Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 16(13)2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-39001392

RESUMO

Rapidly proliferative processes in mammalian tissues including tumorigenesis and embryogenesis rely on the glycolytic pathway for energy and biosynthetic precursors. The enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) plays an important regulatory role in glycolysis by activating the key rate-limiting glycolytic enzyme, 6-phosphofructo-1-kinase (PFK-1). We have previously determined that decreased PFKFB3 expression reduced glycolysis and growth in transformed cells in vitro and suppressed xenograft growth in vivo. In earlier studies, we created a constitutive knockout mouse to interrogate the function of PFKFB3 in vivo but failed to generate homozygous offspring due to the requirement for PFKFB3 for embryogenesis. We have now developed a novel transgenic mouse model that exhibits inducible homozygous pan-tissue Pfkfb3 gene deletion (Pfkfb3fl/fl). We have induced Pfkfb3 genomic deletion in these mice and found that it effectively decreased PFKFB3 expression and activity. To evaluate the functional consequences of Pfkfb3 deletion in vivo, we crossed Cre-bearing Pfkfb3fl/fl mice with oncogene-driven tumor models and found that Pfkfb3 deletion markedly decreased their glucose uptake and growth. In summary, our studies reveal a critical regulatory function for PFKFB3 in glycolysis and tumorigenesis in vivo and characterize an effective and powerful model for further investigation of its role in multiple biological processes.

2.
Neurol Int ; 14(1): 75-88, 2022 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-35076581

RESUMO

Hypoactive sexual desire disorder (HSDD) is a persistent deficiency or absence of sexual fantasies and desire resulting in significant distress or interpersonal difficulty. Women with this disorder may display a lack of motivation for sexual activity, reduced responsiveness to erotic cues, a loss of interest during sexual activity, and avoidance of situations that could lead to sexual activity. The pathophysiology of HSDD is thought to be centered around inhibitory and excitatory hormones, neurotransmitters, and specific brain anatomy. Due to the multifactorial nature of HSDD, treatment can be complex and must attempt to target the biological and psychosocial aspects of the disorder. Bremelanotide is a melanocortin receptor agonist and has been recently approved by the FDA to treat HSDD. Bremelanotide is administered intranasally or as a subcutaneous injection. The recommended dosage of bremelanotide is 1.75 mg injected subcutaneously in the abdomen or thigh at least 45 min before sexual activity. Studies showed improvements in desire, arousal, and orgasm scores when 1.75 mg of bremelanotide was administered before sexual activity compared to a placebo. Bremelanotide is a promising way to treat HSDD.

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.
Diabetes ; 59(2): 519-28, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19934009

RESUMO

OBJECTIVE: The response of ventromedial hypothalamic (VMH) glucose-inhibited neurons to decreased glucose is impaired under conditions where the counterregulatory response (CRR) to hypoglycemia is impaired (e.g., recurrent hypoglycemia). This suggests a role for glucose-inhibited neurons in the CRR. We recently showed that decreased glucose increases nitric oxide (NO) production in cultured VMH glucose-inhibited neurons. These in vitro data led us to hypothesize that NO release from VMH glucose-inhibited neurons is critical for the CRR. RESEARCH DESIGN AND METHODS: The CRR was evaluated in rats and mice in response to acute insulin-induced hypoglycemia and hypoglycemic clamps after modulation of brain NO signaling. The glucose sensitivity of ventromedial nucleus glucose-inhibited neurons was also assessed. RESULTS: Hypoglycemia increased hypothalamic constitutive NO synthase (NOS) activity and neuronal NOS (nNOS) but not endothelial NOS (eNOS) phosphorylation in rats. Intracerebroventricular and VMH injection of the nonselective NOS inhibitor N(G)-monomethyl-l-arginine (l-NMMA) slowed the recovery to euglycemia after hypoglycemia. VMH l-NMMA injection also increased the glucose infusion rate (GIR) and decreased epinephrine secretion during hyperinsulinemic/hypoglycemic clamp in rats. The GIR required to maintain the hypoglycemic plateau was higher in nNOS knockout than wild-type or eNOS knockout mice. Finally, VMH glucose-inhibited neurons were virtually absent in nNOS knockout mice. CONCLUSIONS: We conclude that VMH NO production is necessary for glucose sensing in glucose-inhibited neurons and full generation of the CRR to hypoglycemia. These data suggest that potentiating NO signaling may improve the defective CRR resulting from recurrent hypoglycemia in patients using intensive insulin therapy.


Assuntos
Hipoglicemia/diagnóstico , Hipotálamo/enzimologia , Óxido Nítrico/biossíntese , Núcleo Hipotalâmico Ventromedial/fisiologia , Animais , Catecolaminas/sangue , Epinefrina/metabolismo , Glucagon/sangue , Técnica Clamp de Glucose , Homeostase , Hiperinsulinismo/enzimologia , Hiperinsulinismo/fisiopatologia , Hipoglicemia/induzido quimicamente , Insulina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/metabolismo , Ratos , Ratos Sprague-Dawley
5.
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
6.
Am J Physiol Regul Integr Comp Physiol ; 295(5): R1446-54, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18753263

RESUMO

A limiting factor to the clinical management of diabetes is iatrogenic hypoglycemia. With multiple hypoglycemic episodes, the collective neuroendocrine response that restores euglycemia is impaired. In our animal model of recurrent hypoglycemia (RH), neuroendocrine deficits are accompanied by a decrease in medial hypothalamic activation. Here we tested the hypothesis that the medial hypothalamus may exhibit unique changes in the expression of regulatory proteins in response to RH. We report that expression of the immediate early gene FosB is increased in medial hypothalamic nuclei, anterior hypothalamus, and posterior paraventricular nucleus of the thalamus (THPVN) of the thalamus following RH. We identified the hypothalamic PVN, a key autonomic output site, among the regions expressing FosB. To identify the subtype(s) of neuronal populations that express FosB, we screened candidate neuropeptides of the PVN for coexpression using dual fluorescence immunohistochemistry. Among the neuropeptides analyzed [including oxytocin, vasopressin, thyrotropin-releasing hormone, and corticotropin-releasing factor (CRF)], FosB was only identified in CRF-positive neurons. Inhibitory gamma-aminobutyric acid-positive processes appear to impinge on these FosB-expressing neurons. Finally, we observed a significant decrease in the presynaptic marker synaptophysin within the PVN of RH-treated vs. saline-treated rats, suggesting that rapid alterations of synaptic morphology may occur in association with RH. Collectively, these data suggest that RH stress triggers cellular changes that support synaptic plasticity, in specific neuroanatomical sites, which may contribute to the development of hypoglycemia-associated autonomic failure.


Assuntos
Hipoglicemia/metabolismo , Hipotálamo/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Sinaptofisina/biossíntese , Hormônio Adrenocorticotrópico/metabolismo , Animais , Glicemia/metabolismo , Western Blotting , Epinefrina/metabolismo , Imunofluorescência , Glucagon/metabolismo , Hidrocortisona/metabolismo , Hipotálamo Médio/metabolismo , Imuno-Histoquímica , Masculino , Neurônios/fisiologia , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Ratos Wistar , Recidiva , Hormônio Liberador de Tireotropina/metabolismo , Vasopressinas/metabolismo , Ácido gama-Aminobutírico/metabolismo
7.
Am J Physiol Endocrinol Metab ; 294(5): E853-60, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18334609

RESUMO

Selective serotonin reuptake inhibitors (SSRIs) are widely prescribed for patients with comorbid diabetes and depression. Clinical case studies in diabetic patients, however, suggest that SSRI therapy may exacerbate hypoglycemia. We hypothesized that SSRIs might increase the risk of hypoglycemia by impairing hormonal counterregulatory responses (CRR). We evaluated the effect of the SSRI sertraline on hormonal CRR to single or recurrent hypoglycemia in nondiabetic rats. Since there are time-dependent effects of SSRIs on serotonin neurotransmission that correspond with therapeutic action, we evaluated the effect of 6- or 20-day sertraline treatment on hypoglycemia CRR. We found that 6-day sertraline (SERT) treatment specifically enhanced the epinephrine response to a single bout of hypoglycemia vs. vehicle (VEH)-treated rats (t = 120: VEH, 2,573 +/- 448 vs. SERT, 4,202 +/- 545 pg/ml, P < 0.05). In response to recurrent hypoglycemia, VEH-treated rats exhibited the expected impairment in epinephrine secretion (t = 60: 678 +/- 73 pg/ml) vs. VEH-treated rats experiencing first-time hypoglycemia (t = 60: 2,081 +/- 436 pg/ml, P < 0.01). SERT treatment prevented the impaired epinephrine response in recurrent hypoglycemic rats (t = 60: 1,794 +/- 276 pgl/ml). In 20-day SERT-treated rats, epinephrine, norepinephrine, and glucagon CRR were all significantly elevated above VEH-treated controls in response to hypoglycemia. Similarly to 6-day SERT treatment, 20-day SERT treatment rescued the impaired epinephrine response in recurrent hypoglycemic rats. Our data demonstrate that neither 6- nor 20-day sertraline treatment impaired hormonal CRR to hypoglycemia in nondiabetic rats. Instead, sertraline treatment resulted in an enhancement of hypoglycemia CRR and prevented the impaired adrenomedullary response normally observed in recurrent hypoglycemic rats.


Assuntos
Hipoglicemia/fisiopatologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Sertralina/farmacologia , Hormônio Adrenocorticotrópico/sangue , Animais , Sistema Nervoso Autônomo/efeitos dos fármacos , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Corticosterona/sangue , Ingestão de Alimentos/efeitos dos fármacos , Epinefrina/sangue , Glucagon/sangue , Masculino , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/fisiologia , Norepinefrina/sangue , Ratos , Ratos Sprague-Dawley , Estimulação Química
8.
Brain Res ; 1194: 65-72, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18191818

RESUMO

The posterior paraventricular nucleus of the thalamus (THPVP) has been identified as a forebrain region that modulates the central nervous system (CNS) response to recurrent experiences of stressors. The THPVP is activated in response to a single (SH) or recurrent (RH) experience of the metabolic stress of hypoglycemia. In this study, we evaluated whether temporary experimental inactivation of the THPVP would modify the neuroendocrine response to SH or RH. Infusion of lidocaine (LIDO) or vehicle had no effect on the neuroendocrine response to SH, comparable to findings with other stressors. THPVP vehicle infusion concomitant with RH resulted in a prevention of the expected impairment of neuroendocrine responses, relative to SH. LIDO infusion with RH resulted in significantly decreased glucagon and sympathoadrenal responses, relative to SH. These results suggest that the THPVP may contribute to the sympathoadrenal stimulation induced by hypoglycemia; and emphasizes that the THPVP is a forebrain region that may contribute to the coordinated CNS response to metabolic stressors.


Assuntos
Glucagon/metabolismo , Hipoglicemia/fisiopatologia , Núcleos da Linha Média do Tálamo/fisiologia , Sistemas Neurossecretores/fisiologia , Corticosteroides/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Análise de Variância , Anestésicos Locais/farmacologia , Animais , Glicemia , Epinefrina/metabolismo , Hipoglicemia/induzido quimicamente , Insulina , Lidocaína/farmacologia , Masculino , Núcleos da Linha Média do Tálamo/efeitos dos fármacos , Sistemas Neurossecretores/efeitos dos fármacos , Norepinefrina/metabolismo , Ratos , Ratos Wistar , Fatores de Tempo
9.
Am J Physiol Regul Integr Comp Physiol ; 294(3): R784-92, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18094065

RESUMO

Antecedent insulin-induced hypoglycemia (IIH) reduces adrenomedullary responses (AMR) to subsequent bouts of hypoglycemia. The ventromedial hypothalamus [VMH: arcuate (ARC) + ventromedial nuclei] contains glucosensing neurons, which are thought to be mediators of these AMR. Since type 1 diabetes mellitus often begins in childhood, we used juvenile (4- to 5-wk-old) rats to demonstrate that a single bout of IIH (5 U/kg sc) reduced plasma glucose by 24% and peak epinephrine by 59% 1 day later. This dampened AMR was associated with 46% higher mRNA for VMH glucokinase, a key mediator of neuronal glucosensing. Compared with neurons from saline-injected rats, ventromedial nucleus glucose-excited neurons from insulin-injected rats demonstrated a leftward shift in their glucose responsiveness (EC50 = 0.45 and 0.10 mmol/l for saline and insulin, respectively, P = 0.05) and a 31% higher maximal activation by glucose (P = 0.05), although this maximum occurred at a higher glucose concentration (saline, 0.7 vs. insulin, 1.5 mmol/l). Although EC50 values did not differ, ARC glucose-excited neurons had 19% higher maximal activation, which occurred at a lower glucose concentration in insulin- than saline-injected rats (saline, 2.5 vs. insulin, 1.5 mmol/l). In addition, ARC glucose-inhibited neurons from insulin-injected rats were maximally inhibited at a fivefold lower glucose concentration (saline, 2.5 vs. insulin, 0.5 mmol/l), although this inhibition declined at >0.5 mmol/l glucose. These data suggest that the increased VMH glucokinase after IIH may contribute to the increased responsiveness of VMH glucosensing neurons to glucose and the associated blunting of the AMR.


Assuntos
Glucose/metabolismo , Glucose/farmacologia , Hipoglicemia/fisiopatologia , Neurônios/efeitos dos fármacos , Núcleo Hipotalâmico Ventromedial/fisiopatologia , Medula Suprarrenal/efeitos dos fármacos , Medula Suprarrenal/fisiologia , Animais , Cálcio/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Glucoquinase/biossíntese , Glucoquinase/genética , Hexoquinase/biossíntese , Hexoquinase/genética , Hipoglicemia/induzido quimicamente , Hipoglicemiantes , Insulina , Masculino , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Núcleo Hipotalâmico Ventromedial/citologia , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos
10.
Diabetes ; 56(1): 217-23, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17192485

RESUMO

Recurrent hypoglycemia impairs hormonal counterregulatory responses (CRRs) to further bouts of hypoglycemia. The hypothalamus and hindbrain are both critical for sensing hypoglycemia and triggering CRRs. Hypothalamic glucose sensing sites are implicated in the pathogenesis of defective CRRs; however, the contribution of hindbrain glucose sensing has not been elucidated. Using a rat model, we compared the effect of antecedent glucoprivation targeting hindbrain or hypothalamic glucose sensing sites with the effect of antecedent recurrent hypoglycemia on CRR to hypoglycemia induced 24 h later. Recurrent hypoglycemia decreased sympathoadrenal (1,470 +/- 325 vs. 3,811 +/- 540 pg/ml in controls [t = 60 min], P = 0.001) and glucagon secretion (222 +/- 43 vs. 494 +/- 56 pg/ml in controls [t = 60]), P = 0.003) in response to hypoglycemia. Antecedent 5-thio-glucose (5TG) injected into the hindbrain did not impair sympathoadrenal (3,806 +/- 344 pg/ml [t = 60]) or glucagon (513 +/- 56 pg/ml [t = 60]) responses to subsequent hypoglycemia. However, antecedent 5TG delivered into the third ventricle was sufficient to blunt CRRs to hypoglycemia. These results show that hindbrain glucose sensing is not involved in the development of defective CRRs. However, neural substrates surrounding the third ventricle are particularly sensitive to glucoprivic stimulation and may contribute importantly to the development of defective CRRs.


Assuntos
Glucose/deficiência , Glucose/metabolismo , Hipoglicemia/fisiopatologia , Rombencéfalo/metabolismo , Hormônio Adrenocorticotrópico/sangue , Animais , Glicemia/metabolismo , Ventrículos Cerebrais/metabolismo , Corticosterona/sangue , Epinefrina/sangue , Glucagon/sangue , Homeostase , Masculino , Norepinefrina/sangue , Ratos , Ratos Sprague-Dawley , Recidiva
11.
Physiol Behav ; 87(4): 700-6, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16492385

RESUMO

Prior exposure to hypoglycemia impairs neuroendocrine counterregulatory responses (CRR) during subsequent hypoglycemia. Defective CRR to hypoglycemia is a component of the clinical syndrome hypoglycemia-associated autonomic failure (HAAF). Hypoglycemia also potently stimulates food intake, an important behavioral CRR. Because the increased feeding response to hypoglycemia is behavioral and not hormonal, we hypothesized that it may be regulated differently with recurrent bouts of hypoglycemia. To test this hypothesis, we simultaneously evaluated neuroendocrine CRR and food intake in rats experiencing one or three episodes of insulin-induced hypoglycemia. As expected, recurrent hypoglycemia significantly reduced neuroendocrine hypoglycemic CRR. Epinephrine (E), norepinephrine (NE) and glucagon responses 120 min after insulin injection were significantly reduced in recurrent hypoglycemic rats, relative to rats experiencing hypoglycemia for the first time. Despite these neuroendocrine impairments, food intake was significantly elevated above baseline saline intake whether rats were experiencing a first (hypoglycemia: 3.4+/-0.4 g vs. saline: 0.94+/-0.3 g, P<0.05) or third hypoglycemic episode (hypoglycemia: 3.8+/-0.3 g vs. saline: 1.2+/-0.3 g, P<0.05). These findings demonstrate that food intake elicited in response to hypoglycemia is not impaired as a result of recurrent hypoglycemia. Thus, neuroendocrine and behavioral (stimulation of food intake) CRR are differentially regulated by recurrent hypoglycemia experience.


Assuntos
Catecolaminas/sangue , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Hiperfagia/etiologia , Hipoglicemia/complicações , Adaptação Fisiológica , Hormônio Adrenocorticotrópico/sangue , Análise de Variância , Animais , Glicemia/fisiologia , Corticosterona/sangue , Modelos Animais de Doenças , Ingestão de Alimentos/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Glucagon/sangue , Fome/efeitos dos fármacos , Fome/fisiologia , Hiperfagia/sangue , Hipoglicemia/sangue , Hipoglicemia/induzido quimicamente , Insulina , Masculino , Ratos , Ratos Sprague-Dawley
12.
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
13.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA