Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Physiol Rev ; 95(1): 47-82, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25540138

RESUMO

Obesity and type 2 diabetes mellitus (T2DM) often occur together and affect a growing number of individuals in both the developed and developing worlds. Both are associated with a number of other serious illnesses that lead to increased rates of mortality. There is likely a polygenic mode of inheritance underlying both disorders, but it has become increasingly clear that the pre- and postnatal environments play critical roles in pushing predisposed individuals over the edge into a disease state. This review focuses on the many genetic and environmental variables that interact to cause predisposed individuals to become obese and diabetic. The brain and its interactions with the external and internal environment are a major focus given the prominent role these interactions play in the regulation of energy and glucose homeostasis in health and disease.


Assuntos
Metabolismo Energético/fisiologia , Interação Gene-Ambiente , Glucose/metabolismo , Homeostase/fisiologia , Obesidade/genética , Animais , Diabetes Mellitus/etiologia , Metabolismo Energético/genética , Meio Ambiente , Predisposição Genética para Doença , Humanos , Plasticidade Neuronal/fisiologia , Obesidade/etiologia , Obesidade/metabolismo
2.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1186-92, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27122369

RESUMO

Obesity and Type 2 diabetes are major worldwide public health issues today. A relationship between total fat intake and obesity has been found. In addition, the mechanisms of long-term and excessive high-fat diet (HFD) intake in the development of obesity still need to be elucidated. The ventromedial hypothalamus (VMH) is a major site involved in the regulation of glucose and energy homeostasis where "metabolic sensing neurons" integrate metabolic signals from the periphery. Among these signals, fatty acids (FA) modulate the activity of VMH neurons using the FA translocator/CD36, which plays a critical role in the regulation of energy and glucose homeostasis. During low-fat diet (LFD) intake, FA are oxidized by VMH astrocytes to fuel their ongoing metabolic needs. However, HFD intake causes VMH astrocytes to use FA to generate ketone bodies. We postulate that these astrocyte-derived ketone bodies are exported to neurons where they produce excess ATP and reactive oxygen species, which override CD36-mediated FA sensing and act as a signal to decrease short-term food intake. On a HFD, VMH astrocyte-produced ketones reduce elevated caloric intake to LFD levels after 3 days in rats genetically predisposed to resist (DR) diet-induced obesity (DIO), but not leptin-resistant DIO rats. This suggests that, while VMH ketone production on a HFD can contribute to protection from obesity, the inherent leptin resistance overrides this inhibitory action of ketone bodies on food intake. Thus, astrocytes and neurons form a tight metabolic unit that is able to monitor circulating nutrients to alter food intake and energy homeostasis.


Assuntos
Apetite/fisiologia , Astrócitos/metabolismo , Ingestão de Alimentos/fisiologia , Ácidos Graxos/metabolismo , Corpos Cetônicos/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Regulação do Apetite/fisiologia , Dieta Hiperlipídica , Modelos Biológicos , Neurônios/metabolismo
3.
Am J Physiol Regul Integr Comp Physiol ; 310(1): R66-73, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26511522

RESUMO

While the neural control of glucoregulatory responses to insulin-induced hypoglycemia is beginning to be elucidated, brain sites responsible for behavioral responses to hypoglycemia are relatively poorly understood. To help elucidate central control mechanisms associated with hypoglycemia unawareness, we first evaluated the effect of recurrent hypoglycemia on a simple behavioral measure, the robust feeding response to hypoglycemia, in rats. First, food intake was significantly, and similarly, increased above baseline saline-induced intake (1.1 ± 0.2 g; n = 8) in rats experiencing a first (4.4 ± 0.3; n = 8) or third daily episode of recurrent insulin-induced hypoglycemia (IIH, 3.7 ± 0.3 g; n = 9; P < 0.05). Because food intake was not impaired as a result of prior IIH, we next developed an alternative animal model of hypoglycemia-induced behavioral arousal using a conditioned place preference (CPP) model. We found that hypoglycemia severely blunted previously acquired CPP in rats and that recurrent hypoglycemia prevented this blunting. Pretreatment with a brain penetrant, selective orexin receptor-1 antagonist, SB-334867A, blocked hypoglycemia-induced blunting of CPP. Recurrently hypoglycemic rats also showed decreased preproorexin expression in the perifornical hypothalamus (50%) but not in the adjacent lateral hypothalamus. Pretreatment with sertraline, previously shown to prevent hypoglycemia-associated glucoregulatory failure, did not prevent blunting of hypoglycemia-induced CPP prevention by recurrent hypoglycemia. This work describes the first behavioral model of hypoglycemia unawareness and suggests a role for orexin neurons in mediating behavioral responses to hypoglycemia.


Assuntos
Comportamento Animal , Glicemia/metabolismo , Encéfalo/metabolismo , Condicionamento Psicológico , Comportamento Alimentar , Hipoglicemia/metabolismo , Orexinas/metabolismo , Transdução de Sinais , Animais , Nível de Alerta , Comportamento Animal/efeitos dos fármacos , Benzoxazóis/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Condicionamento Psicológico/efeitos dos fármacos , Modelos Animais de Doenças , Comportamento Alimentar/efeitos dos fármacos , Hipoglicemia/fisiopatologia , Hipoglicemia/psicologia , Masculino , Naftiridinas , Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina/efeitos dos fármacos , Receptores de Orexina/metabolismo , Ratos Sprague-Dawley , Recompensa , Sertralina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Ureia/análogos & derivados , Ureia/farmacologia
4.
Am J Physiol Regul Integr Comp Physiol ; 311(4): R764-R770, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27534878

RESUMO

Rats selectively bred to develop diet-induced obesity (DIO) have an early onset reduction in the sensitivity of their ventromedial hypothalamic nucleus (VMN) neurons to leptin compared with diet-resistant (DR) rats. This reduced sensitivity includes decreased leptin receptor (Lepr-b) mRNA expression, leptin receptor binding, leptin-induced phosphorylation of STAT3 (pSTAT3), and impaired leptin excitation (LepE) of VMN neurons. When administered exogenously, the pancreatic peptide, amylin, acts synergistically to reduce food intake and body weight in obese, leptin-resistant DIO rats by increasing VMN leptin signaling, likely by stimulation of microglia IL-6, which acts on its receptor to increase leptin-induced pSTAT3. Here, we demonstrate that incubation of cultured VMN neurons of outbred rats with IL-6 increases their leptin sensitivity. Control, dissociated DIO VMN neurons express 66% less Lepr-b and 75% less Bardet Biedl Syndrome-6 (BBS6) mRNA and have reduced leptin-induced activation of LepE neurons compared with DR neurons. Incubation for 4 days with IL-6 increased DIO neuron Lepr-b expression by 77% and BBS6 by 290% and corrected their defective leptin activation of LepE neurons to DR levels. Since BBS6 enhances trafficking of Lepr-b to the cell membrane, the increases in Lepr-b and BBS6 expression appear to account for correction of the reduced leptin excitation of DIO LepE neurons to that of control DR rats. These data support prior findings suggesting that IL-6 mediates the leptin-sensitizing effects of amylin on VMN neurons and that the inherent leptin resistance of DIO rats can be effectively reversed at a cellular level by IL-6.


Assuntos
Interleucina-6/imunologia , Leptina/imunologia , Neurônios/metabolismo , Obesidade/imunologia , Receptores para Leptina/metabolismo , Núcleo Hipotalâmico Ventromedial/imunologia , Animais , Células Cultivadas , Gorduras na Dieta , Masculino , Obesidade/induzido quimicamente , Ratos , Ratos Sprague-Dawley
5.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1032-R1044, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27629888

RESUMO

Selectively bred diet-induced obese (DIO) rats become obese on a high-fat diet and are leptin resistant before becoming obese. Compared with diet-resistant (DR) neonates, DIO neonates have impaired leptin-dependent arcuate (ARC) neuropeptide Y/agouti-related peptide (NPY/AgRP) and α-melanocyte-stimulating hormone (α-MSH; from proopiomelanocortin (POMC) neurons) axon outgrowth to the paraventricular nucleus (PVN). Using phosphorylation of STAT3 (pSTAT3) as a surrogate, we show that reduced DIO ARC leptin signaling develops by postnatal day 7 (P7) and is reduced within POMC but not NPY/AgRP neurons. Since amylin increases leptin signaling in adult rats, we treated DIO neonates with amylin during postnatal hypothalamic development and assessed leptin signaling, leptin-dependent ARC-PVN pathway development, and metabolic changes. DIO neonates treated with amylin from P0-6 and from P0-16 increased ARC leptin signaling and both AgRP and α-MSH ARC-PVN pathway development, but increased only POMC neuron number. Despite ARC-PVN pathway correction, P0-16 amylin-induced reductions in body weight did not persist beyond treatment cessation. Since amylin enhances adult DIO ARC signaling via an IL-6-dependent mechanism, we assessed ARC-PVN pathway competency in IL-6 knockout mice and found that the AgRP, but not the α-MSH, ARC-PVN pathway was reduced. These results suggest that both leptin and amylin are important neurotrophic factors for the postnatal development of the ARC-PVN pathway. Amylin might act as a direct neurotrophic factor in DIO rats to enhance both the number of POMC neurons and their α-MSH ARC-PVN pathway development. This suggests important and selective roles for amylin during ARC hypothalamic development.


Assuntos
Hipotálamo/fisiopatologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/administração & dosagem , Leptina/metabolismo , Obesidade/tratamento farmacológico , Obesidade/fisiopatologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/patologia , Núcleo Arqueado do Hipotálamo/fisiopatologia , Peso Corporal/efeitos dos fármacos , Dieta Hiperlipídica , Gorduras na Dieta , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/farmacologia , Masculino , Neurogênese/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Cuidado Pós-Natal , Ratos , Resultado do Tratamento
6.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1045-R1059, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27654396

RESUMO

Maternal overnutrition or associated complications putatively mediate the obesogenic effects of perinatal high-fat diet on developing offspring. Here, we tested the hypothesis that a Western diet developmental environment increases adiposity not only in male offspring from obesity-prone (DIO) mothers, but also in those from obesity-resistant (DR) dams, implicating a deleterious role for the Western diet per se. Selectively bred DIO and DR female rats were fed chow (17% kcal fat) or Western diet (32%) for 54 days before mating and, thereafter, through weaning. As intended, despite chow-like caloric intake, Western diet increased prepregnancy weight gain and circulating leptin levels in DIO, but not DR, dams. Yet, in both genotypes, maternal Western diet increased the weight and adiposity of preweanlings, as early as in DR offspring, and increased plasma leptin, insulin, and adiponectin of weanlings. Although body weight normalized with chow feeding during adolescence, young adult Western diet offspring subsequently showed decreased energy expenditure and, in DR offspring, decreased lipid utilization as a fuel substrate. By mid-adulthood, maternal Western diet DR offspring ate more chow, weighed more, and were fatter than controls. Thus, maternal Western diet covertly programmed increased adiposity in childhood and adulthood, disrupted relations of energy regulatory hormones with body fat, and decreased energy expenditure in offspring of lean, genetically obesity-resistant mothers. Maternal Western diet exposure alone, without maternal obesity or overnutrition, can promote offspring weight gain.


Assuntos
Dieta Ocidental , Resistência à Doença/fisiologia , Hormônios/sangue , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Obesidade/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Adiposidade/fisiologia , Animais , Animais não Endogâmicos , Biomarcadores/sangue , Ingestão de Energia , Feminino , Masculino , Obesidade/sangue , Obesidade/diagnóstico , Gravidez , Ratos , Ratos Sprague-Dawley , Fatores de Risco
7.
Am J Physiol Regul Integr Comp Physiol ; 310(4): R355-65, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26676252

RESUMO

Amylin enhances arcuate (ARC) and ventromedial (VMN) hypothalamic nuclei leptin signaling and synergistically reduces food intake and body weight in selectively bred diet-induced obese (DIO) rats. Since DIO (125)I-amylin dorsomedial nucleus-dorsomedial VMN binding was reduced, we postulated that this contributed to DIO ventromedial hypothalamus (VMH) leptin resistance, and that impairing VMH (ARC + VMN) calcitonin receptor (CTR)-mediated signaling by injecting adeno-associated virus (AAV) expressing a short hairpin portion of the CTR mRNA would predispose diet-resistant (DR) rats to obesity on high-fat (45%) diet (HFD). Depleting VMH CTR by 80-90% in 4-wk-old male DR rats reduced their ARC and VMN (125)I-labeled leptin binding by 57 and 51%, respectively, and VMN leptin-induced phospho-signal transducer and activator of transcription 3-positive neurons by 59% vs. AAV control rats. After 6 wk on chow, VMH CTR-depleted DR rats ate and gained the equivalent amount of food and weight but had 18% heavier fat pads (relative to carcass weight), 144% higher leptin levels, and were insulin resistant compared with control AAV DR rats. After 6 wk more on HFD, VMH CTR-depleted DR rats ate the same amount but gained 28% more weight, had 60% more carcass fat, 254% higher leptin levels, and 132% higher insulin areas under the curve during an oral glucose tolerance test than control DR rats. Therefore, impairing endogenous VMH CTR-mediated signaling reduced leptin signaling and caused DR rats to become more obese and insulin resistant, both on chow and HFD. These results suggest that endogenous VMH amylin signaling is required for full leptin signaling and protection from HFD-induced obesity.


Assuntos
Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Leptina/metabolismo , Obesidade/fisiopatologia , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Dieta Hiperlipídica , Ingestão de Alimentos , Intolerância à Glucose/genética , Resistência à Insulina/genética , Radioisótopos do Iodo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Leptina/genética , Masculino , Obesidade/genética , RNA Interferente Pequeno/genética , Cintilografia , Ratos , Receptores da Calcitonina/genética , Receptores da Calcitonina/metabolismo , Fator de Transcrição STAT3/genética , Núcleo Hipotalâmico Ventromedial/diagnóstico por imagem , Aumento de Peso
8.
Am J Physiol Regul Integr Comp Physiol ; 308(3): R188-98, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25477422

RESUMO

Hypothalamic fatty acid (FA) sensing neurons alter their activity utilizing the FA translocator/receptor, FAT/CD36. Depletion of ventromedial hypothalamus (VMH) CD36 with adeno-associated viral vector expressing CD36 shRNA (AAV CD36 shRNA) leads to redistribution of adipose stores and insulin resistance in outbred rats. This study assessed the requirement of VMH CD36-mediated FA sensing for the regulation of energy and glucose homeostasis in postnatal day 5 (P5) and P21 selectively bred diet-induced obese (DIO) and diet-resistant (DR) rats using VMH AAV CD36 shRNA injections. P5 CD36 depletion altered VMH neuronal FA sensing predominantly in DIO rats. After 10 wk on a 45% fat diet, DIO rats injected with VMH AAV CD36 shRNA at P21 ate more and gained more weight than DIO AAV controls, while DR AAV CD36 shRNA-injected rats gained less weight than DR AAV controls. VMH CD36 depletion increased inguinal fat pad weights and leptin levels in DIO and DR rats. Although DR AAV CD36 shRNA-injected rats became as obese as DIO AAV controls, only DIO control and CD36 depleted rats became insulin-resistant on a 45% fat diet. VMH CD36 depletion stunted linear growth in DIO and DR rats. DIO rats injected with AAV CD36 shRNA at P5 had increased fat mass, mostly due to a 45% increase in subcutaneous fat. They were also insulin-resistant with an associated 71% increase of liver triglycerides. These results demonstrate that VMH CD36-mediated FA sensing is a critical factor in the regulation of energy and glucose homeostasis and fat deposition in DIO and DR rats.


Assuntos
Glicemia , Antígenos CD36/metabolismo , Ingestão de Energia/fisiologia , Ácidos Graxos/metabolismo , Homeostase/fisiologia , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Ração Animal , Animais , Glicemia/metabolismo , Peso Corporal/fisiologia , Modelos Animais de Doenças , Insulina/metabolismo , Leptina , Masculino , Ratos
9.
Am J Physiol Regul Integr Comp Physiol ; 308(10): R872-8, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25786485

RESUMO

The objective of this study was to determine the potential role of astrocyte-derived ketone bodies in regulating the early changes in caloric intake of diet induced-obese (DIO) versus diet-resistant (DR) rats fed a 31.5% fat high-energy (HE) diet. After 3 days on chow or HE diet, DR and DIO rats were assessed for their ventromedial hypothalamic (VMH) ketone bodies levels and neuronal ventromedial hypothalamic nucleus (VMN) sensing using microdialysis coupled to continuous food intake monitoring and calcium imaging in dissociated neurons, respectively. DIO rats ate more than DR rats over 3 days of HE diet intake. On day 3 of HE diet intake, DR rats reduced their caloric intake while DIO rats remained hyperphagic. Local VMH astrocyte ketone bodies production was similar between DR and DIO rats during the first 6 h after dark onset feeding but inhibiting VMH ketone body production in DR rats on day 3 transiently returned their intake of HE diet to the level of DIO rats consuming HE diet. In addition, dissociated VMN neurons from DIO and DR rats were equally sensitive to the largely excitatory effects of ß-hydroxybutyrate. Thus while DR rats respond to increased VMH ketone levels by decreasing their intake after 3 days of HE diet, this is not the case of DIO rats. These data suggest that DIO inherent leptin resistance prevents ketone bodies inhibitory action on food intake.


Assuntos
Ingestão de Energia/fisiologia , Corpos Cetônicos/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Ácido 3-Hidroxibutírico/farmacologia , Animais , Cálcio/metabolismo , Dieta Hiperlipídica , Glucose/farmacologia , Masculino , Neurônios/efeitos dos fármacos , Obesidade/etiologia , Ácido Oleico/farmacologia , Ratos , Ratos Sprague-Dawley , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
10.
Am J Physiol Regul Integr Comp Physiol ; 306(7): R447-56, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24477542

RESUMO

Neurons in the supraoptic nuclei (SON) produce oxytocin and vasopressin and express insulin receptors (InsR) and glucokinase. Since oxytocin is an anorexigenic agent and glucokinase and InsR are hallmarks of cells that function as glucose and/or metabolic sensors, we evaluated the effect of glucose, insulin, and their downstream effector ATP-sensitive potassium (KATP) channels on calcium signaling in SON neurons and on oxytocin and vasopressin release from explants of the rat hypothalamo-neurohypophyseal system. We also evaluated the effect of blocking glucokinase and phosphatidylinositol 3 kinase (PI3K; mediates insulin-induced mobilization of glucose transporter, GLUT4) on responses to glucose and insulin. Glucose and insulin increased intracellular calcium ([Ca(2+)]i). The responses were glucokinase and PI3K dependent, respectively. Insulin and glucose alone increased vasopressin release (P < 0.002). Oxytocin release was increased by glucose in the presence of insulin. The oxytocin (OT) and vasopressin (VP) responses to insulin+glucose were blocked by the glucokinase inhibitor alloxan (4 mM; P ≤ 0.002) and the PI3K inhibitor wortmannin (50 nM; OT: P = 0.03; VP: P ≤ 0.002). Inactivating K ATP channels with 200 nM glibenclamide increased oxytocin and vasopressin release (OT: P < 0.003; VP: P < 0.05). These results suggest that insulin activation of PI3K increases glucokinase-mediated ATP production inducing closure of K ATP channels, opening of voltage-sensitive calcium channels, and stimulation of oxytocin and vasopressin release. The findings are consistent with SON oxytocin and vasopressin neurons functioning as glucose and "metabolic" sensors to participate in appetite regulation.


Assuntos
Sinalização do Cálcio , Glucose/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Neurônios/metabolismo , Ocitocina/metabolismo , Núcleo Supraóptico/metabolismo , Vasopressinas/metabolismo , Animais , Regulação do Apetite , Sinalização do Cálcio/efeitos dos fármacos , Glucoquinase/antagonistas & inibidores , Glucoquinase/genética , Glucoquinase/metabolismo , Sistema Hipotálamo-Hipofisário/citologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Insulina/metabolismo , Canais KATP/antagonistas & inibidores , Canais KATP/metabolismo , Masculino , Neurônios/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Bloqueadores dos Canais de Potássio/farmacologia , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Núcleo Supraóptico/citologia , Núcleo Supraóptico/efeitos dos fármacos , Fatores de Tempo , Técnicas de Cultura de Tecidos
11.
Cell Metab ; 7(2): 179-85, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18249177

RESUMO

The arcuate nucleus of the hypothalamus (ARH) is a key component of hypothalamic pathways regulating energy balance, and leptin is required for normal development of ARH projections. Diet-induced obesity (DIO) has a polygenic mode of inheritance, and DIO individuals develop the metabolic syndrome when a moderate amount of fat is added to the diet. Here we demonstrate that rats selectively bred to develop DIO, which are known to be leptin resistant before they become obese, have defective ARH projections that persist into adulthood. Furthermore, the ability of leptin to activate intracellular signaling in ARH neurons in vivo and to promote ARH neurite outgrowth in vitro is significantly reduced in DIO neonates. Thus, animals that are genetically predisposed toward obesity display an abnormal organization of hypothalamic pathways involved in energy homeostasis that may be the result of diminished responsiveness of ARH neurons to the trophic actions of leptin during postnatal development.


Assuntos
Hipotálamo/patologia , Neuritos , Neurônios/ultraestrutura , Obesidade/etiologia , Animais , Regulação do Apetite , Núcleo Arqueado do Hipotálamo/patologia , Dieta , Metabolismo Energético , Predisposição Genética para Doença , Leptina/fisiologia , Ratos , Ratos Endogâmicos , Transdução de Sinais
12.
Am J Physiol Endocrinol Metab ; 304(2): E131-8, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23169787

RESUMO

A major side effect of insulin treatment of diabetes is weight gain, which limits patient compliance and may pose additional health risks. Although the mechanisms responsible for this weight gain are poorly understood, it has been suggested that there may be a link to the incidence of recurrent episodes of hypoglycemia. Here we present a rodent model of marked weight gain associated with weekly insulin-induced hypoglycemic episodes in the absence of diabetes. Insulin treatment caused a significant increase in both body weight and fat mass, accompanied by reduced motor activity, lowered thermogenesis in response to a cold challenge, and reduced brown fat uncoupling protein mRNA. However, there was no effect of insulin treatment on total food intake nor on hypothalamic neuropeptide Y or proopiomelanocortin mRNA expression, and insulin-treated animals did not become insulin-resistant. Our results suggest that repeated iatrogenic hypoglycemia leads to weight gain, and that such weight gain is associated with a multifaceted deficit in metabolic regulation rather than to a chronic increase in caloric intake.


Assuntos
Hipoglicemia/induzido quimicamente , Hipoglicemia/complicações , Insulina/efeitos adversos , Obesidade/etiologia , Aumento de Peso/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Hiperfagia/complicações , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/efeitos adversos , Insulina/administração & dosagem , Resistência à Insulina/fisiologia , Masculino , Obesidade/induzido quimicamente , Obesidade/patologia , Periodicidade , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Fatores de Tempo
13.
Proc Natl Acad Sci U S A ; 107(33): 14875-80, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20679202

RESUMO

The neuronal circuits involved in the regulation of feeding behavior and energy expenditure are soft-wired, reflecting the relative activity of the postsynaptic neuronal system, including the anorexigenic proopiomelanocortin (POMC)-expressing cells of the arcuate nucleus. We analyzed the synaptic input organization of the melanocortin system in lean rats that were vulnerable (DIO) or resistant (DR) to diet-induced obesity. We found a distinct difference in the quantitative and qualitative synaptology of POMC cells between DIO and DR animals, with a significantly greater number of inhibitory inputs in the POMC neurons in DIO rats compared with DR rats. When exposed to a high-fat diet (HFD), the POMC cells of DIO animals lost synapses, whereas those of DR rats recruited connections. In both DIO rats and mice, the HFD-triggered loss of synapses on POMC neurons was associated with increased glial ensheathment of the POMC perikarya. The altered synaptic organization of HFD-fed animals promoted increased POMC tone and a decrease in the stimulatory connections onto the neighboring neuropeptide Y (NPY) cells. Exposure to HFD was associated with reactive gliosis, and this affected the structure of the blood-brain barrier such that the POMC and NPY cell bodies and dendrites became less accessible to blood vessels. Taken together, these data suggest that consumption of an HFD has a major impact on the cytoarchitecture of the arcuate nucleus in vulnerable subjects, with changes that might be irreversible due to reactive gliosis.


Assuntos
Dieta , Gliose/metabolismo , Melanocortinas/metabolismo , Obesidade/metabolismo , Sinapses/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/patologia , Núcleo Arqueado do Hipotálamo/fisiopatologia , Gorduras na Dieta/efeitos adversos , Feminino , Gliose/etiologia , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica , Neurônios/metabolismo , Neurônios/ultraestrutura , Neuropeptídeo Y/metabolismo , Obesidade/etiologia , Pró-Opiomelanocortina/metabolismo , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica/fisiologia
14.
Cell Metab ; 6(4): 252-4, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17908553

RESUMO

Although hypothalamic glucose sensing is a long-established phenomenon, its physiological role remains unclear. New studies (Parton et al., 2007; Claret et al., 2007) disrupting glucose sensing in pro-opiomelanocortin neurons via differing methods have yielded disparate energy and glucose homeostasis phenotypes, suggesting that neuronal glucose sensing is not critical for these processes.


Assuntos
Glucose/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Hipotálamo/citologia , Camundongos , Transdução de Sinais
15.
Front Neuroendocrinol ; 31(3): 270-83, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20206200

RESUMO

Most human obesity is inherited as a polygenic trait which is largely refractory to medical therapy because obese individuals avidly defend their elevated body weight set-point. This set-point is mediated by an integrated neural network that controls energy homeostasis. Epidemiological studies suggest that perinatal and pre-pubertal environmental factors can promote offspring obesity. Rodent studies demonstrate the important interactions between genetic predisposition and environmental factors in promoting obesity. This review covers issues of development and function of neural systems involved in the regulation of energy homeostasis and the roles of leptin and insulin in these processes, the ways in which interventions at various phases from gestation, lactation and pre-pubertal stages of development can favorably and unfavorably alter the development of obesity n offspring. These studies suggest that early identification of obesity-prone humans and of the factors that can prevent them from becoming obese could provide an effective strategy for preventing the world-wide epidemic of obesity.


Assuntos
Metabolismo Energético/genética , Meio Ambiente , Epistasia Genética/fisiologia , Obesidade/etiologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Homeostase/genética , Humanos , Modelos Biológicos , Obesidade/genética
16.
J Neurosci ; 29(21): 7015-22, 2009 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-19474328

RESUMO

Although several studies implicate small declines in blood glucose levels as stimulus for spontaneous meal initiation, no mechanism is known for how these dips might initiate feeding. To assess the role of ventromedial hypothalamus (VMH) (arcuate plus ventromedial nucleus) glucosensing neurons as potential mediators of spontaneous and glucoprivic feeding, meal patterns were observed, and blood and VMH microdialysis fluid were sampled in 15 rats every 10 min for 3.5 h after dark onset and 2 h after insulin (5 U/kg, i.v.) infusion. Blood glucose levels declined by 11% beginning approximately 5 min before 65% of all spontaneous meals, with no fall in VMH levels. After insulin, blood and VMH glucose reached nadirs by 30-40 min, and the same rats ate 60% faster and spent 84% more time eating during the ensuing hypoglycemia. Although 83% of first hypoglycemic meals were preceded by 5 min dips in VMH (but not blood) glucose levels, neither blood nor VMH levels declined before second meals, suggesting that low glucose, rather than changing levels, was the stimulus for glucoprivic meals. Furthermore, altering VMH glucosensing by raising or lowering glucokinase (GK) activity failed to affect spontaneous feeding, body or adipose weights, or glucose tolerance. However, chronic depletion by 26-70% of VMH GK mRNA reduced glucoprivic feeding. Thus, although VMH glucosensing does not appear to be involved in either spontaneous feeding or long-term body-weight regulation, it does participate in glucoprivic feeding, similar to its role in the counter-regulatory neurohumoral responses to glucoprivation.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Glicemia/fisiologia , Comportamento Alimentar/fisiologia , Glucose/deficiência , Núcleo Hipotalâmico Ventromedial/metabolismo , Análise de Variância , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Comportamento Animal , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Comportamento Alimentar/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Glucoquinase/genética , Glucoquinase/metabolismo , Glucose/análogos & derivados , Glucose/farmacologia , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Masculino , Microdiálise/métodos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
17.
Am J Physiol Regul Integr Comp Physiol ; 297(5): R1351-7, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19710389

RESUMO

Maternal obesity accentuates offspring obesity in dams bred to develop diet-induced obesity (DIO) on a 31% fat, high-sucrose, high-energy (HE) diet but has no effect on offspring of diet-resistant (DR) dams. Also, only DIO dams become obese when they and DR dams are fed HE diet throughout gestation and lactation. We assessed glucose and oleic acid (OA) sensitivity of dissociated ventromedial hypothalamic nucleus (VMN) neurons from 3- to 4-wk old offspring of DIO and DR dams fed chow or HE diet using fura-2 calcium imaging to monitor intracellular calcium fluctuations as an index of neuronal activity. Offspring of DIO dams fed chow had approximately 2-fold more glucose-inhibited (GI) neurons than did DR offspring. This difference was eliminated in offspring of DIO dams fed HE diet. At 2.5 mM glucose, offspring of chow-fed DIO dams had more GI neurons that were either excited or inhibited by OA than did DR offspring. Maternal HE diet intake generally increased the percentage of neurons that were excited and decreased the percentage that were inhibited by OA in both DIO and DR offspring. However, this effect was more pronounced in DIO offspring. These data, as well as concentration-dependent differences in OA sensitivity, suggest that genotype, maternal obesity, and dietary content can all affect the sensitivity of offspring VMN neurons to glucose and long-chain fatty acids. Such altered sensitivities may underlie the propensity of DIO offspring to become obese when fed high-fat, high-sucrose diets.


Assuntos
Glicemia/metabolismo , Gorduras na Dieta/farmacologia , Sacarose Alimentar/farmacologia , Ácidos Graxos/metabolismo , Neurônios/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Animais Recém-Nascidos/metabolismo , Peso Corporal/fisiologia , Cálcio/metabolismo , Modelos Animais de Doenças , Feminino , Genótipo , Insulina/sangue , Leptina/sangue , Masculino , Obesidade/metabolismo , Ácido Oleico/farmacologia , Gravidez , Prenhez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/genética , Ratos , Ratos Endogâmicos , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
18.
Am J Physiol Regul Integr Comp Physiol ; 297(3): R655-64, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19535676

RESUMO

We assessed the mechanisms by which specialized hypothalamic ventromedial nucleus (VMN) neurons utilize both glucose and long-chain fatty acids as signaling molecules to alter their activity as a potential means of regulating energy homeostasis. Fura-2 calcium (Ca(2+)) and membrane potential dye imaging, together with pharmacological agents, were used to assess the mechanisms by which oleic acid (OA) alters the activity of dissociated VMN neurons from 3- to 4-wk-old rats. OA excited up to 43% and inhibited up to 29% of all VMN neurons independently of glucose concentrations. In those neurons excited by both 2.5 mM glucose and OA, OA had a concentration-dependent effective excitatory concentration (EC(50)) of 13.1 nM. Neurons inhibited by both 2.5 mM glucose and OA had an effective inhibitory concentration (IC(50)) of 93 nM. At 0.5 mM glucose, OA had markedly different effects on these same neurons. Inhibition of carnitine palmitoyltransferase, reactive oxygen species formation, long-chain acetyl-CoA synthetase and ATP-sensitive K(+) channel activity or activation of uncoupling protein 2 (UCP2) accounted for only approximately 20% of OA's excitatory effects and approximately 40% of its inhibitory effects. Inhibition of CD36, a fatty acid transporter that can alter cell function independently of intracellular fatty acid metabolism, reduced the effects of OA by up to 45%. Thus OA affects VMN neuronal activity through multiple pathways. In glucosensing neurons, its effects are glucose dependent. This glucose-OA interaction provides a potential mechanism whereby such "metabolic sensing" neurons can respond to differences in the metabolic states associated with fasting and feeding.


Assuntos
Sinalização do Cálcio , Metabolismo Energético , Glucose/metabolismo , Neurônios/metabolismo , Ácido Oleico/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Antígenos CD36/metabolismo , Carnitina O-Palmitoiltransferase/antagonistas & inibidores , Carnitina O-Palmitoiltransferase/metabolismo , Coenzima A Ligases/antagonistas & inibidores , Coenzima A Ligases/metabolismo , Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Homeostase , Canais Iônicos/metabolismo , Canais KATP/antagonistas & inibidores , Canais KATP/metabolismo , Masculino , Potenciais da Membrana , Microscopia de Fluorescência , Proteínas Mitocondriais/metabolismo , Inibição Neural , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo , Proteína Desacopladora 2 , Núcleo Hipotalâmico Ventromedial/citologia , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos , Núcleo Hipotalâmico Ventromedial/enzimologia
19.
Endocrinology ; 149(10): 5146-54, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18556346

RESUMO

Neurons in the ventromedial and arcuate hypothalamic nuclei (VMN and ARC, respectively) mediate many of leptin's effects on energy homeostasis. Some are also glucosensing, whereby they use glucose as a signaling molecule to regulate their firing rate. We used fura-2 calcium (Ca2+) imaging to determine the interactions between these two important mediators of peripheral metabolism on individual VMN neurons and the mechanisms by which leptin regulates neuronal activity in vitro. Leptin excited 24%, inhibited 20%, and had a biphasic response in 10% of VMN neurons. Excitation occurred with a EC50 of 5.2 fmol/liter and inhibition with a IC50 of 4.2 fmol/liter. These effects were independent of the ambient glucose levels, and both glucosensing and non-glucosensing neurons were affected by leptin. In contrast, the ARC showed a very different distribution of leptin-responsive neurons, with 40% leptin excited, 10% leptin inhibited, and 2% having a biphasic response (chi2=60.2; P<0.0001). Using pharmacological manipulations we found that leptin inhibits VMN neurons via activation of phosphoinositol-3 kinase and activation of the ATP-sensitive K+ channel. In addition, leptin inhibition was antagonized by 5'-AMP-activated protein kinase activation in 39% of neurons but was unaffected by 5'-AMP-activated protein kinase inhibition. No mechanism was delineated for leptin-induced excitation. Thus, within the physiological range of brain glucose levels, leptin has a differential effect on VMN vs. ARC neurons, and acts on both glucosensing and non-glucosensing VMN neurons in a glucose-independent fashion with inhibition primarily dependent upon activation of the ATP-sensitive K+ channel.


Assuntos
Leptina/metabolismo , Leptina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Núcleo Hipotalâmico Ventromedial/citologia , Proteínas Quinases Ativadas por AMP , Animais , Núcleo Arqueado do Hipotálamo/citologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Glucose/metabolismo , Canais KATP/metabolismo , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Complexos Multienzimáticos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley
20.
Neuroendocrinology ; 87(2): 65-70, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17374946

RESUMO

Many of the small percentage of previously obese humans who successfully maintain weight loss report high levels of physical activity, suggesting a role for exercise in the maintenance of their lower body weights. The rat model of diet-induced obesity (DIO) has been particularly useful, since it shares several common characteristics with human obesity and, unlike the human condition, allows a thorough investigation of the effects of exercise on the central pathways which regulate energy homeostasis. In rats with DIO, voluntary wheel running selectively reduces adiposity without causing a compensatory increase in energy intake. These effects are likely mediated by signals generated by the exercising body such as interleukin-6, fatty acids, and heat which feed back on the brain to regulate central neuropeptide systems involved in the regulation of energy homeostasis. While exercise provides temporary reductions in obesity in adult rats, early postweaning exercise reduces adiposity in high-fat-fed DIO rats long after exercise is terminated. This suggests that early-onset exercise may permanently alter the development of the central pathways which regulate energy homeostasis. Therefore, identification of exercise-induced central and peripheral factors and elucidation of their interactions with central modulatory pathways may aid in the identification of new targets for the pharmacological treatment of human obesity.


Assuntos
Metabolismo Energético/fisiologia , Exercício Físico/fisiologia , Obesidade/prevenção & controle , Adiposidade/fisiologia , Animais , Modelos Animais de Doenças , Homeostase/fisiologia , Humanos , Obesidade/fisiopatologia , Condicionamento Físico Animal/fisiologia , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA