Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Metab ; 14: 130-138, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29914853

RESUMO

OBJECTIVE: To date, early developmental ablation of leptin receptor (LepRb) expression from circumscribed populations of hypothalamic neurons (e.g., arcuate nucleus (ARC) Pomc- or Agrp-expressing cells) has only minimally affected energy balance. In contrast, removal of LepRb from at least two large populations (expressing vGat or Nos1) spanning multiple hypothalamic regions produced profound obesity and metabolic dysfunction. Thus, we tested the notion that the total number of leptin-responsive hypothalamic neurons (rather than specific subsets of cells with a particular molecular or anatomical signature) subjected to early LepRb deletion might determine energy balance. METHODS: We generated new mouse lines deleted for LepRb in ARC GhrhCre neurons or in Htr2cCre neurons (representing roughly half of all hypothalamic LepRb neurons, distributed across many nuclei). We compared the phenotypes of these mice to previously-reported models lacking LepRb in Pomc, Agrp, vGat or Nos1 cells. RESULTS: The early developmental deletion of LepRb from vGat or Nos1 neurons produced dramatic obesity, but deletion of LepRb from Pomc, Agrp, Ghrh, or Htr2c neurons minimally altered energy balance. CONCLUSIONS: Although early developmental deletion of LepRb from known populations of ARC neurons fails to substantially alter body weight, the minimal phenotype of mice lacking LepRb in Htr2c cells suggests that the phenotype that results from early developmental LepRb deficiency depends not simply upon the total number of leptin-responsive hypothalamic LepRb cells. Rather, specific populations of LepRb neurons must play particularly important roles in body energy homeostasis; these as yet unidentified LepRb cells likely reside in the DMH.


Assuntos
Metabolismo Energético , Hipotálamo/citologia , Neurônios/metabolismo , Obesidade/metabolismo , Receptores para Leptina/genética , Animais , Feminino , Deleção de Genes , Hipotálamo/embriologia , Hipotálamo/metabolismo , Masculino , Camundongos , Neurônios/classificação , Neurônios/citologia , Receptor 5-HT2C de Serotonina/genética , Receptor 5-HT2C de Serotonina/metabolismo , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Receptores para Leptina/metabolismo
2.
Diabetes ; 67(6): 1093-1104, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29535089

RESUMO

Leptin acts via its receptor (LepRb) to modulate gene expression in hypothalamic LepRb-expressing neurons, thereby controlling energy balance and glucose homeostasis. Despite the importance of the control of gene expression in hypothalamic LepRb neurons for leptin action, the transcriptional targets of LepRb signaling have remained undefined because LepRb cells contribute a small fraction to the aggregate transcriptome of the brain regions in which they reside. We thus employed translating ribosome affinity purification followed by RNA sequencing to isolate and analyze mRNA from the hypothalamic LepRb neurons of wild-type or leptin-deficient (Lepob/ob) mice treated with vehicle or exogenous leptin. Although the expression of most of the genes encoding the neuropeptides commonly considered to represent the main targets of leptin action were altered only following chronic leptin deprivation, our analysis revealed other transcripts that were coordinately regulated by leptin under multiple treatment conditions. Among these, acute leptin treatment increased expression of the transcription factor Atf3 in LepRb neurons. Furthermore, ablation of Atf3 from LepRb neurons (Atf3LepRbKO mice) decreased leptin efficacy and promoted positive energy balance in mice. Thus, this analysis revealed the gene targets of leptin action, including Atf3, which represents a cellular mediator of leptin action.


Assuntos
Fator 3 Ativador da Transcrição/agonistas , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , Receptores para Leptina/agonistas , Transdução de Sinais , Fator 3 Ativador da Transcrição/química , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Animais , Cruzamentos Genéticos , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Metabolismo Energético/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/patologia , Leptina/análogos & derivados , Leptina/farmacologia , Leptina/uso terapêutico , Lipotrópicos/farmacologia , Lipotrópicos/uso terapêutico , Masculino , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Obesidade/patologia , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Análise de Sequência de RNA , Transdução de Sinais/efeitos dos fármacos
3.
Endocrinology ; 159(4): 1860-1872, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29522093

RESUMO

The adipocyte-derived hormone leptin acts via its receptor (LepRb) on central nervous system neurons to communicate the repletion of long-term energy stores, to decrease food intake, and to promote energy expenditure. We generated mice that express Cre recombinase from the calcitonin receptor (Calcr) locus (Calcrcre mice) to study Calcr-expressing LepRb (LepRbCalcr) neurons, which reside predominantly in the arcuate nucleus (ARC). Calcrcre-mediated ablation of LepRb in LepRbCalcrknockout (KO) mice caused hyperphagic obesity. Because LepRb-mediated transcriptional control plays a crucial role in leptin action, we used translating ribosome affinity purification followed by RNA sequencing to define the transcriptome of hypothalamic Calcr neurons, along with its alteration in LepRbCalcrKO mice. We found that ARC LepRbCalcr cells include neuropeptide Y (NPY)/agouti-related peptide (AgRP)/γ-aminobutyric acid (GABA) ("NAG") cells as well as non-NAG cells that are distinct from pro-opiomelanocortin cells. Furthermore, although LepRbCalcrKO mice exhibited dysregulated expression of several genes involved in energy balance, neither the expression of Agrp and Npy nor the activity of NAG cells was altered in vivo. Thus, although direct leptin action via LepRbCalcr cells plays an important role in leptin action, our data also suggest that leptin indirectly, as well as directly, regulates these cells.


Assuntos
Ingestão de Alimentos/fisiologia , Hipotálamo/metabolismo , Leptina/análogos & derivados , Neurônios/fisiologia , Receptores da Calcitonina/metabolismo , Receptores para Leptina/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Leptina/farmacologia , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Obesidade/genética , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptores para Leptina/genética
4.
Diabetes ; 65(9): 2711-23, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27207534

RESUMO

The brain influences glucose homeostasis, partly by supplemental control over insulin and glucagon secretion. Without this central regulation, diabetes and its complications can ensue. Yet, the neuronal network linking to pancreatic islets has never been fully mapped. Here, we refine this map using pseudorabies virus (PRV) retrograde tracing, indicating that the pancreatic islets are innervated by efferent circuits that emanate from the hypothalamus. We found that the hypothalamic arcuate nucleus (ARC), ventromedial nucleus (VMN), and lateral hypothalamic area (LHA) significantly overlap PRV and the physiological glucose-sensing enzyme glucokinase. Then, experimentally lowering glucose sensing, specifically in the ARC, resulted in glucose intolerance due to deficient insulin secretion and no significant effect in the VMN, but in the LHA it resulted in a lowering of the glucose threshold that improved glucose tolerance and/or improved insulin sensitivity, with an exaggerated counter-regulatory response for glucagon secretion. No significant effect on insulin sensitivity or metabolic homeostasis was noted. Thus, these data reveal novel direct neuronal effects on pancreatic islets and also render a functional validation of the brain-to-islet neuronal map. They also demonstrate that distinct regions of the hypothalamus differentially control insulin and glucagon secretion, potentially in partnership to help maintain glucose homeostasis and guard against hypoglycemia.


Assuntos
Encéfalo/metabolismo , Glucose/metabolismo , Hipotálamo/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Mapeamento Encefálico , Feminino , Glucoquinase/metabolismo , Região Hipotalâmica Lateral/metabolismo , Imuno-Histoquímica , Ilhotas Pancreáticas/inervação , Masculino , Camundongos Endogâmicos C57BL , Núcleo Hipotalâmico Ventromedial/metabolismo
5.
Endocrinology ; 157(4): 1555-65, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26862996

RESUMO

A variety of data suggest that estrogen action on kisspeptin (Kiss1)-containing arcuate nucleus neurons (which coexpress Kiss1, neurokinin B (the product of Tac2) and dynorphin (KNDy) neurons restrains reproductive onset and function, but roles for estrogen action in these Kiss1 neurons relative to a distinct population of rostral hypothalamic Kiss1 neurons (which does not express Tac2 or dynorphin) have not been directly tested. To test the role for estrogen receptor (ER)α in KNDy cells, we thus generated Tac2(Cre) and Kiss1(Cre) knock-in mice and bred them onto the Esr1(flox) background to ablate ERα specifically in Tac2-expressing cells (ERα(Tac2)KO mice) or all Kiss1 cells (ERα(Kiss1)KO mice), respectively. Most ERα-expressing Tac2 neurons represent KNDy cells. Arcuate nucleus Kiss1 expression was elevated in ERα(Tac2)KO and ERα(Kiss1)KO females independent of gonadal hormones, whereas rostral hypothalamic Kiss1 expression was normal in ERα(Tac2)KO but decreased in ERα(Kiss1)KO females; this suggests that ERα in rostral Kiss1 cells is crucial for control of Kiss1 expression in these cells. Both ERα(Kiss1)KO and ERα(Tac2)KO females displayed early vaginal opening, early and persistent vaginal cornification, increased gonadotropins, uterine hypertrophy, and other evidence of estrogen excess. Thus, deletion of ERα in Tac2 neurons suffices to drive precocious gonadal hyperstimulation, demonstrating that ERα in Tac2 neurons typically restrains pubertal onset and hypothalamic reproductive drive.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Neurônios/metabolismo , Precursores de Proteínas/metabolismo , Maturidade Sexual/fisiologia , Taquicininas/metabolismo , Animais , Composição Corporal/genética , Composição Corporal/fisiologia , Dinorfinas/genética , Dinorfinas/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Feminino , Gonadotropinas/metabolismo , Hipotálamo/citologia , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Ovariectomia , Ovário/metabolismo , Precursores de Proteínas/genética , Reprodução/genética , Reprodução/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Maturidade Sexual/genética , Taquicininas/genética , Fatores de Tempo , Útero/metabolismo
6.
Cell Metab ; 20(6): 1030-7, 2014 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-25470549

RESUMO

Hypoglycemia engenders an autonomically mediated counterregulatory (CR)-response that stimulates endogenous glucose production to maintain concentrations within an appropriate physiological range. Although the involvement of the brain in preserving normoglycemia has been established, the neurocircuitry underlying centrally mediated CR-responses remains unclear. Here we demonstrate that lateral parabrachial nucleus cholecystokinin (CCK(LPBN)) neurons are a population of glucose-sensing cells (glucose inhibited) with counterregulatory capacity. Furthermore, we reveal that steroidogenic-factor 1 (SF1)-expressing neurons of the ventromedial nucleus of the hypothalamus (SF1(VMH)) are the specific target of CCK(LPBN) glucoregulatory neurons. This discrete CCK(LPBN)→SF1(VMH) neurocircuit is both necessary and sufficient for the induction of CR-responses. Together, these data identify CCK(LPBN) neurons, and specifically CCK neuropeptide, as glucoregulatory and provide significant insight into the homeostatic mechanisms controlling CR-responses to hypoglycemia.


Assuntos
Colecistocinina/metabolismo , Hipoglicemia/metabolismo , Hipotálamo/metabolismo , Animais , Glicemia/metabolismo , Masculino , Camundongos , Núcleos Parabraquiais/citologia
7.
Nat Med ; 18(5): 820-3, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22522563

RESUMO

Few effective measures exist to combat the worldwide obesity epidemic(1), and the identification of potential therapeutic targets requires a deeper understanding of the mechanisms that control energy balance. Leptin, an adipocyte-derived hormone that signals the long-term status of bodily energy stores, acts through multiple types of leptin receptor long isoform (LepRb)-expressing neurons (called here LepRb neurons) in the brain to control feeding, energy expenditure and endocrine function(2-4). The modest contributions to energy balance that are attributable to leptin action in many LepRb populations(5-9) suggest that other previously unidentified hypothalamic LepRb neurons have key roles in energy balance. Here we examine the role of LepRb in neuronal nitric oxide synthase (NOS1)-expressing LebRb (LepRb(NOS1)) neurons that comprise approximately 20% of the total hypothalamic LepRb neurons. Nos1(cre)-mediated genetic ablation of LepRb (Lepr(Nos1KO)) in mice produces hyperphagic obesity, decreased energy expenditure and hyperglycemia approaching that seen in whole-body LepRb-null mice. In contrast, the endocrine functions in Lepr(Nos1KO) mice are only modestly affected by the genetic ablation of LepRb in these neurons. Thus, hypothalamic LepRb(NOS1) neurons are a key site of action of the leptin-mediated control of systemic energy balance.


Assuntos
Metabolismo Energético , Hipotálamo/fisiologia , Leptina/fisiologia , Neurônios/fisiologia , Óxido Nítrico Sintase Tipo I/fisiologia , Animais , Camundongos , Receptores para Leptina/fisiologia
8.
Diabetes ; 59(4): 782-90, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20068132

RESUMO

OBJECTIVE: Leptin acts via its receptor (LepRb) to signal the status of body energy stores. Leptin binding to LepRb initiates signaling by activating the associated Janus kinase 2 (Jak2) tyrosine kinase, which promotes the phosphorylation of tyrosine residues on the intracellular tail of LepRb. Two previously examined LepRb phosphorylation sites mediate several, but not all, aspects of leptin action, leading us to hypothesize that Jak2 signaling might contribute to leptin action independently of LepRb phosphorylation sites. We therefore determined the potential role in leptin action for signals that are activated by Jak2 independently of LepRb phosphorylation (Jak2-autonomous signals). RESEARCH DESIGN AND METHODS: We inserted sequences encoding a truncated LepRb mutant (LepRb(Delta65c), which activates Jak2 normally, but is devoid of other LepRb intracellular sequences) into the mouse Lepr locus. We examined the leptin-regulated physiology of the resulting Delta/Delta mice relative to LepRb-deficient db/db animals. RESULTS: The Delta/Delta animals were similar to db/db animals in terms of energy homeostasis, neuroendocrine and immune function, and regulation of the hypothalamic arcuate nucleus, but demonstrated modest improvements in glucose homeostasis. CONCLUSIONS: The ability of Jak2-autonomous LepRb signals to modulate glucose homeostasis in Delta/Delta animals suggests a role for these signals in leptin action. Because Jak2-autonomous LepRb signals fail to mediate most leptin action, however, signals from other LepRb intracellular sequences predominate.


Assuntos
Janus Quinase 2/genética , Leptina/fisiologia , Receptores para Leptina/genética , Animais , DNA Complementar/genética , Estro/fisiologia , Feminino , Vetores Genéticos , Hipotálamo/fisiologia , Janus Quinase 2/metabolismo , Leptina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Sítio-Dirigida , Fosforilação , Plasmídeos , Reação em Cadeia da Polimerase , RNA/genética , RNA/isolamento & purificação , Receptores para Leptina/deficiência , Receptores para Leptina/fisiologia , Baço/citologia , Baço/imunologia , Baço/fisiologia , Linfócitos T/imunologia , Linfócitos T/fisiologia
9.
Cell Metab ; 7(2): 179-85, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18249177

RESUMO

The arcuate nucleus of the hypothalamus (ARH) is a key component of hypothalamic pathways regulating energy balance, and leptin is required for normal development of ARH projections. Diet-induced obesity (DIO) has a polygenic mode of inheritance, and DIO individuals develop the metabolic syndrome when a moderate amount of fat is added to the diet. Here we demonstrate that rats selectively bred to develop DIO, which are known to be leptin resistant before they become obese, have defective ARH projections that persist into adulthood. Furthermore, the ability of leptin to activate intracellular signaling in ARH neurons in vivo and to promote ARH neurite outgrowth in vitro is significantly reduced in DIO neonates. Thus, animals that are genetically predisposed toward obesity display an abnormal organization of hypothalamic pathways involved in energy homeostasis that may be the result of diminished responsiveness of ARH neurons to the trophic actions of leptin during postnatal development.


Assuntos
Hipotálamo/patologia , Neuritos , Neurônios/ultraestrutura , Obesidade/etiologia , Animais , Regulação do Apetite , Núcleo Arqueado do Hipotálamo/patologia , Dieta , Metabolismo Energético , Predisposição Genética para Doença , Leptina/fisiologia , Ratos , Ratos Endogâmicos , Transdução de Sinais
10.
Am J Physiol Regul Integr Comp Physiol ; 294(2): R290-301, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17989137

RESUMO

We assessed the effect of early-onset exercise as a means of preventing childhood obesity using juvenile male rats selectively bred to develop diet-induced obesity (DIO) or to be diet resistant (DR) when fed a 31% fat high-energy diet. Voluntary wheel running begun at 36 days of age selectively reduced adiposity in DIO vs. DR rats. Other 4-wk-old DIO rats fed a high-energy diet and exercised (Ex) for 13 wk increased their core temperature, gained 22% less body weight, and had 39% lighter fat pads compared with sedentary (Sed) rats. When wheels were removed after 6 wk (6 wk Ex/7 wk Sed), rats gained less body weight over the next 7 wk than Sed rats and still had comparable adipose pad weights to 13-wk-exercised rats. In fact, only 3 wk of exercise sufficed to prevent obesity for 10 wk after wheel removal. Terminally, the 6-wk-Ex/7-wk-Sed rats had a 55% increase in arcuate nucleus proopiomelanocortin mRNA expression vs. Sed rats, suggesting that this contributed to their sustained obesity resistance. Finally, when Sed rats were calorically restricted for 6 wk to weight match them to Ex rats (6 wk Rstr/7 wk Al), they increased their intake and body weight when fed ad libitum and, after 7 wk more, had higher leptin levels and adiposity than Sed rats. Thus, early-onset exercise may favorably alter, while early caloric restriction may unfavorably influence, the development of the hypothalamic pathways controlling energy homeostasis during brain development.


Assuntos
Peso Corporal/fisiologia , Hipotálamo/fisiologia , Obesidade/prevenção & controle , Obesidade/fisiopatologia , Condicionamento Físico Animal/fisiologia , Tecido Adiposo/fisiologia , Ração Animal , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Restrição Calórica , Ingestão de Alimentos/fisiologia , Insulina/sangue , Leptina/sangue , Masculino , Neuropeptídeo Y/genética , Pró-Opiomelanocortina/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos , Receptores para Leptina/genética , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA