Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Alzheimers Res Ther ; 13(1): 32, 2021 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-33504364

RESUMO

BACKGROUND: Prominent activation of microglial immune/inflammatory processes is a characteristic feature of brains of patients with tauopathies including Alzheimer's disease (AD), suggesting that neuroinflammation may be a critical factor in their pathogenesis. Strategies aimed at developing new therapeutics for tauopathies based on anti-inflammation or immunomodulation are likely to be promising avenues of research. We previously developed JM4-a 19'mer cyclic peptide derived from the first loop of human erythropoietin. This peptide possesses beneficial immune modulatory and tissue protective effects while lacking the undesirable side effects of full-length erythropoietin. In this preclinical study, we investigated the effect of chronic JM4 treatment on the PS19 mouse that carries the P301S mutant human tau gene, linked to a form of frontotemporal dementia. This transgenic mouse has been widely used as a model of tauopathies including AD and related dementias. METHODS: Daily subcutaneous treatment of female PS19 mice with JM4 was initiated before disease onset and continued on for the animals' lifespan. The progression of neurological deficit and the lifespan of these mice were assessed. To evaluate the effect of JM4 treatment on cognition of these animals, the PS19 mice underwent Barnes maze test and elevated plus maze test. In addition, neuronal loss, phosphorylated tau aggregation, and microglial activation were assessed using immunohistochemistry of PS19 mouse brain sections. RESULTS: JM4 treatment of PS19 mice initiated before disease onset reduced neurological deficit, prolonged lifespan, and rescued memory impairment. The beneficial effects of JM4 were accompanied by reductions in neuronal loss, phosphorylated tau aggregation, and microglial activation in the PS19 mouse brain. LIMITATIONS: Use of a single dose of JM4 and female mice only. CONCLUSION: JM4 is a potential novel therapeutic agent for the treatment of tauopathies including AD and related dementias.


Assuntos
Eritropoetina , Tauopatias , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Tauopatias/tratamento farmacológico , Proteínas tau/genética , Proteínas tau/metabolismo
7.
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
8.
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
9.
Front Aging Neurosci ; 11: 252, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31572168

RESUMO

Many studies of tauopathy use transgenic mice that overexpress the P301S mutant form of tau. Neuronal damage in these mice is associated with astrogliosis and induction of glial fibrillary acidic protein (GFAP) expression. GFAP-luc transgenic mice express firefly luciferase under the GFAP promoter, allowing bioluminescence to be measured non-invasively as a surrogate biomarker for astrogliosis. We bred double transgenic mice possessing both P301S and GFAP-luc cassettes and compared them to control mice bearing only the GFAP-luc transgene. We used serial bioluminescent images to define the onset and the time course of astrogliosis in these mice and this was correlated with the development of clinical deficit. Mice containing both GFAP-luc and P301S transgenes showed increased luminescence indicative of astroglial activation in the brain and spinal cord. Starting at 5 months old, the onset of clinical deterioration in these mice corresponded closely to the initial rise in the luminescent signal. Post mortem analysis showed the elevated luminescence was correlated with hyperphosphorylated tau deposition in the hippocampus of double transgenic mice. We used this method to determine the therapeutic effect of JM4 peptide [a small peptide immunomodulatory agent derived from human erythropoietin (EPO)] on double transgenic mice. JM4 treatment significantly decreased the intensity of luminescence, neurological deficit and hyperphosphorylated tau in mice with both the P301S and GFAP-luc transgenes. These findings indicate that bioluminescence imaging (BLI) is a powerful tool for quantifying GFAP expression in living P301S mice and can be used as a noninvasive biomarker of tau-induced neurodegeneration in preclinical therapeutic trials.

10.
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
11.
Endocrinology ; 148(1): 310-6, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17023527

RESUMO

Rats with a genetic predisposition to develop diet-induced obesity (DIO) have a preexisting reduction in central leptin and insulin sensitivity. High-fat diets also reduce sensitivity to leptin, insulin, and melanocortin agonists. We postulated that such reduced sensitivities would be associated with decreased binding to the hypothalamic leptin, insulin, and melanocortin receptors in selectively bred DIO rats and in rats fed a high-energy (HE; 31% fat) diet for 7 wk. On HE diet, DIO rats gained 15% more weight and had 121% heavier fat pads and 70% higher leptin levels than low fat chow-fed DIO rats. Diet-resistant (DR) rats gained no more weight on HE diet but had 48% heavier fat pads and 70% higher leptin levels than chow-fed DR rats. Compared with DR rats, DIO (125)I-leptin binding was 41, 36, and 40% lower in the hypothalamic dorsomedial, arcuate, and dorsomedial portion of the ventromedial nuclei, respectively, and arcuate (125)I-insulin binding was 31% lower independent of diet. In contrast, hypothalamic melanocortin binding did not differ between DIO and DR rats. However, HE diet intake lowered lateral hypothalamic melanocortin-3 and melanocortin-4 receptor and hippocampal insulin binding of both DIO and DR rats and hypothalamic paraventricular nucleus melanocortin-4 receptor binding only in DR rats. Neither genotype nor diet affected substantia nigra or ventral tegmental area binding. These results corroborate our previous findings demonstrating a preexisting decrease in DIO hypothalamic leptin and insulin signaling and demonstrate that HE diet intake reduces hypothalamic melanocortin and hippocampal insulin binding.


Assuntos
Hipotálamo/metabolismo , Insulina/farmacocinética , Leptina/farmacocinética , Melanocortinas/metabolismo , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Glicemia , Gorduras na Dieta/farmacologia , Núcleo Hipotalâmico Dorsomedial/metabolismo , Insulina/sangue , Radioisótopos do Iodo , Leptina/sangue , Masculino , Obesidade/genética , Ratos , Ratos Mutantes , Receptor Tipo 4 de Melanocortina/metabolismo , Substância Negra/metabolismo , Área Tegmentar Ventral/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Aumento de Peso/fisiologia
12.
Diabetes ; 55(2): 412-20, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16443775

RESUMO

To test the hypothesis that glucokinase is a critical regulator of neuronal glucosensing, glucokinase activity was increased, using a glucokinase activator drug, or decreased, using RNA interference combined with calcium imaging in freshly dissociated ventromedial hypothalamic nucleus (VMN) neurons or primary ventromedial hypothalamus (VMH; VMN plus arcuate nucleus) cultures. To assess the validity of our approach, we first showed that glucose-induced (0.5-2.5 mmol/l) changes in intracellular Ca(2+) concentration ([Ca(2+)](i)) oscillations, using fura-2 and changes in membrane potential (using a membrane potential-sensitive dye), were highly correlated in both glucose-excited and -inhibited neurons. Also, glucose-excited neurons increased (half-maximal effective concentration [EC(50)] = 0.54 mmol/l) and glucose-inhibited neurons decreased (half-maximal inhibitory concentration [IC(50)] = 1.12 mmol/l) [Ca(2+)](i) oscillations to incremental changes in glucose from 0.3 to 5 mmol/l. In untreated primary VMH neuronal cultures, the expression of glucokinase mRNA and the number of demonstrable glucosensing neurons fell spontaneously by half over 12-96 h without loss of viable neurons. Transfection of neurons with small interfering glucokinase RNA did not affect survival but did reduce glucokinase mRNA by 90% in association with loss of all demonstrable glucose-excited neurons and a 99% reduction in glucose-inhibited neurons. A pharmacological glucokinase activator produced a dose-related increase in [Ca(2+)](i) oscillations in glucose-excited neurons (EC(50) = 0.98 mmol/l) and a decrease in glucose-inhibited neurons (IC(50) = 0.025 micromol/l) held at 0.5 mmol/l glucose. Together, these data support a critical role for glucokinase in neuronal glucosensing.


Assuntos
Glucoquinase/metabolismo , Glucose/metabolismo , Neurônios/enzimologia , Neurônios/fisiologia , Núcleo Hipotalâmico Ventromedial/enzimologia , Núcleo Hipotalâmico Ventromedial/fisiologia , Animais , Cálcio/metabolismo , Células Cultivadas , Regulação Enzimológica da Expressão Gênica , Glucoquinase/genética , Hipoglicemiantes/farmacologia , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Tolbutamida/farmacologia , Núcleo Hipotalâmico Ventromedial/citologia , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
13.
J Cereb Blood Flow Metab ; 25(12): 1645-55, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15902194

RESUMO

Intensive insulin therapy in patients with type 1 diabetes mellitus reduces long-term complications; however, intensive therapy is also associated with a three-fold increase in hypoglycemic episodes. The present study in conscious rats characterizes the physiologic and neuropathologic consequences of a single episode of moderate hypoglycemia. In this model, intravenous insulin is used to reduce plasma glucose to 30 to 35 mg/dL for 75 mins. This single hypoglycemic insult acutely induces hypoglycemia-associated autonomic failure (HAAF), with epinephrine responses to hypoglycemia reduced more than 36% from control. Neuropathology after this insult includes the appearance of dying cells, assessed with the marker Fluoro-jade B (FJ). After hypoglycemic insult, FJ+ cells were consistently seen in subdivisions of the medial prefrontal cortex, the orbital cortex, and the piriform cortex. There was a significant correlation between depth of hypoglycemia and number of FJ+ cells, suggesting that there is a critical threshold below which vulnerable cells begin to die. These data suggest that there is a population of cells that are vulnerable to moderate levels of hypoglycemia commonly experienced by patients with insulin-treated diabetes. These cells, which may be neurons, are primarily found in cortical regions implicated in visceral perception and autonomic control, raising the possibility that their loss contributes to clinically reported deficits in autonomic and perceptual responses to hypoglycemia.


Assuntos
Medula Suprarrenal/fisiopatologia , Sistema Nervoso Autônomo/fisiopatologia , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Hipoglicemia/fisiopatologia , Animais , Biomarcadores , Glicemia , Estado de Consciência , Fluoresceínas , Hipoglicemia/induzido quimicamente , Hipoglicemia/patologia , Hipoglicemiantes/sangue , Hipoglicemiantes/farmacologia , Insulina/sangue , Insulina/farmacologia , Ácido Láctico/sangue , Masculino , Compostos Orgânicos , Ratos , Ratos Sprague-Dawley
14.
Diabetes ; 53(5): 1230-6, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15111491

RESUMO

Glucokinase (GK) is hypothesized to be the critical glucosensor of pancreatic beta-cells and hypothalamic glucosensing neurons. To understand the role of GK in glucoprivic counterregulatory responses, we injected alloxan, a GK inhibitor and toxin, into the third ventricle (3v) to target nearby GK-expressing neurons. Four and 6 days after 3v, but not 4v, alloxan injection, alloxan-treated rats ate only 30% and their blood glucose area under the curve was only 28% of saline controls' after systemic 2-deoxy-D-glucose. In addition, their hyperglycemic response to hindbrain glucoprivation induced with 5-thio-glucose was impaired, whereas fasting blood glucose levels and food intake after an overnight fast were elevated. These impaired responses were associated with the destruction of 3v tanycytes, reduced glial fibrillary acidic protein-immunoreactivity surrounding the 3v, neuronal swelling, and decreased arcuate nucleus neuropeptide Y (NPY) mRNA. Nevertheless, hypothalamic GK mRNA was significantly elevated. Two weeks after alloxan injection, 3v tanycyte destruction was reversed along with restoration of feeding and hyperglycemic responses to both systemic and hindbrain glucoprivation. At this time there were significant decreases in GK, NPY, and proopiomelanocortin mRNA. Thus, neural substrates near and around the 3v affected by alloxan may be critically involved in the expression of these glucoprivic responses.


Assuntos
Aloxano/administração & dosagem , Inibidores Enzimáticos/administração & dosagem , Glucoquinase/antagonistas & inibidores , Glucoquinase/fisiologia , Glucose/análogos & derivados , Glucose/metabolismo , Terceiro Ventrículo , Animais , Glicemia/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/intoxicação , Quarto Ventrículo , Glucoquinase/genética , Hiperglicemia/induzido quimicamente , Hipotálamo/metabolismo , Injeções , Injeções Intraventriculares , Masculino , Bulbo , Neuropeptídeo Y/genética , Neuropeptídeo Y/fisiologia , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/fisiologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Terceiro Ventrículo/patologia
15.
Diabetes ; 53(10): 2521-8, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15448079

RESUMO

Glucosensing neurons are specialized cells that use glucose as a signaling molecule to alter their action potential frequency in response to variations in ambient glucose levels. Glucokinase (GK) appears to be the primary regulator of most neuronal glucosensing, but other regulators almost certainly exist. Glucose-excited neurons increase their activity when glucose levels rise, and most use GK and an ATP-sensitive K(+) channel as the ultimate effector of glucose-induced signaling. Glucose-inhibited (GI) neurons increase their activity at low glucose levels. Although many use GK, it is unclear what the final pathway of GI neuronal glucosensing is. Glucosensing neurons are located in brain sites and respond to and integrate a variety of hormonal, metabolic, transmitter, and peptide signals involved in the regulation of energy homeostasis and other biological functions. Although it is still uncertain whether daily fluctuations in blood glucose play a specific regulatory role in these physiological functions, it is clear that large decreases in glucose availability stimulate food intake and counterregulatory responses that restore glucose levels to sustain cerebral function. Finally, glucosensing is altered in obesity and after recurrent bouts of hypoglycemia, and this altered sensing may contribute to the adverse outcomes of these conditions. Thus, although much is known, much remains to be learned about the physiological function of brain glucosensing neurons.


Assuntos
Glucose/análise , Glucose/fisiologia , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Potenciais de Ação/fisiologia , Animais , Glucoquinase/metabolismo , Glicólise , Humanos , Canais de Potássio/fisiologia
16.
Diabetes ; 51(7): 2056-65, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12086933

RESUMO

Specialized neurons utilize glucose as a signaling molecule to alter their firing rate. Glucose-excited (GE) neurons increase and glucose-inhibited (GI) neurons reduce activity as ambient glucose levels rise. Glucose-induced changes in the ATP-to-ADP ratio in GE neurons modulate the activity of the ATP-sensitive K(+) channel, which determines the rate of cell firing. The GI glucosensing mechanism is unknown. We postulated that glucokinase (GK), a high-Michaelis constant (K(m)) hexokinase expressed in brain areas containing populations of GE and GI neurons, is the controlling step in glucosensing. Double-label in situ hybridization demonstrated neuron-specific GK mRNA expression in locus ceruleus norepinephrine and in hypothalamic neuropeptide Y, pro-opiomelanocortin, and gamma-aminobutyric acid neurons, but it did not demonstrate this expression in orexin neurons. GK mRNA was also found in the area postrema/nucleus tractus solitarius region by RT-PCR. Intracarotid glucose infusions stimulated c-fos expression in the same areas that expressed GK. At 2.5 mmol/l glucose, fura-2 Ca(2+) imaging of dissociated ventromedial hypothalamic nucleus neurons demonstrated GE neurons whose intracellular Ca(2+) oscillations were inhibited and GI neurons whose Ca(2+) oscillations were stimulated by four selective GK inhibitors. Finally, GK expression was increased in rats with impaired central glucosensing (posthypoglycemia and diet-induced obesity) but was unaffected by a 48-h fast. These data suggest a critical role for GK as a regulator of glucosensing in both GE and GI neurons in the brain.


Assuntos
Encéfalo/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucoquinase/metabolismo , Glucose/farmacologia , Neurônios/fisiologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Artéria Carótida Interna , Genes fos/efeitos dos fármacos , Glucoquinase/genética , Glucose/administração & dosagem , Hibridização In Situ , Infusões Intra-Arteriais , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Obesidade/metabolismo , Ratos , Ratos Sprague-Dawley , Transcrição Gênica/efeitos dos fármacos , Aumento de Peso
17.
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
18.
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
19.
Cell Metab ; 17(4): 607-17, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23562080

RESUMO

The delivery of blood-borne molecules conveying metabolic information to neural networks that regulate energy homeostasis is restricted by brain barriers. The fenestrated endothelium of median eminence microvessels and tight junctions between tanycytes together compose one of these. Here, we show that the decrease in blood glucose levels during fasting alters the structural organization of this blood-hypothalamus barrier, resulting in the improved access of metabolic substrates to the arcuate nucleus. These changes are mimicked by 2-deoxyglucose-induced glucoprivation and reversed by raising blood glucose levels after fasting. Furthermore, we show that VEGF-A expression in tanycytes modulates these barrier properties. The neutralization of VEGF signaling blocks fasting-induced barrier remodeling and significantly impairs the physiological response to refeeding. These results implicate glucose in the control of blood-hypothalamus exchanges through a VEGF-dependent mechanism and demonstrate a hitherto unappreciated role for tanycytes and the permeable microvessels associated with them in the adaptive metabolic response to fasting.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Barreira Hematoencefálica/metabolismo , Epêndima/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Desoxiglucose/farmacologia , Epêndima/citologia , Jejum , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Junções Íntimas/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
20.
Endocrinology ; 151(9): 4270-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20668022

RESUMO

Because rearing rats in large litters (LLs) protects them from becoming obese, we postulated that LL rearing would protect rats selectively bred to develop diet-induced obesity (DIO) from becoming obese by overcoming their inborn central leptin resistance. Male and female DIO rats were raised in normal litters (NLs; 10 pups/dam) or LLs (16 pups/dam) and assessed for anatomical, biochemical, and functional aspects of leptin sensitivity at various ages when fed low-fat chow or a 31% fat high-energy (HE) diet. LL rearing reduced plasma leptin levels by postnatal day 2 (P2) and body weight gain by P8. At P16, LL DIO neonates had increased arcuate nucleus (ARC) binding of leptin to its extracellular receptors and at P28 an associated increase of their agouti-related peptide and alpha-MSH axonal projections to the paraventricular nucleus. Reduced body weight persisted and was associated with increased ARC leptin receptor binding and sensitivity to the anorectic effects of leptin, reduced adiposity, and enhanced insulin sensitivity in LL DIO rats fed chow until 10 wk of age. The enhanced ARC leptin receptor binding and reduced adiposity of LL DIO rats persisted after an additional 5 wk on the HE diet. Female LL DIO rats had similar reductions in weight gain on both chow and HE diet vs. normal litter DIO rats. We postulate that LL rearing enhances DIO leptin sensitivity by lowering plasma leptin levels and thereby increasing leptin receptor availability and that this both enhances the ARC-paraventricular nucleus pathway development and protects them from becoming obese.


Assuntos
Dieta , Leptina/sangue , Tamanho da Ninhada de Vivíparos/fisiologia , Obesidade/fisiopatologia , Adiposidade/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Animais Recém-Nascidos , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal/fisiologia , Cruzamento , Ingestão de Alimentos/fisiologia , Feminino , Idade Gestacional , Imuno-Histoquímica , Lactação/fisiologia , Leptina/metabolismo , Masculino , Obesidade/sangue , Obesidade/etiologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Ligação Proteica , Ratos , Receptores para Leptina/metabolismo , Fatores de Tempo , Desmame , Aumento de Peso/fisiologia , alfa-MSH/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA