Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 2(3): e91001, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28194444

RESUMO

For nearly 100 years, growth hormone (GH) has been known to affect insulin sensitivity and risk of diabetes. However, the tissue governing the effects of GH signaling on insulin and glucose homeostasis remains unknown. Excess GH reduces fat mass and insulin sensitivity. Conversely, GH insensitivity (GHI) is associated with increased adiposity, augmented insulin sensitivity, and protection from diabetes. Here, we induce adipocyte-specific GHI through conditional deletion of Jak2 (JAK2A), an obligate transducer of GH signaling. Similar to whole-body GHI, JAK2A mice had increased adiposity and extreme insulin sensitivity. Loss of adipocyte Jak2 augmented hepatic insulin sensitivity and conferred resistance to diet-induced metabolic stress without overt changes in circulating fatty acids. While GH injections induced hepatic insulin resistance in control mice, the diabetogenic action was absent in JAK2A mice. Adipocyte GH signaling directly impinged on both adipose and hepatic insulin signal transduction. Collectively, our results show that adipose tissue governs the effects of GH on insulin and glucose homeostasis. Further, we show that JAK2 mediates liver insulin sensitivity via an extrahepatic, adipose tissue-dependent mechanism.


Assuntos
Adipócitos/efeitos dos fármacos , Hormônio do Crescimento/efeitos adversos , Resistência à Insulina , Janus Quinase 2/genética , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Camundongos , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico
2.
Mol Endocrinol ; 27(8): 1333-42, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23782652

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is considered the hepatic expression of the metabolic syndrome, and its prevalence is increasing. The factors that influence the development of fatty liver and its progression to steatohepatitis and cirrhosis are not well understood. The pleiotropic hormone, GH, has been associated with an increased risk of NAFLD in humans and mice. GH is known to have diverse effects on lipid metabolism including decreasing body fat in vivo, presumably through stimulation of lipolysis via an undefined mechanism. Previously we described mice with hepatocyte-specific deletion of the GH signaling mediator, Janus kinase 2 (JAK2L). JAK2L animals have elevated serum GH, reduced body fat, high liver triglyceride content, and increased serum markers of hepatocyte injury (alanine transaminase and aspartate transaminase). We aimed to determine whether the elevation of GH in JAK2L mice contributed to fatty liver by promoting lipolysis directly in adipocytes. We generated mice with adipocyte-specific disruption of JAK2 (JAK2A) and found that GH resistance in adipocytes reduced lipolysis and increased body fat. JAK2A mice were then crossed to JAK2L mice, and the resultant JAK2L/A animals had increased body fat and decreased lipolysis, despite elevated circulating GH. Furthermore, the increased triglyceride content, serum alanine transaminase, and serum aspartate transaminase observed in JAK2L mice were nearly normalized with the additional disruption of JAK2 in adipocytes (JAK2L/A mice). Our results offer novel mechanistic insights into the long-recognized effects of GH on lipid flux and suggest that GH signaling may play an important regulatory role in the development of NAFLD.


Assuntos
Fígado Gorduroso/metabolismo , Hormônio do Crescimento/metabolismo , Janus Quinase 2/genética , Metabolismo dos Lipídeos/genética , Lipólise/genética , Adipócitos/citologia , Tecido Adiposo/citologia , Tecido Adiposo/fisiologia , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Composição Corporal , Fígado Gorduroso/genética , Fibrose , Expressão Gênica , Hormônio do Crescimento/sangue , Janus Quinase 2/deficiência , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica , Receptores da Somatotropina , Transdução de Sinais , Triglicerídeos/análise , Triglicerídeos/metabolismo
3.
Clin Infect Dis ; 54(8): 1196-203, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22438347

RESUMO

BACKGROUND: Excess risk of cardiovascular disease occurs in effectively treated individuals with human immunodeficiency virus (HIV) infection. Although elevated plasma D-dimer levels are associated with increased morbidity and mortality, the impact of HIV infection on coagulation in vivo has not been well studied. METHODS: We measured D-dimers, antithrombin, endogenous thrombin potential (ETP; a functional measure of thrombin generation in vitro), thrombin/antithrombin complexes (TAT; a measure of thrombin generation in vivo), tissue factor, prothrombin fragment 1 + 2 (F1+2), and normalized APC sensitivity ratio (nAPCsr) in 199 HIV-positive men who were receiving antiretroviral therapy and had an undetectable HIV RNA level, in 79 HIV-positive untreated men, and in 39 uninfected controls. RESULTS: Median antithrombin levels were higher while the ETP was lower among HIV-infected adults (treated and untreated), compared with controls. There were few differences between coagulation markers in the 2 HIV groups. Compared with controls, the nAPCsr was lower in treated men and the TAT level was lower in untreated individuals. We observed little difference among measured levels of D-dimer, tissue factor, or F1+2 between HIV-infected individuals and controls. Antiretroviral therapy exposure was associated with a lower antithrombin level, a lower nAPCsr, and a lower ETP, while history of opportunistic infection was associated with a higher nAPCsr. CONCLUSIONS: HIV infection is associated with decreased thrombin generation, as measured by the ETP, and an increased antithrombin level. These data suggest that HIV infection may not be associated with increased propensity toward clotting, as has been suggested on the basis of isolated measures of D-dimer levels.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , Infecções por HIV/fisiopatologia , Trombina/análise , Adulto , Antitrombinas/análise , Transtornos da Coagulação Sanguínea/epidemiologia , Infecções por HIV/complicações , Humanos , Masculino , Pessoa de Meia-Idade
4.
Mol Endocrinol ; 25(7): 1223-30, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21527499

RESUMO

The relative contributions of circulating and locally produced IGF-I in growth remain controversial. The majority of circulating IGF-I is produced by the liver, and numerous mouse models have been developed to study the endocrine actions of IGF-I. A common drawback to these models is that the elimination of circulating IGF-I disrupts a negative feedback pathway, resulting in unregulated GH secretion. We generated a mouse with near total abrogation of circulating IGF-I by disrupting the GH signaling mediator, Janus kinase (JAK)2, in hepatocytes. We then crossed these mice, termed JAK2L, to GH-deficient little mice (Lit). Compound mutant (Lit-JAK2L) and control (Lit-Con) mice were treated with equal amounts of GH such that the only difference between the two groups was hepatic GH signaling. Both groups gained weight in response to GH but there was a reduction in the final weight of GH-treated Lit-JAK2L vs. Lit-Con mice. Similarly, lean mass increased in both groups, but there was a reduction in the final lean mass of Lit-JAK2L vs. Lit-Con mice. There was an equivalent increase in skeletal length in response to GH in Lit-Con and Lit-JAK2L mice. There was an increase in bone mineral density (BMD) in both groups, but Lit-JAK2L had lower BMD than Lit-Con mice. In addition, GH-mediated increases in spleen and kidney mass were absent in Lit-JAK2L mice. Taken together, hepatic GH-dependent production of IGF-I had a significant and nonredundant role in GH-mediated acquisition of lean mass, BMD, spleen mass, and kidney mass; however, skeletal length was dependent upon or compensated for by locally produced IGF-I.


Assuntos
Hormônio do Crescimento/farmacologia , Fator de Crescimento Insulin-Like I/metabolismo , Animais , Densidade Óssea , Feminino , Expressão Gênica , Hormônio do Crescimento/sangue , Hormônio do Crescimento/fisiologia , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Fator de Crescimento Insulin-Like I/genética , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Rim/anatomia & histologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Receptores da Somatotropina/genética , Receptores da Somatotropina/metabolismo , Baço/anatomia & histologia , Aumento de Peso
5.
J Clin Invest ; 121(4): 1412-23, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21364286

RESUMO

Non-alcoholic fatty liver disease is associated with multiple comorbid conditions, including diabetes, obesity, infection, and malnutrition. Mice with hepatocyte-specific disruption of growth hormone (GH) signaling develop fatty liver (FL), although the precise mechanism underlying this finding remains unknown. Because GH signals through JAK2, we developed mice bearing hepatocyte-specific deletion of JAK2 (referred to herein as JAK2L mice). These mice were lean, but displayed markedly elevated levels of GH, liver triglycerides (TGs), and plasma FFAs. Because GH is known to promote lipolysis, we crossed GH-deficient little mice to JAK2L mice, and this rescued the FL phenotype. Expression of the fatty acid transporter CD36 was dramatically increased in livers of JAK2L mice, as was expression of Pparg. Since GH signaling represses PPARγ expression and Cd36 is a known transcriptional target of PPARγ, we treated JAK2L mice with the PPARγ-specific antagonist GW9662. This resulted in reduced expression of liver Cd36 and decreased liver TG content. These results provide a mechanism for the FL observed in mice with liver-specific disruption in GH signaling and suggest that the development of FL depends on both GH-dependent increases in plasma FFA and increased hepatic uptake of FFA, likely mediated by increased expression of CD36.


Assuntos
Fígado Gorduroso/genética , Fígado Gorduroso/fisiopatologia , Hormônio do Crescimento/metabolismo , Janus Quinase 2/deficiência , Fígado/fisiopatologia , Anilidas/farmacologia , Animais , Antígenos CD36/metabolismo , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/metabolismo , Fígado Gorduroso/etiologia , Fígado Gorduroso/patologia , Feminino , Deleção de Genes , Hepatócitos/fisiologia , Janus Quinase 2/genética , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Especificidade de Órgãos , PPAR gama/antagonistas & inibidores , Transdução de Sinais , Triglicerídeos/metabolismo
6.
Blood ; 116(25): 5724-33, 2010 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-20823455

RESUMO

To explore the effect(s) of growth hormone signaling on thrombosis, we studied signal transduction and transcription factor 5 (STAT5)-deficient mice and found markedly reduced survival in an in vivo thrombosis model. These findings were not explained by a compensatory increase in growth hormone secretion. There was a modest increase in the activity of several procoagulant factors, but there was no difference in the rate or magnitude of thrombin generation in STAT5-deficient mice relative to control. However, thrombin-triggered clot times were markedly shorter, and fibrin polymerization occurred more rapidly in plasma from STAT5-deficient mice. Fibrinogen depletion and mixing studies indicated that the effect on fibrin polymerization was not due to intrinsic changes in fibrinogen, but resulted from changes in the concentration of a circulating plasma inhibitor. While thrombin-triggered clot times were significantly shorter in STAT5-deficient animals, reptilase-triggered clot times were unchanged. Accordingly, while the rate of thrombin-catalyzed release of fibrinopeptide A was similar, the release of fibrinopeptide B was accelerated in STAT5-deficient plasma versus control. Taken together, these studies demonstrated that the loss of STAT5 resulted in a decrease in the concentration of a plasma inhibitor affecting thrombin-triggered cleavage of fibrinopeptide B. This ultimately resulted in accelerated fibrin polymerization and greater thrombosis susceptibility in STAT5-deficient animals.


Assuntos
Fibrina/metabolismo , Embolia Pulmonar/metabolismo , Fator de Transcrição STAT5/fisiologia , Trombose/metabolismo , Animais , Coagulação Sanguínea , Modelos Animais de Doenças , Fator XIII/metabolismo , Fibrinopeptídeo B/metabolismo , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Embolia Pulmonar/patologia , Transdução de Sinais , Tempo de Trombina , Trombose/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA