Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 318
Filtrar
1.
Nature ; 619(7969): 332-337, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37380765

RESUMO

Fast-acting neurotransmitters and slow, modulatory neuropeptides are co-released from neurons in the central nervous system, albeit from distinct synaptic vesicles1. The mechanisms of how co-released neurotransmitters and neuropeptides that have opposing actions-for example, stimulatory versus inhibitory-work together to exert control of neural circuit output remain unclear. This has been difficult to resolve owing to the inability to selectively isolate these signalling pathways in a cell- and circuit-specific manner. Here we developed a genetic-based anatomical disconnect procedure that utilizes distinct DNA recombinases to independently facilitate CRISPR-Cas9 mutagenesis2 of neurotransmitter- and neuropeptide-related genes in distinct cell types in two different brain regions simultaneously. We demonstrate that neurons within the lateral hypothalamus that produce the stimulatory neuropeptide neurotensin and the inhibitory neurotransmitter GABA (γ-aminobutyric acid) utilize these signals to coordinately activate dopamine-producing neurons of the ventral tegmental area. We show that GABA release from lateral hypothalamus neurotensin neurons inhibits GABA neurons within the ventral tegmental area, disinhibiting dopamine neurons and causing a rapid rise in calcium, whereas neurotensin directly generates a slow inactivating calcium signal in dopamine neurons that is dependent on the expression of neurotensin receptor 1 (Ntsr1). We further show that these two signals work together to regulate dopamine neuron responses to maximize behavioural responding. Thus, a neurotransmitter and a neuropeptide with opposing signals can act on distinct timescales through different cell types to enhance circuit output and optimize behaviour.


Assuntos
Encéfalo , Vias Neurais , Neurotensina , Neurotransmissores , Transdução de Sinais , Encéfalo/citologia , Encéfalo/metabolismo , Cálcio/metabolismo , Sistemas CRISPR-Cas , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios GABAérgicos , Ácido gama-Aminobutírico/metabolismo , Edição de Genes , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/metabolismo , Neurotensina/metabolismo , Neurotransmissores/metabolismo , Receptores de Neurotensina/metabolismo , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo
2.
Cell ; 184(26): 6361-6377.e24, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34875226

RESUMO

Determining the spatial organization and morphological characteristics of molecularly defined cell types is a major bottleneck for characterizing the architecture underpinning brain function. We developed Expansion-Assisted Iterative Fluorescence In Situ Hybridization (EASI-FISH) to survey gene expression in brain tissue, as well as a turnkey computational pipeline to rapidly process large EASI-FISH image datasets. EASI-FISH was optimized for thick brain sections (300 µm) to facilitate reconstruction of spatio-molecular domains that generalize across brains. Using the EASI-FISH pipeline, we investigated the spatial distribution of dozens of molecularly defined cell types in the lateral hypothalamic area (LHA), a brain region with poorly defined anatomical organization. Mapping cell types in the LHA revealed nine spatially and molecularly defined subregions. EASI-FISH also facilitates iterative reanalysis of scRNA-seq datasets to determine marker-genes that further dissociated spatial and morphological heterogeneity. The EASI-FISH pipeline democratizes mapping molecularly defined cell types, enabling discoveries about brain organization.


Assuntos
Região Hipotalâmica Lateral/metabolismo , Hibridização in Situ Fluorescente , Animais , Biomarcadores/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Região Hipotalâmica Lateral/citologia , Imageamento Tridimensional , Masculino , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA/metabolismo , RNA-Seq , Análise de Célula Única , Transcrição Gênica
3.
J Neurosci ; 41(46): 9539-9560, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34642212

RESUMO

The lateral hypothalamic area (LHA) is a highly conserved brain region critical for maintaining physiological homeostasis and goal-directed behavior. LHA neurons that express melanin-concentrating hormone (MCH) are key regulators of arousal, energy balance, and motivated behavior. However, cellular and functional diversity among LHAMCH neurons is not well understood. Previous anatomic and molecular data suggest that LHAMCH neurons may be parsed into at least two distinct subpopulations, one of which is enriched in neurokinin-3 receptor (NK3R), the receptor for neurokinin B (NKB), encoded by the Tac2 gene. This tachykininergic ligand-receptor system has been implicated in reproduction, fear memory, and stress in other brain regions, but NKB interactions with LHAMCH neurons are poorly understood. We first identified how LHAMCH subpopulations may be distinguished anatomically and electrophysiologically. To dissect functional connectivity between NKB-expressing neurons and LHAMCH neurons, we used Cre-dependent retrograde and anterograde viral tracing in male Tac2-Cre mice and identified Tac2/EYFP+ neurons in the bed nucleus of the stria terminalis and central nucleus of the amygdala, the central extended amygdala, as major sources of NKB input onto LHAMCH neurons. In addition to innervating the LHA, these limbic forebrain NKB neurons also project to midbrain and brainstem targets. Finally, using a dual-virus approach, we found that optogenetic activation of these inputs in slices evokes GABA release onto a subset of LHAMCH neurons but lacked specificity for the NK3R+ subpopulation. Overall, these data define parallel tachykininergic/GABAergic limbic forebrain projections that are positioned to modulate multiple nodes of homeostatic and behavioral control.SIGNIFICANCE STATEMENT The LHA orchestrates fundamental behavioral states in the mammalian hypothalamus, including arousal, energy balance, memory, stress, and motivated behavior. The neuropeptide MCH defines one prominent population of LHA neurons, with multiple roles in the regulation of homeostatic behavior. Outstanding questions remain concerning the upstream inputs that control MCH neurons. We sought to define neurochemically distinct pathways in the mouse brain that may communicate with specific MCH neuron subpopulations using viral-based retrograde and anterograde neural pathway tracing and optogenetics in brain slices. Here, we identify a specific neuropeptide-defined forebrain circuit that makes functional synaptic connections with MCH neuron subpopulations. This work lays the foundation for further manipulating molecularly distinct neural circuits that modulate innate behavioral states.


Assuntos
Núcleo Central da Amígdala/citologia , Região Hipotalâmica Lateral/citologia , Vias Neurais/citologia , Neurônios/citologia , Animais , Hormônios Hipotalâmicos/metabolismo , Masculino , Melaninas/metabolismo , Camundongos , Camundongos Transgênicos , Vias Neurais/metabolismo , Neurocinina B/metabolismo , Neurônios/metabolismo , Hormônios Hipofisários/metabolismo
4.
Nat Commun ; 12(1): 5249, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34475397

RESUMO

The wake-active orexin system plays a central role in the dynamic regulation of glucose homeostasis. Here we show orexin receptor type 1 and 2 are predominantly expressed in dorsal raphe nucleus-dorsal and -ventral, respectively. Serotonergic neurons in ventral median raphe nucleus and raphe pallidus selectively express orexin receptor type 1. Inactivation of orexin receptor type 1 in serotonin transporter-expressing cells of mice reduced insulin sensitivity in diet-induced obesity, mainly by decreasing glucose utilization in brown adipose tissue and skeletal muscle. Selective inactivation of orexin receptor type 2 improved glucose tolerance and insulin sensitivity in obese mice, mainly through a decrease in hepatic gluconeogenesis. Optogenetic activation of orexin neurons in lateral hypothalamus or orexinergic fibers innervating raphe pallidus impaired or improved glucose tolerance, respectively. Collectively, the present study assigns orexin signaling in serotonergic neurons critical, yet differential orexin receptor type 1- and 2-dependent functions in the regulation of systemic glucose homeostasis.


Assuntos
Glucose/metabolismo , Obesidade/metabolismo , Receptores de Orexina/metabolismo , Neurônios Serotoninérgicos/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Homeostase , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/metabolismo , Resistência à Insulina , Fígado/metabolismo , Camundongos , Fibras Nervosas/metabolismo , Obesidade/etiologia , Receptores de Orexina/genética , Orexinas/metabolismo , Núcleos da Rafe/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Transdução de Sinais
5.
J Neurosci ; 41(1): 61-72, 2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33188067

RESUMO

Persistent avoidance of stress-related stimuli following acute stress exposure predicts negative outcomes such as substance abuse and traumatic stress disorders. Previous work using a rat model showed that the central amygdala (CeA) plays an important role in avoidance of a predator odor stress-paired context. Here, we show that CeA projections to the lateral hypothalamus (LH) are preferentially activated in male rats that show avoidance of a predator odor-paired context (termed Avoider rats), that chemogenetic inhibition of CeA-LH projections attenuates avoidance in male Avoider rats, that chemogenetic stimulation of the CeA-LH circuit produces conditioned place avoidance (CPA) in otherwise naive male rats, and that avoidance behavior is associated with intrinsic properties of LH-projecting CeA cells. Collectively, these data show that CeA-LH projections are important for persistent avoidance of stress-related stimuli following acute stress exposure.SIGNIFICANCE STATEMENT This study in rats shows that a specific circuit in the brain [i.e., neurons that project from the central amygdala (CeA) to the lateral hypothalamus (LH)] mediates avoidance of stress-associated stimuli. In addition, this study shows that intrinsic physiological properties of cells in this brain circuit are associated with avoidance of stress-associated stimuli. Further characterization of the CeA-LH circuit may improve our understanding of the neural mechanisms underlying specific aspects of stress-related disorders in humans.


Assuntos
Aprendizagem da Esquiva/fisiologia , Núcleo Central da Amígdala/fisiologia , Região Hipotalâmica Lateral/fisiologia , Vias Neurais/fisiologia , Animais , Comportamento Animal/fisiologia , Núcleo Central da Amígdala/citologia , Fenômenos Eletrofisiológicos , Região Hipotalâmica Lateral/citologia , Masculino , Vias Neurais/citologia , Neurônios , Odorantes , Ratos , Ratos Wistar
6.
Curr Biol ; 30(20): 4063-4070.e2, 2020 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-32822604

RESUMO

The lateral hypothalamic area (LH) is a vital controller of arousal, feeding, and metabolism [1, 2], which integrates external and internal sensory information. Whereas sensory and whole-body output properties of LH cell populations have received much interest, their intrinsic synaptic organization has remained largely unstudied. Local inhibitory and excitatory connections could help integrate and filter sensory information and mutually inhibitory connections [3] could allow coordinating activity between LH cell types, some of which have mutually exclusive behavioral effects, such as LH VGLUT2 and VGAT neurons [4-7] and orexin- (ORX) and melanin-concentrating hormone (MCH) neurons [8-10]. However, classical Golgi staining studies did not find interneurons with locally ramifying axons in the LH [11, 12], and nearby subthalamic and thalamic areas lack local synaptic connectivity [13, 14]. Studies with optogenetic circuit mapping within the LH have demonstrated only a minority of connections when a large pool of presynaptic neurons was activated [15-19]. Because multiple patch clamp has not been used to study LH connectivity, aside from a limited dataset of MCH neurons where no connections were discovered [15], we used quadruple whole-cell recordings to screen connectivity within the LH with standard methodology we previously used in the neocortex [20-22]. Finding a lack of local connectivity, we used optogenetic circuit mapping to study the strength of LH optogenetic responses and network oscillations, which were consistent with ultra-sparse intrinsic connectivity within the LH. These results suggest that input from other brain structures is decisive for selecting active populations in the LH.


Assuntos
Potenciais de Ação/fisiologia , Conectoma , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/fisiologia , Neocórtex/fisiologia , Animais , Ondas Encefálicas/fisiologia , Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/fisiologia , Optogenética , Orexinas/metabolismo , Técnicas de Patch-Clamp , Hormônios Hipofisários/metabolismo
7.
Horm Metab Res ; 52(10): 747-754, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32731263

RESUMO

The objective of the study was to investigate the regulatory actions of unacylated ghrelin (UAG) on glucose-sensitive (GS) neurons and glycolipid metabolism in the lateral hypothalamus area (LHA) and its involvement with orexin-A-immunopositive neurons. The effects of UAG administered into the LHA on GS neurons discharges and glycolipid metabolism were detected by single neuron discharge recording, biochemical index analysis and quantitative real-time PCR; the level of c-fos protein in orexin-A-immunopositive neurons was observed using immunofluorescence staining. UAG microinjected into the LHA activated glucose-inhibited neurons, which were partially blocked by pre-administration of anti-orexin-A antibody in the LHA. Furthermore, UAG microinjected into the LHA significantly reduced serum triglycerides (TG), total cholesterol, low-density lipoprotein cholesterol, blood glucose, insulin and hepatic TG levels, while elevated serum high-density lipoprotein cholesterol levels. UAG elevated the mRNA expression of carnitine palmitoyltransferase-1 and reduced the mRNA expression of acetyl-CoA carboxylase-1 in the liver. The above-mentioned effects of UAG were partially blocked by pre-administration of anti-orexin-A antibody. The expressions of orexin-A and c-fos were observed in the LHA. After UAG injection into the LHA, some neurons showed double labeling, and the percentage of double-labeled orexin-A/c-fos neurons in orexin-A-immunopositive neurons increased significantly. UAG in the LHA regulates glycolipid metabolism by activating orexin-A-immunopositive neurons in the LHA.


Assuntos
Grelina/metabolismo , Glucose/farmacologia , Glicolipídeos/metabolismo , Região Hipotalâmica Lateral/fisiologia , Neurônios/fisiologia , Orexinas/metabolismo , Acilação , Animais , Grelina/química , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/efeitos dos fármacos , Masculino , Neurônios/citologia , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Edulcorantes/farmacologia
8.
Neuron ; 107(2): 368-382.e8, 2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32442399

RESUMO

The ventral tegmental area (VTA) has dopamine, GABA, and glutamate neurons, which have been implicated in reward and aversion. Here, we determined whether VTA-glutamate or -GABA neurons play a role in innate defensive behavior. By VTA cell-type-specific genetic ablation, we found that ablation of glutamate, but not GABA, neurons abolishes escape behavior in response to threatening stimuli. We found that escape behavior is also decreased by chemogenetic inhibition of VTA-glutamate neurons and detected increases in activity in VTA-glutamate neurons in response to the threatening stimuli. By ultrastructural and electrophysiological analysis, we established that VTA-glutamate neurons receive a major monosynaptic glutamatergic input from the lateral hypothalamic area (LHA) and found that photoinhibition of this input decreases escape responses to threatening stimuli. These findings indicate that VTA-glutamate neurons are activated by and required for innate defensive responses and that information on threatening stimuli to VTA-glutamate neurons is relayed by LHA-glutamate neurons.


Assuntos
Agressão/fisiologia , Ácido Glutâmico/fisiologia , Neurônios/fisiologia , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/fisiologia , Animais , Reação de Fuga , Humanos , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/fisiologia , Hipotálamo/citologia , Hipotálamo/fisiologia , Camundongos , Neurônios/ultraestrutura , Optogenética , Estimulação Luminosa , Reflexo Monosináptico/fisiologia , Área Tegmentar Ventral/ultraestrutura , Ácido gama-Aminobutírico/fisiologia
9.
Mol Metab ; 35: 100956, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32244183

RESUMO

OBJECTIVE: Histaminergic neurons of the tuberomammillary nucleus (TMN) are wake-promoting and contribute to the regulation of energy homeostasis. Evidence indicates that melanocortin 4 receptors (MC4R) are expressed within the TMN. However, whether the melanocortin system influences the activity and function of TMN neurons expressing histidine decarboxylase (HDC), the enzyme required for histamine synthesis, remains undefined. METHODS: We utilized Hdc-Cre mice in combination with whole-cell patch-clamp electrophysiology and in vivo chemogenetic techniques to determine whether HDC neurons receive metabolically relevant information via the melanocortin system. RESULTS: We found that subsets of HDC-expressing neurons were excited by melanotan II (MTII), a non-selective melanocortin receptor agonist. Use of melanocortin receptor selective agonists (THIQ, [D-Trp8]-γ-MSH) and inhibitors of synaptic transmission (TTX, CNQX, AP5) indicated that the effect was mediated specifically by MC4Rs and involved a glutamatergic dependent presynaptic mechanism. MTII enhanced evoked excitatory post-synaptic currents (EPSCs) originating from electrical stimulation of the perifornical lateral hypothalamic area (PeFLH), supportive of melanocortin effects on the glutamatergic PeFLH projection to the TMN. Finally, in vivo chemogenetic inhibition of HDC neurons strikingly enhanced the anorexigenic effects of intracerebroventricular administration of MTII, suggesting that MC4R activation of histaminergic neurons may restrain the anorexigenic effects of melanocortin system activation. CONCLUSIONS: These experiments identify a functional interaction between the melanocortin and histaminergic systems and suggest that HDC neurons act naturally to restrain the anorexigenic effect of melanocortin system activation. These findings may have implications for the control of arousal and metabolic homeostasis, especially in the context of obesity, in which both processes are subjected to alterations.


Assuntos
Histamina/metabolismo , Histidina Descarboxilase/metabolismo , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/metabolismo , Melanocortinas/metabolismo , Neurônios/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Histidina Descarboxilase/genética , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/genética , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
10.
Proc Natl Acad Sci U S A ; 117(15): 8611-8615, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32229573

RESUMO

Electrical or optogenetic stimulation of lateral hypothalamic (LH) GABA neurons induces rapid vigorous eating in sated animals. The dopamine system has been implicated in the regulation of feeding. Previous work has suggested that a subset of LH GABA neurons projects to the ventral tegmental area (VTA) and targets GABA neurons, inhibiting them and thereby disinhibiting dopaminergic activity and release. Furthermore, stimulation-induced eating is attenuated by dopamine lesions or receptor antagonists. Here we explored the involvement of dopamine in LH stimulation-induced eating. LH stimulation caused sated mice to pick up pellets of standard chow with latencies that varied based on stimulation intensity; once food was picked up, animals ate for the remainder of the 60-s stimulation period. However, lesion of VTA GABA neurons failed to disrupt this effect. Moreover, direct stimulation of VTA or substantia nigra dopamine cell bodies failed to induce food approach or eating. Looking further, we found that some LH GABA fibers pass through the VTA to more caudal sites, where they synapse onto neurons near the locus coeruleus (LC). Similar eating was induced by stimulation of LH GABA terminals or GABA cell bodies in this peri-LC region. Lesion of peri-LC GABA neurons blocked LH stimulation-induced eating, establishing them as a critical downstream circuit element for LH neurons. Surprisingly, lesions did not alter body weight, suggesting that this system is not involved in the hunger or satiety mechanisms that govern normal feeding. Thus, we present a characterization of brain circuitry that may promote overeating and contribute to obesity.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Neurônios GABAérgicos/metabolismo , Região Hipotalâmica Lateral/fisiologia , Área Tegmentar Ventral/fisiologia , Animais , Comportamento Animal , Dopamina/metabolismo , Neurônios Dopaminérgicos/citologia , Feminino , Neurônios GABAérgicos/citologia , Região Hipotalâmica Lateral/citologia , Masculino , Camundongos , Vias Neurais , Receptores de GABA-A/metabolismo , Recompensa , Área Tegmentar Ventral/citologia , Ácido gama-Aminobutírico/metabolismo
11.
Alcohol Clin Exp Res ; 44(4): 866-879, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32020622

RESUMO

BACKGROUND: Prenatal exposure to ethanol (EtOH) has lasting effects on neuropeptide and neuroimmune systems in the brain alongside detrimental alcohol-related behaviors. At low-to-moderate doses, prenatal EtOH stimulates neurogenesis in lateral hypothalamus (LH) and increases neurons that express the orexigenic peptides hypocretin/orexin (Hcrt/OX) and melanin-concentrating hormone (MCH), and the proinflammatory chemokine CCL2, which through its receptor CCR2 stimulates cell differentiation and movement. Our recent studies demonstrated that CCL2 and CCR2 colocalize with MCH neurons and are involved in EtOH's stimulatory effect on their development but show no relation to Hcrt/OX. Here, we investigated another chemokine, CXCL12, and its receptor, CXCR4, which promote neurogenesis and neuroprogenitor cell proliferation, to determine if they also exhibit peptide specificity in their response to EtOH exposure. METHODS: Pregnant rats were intraorally administered a moderate dose of EtOH (2 g/kg/d) from embryonic day 10 (E10) to E15. Their embryos and postnatal offspring were examined using real-time quantitative PCR and immunofluorescence histochemistry, to determine if EtOH affects CXCL12 and CXCR4 and the colocalization of CXCR4 with Hcrt/OX and MCH neurons in the LH and with radial glia neuroprogenitor cells in the hypothalamic neuroepithelium (NEP). RESULTS: Prenatal EtOH strongly stimulated CXCL12 and CXCR4 in LH neurons of embryos and postnatal offspring. This stimulation was significantly stronger in Hcrt/OX than MCH neurons in LH and also occurred in radial glia neuroprogenitor cells dense in the NEP. These effects were sexually dimorphic, consistently stronger in females than males. CONCLUSIONS: While showing prenatal EtOH exposure to have a sexually dimorphic, stimulatory effect on CXCL12 and CXCR4 in LH similar to CCL2 and its receptor, these results reveal their distinct relationship to the peptide neurons, with the former closely related to Hcrt/OX and the latter to MCH, and they link EtOH's actions in LH to a stimulatory effect on neuroprogenitor cells in the NEP.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Quimiocina CXCL12/efeitos dos fármacos , Células Ependimogliais/efeitos dos fármacos , Etanol/farmacologia , Região Hipotalâmica Lateral/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptores CXCR4/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Embrião de Mamíferos , Células Ependimogliais/metabolismo , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/metabolismo , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Imuno-Histoquímica , Melaninas/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Neurônios/metabolismo , Orexinas/metabolismo , Hormônios Hipofisários/metabolismo , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Receptores CXCR4/metabolismo
12.
Addict Biol ; 25(4): e12795, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31297913

RESUMO

Lateral hypothalamus (LH) orexin neuron signaling has been implicated in the motivation to seek and take drugs of abuse. The number of LH orexin neurons has been shown to be upregulated with exposure to drugs of abuse. We sought to determine if the number of LH orexin neurons related to individual differences in motivation (demand) for cocaine in our behavioral economics (BE) paradigm, and whether knockdown of these cells predicted changes in economic demand. We quantified LH orexin cell numbers in animals immediately following our BE paradigm, as well as after a 2-week period of abstinence, to relate the number of LH orexin cells to economic demand for cocaine. We also knocked down LH orexin expression with an orexin morpholino antisense to determine how reduced orexin numbers impacted cocaine demand. We found that animals with greater baseline motivation for cocaine (lower demand elasticity) had more LH orexin neurons. Following a 2-week abstinence from cocaine, the number of LH orexin neurons predicted economic demand for cocaine prior to abstinence, indicating that orexin expression is a persistent marker for demand. Reducing LH orexin cell numbers with antisense decreased motivation for cocaine (increased demand elasticity) without affecting baseline consumption. In addition, the number of spared LH orexin neurons after antisense treatment correlated with individual motivation for cocaine. These studies point to a role for the endogenous number of LH orexin neurons in individual differences in motivation for cocaine.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/metabolismo , Cocaína , Inibidores da Captação de Dopamina , Região Hipotalâmica Lateral/citologia , Motivação , Neurônios/citologia , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Animais , Contagem de Células , Economia Comportamental , Região Hipotalâmica Lateral/metabolismo , Individualidade , Masculino , Morfolinos , Neurônios/metabolismo , Ratos
13.
Pharmacol Biochem Behav ; 187: 172794, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31654653

RESUMO

Orexin-producing neurons in the lateral hypothalamus are a critical regulator of sleep/wake states, and their loss is associated with narcolepsy type 1 (NT1). Orexin peptides act on two G protein-coupled receptors: orexin 1 receptor (OX1R) and orexin 2 receptor (OX2R). OX2R knockout (KO) mice, but not OX1R KO mice, showed clear narcolepsy-like phenotypes, including fragmented sleep-wake cycles. Moreover, OX2R-selective antagonists have been shown to induce sleepiness in mice, and activation of OX2R has been reported to increase wakefulness. In this study, we characterized in vitro and in vivo profiles of a novel, highly selective OX2R agonist, TAK-925 [methyl (2R,3S)-3-[(methylsulfonyl)amino]-2-{[(cis-4-phenylcyclohexyl)oxy]methyl}piperidine-1-carboxylate]. TAK-925 activated human recombinant OX2R with 50% effective concentration value of 5.5 nM, and showed >5,000-fold selectivity over OX1R in calcium mobilization assays. TAK-925 induced OX2R-downstream signals similar to those displayed by orexin peptides in Chinese hamster ovary cells stably expressing human OX2R. In an electrophysiological study, TAK-925 activated physiological OX2R on histaminergic neurons in the mouse tuberomammillary nucleus (TMN). Subcutaneous (SC) administration of TAK-925 also modulated neuronal activity in various brain regions, including TMN, as measured by an immunohistochemical analysis using an anti-c-fos antibody. TAK-925 (SC) increased wakefulness in wild-type mice, but not in OX2R KO mice, during their sleep phase, demonstrating that a highly selective OX2R agonist can increase wakefulness in mice via OX2R activation. TAK-925 may have therapeutic potential to reduce hypersomnia in multiple disorders including NT1.


Assuntos
Receptores de Orexina/agonistas , Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos , Animais , Células CHO , Cricetulus , Humanos , Região Hipotalâmica Lateral/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Narcolepsia/tratamento farmacológico , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Sono/genética , Transfecção , Vigília/genética
14.
Nat Commun ; 10(1): 4560, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31594935

RESUMO

The gustatory system plays a critical role in sensing appetitive and aversive taste stimuli for evaluating food quality. Although taste preference is known to change depending on internal states such as hunger, a mechanistic insight remains unclear. Here, we examine the neuronal mechanisms regulating hunger-induced taste modification. Starved mice exhibit an increased preference for sweetness and tolerance for aversive taste. This hunger-induced taste modification is recapitulated by selective activation of orexigenic Agouti-related peptide (AgRP)-expressing neurons in the hypothalamus projecting to the lateral hypothalamus, but not to other regions. Glutamatergic, but not GABAergic, neurons in the lateral hypothalamus function as downstream neurons of AgRP neurons. Importantly, these neurons play a key role in modulating preferences for both appetitive and aversive tastes by using distinct pathways projecting to the lateral septum or the lateral habenula, respectively. Our results suggest that these hypothalamic circuits would be important for optimizing feeding behavior under fasting.


Assuntos
Habenula/fisiologia , Fome/fisiologia , Região Hipotalâmica Lateral/fisiologia , Núcleos Septais/fisiologia , Paladar/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Comportamento Apetitivo/fisiologia , Neurônios GABAérgicos/metabolismo , Habenula/citologia , Região Hipotalâmica Lateral/citologia , Masculino , Camundongos , Modelos Animais , Vias Neurais/fisiologia , Optogenética , Técnicas de Patch-Clamp , Núcleos Septais/citologia , Técnicas Estereotáxicas
15.
J Neurosci ; 39(45): 8929-8939, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31548232

RESUMO

The histaminergic neurons of the tuberomammillary nucleus (TMNHDC) of the posterior hypothalamus have long been implicated in promoting arousal. More recently, a role for GABAergic signaling by the TMNHDC neurons in arousal control has been proposed. Here, we investigated the effects of selective chronic disruption of GABA synthesis (via genetic deletion of the GABA synthesis enzyme, glutamic acid decarboxylase 67) or GABAergic transmission (via genetic deletion of the vesicular GABA transporter (VGAT)) in the TMNHDC neurons on sleep-wake in male mice. We also examined the effects of acute chemogenetic activation and optogenetic inhibition of TMNHDC neurons upon arousal in male mice. Unexpectedly, we found that neither disruption of GABA synthesis nor GABAergic transmission altered hourly sleep-wake quantities, perhaps because very few TMNHDC neurons coexpressed VGAT. Acute chemogenetic activation of TMNHDC neurons did not increase arousal levels above baseline but did enhance vigilance when the mice were exposed to a behavioral cage change challenge. Similarly, acute optogenetic inhibition had little effect upon baseline levels of arousal. In conclusion, we could not identify a role for GABA release by TMNHDC neurons in arousal control. Further, if TMNHDC neurons do release GABA, the mechanism by which they do so remains unclear. Our findings support the view that TMNHDC neurons may be important for enhancing arousal under certain conditions, such as exposure to a novel environment, but play only a minor role in behavioral and EEG arousal under baseline conditions.SIGNIFICANCE STATEMENT The histaminergic neurons of the tuberomammillary nucleus of the hypothalamus (TMNHDC) have long been thought to promote arousal. Additionally, TMNHDC neurons may counter-regulate the wake-promoting effects of histamine through co-release of the inhibitory neurotransmitter, GABA. Here, we show that impairing GABA signaling from TMNHDC neurons does not impact sleep-wake amounts and that few TMNHDC neurons contain the vesicular GABA transporter, which is presumably required to release GABA. We further show that acute activation or inhibition of TMNHDC neurons has limited effects upon baseline arousal levels and that activation enhances vigilance during a behavioral challenge. Counter to general belief, our findings support the view that TMNHDC neurons are neither necessary nor sufficient for the initiation and maintenance of arousal under baseline conditions.


Assuntos
Nível de Alerta , Histamina/metabolismo , Região Hipotalâmica Lateral/fisiologia , Neurônios/metabolismo , Ácido gama-Aminobutírico/metabolismo , Potenciais de Ação , Animais , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Sono , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
16.
Cell Rep ; 28(3): 616-624.e5, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31315042

RESUMO

Overeating is a serious issue in modern society, causing many health problems, including obesity. Although the hypothalamus has been previously identified as the key brain structure that regulates body weight homeostasis, the downstream pathways and non-canonical neural circuitry involved in feeding behavior remain largely uncharacterized. Here, we discover that suppressing the activity of GABAergic cells in the anterior ventrolateral periaqueductal gray (vlPAG), whether directly or through long-projection GABAergic inputs from either the bed nucleus of the stria terminalis (BNST) or the lateral hypothalamus (LH), is sufficient to promptly induce feeding behavior in well-fed mice. In contrast, optogenetic activation of these cells interrupts food intake in starved mice. Long-term chemogenetic manipulation of vlPAG GABAergic cell activity elicits a corresponding change in mouse body weight. Our studies reveal distinct midbrain GABAergic pathways and highlight an important role of GABAergic cells in the anterior vlPAG in feeding behavior.


Assuntos
Comportamento Alimentar/psicologia , Neurônios GABAérgicos/fisiologia , Região Hipotalâmica Lateral/fisiologia , Vias Neurais/fisiologia , Substância Cinzenta Periaquedutal/fisiologia , Núcleos Septais/fisiologia , Animais , Antipsicóticos/farmacologia , Peso Corporal/efeitos dos fármacos , Peso Corporal/genética , Peso Corporal/fisiologia , Núcleo Central da Amígdala/efeitos dos fármacos , Núcleo Central da Amígdala/fisiologia , Clozapina/análogos & derivados , Clozapina/farmacologia , Comportamento Alimentar/fisiologia , Agonistas de Receptores de GABA-A/farmacologia , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/metabolismo , Região Hipotalâmica Lateral/citologia , Camundongos , Muscimol/farmacologia , Optogenética , Substância Cinzenta Periaquedutal/citologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Substância Cinzenta Periaquedutal/efeitos da radiação , Núcleos Septais/citologia
17.
PLoS One ; 14(7): e0219522, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31291348

RESUMO

Across species, motivated states such as food-seeking and consumption are essential for survival. The lateral hypothalamus (LH) is known to play a fundamental role in regulating feeding and reward-related behaviors. However, the contributions of neuronal subpopulations in the LH have not been thoroughly identified. Here we examine how lateral hypothalamic leptin receptor-expressing (LHLEPR) neurons, a subset of GABAergic cells, regulate motivation in mice. We find that LHLEPR neuronal activation significantly increases progressive ratio (PR) performance, while inhibition decreases responding. Moreover, we mapped LHLEPR axonal projections and demonstrated that they target the ventral tegmental area (VTA), form functional inhibitory synapses with non-dopaminergic VTA neurons, and their activation promotes motivation for food. Finally, we find that LHLEPR neurons also regulate motivation to obtain water, suggesting that they may play a generalized role in motivation. Together, these results identify LHLEPR neurons as modulators within a hypothalamic-ventral tegmental circuit that gates motivation.


Assuntos
Região Hipotalâmica Lateral/fisiologia , Motivação/fisiologia , Área Tegmentar Ventral/fisiologia , Animais , Condicionamento Operante/fisiologia , Comportamento Alimentar/psicologia , Feminino , Região Hipotalâmica Lateral/citologia , Masculino , Camundongos , Modelos Animais , Vias Neurais/fisiologia , Neurônios/fisiologia , Recompensa , Técnicas Estereotáxicas , Sinapses , Área Tegmentar Ventral/citologia
18.
Neuron ; 103(3): 423-431.e4, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31196673

RESUMO

The paraventricular thalamus (PVT) is an interface for brain reward circuits, with input signals arising from structures, such as prefrontal cortex and hypothalamus, that are broadcast to downstream limbic targets. However, the precise synaptic connectivity, activity, and function of PVT circuitry for reward processing are unclear. Here, using in vivo two-photon calcium imaging, we find that PVT neurons projecting to the nucleus accumbens (PVT-NAc) develop inhibitory responses to reward-predictive cues coding for both cue-reward associative information and behavior. The multiplexed activity in PVT-NAc neurons is directed by opposing activity patterns in prefrontal and lateral hypothalamic afferent axons. Further, we find that prefrontal cue encoding may maintain accurate cue-reward processing, as optogenetic disruption of this encoding induced long-lasting effects on downstream PVT-NAc cue responses and behavioral cue discrimination. Together, these data reveal that PVT-NAc neurons act as an interface for reward processing by integrating relevant inputs to accurately inform reward-seeking behavior.


Assuntos
Aprendizagem por Associação/fisiologia , Região Hipotalâmica Lateral/fisiologia , Núcleos da Linha Média do Tálamo/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Condicionamento Clássico , Fissura/fisiologia , Sinais (Psicologia) , Ácido Glutâmico/fisiologia , Região Hipotalâmica Lateral/citologia , Camundongos , Núcleos da Linha Média do Tálamo/citologia , Vias Neurais/fisiologia , Optogenética , Técnicas de Patch-Clamp , Córtex Pré-Frontal/citologia , Recompensa , Ácido gama-Aminobutírico/fisiologia
19.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R571-R583, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30726119

RESUMO

Aging affects numerous physiological processes, as well as behavior. A large number of these processes are regulated, at least partially, by hypothalamic orexin neurons, and orexin tone may decrease with normal aging. In this study, we hypothesized that designer receptors exclusively activated by designer drugs (DREADD) stimulation of orexin neuronal activity will ameliorate the effect of aging on behavioral and metabolic alterations in young and middle-aged mice. DREADD targeting was achieved by stereotaxic injection of AAV vectors (AAV2-hSyn-DIO-hM3D(Gq)-mCherry) into the lateral hypothalamus of 5- and 12-mo old orexin-cre female mice and was confirmed by immunohistochemistry (IHC) analysis of orexin A and mCherry expression. After recovery, animals were subjected to a behavioral test battery consisting of the elevated plus maze (EPM), open field (OFT), and novel object recognition tests (NORT) to assess effects of aging on anxiety-like behavior, general locomotion, and working memory. A comprehensive laboratory animal monitoring system (CLAMS) was used to measure spontaneous physical activity (SPA) and energy expenditure (EE). The results indicate that activation of orexin neurons mitigates aging-induced reductions in anxiety-like behavior in middle-aged mice (P < 0.005) and increases locomotion in both young and middle-aged mice (P < 0.05). Activation of orexin neurons increases SPA (P < 0.01) and EE (P < 0.005) in middle-aged mice, restoring the levels to that observed in young animals. Results from this study identify orexin neurons as potential therapeutic targets for age-related impairments in cognitive and anxiety-related behavior, and energy balance.


Assuntos
Envelhecimento , Comportamento Animal , Metabolismo Energético , Região Hipotalâmica Lateral/metabolismo , Neurônios/metabolismo , Orexinas/metabolismo , Envelhecimento/genética , Envelhecimento/metabolismo , Envelhecimento/psicologia , Animais , Comportamento Exploratório , Feminino , Região Hipotalâmica Lateral/citologia , Locomoção , Aprendizagem em Labirinto , Memória de Curto Prazo , Camundongos Endogâmicos C57BL , Orexinas/deficiência , Orexinas/genética
20.
Neuropeptides ; 74: 24-33, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30700376

RESUMO

OBJECTIVE: Abnormal gastric acid secretion and gastric dyskinesia are common gastroenterological ailments. Our study aims to investigate the effect of orexin-A in the paraventricular nucleus (PVN) gastric motility and gastric acid secretion. METHODS: The source of orexin-A neuronal projections to the PVN were explored by retrograde tracing and fluorescence immunohistochemistry experiments. Neuronal discharge recordings of single cells were taken within the PVN. Gastric motility was recorded using a force transducer implanted into the stomach, and gastric acid secretion measured through a pyloric catheter. RESULTS: Orexin-A-positive neuronal projections from LHA to PVN were found. Administration of orexin-A to PVN activated the firing of 63.2% NPY-excited/GD-excitatory (GD-E) neurons but suppressed the firing of 55.9% NPY-inhibited/GD-inhibitory (GD-I) neurons, promoted gastric motility and gastric acid secretion in a dose-dependent manner. Responses produced by orexin-A could be partially blocked by Y1 receptor antagonist GR-231118; Electrical stimulation to the the hypothalamic lateral area (LHA) altered NPY-sensitive/GD neuronal activity in the PVN, stimulated gastric motility and gastric acid secretion. Additionally, these effects induced by LHA electrical stimulation were blocked by administration of the OX1R antagonist SB-334867 to the PVN. CONCLUSION: Orexin-A from LHA neurons act on the PVN to enhance gastric motility and gastric acid secretion, with Y1 receptor signaling playing a critical role.


Assuntos
Ácido Gástrico/metabolismo , Motilidade Gastrointestinal , Região Hipotalâmica Lateral/metabolismo , Neurônios/metabolismo , Orexinas/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Potenciais de Ação , Animais , Região Hipotalâmica Lateral/citologia , Masculino , Vias Neurais/citologia , Vias Neurais/metabolismo , Neurônios/citologia , Núcleo Hipotalâmico Paraventricular/citologia , Ratos Sprague-Dawley , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA