Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Endocrinology ; 157(2): 799-809, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26646203

RESUMO

The WNT pathway was shown to play an important role in the adult central nervous system. We previously identified the WNT pathway as a novel integration site of the adipokine leptin in mediating its neuroendocrine control of metabolism in obese mice. Here we investigated the implication of WNT signaling in seasonal body weight regulation exhibited by the Djungarian hamster (Phodopus sungorus), a seasonal mammal that exhibits profound annual changes in leptin sensitivity. We furthermore investigated whether crucial components of the WNT pathway are regulated in a diurnal manner. Gene expression of key components of the WNT pathway in the hypothalamus of hamsters acclimated to either long day (LD) or short day (SD) photoperiod was analyzed by in situ hybridization. We detected elevated expression of the genes WNT-4, Axin-2, Cyclin-D1, and SFRP-2, in the hypothalamic arcuate nucleus, a key energy balance integration site, during LD compared with SD as well as a diurnal regulation of Axin-2, Cyclin-D1, and DKK-3. Investigating the effect of photoperiod as well as leptin on the activation (phosphorylation) of the WNT coreceptor LRP-6-(Ser1490) by immunohistochemistry, we found elevated activity in the arcuate nucleus during LD relative to SD as well as after leptin treatment (2 mg/kg body weight). These findings indicate that differential WNT signaling may be associated with seasonal body weight regulation and is partially regulated in a diurnal manner in the adult brain. Furthermore, they suggest that this pathway plays a key role in the neuroendocrine regulation of body weight and integration of the leptin signal.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Proteína Axina/genética , Peso Corporal/genética , Ritmo Circadiano/genética , Ciclina D1/genética , Fotoperíodo , Via de Sinalização Wnt/genética , Proteína Wnt4/genética , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Proteína Axina/efeitos dos fármacos , Proteína Axina/metabolismo , Peso Corporal/efeitos dos fármacos , Ritmo Circadiano/efeitos dos fármacos , Cricetinae , Ciclina D1/efeitos dos fármacos , Ciclina D1/metabolismo , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Feminino , Perfilação da Expressão Gênica , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Leptina/farmacologia , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Phodopus , Estações do Ano , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt4/efeitos dos fármacos , Proteína Wnt4/metabolismo
2.
Diabetes ; 64(6): 2015-27, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25626735

RESUMO

Metabolic inflammation in the central nervous system might be causative for the development of overnutrition-induced metabolic syndrome and related disorders, such as obesity, leptin and insulin resistance, and type 2 diabetes. Here we investigated whether nutritive and genetic inhibition of the central IκB kinase ß (IKKß)/nuclear factor-κB (NF-κB) pathway in diet-induced obese (DIO) and leptin-deficient mice improves these metabolic impairments. A known prominent inhibitor of IKKß/NF-κB signaling is the dietary flavonoid butein. We initially determined that oral, intraperitoneal, and intracerebroventricular administration of this flavonoid improved glucose tolerance and hypothalamic insulin signaling. The dose-dependent glucose-lowering capacity was profound regardless of whether obesity was caused by leptin deficiency or high-fat diet (HFD). To confirm the apparent central role of IKKß/NF-κB signaling in the control of glucose and energy homeostasis, we genetically inhibited this pathway in neurons of the arcuate nucleus, one key center for control of energy homeostasis, via specific adeno-associated virus serotype 2-mediated overexpression of IκBα, which inhibits NF-κB nuclear translocation. This treatment attenuated HFD-induced body weight gain, body fat mass accumulation, increased energy expenditure, and reduced arcuate suppressor of cytokine signaling 3 expression, indicative for enhanced leptin signaling. These results reinforce a specific role of central proinflammatory IKKß/NF-κB signaling in the development and potential treatment of DIO-induced comorbidities.


Assuntos
Glicemia/metabolismo , Gorduras na Dieta/efeitos adversos , Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Obesidade/metabolismo , Animais , Composição Corporal/fisiologia , Linhagem Celular , Intolerância à Glucose , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Obesidade/etiologia , Transdução de Sinais/fisiologia
3.
Endocrinology ; 154(12): 4737-45, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24105484

RESUMO

The WNT pathway has been well characterized in embryogenesis and tumorigenesis. In humans, specific polymorphisms in the T cell-specific transcription factor 7 and the WNT coreceptor, low-density lipoprotein receptor-related protein-6 (LRP-6), both prominent components of this pathway, correlate with a higher incidence of type 2 diabetes, suggesting that the WNT pathway might be involved in the control of adult glucose homeostasis. We previously demonstrated that glycogen-synthase-kinase-3ß (GSK-3ß), the key enzyme of the WNT pathway, is increased in the hypothalamus during obesity and exacerbates high-fat diet-induced weight gain as well as glucose intolerance. These data suggest that WNT action in the hypothalamus might be required for normal glucose homeostasis. Here we characterized whether WNT signaling in general is altered in the hypothalamus of adult obese mice relative to controls. First we identified expression of multiple components of this pathway in the murine arcuate nucleus by in situ hybridization. In this region mRNA of ligands and target genes of the WNT pathway were down-regulated in obese and glucose-intolerant leptin-deficient mice. Similarly, the number of cells immunoreactive for the phosphorylated (active) form of the WNT-coreceptor LRP-6 was also decreased in leptin-deficient mice. Leptin treatment normalized expression of the WNT-target genes Axin-2 and Cylin-D1 and increased the number of phospho-LRP-6-immunoreactive cells reaching levels of lean controls. Leptin also increased the levels of phosphorylated (inactive) GSK-3ß in the arcuate nucleus, and this effect was colocalized to neuropeptide Y neurons, suggesting that inactivation of GSK-3ß may contribute to the neuroendocrine control of energy homeostasis. Taken together our findings identify hypothalamic WNT signaling as an important novel pathway that integrates peripheral information of the body's energy status encoded by leptin.


Assuntos
Hipotálamo/fisiologia , Leptina/farmacologia , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Via de Sinalização Wnt/fisiologia , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Cromatina , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos , Miocárdio/metabolismo , Obesidade/genética , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Testículo/metabolismo , Testosterona/farmacologia
4.
Nat Commun ; 4: 2140, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23860571

RESUMO

Endothermy has facilitated mammalian species radiation, but the sequence of events leading to sustained thermogenesis is debated in multiple evolutionary models. Here we study the Lesser hedgehog tenrec (Echinops telfairi), a phylogenetically ancient, 'protoendothermic' eutherian mammal, in which constantly high body temperatures are reported only during reproduction. Evidence for nonshivering thermogenesis is found in vivo during periodic ectothermic-endothermic transitions. Anatomical studies reveal large brown fat-like structures in the proximity of the reproductive organs, suggesting physiological significance for parental care. Biochemical analysis demonstrates high mitochondrial proton leak catalysed by an uncoupling protein 1 ortholog. Strikingly, bioenergetic profiling of tenrec uncoupling protein 1 reveals similar thermogenic potency as modern mouse uncoupling protein 1, despite the large phylogenetic distance. The discovery of functional brown adipose tissue in this 'protoendothermic' mammal links nonshivering thermogenesis directly to the roots of eutherian evolution, suggesting physiological importance prior to sustained body temperatures and migration to the cold.


Assuntos
Tecido Adiposo Marrom/fisiologia , Eulipotyphla/fisiologia , Canais Iônicos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Reprodução/fisiologia , Termogênese/fisiologia , Adaptação Fisiológica , Animais , Evolução Biológica , Temperatura Corporal/fisiologia , Feminino , Expressão Gênica , Células HEK293 , Humanos , Canais Iônicos/genética , Masculino , Camundongos , Proteínas Mitocondriais/genética , Filogenia , Proteína Desacopladora 1
5.
J Comp Physiol B ; 183(8): 1101-11, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23860586

RESUMO

The profound seasonal cycle in body weight exhibited by the Djungarian hamster (Phodopus sungorus) is associated with the development of hypothalamic leptin resistance during long day photoperiod (LD, 16:8 h light dark cycle), when body weight is elevated relative to short day photoperiod (SD, 8:16 h light dark cycle). We previously have shown that this seasonal change in physiology is associated with higher levels of mRNA for the potent inhibitor of leptin signaling, suppressor of cytokine signaling-3 (SOCS3), in the arcuate nucleus (ARC) of LD hamsters relative to hamsters in SD. The alteration in SOCS3 gene expression preceded the body weight change suggesting that SOCS3 might be the molecular switch of seasonal body weight changes. To functionally characterize the role of SOCS3 in seasonal body weight regulation, we injected SOCS3 expressing recombinant adeno-associated virus type-2 (rAAV2-SOCS3) constructs into the ARC of leptin sensitive SD hamsters immediately after weaning. Hamsters that received rAAV2 expressing enhanced green fluorescent protein (rAAV2-EGFP) served as controls. ARC-directed SOCS3 overexpression led to a significant increase in body weight over a period of 12 weeks without fully restoring the LD phenotype. This increase was partially due to elevated brown and white adipose tissue mass. Gene expression of pro-opiomelanocortin was increased while thyroid hormone converting enzyme DIO3 mRNA levels were reduced in SD hamsters with SOCS3 overexpression. In conclusion, our data suggest that ARC-directed SOCS3 overexpression partially overcomes the profound seasonal body weight cycle exhibited by the hamster which is associated with altered pro-opiomelanocortin and DIO3 gene expression.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal/fisiologia , Regulação da Expressão Gênica/fisiologia , Phodopus/metabolismo , Estações do Ano , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Tecido Adiposo/fisiologia , Análise de Variância , Animais , Clonagem Molecular , Cricetinae , Primers do DNA/genética , Dependovirus , Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde/metabolismo , Hibridização In Situ , Fotoperíodo , Pró-Opiomelanocortina/metabolismo , Proteínas Supressoras da Sinalização de Citocina/administração & dosagem
6.
Br J Nutr ; 109(6): 1040-51, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-22850125

RESUMO

Secondary metabolites of herbs and spices are widely used as an alternative strategy in the therapy of various diseases. The polyphenols naringenin, quercetin and curcumin have been characterised as anti-diabetic agents. Conversely, in vitro, naringenin and quercetin are described to inhibit phosphoinositide-3-kinase (PI3K), an enzyme that is essential for the neuronal control of whole body glucose homoeostasis. Using both in vitro and in vivo experiments, we tested whether the inhibitory effect on PI3K occurs in neurons and if it might affect whole body glucose homoeostasis. Quercetin was found to inhibit basal and insulin-induced phosphorylation of Akt (Ser473), a downstream target of PI3K, in HT-22 cells, whereas naringenin and curcumin had no effect. In Djungarian hamsters (Phodopus sungorus) naringenin and quercetin (10 mg/kg administered orally) diminished insulin-induced phosphorylation of Akt (Ser473) in the arcuate nucleus, indicating a reduction in hypothalamic PI3K activity. In agreement with this finding, glucose tolerance in naringenin-treated hamsters (oral) and mice (oral and intracerebroventricular) was reduced compared with controls. Dietary quercetin also impaired glucose tolerance, whereas curcumin was ineffective. Circulating levels of insulin and insulin-like growth factor-binding protein were not affected by the polyphenols. Oral quercetin reduced the respiratory quotient, suggesting that glucose utilisation was impaired after treatment. These data demonstrate that low doses of naringenin and quercetin acutely and potently impair glucose homoeostasis. This effect may be mediated by inhibition of hypothalamic PI3K signalling. Whether chronic impairments in glucose homoeostasis occur after long-term application remains to be identified.


Assuntos
Flavanonas/farmacologia , Glucose/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Quercetina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Linhagem Celular , Cricetinae , Dieta , Inibidores Enzimáticos/farmacologia , Feminino , Intolerância à Glucose/induzido quimicamente , Homeostase/efeitos dos fármacos , Hipoglicemiantes , Hipotálamo/efeitos dos fármacos , Insulina/sangue , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Camundongos , Phodopus , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo
7.
Biochem J ; 447(1): 175-84, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22849606

RESUMO

GSK3ß (glycogen synthase kinase 3ß) is a ubiquitous kinase that plays a key role in multiple intracellular signalling pathways, and increased GSK3ß activity is implicated in disorders ranging from cancer to Alzheimer's disease. In the present study, we provide the first evidence of increased hypothalamic signalling via GSK3ß in leptin-deficient Lep(ob/ob) mice and show that intracerebroventricular injection of a GSK3ß inhibitor acutely improves glucose tolerance in these mice. The beneficial effect of the GSK3ß inhibitor was dependent on hypothalamic signalling via PI3K (phosphoinositide 3-kinase), a key intracellular mediator of both leptin and insulin action. Conversely, neuron-specific overexpression of GSK3ß in the mediobasal hypothalamus exacerbated the hyperphagia, obesity and impairment of glucose tolerance induced by a high-fat diet, while having little effect in controls fed standard chow. These results demonstrate that increased hypothalamic GSK3ß signalling contributes to deleterious effects of leptin deficiency and exacerbates high-fat diet-induced weight gain and glucose intolerance.


Assuntos
Ingestão de Alimentos/fisiologia , Glucose/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Hipotálamo/enzimologia , Animais , Núcleo Arqueado do Hipotálamo/enzimologia , Núcleo Arqueado do Hipotálamo/fisiologia , Sequência de Bases , Primers do DNA/genética , Dieta Hiperlipídica/efeitos adversos , Intolerância à Glucose/enzimologia , Intolerância à Glucose/etiologia , Quinase 3 da Glicogênio Sintase/deficiência , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Hipotálamo/fisiologia , Leptina/deficiência , Leptina/genética , Masculino , Camundongos , Camundongos Knockout , Obesidade/enzimologia , Obesidade/etiologia , Transdução de Sinais , Aumento de Peso/fisiologia
8.
J Comp Physiol B ; 182(4): 553-67, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22198805

RESUMO

The Siberian hamster, Phodopus sungorus, undergoes a striking seasonal cycle of leptin sensitivity and body weight regulation, but the molecular mechanism and relevance to human leptin insensitivity are unknown. Here we show that nuclear translocation of phospho-STAT3 in the hypothalamus is rapidly stimulated by leptin to a greater extent in hamsters held in short-day length (SD) as compared to long-day length (LD). Intriguingly, effects of leptin on STAT3 appeared to be in part limited to nuclear translocation of phospho-STAT3 associated with the cell surface rather than phosphorylation of STAT3. The number of phospho-ERK cells within the hypothalamus was unaffected by either photoperiod or leptin. However, proximal to ERK phosphorylation, hypothalamic SH2-containing tyrosine phosphatase (SHP2) and the small growth factor receptor-binding protein (GRB2), which act as competitive negative modulators on binding of SOCS3 to leptin receptor (LRb)-associated Tyr985, were increased in SD compared to LD. Our findings suggest that activation of STAT3 by leptin may be dependent on interaction of stimulatory SHP2/GRB2 as well as inhibitory SOCS3 on the level of competitive binding to LRb-associated Tyr985. This hypothetical mechanism may represent the molecular identity of seasonally induced adjustments in leptin sensitivity and may be applied to investigating leptin sensitivity in other rodent models.


Assuntos
Proteína Adaptadora GRB2/metabolismo , Hipotálamo/metabolismo , Janus Quinase 2/metabolismo , Leptina/metabolismo , Phodopus/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Animais , Núcleo Celular , Cricetinae , Feminino , Regulação da Expressão Gênica , Hipotálamo/citologia , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fotoperíodo , Isoformas de Proteínas/metabolismo , Transporte Proteico , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , RNA Mensageiro/metabolismo , Receptores para Leptina/metabolismo , Fator de Transcrição STAT3/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo
9.
Cell Metab ; 13(4): 389-400, 2011 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-21459324

RESUMO

The aberrant accumulation of lipids in the liver ("fatty liver") is tightly associated with several components of the metabolic syndrome, including type 2 diabetes, coronary heart disease, and atherosclerosis. Here we show that the impaired hepatic expression of transcriptional cofactor transducin beta-like (TBL) 1 represents a common feature of mono- and multigenic fatty liver mouse models. Indeed, the liver-specific ablation of TBL1 gene expression in healthy mice promoted hypertriglyceridemia and hepatic steatosis under both normal and high-fat dietary conditions. TBL1 deficiency resulted in inhibition of fatty acid oxidation due to impaired functional cooperation with its heterodimerization partner TBL-related (TBLR) 1 and the nuclear receptor peroxisome proliferator-activated receptor (PPAR) α. As TBL1 expression levels were found to also inversely correlate with liver fat content in human patients, the lack of hepatic TBL1/TBLR1 cofactor activity may represent a molecular rationale for hepatic steatosis in subjects with obesity and the metabolic syndrome.


Assuntos
Fígado Gorduroso/etiologia , Hipertrigliceridemia/etiologia , Fígado/metabolismo , Transducina/metabolismo , Animais , Gorduras na Dieta/farmacologia , Dimerização , Modelos Animais de Doenças , Humanos , Metabolismo dos Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Camundongos Transgênicos , Proteínas Nucleares/metabolismo , PPAR alfa/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Transducina/antagonistas & inibidores , Transducina/genética
10.
Diabetes ; 58(5): 1040-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19188430

RESUMO

OBJECTIVE: In mammals, proper storage and distribution of lipids in and between tissues is essential for the maintenance of energy homeostasis. In contrast, aberrantly high levels of triglycerides in the blood ("hypertriglyceridemia") represent a hallmark of the metabolic syndrome and type 2 diabetes. As hypertriglyceridemia has been identified as an important risk factor for cardiovascular complications, in this study we aimed to identify molecular mechanisms in aberrant triglyceride elevation under these conditions. RESEARCH DESIGN AND METHODS: To determine the importance of hepatic lipid handling for systemic dyslipidemia, we profiled the expression patterns of various hepatic lipid transporters and receptors under healthy and type 2 diabetic conditions. A differentially expressed lipoprotein receptor was functionally characterized by generating acute, liver-specific loss- and gain-of-function animal models. RESULTS: We show that the hepatic expression of lipid transporter lipolysis-stimulated lipoprotein receptor (LSR) is specifically impaired in mouse models of obesity and type 2 diabetes and can be restored by leptin replacement. Experimental imitation of this pathophysiological situation by liver-specific knockdown of LSR promotes hypertriglyceridemia and elevated apolipoprotein (Apo)B and E serum levels in lean wild-type and ApoE knockout mice. In contrast, genetic restoration of LSR expression in obese animals to wild-type levels improves serum triglyceride levels and serum profiles in these mice. CONCLUSIONS: The dysregulation of hepatic LSR under obese and diabetic conditions may provide a molecular rationale for systemic dyslipidemia in type 2 diabetes and the metabolic syndrome and represent a novel target for alternative treatment strategies in these patients.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Hiperlipidemias/fisiopatologia , Fígado/fisiologia , Receptores de LDL/genética , Receptores de Lipoproteínas/fisiologia , Animais , Apolipoproteínas E/deficiência , Glicemia/metabolismo , Colesterol/sangue , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/sangue , Corpos Cetônicos/sangue , Lipólise , Lipoproteínas VLDL/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Interferência de RNA , Receptores de LDL/deficiência , Receptores de LDL/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Triglicerídeos/sangue
11.
Am J Physiol Regul Integr Comp Physiol ; 291(3): R643-50, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16601260

RESUMO

During the last 5 years it has been well established that photoperiod-induced changes in body weight in the seasonal hamster, Phodopus sungorus, are accompanied by a marked seasonal cycle in leptin sensitivity. In the present study, we investigated the possible involvement of insulin signaling in seasonal body weight regulation. We analyzed the expression pattern and relative intensity of insulin receptor (IR), phosphatidylinositol 3-kinase (PI3-kinase), and protein tyrosine phosphatase 1B (PTP1B) mRNAs by in situ hybridization in the brains of juvenile female hamsters acclimated to either long- (LD) or short-day length (SD) for 8 wk, with or without superimposed food deprivation for 48 h. Furthermore, the hypothalamic concentration and distribution of phospho-AKT, a marker of PI3-kinase activity was determined by immunoblotting and immunohistochemistry. Eight weeks of acclimation to SD led to a substantial downregulation of IR, PTP1B gene expression, and phospho-AKT concentration in this brain region, whereas PI3-kinase mRNA was unchanged. Food deprivation induced a decrease in PTP1B and a trend toward lowered IR gene expression in LD but not in SD. Additionally, a striking increase in PTP1B gene expression in the thalamus was observed after food deprivation in both photoperiods. The direction of change in neuronal insulin signaling contrasts to the central catabolic nature of this pathway described in other species. SD-induced reduction in insulin signaling may be due to decline in body fat stores mediated by enhanced central leptin sensitivity. Increased anorexigenic tone of leptin may overwrite central insulin signaling to prevent catabolic overdrive.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Insulina/metabolismo , Phodopus/fisiologia , Fotoperíodo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transdução de Sinais , Animais , Núcleo Arqueado do Hipotálamo/anatomia & histologia , Cricetinae , Feminino , Privação de Alimentos , Regulação Enzimológica da Expressão Gênica , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA