitis and were rapidly destroyed. These results suggest that the delayed expression of a beta cell-specific autoantigen may result in the initiation of beta cell-specific autoimmunity. The islet cell-specific 37 kd autoantigen is not expressed early in the life of BB rats, but is expressed at around 30 days of age. This islet-specific autoantigen might be recognized and attacked by the immune effectors. In contrast, nondiabetic Wistar Furth rats express the autoantigen from birth.


Asunto(s)
Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Diabetes Mellitus Tipo 1/inmunología , Islotes Pancreáticos/inmunología , Animales , Autoanticuerpos/inmunología , Autoantígenos/genética , Autoantígenos/aislamiento & purificación , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/prevención & control , Secuencia de Bases , Clonación Molecular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/genética , Genes , Supervivencia de Injerto , Humanos , Trasplante de Islotes Pancreáticos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Ratas , Ratas Endogámicas BB , Proteínas Recombinantes de Fusión/inmunología , Dióxido de Silicio/farmacología , Dióxido de Silicio/uso terapéutico , Tiroiditis Autoinmune/genética , Tiroiditis Autoinmune/inmunología , Tiroiditis Autoinmune/prevención & control
17.
Arch Pharm Res ; 22(5): 437-47, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10549569

RESUMEN

Type I diabetes, also known as insulin-dependent diabetes mellitus (IDDM) results from the destruction of insulin-producing pancreatic beta cells by a progressive beta cell-specific autoimmune process. The pathogenesis of autoimmune IDDM has been extensively studied for the past two decades using animal models such as the non-obese diabetic (NOD) mouse and the BioBreeding (BB) rat. However, the initial events that trigger the immune responses leading to the selective destruction of the beta cells are poorly understood. It is thought that beta cell autoantigens are involved in the triggering of beta cell-specific autoimmunity. Among a dozen putative beta cell autoantigens, glutamic acid decarboxylase (GAD) has been proposed as perhaps the strongest candidate in both humans and the NOD mouse. In the NOD mouse, GAD, as compared with other beta cell autoantigens, provokes the earliest T cell proliferative response. The suppression of GAD expression in the beta cells results in the prevention of autoimmune diabetes in NOD mice. In addition, the major populations of cells infiltrating the islets during the early stage of insulitis in BB rats and NOD mice are macrophages and dendritic cells. The inactivation of macrophages in NOD mice results in the prevention of T cell mediated autoimmune diabetes. Macrophages are primary contributors to the creation of the immune environment conducive to the development and activation of beta cell-specific Th1-type CD4+ T cells and CD8+ cytotoxic T cells that cause autoimmune diabetes in NOD mice. CD4+ and CD8+ T cells are both believed to be important for the destruction of beta cells. These cells, as final effectors, can kill the insulin-producing beta cells by the induction of apoptosis. In addition, CD8+ cytotoxic T cells release granzyme and cytolysin (perforin), which are also toxic to beta cells. In this way, macrophages, CD4+ T cells and CD8+ T cells act synergistically to kill the beta cells in conjunction with beta cell autoantigens and MHC class I and class II antigens, resulting in the onset of autoimmune type I diabetes.


Asunto(s)
Autoantígenos/fisiología , Diabetes Mellitus Tipo 1/patología , Glutamato Descarboxilasa/fisiología , Macrófagos/fisiología , Linfocitos T/fisiología , Animales , Humanos
18.
Obesity (Silver Spring) ; 17(2): 355-62, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19039313

RESUMEN

To investigate the associations of uncoupling protein (UCP)2 and UCP3 gene variants with overweight and related traits, we genotyped UCP2-866G>A, UCP2Ala55Val, and UCP3-55C>T in 737 Korean children and 732 adults and collected data regarding anthropometric status and blood biochemistry. Information concerning the children's lifestyles and dietary habits was collected. The UCP2-866G>A and UCP3-55C>T gene variants showed significant associations with BMI level, waist circumference, and body weight in the children but not in the adults. Compared with -866GG carriers, the -866GA and AA carriers showed a strong decreasing trend in the risk for overweight (odds ratio (OR), 0.67; 95% confidence interval (CI), 0.45-1.01; P = 0.053). In comparison with UCP3-55CC carriers, children carrying -55CT and TT showed a significant reduction in the risk of overweight (OR, 0.67; 95% CI, 0.46-0.98; P = 0.039). There was also evidence of interactions between the effects of the combined UCP2-UCP3 genotype and obesity-related metabolic traits. The greatest protective effect against overweight was seen in those with the combined genotype non-UCP2-866GG and non-UCP3-55CC, as compared with those carrying both UCP2-866GG and UCP3-55CC (OR,0.60; 95% CI, 0.38-0.95; P = 0.030). In the subgroup with a low level of physical activity, UCP3-55CC carriers had higher BMI values than UCP3-55T carriers (16.6 +/- 2.3 kg/m(2) vs. 16.1 +/- 1.9 kg/m(2), P = 0.016). Low physical activity may aggravate the susceptibility to overweight in UCP2-866GG and UCP3-55CC carriers.


Asunto(s)
Canales Iónicos/genética , Proteínas Mitocondriales/genética , Obesidad/epidemiología , Obesidad/genética , Sobrepeso/epidemiología , Sobrepeso/genética , Adulto , Anciano , Índice de Masa Corporal , Peso Corporal/genética , Peso Corporal/fisiología , Niño , Ingestión de Alimentos/fisiología , Femenino , Predisposición Genética a la Enfermedad/genética , Genotipo , Encuestas Epidemiológicas , Humanos , Corea (Geográfico)/epidemiología , Masculino , Persona de Mediana Edad , Actividad Motora/fisiología , Obesidad/etnología , Sobrepeso/etnología , Factores de Riesgo , Proteína Desacopladora 2 , Proteína Desacopladora 3 , Circunferencia de la Cintura/genética , Circunferencia de la Cintura/fisiología
19.
Diabetologia ; 51(6): 1025-34, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18408913

RESUMEN

AIMS/HYPOTHESIS: Type 2 diabetes mellitus is a common age-dependent disease. We discovered that male offspring of non-diabetic C57BL/6 and DBA/2 mice, called JYD mice, develop type 2 diabetes when they grow old. JYD mice show characteristics of insulin resistance, hyperglycaemia and hyperinsulinaemia in old age without obesity. We postulated that the mechanism of age-dependent type 2 diabetes in this model relates to caveolin-1 status in skeletal muscle, which appears to regulate insulin sensitivity in the mice. METHODS: We compared insulin sensitivity in aged C57BL/6 and JYD mice using glucose and insulin tolerance tests and (18)F-fluorodeoxyglucose positron emission tomography. We also determined insulin signalling molecules and caveolin proteins using western blotting, and altered caveolin-1 levels in skeletal muscle of C57BL/6 and JYD mice using viral vector systems, to examine the effect of this on insulin sensitivity. RESULTS: In 30-week-old C57BL/6 and JYD mice, the basal levels of IRS-1, Akt and peroxisome proliferator-activated receptor-gamma decreased, as did insulin-stimulated phosphorylation of Akt and insulin receptor beta. However, caveolin-1 was only increased about twofold in 30-week-old JYD mice as compared with 3-week-old mice, whereas an eightfold increase was seen in C57BL/6 mice. Downregulation of caveolin-1 production in C57BL/6 mice caused severe impairment of glucose and insulin tolerance. Upregulation of caveolin-1 in aged diabetic JYD mice significantly improved insulin sensitivity with a concomitant increase of glucose uptake in the skeletal muscle. CONCLUSIONS/INTERPRETATION: The level of skeletal muscle caveolin-1 is correlated with the progression of age-dependent type 2 diabetes in JYD mice.


Asunto(s)
Envejecimiento/fisiología , Caveolina 1/fisiología , Diabetes Mellitus Tipo 2/fisiopatología , Músculo Esquelético/fisiopatología , Animales , Transporte Biológico , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Femenino , Fluorodesoxiglucosa F18/metabolismo , Prueba de Tolerancia a la Glucosa , Insulina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Tomografía de Emisión de Positrones
20.
Diabetologia ; 50(10): 2147-55, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17676307

RESUMEN

AIMS/HYPOTHESIS: Expression of T helper (Th)1 cytokine mRNA in pregnant women is known to be inversely correlated with serum human chorionic gonadotropin (hCG). Type 1 diabetes is a Th1-mediated autoimmune disease, in which intervention at an early stage of the autoimmune process can prevent disease progression. We hypothesised that immune modulation by treating young NOD mice with hCG may prevent diabetes. METHODS: Female NOD mice were treated with hCG or recombinant hCG from 3 to 15 weeks of age and the incidence of diabetes and development of insulitis was determined. CD4(+) and CD8(+) T cell populations, T cell proliferation, cytokine production and CD4(+)CD25(+) regulatory T cells were examined and adoptive transfer experiments were performed. RESULTS: Both purified and recombinant hCG prevented development of diabetes in NOD mice. hCG decreased the proportion and number of CD4(+) and CD8(+) T cells and inhibited T cell proliferative responses against beta cell antigens. hCG treatment suppressed IFN-gamma production, but increased IL-10 and TGF-beta production in splenocytes stimulated with anti-CD3 antibody. hCG treatment also suppressed TNF-alpha production in splenocytes stimulated with lipopolysaccharide. Furthermore, hCG treatment increased the CD4(+)CD25(+)/CD4(+) T cell ratio in spleen and pancreatic lymph nodes. Depletion of CD4(+)CD25(+) T cells from splenocytes of hCG-treated NOD mice abolished their preventive effect on diabetes transfer. CONCLUSIONS/INTERPRETATION: We conclude that hCG has an immunomodulatory effect by downregulating effector cells, including Th1 cells, CD8(+) T cells and macrophages, and increasing the CD4(+)CD25(+)/CD4(+) T cell ratio, thus preventing autoimmune diabetes in NOD mice.


Asunto(s)
Gonadotropina Coriónica/uso terapéutico , Diabetes Mellitus Tipo 1/prevención & control , Factores Inmunológicos/uso terapéutico , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Gonadotropina Coriónica/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Femenino , Citometría de Flujo , Ganglios Linfáticos/inmunología , Transfusión de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Embarazo , Bazo/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA