Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 1966, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38438345

RESUMO

The "dorsal pons", or "dorsal pontine tegmentum" (dPnTg), is part of the brainstem. It is a complex, densely packed region whose nuclei are involved in regulating many vital functions. Notable among them are the parabrachial nucleus, the Kölliker Fuse, the Barrington nucleus, the locus coeruleus, and the dorsal, laterodorsal, and ventral tegmental nuclei. In this study, we applied single-nucleus RNA-seq (snRNA-seq) to resolve neuronal subtypes based on their unique transcriptional profiles and then used multiplexed error robust fluorescence in situ hybridization (MERFISH) to map them spatially. We sampled ~1 million cells across the dPnTg and defined the spatial distribution of over 120 neuronal subtypes. Our analysis identified an unpredicted high transcriptional diversity in this region and pinpointed the unique marker genes of many neuronal subtypes. We also demonstrated that many neuronal subtypes are transcriptionally similar between humans and mice, enhancing this study's translational value. Finally, we developed a freely accessible, GPU and CPU-powered dashboard ( http://harvard.heavy.ai:6273/ ) that combines interactive visual analytics and hardware-accelerated SQL into a data science framework to allow the scientific community to query and gain insights into the data.


Assuntos
Ascomicetos , Núcleos Parabraquiais , Tegmento Pontino , Humanos , Animais , Camundongos , Hibridização in Situ Fluorescente , Tronco Encefálico , Locus Cerúleo
2.
bioRxiv ; 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-38014113

RESUMO

The "dorsal pons", or "dorsal pontine tegmentum" (dPnTg), is part of the brainstem. It is a complex, densely packed region whose nuclei are involved in regulating many vital functions. Notable among them are the parabrachial nucleus, the Kölliker Fuse, the Barrington nucleus, the locus coeruleus, and the dorsal, laterodorsal, and ventral tegmental nuclei. In this study, we applied single-nucleus RNA-seq (snRNA-seq) to resolve neuronal subtypes based on their unique transcriptional profiles and then used multiplexed error robust fluorescence in situ hybridization (MERFISH) to map them spatially. We sampled ~1 million cells across the dPnTg and defined the spatial distribution of over 120 neuronal subtypes. Our analysis identified an unpredicted high transcriptional diversity in this region and pinpointed many neuronal subtypes' unique marker genes. We also demonstrated that many neuronal subtypes are transcriptionally similar between humans and mice, enhancing this study's translational value. Finally, we developed a freely accessible, GPU and CPU-powered dashboard (http://harvard.heavy.ai:6273/) that combines interactive visual analytics and hardware-accelerated SQL into a data science framework to allow the scientific community to query and gain insights into the data.

3.
Nature ; 620(7972): 154-162, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37495689

RESUMO

Fasting initiates a multitude of adaptations to allow survival. Activation of the hypothalamic-pituitary-adrenal (HPA) axis and subsequent release of glucocorticoid hormones is a key response that mobilizes fuel stores to meet energy demands1-5. Despite the importance of the HPA axis response, the neural mechanisms that drive its activation during energy deficit are unknown. Here, we show that fasting-activated hypothalamic agouti-related peptide (AgRP)-expressing neurons trigger and are essential for fasting-induced HPA axis activation. AgRP neurons do so through projections to the paraventricular hypothalamus (PVH), where, in a mechanism not previously described for AgRP neurons, they presynaptically inhibit the terminals of tonically active GABAergic afferents from the bed nucleus of the stria terminalis (BNST) that otherwise restrain activity of corticotrophin-releasing hormone (CRH)-expressing neurons. This disinhibition of PVHCrh neurons requires γ-aminobutyric acid (GABA)/GABA-B receptor signalling and potently activates the HPA axis. Notably, stimulation of the HPA axis by AgRP neurons is independent of their induction of hunger, showing that these canonical 'hunger neurons' drive many distinctly different adaptations to the fasted state. Together, our findings identify the neural basis for fasting-induced HPA axis activation and uncover a unique means by which AgRP neurons activate downstream neurons: through presynaptic inhibition of GABAergic afferents. Given the potency of this disinhibition of tonically active BNST afferents, other activators of the HPA axis, such as psychological stress, may also work by reducing BNST inhibitory tone onto PVHCrh neurons.


Assuntos
Jejum , Sistema Hipotálamo-Hipofisário , Neurônios , Sistema Hipófise-Suprarrenal , Proteína Relacionada com Agouti/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Jejum/fisiologia , Neurônios GABAérgicos/metabolismo , Ácido gama-Aminobutírico/metabolismo , Sistema Hipotálamo-Hipofisário/citologia , Sistema Hipotálamo-Hipofisário/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Hipófise-Suprarrenal/citologia , Sistema Hipófise-Suprarrenal/inervação , Sistema Hipófise-Suprarrenal/metabolismo , Terminações Pré-Sinápticas/metabolismo , Núcleos Septais/citologia , Núcleos Septais/metabolismo
4.
J Physiol ; 601(16): 3499-3532, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37291801

RESUMO

In addition to its renal and cardiovascular functions, angiotensin signalling is thought to be responsible for the increases in salt and water intake caused by hypovolaemia. However, it remains unclear whether these behaviours require angiotensin production in the brain or liver. Here, we use in situ hybridization to identify tissue-specific expression of the genes required for producing angiotensin peptides, and then use conditional genetic deletion of the angiotensinogen gene (Agt) to test whether production in the brain or liver is necessary for sodium appetite and thirst. In the mouse brain, we identified expression of Agt (the precursor for all angiotensin peptides) in a large subset of astrocytes. We also identified Ren1 and Ace (encoding enzymes required to produce angiotensin II) expression in the choroid plexus, and Ren1 expression in neurons within the nucleus ambiguus compact formation. In the liver, we confirmed that Agt is widely expressed in hepatocytes. We next tested whether thirst and sodium appetite require angiotensinogen production in astrocytes or hepatocytes. Despite virtually eliminating expression in the brain, deleting astrocytic Agt did not reduce thirst or sodium appetite. Despite markedly reducing angiotensinogen in the blood, eliminating Agt from hepatocytes did not reduce thirst or sodium appetite, and in fact, these mice consumed the largest amounts of salt and water after sodium deprivation. Deleting Agt from both astrocytes and hepatocytes also did not prevent thirst or sodium appetite. Our findings suggest that angiotensin signalling is not required for sodium appetite or thirst and highlight the need to identify alternative signalling mechanisms. KEY POINTS: Angiotensin signalling is thought to be responsible for the increased thirst and sodium appetite caused by hypovolaemia, producing elevated water and sodium intake. Specific cells in separate brain regions express the three genes needed to produce angiotensin peptides, but brain-specific deletion of the angiotensinogen gene (Agt), which encodes the lone precursor for all angiotensin peptides, did not reduce thirst or sodium appetite. Double-deletion of Agt from brain and liver also did not reduce thirst or sodium appetite. Liver-specific deletion of Agt reduced circulating angiotensinogen levels without reducing thirst or sodium appetite. Instead, these angiotensin-deficient mice exhibited an enhanced sodium appetite. Because the physiological mechanisms controlling thirst and sodium appetite continued functioning without angiotensin production in the brain and liver, understanding these mechanisms requires a renewed search for the hypovolaemic signals necessary for activating each behaviour.


Assuntos
Angiotensinogênio , Sódio , Camundongos , Animais , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Apetite/fisiologia , Sede/fisiologia , Hipovolemia , Astrócitos/metabolismo , Hepatócitos/metabolismo , Angiotensina II/metabolismo , Cloreto de Sódio , Água
5.
J Neurophysiol ; 129(2): 347-355, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36542422

RESUMO

The parabrachial nucleus (PB) in the upper brainstem receives interoceptive information and sends a massive output projection directly to the cerebral cortex. Its glutamatergic axons primarily target the midinsular cortex, and we have proposed that this PB-insular projection promotes arousal. Here, we test whether stimulating this projection causes wakefulness. We combined optogenetics and video-electroencephalography (vEEG) in mice to test this hypothesis by stimulating PB axons in the insular cortex. Stimulating this projection did not alter the cortical EEG or awaken mice. Also, despite a tendency toward aversion, PB-insular stimulation did not significantly alter real-time place preference (RTPP). These results are not consistent with the hypothesis that the direct PB-insular projection is part of the ascending arousal system.NEW & NOTEWORTHY A brainstem region critical for wakefulness overlaps the medial parabrachial nucleus (PB) and has functional and direct axonal connectivity with the insular cortex. In this study, we hypothesized that this direct projection from the PB to the insular cortex promotes arousal. However, photostimulating PB axons in the insular cortex did not alter the cortical EEG or awaken mice. This information constrains the possible circuit connections through which brainstem neurons may sustain arousal.


Assuntos
Tronco Encefálico , Córtex Cerebral , Camundongos , Animais , Tronco Encefálico/fisiologia , Eletroencefalografia , Nível de Alerta , Vigília
6.
Mol Metab ; 66: 101622, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36307046

RESUMO

OBJECTIVE: RGS2 is a GTPase activating protein that modulates GPCR-Gα signaling and mice lacking RGS2 globally exhibit metabolic alterations. While RGS2 is known to be broadly expressed throughout the body including the brain, the relative contribution of brain RGS2 to metabolic homeostasis remains unknown. The purpose of this study was to characterize RGS2 expression in the paraventricular nucleus of hypothalamus (PVN) and test its role in metabolic homeostasis. METHODS: We used a combination of RNAscope in situ hybridization (ISH), immunohistochemistry, and bioinformatic analyses to characterize the pattern of Rgs2 expression in the PVN. We then created mice lacking Rgs2 either prenatally or postnatally in the PVN and evaluated their metabolic consequences. RESULTS: RNAscope ISH analysis revealed a broad but regionally enriched Rgs2 mRNA expression throughout the mouse brain, with the highest expression being observed in the PVN along with several other brain regions, such as the arcuate nucleus of hypothalamus and the dorsal raphe nucleus. Within the PVN, we found that Rgs2 is specifically enriched in CRH+ endocrine neurons and is further increased by calorie restriction. Functionally, although Sim1-Cre-mediated prenatal deletion of Rgs2 in PVN neurons had no major effects on metabolic homeostasis, AAV-mediated adult deletion of Rgs2 in the PVN led to significantly increased food intake, body weight (both fat and fat-free masses), body length, and blood glucose levels in both male and female mice. Strikingly, we found that prolonged postnatal loss of Rgs2 leads to neuronal cell death in the PVN, while rapid body weight gain in the early phase of viral-mediated PVN Rgs2 deletion is independent of PVN neuronal loss. CONCLUSIONS: Our results provide the first evidence to show that PVN Rgs2 expression is not only sensitive to metabolic challenge but also critically required for PVN endocrine neurons to function and maintain metabolic homeostasis.


Assuntos
Metabolismo Energético , Núcleo Hipotalâmico Paraventricular , Camundongos , Animais , Masculino , Feminino , Núcleo Hipotalâmico Paraventricular/metabolismo , Metabolismo Energético/fisiologia , Obesidade/metabolismo , Homeostase , Peso Corporal
8.
Am J Physiol Regul Integr Comp Physiol ; 320(3): R342-R361, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33296280

RESUMO

Previously, we identified a population of neurons in the hindbrain tegmentum, bordering the locus coeruleus (LC). We named this population the pre-locus coeruleus (pre-LC) because in rats its neurons lie immediately rostral to the LC. In mice, however, pre-LC and LC neurons intermingle, making them difficult to distinguish. Here, we use molecular markers and anterograde tracing to clarify the location and distribution of pre-LC neurons in mice, relative to rats. First, we colocalized the transcription factor FoxP2 with the activity marker Fos to identify pre-LC neurons in sodium-deprived rats and show their distribution relative to surrounding catecholaminergic and cholinergic neurons. Next, we used sodium depletion and chemogenetic activation of the aldosterone-sensitive HSD2 neurons in the nucleus of the solitary tract (NTS) to identify the homologous population of pre-LC neurons in mice, along with a related population in the central lateral parabrachial nucleus. Using Cre-reporter mice for Pdyn, we confirmed that most of these sodium-depletion-activated neurons are dynorphinergic. Finally, after confirming that these neurons receive excitatory input from the NTS and paraventricular hypothalamic nucleus, plus convergent input from the inhibitory AgRP neurons in the arcuate hypothalamic nucleus, we identify a major, direct input projection from the medial prefrontal cortex. This new information on the location, distribution, and input to pre-LC neurons provides a neuroanatomical foundation for cell-type-specific investigation of their properties and functions in mice. Pre-LC neurons likely integrate homeostatic information from the brainstem and hypothalamus with limbic, contextual information from the cerebral cortex to influence ingestive behavior.


Assuntos
Encéfalo/fisiologia , Vias Neurais/fisiologia , Neurônios/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Neurônios Adrenérgicos/fisiologia , Ração Animal , Animais , Regulação do Apetite , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Biomarcadores/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Neurônios Colinérgicos/fisiologia , Dieta Hipossódica , Encefalinas/genética , Comportamento Alimentar , Feminino , Locus Cerúleo/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/citologia , Vias Neurais/metabolismo , Técnicas de Rastreamento Neuroanatômico , Neurônios/metabolismo , Precursores de Proteínas/genética , Ratos Sprague-Dawley , Proteínas Repressoras/genética
9.
Nature ; 578(7796): 610-614, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32076265

RESUMO

The sympathetic nervous system innervates peripheral organs to regulate their function and maintain homeostasis, whereas target cells also produce neurotrophic factors to promote sympathetic innervation1,2. The molecular basis of this bi-directional communication remains to be fully determined. Here we use thermogenic adipose tissue from mice as a model system to show that T cells, specifically γδ T cells, have a crucial role in promoting sympathetic innervation, at least in part by driving the expression of TGFß1 in parenchymal cells via the IL-17 receptor C (IL-17RC). Ablation of IL-17RC specifically in adipose tissue reduces expression of TGFß1 in adipocytes, impairs local sympathetic innervation and causes obesity and other metabolic phenotypes that are consistent with defective thermogenesis; innervation can be fully rescued by restoring TGFß1 expression. Ablating γδ Τ cells and the IL-17RC signalling pathway also impairs sympathetic innervation in other tissues such as salivary glands. These findings demonstrate coordination between T cells and parenchymal cells to regulate sympathetic innervation.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/inervação , Tecido Adiposo/metabolismo , Interleucina-17/metabolismo , Sistema Nervoso Simpático/fisiologia , Linfócitos T/metabolismo , Termogênese , Tecido Adiposo Marrom/metabolismo , Animais , Interleucina-17/deficiência , Interleucina-17/genética , Masculino , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Tecido Parenquimatoso/citologia , Transdução de Sinais , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
10.
Neuron ; 105(6): 1094-1111.e10, 2020 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-31955944

RESUMO

Interoception, the sense of internal bodily signals, is essential for physiological homeostasis, cognition, and emotions. While human insular cortex (InsCtx) is implicated in interoception, the cellular and circuit mechanisms remain unclear. We imaged mouse InsCtx neurons during two physiological deficiency states: hunger and thirst. InsCtx ongoing activity patterns reliably tracked the gradual return to homeostasis but not changes in behavior. Accordingly, while artificial induction of hunger or thirst in sated mice via activation of specific hypothalamic neurons (AgRP or SFOGLUT) restored cue-evoked food- or water-seeking, InsCtx ongoing activity continued to reflect physiological satiety. During natural hunger or thirst, food or water cues rapidly and transiently shifted InsCtx population activity to the future satiety-related pattern. During artificial hunger or thirst, food or water cues further shifted activity beyond the current satiety-related pattern. Together with circuit-mapping experiments, these findings suggest that InsCtx integrates visceral-sensory signals of current physiological state with hypothalamus-gated amygdala inputs that signal upcoming ingestion of food or water to compute a prediction of future physiological state.


Assuntos
Córtex Cerebral/fisiologia , Fome/fisiologia , Interocepção/fisiologia , Sede/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Clozapina/análogos & derivados , Clozapina/farmacologia , Sinais (Psicologia) , Feminino , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Vias Neurais/fisiologia , Imagem Óptica , Optogenética , Órgão Subfornical/fisiologia
11.
Proc Natl Acad Sci U S A ; 116(27): 13670-13679, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31213533

RESUMO

Leptin informs the brain about sufficiency of fuel stores. When insufficient, leptin levels fall, triggering compensatory increases in appetite. Falling leptin is first sensed by hypothalamic neurons, which then initiate adaptive responses. With regard to hunger, it is thought that leptin-sensing neurons work entirely via circuits within the central nervous system (CNS). Very unexpectedly, however, we now show this is not the case. Instead, stimulation of hunger requires an intervening endocrine step, namely activation of the hypothalamic-pituitary-adrenocortical (HPA) axis. Increased corticosterone then activates AgRP neurons to fully increase hunger. Importantly, this is true for 2 forms of low leptin-induced hunger, fasting and poorly controlled type 1 diabetes. Hypoglycemia, which also stimulates hunger by activating CNS neurons, albeit independently of leptin, similarly recruits and requires this pathway by which HPA axis activity stimulates AgRP neurons. Thus, HPA axis regulation of AgRP neurons is a previously underappreciated step in homeostatic regulation of hunger.


Assuntos
Fome/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Leptina/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Hormônio Adrenocorticotrópico/sangue , Animais , Ingestão de Alimentos/fisiologia , Jejum/fisiologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Insulina/farmacologia , Leptina/sangue , Masculino , Mifepristona/farmacologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Ratos , Receptores de Glucocorticoides/antagonistas & inibidores
12.
Nature ; 569(7755): 229-235, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31043739

RESUMO

The sympathetic nervous system drives brown and beige adipocyte thermogenesis through the release of noradrenaline from local axons. However, the molecular basis of higher levels of sympathetic innervation of thermogenic fat, compared to white fat, has remained unknown. Here we show that thermogenic adipocytes express a previously unknown, mammal-specific protein of the endoplasmic reticulum that we term calsyntenin 3ß. Genetic loss or gain of expression of calsyntenin 3ß in adipocytes reduces or enhances functional sympathetic innervation, respectively, in adipose tissue. Ablation of calsyntenin 3ß predisposes mice on a high-fat diet to obesity. Mechanistically, calsyntenin 3ß promotes endoplasmic-reticulum localization and secretion of S100b-a protein that lacks a signal peptide-from brown adipocytes. S100b stimulates neurite outgrowth from sympathetic neurons in vitro. A deficiency of S100b phenocopies deficiency of calsyntenin 3ß, and forced expression of S100b in brown adipocytes rescues the defective sympathetic innervation that is caused by ablation of calsyntenin 3ß. Our data reveal a mammal-specific mechanism of communication between thermogenic adipocytes and sympathetic neurons.


Assuntos
Tecido Adiposo Marrom/inervação , Tecido Adiposo Marrom/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo , Sistema Nervoso Simpático/citologia , Termogênese , Adipócitos/metabolismo , Animais , Proteínas de Ligação ao Cálcio/deficiência , Proteínas de Ligação ao Cálcio/genética , Dieta Hiperlipídica , Retículo Endoplasmático/metabolismo , Feminino , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Neuritos/metabolismo , Obesidade/metabolismo , Especificidade de Órgãos , Sistema Nervoso Simpático/metabolismo , Termogênese/genética
13.
Nature ; 570(7760): E32, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31114060

RESUMO

In Fig. 6a of this Article, the two dots corresponding to Cidea and S100b were erroneously moved to the top left of the volcano plot; this figure has been corrected online.An amendment to this paper has been published and can be accessed via a link at the top of the paper.

14.
Neuron ; 102(3): 653-667.e6, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-30879785

RESUMO

SIM1-expressing paraventricular hypothalamus (PVH) neurons are key regulators of energy balance. Within the PVHSIM1 population, melanocortin-4 receptor-expressing (PVHMC4R) neurons are known to regulate satiety and bodyweight, yet they account for only half of PVHSIM1 neuron-mediated regulation. Here we report that PVH prodynorphin-expressing (PVHPDYN) neurons, which notably lack MC4Rs, function independently and additively with PVHMC4R neurons to account for the totality of PVHSIM1 neuron-mediated satiety. Moreover, PVHPDYN neurons are necessary for prevention of obesity in an independent but equipotent manner to PVHMC4R neurons. While PVHPDYN and PVHMC4R neurons both project to the parabrachial complex (PB), they synaptically engage distinct efferent nodes, the pre-locus coeruleus (pLC), and central lateral parabrachial nucleus (cLPBN), respectively. PB-projecting PVHPDYN neurons, like PVHMC4R neurons, receive input from interoceptive ARCAgRP neurons, respond to caloric state, and are sufficient and necessary to control food intake. This expands the CNS satiety circuitry to include two non-overlapping PVH to hindbrain circuits.


Assuntos
Comportamento Alimentar/fisiologia , Neurônios/citologia , Obesidade/fisiopatologia , Núcleo Hipotalâmico Paraventricular/citologia , Resposta de Saciedade/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Metabolismo Energético , Encefalinas/metabolismo , Locus Cerúleo/citologia , Locus Cerúleo/metabolismo , Locus Cerúleo/fisiologia , Camundongos , Neurônios/metabolismo , Neurônios/fisiologia , Núcleos Parabraquiais/citologia , Núcleos Parabraquiais/metabolismo , Núcleos Parabraquiais/fisiologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Precursores de Proteínas/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Proteínas Repressoras/metabolismo
15.
Brain Struct Funct ; 224(1): 387-417, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30343334

RESUMO

Sodium deficiency elevates aldosterone, which in addition to epithelial tissues acts on the brain to promote dysphoric symptoms and salt intake. Aldosterone boosts the activity of neurons that express 11-beta-hydroxysteroid dehydrogenase type 2 (HSD2), a hallmark of aldosterone-sensitive cells. To better characterize these neurons, we combine immunolabeling and in situ hybridization with fate mapping and Cre-conditional axon tracing in mice. Many cells throughout the brain have a developmental history of Hsd11b2 expression, but in the adult brain one small brainstem region with a leaky blood-brain barrier contains HSD2 neurons. These neurons express Hsd11b2, Nr3c2 (mineralocorticoid receptor), Agtr1a (angiotensin receptor), Slc17a6 (vesicular glutamate transporter 2), Phox2b, and Nxph4; many also express Cartpt or Lmx1b. No HSD2 neurons express cholinergic, monoaminergic, or several other neuropeptidergic markers. Their axons project to the parabrachial complex (PB), where they intermingle with AgRP-immunoreactive axons to form dense terminal fields overlapping FoxP2 neurons in the central lateral subnucleus (PBcL) and pre-locus coeruleus (pLC). Their axons also extend to the forebrain, intermingling with AgRP- and CGRP-immunoreactive axons to form dense terminals surrounding GABAergic neurons in the ventrolateral bed nucleus of the stria terminalis (BSTvL). Sparse axons target the periaqueductal gray, ventral tegmental area, lateral hypothalamic area, paraventricular hypothalamic nucleus, and central nucleus of the amygdala. Dual retrograde tracing revealed that largely separate HSD2 neurons project to pLC/PB or BSTvL. This projection pattern raises the possibility that a subset of HSD2 neurons promotes the dysphoric, anorexic, and anhedonic symptoms of hyperaldosteronism via AgRP-inhibited relay neurons in PB.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Aldosterona/farmacologia , Tronco Encefálico/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Núcleo Solitário/efeitos dos fármacos , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Animais , Regulação do Apetite , Axônios/efeitos dos fármacos , Axônios/enzimologia , Tronco Encefálico/citologia , Tronco Encefálico/enzimologia , Encefalinas/genética , Encefalinas/metabolismo , Comportamento Alimentar , Imunofluorescência , Regulação da Expressão Gênica , Genes Reporter , Hibridização in Situ Fluorescente , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Vias Neurais/efeitos dos fármacos , Vias Neurais/enzimologia , Técnicas de Rastreamento Neuroanatômico , Neurônios/enzimologia , Prosencéfalo/citologia , Prosencéfalo/enzimologia , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Núcleo Solitário/citologia , Núcleo Solitário/enzimologia
16.
Curr Biol ; 28(14): 2291-2301.e5, 2018 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-30017482

RESUMO

Stress elicits a variety of autonomic responses, including hyperthermia (stress fever) in humans and animals. In this present study, we investigated the circuit basis for thermogenesis and heat conservation during this response. We first demonstrated the glutamatergic identity of the dorsal hypothalamic area (DHAVglut2) neurons that innervate the raphe pallidus nucleus (RPa) to regulate core temperature (Tc) and mediate stress-induced hyperthermia. Then, using chemogenetic and optogenetic methods to manipulate this hypothalamomedullary circuit, we found that activation of DHAVglut2 neurons potently drove an increase in Tc, but surprisingly, stress-induced hyperthermia was only reduced by about one-third when they were inhibited. Further investigation showed that DHAVglut2 neurons activate brown adipose tissue (BAT) but do not cause vasoconstriction, instead allowing reflex tail artery vasodilation as a response to BAT-induced hyperthermia. Retrograde rabies virus tracing revealed projections from DHAVglut2 neurons to RPaVglut3, but not to RPaGABA neurons, and identified a set of inputs to DHAVglut2 → RPa neurons that are likely to mediate BAT activation. The dissociation of the DHAVglut2 thermogenic pathway from the thermoregulatory vasoconstriction (heat-conserving) pathway may explain stress flushing (skin vasodilation but a feeling of being too hot) during stressful times.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Febre/fisiopatologia , Hipotálamo/metabolismo , Neurônios/fisiologia , Termogênese , Animais , Feminino , Masculino , Camundongos , Núcleo Pálido da Rafe/fisiologia , Optogenética , Estresse Fisiológico
17.
Neuron ; 96(1): 190-206.e7, 2017 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-28957668

RESUMO

Sodium deficiency increases angiotensin II (ATII) and aldosterone, which synergistically stimulate sodium retention and consumption. Recently, ATII-responsive neurons in the subfornical organ (SFO) and aldosterone-sensitive neurons in the nucleus of the solitary tract (NTSHSD2 neurons) were shown to drive sodium appetite. Here we investigate the basis for NTSHSD2 neuron activation, identify the circuit by which NTSHSD2 neurons drive appetite, and uncover an interaction between the NTSHSD2 circuit and ATII signaling. NTSHSD2 neurons respond to sodium deficiency with spontaneous pacemaker-like activity-the consequence of "cardiac" HCN and Nav1.5 channels. Remarkably, NTSHSD2 neurons are necessary for sodium appetite, and with concurrent ATII signaling their activity is sufficient to produce rapid consumption. Importantly, NTSHSD2 neurons stimulate appetite via projections to the vlBNST, which is also the effector site for ATII-responsive SFO neurons. The interaction between angiotensin signaling and NTSHSD2 neurons provides a neuronal context for the long-standing "synergy hypothesis" of sodium appetite regulation.


Assuntos
Aldosterona/fisiologia , Angiotensina II/fisiologia , Relógios Biológicos/fisiologia , Neurônios/fisiologia , Transdução de Sinais , Sódio/fisiologia , Núcleo Solitário/fisiologia , Animais , Ingestão de Alimentos/fisiologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Vias Neurais/fisiologia , Núcleos Septais/fisiologia , Sódio/deficiência
19.
Nature ; 546(7660): 611-616, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28614299

RESUMO

Physiological needs bias perception and attention to relevant sensory cues. This process is 'hijacked' by drug addiction, causing cue-induced cravings and relapse. Similarly, its dysregulation contributes to failed diets, obesity, and eating disorders. Neuroimaging studies in humans have implicated insular cortex in these phenomena. However, it remains unclear how 'cognitive' cortical representations of motivationally relevant cues are biased by subcortical circuits that drive specific motivational states. Here we develop a microprism-based cellular imaging approach to monitor visual cue responses in the insular cortex of behaving mice across hunger states. Insular cortex neurons demonstrate food-cue-biased responses that are abolished during satiety. Unexpectedly, while multiple satiety-related visceral signals converge in insular cortex, chemogenetic activation of hypothalamic 'hunger neurons' (expressing agouti-related peptide (AgRP)) bypasses these signals to restore hunger-like response patterns in insular cortex. Circuit mapping and pathway-specific manipulations uncover a pathway from AgRP neurons to insular cortex via the paraventricular thalamus and basolateral amygdala. These results reveal a neural basis for state-specific biased processing of motivationally relevant cues.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Alimentos , Homeostase , Vias Neurais , Estimulação Luminosa , Proteína Relacionada com Agouti/metabolismo , Animais , Sinais (Psicologia) , Fome/fisiologia , Hipotálamo/citologia , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Resposta de Saciedade/fisiologia
20.
Nat Neurosci ; 20(3): 484-496, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28166221

RESUMO

The hypothalamic arcuate-median eminence complex (Arc-ME) controls energy balance, fertility and growth through molecularly distinct cell types, many of which remain unknown. To catalog cell types in an unbiased way, we profiled gene expression in 20,921 individual cells in and around the adult mouse Arc-ME using Drop-seq. We identify 50 transcriptionally distinct Arc-ME cell populations, including a rare tanycyte population at the Arc-ME diffusion barrier, a new leptin-sensing neuron population, multiple agouti-related peptide (AgRP) and pro-opiomelanocortin (POMC) subtypes, and an orexigenic somatostatin neuron population. We extended Drop-seq to detect dynamic expression changes across relevant physiological perturbations, revealing cell type-specific responses to energy status, including distinct responses in AgRP and POMC neuron subtypes. Finally, integrating our data with human genome-wide association study data implicates two previously unknown neuron populations in the genetic control of obesity. This resource will accelerate biological discovery by providing insights into molecular and cell type diversity from which function can be inferred.


Assuntos
Núcleo Arqueado do Hipotálamo/anatomia & histologia , Eminência Mediana/anatomia & histologia , Neurônios/metabolismo , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Metabolismo Energético/fisiologia , Células Ependimogliais/metabolismo , Feminino , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Leptina/fisiologia , Masculino , Eminência Mediana/metabolismo , Camundongos , Camundongos Transgênicos , Obesidade/metabolismo , Orexinas/metabolismo , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/fisiologia , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/fisiologia , Somatostatina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...