Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Clin Invest ; 127(4): 1172-1180, 2017 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-28218618

RESUMO

Neural pathways, especially those in the hypothalamus, integrate multiple nutritional, hormonal, and neural signals, resulting in the coordinated control of body weight balance and glucose homeostasis. Nuclear receptors (NRs) sense changing levels of nutrients and hormones, and therefore play essential roles in the regulation of energy homeostasis. Understanding the role and the underlying mechanisms of NRs in the context of energy balance control may facilitate the identification of novel targets to treat obesity. Notably, NRs are abundantly expressed in the brain, and emerging evidence indicates that a number of these brain NRs regulate multiple aspects of energy balance, including feeding, energy expenditure and physical activity. In this Review we summarize some of the recent literature regarding effects of brain NRs on body weight regulation and discuss mechanisms underlying these effects.


Assuntos
Metabolismo Energético , Exercício Físico , Glucose/metabolismo , Hipotálamo/metabolismo , Obesidade/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Humanos , Obesidade/patologia
2.
Nature ; 519(7541): 45-50, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25707796

RESUMO

Hypothalamic pro-opiomelanocortin (POMC) neurons promote satiety. Cannabinoid receptor 1 (CB1R) is critical for the central regulation of food intake. Here we test whether CB1R-controlled feeding in sated mice is paralleled by decreased activity of POMC neurons. We show that chemical promotion of CB1R activity increases feeding, and notably, CB1R activation also promotes neuronal activity of POMC cells. This paradoxical increase in POMC activity was crucial for CB1R-induced feeding, because designer-receptors-exclusively-activated-by-designer-drugs (DREADD)-mediated inhibition of POMC neurons diminishes, whereas DREADD-mediated activation of POMC neurons enhances CB1R-driven feeding. The Pomc gene encodes both the anorexigenic peptide α-melanocyte-stimulating hormone, and the opioid peptide ß-endorphin. CB1R activation selectively increases ß-endorphin but not α-melanocyte-stimulating hormone release in the hypothalamus, and systemic or hypothalamic administration of the opioid receptor antagonist naloxone blocks acute CB1R-induced feeding. These processes involve mitochondrial adaptations that, when blocked, abolish CB1R-induced cellular responses and feeding. Together, these results uncover a previously unsuspected role of POMC neurons in the promotion of feeding by cannabinoids.


Assuntos
Canabinoides/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Hipotálamo/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Metabolismo Energético/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiologia , Canais Iônicos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Naloxona/farmacologia , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/metabolismo , Resposta de Saciedade/efeitos dos fármacos , Resposta de Saciedade/fisiologia , Proteína Desacopladora 2 , alfa-MSH/metabolismo , beta-Endorfina/metabolismo
3.
Cell Metab ; 18(3): 431-44, 2013 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-24011077

RESUMO

The dogma that life without insulin is incompatible has recently been challenged by results showing the viability of insulin-deficient rodents undergoing leptin monotherapy. Yet, the mechanisms underlying these actions of leptin are unknown. Here, the metabolic outcomes of intracerebroventricular (i.c.v.) administration of leptin in mice devoid of insulin and lacking or re-expressing leptin receptors (LEPRs) only in selected neuronal groups were assessed. Our results demonstrate that concomitant re-expression of LEPRs only in hypothalamic γ-aminobutyric acid (GABA) and pro-opiomelanocortin (POMC) neurons is sufficient to fully mediate the lifesaving and antidiabetic actions of leptin in insulin deficiency. Our analyses indicate that enhanced glucose uptake by brown adipose tissue and soleus muscle, as well as improved hepatic metabolism, underlies these effects of leptin. Collectively, our data elucidate a hypothalamic-dependent pathway enabling life without insulin and hence pave the way for developing better treatments for diseases of insulin deficiency.


Assuntos
Hipotálamo/efeitos dos fármacos , Insulina/metabolismo , Leptina/farmacologia , Neurônios/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/metabolismo , Glucose/análise , Hiperglicemia/tratamento farmacológico , Hiperglicemia/mortalidade , Hipotálamo/metabolismo , Estimativa de Kaplan-Meier , Leptina/uso terapêutico , Fígado/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Neurônios/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo
4.
Nat Med ; 19(9): 1147-52, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23933984

RESUMO

Fibroblast growth factor 21 (FGF21) is a hepatokine that acts as a global starvation signal to modulate fuel partitioning and metabolism and repress growth; however, the site of action of these diverse effects remains unclear. FGF21 signals through a heteromeric cell-surface receptor composed of one of three FGF receptors (FGFR1c, FGFR2c or FGFR3c) in complex with ß-Klotho, a single-pass transmembrane protein that is enriched in metabolic tissues. Here we show that in addition to its known effects on peripheral metabolism, FGF21 increases systemic glucocorticoid levels, suppresses physical activity and alters circadian behavior, which are all features of the adaptive starvation response. These effects are mediated through ß-Klotho expression in the suprachiasmatic nucleus of the hypothalamus and the dorsal vagal complex of the hindbrain. Mice lacking the gene encoding ß-Klotho (Klb) in these regions are refractory to these effects, as well as those on metabolism, insulin and growth. These findings demonstrate a crucial role for the nervous system in mediating the diverse physiologic and pharmacologic actions of FGF21.


Assuntos
Ritmo Circadiano/fisiologia , Metabolismo Energético , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas de Membrana/metabolismo , Sistema Nervoso/metabolismo , Animais , Glucocorticoides/metabolismo , Hipotálamo/metabolismo , Proteínas Klotho , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Inanição , Núcleo Supraquiasmático/metabolismo
5.
Mol Endocrinol ; 27(4): 598-605, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23504956

RESUMO

The differential expression and secretion of the neuropeptide kisspeptin from neurons in the arcuate (Arc) and anteroventral periventricular (AVPV) nuclei of the hypothalamus coordinate the temporal release of pituitary gonadotropins that control the female reproductive cycle. However, the molecular basis for this differential regulation is incompletely understood. Here, we report that liver receptor homolog-1 (LRH-1), a member of the nuclear receptor superfamily, is expressed in kisspeptin neurons in the Arc but not in the AVPV in female mice. LRH-1 binds directly to the kisspeptin (Kiss1) promoter and stimulates Kiss1 transcription. Deletion of LRH-1 from kisspeptin neurons in mice decreased Kiss1 expression in the Arc, leading to reduced plasma FSH levels, dysregulated follicle maturation, and prolongation of the estrous cycle. Conversely, overexpression of LRH-1 in kisspeptin neurons increased Arc Kiss1 expression and plasma FSH concentrations. These studies provide a molecular basis for the differential regulation of basal kisspeptin expression in Arc and AVPV neurons and reveal a prominent role for LRH-1 in hypothalamus in regulating the female reproductive axis.


Assuntos
Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neuropeptídeos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Reprodução , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Feminino , Kisspeptinas/genética , Camundongos , Neurônios/metabolismo , Neuropeptídeos/genética , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/genética
6.
J Clin Invest ; 122(11): 3840-2, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23093788

RESUMO

Obesity has reached epidemic proportions in the United States, and obesity-related illnesses have become a leading preventable cause of death. Childhood obesity is also growing in frequency, and the impact of a lifetime spent in the overweight state is only beginning to emerge in the literature. In this issue of the JCI, Bumaschny et al. used a genetic mouse model to investigate the self-perpetuating nature of obesity and shed some light on why it can become increasingly difficult to lose weight over time.


Assuntos
Tecido Adiposo/fisiopatologia , Adiposidade , Ingestão de Alimentos , Hipotálamo/fisiopatologia , Obesidade/prevenção & controle , Obesidade/fisiopatologia , Animais
7.
J Clin Invest ; 122(3): 1000-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22326958

RESUMO

Leptin action on its receptor (LEPR) stimulates energy expenditure and reduces food intake, thereby lowering body weight. One leptin-sensitive target cell mediating these effects on energy balance is the proopiomelano-cortin (POMC) neuron. Recent evidence suggests that the action of leptin on POMC neurons regulates glucose homeostasis independently of its effects on energy balance. Here, we have dissected the physiological impact of direct leptin action on POMC neurons using a mouse model in which endogenous LEPR expression was prevented by a LoxP-flanked transcription blocker (loxTB), but could be reactivated by Cre recombinase. Mice homozygous for the Lepr(loxTB) allele were obese and exhibited defects characteristic of LEPR deficiency. Reexpression of LEPR only in POMC neurons in the arcuate nucleus of the hypothalamus did not reduce food intake, but partially normalized energy expenditure and modestly reduced body weight. Despite the moderate effects on energy balance and independent of changes in body weight, restoring LEPR in POMC neurons normalized blood glucose and ameliorated hepatic insulin resistance, hyperglucagonemia, and dyslipidemia. Collectively, these results demonstrate that direct leptin action on POMC neurons does not reduce food intake, but is sufficient to normalize glucose and glucagon levels in mice otherwise lacking LEPR.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal , Metabolismo Energético , Feminino , Glucagon/química , Homeostase , Homozigoto , Hipotálamo/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores para Leptina/metabolismo
8.
Cell Metab ; 14(4): 453-65, 2011 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-21982706

RESUMO

Estrogens regulate body weight and reproduction primarily through actions on estrogen receptor-α (ERα). However, ERα-expressing cells mediating these effects are not identified. We demonstrate that brain-specific deletion of ERα in female mice causes abdominal obesity stemming from both hyperphagia and hypometabolism. Hypometabolism and abdominal obesity, but not hyperphagia, are recapitulated in female mice lacking ERα in hypothalamic steroidogenic factor-1 (SF1) neurons. In contrast, deletion of ERα in hypothalamic pro-opiomelanocortin (POMC) neurons leads to hyperphagia, without directly influencing energy expenditure or fat distribution. Further, simultaneous deletion of ERα from both SF1 and POMC neurons causes hypometabolism, hyperphagia, and increased visceral adiposity. Additionally, female mice lacking ERα in SF1 neurons develop anovulation and infertility, while POMC-specific deletion of ERα inhibits negative feedback regulation of estrogens and impairs fertility in females. These results indicate that estrogens act on distinct hypothalamic ERα neurons to regulate different aspects of energy homeostasis and reproduction.


Assuntos
Metabolismo Energético/fisiologia , Receptor alfa de Estrogênio/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Animais , Estradiol/sangue , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/genética , Feminino , Hiperfagia/etiologia , Infertilidade Feminina/etiologia , Masculino , Camundongos , Camundongos Knockout , Obesidade/etiologia , Pró-Opiomelanocortina/metabolismo , Fator Esteroidogênico 1/metabolismo
9.
J Neurosci ; 31(37): 13147-56, 2011 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-21917798

RESUMO

Evidence suggests that the role played by the adipocyte-derived hormone leptin in female reproductive physiology is mediated in part by neurons located within the ventral premammillary nucleus (PMV). Leptin activates PMV neurons; however, the intracellular signaling pathway and channel(s) involved remain undefined. Notably, leptin's excitatory and inhibitory effects within hypothalamic and brainstem nuclei share the intracellular signaling cascade phosphoinositide 3 kinase (PI3K). Therefore, we assessed whether PI3K signaling is required for the acute effect of leptin to alter cellular activity of PMV neurons that express leptin receptors (LepR PMV neurons). Leptin caused a rapid depolarization in the majority of LepR PMV neurons in patch-clamp recordings of hypothalamic slices, while a subset of LepR PMV neurons were hyperpolarized in response to leptin. Data were obtained from both male and female mice and results demonstrate that the acute effect of leptin on LepR PMV neurons was identical for both sexes. Pharmacological inhibition of PI3K prevented the acute leptin-induced change in neuronal activity of LepR PMV neurons, indicating a PI3K-dependent mechanism of leptin action. Similarly, mice with genetically disrupted PI3K signaling in LepR PMV neurons failed to alter cellular activity in response to leptin. Moreover, the leptin-induced depolarization was dependent on a putative TRPC channel. In contrast, the leptin-induced-hyperpolarization required the activation of a putative Katp channel. Collectively, these results suggest that PI3K signaling in LepR PMV neurons is essential for leptin-induced alteration in cellular activity, and these data may suggest a cellular correlate in which leptin contributes to the initiation of reproductive development.


Assuntos
Hipotálamo/fisiologia , Leptina/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/fisiologia , Androstadienos/farmacologia , Animais , Compostos de Boro/farmacologia , Cromonas/farmacologia , Feminino , Genes Reporter/genética , Hipotálamo/efeitos dos fármacos , Hipotálamo/enzimologia , Imidazóis/farmacologia , Canais KATP/fisiologia , Leptina/administração & dosagem , Leptina/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfolinas/farmacologia , Neurônios/fisiologia , Técnicas de Patch-Clamp , Fosfatidilinositol 3-Quinases/biossíntese , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Subunidades Proteicas/biossíntese , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/fisiologia , Wortmanina
10.
J Clin Invest ; 121(6): 2087-93, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21633176

RESUMO

Cloned in 1994, the ob gene encodes the protein hormone leptin, which is produced and secreted by white adipose tissue. Since its discovery, leptin has been found to have profound effects on behavior, metabolic rate, endocrine axes, and glucose fluxes. Leptin deficiency in mice and humans causes morbid obesity, diabetes, and various neuroendocrine anomalies, and replacement leads to decreased food intake, normalized glucose homeostasis, and increased energy expenditure. Here, we provide an update on the most current understanding of leptin-sensitive neural pathways in terms of both anatomical organization and physiological roles.


Assuntos
Metabolismo Energético/fisiologia , Leptina/fisiologia , Receptores para Leptina/fisiologia , Adaptação Fisiológica , Tecido Adiposo Branco/metabolismo , Animais , Peso Corporal/fisiologia , Encéfalo/fisiologia , Modelos Animais de Doenças , Comportamento Alimentar/fisiologia , Glucose/metabolismo , Humanos , Hipotálamo/fisiologia , Leptina/deficiência , Leptina/genética , Camundongos , Camundongos Obesos , Modelos Biológicos , Hipernutrição/fisiopatologia , Pró-Opiomelanocortina/fisiologia , Receptor Tipo 4 de Melanocortina/fisiologia , Receptores para Leptina/deficiência , Receptores para Leptina/genética , Inanição/fisiopatologia , Sistema Nervoso Simpático/fisiologia
11.
Cell Metab ; 13(3): 331-9, 2011 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-21356522

RESUMO

Leptin regulates energy balance and glucose homeostasis. Shortly after leptin was identified, it was established that obesity is commonly associated with leptin resistance, though the molecular mechanisms remain to be identified. To explore potential mechanisms of leptin resistance, we employed organotypic brain slices to identify candidate signaling pathways that negatively regulate leptin sensitivity. We found that elevation of adenosine 3', 5'-monophosphate (cAMP) levels impairs multiple signaling cascades activated by leptin within the hypothalamus. Notably, this effect is independent of protein kinase A activation. In contrast, activation of Epac, a cAMP-regulated guanine nucleotide exchange factor for the small G protein Rap1, was sufficient to impair leptin signaling with concomitant induction of SOCS-3 expression. Epac activation also blunted leptin-induced depolarization of hypothalamic POMC neurons. Finally, central infusion of an Epac activator blunted the anorexigenic actions of leptin. Thus, activation of hypothalamic cAMP-Epac pathway is sufficient to induce multiple indices of leptin resistance.


Assuntos
AMP Cíclico/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Leptina/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Hipotálamo/citologia , Hipotálamo/metabolismo , Camundongos , Neurônios/metabolismo , Fosforilação , Receptores para Leptina/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo
12.
Proc Natl Acad Sci U S A ; 108(11): 4471-6, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21368172

RESUMO

Narcolepsy is caused by a loss of orexin/hypocretin signaling, resulting in chronic sleepiness, fragmented non-rapid eye movement sleep, and cataplexy. To identify the neuronal circuits underlying narcolepsy, we produced a mouse model in which a loxP-flanked gene cassette disrupts production of the orexin receptor type 2 (OX2R; also known as HCRTR2), but normal OX2R expression can be restored by Cre recombinase. Mice lacking OX2R signaling had poor maintenance of wakefulness indicative of sleepiness and fragmented sleep and lacked any electrophysiological response to orexin-A in the wake-promoting neurons of the tuberomammillary nucleus. These defects were completely recovered by crossing them with mice that express Cre in the female germline, thus globally deleting the transcription-disrupter cassette. Then, by using an adeno-associated viral vector coding for Cre recombinase, we found that focal restoration of OX2R in neurons of the tuberomammillary nucleus and adjacent parts of the posterior hypothalamus completely rescued the sleepiness of these mice, but their fragmented sleep was unimproved. These observations demonstrate that the tuberomammillary region plays an essential role in the wake-promoting effects of orexins, but orexins must stabilize sleep through other targets.


Assuntos
Antígenos de Superfície/metabolismo , Hipotálamo/metabolismo , Narcolepsia/prevenção & controle , Narcolepsia/fisiopatologia , Receptores de Superfície Celular/metabolismo , Sono/fisiologia , Animais , Dependovirus/genética , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Feminino , Região Hipotalâmica Lateral/efeitos dos fármacos , Região Hipotalâmica Lateral/patologia , Região Hipotalâmica Lateral/fisiopatologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Integrases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Camundongos , Camundongos Transgênicos , Microinjeções , Narcolepsia/patologia , Neuropeptídeos/farmacologia , Receptores de Orexina , Orexinas , Transdução de Sinais/efeitos dos fármacos , Sono/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Vigília/efeitos dos fármacos , Vigília/fisiologia
14.
J Clin Invest ; 121(1): 355-68, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21183787

RESUMO

Studies in humans and rodents indicate that a minimum amount of stored energy is required for normal pubertal development. The adipocyte-derived hormone leptin is a key metabolic signal to the neuroendocrine reproductive axis. Humans and mice lacking leptin or the leptin receptor (LepR) (ob/ob and db/db mice, respectively) are infertile and fail to enter puberty. Leptin administration to leptin-deficient subjects and ob/ob mice induces puberty and restores fertility, but the exact site or sites of leptin action are unclear. Here, we found that genetic deletion of LepR selectively from hypothalamic Kiss1 neurons in mice had no effect on puberty or fertility, indicating that direct leptin signaling in Kiss1 neurons is not required for these processes. However, bilateral lesions of the ventral premammillary nucleus (PMV) of ob/ob mice blunted the ability of exogenous leptin to induce sexual maturation. Moreover, unilateral reexpression of endogenous LepR in PMV neurons was sufficient to induce puberty and improve fertility in female LepR-null mice. This LepR reexpression also normalized the increased hypothalamic GnRH content characteristic of leptin-signaling deficiency. These data suggest that the PMV is a key site for leptin's permissive action at the onset of puberty and support the hypothesis that the multiple actions of leptin to control metabolism and reproduction are anatomically dissociated.


Assuntos
Hipotálamo/metabolismo , Leptina/metabolismo , Proteínas/metabolismo , Maturidade Sexual/fisiologia , Animais , Sequência de Bases , Feminino , Fertilidade/genética , Fertilidade/fisiologia , Expressão Gênica , Humanos , Kisspeptinas , Leptina/deficiência , Leptina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Camundongos Transgênicos , Neurônios/metabolismo , Gravidez , Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores para Leptina/deficiência , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Maturidade Sexual/genética , Transdução de Sinais
15.
Nat Neurosci ; 13(12): 1457-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21037584

RESUMO

Mice lacking 5-HT 2C receptors (5-HT(2C)Rs) displayed hepatic insulin resistance, a phenotype normalized by re-expression of 5-HT(2C)Rs only in pro-opiomelanocortin (POMC) neurons. 5-HT(2C)R deficiency also abolished the anti-diabetic effects of meta-chlorophenylpiperazine (a 5-HT(2C)R agonist); these effects were restored when 5-HT(2C)Rs were re-expressed in POMC neurons. Our findings indicate that 5-HT(2C)Rs expressed by POMC neurons are physiologically relevant regulators of insulin sensitivity and glucose homeostasis in the liver.


Assuntos
Regulação da Expressão Gênica , Resistência à Insulina/fisiologia , Fígado/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/biossíntese , Receptor 5-HT2C de Serotonina/biossíntese , Animais , Tronco Encefálico/metabolismo , Glucose/metabolismo , Homeostase/fisiologia , Hipotálamo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neurônios/fisiologia , Pró-Opiomelanocortina/fisiologia , Receptor 5-HT2C de Serotonina/genética , Receptor 5-HT2C de Serotonina/fisiologia
16.
J Comp Neurol ; 518(11): 2090-108, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20394060

RESUMO

Tracing the axonal projections of selected neurons is labor intensive and inherently limited by currently available neuroanatomical methods. We developed an adeno-associated virus (AAV) that can be used for efficiently tracing identified neuronal populations. The virus encodes a humanized Renilla green fluorescent protein (hrGFP) that is transcriptionally silenced by a neo cassette flanked by LoxH/LoxP sites (AAV-lox-Stop-hrGFP). Thus, hrGFP is expressed only in neurons with Cre recombinase activity. To demonstrate the utility of this approach, the virus was injected unilaterally into the dorsomedial hypothalamus (DMH) of mice that express Cre in neurons expressing the leptin receptor. Animals with DMH injections showed robust hrGFP expression in DMH neurons, as visualized by its endogenous fluorescence or following immunolabeling. We found that hrGFP was expressed in approximately one-third to one-half of Cre-expressing neurons at the site of injection, but not in non-Cre-expressing neurons. The expression of GFP allowed us to identify the projection fields of DMH leptin-responsive neurons. Our results show hrGFP-positive axonal projections and terminals in the paraventricular nucleus of the hypothalamus, arcuate nucleus, preoptic area, bed nucleus of the stria terminalis, paraventricular thalamus, periaqueductal gray, and precoeruleus. The aforementioned pattern of projections was similar to DMH projections determined by injections of biotinylated dextran amine in the mouse DMH. Interestingly, some hrGFP-positive terminals were seen contacting the ependymal layer of the third and fourth ventricles. In summary, this approach is an effective tool for tracing axonal projections of chemically identified neurons, including leptin-responsive neurons.


Assuntos
Vias Eferentes/anatomia & histologia , Hipotálamo/citologia , Leptina/metabolismo , Neurônios , Coloração e Rotulagem/métodos , Animais , Dependovirus/genética , Dependovirus/metabolismo , Vias Eferentes/fisiologia , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Integrases/metabolismo , Camundongos , Neurônios/citologia , Neurônios/fisiologia
17.
Cell Metab ; 11(4): 286-97, 2010 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-20374961

RESUMO

Circulating leptin and insulin convey information regarding energy stores to the central nervous system, particularly the hypothalamus. Hypothalamic pro-opiomelanocortin (POMC) neurons regulate energy balance and glucose homeostasis and express leptin and insulin receptors. However, the physiological significance of concomitant leptin and insulin action on POMC neurons remains to be established. Here, we show that mice lacking both leptin and insulin receptors in POMC neurons (Pomc-Cre, Lepr(flox/flox) IR(flox/flox) mice) display systemic insulin resistance, which is distinct from the single deletion of either receptor. In addition, Pomc-Cre, Lepr(flox/flox) IR(flox/flox) female mice display elevated serum testosterone levels and ovarian abnormalities, resulting in reduced fertility. We conclude that direct action of insulin and leptin on POMC neurons is required to maintain normal glucose homeostasis and reproductive function.


Assuntos
Glicemia/metabolismo , Fertilidade/fisiologia , Homeostase/fisiologia , Hipotálamo/metabolismo , Resistência à Insulina/fisiologia , Insulina/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , Análise de Variância , Animais , Feminino , Hipotálamo/citologia , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Pró-Opiomelanocortina/metabolismo , Testosterona/sangue
18.
Mol Endocrinol ; 24(6): 1240-50, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20339005

RESUMO

The ventromedial hypothalamic nucleus (VMH) regulates a variety of homeostatic processes including female sexual behavior and reproduction. In the current study, we assessed the roles of steroidogenic factor 1 (SF-1) on reproductive function in the VMH using central nervous system-specific SF-1 knockout (SF-1 KO(nCre;F/-)) mice. Here we show that SF-1 KO(nCre;F/-) females exhibited marked impairment in female reproduction. Although male mice appeared to be normal in all aspects studied, including sexual behavior, SF-1 KO(nCre;F/-) females showed infertility or subfertility. Although adult SF-1 KO(nCre;F/-) females showed decreased or lacked corpora lutea, exogenous administration of gonadotropins induced the formation of multiple corpora lutea and induced normal ovulation, demonstrating that the ovaries are functionally intact. In addition, SF-1 KO(nCre;F/-) females stimulated with a synthetic GnRH agonist after priming exhibited markedly reduced LH secretion compared with wild-type littermates, arguing that disorganization in and around the VMH caused by SF-1 ablation interferes with the GnRH priming process or gonadotrope LH capacity. Furthermore, the SF-1 KO(nCre;F/-) females primed with estrogen benzoate and progesterone failed to induce steroid receptors around the VMH, consistent with impaired lordosis behavior in the SF-1 KO(nCre;F/-) females. Collectively, our results highlight that SF-1 in the VMH plays crucial roles in regulation of female reproductive function, presumably by organizing a precise neuronal connection and communication in and around the VMH.


Assuntos
Sistema Nervoso Central/metabolismo , Reprodução/fisiologia , Fator Esteroidogênico 1/metabolismo , Animais , Estradiol/sangue , Ciclo Estral/metabolismo , Feminino , Fertilidade/fisiologia , Hormônio Foliculoestimulante/sangue , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Hipotálamo/patologia , Integrases/metabolismo , Hormônio Luteinizante/sangue , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Ovário/metabolismo , Ovário/patologia , Ovulação/sangue , Progesterona/sangue , Comportamento Sexual Animal
19.
J Neurosci ; 30(10): 3803-12, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20220015

RESUMO

Single-minded 1 (SIM1) mutations are one of the few known causes of nonsyndromic monogenic obesity in both humans and mice. Although the role of Sim1 in the formation of the hypothalamus has been described, its postdevelopmental, physiological functions have not been well established. Here we demonstrate that postnatal CNS deficiency of Sim1 is sufficient to cause hyperphagic obesity. We conditionally deleted Sim1 after birth using CaMKII-Cre (alpha-calcium/calmodulin-dependent protein kinase II-Cre) lines to recombine a floxed Sim1 allele. Conditional Sim1 heterozygotes phenocopied germ line Sim1 heterozygotes, displaying hyperphagic obesity and increased length. We also generated viable conditional Sim1 homozygotes, demonstrating that adult Sim1 expression is not essential for mouse or neuron survival and revealing a dosage-dependent effect of Sim1 on obesity. Using stereological cell counting, we showed that the phenotype of both germ line heterozygotes and conditional Sim1 homozygotes was not attributable to global hypocellularity of the paraventricular nucleus (PVN) of the hypothalamus. We also used retrograde tract tracing to demonstrate that the PVN of germ line heterozygous mice projects normally to the dorsal vagal complex and the median eminence. Finally, we showed that conditional Sim1 homozygotes and germ line Sim1 heterozygotes exhibit a remarkable decrease in hypothalamic oxytocin (Oxt) and PVN melanocortin 4 receptor (Mc4r) mRNA. These results demonstrate that the role of Sim1 in feeding regulation is not limited to formation of the PVN or its projections and that the hyperphagic obesity in Sim1-deficient mice may be attributable to changes in the leptin-melanocortin-oxytocin pathway.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Regulação da Expressão Gênica no Desenvolvimento , Hiperfagia/genética , Obesidade/genética , Ocitocina/antagonistas & inibidores , Receptor Tipo 4 de Melanocortina/antagonistas & inibidores , Proteínas Repressoras/deficiência , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Ingestão de Alimentos/genética , Feminino , Inativação Gênica , Hiperfagia/metabolismo , Hiperfagia/patologia , Hiperfagia/fisiopatologia , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Ocitocina/biossíntese , Ocitocina/genética , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/patologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Receptor Tipo 4 de Melanocortina/biossíntese , Receptor Tipo 4 de Melanocortina/genética , Proteínas Repressoras/genética , Reprodutibilidade dos Testes , Transdução de Sinais/genética
20.
J Neurosci ; 30(7): 2472-9, 2010 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-20164331

RESUMO

Acute leptin administration results in a depolarization and concomitant increase in the firing rate of a subpopulation of arcuate proopiomelanocortin (POMC) cells. This rapid activation of POMC cells has been implicated as a cellular correlate of leptin effects on energy balance. In contrast to leptin, insulin inhibits the activity of some POMC neurons. Several studies have described a "cross talk" between leptin and insulin within the mediobasal hypothalamus via the intracellular enzyme, phosphoinositol-3-kinase (PI3K). Interestingly, both insulin and leptin regulate POMC cellular activity by activation of PI3K; however, it is unclear whether leptin and insulin effects are observed in similar or distinct populations of POMC cells. We therefore used dual label immunohistochemistry/in situ hybridization and whole-cell patch-clamp electrophysiology to map insulin and leptin responsive arcuate POMC neurons. Leptin-induced Fos activity within arcuate POMC neurons was localized separate from POMC neurons that express insulin receptor. Moreover, acute responses to leptin and insulin were largely segregated in distinct subpopulations of POMC cells. Collectively, these data suggest that cross talk between leptin and insulin occurs within a network of cells rather than within individual POMC neurons.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Insulina/farmacologia , Leptina/farmacologia , Neurônios/classificação , Neurônios/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Proteínas de Fluorescência Verde/genética , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Oncogênicas v-fos/genética , Proteínas Oncogênicas v-fos/metabolismo , Técnicas de Patch-Clamp/métodos , Pró-Opiomelanocortina/genética , RNA Mensageiro/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA