Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Genet ; 31(4): 391-4, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12118252

RESUMO

The autoimmune disease type 1 diabetes mellitus (insulin-dependent diabetes mellitus, IDDM) has a multifactorial etiology. So far, the major histocompatibility complex (MHC) is the only major susceptibility locus that has been identified for this disease and its animal models. The Komeda diabetes-prone (KDP) rat is a spontaneous animal model of human type 1 diabetes in which the major susceptibility locus Iddm/kdp1 accounts, in combination with MHC, for most of the genetic predisposition to diabetes. Here we report the positional cloning of Iddm/kdp1 and identify a nonsense mutation in Cblb, a member of the Cbl/Sli family of ubiquitin-protein ligases. Lymphocytes of the KDP rat infiltrate into pancreatic islets and several tissues including thyroid gland and kidney, indicating autoimmunity. Similar findings in Cblb-deficient mice are caused by enhanced T-cell activation. Transgenic complementation with wildtype Cblb significantly suppresses development of the KDP phenotype. Thus, Cblb functions as a negative regulator of autoimmunity and Cblb is a major susceptibility gene for type 1 diabetes in the rat. Impairment of the Cblb signaling pathway may contribute to human autoimmune diseases, including type 1 diabetes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Diabetes Mellitus Tipo 1/genética , Ligases/genética , Ligases/metabolismo , Ubiquitina-Proteína Ligases , Molécula de Adesão de Leucócito Ativado/genética , Animais , Animais Geneticamente Modificados , Autoimunidade/genética , Mapeamento Cromossômico , Clonagem Molecular , Diabetes Mellitus Tipo 1/patologia , Feminino , Predisposição Genética para Doença , Heterozigoto , Ativação Linfocitária , Masculino , Camundongos , Dados de Sequência Molecular , Mutação , Proteínas Proto-Oncogênicas c-cbl , Ratos , Ratos Mutantes , Ratos Sprague-Dawley , Linfócitos T/metabolismo , Linfócitos T/patologia
2.
Nat Genet ; 30(2): 221-6, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11818964

RESUMO

The CBP protein (cAMP response element binding protein (CREB) binding protein) is a co-activator for several transcription factors with a wide range of important biological functions, such as sterol regulatory element binding proteins (SREBPs), CCAAT/enhancer-binding proteins (C/EBPs), nuclear receptors (including peroxisome proliferator-activated receptors, PPARs), and signal transducers and activators of transcription (STATs). In contrast to these individual transcription factors, the biological roles of CBP are poorly understood. CBP enhances transcriptional activities via histone acetylation and the recruitment of additional co-activators such as SRC (steroid coactivator)-1 (ref. 9). To identify its physiological functions using a loss-of-function mutant, we analyzed CBP-deficient mice. As Crebbp null mice (Crebbp-/-) died during embryogenesis, we used Crebbp+/- mice. Unexpectedly, Crebbp+/- mice showed markedly reduced weight of white adipose tissue (WAT) but not of other tissues. Despite this lipodystrophy, Crebbp+/- mice showed increased insulin sensitivity and glucose tolerance and were completely protected from body weight gain induced by a high-fat (HF) diet. We observed increased leptin sensitivity and increased serum adiponectin levels in Crebbp+/- mice. These increased effects of insulin-sensitizing hormones secreted from WAT may explain, at least in part, the phenotypes of Crebbp+/- mice. This study demonstrates that CBP may function as a 'master-switch' between energy storage and expenditure.


Assuntos
Resistência à Insulina/genética , Lipodistrofia/genética , Proteínas Nucleares/genética , Transativadores/genética , Adipócitos/metabolismo , Adipócitos/patologia , Tecido Adiposo/patologia , Tecido Adiposo Marrom/patologia , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteína de Ligação a CREB , Tamanho Celular , Proteínas de Ligação a DNA/metabolismo , Gorduras na Dieta/administração & dosagem , Metabolismo Energético , Heterozigoto , Resistência à Insulina/fisiologia , Lipodistrofia/patologia , Lipodistrofia/fisiopatologia , Camundongos , Camundongos Mutantes , Proteínas Nucleares/deficiência , Proteínas Nucleares/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1 , Transativadores/deficiência , Transativadores/fisiologia , Fatores de Transcrição/metabolismo
3.
J Clin Invest ; 117(1): 246-57, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17200721

RESUMO

Glucokinase (Gck) functions as a glucose sensor for insulin secretion, and in mice fed standard chow, haploinsufficiency of beta cell-specific Gck (Gck(+/-)) causes impaired insulin secretion to glucose, although the animals have a normal beta cell mass. When fed a high-fat (HF) diet, wild-type mice showed marked beta cell hyperplasia, whereas Gck(+/-) mice demonstrated decreased beta cell replication and insufficient beta cell hyperplasia despite showing a similar degree of insulin resistance. DNA chip analysis revealed decreased insulin receptor substrate 2 (Irs2) expression in HF diet-fed Gck(+/-) mouse islets compared with wild-type islets. Western blot analyses confirmed upregulated Irs2 expression in the islets of HF diet-fed wild-type mice compared with those fed standard chow and reduced expression in HF diet-fed Gck(+/-) mice compared with those of HF diet-fed wild-type mice. HF diet-fed Irs2(+/-) mice failed to show a sufficient increase in beta cell mass, and overexpression of Irs2 in beta cells of HF diet-fed Gck(+/-) mice partially prevented diabetes by increasing beta cell mass. These results suggest that Gck and Irs2 are critical requirements for beta cell hyperplasia to occur in response to HF diet-induced insulin resistance.


Assuntos
Gorduras na Dieta/farmacologia , Glucoquinase/fisiologia , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/patologia , Insulina/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Fosfoproteínas/fisiologia , Animais , Glucoquinase/deficiência , Glucoquinase/genética , Humanos , Hiperplasia , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Transdução de Sinais
4.
Comp Med ; 58(6): 560-7, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19149413

RESUMO

The Komeda miniature rat Ishikawa (KMI) is a spontaneous animal model of dwarfism caused by a mutation in Prkg2, which encodes cGMP-dependent protein kinase type II (cGKII). This strain has been maintained as a segregating inbred strain for the mutated allele mri. In this study, we characterized the phenotype of the KMI strain, particularly growth traits, craniofacial measurements, and organ weights. The homozygous mutant (mri/mri) animals were approximately 70% to 80% of the size of normal, heterozygous (mri/+) animals in regard to body length, weight, and naso-occipital length of the calvarium, and the retroperitoneal fat of mri/mri rats was reduced greatly. In addition, among progeny of the (BNxKMI-mri/mri)F1xKMI-mri/mri backcross, animals with the KMI phenotype (mri/mri) were easily distinguished from those showing the wild-type phenotype (mri/+) by using growth traits such as body length and weight. Genetic analysis revealed that all of the backcrossed progeny exhibiting the KMI phenotype were homozygous for the KMI allele in the 1.2-cM region between D14Rat5 and D14Rat80 on chromosome 14, suggesting strongly that mri acts in a completely recessive manner. The KMI strain is the first and only rat model with a confirmed mutation in Prkg2 and is a valuable model for studying dwarfism and longitudinal growth traits in humans and for functional studies of cGKII.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/genética , Nanismo/genética , Mutação , Animais , Peso Corporal/genética , Mapeamento Cromossômico , Proteína Quinase Dependente de GMP Cíclico Tipo II , Nanismo/enzimologia , Nanismo/patologia , Feminino , Masculino , Tamanho do Órgão/genética , Fenótipo , Ratos , Ratos Endogâmicos BN , Ratos Mutantes
5.
Diabetes ; 53(11): 2844-54, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15504964

RESUMO

Heterozygous peroxisome proliferator-activated receptor-gamma (PPAR-gamma)-deficient (PPARgamma(+/-)) mice were protected from high-fat diet-induced insulin resistance. To determine the impact of systemic reduction of PPAR-gamma activity on beta-cell function, we investigated insulin secretion in PPARgamma(+/-) mice on a high-fat diet. Glucose-induced insulin secretion in PPARgamma(+/-) mice was impaired in vitro. The tissue triglyceride (TG) content of the white adipose tissue, skeletal muscle, and liver was decreased in PPARgamma(+/-) mice, but it was unexpectedly increased in the islets, and the increased TG content in the islets was associated with decreased glucose oxidation. Administration of a PPAR-gamma agonist, pioglitazone, reduced the islet TG content in PPARgamma(+/-) mice on a high-fat diet and ameliorated the impaired insulin secretion in vitro. Our results demonstrate that PPAR-gamma protects islets from lipotoxicity by regulating TG partitioning among tissues and that a PPAR-gamma agonist can restore impaired insulin secretion under conditions of islet fat accumulation.


Assuntos
Gorduras na Dieta , Intolerância à Glucose/fisiopatologia , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , PPAR gama/fisiologia , Tiazolidinedionas/farmacologia , Triglicerídeos/metabolismo , Animais , Sequência de Bases , Primers do DNA , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos ICR , Camundongos Knockout , PPAR gama/deficiência , PPAR gama/genética , Pioglitazona
6.
Diabetes ; 53(9): 2261-70, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15331535

RESUMO

Mice lacking the p85alpha regulatory subunit of phosphoinositide (PI) 3-kinase (Pik3r1(-/-)) showed increased glucose uptake in white adipose tissue (WAT) and skeletal muscle due to increased phosphatidylinositol (3,4,5)-triphosphate [PtdIns(3,4,5)P3] production and on a normal diet had a body weight and fat mass similar to wild-type mice. After 3 months on a high-fat diet, Pik3r1(-/-) mice still had increased insulin sensitivity and better glucose tolerance than wild-type mice, but showed markedly greater increases in body weight and WAT mass than wild-type mice. On the normal diet, serum leptin levels of Pik3r1(-/-) mice were significantly higher than in wild-type mice as a result of increased leptin secretion from adipocytes, presumably due to the increased PtdIns(3,4,5)P3 production in adipocytes. Leptin (5 microg/g body wt per day) caused a reduction in food intake and decrease in body weight by the wild-type mice as well as Pik3r1(-/-) mice, suggesting Pik3r1(-/-) mice having leptin sensitivity similar to wild-type mice. The slightly increased serum leptin compensated for the increased glucose uptake by adipocytes in Pik3r1(-/-) mice, thereby preventing adiposity on the normal diet. On the high-fat diet, leptin (5 microg/g body wt per day) failed to decrease food intake or body weight in either genotype, indicating that both genotypes had indeed become severely leptin resistant. Consequently, leptin secretion was unable to sufficiently compensate for the severe leptin resistance caused by the high-fat diet, thereby failing to prevent obesity in Pik3r1(-/-) mice. Our findings suggest that primary increase in serum leptin on the normal diet play a role in the protection from adiposity in Pik3r1(-/-) mice.


Assuntos
Tecido Adiposo/enzimologia , Glucose/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Leptina/sangue , Obesidade/fisiopatologia , Fosfatidilinositol 3-Quinases/genética , Adipócitos/metabolismo , Adiponectina , Animais , Peso Corporal , Gorduras na Dieta/farmacologia , Feminino , Resistência à Insulina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Mutantes , Obesidade/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas/metabolismo , Uridina Difosfato N-Acetilgalactosamina/metabolismo
7.
Brain Res Mol Brain Res ; 112(1-2): 1-7, 2003 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-12670697

RESUMO

Reelin (Reln) is an extracellular matrix protein secreted from distinct neuronal populations and controls neural cell positioning during brain development. Alterations of human RELN have been reported in two pedigrees with an autosomal recessive lissencephaly. Although several alleles of the mouse reeler mutation were identified as disruptions of Reln, there is no other animal model with a confirmed mutation in Reln. We recently established the Komeda Zucker creeping (KZC) rat strain with an autosomal recessive mutation creeping (cre), showing a reeler-like phenotype. We also found that creeping was located in the genomic segment on rat chromosome 4 containing Reln and that the expression level of Reln mRNA was markedly reduced in cre/cre homozygous mutant animals. Here we report positional candidate cloning of creeping, and identify a nucleotide insertion mutation in Reln. This mutation leads to a translational frameshift and results in truncation of the predicted protein in the fourth reelin-specific repeat, removing the C-terminal region required for secretion and function of the protein. We conclude that the mutation detected here is causative and is probably a null allele. The KZC rat is the first rat model with a confirmed Reln mutation and would therefore contribute to the understanding of the Reln function.


Assuntos
Encéfalo/anormalidades , Moléculas de Adesão Celular Neuronais/deficiência , Proteínas da Matriz Extracelular/deficiência , Transtornos dos Movimentos/genética , Mutação/genética , Neurônios/metabolismo , Animais , Sequência de Bases/genética , Encéfalo/metabolismo , Encéfalo/patologia , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/isolamento & purificação , Mapeamento Cromossômico , Clonagem Molecular , Códon sem Sentido/genética , Análise Mutacional de DNA , DNA Complementar/análise , DNA Complementar/genética , Modelos Animais de Doenças , Éxons/genética , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/isolamento & purificação , Genótipo , Dados de Sequência Molecular , Transtornos dos Movimentos/metabolismo , Proteínas do Tecido Nervoso , Neurônios/patologia , Fenótipo , Ratos , Ratos Endogâmicos , Proteína Reelina , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Serina Endopeptidases
8.
Exp Anim ; 52(4): 295-301, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-14562605

RESUMO

The Komeda diabetes-prone (KDP) rat is a spontaneous animal model of human autoimmune type 1 diabetes. By positional cloning of the non-MHC major susceptibility locus lddm/kdp1, we recently identified a nonsense mutation in Cblb and also found that lymphocytes of KDP rats infiltrate into various tissues, indicating autoimmunity. The maintenance and production of KDP rats has been a critical problem owing to the poor reproductive ability of diabetic animals. To solve the problem, we here established the KDP rat as a segregating inbred strain. We first identified animals that were heterozygous at the lddm/kdp1 region in a breeding colony of KDP rats. The heterozygous region spans at least from D11Yok1 to Cblb on rat chromosome 11. By mating between the heterozygous rats, we obtained homozygotes, heterozygotes and wild-types with the expected ratio of 1:2:1 and found that only the homozygotes developed diabetes, suggesting that these genotypes represent those of lddm/kdp1. We then tried to maintain KDP rats by mating between the heterozygotes, which resulted in a segregating inbred strain. Within 210 d of age, about 80% of lddm/kdp1 homozygotes developed diabetes with severe insulitis, while neither heterozygotes nor wild-types developed diabetes. The phenotypic characteristics of the homozygotes are the same as those of progeny of diabetic parents in the original KDP rats. The segregating inbred KDP rat strain described here would serve as a useful animal model for autoimmune diseases, including type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/genética , Modelos Animais de Doenças , Ratos Endogâmicos/genética , Animais , Doenças Autoimunes/genética , Códon sem Sentido , Ratos
9.
Biochem Biophys Res Commun ; 314(3): 870-7, 2004 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-14741717

RESUMO

To characterize the underlying mechanisms of diabetes development in males of the Spontaneously Diabetic Torii (SDT) rat, a novel spontaneous model for diabetes, we chronologically examined them, focusing on their diabetic features and the pathological changes in the pancreatic islets. Male SDT rats exhibited glucose intolerance with impaired insulin secretion after 14 weeks and developed diabetes with remarkable hyperglycemia and marked hypoinsulinemia after 20 weeks. At prediabetic stage (10-20 weeks), they were normoglycemic, but had significantly lower insulin levels of plasma and pancreas than the normal rats. Their beta-cell volume was already smaller significantly at 10 weeks than that of normal rats. The primary changes of the pancreatic islets were microvascular events such as congestion and hemorrhage at 8-10 weeks. Thereafter, the SDT rat islets were affected with inflammation and progressive fibrosis (at 10-20 weeks), and eventually atrophied with a loss of beta-cells (at 38 weeks). These results indicate that the male SDT rats develop spontaneous diabetes with an absolute decrease in the insulin secretory capacity of the islets.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Animais , Área Sob a Curva , Glicemia/análise , Índice de Massa Corporal , Diabetes Mellitus Tipo 2/genética , Progressão da Doença , Teste de Tolerância a Glucose , Imuno-Histoquímica , Incidência , Insulina/sangue , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/ultraestrutura , Masculino , Tamanho do Órgão , Pâncreas/patologia , Ratos , Ratos Endogâmicos , Ratos Sprague-Dawley , Fatores de Tempo
10.
Biochem Biophys Res Commun ; 304(1): 196-206, 2003 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-12705906

RESUMO

The Spontaneously Diabetic Torii (SDT) rat has recently been established as a new rat model of nonobese type 2 diabetes. In this study, we characterized diabetic features in SDT rats, and performed quantitative trait locus (QTL) analysis for glucose intolerance using 319 male (BNxSDT)xSDT backcrosses. Male SDT rats exhibited glucose intolerance at 20 weeks, and spontaneously developed diabetes with the incidence of 100% at 38 weeks, and glucose intolerance is well associated with the development of diabetes. The QTL analysis identified three highly significant QTLs (Gisdt1, Gisdt2, and Gisdt3) for glucose intolerance on rat chromosomes 1, 2, and X, respectively. The SDT allele for these QTLs significantly exacerbated glucose intolerance. Furthermore, synergistic interactions among these QTLs were detected. These findings indicate that diabetic features in SDT rats are inherited as polygenic traits and that SDT rats would provide insights into genetics of human type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/genética , Modelos Animais de Doenças , Ratos Endogâmicos , Idade de Início , Animais , Glicemia/análise , Peso Corporal/genética , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/patologia , Genótipo , Intolerância à Glucose/sangue , Intolerância à Glucose/genética , Masculino , Fenótipo , Locos de Características Quantitativas , Ratos , Ratos Sprague-Dawley
11.
J Biol Chem ; 278(16): 14284-90, 2003 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-12493745

RESUMO

Although we and others have generated IRS-2 knock-out (IRS-2(-/-)) mice, significant differences were seen between the two lines of IRS-2(-/-) mice in the severity of diabetes and alterations of beta-cell mass. It has been reported that although IRS-1 and IRS-3 knock-out mice showed normal blood glucose levels, IRS-1/IRS-3 double knock-out mice exhibited marked hyperglycemia. Thus, IRS-1 and IRS-3 compensate each other's functions in maintaining glucose homeostasis. To assess the effect of genetic background and also ablation of IRS-3 on IRS-2(-/-), we generated IRS-2/IRS-3 double knock-out (IRS-2(-/-)IRS-3(-/-)) mice by crossing IRS-3(-/-) mice (129/Sv and C57Bl/6 background) with our IRS-2(-/-) mice (CBA and C57Bl/6 background). Intercrosses of IRS-2(+/-)IRS-3(+/-) mice yielded nine genotypes, and all of them including IRS-2(-/-)IRS-3(-/-) mice were apparently healthy and showed normal growth. However, at 10-20 weeks of age, 20-30% mice carrying a null mutation for the IRS-2 gene, irrespective of the IRS-3 genotype, developed diabetes. When mice with diabetes were excluded from the analysis of glucose and insulin tolerance test, IRS-2(-/-)IRS-3(-/-) showed a degree of glucose intolerance and insulin resistance similar to those of IRS-2(-/-) mice. Both IRS-2(-/-) and IRS-2(-/-)IRS-3(-/-) mice had moderately reduced beta-cell mass despite having insulin resistance. Insulin-positive beta-cells were decreased to nearly zero in IRS-2(-/-) mice with diabetes. Although Pdx1 and glucose transporter 2 expressions were essentially unaltered in islets from IRS-2(-/-) mice without diabetes, they were dramatically decreased in IRS-2(-/-) mice with diabetes. Taken together, these observations indicate that IRS-3 does not play a role compensating for the loss of IRS-2 in maintaining glucose homeostasis and that the severity of diabetes in IRS-2(-/-) mice depends upon genetic background, suggesting the existence of modifier gene(s) for diabetes in mice of the 129/Sv genetic strain.


Assuntos
Proteínas de Homeodomínio , Fosfoproteínas/genética , Animais , Glicemia/metabolismo , Cruzamentos Genéticos , Diabetes Mellitus/genética , Relação Dose-Resposta a Droga , Feminino , Genótipo , Glucose/metabolismo , Transportador de Glucose Tipo 2 , Hiperglicemia/genética , Imuno-Histoquímica , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Proteínas de Transporte de Monossacarídeos/metabolismo , Mutação , Fosfoproteínas/fisiologia , Especificidade da Espécie , Fatores de Tempo , Transativadores/metabolismo
12.
Genes Dev ; 18(19): 2418-29, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15466490

RESUMO

The Komeda miniature rat Ishikawa (KMI) is a naturally occurring mutant caused by an autosomal recessive mutation mri, which exhibits longitudinal growth retardation. Here we identified the mri mutation as a deletion in the rat gene encoding cGMP-dependent protein kinase type II (cGKII). KMIs showed an expanded growth plate and impaired bone healing with abnormal accumulation of postmitotic but nonhypertrophic chondrocytes. Ex vivo culture of KMI chondrocytes reproduced the differentiation impairment, which was restored by introducing the adenovirus-mediated cGKII gene. The expression of Sox9, an inhibitory regulator of hypertrophic differentiation, persisted in the nuclei of postmitotic chondrocytes of the KMI growth plate. Transfection experiments in culture systems revealed that cGKII attenuated the Sox9 functions to induce the chondrogenic differentiation and to inhibit the hypertrophic differentiation of chondrocytes. This attenuation of Sox9 was due to the cGKII inhibition of nuclear entry of Sox9. The impaired differentiation of cultured KMI chondrocytes was restored by the silencing of Sox9 through RNA interference. Hence, the present study for the first time shed light on a novel role of cGKII as a molecular switch, coupling the cessation of proliferation and the start of hypertrophic differentiation of chondrocytes through attenuation of Sox9 function.


Assuntos
Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Condrócitos/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Animais , Sequência de Bases , Proteína Quinase Dependente de GMP Cíclico Tipo II , Primers do DNA , Feminino , Lâmina de Crescimento/citologia , Proteínas de Grupo de Alta Mobilidade/fisiologia , Masculino , Ratos , Fatores de Transcrição SOX9 , Fatores de Transcrição/fisiologia
13.
J Biol Chem ; 278(44): 43691-8, 2003 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-12869553

RESUMO

We previously demonstrated that Irs2-/- mice develop diabetes due to beta-cell growth failure and insulin resistance; however, glucose-induced insulin secretion was increased in islets isolated from Irs2-/- mice. Pdx-1, a transcription factor important for maintenance of the beta-cell function, was recently reported to be severely reduced in Irs2-/- murine beta-cells. We report herein that Pdx-1 expression, including the amount of Pdx-1 localized in the nucleus, is not down-regulated in our Irs2-/- murine beta-cells with a C57BL/6 background. We have also demonstrated the expression of upstream genes of Pdx-1, such as HNF3beta and HNF1alpha, as well as its downstream genes, including insulin, Glut2, and Nkx6.1, to be well preserved. We have further demonstrated Pdx-1 expression to also be preserved in beta-cells of 30-week-old diabetic Irs2-/- mice. In addition, surprisingly, even in Irs2-/- mice on a high fat diet with markedly elevated blood glucose, exceeding 400 mg/dl, Pdx-1 expression was not reduced. Furthermore, we found Pdx-1 to be markedly decreased in certain severely diabetic Irs2-/- mice with a mixed C57BL/6J x 129Sv background. We conclude that 1) Pdx-1 expression in Irs2-/- mice is regulated in a strain-dependent manner, 2) Irs2-/- mice develop diabetes associated with beta-cell growth failure even when Pdx1 expression is preserved, and 3) Pdx-1 expression is preserved in severely hyperglycemic Irs2-/- mice with a C57BL/6 background on a high fat diet.


Assuntos
Regulação da Expressão Gênica , Ilhotas Pancreáticas/metabolismo , Fosfoproteínas/genética , Transativadores/biossíntese , Transativadores/fisiologia , Ração Animal , Animais , Western Blotting , Regulação para Baixo , Feminino , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Hipoglicemia/metabolismo , Imuno-Histoquímica , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina , Secreção de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfoproteínas/fisiologia , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Fatores de Tempo
14.
J Biol Chem ; 278(4): 2461-8, 2003 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-12431986

RESUMO

The adipocyte-derived hormone adiponectin has been shown to play important roles in the regulation of energy homeostasis and insulin sensitivity. In this study, we analyzed globular domain adiponectin (gAd) transgenic (Tg) mice crossed with leptin-deficient ob/ob or apoE-deficient mice. Interestingly, despite an unexpected similar body weight, gAd Tg ob/ob mice showed amelioration of insulin resistance and beta-cell degranulation as well as diabetes, indicating that globular adiponectin and leptin appeared to have both distinct and overlapping functions. Amelioration of diabetes and insulin resistance was associated with increased expression of molecules involved in fatty acid oxidation such as acyl-CoA oxidase, and molecules involved in energy dissipation such as uncoupling proteins 2 and 3 and increased fatty acid oxidation in skeletal muscle of gAd Tg ob/ob mice. Moreover, despite similar plasma glucose and lipid levels on an apoE-deficient background, gAd Tg apoE-deficient mice showed amelioration of atherosclerosis, which was associated with decreased expression of class A scavenger receptor and tumor necrosis factor alpha. This is the first demonstration that globular adiponectin can protect against atherosclerosis in vivo. In conclusion, replenishment of globular adiponectin may provide a novel treatment modality for both type 2 diabetes and atherosclerosis.


Assuntos
Apolipoproteínas E/genética , Arteriosclerose/genética , Diabetes Mellitus/genética , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas/química , Proteínas/fisiologia , Adiponectina , Animais , Apolipoproteínas E/fisiologia , Arteriosclerose/metabolismo , Arteriosclerose/patologia , Northern Blotting , DNA Complementar/metabolismo , Diabetes Mellitus/patologia , Relação Dose-Resposta a Droga , Immunoblotting , Insulina/sangue , Resistência à Insulina , Ilhotas Pancreáticas/metabolismo , Ligantes , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Obesos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Proteínas/genética , Proteínas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Risco , Fatores de Tempo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA