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










Base de dados
Intervalo de ano de publicação
2.
Diabetes ; 67(5): 805-817, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29467172

RESUMO

Amylin phosphorylates ERK (p-ERK) in the area postrema to reduce eating and synergizes with leptin to phosphorylate STAT3 in the arcuate (ARC) and ventromedial (VMN) hypothalamic nuclei to reduce food intake and body weight. The current studies assessed potential amylin and amylin-leptin ARC/VMN interactions on ERK signaling and their roles in postnatal hypothalamic pathway development. In amylin knockout mice, the density of agouti-related protein (AgRP)-immunoreactive (IR) fibers in the hypothalamic paraventricular nucleus (PVN) was increased, while the density of α-melanocyte-stimulating hormone (αMSH) fibers was decreased. In mice deficient of the amylin receptor components RAMP1/3, both AgRP and αMSH-IR fiber densities were decreased, while only αMSH-IR fiber density was decreased in rats injected neonatally in the ARC/VMN with an adeno-associated virus short hairpin RNA against the amylin core receptor. Amylin induced p-ERK in ARC neurons, 60% of which was present in POMC-expressing neurons, with none in NPY neurons. An amylin-leptin interaction was shown by an additive effect on ARC ERK signaling in neonatal rats and a 44% decrease in amylin-induced p-ERK in the ARC of leptin receptor-deficient and of ob/ob mice. Together, these results suggest that amylin directly acts, through a p-ERK-mediated process, on POMC neurons to enhance ARC-PVN αMSH pathway development.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Leptina/metabolismo , Sistema de Sinalização das MAP Quinases , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Pró-Opiomelanocortina/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Animais Recém-Nascidos , Comportamento Alimentar , Feminino , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Masculino , Camundongos , Camundongos Knockout , Camundongos Obesos , Neuropeptídeo Y/metabolismo , Ratos Sprague-Dawley , Proteína 1 Modificadora da Atividade de Receptores/genética , Proteína 1 Modificadora da Atividade de Receptores/metabolismo , Proteína 3 Modificadora da Atividade de Receptores/genética , Proteína 3 Modificadora da Atividade de Receptores/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , alfa-MSH/metabolismo
3.
Trends Endocrinol Metab ; 28(2): 153-164, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27938937

RESUMO

While the regulation of energy homeostasis by amylin is already well-characterized, emerging data suggest that amylin is also crucial for the development of neural pathways in the hypothalamus and caudal hindbrain (area postrema, AP; nucleus tractus solitarius, NTS). Exciting new findings demonstrate crucial amylin-leptin interactions in altering the activity of specific hypothalamic and AP neurons, and a role for amylin as a novel class of 'leptin sensitizers' which enhance leptin signaling in both leptin-sensitive and -resistant individuals, in part by stimulating IL-6 production by hypothalamic microglia. This review summarizes these findings and provides a hypothetical framework for future studies to elucidate the mechanisms by which amylin and leptin act individually and as co-conspirators to alter energy homeostasis and neuronal development.


Assuntos
Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Leptina/metabolismo , Animais , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , Homeostase/genética , Homeostase/fisiologia , Humanos , Hipotálamo/metabolismo , Neurônios/metabolismo , Transdução de Sinais
4.
Physiol Behav ; 176: 217-222, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28034577

RESUMO

It was a great and humbling honor to receive the 2016 Distinguished Career Award from my SSIB colleagues. This paper summarizes the major points of my DCA talk at the 2016 annual meeting. It is a reflection on my 50year medical and research career and 10 lessons I have learned over those years which might be of help to young investigators near the beginning of their own research careers. These lessons include: the value of being receptive to the opportunities provided you; how clinician-scientists can serve as critical role models for young investigators like me and a history of how my career developed as a result of their influence; the importance of carefully examining your own data, particularly when it doesn't agree with your preconceived ideas; the critical role that students, postdocs and PhD (and even veterinarian) colleagues can play in developing one's career; the likelihood that your career path will have many interesting twists and turns determined by changes in your own scientific interests and how rewarding various areas of research focus are to you; the importance of building a close-knit laboratory staff family; the fact that science and romance can mix. Finally, I offer 3 somewhat self-evident free pieces of advice for building and maintaining a rewarding career.


Assuntos
Pesquisa Biomédica , Aprendizagem , Médicos , Animais , Distinções e Prêmios , Pesquisa Biomédica/história , Pesquisa Biomédica/métodos , Comportamento Alimentar/fisiologia , História do Século XXI , Humanos
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 ; 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
8.
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
9.
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
10.
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
11.
Mol Cell Endocrinol ; 418 Pt 1: 3-8, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26415589

RESUMO

Fatty acid (FA) -sensitive neurons are present in the brain, especially the hypothalamus, and play a key role in the neural control of energy and glucose homeostasis including feeding behavior, secretion insulin and action. Subpopulations of neurons in the arcuate and ventromedial hypothalamic nuclei are selectively either activated or inhibited by FA. Molecular effectors of these FA effects include ion channels such as chloride, potassium or calcium. In addition, at least half of the responses in the hypothalamic ventromedial FA neurons are mediated through interaction with the FA translocator/receptor, FAT/CD36, that does not require metabolism to activate intracellular signaling downstream. Recently, an important role of lipoprotein lipase in FA detection has also been demonstrated not only in the hypothalamus, but also in the hippocampus and striatum. Finally, FA could overload energy homeostasis via increased hypothalamic ceramide synthesis which could, in turn, contribute to the pathogenesis of diabetes of obesity and/or type 2 in predisposed individuals by disrupting the endocrine signaling pathways of insulin and/or leptin.


Assuntos
Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Ácidos Graxos/metabolismo , Neurônios/metabolismo , Animais , Homeostase/fisiologia , Humanos , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo
12.
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
13.
Obesity (Silver Spring) ; 23(1): 7-15, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25469998

RESUMO

OBJECTIVES: The National Institutes of Health, led by the National Heart, Lung, and Blood Institute, organized a working group of experts to discuss the problem of weight regain after weight loss. A number of experts in integrative physiology and behavioral psychology were convened with the goal of merging their perspectives regarding the barriers to scientific progress and the development of novel ways to improve long-term outcomes in obesity therapeutics. The specific objectives of this working group were to: (1) identify the challenges that make maintaining a reduced weight so difficult; (2) review strategies that have been used to improve success in previous studies; and (3) recommend novel solutions that could be examined in future studies of long-term weight control. RESULTS: Specific barriers to successful weight loss maintenance include poor adherence to behavioral regimens and physiological adaptations that promote weight regain. A better understanding of how these behavioral and physiological barriers are related, how they vary between individuals, and how they can be overcome will lead to the development of novel strategies with improved outcomes. CONCLUSIONS: Greater collaboration and cross-talk between physiological and behavioral researchers is needed to advance the science and develop better strategies for weight loss maintenance.


Assuntos
Pesquisa Biomédica , Obesidade/terapia , Terapias em Estudo/métodos , Redução de Peso , Pesquisa Biomédica/tendências , Peso Corporal , Comportamentos Relacionados com a Saúde , Humanos , National Institutes of Health (U.S.) , Cooperação do Paciente/psicologia , Relatório de Pesquisa , Estados Unidos
14.
Diabetes ; 64(5): 1621-31, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25409701

RESUMO

Amylin acts acutely via the area postrema to reduce food intake and body weight, but it also interacts with leptin over longer periods of time, possibly via the ventromedial hypothalamus (VMH), to increase leptin signaling and phosphorylation of STAT3. We postulated that amylin enhances VMH leptin signaling by inducing interleukin (IL)-6, which then interacts with its gp130 receptor to activate STAT3 signaling and gene transcription downstream of the leptin receptor. We found that components of the amylin receptor (RAMPs1-3, CTR1a,b) are expressed in cultured VMH astrocytes, neurons, and microglia, as well as in micropunches of arcuate and ventromedial hypothalamic nuclei (VMN). Amylin exposure for 5 days increased IL-6 mRNA expression in VMH explants and microglia by two- to threefold, respectively, as well as protein abundance in culture supernatants by five- and twofold, respectively. Amylin had no similar effects on cultured astrocytes or neurons. In rats, 5 days of amylin treatment decreased body weight gain and/or food intake and increased IL-6 mRNA expression in the VMN. Similar 5-day amylin treatment increased VMN leptin-induced phosphorylation of STAT3 expression in wild-type mice and rats infused with lateral ventricular IgG but not in IL-6 knockout mice or rats infused with ventricular IL-6 antibody. Lateral ventricular infusion of IL-6 antibody also prevented the amylin-induced decrease of body weight gain. These results show that amylin-induced VMH microglial IL-6 production is the likely mechanism by which amylin treatment interacts with VMH leptin signaling to increase its effect on weight loss.


Assuntos
Interleucina-6/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/farmacologia , Leptina/metabolismo , Transdução de Sinais/fisiologia , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Astrócitos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Interleucina-6/genética , Leptina/genética , Masculino , Camundongos , Camundongos Knockout , Microglia/metabolismo , Neurônios/metabolismo , RNA Mensageiro , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Núcleo Hipotalâmico Ventromedial/citologia
15.
Diabetes ; 64(1): 226-35, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25114294

RESUMO

Previous reports suggested an important role for serotonin (5-hydroxytryptamine [5-HT]) in enhancing the counterregulatory response (CRR) to hypoglycemia. To elucidate the sites of action mediating this effect, we initially found that insulin-induced hypoglycemia stimulates 5-HT release in widespread forebrain regions, including the perifornical hypothalamus (PFH; 30%), ventromedial hypothalamus (34%), paraventricular hypothalamus (34%), paraventricular thalamic nucleus (64%), and cerebral cortex (63%). Of these, we focused on the PFH because of its known modulation of diverse neurohumoral and behavioral responses. In awake, behaving rats, bilateral PFH glucoprivation with 5-thioglucose stimulated adrenal medullary epinephrine (Epi) release (3,153%) and feeding (400%), while clamping PFH glucose at postprandial brain levels blunted the Epi response to hypoglycemia by 30%. The PFH contained both glucose-excited (GE) and glucose-inhibited (GI) neurons; GE neurons were primarily excited, while GI neurons were equally excited or inhibited by 5-HT at hypoglycemic glucose levels in vitro. Also, 5-HT stimulated lactate production by cultured hypothalamic astrocytes. Depleting PFH 5-HT blunted the Epi (but not feeding) response to focal PFH (69%) and systemic glucoprivation (39%), while increasing PFH 5-HT levels amplified the Epi response to hypoglycemia by 32%. Finally, the orexin 1 receptor antagonist SB334867A attenuated both the Epi (65%) and feeding (47%) responses to focal PFH glucoprivation. Thus we have identified the PFH as a glucoregulatory region where both 5-HT and orexin modulate the CRR and feeding responses to glucoprivation.


Assuntos
Fórnice/metabolismo , Hipoglicemia/metabolismo , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neuropeptídeos/metabolismo , Serotonina/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Benzoxazóis/farmacologia , Glicemia/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Hipoglicemia/fisiopatologia , Masculino , Núcleos da Linha Média do Tálamo/metabolismo , Naftiridinas , Neurônios/citologia , Neurônios/metabolismo , Orexinas , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos Sprague-Dawley , Receptores de Neuropeptídeos/antagonistas & inibidores , Ureia/análogos & derivados , Ureia/farmacologia , Núcleo Hipotalâmico Ventromedial/metabolismo
16.
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
17.
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
18.
Diab Vasc Dis Res ; 12(1): 2-12, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25268022

RESUMO

In Type 1 and advanced Type 2 diabetes mellitus, elevation of plasma epinephrine plays a key role in normalizing plasma glucose during hypoglycaemia. However, recurrent hypoglycaemia blunts this elevation of plasma epinephrine. To determine whether recurrent hypoglycaemia affects peripheral components of the sympatho-adrenal system responsible for epinephrine release, male rats were administered subcutaneous insulin daily for 3 days. These recurrent hypoglycaemic animals showed a smaller elevation of plasma epinephrine than saline-injected controls when subjected to insulin-induced hypoglycaemia. Electrical stimulation of an adrenal branch of the splanchnic nerve in recurrent hypoglycaemic animals elicited less release of epinephrine and norepinephrine than in controls, without a change in adrenal catecholamine content. Responsiveness of isolated, perfused adrenal glands to acetylcholine and other acetylcholine receptor agonists was also unchanged. These results indicate that recurrent hypoglycaemia compromised the efficacy with which peripheral neuronal activity stimulates adrenal catecholamine release and demonstrate that peripheral components of the sympatho-adrenal system were directly affected by recurrent hypoglycaemia.


Assuntos
Glândulas Suprarrenais/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Epinefrina/metabolismo , Hipoglicemia/fisiopatologia , Norepinefrina/metabolismo , Acetilcolina/metabolismo , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/inervação , Animais , Glicemia/análise , Agonistas Colinérgicos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Estimulação Elétrica , Epinefrina/sangue , Hipoglicemia/sangue , Hipoglicemia/metabolismo , Masculino , Agonistas Muscarínicos/farmacologia , Nicotina/farmacologia , Norepinefrina/sangue , Perfusão , Pilocarpina/farmacologia , Ratos Sprague-Dawley , Receptores Colinérgicos/química , Receptores Colinérgicos/metabolismo , Recidiva , Nervos Esplâncnicos/metabolismo
19.
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
20.
Diabetes ; 63(4): 1259-69, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24379353

RESUMO

Metabolic sensing neurons in the ventromedial hypothalamus (VMH) alter their activity when ambient levels of metabolic substrates, such as glucose and fatty acids (FA), change. To assess the relationship between a high-fat diet (HFD; 60%) intake on feeding and serum and VMH FA levels, rats were trained to eat a low-fat diet (LFD; 13.5%) or an HFD in 3 h/day and were monitored with VMH FA microdialysis. Despite having higher serum levels, HFD rats had lower VMH FA levels but ate less from 3 to 6 h of refeeding than did LFD rats. However, VMH ß-hydroxybutyrate (ß-OHB) and VMH-to-serum ß-OHB ratio levels were higher in HFD rats during the first 1 h of refeeding, suggesting that VMH astrocyte ketone production mediated their reduced intake. In fact, using calcium imaging in dissociated VMH neurons showed that ketone bodies overrode normal FA sensing, primarily by exciting neurons that were activated or inhibited by oleic acid. Importantly, bilateral inhibition of VMH ketone production with a 3-hydroxy-3-methylglutaryl-CoA synthase inhibitor reversed the 3- to 6-h HFD-induced inhibition of intake but had no effect in LFD-fed rats. These data suggest that a restricted HFD intake regimen inhibits caloric intake as a consequence of FA-induced VMH ketone body production by astrocytes.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Ácidos Graxos/farmacologia , Hipotálamo/fisiologia , Corpos Cetônicos/farmacologia , Neurônios/fisiologia , Ácido 3-Hidroxibutírico/sangue , Animais , Hipotálamo/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...