Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 151(3): 645-57, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-23101631

RESUMO

Neural regulation of energy expenditure is incompletely understood. By genetically disrupting GABAergic transmission in a cell-specific fashion, and by combining this with selective pharmacogenetic activation and optogenetic mapping techniques, we have uncovered an arcuate-based circuit that selectively drives energy expenditure. Specifically, mice lacking synaptic GABA release from RIP-Cre neurons have reduced energy expenditure, become obese and are extremely sensitive to high-fat diet-induced obesity, the latter due to defective diet-induced thermogenesis. Leptin's ability to stimulate thermogenesis, but not to reduce feeding, is markedly attenuated. Acute, selective activation of arcuate GABAergic RIP-Cre neurons, which monosynaptically innervate PVH neurons projecting to the NTS, rapidly stimulates brown fat and increases energy expenditure but does not affect feeding. Importantly, this response is dependent upon GABA release from RIP-Cre neurons. Thus, GABAergic RIP-Cre neurons in the arcuate selectively drive energy expenditure, contribute to leptin's stimulatory effect on thermogenesis, and protect against diet-induced obesity.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Metabolismo Energético , Neurônios GABAérgicos/metabolismo , Vias Neurais , Tecido Adiposo Marrom/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Dieta , Integrases/metabolismo , Leptina/metabolismo , Camundongos , Obesidade/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33827930

RESUMO

The ventromedial hypothalamus (VMH) is a critical neural node that senses blood glucose and promotes glucose utilization or mobilization during hypoglycemia. The VMH neurons that control these distinct physiologic processes are largely unknown. Here, we show that melanocortin 3 receptor (Mc3R)-expressing VMH neurons (VMHMC3R) sense glucose changes both directly and indirectly via altered excitatory input. We identify presynaptic nodes that potentially regulate VMHMC3R neuronal activity, including inputs from proopiomelanocortin (POMC)-producing neurons in the arcuate nucleus. We find that VMHMC3R neuron activation blunts, and their silencing enhances glucose excursion following a glucose load. Overall, these findings demonstrate that VMHMC3R neurons are a glucose-responsive hypothalamic subpopulation that promotes glucose disposal upon activation; this highlights a potential site for targeting dysregulated glycemia.


Assuntos
Glucose/metabolismo , Hiperglicemia/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Receptor Tipo 3 de Melanocortina/metabolismo , Animais , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Pró-Opiomelanocortina/metabolismo , Receptor Tipo 3 de Melanocortina/genética , Potenciais Sinápticos
3.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33593916

RESUMO

The TGFß cytokine family member, GDF-15, reduces food intake and body weight and represents a potential treatment for obesity. Because the brainstem-restricted expression pattern of its receptor, GDNF Family Receptor α-like (GFRAL), presents an exciting opportunity to understand mechanisms of action for area postrema neurons in food intake; we generated GfralCre and conditional GfralCreERT mice to visualize and manipulate GFRAL neurons. We found infection or pathophysiologic states (rather than meal ingestion) stimulate GFRAL neurons. TRAP-Seq analysis of GFRAL neurons revealed their expression of a wide range of neurotransmitters and neuropeptides. Artificially activating GfralCre -expressing neurons inhibited feeding, decreased gastric emptying, and promoted a conditioned taste aversion (CTA). GFRAL neurons most strongly innervate the parabrachial nucleus (PBN), where they target CGRP-expressing (CGRPPBN) neurons. Silencing CGRPPBN neurons abrogated the aversive and anorexic effects of GDF-15. These findings suggest that GFRAL neurons link non-meal-associated pathophysiologic signals to suppress nutrient uptake and absorption.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator 15 de Diferenciação de Crescimento/farmacologia , Neurônios/fisiologia , Núcleos Parabraquiais/fisiologia , Animais , Peso Corporal , Feminino , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Núcleos Parabraquiais/efeitos dos fármacos , Ratos , Ratos Long-Evans
4.
Nature ; 532(7597): 103-6, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27001694

RESUMO

Instinctive reactions to danger are critical to the perpetuation of species and are observed throughout the animal kingdom. The scent of predators induces an instinctive fear response in mice that includes behavioural changes, as well as a surge in blood stress hormones that mobilizes multiple body systems to escape impending danger. How the olfactory system routes predator signals detected in the nose to achieve these effects is unknown. Here we identify a specific area of the olfactory cortex in mice that induces stress hormone responses to volatile predator odours. Using monosynaptic and polysynaptic viral tracers, we found that multiple olfactory cortical areas transmit signals to hypothalamic corticotropin-releasing hormone (CRH) neurons, which control stress hormone levels. However, only one minor cortical area, the amygdalo-piriform transition area (AmPir), contained neurons upstream of CRH neurons that were activated by volatile predator odours. Chemogenetic stimulation of AmPir activated CRH neurons and induced an increase in blood stress hormones, mimicking an instinctive fear response. Moreover, chemogenetic silencing of AmPir markedly reduced the stress hormone response to predator odours without affecting a fear behaviour. These findings suggest that AmPir, a small area comprising <5% of the olfactory cortex, plays a key part in the hormonal component of the instinctive fear response to volatile predator scents.


Assuntos
Hormônios/metabolismo , Odorantes/análise , Córtex Olfatório/anatomia & histologia , Córtex Olfatório/fisiologia , Condutos Olfatórios , Comportamento Predatório , Olfato/fisiologia , Estresse Psicológico , Hormônio Adrenocorticotrópico/sangue , Animais , Corticosterona/sangue , Hormônio Liberador da Corticotropina/sangue , Hormônio Liberador da Corticotropina/metabolismo , Reação de Fuga , Medo , Feminino , Hipocampo/citologia , Hipocampo/fisiologia , Hormônios/sangue , Instinto , Masculino , Camundongos , Neurônios/metabolismo , Córtex Olfatório/citologia , Percepção Olfatória/fisiologia , Telencéfalo/anatomia & histologia , Telencéfalo/citologia , Telencéfalo/fisiologia
5.
Psychol Med ; 50(16): 2790-2798, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-31615590

RESUMO

BACKGROUND: Preclinical and human studies suggest an association between chronic inflammation and the development of depressive behaviors. This is proposed to occur through downstream effects of inflammatory cytokines on neuroplasticity, neurogenesis and neurotransmitter function, although the neural correlates remain poorly understood in humans. METHODS: In Study 1, structural magnetic resonance imaging and serum inflammatory cytokine data were analyzed from 53 psychiatrically healthy female participants. Correlational analyses were conducted between interleukin-6 (IL-6) and volume in a priori regions implicated in the pathophysiology of major depressive disorder (MDD). In Study 2, medical data [including serum inflammatory acute phase reactants (C-reactive protein)] were analyzed for 12 589 participants. Participants were classified as having (n = 2541) v. not having (n = 10 048) probable lifetime MDD using phenotypes derived using machine-learning approaches. Non-parametric analyses compared inflammation between groups, whereas regression analyses probed whether inflammation predicted probable MDD classification while accounting for other variables. RESULTS: In Study 1, significant negative correlations emerged between IL-6 and hippocampal, caudate, putamen and amygdalar volume. In Study 2, the MDD group showed a higher probability of elevated inflammation than the non-MDD group. Moreover, elevated inflammation was a significant predictor of probable MDD classification. CONCLUSIONS: Findings indicate that inflammation is cross-sectionally related to reduced volume in brain regions implicated in MDD phenotypes among a sample of psychiatrically healthy women, and is associated with the presence of probable MDD in a large clinical dataset. Future investigations may identify specific inflammatory markers predicting first MDD onset.


Assuntos
Encéfalo/fisiopatologia , Transtorno Depressivo Maior/sangue , Transtorno Depressivo Maior/fisiopatologia , Inflamação/psicologia , Adulto , Biomarcadores/sangue , Proteína C-Reativa/análise , Estudos de Casos e Controles , Doença Crônica , Comorbidade , Feminino , Humanos , Inflamação/sangue , Interleucina-6/sangue , Modelos Logísticos , Imageamento por Ressonância Magnética , Masculino , Prontuários Médicos , Fenótipo , Adulto Jovem
6.
Nature ; 507(7491): 238-42, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24487620

RESUMO

Hunger is a hard-wired motivational state essential for survival. Agouti-related peptide (AgRP)-expressing neurons in the arcuate nucleus (ARC) at the base of the hypothalamus are crucial to the control of hunger. They are activated by caloric deficiency and, when naturally or artificially stimulated, they potently induce intense hunger and subsequent food intake. Consistent with their obligatory role in regulating appetite, genetic ablation or chemogenetic inhibition of AgRP neurons decreases feeding. Excitatory input to AgRP neurons is important in caloric-deficiency-induced activation, and is notable for its remarkable degree of caloric-state-dependent synaptic plasticity. Despite the important role of excitatory input, its source(s) has been unknown. Here, through the use of Cre-recombinase-enabled, cell-specific neuron mapping techniques in mice, we have discovered strong excitatory drive that, unexpectedly, emanates from the hypothalamic paraventricular nucleus, specifically from subsets of neurons expressing thyrotropin-releasing hormone (TRH) and pituitary adenylate cyclase-activating polypeptide (PACAP, also known as ADCYAP1). Chemogenetic stimulation of these afferent neurons in sated mice markedly activates AgRP neurons and induces intense feeding. Conversely, acute inhibition in mice with caloric-deficiency-induced hunger decreases feeding. Discovery of these afferent neurons capable of triggering hunger advances understanding of how this intense motivational state is regulated.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Fome/fisiologia , Vias Neurais/fisiologia , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Proteína Relacionada com Agouti/deficiência , Animais , Apetite/efeitos dos fármacos , Apetite/fisiologia , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Mapeamento Encefálico , Rastreamento de Células , Clozapina/análogos & derivados , Clozapina/farmacologia , Dependovirus/genética , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Feminino , Privação de Alimentos , Fome/efeitos dos fármacos , Integrases/metabolismo , Masculino , Camundongos , Vias Neurais/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Fragmentos de Peptídeos/deficiência , Fragmentos de Peptídeos/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Vírus da Raiva/genética , Resposta de Saciedade/fisiologia , Hormônio Liberador de Tireotropina/metabolismo
7.
Annu Rev Physiol ; 78: 207-21, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26863324

RESUMO

Although it has been known for more than a century that the brain controls overall energy balance and adiposity by regulating feeding behavior and energy expenditure, the roles for individual brain regions and neuronal subtypes were not fully understood until recently. This area of research is active, and as such our understanding of the central regulation of energy balance is continually being refined as new details emerge. Much of what we now know stems from the discoveries of leptin and the hypothalamic melanocortin system. Hypothalamic circuits play a crucial role in the control of feeding and energy expenditure, and within the hypothalamus, the arcuate nucleus (ARC) functions as a gateway for hormonal signals of energy balance, such as leptin. It is also well established that the ARC is a primary residence for hypothalamic melanocortinergic neurons. The paraventricular hypothalamic nucleus (PVH) receives direct melanocortin input, along with other integrated signals that affect energy balance, and mediates the majority of hypothalamic output to control both feeding and energy expenditure. Herein, we review in detail the structure and function of the ARC-PVH circuit in mediating leptin signaling and in regulating energy balance.


Assuntos
Metabolismo Energético/fisiologia , Leptina/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Ingestão de Alimentos/fisiologia , Humanos , Neurônios/metabolismo , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/metabolismo
8.
J Physiol ; 597(12): 3217-3232, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31054267

RESUMO

KEY POINTS: Alpha-melanocyte stimulating hormone (α-MSH) is an anorexigenic peptide. Injection of the α-MSH analog MTII into the ventral tegmental area (VTA) decreases food and sucrose intake and food reward. Melanocortin-3 receptors (MC3R) are highly expressed in the VTA, suggesting that the effects of intra-VTA α-MSH may be mediated by α-MSH changing the activity of MC3R-expressing VTA neurons. α-MSH increased the firing rate of MC3R VTA neurons in acute brain slices from mice, although it did not affect the firing rate of non-MC3R VTA neurons. The α-MSH induced increase in MC3R neuron firing rate is probably activity-dependent, and was independent of fast synaptic transmission and intracellular Ca2+ levels. These results help us to better understand how α-MSH acts in the VTA to affect feeding and other dopamine-dependent behaviours. ABSTRACT: The mesocorticolimbic dopamine system, the brain's reward system, regulates multiple behaviours, including food intake and food reward. There is substantial evidence that the melanocortin system of the hypothalamus, an important neural circuit controlling feeding and body weight, interacts with the mesocorticolimbic dopamine system to affect feeding, food reward and body weight. For example, melanocortin-3 receptors (MC3Rs) are expressed in the ventral tegmental area (VTA) and our laboratory previously showed that intra-VTA injection of the MC3R agonist, MTII, decreases home-cage food intake and operant responding for sucrose pellets. However, the cellular mechanisms underlying the effects of intra-VTA alpha-melanocyte stimulating hormone (α-MSH) on feeding and food reward are unknown. To determine how α-MSH acts in the VTA to affect feeding, we performed electrophysiological recordings in acute brain slices from mice expressing enhanced yellow fluorescent protein in MC3R neurons to test how α-MSH affects the activity of VTA MC3R neurons. α-MSH significantly increased the firing rate of VTA MC3R neurons without altering the activity of non-MC3R expressing VTA neurons. In addition, the α-MSH-induced increase in MC3R neuron activity was independent of fast synaptic transmission and intracellular Ca2+ levels. Finally, we show that the effect of α-MSH on MC3R neuron firing rate is probably activity-dependent. Overall, these studies provide an important advancement in the understanding of how α-MSH acts in the VTA to affect feeding and food reward.


Assuntos
Receptor Tipo 3 de Melanocortina/fisiologia , Área Tegmentar Ventral/fisiologia , alfa-MSH/fisiologia , Animais , Feminino , Técnicas In Vitro , Masculino , Camundongos Transgênicos , Neurônios/fisiologia
9.
Proc Natl Acad Sci U S A ; 113(14): E2073-82, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27001850

RESUMO

Previous studies implicate the hypothalamic ventromedial nucleus (VMN) in glycemic control. Here, we report that selective inhibition of the subset of VMN neurons that express the transcription factor steroidogenic-factor 1 (VMN(SF1) neurons) blocks recovery from insulin-induced hypoglycemia whereas, conversely, activation of VMN(SF1) neurons causes diabetes-range hyperglycemia. Moreover, this hyperglycemic response is reproduced by selective activation of VMN(SF1) fibers projecting to the anterior bed nucleus of the stria terminalis (aBNST), but not to other brain areas innervated by VMN(SF1) neurons. We also report that neurons in the lateral parabrachial nucleus (LPBN), a brain area that is also implicated in the response to hypoglycemia, make synaptic connections with the specific subset of glucoregulatory VMN(SF1) neurons that project to the aBNST. These results collectively establish a physiological role in glucose homeostasis for VMN(SF1) neurons and suggest that these neurons are part of an ascending glucoregulatory LPBN→VMN(SF1)→aBNST neurocircuit.


Assuntos
Glicemia/metabolismo , Neurônios Aferentes/fisiologia , Núcleo Hipotalâmico Ventromedial/fisiologia , Animais , Insulina/administração & dosagem , Camundongos , Núcleo Hipotalâmico Ventromedial/citologia
10.
Nature ; 491(7424): 357-63, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23151578

RESUMO

Although it is a widely held thought that direct hormone action on peripheral tissues is sufficient to mediate the control of nutrient handling, the role of the central nervous system in certain aspects of metabolism has long been recognized. Furthermore, recent findings have suggested a more general role for the central nervous system in metabolic control, and have revealed the importance of a number of cues and hypothalamic circuits. The brain's contributions to metabolic control are more readily revealed and play a crucial part in catabolic states or in hormone deficiencies that mimic starvation.


Assuntos
Sistema Nervoso Central/metabolismo , Metabolismo Energético/fisiologia , Animais , Sistema Nervoso Autônomo/fisiologia , Humanos , Melanocortinas/metabolismo , Sistemas Neurossecretores/fisiologia , Transdução de Sinais/fisiologia
11.
Proc Natl Acad Sci U S A ; 111(36): 13193-8, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25157144

RESUMO

Activation of melanocortin-4 receptors (MC4Rs) restrains feeding and prevents obesity; however, the identity, location, and axonal projections of the neurons bearing MC4Rs that control feeding remain unknown. Reexpression of MC4Rs on single-minded 1 (SIM1)(+) neurons in mice otherwise lacking MC4Rs is sufficient to abolish hyperphagia. Thus, MC4Rs on SIM1(+) neurons, possibly in the paraventricular hypothalamus (PVH) and/or amygdala, regulate food intake. It is unknown, however, whether they are also necessary, a distinction required for excluding redundant sites of action. Hence, the location and nature of obesity-preventing MC4R-expressing neurons are unknown. Here, by deleting and reexpressing MC4Rs from cre-expressing neurons, establishing both necessity and sufficiency, we demonstrate that the MC4R-expressing neurons regulating feeding are SIM1(+), located in the PVH, glutamatergic and not GABAergic, and do not express oxytocin, corticotropin-releasing hormone, vasopressin, or prodynorphin. Importantly, these excitatory MC4R-expressing PVH neurons are synaptically connected to neurons in the parabrachial nucleus, which relays visceral information to the forebrain. This suggests a basis for the feeding-regulating effects of MC4Rs.


Assuntos
Comportamento Alimentar , Glutamatos/metabolismo , Neurônios/metabolismo , Núcleos Parabraquiais/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Sinapses/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Peso Corporal , Dependovirus/metabolismo , Metabolismo Energético , Neurônios GABAérgicos/metabolismo , Deleção de Genes , Injeções , Integrases/metabolismo , Camundongos , Neuropeptídeos/metabolismo , Proteínas Repressoras/metabolismo , Reprodutibilidade dos Testes , Técnicas Estereotáxicas , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
12.
J Neurosci ; 35(5): 1905-20, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25653351

RESUMO

GABAergic synaptic transmission plays an important role in resetting and synchronizing circadian rhythms in the suprachiasmatic nucleus (SCN). Although the circadian modulation of intrinsic membrane currents and biochemical signaling have been examined in the SCN, the modulation of specific synaptic pathways within the SCN is unexplored. In addition, little is known about the functional properties of these pathways, including which ones involve GABAA receptors (GABAA-Rs). In brain slices obtained from mice, we examined synaptic responses originating from the SCN neurons expressing vasoactive intestinal peptide (VIP+ neurons). Focusing on the local projection within the ventromedial SCN, we found that VIP+ afferents provided input onto 49% of neurons with a preference for VIP-negative (VIP-) neurons. Responses were mediated by GABAA-Rs. The projection was sparsely connected and preferentially targeted a subset of SCN neurons unrelated to postsynaptic VIP expression. For most aspects of VIP+ network output, there was no circadian regulation. Excitability and spontaneous firing of the presynaptic VIP+ neurons were unchanged between day and night, and their network connectivity and synaptic function up through the evoked synaptic conductance were also unchanged. On the other hand, VIP+ input onto VIP- neurons became less inhibitory at night suggesting a postsynaptic alteration in the coupling of GABAA-R conductances to action potential firing. These data suggest that components of the VIP network and its synaptic output up through GABAA-R opening are invariant during the circadian cycle, but the effect on action potential firing is modulated by postsynaptic processes occurring after GABAA-R channel opening.


Assuntos
Ritmo Circadiano , Neurônios GABAérgicos/metabolismo , Neurônios Aferentes/metabolismo , Receptores de GABA-A/metabolismo , Núcleo Supraquiasmático/metabolismo , Potenciais Sinápticos , Peptídeo Intestinal Vasoativo/metabolismo , Potenciais de Ação , Animais , Feminino , Neurônios GABAérgicos/fisiologia , Masculino , Camundongos , Neurônios Aferentes/fisiologia , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/fisiologia , Sinapses/metabolismo , Sinapses/fisiologia , Peptídeo Intestinal Vasoativo/genética , Ácido gama-Aminobutírico/metabolismo
13.
J Neurosci ; 34(46): 15306-18, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25392498

RESUMO

The paraventricular nucleus of the hypothalamus (PVH) contains a heterogeneous cluster of Sim1-expressing cell types that comprise a major autonomic output nucleus and play critical roles in the control of food intake and energy homeostasis. The roles of specific PVH neuronal subtypes in energy balance have yet to be defined, however. The PVH contains nitric oxide synthase-1 (Nos1)-expressing (Nos1(PVH)) neurons of unknown function; these represent a subset of the larger population of Sim1-expressing PVH (Sim1(PVH)) neurons. To determine the role of Nos1(PVH) neurons in energy balance, we used Cre-dependent viral vectors to both map their efferent projections and test their functional output in mice. Here we show that Nos1(PVH) neurons project to hindbrain and spinal cord regions important for food intake and energy expenditure control. Moreover, pharmacogenetic activation of Nos1(PVH) neurons suppresses feeding to a similar extent as Sim1(PVH) neurons, and increases energy expenditure and activity. Furthermore, we found that oxytocin-expressing PVH neurons (OXT(PVH)) are a subset of Nos1(PVH) neurons. OXT(PVH) cells project to preganglionic, sympathetic neurons in the thoracic spinal cord and increase energy expenditure upon activation, though not to the same extent as Nos1(PVH) neurons; their activation fails to alter feeding, however. Thus, Nos1(PVH) neurons promote negative energy balance through changes in feeding and energy expenditure, whereas OXT(PVH) neurons regulate energy expenditure alone, suggesting a crucial role for non-OXT Nos1(PVH) neurons in feeding regulation.


Assuntos
Regulação do Apetite/fisiologia , Metabolismo Energético/fisiologia , Neurônios/fisiologia , Óxido Nítrico Sintase Tipo I/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Animais , Regulação do Apetite/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Temperatura Corporal/fisiologia , Ingestão de Alimentos/fisiologia , Metabolismo Energético/genética , Masculino , Camundongos , Camundongos Transgênicos , Vias Neurais/citologia , Vias Neurais/fisiologia , Técnicas de Rastreamento Neuroanatômico , Óxido Nítrico Sintase Tipo I/genética , Ocitocina/fisiologia , Núcleo Hipotalâmico Paraventricular/anatomia & histologia , Proteínas Repressoras/fisiologia , Rombencéfalo/anatomia & histologia , Rombencéfalo/citologia , Rombencéfalo/fisiologia , Medula Espinal/anatomia & histologia , Medula Espinal/citologia , Medula Espinal/fisiologia
14.
Circ Res ; 111(6): 728-38, 2012 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-22730442

RESUMO

RATIONALE: Decreased fatty acid oxidation (FAO) with increased reliance on glucose are hallmarks of metabolic remodeling that occurs in pathological cardiac hypertrophy and is associated with decreased myocardial energetics and impaired cardiac function. To date, it has not been tested whether prevention of the metabolic switch that occurs during the development of cardiac hypertrophy has unequivocal benefits on cardiac function and energetics. OBJECTIVE: Because malonyl CoA production via acetyl CoA carboxylase 2 (ACC2) inhibits the entry of long chain fatty acids into the mitochondria, we hypothesized that mice with a cardiac-specific deletion of ACC2 (ACC2H-/-) would maintain cardiac FAO and improve function and energetics during the development of pressure-overload hypertrophy. METHODS AND RESULTS: ACC2 deletion led to a significant reduction in cardiac malonyl CoA levels. In isolated perfused heart experiments, left ventricular function and oxygen consumption were similar in ACC2H-/- mice despite an ≈60% increase in FAO compared with controls (CON). After 8 weeks of pressure overload via transverse aortic constriction (TAC), ACC2H-/- mice exhibited a substrate utilization profile similar to sham animals, whereas CON-TAC hearts had decreased FAO with increased glycolysis and anaplerosis. Myocardial energetics, assessed by 31P nuclear magnetic resonance spectroscopy, and cardiac function were maintained in ACC2H-/- after 8 weeks of TAC. Furthermore, ACC2H-/--TAC demonstrated an attenuation of cardiac hypertrophy with a significant reduction in fibrosis relative to CON-TAC. CONCLUSIONS: These data suggest that reversion to the fetal metabolic profile in chronic pathological hypertrophy is associated with impaired myocardial function and energetics and maintenance of the inherent cardiac metabolic profile and mitochondrial oxidative capacity is a viable therapeutic strategy.


Assuntos
Acetil-CoA Carboxilase/metabolismo , Cardiomegalia/metabolismo , Miocárdio/enzimologia , Remodelação Ventricular , Acetil-CoA Carboxilase/genética , Animais , Aorta/patologia , Western Blotting , Cardiomegalia/genética , Carnitina/análogos & derivados , Carnitina/metabolismo , Constrição Patológica , Ácidos Graxos/metabolismo , Feminino , Fibrose , Coração/fisiopatologia , Técnicas In Vitro , Masculino , Malonil Coenzima A/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Cardíacas/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Oxirredução , Pressão
15.
Adv Sci (Weinh) ; 11(31): e2400437, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38885417

RESUMO

SH2B1 mutations are associated with obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD) in humans. Global deletion of Sh2b1 results in severe obesity, type 2 diabetes, and MASLD in mice. Neuron-specific restoration of SH2B1 rescues the obesity phenotype of Sh2b1-null mice, indicating that the brain is a main SH2B1 target. However, SH2B1 neurocircuits remain elusive. SH2B1-expressing neurons in the paraventricular hypothalamus (PVHSH2B1) and a PVHSH2B1→dorsal raphe nucleus (DRN) neurocircuit are identified here. PVHSH2B1 axons monosynaptically innervate DRN neurons. Optogenetic stimulation of PVHSH2B1 axonal fibers in the DRN suppresses food intake. Chronic inhibition of PVHSH2B1 neurons causes obesity. In male and female mice, either embryonic-onset or adult-onset deletion of Sh2b1 in PVH neurons causes energy imbalance, obesity, insulin resistance, glucose intolerance, and MASLD. Ablation of Sh2b1 in the DRN-projecting PVHSH2B1 subpopulation also causes energy imbalance, obesity, and metabolic disorders. Conversely, SH2B1 overexpression in either total or DRN-projecting PVHSH2B1 neurons protects against diet-induced obesity. SH2B1 binds to TrkB and enhances brain-derived neurotrophic factor (BDNF) signaling. Ablation of Sh2b1 in PVHSH2B1 neurons induces BDNF resistance in the PVH, contributing to obesity. In conclusion, these results unveil a previously unrecognized PVHSH2B1→DRN neurocircuit through which SH2B1 defends against obesity by enhancing BDNF/TrkB signaling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Obesidade , Núcleo Hipotalâmico Paraventricular , Animais , Obesidade/metabolismo , Obesidade/genética , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Masculino , Feminino , Núcleo Hipotalâmico Paraventricular/metabolismo , Modelos Animais de Doenças , Doenças Metabólicas/metabolismo , Doenças Metabólicas/genética , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , Núcleo Dorsal da Rafe/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Neurônios/metabolismo
16.
Sci Transl Med ; 16(738): eadk1866, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38478630

RESUMO

Metabolic dysfunction-associated steatohepatitis (MASH), formerly known as nonalcoholic steatohepatitis (NASH), is an advanced stage of metabolic fatty liver disease. The pathogenic mechanisms of MASH center on hepatocyte injury and the ensuing immune response within the liver microenvironment. Recent work has implicated TREM2+ macrophages in various disease conditions, and substantial induction of TREM2+ NASH-associated macrophages (NAMs) serves as a hallmark of metabolic liver disease. Despite this, the mechanisms through which NAMs contribute to MASH pathogenesis remain poorly understood. Here, we identify membrane-spanning 4-domains a7 (MS4A7) as a NAM-specific pathogenic factor that exacerbates MASH progression in mice. Hepatic MS4A7 expression was strongly induced in mouse and human MASH and associated with the severity of liver injury. Whole-body and myeloid-specific ablation of Ms4a7 alleviated diet-induced MASH pathologies in male mice. We demonstrate that exposure to lipid droplets (LDs), released upon injury of steatotic hepatocytes, triggered NAM induction and exacerbated MASH-associated liver injury in an MS4A7-dependent manner. Mechanistically, MS4A7 drove NLRP3 inflammasome activation via direct physical interaction and shaped disease-associated cell states within the liver microenvironment. This work reveals the LD-MS4A7-NLRP3 inflammasome axis as a pathogenic driver of MASH progression and provides insights into the role of TREM2+ macrophages in disease pathogenesis.


Assuntos
Inflamassomos , Hepatopatia Gordurosa não Alcoólica , Animais , Humanos , Masculino , Camundongos , Inflamassomos/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Receptores Imunológicos/metabolismo
17.
Proc Natl Acad Sci U S A ; 107(16): 7598-603, 2010 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-20368432

RESUMO

Deletion of acetyl CoA carboxylase-2 (Acc2) reportedly causes leanness in the setting of hyperphagia. To determine the cellular basis for these effects, we generated a mouse model in which Acc2 can be selectively deleted by the action of Cre recombinase. Deletion of Acc2 from skeletal muscle, the predominant site of Acc2 expression, had no effect on body weight, food intake, or body composition. When Acc2 was inactivated in the germline, Acc2 knockout (Acc2KO) mice displayed no differences in body weight, food intake, body composition, or glucose homeostasis as compared to controls on chow or high fat diet. Total malonyl CoA content and fatty acid oxidation rates in skeletal muscle of Acc2KO mice were unchanged, suggesting metabolic compensation in response to the loss of Acc2. The limited impact of Acc2 deletion on energy balance raises the possibility that selective pharmacological inhibition of Acc2 for the treatment of obesity may be ineffective.


Assuntos
Acetil-CoA Carboxilase/genética , Acetil-CoA Carboxilase/fisiologia , Alelos , Animais , Composição Corporal , Peso Corporal , Éxons , Deleção de Genes , Genótipo , Integrases/metabolismo , Malonil Coenzima A/metabolismo , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Obesidade , Fenótipo
18.
J Clin Invest ; 133(19)2023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37581939

RESUMO

The adipose-derived hormone leptin acts via its receptor (LepRb) in the brain to control energy balance. A potentially unidentified population of GABAergic hypothalamic LepRb neurons plays key roles in the restraint of food intake and body weight by leptin. To identify markers for candidate populations of LepRb neurons in an unbiased manner, we performed single-nucleus RNA-Seq of enriched mouse hypothalamic LepRb cells, identifying several previously unrecognized populations of hypothalamic LepRb neurons. Many of these populations displayed strong conservation across species, including GABAergic Glp1r-expressing LepRb (LepRbGlp1r) neurons, which expressed more Lepr than other LepRb cell populations. Ablating Lepr from LepRbGlp1r cells provoked hyperphagic obesity without impairing energy expenditure. Similarly, improvements in energy balance caused by Lepr reactivation in GABA neurons of otherwise Lepr-null mice required Lepr expression in GABAergic Glp1r-expressing neurons. Furthermore, restoration of Glp1r expression in LepRbGlp1r neurons in otherwise Glp1r-null mice enabled food intake suppression by the GLP1R agonist, liraglutide. Thus, the conserved GABAergic LepRbGlp1r neuron population plays crucial roles in the suppression of food intake by leptin and GLP1R agonists.


Assuntos
Leptina , Obesidade , Camundongos , Animais , Leptina/genética , Leptina/metabolismo , Obesidade/genética , Obesidade/prevenção & controle , Obesidade/metabolismo , Hipotálamo/metabolismo , Camundongos Knockout , Neurônios GABAérgicos/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Ingestão de Alimentos/genética
19.
Neuroimage Clin ; 36: 103164, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36044792

RESUMO

Early life stress (ELS) and major depressive disorder (MDD) share neural network abnormalities. However, it is unclear how ELS and MDD may separately and/or jointly relate to brain networks, and whether neural differences exist between depressed individuals with vs without ELS. Moreover, prior work evaluated static versus dynamic network properties, a critical gap considering brain networks show changes in coordinated activity over time. Seventy-one unmedicated females with and without childhood sexual abuse (CSA) histories and/or MDD completed a resting state scan and a stress task in which cortisol and affective ratings were collected. Recurring functional network co-activation patterns (CAPs) were examined and time in CAP (number of times each CAP is expressed) and transition frequencies (transitioning between different CAPs) were computed. The effects of MDD and CSA on CAP metrics were examined and CAP metrics were correlated with depression and stress-related variables. Results showed that MDD, but not CSA, related to CAP metrics. Specifically, individuals with MDD (N = 35) relative to HCs (N = 36), spent more time in a posterior default mode (DMN)-frontoparietal network (FPN) CAP and transitioned more frequently between posterior DMN-FPN and prototypical DMN CAPs. Across groups, more time spent in a posterior DMN-FPN CAP and greater DMN-FPN and prototypical DMN CAP transition frequencies were linked to higher rumination. Imbalances between the DMN and the FPN appear central to MDD and might contribute to MDD-related cognitive dysfunction, including rumination. Unexpectedly, CSA did not modulate such dysfunctions, a finding that needs to be replicated by future studies with larger sample sizes.


Assuntos
Transtorno Depressivo Maior , Delitos Sexuais , Feminino , Criança , Humanos , Vias Neurais , Imageamento por Ressonância Magnética/métodos , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico/métodos
20.
Sci Signal ; 15(733): eabj8204, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35536884

RESUMO

Variants in the gene encoding ankyrin repeat and SOCS box-containing 4 (ASB4) are linked to human obesity. Here, we characterized the pathways underlying the metabolic functions of ASB4. Hypothalamic Asb4 expression was suppressed by fasting in wild-type mice but not in mice deficient in AgRP, which encodes Agouti-related protein (AgRP), an appetite-stimulating hormone, suggesting that ASB4 is a negative target of AgRP. Many ASB4 neurons in the brain were adjacent to AgRP terminals, and feeding induced by AgRP neuronal activation was disrupted in Asb4-deficient mice. Acute knockdown of Asb4 in the brain caused marked hyperphagia due to increased meal size, and Asb4 deficiency led to increased meal size and food intake at the onset of refeeding, when very large meals were consumed. Asb4-deficient mice were resistant to the meal-terminating effects of exogenously administered calcitonin and showed decreased neuronal expression of Calcr, which encodes the calcitonin receptor. Pro-opiomelanocortin (POMC) neurons in the arcuate nucleus in mice are involved in glucose homeostasis, and Asb4 deficiency specifically in POMC neurons resulted in glucose intolerance that was independent of obesity. Furthermore, individuals with type 2 diabetes showed reduced ASB4 abundance in the infundibular nuclei, the human equivalent of the arcuate nucleus. Together, our results indicate that ASB4 acts in the brain to improve glucose homeostasis and to induce satiety after substantial meals, particularly those after food deprivation.


Assuntos
Diabetes Mellitus Tipo 2 , Neuropeptídeos , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/farmacologia , Animais , Calcitonina/metabolismo , Calcitonina/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Homeostase , Hipotálamo/metabolismo , Camundongos , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Obesidade/genética , Obesidade/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA