Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 15(723): eadd4897, 2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-37992152

RESUMO

Deficiency in the adipose-derived hormone leptin or leptin receptor signaling causes class 3 obesity in individuals with genetic loss-of-function mutations in leptin or its receptor LEPR and metabolic and liver disease in individuals with hypoleptinemia secondary to lipoatrophy such as in individuals with generalized lipodystrophy. Therapies that restore leptin-LEPR signaling may resolve these metabolic sequelae. We developed a fully human monoclonal antibody (mAb), REGN4461 (mibavademab), that activates the human LEPR in the absence or presence of leptin. In obese leptin knockout mice, REGN4461 normalized body weight, food intake, blood glucose, and insulin sensitivity. In a mouse model of generalized lipodystrophy, REGN4461 alleviated hyperphagia, hyperglycemia, insulin resistance, dyslipidemia, and hepatic steatosis. In a phase 1, randomized, double-blind, placebo-controlled two-part study, REGN4461 was well tolerated with an acceptable safety profile. Treatment of individuals with overweight or obesity with REGN4461 decreased body weight over 12 weeks in those with low circulating leptin concentrations (<8 ng/ml) but had no effect on body weight in individuals with higher baseline leptin. Furthermore, compassionate-use treatment of a single patient with atypical partial lipodystrophy and a history of undetectable leptin concentrations associated with neutralizing antibodies to metreleptin was associated with noteable improvements in circulating triglycerides and hepatic steatosis. Collectively, these translational data unveil an agonist LEPR mAb that may provide clinical benefit in disorders associated with relatively low leptin concentrations.


Assuntos
Resistência à Insulina , Lipodistrofia Generalizada Congênita , Animais , Camundongos , Humanos , Leptina/uso terapêutico , Ensaios de Uso Compassivo , Receptores para Leptina/metabolismo , Lipodistrofia Generalizada Congênita/tratamento farmacológico , Obesidade/tratamento farmacológico , Anticorpos/uso terapêutico , Peso Corporal
2.
Proc Natl Acad Sci U S A ; 120(32): e2309967120, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37523551

RESUMO

Body fat distribution is a heritable risk factor for cardiovascular and metabolic disease. In humans, rare Inhibin beta E (INHBE, activin E) loss-of-function variants are associated with a lower waist-to-hip ratio and protection from type 2 diabetes. Hepatic fatty acid sensing promotes INHBE expression during fasting and in obese individuals, yet it is unclear how the hepatokine activin E governs body shape and energy metabolism. Here, we uncover activin E as a regulator of adipose energy storage. By suppressing ß-agonist-induced lipolysis, activin E promotes fat accumulation and adipocyte hypertrophy and contributes to adipose dysfunction in mice. Mechanistically, we demonstrate that activin E elicits its effect on adipose tissue through ACVR1C, activating SMAD2/3 signaling and suppressing PPARG target genes. Conversely, loss of activin E or ACVR1C in mice increases fat utilization, lowers adiposity, and drives PPARG-regulated gene signatures indicative of healthy adipose function. Our studies identify activin E-ACVR1C as a metabolic rheostat promoting liver-adipose cross talk to restrain excessive fat breakdown and preserve fat mass during prolonged fasting, a mechanism that is maladaptive in obese individuals.


Assuntos
Diabetes Mellitus Tipo 2 , Lipólise , Humanos , Camundongos , Animais , Ativinas/metabolismo , Adiposidade/genética , Diabetes Mellitus Tipo 2/metabolismo , PPAR gama/metabolismo , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo
3.
Stem Cell Reports ; 18(1): 394-409, 2023 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-36525967

RESUMO

Rats were more frequently used than mice to model human disease before mouse embryonic stem cells (mESCs) revolutionized genetic engineering in mice. Rat ESCs (rESCs) were first reported over 10 years ago, yet they are not as frequently used as mESCs. CRISPR-based gene editing in zygotes is widely used in rats but is limited by the difficulty of inserting or replacing DNA sequences larger than about 10 kb. We report here the generation of germline-competent rESC lines from several rat strains. These rESC lines maintain their potential for germline transmission after serial targeting with bacterial artificial chromosome (BAC)-based targeting vectors, and CRISPR-Cas9 cutting can increase targeting efficiency. Using these methods, we have successfully replaced entire rat genes spanning up to 101 kb with the human ortholog.


Assuntos
Células-Tronco Embrionárias , Degeneração Retiniana , Humanos , Ratos , Animais , Camundongos , Edição de Genes , Engenharia Genética , Sistemas CRISPR-Cas/genética
4.
J Bone Miner Res ; 36(4): 739-756, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33249643

RESUMO

Osteogenesis imperfecta (OI) is a genetic connective tissue disorder characterized by compromised skeletal integrity, altered microarchitecture, and bone fragility. Current OI treatment strategies focus on bone antiresorptives and surgical intervention with limited effectiveness, and thus identifying alternative therapeutic options remains critical. Muscle is an important stimulus for bone formation. Myostatin, a TGF-ß superfamily myokine, acts through ActRIIB to negatively regulate muscle growth. Recent studies demonstrated the potential benefit of myostatin inhibition with the soluble ActRIIB fusion protein on skeletal properties, although various OI mouse models exhibited variable skeletal responses. The genetic and clinical heterogeneity associated with OI, the lack of specificity of the ActRIIB decoy molecule for myostatin alone, and adverse events in human clinical trials further the need to clarify myostatin's therapeutic potential and role in skeletal integrity. In this study, we determined musculoskeletal outcomes of genetic myostatin deficiency and postnatal pharmacological myostatin inhibition by a monoclonal anti-myostatin antibody (Regn647) in the G610C mouse, a model of mild-moderate type I/IV human OI. In the postnatal study, 5-week-old wild-type and +/G610C male and female littermates were treated with Regn647 or a control antibody for 11 weeks or for 7 weeks followed by a 4-week treatment holiday. Inhibition of myostatin, whether genetically or pharmacologically, increased muscle mass regardless of OI genotype, although to varying degrees. Genetic myostatin deficiency increased hindlimb muscle weights by 6.9% to 34.4%, whereas pharmacological inhibition increased them by 13.5% to 29.6%. Female +/mstn +/G610C (Dbl.Het) mice tended to have similar trabecular and cortical bone parameters as Wt showing reversal of +/G610C characteristics but with minimal effect of +/mstn occurring in male mice. Pharmacologic myostatin inhibition failed to improve skeletal bone properties of male or female +/G610C mice, although skeletal microarchitectural and biomechanical improvements were observed in male wild-type mice. Four-week treatment holiday did not alter skeletal outcomes. © 2020 American Society for Bone and Mineral Research (ASBMR).


Assuntos
Osteogênese Imperfeita , Animais , Osso e Ossos , Colágeno Tipo I , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Miostatina/genética , Osteogênese , Osteogênese Imperfeita/tratamento farmacológico , Osteogênese Imperfeita/genética
5.
Nat Commun ; 8: 15153, 2017 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-28452368

RESUMO

Growth and differentiation factor 8 (GDF8) is a TGF-ß superfamily member, and negative regulator of skeletal muscle mass. GDF8 inhibition results in prominent muscle growth in mice, but less impressive hypertrophy in primates, including man. Broad TGF-ß inhibition suggests another family member negatively regulates muscle mass, and its blockade enhances muscle growth seen with GDF8-specific inhibition. Here we show that activin A is the long-sought second negative muscle regulator. Activin A specific inhibition, on top of GDF8 inhibition, leads to pronounced muscle hypertrophy and force production in mice and monkeys. Inhibition of these two ligands mimics the hypertrophy seen with broad TGF-ß blockers, while avoiding the adverse effects due to inhibition of multiple family members. Altogether, we identify activin A as a second negative regulator of muscle mass, and suggest that inhibition of both ligands provides a preferred therapeutic approach, which maximizes the benefit:risk ratio for muscle diseases in man.


Assuntos
Ativinas/metabolismo , Hipertrofia/patologia , Hipotonia Muscular/patologia , Músculo Esquelético/crescimento & desenvolvimento , Miostatina/metabolismo , Receptores de Activinas Tipo II/metabolismo , Ativinas/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Índice de Massa Corporal , Dexametasona/farmacologia , Humanos , Contração Isométrica/fisiologia , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Músculo Esquelético/fisiologia , Miostatina/antagonistas & inibidores , Ratos
6.
Skelet Muscle ; 5: 34, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26457176

RESUMO

BACKGROUND: Loss of skeletal muscle mass and function in humans is associated with significant morbidity and mortality. The role of myostatin as a key negative regulator of skeletal muscle mass and function has supported the concept that inactivation of myostatin could be a useful approach for treating muscle wasting diseases. METHODS: We generated a myostatin monoclonal blocking antibody (REGN1033) and characterized its effects in vitro using surface plasmon resonance biacore and cell-based Smad2/3 signaling assays. REGN1033 was tested in mice for the ability to induce skeletal muscle hypertrophy and prevent atrophy induced by immobilization, hindlimb suspension, or dexamethasone. The effect of REGN1033 on exercise training was tested in aged mice. Messenger RNA sequencing, immunohistochemistry, and ex vivo force measurements were performed on skeletal muscle samples from REGN1033-treated mice. RESULTS: The human monoclonal antibody REGN1033 is a specific and potent myostatin antagonist. Chronic treatment of mice with REGN1033 increased muscle fiber size, muscle mass, and force production. REGN1033 prevented the loss of muscle mass induced by immobilization, glucocorticoid treatment, or hindlimb unweighting and increased the gain of muscle mass during recovery from pre-existing atrophy. In aged mice, REGN1033 increased muscle mass and strength and improved physical performance during treadmill exercise. CONCLUSIONS: We show that specific myostatin antagonism with the human antibody REGN1033 enhanced muscle mass and function in young and aged mice and had beneficial effects in models of skeletal muscle atrophy.

7.
Endocrinology ; 156(12): 4502-10, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26406932

RESUMO

Secreted frizzled-related protein 4 (SFRP4) is an extracellular regulator of the wingless-type mouse mammary tumor virus integration site family (WNT) pathway. SFRP4 has been implicated in adipocyte dysfunction, obesity, insulin resistance, and impaired insulin secretion in patients with type 2 diabetes. However, the exact role of SFRP4 in regulating whole-body metabolism and glucose homeostasis is unknown. We show here that male Sfrp4(-/-) mice have increased spine length and gain more weight when fed a high-fat diet. The body composition and body mass per spine length of diet-induced obese Sfrp4(-/-) mice is similar to wild-type littermates, suggesting that the increase in body weight can be accounted for by their longer body size. The diet-induced obese Sfrp4(-/-) mice have reduced energy expenditure, food intake, and bone mineral density. Sfrp4(-/-) mice have normal glucose and insulin tolerance and ß-cell mass. Diet-induced obese Sfrp4(-/-) and control mice show similar impairments of glucose tolerance and a 5-fold compensatory expansion of their ß-cell mass. In summary, our data suggest that loss of SFRP4 alters body length and bone mineral density as well as energy expenditure and food intake. However, SFRP4 does not control glucose homeostasis and ß-cell mass in mice.


Assuntos
Tamanho Corporal/genética , Densidade Óssea/genética , Dieta Hiperlipídica , Ingestão de Alimentos/genética , Metabolismo Energético/genética , Células Secretoras de Insulina/metabolismo , Obesidade , Proteínas Proto-Oncogênicas/genética , Animais , Glicemia/metabolismo , Composição Corporal/genética , Comportamento Alimentar , Técnicas de Introdução de Genes , Teste de Tolerância a Glucose , Células HEK293 , Homeostase/genética , Humanos , Insulina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Via de Sinalização Wnt , Microtomografia por Raio-X
8.
Diabetes ; 64(10): 3564-72, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26130763

RESUMO

The objective of this study was to determine whether the sodium-glucose transporter SGLT1 in the ventromedial hypothalamus (VMH) plays a role in glucose sensing and in regulating the counterregulatory response to hypoglycemia, and if so, whether knockdown of in the VMH can improve counterregulatory responses to hypoglycemia in diabetic rats or rats exposed to recurrent bouts of hypoglycemia (RH). Normal Sprague-Dawley rats as well as RH or streptozotocin (STZ)-diabetic rats received bilateral VMH microinjections of an adenoassociated viral vector containing either the SGLT1 short hairpin RNA (shRNA) or a scrambled RNA sequence. Subsequently, these rats underwent a hypoglycemic clamp to assess hormone responses. In a subgroup of rats, glucose kinetics was determined using tritiated glucose. The shRNA reduced VMH SGLT1 expression by 53% in nondiabetic rats, and this augmented glucagon and epinephrine responses and hepatic glucose production during hypoglycemia. Similarly, SGLT1 knockdown improved the glucagon and epinephrine responses in RH rats and restored the impaired epinephrine response to hypoglycemia in STZ-diabetic animals. These findings suggest that SGLT1 in the VMH plays a significant role in the detection and activation of counterregulatory responses to hypoglycemia. Inhibition of SGLT1 may offer a potential therapeutic target to diminish the risk of hypoglycemia in diabetes.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Regulação da Expressão Gênica/fisiologia , Hipoglicemia/metabolismo , RNA Mensageiro/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Glicemia , Masculino , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Transportador 1 de Glucose-Sódio/genética
9.
Proc Natl Acad Sci U S A ; 112(6): 1845-9, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25624481

RESUMO

G protein-coupled receptor 17 (GPR17) was recently reported to be a Foxo1 target in agouti-related peptide (AGRP) neurons. Intracerebroventricular injection of GPR17 agonists induced food intake, whereas administration of an antagonist to the receptor reduced feeding. These data lead to the conclusion that pharmacological modulation of GPR17 has therapeutic potential to treat obesity. Here we report that mice deficient in Gpr17 (Gpr17(-/-)) have similar food intake and body weight compared with their wild-type littermates. Gpr17(-/-) mice have normal hypothalamic Agrp mRNA expression, AGRP plasma levels, and metabolic rate. GPR17 deficiency in mice did not affect glucose homeostasis or prevent fat-induced insulin resistance. These data do not support a role for GPR17 in the control of food intake, body weight, or glycemic control.


Assuntos
Ingestão de Alimentos/genética , Glucose/metabolismo , Proteínas do Tecido Nervoso/genética , Receptores Acoplados a Proteínas G/genética , Proteína Relacionada com Agouti/metabolismo , Análise de Variância , Animais , Sequência de Bases , Composição Corporal/efeitos dos fármacos , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Neurônios/metabolismo , Análise de Sequência de RNA , Fatores de Tempo , Microtomografia por Raio-X
10.
J Child Neurol ; 28(8): 1009-14, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23680948

RESUMO

Accumulating evidence suggests that low-carbohydrate, high-fat diets are safe and effective to reduce glycemia in diabetic patients without producing significant cardiovascular risks. Most of these studies have been carried out specifically restricting carbohydrates, which tends to lead to increased protein intake, thus reducing the ketosis. However, diets that limit protein as well as carbohydrates, entailing a composition very high in fat, appear even more effective to reduce glucose and whole-body glucose metabolism in humans. In animal models, low-carbohydrate, high-protein diets do not produce ketosis or reduce glycemia but rather cause obesity. However, limiting both protein and carbohydrates as in a classic ketogenic diet remarkably reduces blood glucose in animal models of type 1 and type 2 diabetes and reverses diabetic nephropathy. Future studies should assess if ketogenic diets would be effective to reverse diabetic complications in humans.


Assuntos
Complicações do Diabetes/dietoterapia , Dieta Cetogênica/métodos , Obesidade/dietoterapia , Animais , Glicemia/metabolismo , Modelos Animais de Doenças , Metabolismo Energético , Humanos , Obesidade/sangue
11.
Am J Physiol Endocrinol Metab ; 302(8): E987-91, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22318949

RESUMO

To discover hypothalamic genes that might play a role in regulating energy balance, we carried out a microarray screen for genes induced by a 48-h fast in male C57Bl/6J mouse hypothalamus. One such gene was Fkbp51 (FK506 binding protein 5; Locus NP_034350). The product of this gene is of interest because it blocks glucocorticoid action, suggesting that fasting-induced elevation of this gene in the hypothalamus may reduce glucocorticoid negative feedback, leading to elevated glucocorticoid levels, thus promoting obese phenotypes. Subsequent analysis demonstrated that a 48-h fast induces Fkbp51 in ventromedial, paraventricular, and arcuate hypothalamic nuclei of mice and rats. To assess if hypothalamic Fkbp51 promotes obesity, the gene was transferred to the hypothalamus via an adeno-associated virus vector. Within 2 wk following Fkbp51 overexpression, mice on a high-fat diet exhibited elevated body weight, without hyperphagia, relative to mice receiving the control mCherry vector. Body weight remained elevated for more than 8 wk and was associated with elevated corticosterone and impaired glucose tolerance. These studies suggest that elevated hypothalamic Fkbp51 promotes obese phenotypes.


Assuntos
Jejum/metabolismo , Hipotálamo/metabolismo , Obesidade/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Regulação para Cima , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Corticosterona/sangue , Ingestão de Energia , Perfilação da Expressão Gênica , Intolerância à Glucose/sangue , Intolerância à Glucose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/sangue , Análise de Sequência com Séries de Oligonucleotídeos , Núcleo Hipotalâmico Paraventricular/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas de Ligação a Tacrolimo/antagonistas & inibidores , Proteínas de Ligação a Tacrolimo/genética , Núcleo Hipotalâmico Ventromedial/metabolismo , Aumento de Peso
12.
PLoS One ; 6(4): e18604, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21533091

RESUMO

Intensive insulin therapy and protein restriction delay the development of nephropathy in a variety of conditions, but few interventions are known to reverse nephropathy. Having recently observed that the ketone 3-beta-hydroxybutyric acid (3-OHB) reduces molecular responses to glucose, we hypothesized that a ketogenic diet, which produces prolonged elevation of 3-OHB, may reverse pathological processes caused by diabetes. To address this hypothesis, we assessed if prolonged maintenance on a ketogenic diet would reverse nephropathy produced by diabetes. In mouse models for both Type 1 (Akita) and Type 2 (db/db) diabetes, diabetic nephropathy (as indicated by albuminuria) was allowed to develop, then half the mice were switched to a ketogenic diet. After 8 weeks on the diet, mice were sacrificed to assess gene expression and histology. Diabetic nephropathy, as indicated by albumin/creatinine ratios as well as expression of stress-induced genes, was completely reversed by 2 months maintenance on a ketogenic diet. However, histological evidence of nephropathy was only partly reversed. These studies demonstrate that diabetic nephropathy can be reversed by a relatively simple dietary intervention. Whether reduced glucose metabolism mediates the protective effects of the ketogenic diet remains to be determined.


Assuntos
Nefropatias Diabéticas/dietoterapia , Dieta Cetogênica , Animais , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
13.
Diabetes ; 60(1): 39-46, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20811039

RESUMO

OBJECTIVE: Hypoglycemia-associated autonomic failure (HAAF) constitutes one of the main clinical obstacles to optimum treatment of type 1 diabetes. Neurons in the ventromedial hypothalamus are thought to mediate counterregulatory responses to hypoglycemia. We have previously hypothesized that hypoglycemia-induced hypothalamic angiotensin might contribute to HAAF, suggesting that the angiotensin blocker valsartan might prevent HAAF. On the other hand, clinical studies have demonstrated that the opioid receptor blocker naloxone ameliorates HAAF. The goal of this study was to generate novel hypothalamic markers of hypoglycemia and use them to assess mechanisms mediating HAAF and its reversal. RESEARCH DESIGN AND METHODS: Quantitative PCR was used to validate a novel panel of hypothalamic genes regulated by hypoglycemia. Mice were exposed to one or five episodes of insulin-induced hypoglycemia, with or without concurrent exposure to valsartan or naloxone. Corticosterone, glucagon, epinephrine, and hypothalamic gene expression were assessed after the final episode of hypoglycemia. RESULTS: A subset of hypothalamic genes regulated acutely by hypoglycemia failed to respond after repetitive hypoglycemia. Responsiveness of a subset of these genes was preserved by naloxone but not valsartan. Notably, hypothalamic expression of four genes, including pyruvate dehydrogenase kinase 4 and glycerol 3-phosphate dehydrogenase 1, was acutely induced by a single episode of hypoglycemia, but not after antecedent hypoglycemia; naloxone treatment prevented this failure. Similarly, carnitine palmitoyltransferase-1 was inhibited after repetitive hypoglycemia, and this inhibition was prevented by naloxone. Repetitive hypoglycemia also caused a loss of hypoglycemia-induced elevation of glucocorticoid secretion, a failure prevented by naloxone but not valsartan. CONCLUSIONS: Based on these observations we speculate that acute hypoglycemia induces reprogramming of hypothalamic metabolism away from glycolysis toward ß-oxidation, HAAF is associated with a reversal of this reprogramming, and naloxone preserves some responses to hypoglycemia by preventing this reversal.


Assuntos
Hipoglicemia/complicações , Naloxona/farmacologia , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Animais , Anti-Hipertensivos/farmacologia , Proteínas Reguladoras de Apoptose , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Proteínas de Transporte , Inibidor de Quinase Dependente de Ciclina p21/genética , Transportador de Glucose Tipo 2/genética , Glicerolfosfato Desidrogenase/genética , Homeostase/efeitos dos fármacos , Hipoglicemia/induzido quimicamente , Hipoglicemia/etiologia , Hipoglicemia/genética , Insulina/farmacologia , Proteínas de Membrana/genética , Camundongos , Naloxona/uso terapêutico , Antagonistas de Entorpecentes/farmacologia , Perilipina-4 , Proteínas Serina-Treonina Quinases/genética , Insuficiência Autonômica Pura/etiologia , Insuficiência Autonômica Pura/prevenção & controle , Piruvato Desidrogenase Quinase de Transferência de Acetil , Proteínas Repressoras , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tetrazóis/farmacologia , Fatores de Transcrição/genética , Valina/análogos & derivados , Valina/farmacologia , Valsartana
14.
Endocrinology ; 151(11): 5206-17, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20881243

RESUMO

Nutrient-sensitive hypothalamic neurons regulate energy balance and glucose homeostasis, but the molecular mechanisms mediating hypothalamic responses to nutritional state remain incompletely characterized. To address these mechanisms, the present studies used quantitative PCR to characterize the expression of a panel of genes the hypothalamic expression by nutritional status of which had been suggested by DNA microarray studies. Although these genes regulate a variety of function, the most prominent set regulate intermediary metabolism, and the overall pattern clearly indicated that a 48-h fast produced a metabolic reprogramming away from glucose metabolism and toward the utilization of alternative fuels, particularly lipid metabolism. This general reprogramming of intermediary metabolism by fasting was observed both in cortex and hypothalamus but most prominently in hypothalamus. The effect of fasting on the expression of these genes may be mediated by reduction in plasma glucose or glucose metabolism, rather than leptin, because they were generally recapitulated by hypoglycemia even in the presence of elevated insulin and in vitro by low glucose but were not recapitulated in ob/ob mice. These studies suggest that fasting reduces glucose metabolism and thus minimizes the production of hypothalamic malonyl-coenzyme A. However, because the reprogramming of glucose metabolism by fasting was also observed in cortex, this apparent substrate competition may mediate more general responses to nutritional deprivation, including those responsible for the protective effects of dietary restriction. The present studies also provide a large panel of novel glucose-regulated genes that can be used as markers of glucose action to address mechanisms mediating hypothalamic responses to nutritional state.


Assuntos
Jejum/metabolismo , Glicólise/fisiologia , Hipotálamo/metabolismo , Metabolismo dos Lipídeos/fisiologia , Neurônios/metabolismo , Estado Nutricional/fisiologia , Análise de Variância , Animais , Córtex Cerebral/metabolismo , Expressão Gênica , Insulina/metabolismo , Leptina/metabolismo , Masculino , Camundongos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Endocrinology ; 148(4): 1928-32, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17218412

RESUMO

Because appetite, hypothalamic gene expression, reproductive function, and adrenal function are highly sensitive to acute changes in plasma glucose levels, it has been hypothesized hypothalamic neurons sensitive to glucose play a role in regulating these functions. To assess this hypothesis, we examined these neuronendocrine functions in mice in which the glucokinase gene, which plays an essential role in neuroendocrine glucose sensing, has been ablated. Haploinsufficiency in heterozygous glucokinase knockout mice produced effects similar to those produced by hypoglycemia: impaired reproductive function, elevated plasma corticosterone, increased food intake, and hypothalamic gene expression similar to that observed in fasted or leptin-deficient obese mice (increased hypothalamic neuropeptide Y mRNA and reduced hypothalamic proopiomelanocortin mRNA). Plasma glucose was elevated 2-fold in glucokinase knockout mice, consistent with a maturity-onset diabetes of the young phenotype, but plasma insulin and leptin levels were normal. These data support the hypothesis that glucokinase plays a key role in the neuroendocrine regulation of metabolic economy.


Assuntos
Ingestão de Alimentos/genética , Glucocorticoides/metabolismo , Glucoquinase/fisiologia , Hipotálamo/metabolismo , Reprodução/genética , Animais , Feminino , Fertilidade/genética , Regulação da Expressão Gênica , Glucoquinase/genética , Hiperglicemia/genética , Insulina/sangue , Insulina/genética , Leptina/sangue , Masculino , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas
16.
Appetite ; 48(2): 135-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17141367

RESUMO

High-fat diets produce obesity in part because, per calorie, glucose produces greater post-prandial thermogenesis than lipids, an effect probably mediated by glucose-sensing neurons. A very low-carbohydrate/high-fat/high-protein Atkins-type diet produces obesity but is marginally ketogenic in mice. In contrast, high-sucrose/low-fat diets, and very low-carbohydrate/high-fat/low-protein (anti-epileptic) ketogenic diets reverse diet-induced obesity independent of caloric intake. We propose that a non-ketogenic high-fat diet reduces glucose metabolism and signaling in glucose-sensing neurons, thereby reducing post-prandial thermogenesis, and that a ketogenic high-fat diet does not reduce glucose signaling, thereby preventing and/or reversing obesity.


Assuntos
Dieta com Restrição de Carboidratos , Dieta Redutora , Metabolismo Energético/fisiologia , Obesidade/dietoterapia , Obesidade/etiologia , Glicemia/metabolismo , Restrição Calórica , Dieta com Restrição de Carboidratos/efeitos adversos , Carboidratos da Dieta/administração & dosagem , Ingestão de Energia/fisiologia , Humanos , Obesidade/epidemiologia , Obesidade/metabolismo , Resultado do Tratamento
17.
Interdiscip Top Gerontol ; 35: 39-68, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17063032

RESUMO

Elevated blood glucose associated with diabetes produces progressive and apparently irreversible damage to many cell types. Conversely, reduction of glucose extends life span in yeast, and dietary restriction reduces blood glucose. Therefore it has been hypothesized that cumulative toxic effects of glucose drive at least some aspects of the aging process and, conversely, that protective effects of dietary restriction are mediated by a reduction in exposure to glucose. The mechanisms mediating cumulative toxic effects of glucose are suggested by two general principles of metabolic processes, illustrated by the lac operon but also observed with glucose-induced gene expression. First, metabolites induce the machinery of their own metabolism. Second, induction of gene expression by metabolites can entail a form of molecular memory called hysteresis. When applied to glucose-regulated gene expression, these two principles suggest a mechanism whereby repetitive exposure to postprandial excursions of glucose leads to an age-related increase in glycolytic capacity (and reduction in beta-oxidation of free fatty acids), which in turn leads to an increased generation of oxidative damage and a decreased capacity to respond to oxidative damage, independent of metabolic rate. According to this mechanism, dietary restriction increases life span and reduces pathology by reducing exposure to glucose and therefore delaying the development of glucose-induced glycolytic capacity.


Assuntos
Envelhecimento/metabolismo , Glicemia/metabolismo , Restrição Calórica , Metabolismo Energético/genética , Privação de Alimentos , Óperon Lac , Longevidade/genética , Doenças Metabólicas/genética , Envelhecimento/fisiologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Glicemia/genética , Ingestão de Energia , Metabolismo Energético/fisiologia , Glicólise , Humanos , Longevidade/fisiologia , Doenças Metabólicas/fisiopatologia , Estresse Oxidativo
18.
Physiol Behav ; 85(1): 3-23, 2005 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-15924903

RESUMO

Since nutrition-sensitive feedback signals normally act to maintain relatively stable levels of both available and stored nutritional resources, failure in one or more of these feedback signals could plausibly lead to obese phenotypes. The glucostatic hypothesis in its original form posited that glucose serves as a physiological satiety factor (in the sense that post-prandial increases in plasma glucose cause meal termination), but in this form the hypothesis has been difficult to prove, and, especially since the discovery of leptin, the glucostatic hypothesis has largely been abandoned. Nevertheless, reduction of plasma glucose levels or glucose signaling produces a profile of neuroendocrine responses similar to those produced by leptin deficiency. Since leptin is not a physiological satiety factor (because it does not increase before meal termination), yet leptin deficiency causes obesity, we suggest that the glucostatic hypothesis be re-formulated without reference to satiety (i.e., short-term effects on food intake). Instead we argue that like leptin signaling, glucose signaling regulates long-term energy balance, in part by regulating metabolic rate but also by chronically regulating food intake. We further speculate that high-fat diets produce obesity in part because carbohydrates are, per calorie, more effective than lipids to reduce food intake and increase metabolic rate. In support of this glucoadipostatic hypothesis, the 5 present review examines evidence that obesity and the metabolic syndrome may be due to reduction in neuroendocrine sensitivity to glucose leading to increased metabolic efficiency.


Assuntos
Metabolismo Energético , Glucose/metabolismo , Obesidade/etiologia , Transdução de Sinais , Adenilato Quinase/metabolismo , Animais , Glicemia/metabolismo , Dieta , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Insulina/metabolismo , Leptina/metabolismo , Obesidade/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Pró-Opiomelanocortina/fisiologia , Resposta de Saciedade/fisiologia , Termogênese/fisiologia
19.
Diabetes ; 54(4): 952-8, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15793232

RESUMO

The robust neuroendocrine counterregulatory responses induced by hypoglycemia protect the brain by restoring plasma glucose, but little is known about molecular responses to hypoglycemia that may also be neuroprotective. To clarify these mechanisms, we examined gene expression in hypothalamus, cortex, and liver 3 h after induction of mild hypoglycemia by a single injection of insulin, using cDNA microarray analysis and quantitative real-time PCR. Real-time PCR corroborated the induction of six genes (angiotensinogen, GLUT-1, inhibitor of kappaB, inhibitor of DNA binding 1 [ID-1], Ubp41, and mitogen-activated protein kinase phosphatase-1 [MKP-1]) by insulin-induced hypoglycemia in the hypothalamus: five of these six genes in cortex and three (GLUT-1, angiotensinogen, and MKP-1) in liver. The induction was due to hypoglycemia and not hyperinsulinemia, since fasting (characterized by low insulin and glucose) also induced these genes. Four of these genes (angiotensinogen, GLUT-1, ID-1, and MKP-1) have been implicated in enhancement of glucose availability, which could plausibly serve a neuroprotective role during acute hypoglycemia but, if persistent, could also cause glucose-sensing mechanisms to overestimate plasma glucose levels, potentially causing hypoglycemia-induced counterregulatory failure. Although using cDNA microarrays with more genes, or microdissection, would presumably reveal further responses to hypoglycemia, these hypoglycemia-induced genes represent useful markers to assess molecular mechanisms mediating cellular responses to hypoglycemia.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hipoglicemia/induzido quimicamente , Hipoglicemia/metabolismo , Insulina/farmacologia , Fígado/metabolismo , Animais , Regulação para Baixo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Regulação para Cima
20.
Neurochem Res ; 29(6): 1093-103, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15176466

RESUMO

DNA microarray analysis has been used to investigate relative changes in the level of gene expression in the CNS, including changes that are associated with disease, injury, psychiatric disorders, drug exposure or withdrawal, and memory formation. We have used oligonucleotide microarrays to identify hypothalamic genes that respond to nutritional manipulation. In addition to commonly used microarray analysis based on criteria such as fold-regulation, we have also found that simply carrying out multiple t tests then sorting by P value constitutes a highly reliable method to detect true regulation, as assessed by real-time polymerase chain reaction (PCR), even for relatively low abundance genes or relatively low magnitude of regulation. Such analyses directly suggested novel mechanisms that mediate effects of nutritional state on neuroendocrine function and are being used to identify regulated gene products that may elucidate the metabolic pathology of obese ob/ob, lean Vgf-/Vgf-, and other models with profound metabolic impairments.


Assuntos
Regulação da Expressão Gênica/genética , Hipotálamo/fisiologia , Metabolismo/genética , Fenômenos Fisiológicos da Nutrição , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Fenômenos Fisiológicos da Nutrição Animal , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...