Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(18): e2322692121, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38652744

RESUMO

Food intake and energy balance are tightly regulated by a group of hypothalamic arcuate neurons expressing the proopiomelanocortin (POMC) gene. In mammals, arcuate-specific POMC expression is driven by two cis-acting transcriptional enhancers known as nPE1 and nPE2. Because mutant mice lacking these two enhancers still showed hypothalamic Pomc mRNA, we searched for additional elements contributing to arcuate Pomc expression. By combining molecular evolution with reporter gene expression in transgenic zebrafish and mice, here, we identified a mammalian arcuate-specific Pomc enhancer that we named nPE3, carrying several binding sites also present in nPE1 and nPE2 for transcription factors known to activate neuronal Pomc expression, such as ISL1, NKX2.1, and ERα. We found that nPE3 originated in the lineage leading to placental mammals and remained under purifying selection in all mammalian orders, although it was lost in Simiiformes (monkeys, apes, and humans) following a unique segmental deletion event. Interestingly, ablation of nPE3 from the mouse genome led to a drastic reduction (>70%) in hypothalamic Pomc mRNA during development and only moderate (<33%) in adult mice. Comparison between double (nPE1 and nPE2) and triple (nPE1, nPE2, and nPE3) enhancer mutants revealed the relative contribution of nPE3 to hypothalamic Pomc expression and its importance in the control of food intake and adiposity in male and female mice. Altogether, these results demonstrate that nPE3 integrates a tripartite cluster of partially redundant enhancers that originated upon a triple convergent evolutionary process in mammals and that is critical for hypothalamic Pomc expression and body weight homeostasis.


Assuntos
Peso Corporal , Ingestão de Alimentos , Elementos Facilitadores Genéticos , Hipotálamo , Pró-Opiomelanocortina , Peixe-Zebra , Animais , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/genética , Camundongos , Hipotálamo/metabolismo , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Feminino , Masculino , Camundongos Transgênicos , Humanos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Mamíferos/metabolismo , Mamíferos/genética
2.
Elife ; 112022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-35044906

RESUMO

Proopiomelanocortin (POMC) neurons of the hypothalamic arcuate nucleus are essential to regulate food intake and energy balance. However, the ontogenetic transcriptional programs that specify the identity and functioning of these neurons are poorly understood. Here, we use single-cell RNA-sequencing (scRNA-seq) to define the transcriptomes characterizing Pomc-expressing cells in the developing hypothalamus and translating ribosome affinity purification with RNA-sequencing (TRAP-seq) to analyze the subsequent translatomes of mature POMC neurons. Our data showed that Pomc-expressing neurons give rise to multiple developmental pathways expressing different levels of Pomc and unique combinations of transcription factors. The predominant cluster, featured by high levels of Pomc and Prdm12 transcripts, represents the canonical arcuate POMC neurons. Additional cell clusters expressing medium or low levels of Pomc mature into different neuronal phenotypes featured by distinct sets of transcription factors, neuropeptides, processing enzymes, cell surface, and nuclear receptors. We conclude that the genetic programs specifying the identity and differentiation of arcuate POMC neurons are diverse and generate a heterogeneous repertoire of neuronal phenotypes early in development that continue to mature postnatally.


Assuntos
Hipotálamo/metabolismo , Neurônios/metabolismo , Fenótipo , Transcriptoma , Animais , Camundongos , Pró-Opiomelanocortina/metabolismo , RNA-Seq , Análise de Célula Única
3.
Mol Metab ; 37: 100985, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32311511

RESUMO

OBJECTIVE: Hypothalamic arcuate proopiomelanocortin (Arc-POMC) neurons are involved in different physiological processes such as the regulation of energy balance, glucose homeostasis, and stress-induced analgesia. Since these neurons heterogeneously express different biological markers and project to many hypothalamic and extrahypothalamic areas, it is proposed that Arc-POMC neurons could be classified into different subpopulations having diverse physiological roles. The aim of the present study was to characterize the contribution of the subpopulation of Arc-POMC neurons cosecreting gamma-aminobutyric acid (GABA) neurotransmitter in the control of energy balance. METHODS: Arc-Pomc expression restricted to GABAergic-POMC neurons was achieved by crossing a reversible Pomc-deficient mouse line (arcPomc-) with a tamoxifen-inducible Gad2-CreER transgenic line. Pomc expression was rescued in the compound arcPomc-/-:Gad2-CreER female and male mice by tamoxifen treatment at postnatal days 25 (P25) or 60 (P60), and body weight, daily food intake, fasting glycemia, and fasting-induced hyperphagia were measured. POMC recovery was quantified by immunohistochemistry and semiquantitative RT-PCR. Neuropeptide Y (NPY) and GABAergic neurons were identified by in situ hybridization. Arc-POMC neurons projecting to the dorsomedial hypothalamic nucleus (DMH) were studied by stereotactic intracerebral injection of fluorescent retrobeads into the DMH. RESULTS: Tamoxifen treatment of arcPomc-/-:Gad2-CreER mice at P60 resulted in Pomc expression in ∼23-25% of Arc-POMC neurons and ∼15-23% of Pomc mRNA levels, compared to Gad2-CreER control mice. Pomc rescue in GABAergic-POMC neurons at P60 normalized food intake, glycemia, and fasting-induced hyperphagia, while significantly reducing body weight. Energy balance was also improved in arcPomc-/-:Gad2-CreER mice treated with tamoxifen at P25. Distribution analysis of rescued POMC immunoreactive fibers revealed that the DMH is a major target site of GABAergic-POMC neurons. Further, the expression of the orexigenic neuropeptide Y (NPY) in the DMH was increased in arcPomc-/- obese mice but was completely restored after Pomc rescue in arcPomc-/-:Gad2-CreER mice. Finally, we found that ∼75% of Arc-POMC neurons projecting to the DMH are GABAergic. CONCLUSIONS: In the present study, we show that the expression of Pomc in the subpopulation of Arc-GABAergic-POMC neurons is sufficient to maintain normal food intake. In addition, we found that DMH-NPY expression is negatively correlated with Pomc expression in GABAergic-POMC neurons, suggesting that food intake may be regulated by an Arc-GABAergic-POMC → DMH-NPY pathway.


Assuntos
Ingestão de Alimentos/genética , Neurônios GABAérgicos/classificação , Pró-Opiomelanocortina/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal , Núcleo Hipotalâmico Dorsomedial/metabolismo , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Camundongos Obesos , Neuropeptídeo Y/metabolismo , Pró-Opiomelanocortina/genética
4.
Mol Metab ; 34: 43-53, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32180559

RESUMO

OBJECTIVE: Regulation of food intake and energy balance depends on a group of hypothalamic neurons that release anorexigenic melanocortins encoded by the Pomc gene. Although the physiological importance of central melanocortins is well appreciated, the genetic program that defines the functional identity of melanocortin neurons and assures high levels of hypothalamic Pomc expression is only beginning to be understood. This study assessed whether the transcriptional regulator PRDM12, identified as a highly expressed gene in adult mouse POMC neurons, plays an important role in the identity and function of melanocortin neurons. METHODS: We first determined the cellular distribution of PRDM12 in the developing hypothalamus. Then we studied mutant mice with constitutively inactivated Prdm12 to evaluate possible changes in hypothalamic Pomc expression. In addition, we characterized conditional mutant mice specifically lacking Prdm12 in ISL1-positive or POMC neurons during development. Finally, we measured food intake, body weight progression up to 16 weeks of age, adiposity, and glucose tolerance in adult mice lacking Prdm12 selectively from POMC neurons. RESULTS: PRDM12 co-expressed with POMC in mouse hypothalamic neurons from early development to adulthood. Mice lacking Prdm12 displayed greatly reduced Pomc expression in the developing hypothalamus. Selective ablation of Prdm12 from ISL1 neurons prevented hypothalamic Pomc expression. The conditional ablation of Prdm12 limited to POMC neurons greatly reduced Pomc expression in the developing hypothalamus and in adult mice led to increased food intake, adiposity, and obesity. CONCLUSIONS: Altogether, our results demonstrate that PRDM12 plays an essential role in the early establishment of hypothalamic melanocortin neuron identity and the maintenance of high expression levels of Pomc. Its absence in adult mice greatly impairs Pomc expression and leads to increased food intake, adiposity, and obesity.


Assuntos
Adiposidade , Peso Corporal , Proteínas de Transporte/metabolismo , Ingestão de Alimentos , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Pró-Opiomelanocortina/genética , Animais , Proteínas de Transporte/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Gravidez , Pró-Opiomelanocortina/metabolismo
5.
J Neurosci ; 39(21): 4023-4035, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-30886014

RESUMO

Food intake is tightly regulated by a group of neurons present in the arcuate nucleus of the hypothalamus, which release Pomc-encoded melanocortins, the absence of which induces marked hyperphagia and early-onset obesity. Although the relevance of hypothalamic POMC neurons in the regulation of body weight and energy balance is well appreciated, little is known about the transcription factors that establish the melanocortin neuron identity during brain development and its phenotypic maintenance in postnatal life. Here, we report that the transcription factor NKX2.1 is present in mouse hypothalamic POMC neurons from early development to adulthood. Electromobility shift assays showed that NKX2.1 binds in vitro to NKX binding motifs present in the neuronal Pomc enhancers nPE1 and nPE2 and chromatin immunoprecipitation assays detected in vivo binding of NKX2.1 to nPE1 and nPE2 in mouse hypothalamic extracts. Transgenic and mutant studies performed in mouse embryos of either sex and adult males showed that the NKX motifs present in nPE1 and nPE2 are essential for their transcriptional enhancer activity. The conditional early inactivation of Nkx2.1 in the ventral hypothalamus prevented the onset of Pomc expression. Selective Nkx2.1 ablation from POMC neurons decreased Pomc expression in adult males and mildly increased their body weight and adiposity. Our results demonstrate that NKX2.1 is necessary to activate Pomc expression by binding to conserved canonical NKX motifs present in nPE1 and nPE2. Therefore, NKX2.1 plays a critical role in the early establishment of hypothalamic melanocortin neuron identity and participates in the maintenance of Pomc expression levels during adulthood.SIGNIFICANCE STATEMENT Food intake and body weight regulation depend on hypothalamic neurons that release satiety-inducing neuropeptides, known as melanocortins. Central melanocortins are encoded byPomc, and Pomc mutations may lead to hyperphagia and severe obesity. Although the importance of central melanocortins is well appreciated, the genetic program that establishes and maintains fully functional POMC neurons remains to be explored. Here, we combined molecular, genetic, developmental, and functional studies that led to the discovery of NKX2.1, a transcription factor that participates in the early morphogenesis of the developing hypothalamus, as a key player in establishing the early identity of melanocortin neurons by activating Pomc expression. Thus, Nkx2.1 adds to the growing list of genes that participate in body weight regulation and adiposity.


Assuntos
Melanocortinas/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Fator Nuclear 1 de Tireoide/metabolismo , Animais , Peso Corporal/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipotálamo/embriologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
6.
Aging Cell ; 17(1)2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29168299

RESUMO

Weight loss is an effective intervention for diminishing disease burden in obese older adults. Pharmacological interventions that reduce food intake and thereby promote weight loss may offer effective strategies to reduce age-related disease. We previously reported that 17α-estradiol (17α-E2) administration elicits beneficial effects on metabolism and inflammation in old male mice. These observations were associated with reduced calorie intake. Here, we demonstrate that 17α-E2 acts through pro-opiomelanocortin (Pomc) expression in the arcuate nucleus (ARC) to reduce food intake and body mass in mouse models of obesity. These results confirm that 17α-E2 modulates appetite through selective interactions within hypothalamic anorexigenic pathways. Interestingly, some peripheral markers of metabolic homeostasis were also improved in animals with near complete loss of ARC Pomc transcription. This suggests that 17α-E2 might have central and peripheral actions that can beneficially affect metabolism cooperatively or independently.


Assuntos
Estradiol/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Pró-Opiomelanocortina/farmacologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Comportamento Animal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos Transgênicos , Neurônios/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo
7.
Dev Biol ; 416(1): 212-224, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27212025

RESUMO

The hypothalamus is a region of the anterior forebrain that controls basic aspects of vertebrate physiology, but the genes involved in its development are still poorly understood. Here, we investigate the function of the homeobox gene Rax/Rx in early hypothalamic development using a conditional targeted inactivation strategy in the mouse. We found that lack of Rax expression prior to embryonic day 8.5 (E8.5) caused a general underdevelopment of the hypothalamic neuroepithelium, while inactivation at later timepoints had little effect. The early absence of Rax impaired neurogenesis and prevented the expression of molecular markers of the dorsomedial hypothalamus, including neuropeptides Proopiomelanocortin and Somatostatin. Interestingly, the expression domains of genes expressed in the ventromedial hypothalamus and infundibulum invaded dorsal hypothalamic territory, showing that Rax is needed for the proper dorsoventral patterning of the developing medial hypothalamus. The phenotypes caused by the early loss of Rax are similar to those of eliminating the expression of the morphogen Sonic hedgehog (Shh) specifically from the hypothalamus. Consistent with this similarity in phenotypes, we observed that Shh and Rax are coexpressed in the rostral forebrain at late head fold stages and that loss of Rax caused a downregulation of Shh expression in the dorsomedial portion of the hypothalamus.


Assuntos
Proteínas do Olho/fisiologia , Proteínas de Homeodomínio/fisiologia , Hipotálamo/embriologia , Fatores de Transcrição/fisiologia , Animais , Padronização Corporal , Desenvolvimento Embrionário/genética , Proteínas do Olho/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Knockout , Fatores de Transcrição/genética
8.
Diabetes ; 65(3): 660-72, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26467632

RESUMO

Hypothalamic proopiomelanocortin (POMC) is essential for the physiological regulation of energy balance; however, its role in glucose homeostasis remains less clear. We show that hypothalamic arcuate nucleus (Arc)POMC-deficient mice, which develop severe obesity and insulin resistance, unexpectedly exhibit improved glucose tolerance and remain protected from hyperglycemia. To explain these paradoxical phenotypes, we hypothesized that an insulin-independent pathway is responsible for the enhanced glucose tolerance. Indeed, the mutant mice demonstrated increased glucose effectiveness and exaggerated glycosuria relative to wild-type littermate controls at comparable blood glucose concentrations. Central administration of the melanocortin receptor agonist melanotan II in mutant mice reversed alterations in glucose tolerance and glycosuria, whereas, conversely, administration of the antagonist Agouti-related peptide (Agrp) to wild-type mice enhanced glucose tolerance. The glycosuria of ArcPOMC-deficient mice was due to decreased levels of renal GLUT 2 (rGLUT2) but not sodium-glucose cotransporter 2 and was associated with reduced renal catecholamine content. Epinephrine treatment abolished the genotype differences in glucose tolerance and rGLUT2 levels, suggesting that reduced renal sympathetic nervous system (SNS) activity is the underlying mechanism for the observed glycosuria and improved glucose tolerance in ArcPOMC-deficient mice. Therefore, the ArcPOMC-SNS-rGLUT2 axis is potentially an insulin-independent therapeutic target to control diabetes.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Glicemia/metabolismo , Transportador de Glucose Tipo 2/genética , Glicosúria Renal/genética , Resistência à Insulina/genética , Rim/metabolismo , Obesidade/genética , Pró-Opiomelanocortina/genética , Sistema Nervoso Simpático/metabolismo , Proteína Relacionada com Agouti/farmacologia , Animais , Glicemia/efeitos dos fármacos , Western Blotting , Epinefrina/metabolismo , Teste de Tolerância a Glucose , Transportador de Glucose Tipo 2/metabolismo , Glicosúria Renal/metabolismo , Hipotálamo/metabolismo , Injeções Intraventriculares , Camundongos , Camundongos Knockout , Norepinefrina/metabolismo , Obesidade/metabolismo , Peptídeos Cíclicos/farmacologia , Pró-Opiomelanocortina/deficiência , Pró-Opiomelanocortina/metabolismo , Receptores de Melanocortina/agonistas , Receptores de Melanocortina/antagonistas & inibidores , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
9.
Proc Natl Acad Sci U S A ; 112(15): E1861-70, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25825735

RESUMO

Food intake and body weight regulation depend on proper expression of the proopiomelanocortin gene (Pomc) in a group of neurons located in the mediobasal hypothalamus of all vertebrates. These neurons release POMC-encoded melanocortins, which are potent anorexigenic neuropeptides, and their absence from mice or humans leads to hyperphagia and severe obesity. Although the pathophysiology of hypothalamic POMC neurons is well understood, the genetic program that establishes the neuronal melanocortinergic phenotype and maintains a fully functional neuronal POMC phenotype throughout adulthood remains unknown. Here, we report that the early expression of the LIM-homeodomain transcription factor Islet 1 (ISL1) in the developing hypothalamus promotes the terminal differentiation of melanocortinergic neurons and is essential for hypothalamic Pomc expression since its initial onset and throughout the entire lifetime. We detected ISL1 in the prospective hypothalamus just before the onset of Pomc expression and, from then on, Pomc and Isl1 coexpress. ISL1 binds in vitro and in vivo to critical homeodomain binding DNA motifs present in the neuronal Pomc enhancers nPE1 and nPE2, and mutations of these sites completely disrupt the ability of these enhancers to drive reporter gene expression to hypothalamic POMC neurons in transgenic mice and zebrafish. ISL1 is necessary for hypothalamic Pomc expression during mouse and zebrafish embryogenesis. Furthermore, conditional Isl1 inactivation from POMC neurons impairs Pomc expression, leading to hyperphagia and obesity. Our results demonstrate that ISL1 specifies the identity of hypothalamic melanocortin neurons and is required for melanocortin-induced satiety and normal adiposity throughout the entire lifespan.


Assuntos
Adiposidade/fisiologia , Ingestão de Alimentos/fisiologia , Proteínas com Homeodomínio LIM/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Fatores de Transcrição/metabolismo , Adiposidade/genética , Animais , Sequência de Bases , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Ingestão de Alimentos/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Hiperfagia/genética , Hiperfagia/fisiopatologia , Hipotálamo/citologia , Hipotálamo/embriologia , Hipotálamo/metabolismo , Proteínas com Homeodomínio LIM/genética , Masculino , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Dados de Sequência Molecular , Neurônios/citologia , Obesidade/genética , Obesidade/fisiopatologia , Pró-Opiomelanocortina/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Fatores de Transcrição/genética , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
10.
Endocrinology ; 156(4): 1292-302, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25594696

RESUMO

Peptides derived from the proopiomelanocortin (POMC) precursor are critical for the normal regulation of many physiological parameters, and POMC deficiency results in severe obesity and metabolic dysfunction. Conversely, augmentation of central nervous system melanocortin function is a promising therapeutic avenue for obesity and diabetes but is confounded by detrimental cardiovascular effects including hypertension. Because the hypothalamic population of POMC-expressing neurons is neurochemically and neuroanatomically heterogeneous, there is interest in the possible dissociation of functionally distinct POMC neuron subpopulations. We used a Cre recombinase-dependent and hypothalamus-specific reactivatable PomcNEO allele to restrict Pomc expression to hypothalamic neurons expressing leptin receptor (Lepr) in mice. In contrast to mice with total hypothalamic Pomc deficiency, which are severely obese, mice with Lepr-restricted Pomc expression displayed fully normal body weight, food consumption, glucose homeostasis, and locomotor activity. Thus, Lepr+ POMC neurons, which constitute approximately two-thirds of the total POMC neuron population, are sufficient for normal regulation of these parameters. This functional dissociation approach represents a promising avenue for isolating therapeutically relevant POMC neuron subpopulations.


Assuntos
Metabolismo Energético/fisiologia , Homeostase/fisiologia , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptores para Leptina/metabolismo , Animais , Glicemia/metabolismo , Feminino , Teste de Tolerância a Glucose , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Camundongos , Camundongos Knockout , Atividade Motora/fisiologia , Pró-Opiomelanocortina/genética , Receptores para Leptina/genética
11.
J Clin Invest ; 122(11): 4203-12, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23093774

RESUMO

Obesity is a chronic metabolic disorder affecting half a billion people worldwide. Major difficulties in managing obesity are the cessation of continued weight loss in patients after an initial period of responsiveness and rebound to pretreatment weight. It is conceivable that chronic weight gain unrelated to physiological needs induces an allostatic regulatory state that defends a supranormal adipose mass despite its maladaptive consequences. To challenge this hypothesis, we generated a reversible genetic mouse model of early-onset hyperphagia and severe obesity by selectively blocking the expression of the proopiomelanocortin gene (Pomc) in hypothalamic neurons. Eutopic reactivation of central POMC transmission at different stages of overweight progression normalized or greatly reduced food intake in these obesity-programmed mice. Hypothalamic Pomc rescue also attenuated comorbidities such as hyperglycemia, hyperinsulinemia, and hepatic steatosis and normalized locomotor activity. However, effectiveness of treatment to normalize body weight and adiposity declined progressively as the level of obesity at the time of Pomc induction increased. Thus, our study using a novel reversible monogenic obesity model reveals the critical importance of early intervention for the prevention of subsequent allostatic overload that auto-perpetuates obesity.


Assuntos
Tecido Adiposo/fisiopatologia , Adiposidade , Ingestão de Alimentos , Hipotálamo/fisiopatologia , Obesidade/prevenção & controle , Obesidade/fisiopatologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Modelos Animais de Doenças , Hiperfagia/genética , Hiperfagia/metabolismo , Hiperfagia/patologia , Hiperfagia/fisiopatologia , Hiperfagia/prevenção & controle , Hipotálamo/metabolismo , Hipotálamo/patologia , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Obesidade/genética , Obesidade/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo
12.
Endocrinology ; 149(7): 3346-54, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18403487

RESUMO

Signal transducer and activator of transcription (Stat)-3 signals mediate many of the metabolic effects of the fat cell-derived hormone, leptin. In mice, brain-specific depletion of either the long form of the leptin receptor (Lepr) or Stat3 results in comparable obese phenotypes as does replacement of Lepr with an altered leptin receptor locus that codes for a Lepr unable to interact with Stat3. Among the multiple brain regions containing leptin-sensitive Stat3 sites, cells expressing feeding-related neuropeptides in the arcuate nucleus of the hypothalamus have received much of the focus. To determine the contribution to energy homeostasis of Stat3 expressed in agouti-related protein (Agrp)/neuropeptide Y (Npy) arcuate neurons, Stat3 was deleted specifically from these cells, and several metabolic indices were measured. It was found that deletion of Stat3 from Agrp/Npy neurons resulted in modest weight gain that was accounted for by increased adiposity. Agrp/Stat3-deficient mice also showed hyperleptinemia, and high-fat diet-induced hyperinsulinemia. Stat3 deletion in Agrp/Npy neurons also resulted in altered hypothalamic gene expression indicated by increased Npy mRNA and decreased induction of suppressor of cytokine signaling-3 in response to leptin. Agrp mRNA levels in the fed or fasted state were unaffected. Behaviorally, mice without Stat3 in Agrp/Npy neurons were mildly hyperphagic and hyporesponsive to leptin. We conclude that Stat3 in Agrp/Npy neurons is required for normal energy homeostasis, but Stat3 signaling in other brain areas also contributes to the regulation of energy homeostasis.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Fator de Transcrição STAT3/fisiologia , Adiposidade/efeitos dos fármacos , Adiposidade/genética , Animais , Peso Corporal/efeitos dos fármacos , Gorduras na Dieta/farmacologia , Metabolismo Energético/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Hipotálamo/citologia , Hipotálamo/metabolismo , Imuno-Histoquímica , Leptina/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Aumento de Peso/efeitos dos fármacos
13.
Ann N Y Acad Sci ; 994: 169-74, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12851313

RESUMO

It is now established that the hypothalamus is essential in coordinating endocrine, autonomic, and behavioral responses to changes in energy availability. However, the interaction of key peptides, neuropeptides, and neurotransmitters systems within the hypothalamus has yet to be delineated. Recently, we investigated the mechanisms through which central serotonergic (5-hydroxytryptamine, 5-HT) systems recruit leptin-responsive hypothalamic pathways, such as the melanocortin systems, to affect energy balance. Through a combination of functional neuroanatomy, feeding, and electrophysiology studies in rodents, we found that 5-HT drugs require functional melanocortin pathways to exert their effects on food intake. Specifically, we observed that anorectic 5-HT drugs activate pro-opiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (Arc). We provide evidence that the serotonin 2C receptor (5-HT(2C)R) is expressed on POMC neurons and contributes to this effect. Finally, we found that 5-HT drug-induced hypophagia is attenuated by pharmacological or genetic blockade of downstream melanocortin 3 and 4 receptors. We review candidate brain regions expressing melanocortin 3 and 4 receptors that play a role in energy balance. A model is presented in which activation of the melanocortin system is downstream of 5-HT and is necessary to produce the complete anorectic effect of 5-HT drugs. The data reviewed in this paper incorporate the central 5-HT system to the growing list of metabolic signals that converge on melanocortin neurons in the hypothalamus.


Assuntos
Metabolismo Energético/fisiologia , Homeostase , Pró-Opiomelanocortina/metabolismo , Serotonina/metabolismo , Animais , Fenfluramina/metabolismo , Humanos , Hipotálamo/metabolismo , Neurônios/metabolismo , Serotoninérgicos/metabolismo , Transdução de Sinais/fisiologia
14.
Endocrinology ; 144(5): 1753-60, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12697680

RESUMO

Proopiomelanocortin (POMC) neurons in the hypothalamus are direct targets of the adipostatic hormone leptin and contribute to energy homeostasis by integrating peripheral and central information. The melanocortin and beta-endorphin neuropeptides are processed from POMC and putatively coreleased at axon terminals. Melanocortins have been shown by a combination of pharmacological and genetic methods to have inhibitory effects on appetite and body weight. In contrast, pharmacological studies have generally indicated that opioids stimulate food intake. Here we report that male mice engineered to selectively lack beta-endorphin, but that retained normal melanocortin signaling, were hyperphagic and obese. Furthermore, beta-endorphin mutant and wild-type mice had identical orexigenic responses to exogenous opioids and identical anorectic responses to the nonselective opioid antagonist naloxone, implicating an alternative endogenous opioid tone to beta-endorphin that physiologically stimulates feeding. These genetic data indicate that beta-endorphin is required for normal regulation of feeding, but, in contrast to earlier reports suggesting opposing actions of beta-endorphin and melanocortins on appetite, our results suggest a more complementary interaction between the endogenously released POMC-derived peptides in the regulation of energy homeostasis.


Assuntos
Metabolismo Energético/fisiologia , Homeostase/fisiologia , beta-Endorfina/fisiologia , Animais , Ingestão de Alimentos/efeitos dos fármacos , Glucose/metabolismo , Hiperinsulinismo/etiologia , Hiperfagia/etiologia , Leptina/sangue , Masculino , Camundongos , Camundongos Knockout , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neuropeptídeo Y/farmacologia , Obesidade/etiologia , Valores de Referência , beta-Endorfina/deficiência , beta-Endorfina/farmacologia
15.
Endocrinology ; 144(4): 1331-40, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12639916

RESUMO

Hypothalamic proopiomelanocortin (POMC) neurons are critical for controlling homeostatic functions in the mammal. We used a transgenic mouse model in which the POMC neurons were labeled with enhanced green fluorescent protein to perform visualized, whole-cell patch recordings from prepubertal female hypothalamic slices. The mouse POMC-enhanced green fluorescent protein neurons expressed the same endogenous conductances (a transient outward K(+) current and a hyperpolarization-activated, cation current) that have been described for guinea pig POMC neurons. In addition, the selective micro -opioid receptor agonist DAMGO induced an outward current (maximum of 12.8 +/- 1.2 pA), which reversed at K(+) equilibrium potential (E(K+)), in the majority (85%) of POMC neurons with an EC(50) of 102 nM. This response was blocked by the opioid receptor antagonist naloxone with an inhibition constant of 3.1 nM. In addition, the gamma-aminobutyric acid(B) receptor agonist baclofen (40 micro M) caused an outward current (21.6 +/- 4.0 pA) that reversed at E(K+) in these same neurons. The ATP-sensitive potassium channel opener diazoxide also induced an outward K(+) current (maximum of 18.7 +/- 2.2 pA) in the majority (92%) of POMC neurons with an EC(50) of 61 micro M. The response to diazoxide was blocked by the sulfonylurea tolbutamide, indicating that the POMC neurons express both Kir6.2 and sulfonylurea receptor 1 channel subunits, which was verified using single cell RT-PCR. This pharmacological and molecular profile suggested that POMC neurons might be sensitive to metabolic inhibition, and indeed, we found that their firing rate varied with changes in glucose concentrations. Therefore, it appears that POMC neurons may function as an integrator of metabolic cues and synaptic input for controlling homeostasis in the mammal.


Assuntos
Glucose/farmacologia , Hipotálamo/citologia , Neurônios/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , Pró-Opiomelanocortina/genética , Transportadores de Cassetes de Ligação de ATP , Trifosfato de Adenosina/metabolismo , Analgésicos Opioides/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Baclofeno/farmacologia , Diazóxido/farmacologia , Condutividade Elétrica , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Feminino , Agonistas GABAérgicos/farmacologia , Agonistas dos Receptores de GABA-B , Proteínas de Fluorescência Verde , Indicadores e Reagentes/metabolismo , Proteínas Luminescentes/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Canais de Potássio/genética , Canais de Potássio Corretores do Fluxo de Internalização/agonistas , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro/análise , Receptores de Droga , Receptores de GABA-B/fisiologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiologia , Receptores de Sulfonilureias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA