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










Base de dados
Intervalo de ano de publicação
1.
Nature ; 630(8018): 926-934, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38898273

RESUMO

Krause corpuscles, which were discovered in the 1850s, are specialized sensory structures found within the genitalia and other mucocutaneous tissues1-4. The physiological properties and functions of Krause corpuscles have remained unclear since their discovery. Here we report the anatomical and physiological properties of Krause corpuscles of the mouse clitoris and penis and their roles in sexual behaviour. We observed a high density of Krause corpuscles in the clitoris compared with the penis. Using mouse genetic tools, we identified two distinct somatosensory neuron subtypes that innervate Krause corpuscles of both the clitoris and penis and project to a unique sensory terminal region of the spinal cord. In vivo electrophysiology and calcium imaging experiments showed that both Krause corpuscle afferent types are A-fibre rapid-adapting low-threshold mechanoreceptors, optimally tuned to dynamic, light-touch and mechanical vibrations (40-80 Hz) applied to the clitoris or penis. Functionally, selective optogenetic activation of Krause corpuscle afferent terminals evoked penile erection in male mice and vaginal contraction in female mice, while genetic ablation of Krause corpuscles impaired intromission and ejaculation of males and reduced sexual receptivity of females. Thus, Krause corpuscles of the clitoris and penis are highly sensitive mechanical vibration detectors that mediate sexually dimorphic mating behaviours.


Assuntos
Clitóris , Mecanorreceptores , Pênis , Comportamento Sexual Animal , Tato , Vibração , Animais , Feminino , Masculino , Camundongos , Clitóris/inervação , Clitóris/fisiologia , Ejaculação/fisiologia , Mecanorreceptores/metabolismo , Mecanorreceptores/fisiologia , Optogenética , Ereção Peniana/fisiologia , Pênis/inervação , Pênis/fisiologia , Comportamento Sexual Animal/fisiologia , Medula Espinal/fisiologia , Medula Espinal/citologia , Tato/fisiologia , Vagina/fisiologia , Neurônios/fisiologia
2.
Mol Metab ; 84: 101945, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38653401

RESUMO

OBJECTIVE: Glucose dependent insulinotropic polypeptide (GIP) is well established as an incretin hormone, boosting glucose-dependent insulin secretion. However, whilst anorectic actions of its sister-incretin glucagon-like peptide-1 (GLP-1) are well established, a physiological role for GIP in appetite regulation is controversial, despite the superior weight loss seen in preclinical models and humans with GLP-1/GIP dual receptor agonists compared with GLP-1R agonism alone. METHODS: We generated a mouse model in which GIP expressing K-cells can be activated through hM3Dq Designer Receptor Activated by Designer Drugs (DREADD, GIP-Dq) to explore physiological actions of intestinally-released GIP. RESULTS: In lean mice, Dq-stimulation of GIP expressing cells increased plasma GIP to levels similar to those found postprandially. The increase in GIP was associated with improved glucose tolerance, as expected, but also triggered an unexpected robust inhibition of food intake. Validating that this represented a response to intestinally-released GIP, the suppression of food intake was prevented by injecting mice peripherally or centrally with antagonistic GIPR-antibodies, and was reproduced in an intersectional model utilising Gip-Cre/Villin-Flp to limit Dq transgene expression to K-cells in the intestinal epithelium. The effects of GIP cell activation were maintained in diet induced obese mice, in which chronic K-cell activation reduced food intake and attenuated body weight gain. CONCLUSIONS: These studies establish a physiological gut-brain GIP-axis regulating food intake in mice, adding to the multi-faceted metabolic effects of GIP which need to be taken into account when developing GIPR-targeted therapies for obesity and diabetes.


Assuntos
Peso Corporal , Ingestão de Alimentos , Polipeptídeo Inibidor Gástrico , Animais , Polipeptídeo Inibidor Gástrico/metabolismo , Camundongos , Masculino , Camundongos Endogâmicos C57BL , Receptores dos Hormônios Gastrointestinais/metabolismo , Receptores dos Hormônios Gastrointestinais/genética , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Mucosa Intestinal/metabolismo , Obesidade/metabolismo , Incretinas/metabolismo
3.
Nature ; 627(8005): 830-838, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38448588

RESUMO

Airway integrity must be continuously maintained throughout life. Sensory neurons guard against airway obstruction and, on a moment-by-moment basis, enact vital reflexes to maintain respiratory function1,2. Decreased lung capacity is common and life-threatening across many respiratory diseases, and lung collapse can be acutely evoked by chest wall trauma, pneumothorax or airway compression. Here we characterize a neuronal reflex of the vagus nerve evoked by airway closure that leads to gasping. In vivo vagal ganglion imaging revealed dedicated sensory neurons that detect airway compression but not airway stretch. Vagal neurons expressing PVALB mediate airway closure responses and innervate clusters of lung epithelial cells called neuroepithelial bodies (NEBs). Stimulating NEBs or vagal PVALB neurons evoked gasping in the absence of airway threats, whereas ablating NEBs or vagal PVALB neurons eliminated gasping in response to airway closure. Single-cell RNA sequencing revealed that NEBs uniformly express the mechanoreceptor PIEZO2, and targeted knockout of Piezo2 in NEBs eliminated responses to airway closure. NEBs were dispensable for the Hering-Breuer inspiratory reflex, which indicated that discrete terminal structures detect airway closure and inflation. Similar to the involvement of Merkel cells in touch sensation3,4, NEBs are PIEZO2-expressing epithelial cells and, moreover, are crucial for an aspect of lung mechanosensation. These findings expand our understanding of neuronal diversity in the airways and reveal a dedicated vagal pathway that detects airway closure to help preserve respiratory function.


Assuntos
Pulmão , Reflexo , Respiração , Mecânica Respiratória , Nervo Vago , Animais , Feminino , Masculino , Camundongos , Células Epiteliais/metabolismo , Pulmão/citologia , Pulmão/inervação , Pulmão/fisiologia , Mecanorreceptores/metabolismo , Parvalbuminas/metabolismo , Reflexo/fisiologia , Células Receptoras Sensoriais/metabolismo , Nervo Vago/fisiologia , Complacência Pulmonar/fisiologia , Mecânica Respiratória/fisiologia
4.
Curr Biol ; 34(6): 1206-1221.e6, 2024 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-38320553

RESUMO

The physiological performance of any sensory organ is determined by its anatomy and physical properties. Consequently, complex sensory structures with elaborate features have evolved to optimize stimulus detection. Understanding these structures and their physical nature forms the basis for mechanistic insights into sensory function. Despite its crucial role as a sensor for pheromones and other behaviorally instructive chemical cues, the vomeronasal organ (VNO) remains a poorly characterized mammalian sensory structure. Fundamental principles of its physico-mechanical function, including basic aspects of stimulus sampling, remain poorly explored. Here, we revisit the classical vasomotor pump hypothesis of vomeronasal stimulus uptake. Using advanced anatomical, histological, and physiological methods, we demonstrate that large parts of the lateral mouse VNO are composed of smooth muscle. Vomeronasal smooth muscle tissue comprises two subsets of fibers with distinct topography, structure, excitation-contraction coupling, and, ultimately, contractile properties. Specifically, contractions of a large population of noradrenaline-sensitive cells mediate both transverse and longitudinal lumen expansion, whereas cholinergic stimulation targets an adluminal group of smooth muscle fibers. The latter run parallel to the VNO's rostro-caudal axis and are ideally situated to mediate antagonistic longitudinal constriction of the lumen. This newly discovered arrangement implies a novel mode of function. Single-cell transcriptomics and pharmacological profiling reveal the receptor subtypes involved. Finally, 2D/3D tomography provides non-invasive insight into the intact VNO's anatomy and mechanics, enables measurement of luminal fluid volume, and allows an assessment of relative volume change upon noradrenergic stimulation. Together, we propose a revised conceptual framework for mouse vomeronasal pumping and, thus, stimulus sampling.


Assuntos
Órgão Vomeronasal , Camundongos , Animais , Órgão Vomeronasal/fisiologia , Mamíferos , Feromônios/fisiologia
5.
Nature ; 618(7963): 193-200, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37225986

RESUMO

Odorants are detected as smell in the nasal epithelium of mammals by two G-protein-coupled receptor families, the odorant receptors and the trace amine-associated receptors1,2 (TAARs). TAARs emerged following the divergence of jawed and jawless fish, and comprise a large monophyletic family of receptors that recognize volatile amine odorants to elicit both intraspecific and interspecific innate behaviours such as attraction and aversion3-5. Here we report cryo-electron microscopy structures of mouse TAAR9 (mTAAR9) and mTAAR9-Gs or mTAAR9-Golf trimers in complex with ß-phenylethylamine, N,N-dimethylcyclohexylamine or spermidine. The mTAAR9 structures contain a deep and tight ligand-binding pocket decorated with a conserved D3.32W6.48Y7.43 motif, which is essential for amine odorant recognition. In the mTAAR9 structure, a unique disulfide bond connecting the N terminus to ECL2 is required for agonist-induced receptor activation. We identify key structural motifs of TAAR family members for detecting monoamines and polyamines and the shared sequence of different TAAR members that are responsible for recognition of the same odour chemical. We elucidate the molecular basis of mTAAR9 coupling to Gs and Golf by structural characterization and mutational analysis. Collectively, our results provide a structural basis for odorant detection, receptor activation and Golf coupling of an amine olfactory receptor.


Assuntos
Aminas Biogênicas , Odorantes , Percepção Olfatória , Poliaminas , Receptores Odorantes , Animais , Camundongos , Aminas Biogênicas/análise , Aminas Biogênicas/química , Aminas Biogênicas/metabolismo , Microscopia Crioeletrônica , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/ultraestrutura , Odorantes/análise , Percepção Olfatória/fisiologia , Poliaminas/análise , Poliaminas/química , Poliaminas/metabolismo , Receptores de Amina Biogênica/química , Receptores de Amina Biogênica/genética , Receptores de Amina Biogênica/metabolismo , Receptores de Amina Biogênica/ultraestrutura , Receptores Odorantes/química , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Receptores Odorantes/ultraestrutura , Olfato/fisiologia , Espermidina/análise , Espermidina/química , Espermidina/metabolismo
6.
Neuron ; 111(14): 2184-2200.e7, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37192624

RESUMO

Vagal sensory neurons monitor mechanical and chemical stimuli in the gastrointestinal tract. Major efforts are underway to assign physiological functions to the many distinct subtypes of vagal sensory neurons. Here, we use genetically guided anatomical tracing, optogenetics, and electrophysiology to identify and characterize vagal sensory neuron subtypes expressing Prox2 and Runx3 in mice. We show that three of these neuronal subtypes innervate the esophagus and stomach in regionalized patterns, where they form intraganglionic laminar endings. Electrophysiological analysis revealed that they are low-threshold mechanoreceptors but possess different adaptation properties. Lastly, genetic ablation of Prox2 and Runx3 neurons demonstrated their essential roles for esophageal peristalsis in freely behaving mice. Our work defines the identity and function of the vagal neurons that provide mechanosensory feedback from the esophagus to the brain and could lead to better understanding and treatment of esophageal motility disorders.


Assuntos
Subunidade alfa 3 de Fator de Ligação ao Core , Esôfago , Motilidade Gastrointestinal , Proteínas de Homeodomínio , Células Receptoras Sensoriais , Nervo Vago , Animais , Camundongos , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Esôfago/inervação , Esôfago/metabolismo , Esôfago/fisiologia , Motilidade Gastrointestinal/genética , Motilidade Gastrointestinal/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Mecanorreceptores/fisiologia , Neurônios Aferentes/fisiologia , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/fisiologia , Estômago/inervação , Estômago/metabolismo , Estômago/fisiologia , Nervo Vago/fisiologia
7.
Nature ; 615(7953): 660-667, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36890237

RESUMO

Pathogen infection causes a stereotyped state of sickness that involves neuronally orchestrated behavioural and physiological changes1,2. On infection, immune cells release a 'storm' of cytokines and other mediators, many of which are detected by neurons3,4; yet, the responding neural circuits and neuro-immune interaction mechanisms that evoke sickness behaviour during naturalistic infections remain unclear. Over-the-counter medications such as aspirin and ibuprofen are widely used to alleviate sickness and act by blocking prostaglandin E2 (PGE2) synthesis5. A leading model is that PGE2 crosses the blood-brain barrier and directly engages hypothalamic neurons2. Here, using genetic tools that broadly cover a peripheral sensory neuron atlas, we instead identified a small population of PGE2-detecting glossopharyngeal sensory neurons (petrosal GABRA1 neurons) that are essential for influenza-induced sickness behaviour in mice. Ablating petrosal GABRA1 neurons or targeted knockout of PGE2 receptor 3 (EP3) in these neurons eliminates influenza-induced decreases in food intake, water intake and mobility during early-stage infection and improves survival. Genetically guided anatomical mapping revealed that petrosal GABRA1 neurons project to mucosal regions of the nasopharynx with increased expression of cyclooxygenase-2 after infection, and also display a specific axonal targeting pattern in the brainstem. Together, these findings reveal a primary airway-to-brain sensory pathway that detects locally produced prostaglandins and mediates systemic sickness responses to respiratory virus infection.


Assuntos
Barreira Hematoencefálica , Encéfalo , Dinoprostona , Nasofaringe , Infecções por Orthomyxoviridae , Células Receptoras Sensoriais , Animais , Humanos , Camundongos , Comportamento Animal , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Tronco Encefálico/fisiopatologia , Dinoprostona/metabolismo , Ingestão de Líquidos , Ingestão de Alimentos , Influenza Humana/complicações , Influenza Humana/metabolismo , Movimento , Nasofaringe/inervação , Orthomyxoviridae/patogenicidade , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Células Receptoras Sensoriais/metabolismo , Taxa de Sobrevida
8.
Elife ; 122023 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-36810133

RESUMO

Enteroendocrine cells are specialized sensory cells of the gut-brain axis that are sparsely distributed along the intestinal epithelium. The functions of enteroendocrine cells have classically been inferred by the gut hormones they release. However, individual enteroendocrine cells typically produce multiple, sometimes apparently opposing, gut hormones in combination, and some gut hormones are also produced elsewhere in the body. Here, we developed approaches involving intersectional genetics to enable selective access to enteroendocrine cells in vivo in mice. We targeted FlpO expression to the endogenous Villin1 locus (in Vil1-p2a-FlpO knock-in mice) to restrict reporter expression to intestinal epithelium. Combined use of Cre and Flp alleles effectively targeted major transcriptome-defined enteroendocrine cell lineages that produce serotonin, glucagon-like peptide 1, cholecystokinin, somatostatin, or glucose-dependent insulinotropic polypeptide. Chemogenetic activation of different enteroendocrine cell types variably impacted feeding behavior and gut motility. Defining the physiological roles of different enteroendocrine cell types provides an essential framework for understanding sensory biology of the intestine.


Assuntos
Células Enteroendócrinas , Peptídeo 1 Semelhante ao Glucagon , Camundongos , Animais , Células Enteroendócrinas/metabolismo , Linhagem da Célula , Peptídeo 1 Semelhante ao Glucagon/genética , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Polipeptídeo Inibidor Gástrico/metabolismo , Colecistocinina/metabolismo
10.
Nature ; 609(7926): 320-326, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36045291

RESUMO

The nervous system uses various coding strategies to process sensory inputs. For example, the olfactory system uses large receptor repertoires and is wired to recognize diverse odours, whereas the visual system provides high acuity of object position, form and movement1-5. Compared to external sensory systems, principles that underlie sensory processing by the interoceptive nervous system remain poorly defined. Here we developed a two-photon calcium imaging preparation to understand internal organ representations in the nucleus of the solitary tract (NTS), a sensory gateway in the brainstem that receives vagal and other inputs from the body. Focusing on gut and upper airway stimuli, we observed that individual NTS neurons are tuned to detect signals from particular organs and are topographically organized on the basis of body position. Moreover, some mechanosensory and chemosensory inputs from the same organ converge centrally. Sensory inputs engage specific NTS domains with defined locations, each containing heterogeneous cell types. Spatial representations of different organs are further sharpened in the NTS beyond what is achieved by vagal axon sorting alone, as blockade of brainstem inhibition broadens neural tuning and disorganizes visceral representations. These findings reveal basic organizational features used by the brain to process interoceptive inputs.


Assuntos
Tronco Encefálico , Sensação , Tronco Encefálico/anatomia & histologia , Tronco Encefálico/citologia , Tronco Encefálico/fisiologia , Cálcio/metabolismo , Postura/fisiologia , Sensação/fisiologia , Células Receptoras Sensoriais/fisiologia , Núcleo Solitário/anatomia & histologia , Núcleo Solitário/citologia , Núcleo Solitário/fisiologia , Nervo Vago/fisiologia
11.
Cell Rep ; 39(11): 110953, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35705049

RESUMO

Nausea is a discomforting sensation of gut malaise that remains a major clinical challenge. Several visceral poisons induce nausea through the area postrema, a sensory circumventricular organ that detects bloodborne factors. Here, we use genetic approaches based on an area postrema cell atlas to reveal inhibitory neurons that counteract nausea-associated poison responses. The gut hormone glucose insulinotropic peptide (GIP) activates area postrema inhibitory neurons that project locally and elicit inhibitory currents in nausea-promoting excitatory neurons through γ-aminobutyric acid (GABA) receptors. Moreover, GIP blocks behavioral responses to poisons in wild-type mice, with protection eliminated by targeted area postrema neuron ablation. These findings provide insights into the basic organization of nausea-associated brainstem circuits and reveal that area postrema inhibitory neurons are an effective pharmacological target for nausea intervention.


Assuntos
Área Postrema , Venenos , Animais , Área Postrema/fisiologia , Tronco Encefálico , Camundongos , Náusea , Neurônios/fisiologia , Venenos/farmacologia
12.
Neuron ; 110(4): 579-599, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35051375

RESUMO

The vagus nerve is an indispensable body-brain connection that controls vital aspects of autonomic physiology like breathing, heart rate, blood pressure, and gut motility, reflexes like coughing and swallowing, and survival behaviors like feeding, drinking, and sickness responses. Classical physiological studies and recent molecular/genetic approaches have revealed a tremendous diversity of vagal sensory neuron types that innervate different internal organs, with many cell types remaining poorly understood. Here, we review the state of knowledge related to vagal sensory neurons that innervate the respiratory, cardiovascular, and digestive systems. We focus on cell types and their response properties, physiological/behavioral roles, engaged neural circuits and, when possible, sensory receptors. We are only beginning to understand the signal transduction mechanisms used by vagal sensory neurons and upstream sentinel cells, and future studies are needed to advance the field of interoception to the level of mechanistic understanding previously achieved for our external senses.


Assuntos
Interocepção , Nervo Vago , Reflexo , Sensação , Células Receptoras Sensoriais/fisiologia , Nervo Vago/fisiologia
13.
Neuron ; 109(13): 2106-2115.e4, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34077742

RESUMO

The vagus nerve innervates many organs, and most, if not all, of its motor fibers are cholinergic. However, no one knows its organizing principles-whether or not there are dedicated neurons with restricted targets that act as "labeled lines" to perform certain functions, including two opposing ones (gastric contraction versus relaxation). By performing unbiased transcriptional profiling of DMV cholinergic neurons, we discovered seven molecularly distinct subtypes of motor neurons. Then, by using subtype-specific Cre driver mice, we show that two of these subtypes exclusively innervate the glandular domain of the stomach where, remarkably, they contact different enteric neurons releasing functionally opposing neurotransmitters (acetylcholine versus nitric oxide). Thus, the vagus motor nerve communicates via genetically defined labeled lines to control functionally unique enteric neurons within discrete subregions of the gastrointestinal tract. This discovery reveals that the parasympathetic nervous system utilizes a striking division of labor to control autonomic function.


Assuntos
Encéfalo/metabolismo , Neurônios Colinérgicos/metabolismo , Sistema Nervoso Entérico/metabolismo , Mucosa Gástrica/metabolismo , Neurônios Motores/metabolismo , Estômago/inervação , Nervo Vago/metabolismo , Animais , Perfilação da Expressão Gênica , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/metabolismo
14.
Nat Commun ; 12(1): 3798, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145235

RESUMO

Olfactory sensory neurons (OSNs) are functionally defined by their expression of a unique odorant receptor (OR). Mechanisms underlying singular OR expression are well studied, and involve a massive cross-chromosomal enhancer interaction network. Trace amine-associated receptors (TAARs) form a distinct family of olfactory receptors, and here we find that mechanisms regulating Taar gene choice display many unique features. The epigenetic signature of Taar genes in TAAR OSNs is different from that in OR OSNs. We further identify that two TAAR enhancers conserved across placental mammals are absolutely required for expression of the entire Taar gene repertoire. Deletion of either enhancer dramatically decreases the expression probabilities of different Taar genes, while deletion of both enhancers completely eliminates the TAAR OSN populations. In addition, both of the enhancers are sufficient to drive transgene expression in the partially overlapped TAAR OSNs. We also show that the TAAR enhancers operate in cis to regulate Taar gene expression. Our findings reveal a coordinated control of Taar gene choice in OSNs by two remote enhancers, and provide an excellent model to study molecular mechanisms underlying formation of an olfactory subsystem.


Assuntos
Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica/genética , Neurônios Receptores Olfatórios/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Odorantes/metabolismo , Animais , Animais Geneticamente Modificados , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mucosa Olfatória/metabolismo , Imagem Óptica , Receptores Acoplados a Proteínas G/metabolismo , Olfato/genética , Peixe-Zebra/genética
15.
Neuron ; 109(12): 1979-1995.e6, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-34015253

RESUMO

Nutrient sensors allow animals to identify foods rich in specific nutrients. The Drosophila nutrient sensor, diuretic hormone 44 (DH44) neurons, helps the fly to detect nutritive sugar. This sensor becomes operational during starvation; however, the mechanisms by which DH44 neurons or other nutrient sensors are regulated remain unclear. Here, we identified two satiety signals that inhibit DH44 neurons: (1) Piezo-mediated stomach/crop stretch after food ingestion and (2) Neuromedin/Hugin neurosecretory neurons in the ventral nerve cord (VNC) activated by an increase in the internal glucose level. A subset of Piezo+ neurons that express DH44 neuropeptide project to the crop. We found that DH44 neuronal activity and food intake were stimulated following a knockdown of piezo in DH44 neurons or silencing of Hugin neurons in the VNC, even in fed flies. Together, we propose that these two qualitatively distinct peripheral signals work in concert to regulate the DH44 nutrient sensor during the fed state.


Assuntos
Proteínas de Drosophila/metabolismo , Trato Gastrointestinal/fisiologia , Glucose/metabolismo , Canais Iônicos/metabolismo , Inibição Neural/fisiologia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Resposta de Saciedade/fisiologia , Animais , Drosophila , Drosophila melanogaster , Comportamento Alimentar/fisiologia , Trato Gastrointestinal/inervação , Hormônios de Inseto , Mecanotransdução Celular/fisiologia , Neurônios/fisiologia , Estômago/inervação , Estômago/fisiologia
16.
Nature ; 592(7853): 262-266, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33658716

RESUMO

Internal state controls olfaction through poorly understood mechanisms. Odours that represent food, mates, competitors and predators activate parallel neural circuits that may be flexibly shaped by physiological need to alter behavioural outcome1. Here we identify a neuronal mechanism by which hunger selectively promotes attraction to food odours over other olfactory cues. Optogenetic activation of hypothalamic agouti-related peptide (AGRP) neurons enhances attraction to food odours but not to pheromones, and branch-specific activation and inhibition reveal a key role for projections to the paraventricular thalamus. Mice that lack neuropeptide Y (NPY) or NPY receptor type 5 (NPY5R) fail to prefer food odours over pheromones after fasting, and hunger-dependent food-odour attraction is restored by cell-specific NPY rescue in AGRP neurons. Furthermore, acute NPY injection immediately rescues food-odour preference without additional training, indicating that NPY is required for reading olfactory circuits during behavioural expression rather than writing olfactory circuits during odour learning. Together, these findings show that food-odour-responsive neurons comprise an olfactory subcircuit that listens to hunger state through thalamic NPY release, and more generally, provide mechanistic insights into how internal state regulates behaviour.


Assuntos
Alimentos , Fome/fisiologia , Neuropeptídeo Y/metabolismo , Odorantes , Proteína Relacionada com Agouti/metabolismo , Animais , Feminino , Hipotálamo/citologia , Hipotálamo/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Optogenética , Feromônios/metabolismo , Receptores de Neuropeptídeo Y/metabolismo
17.
Neuron ; 109(3): 461-472.e5, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33278342

RESUMO

Nausea, the unpleasant sensation of visceral malaise, remains a mysterious process. The area postrema is implicated in some nausea responses and is anatomically privileged to detect blood-borne signals. To investigate nausea mechanisms, we built an area postrema cell atlas through single-nucleus RNA sequencing, revealing a few neuron types. Using mouse genetic tools for cell-specific manipulation, we discovered excitatory neurons that induce nausea-related behaviors, with one neuron type mediating aversion imposed by multiple poisons. Nausea-associated responses to agonists of identified area postrema receptors were observed and suppressed by targeted cell ablation and/or gene knockout. Anatomical mapping revealed a distributed network of long-range excitatory but not inhibitory projections with subtype-specific patterning. These studies reveal the basic organization of area postrema nausea circuitry and provide a framework toward understanding and therapeutically controlling nausea.


Assuntos
Área Postrema/metabolismo , Comportamento Animal/fisiologia , Náusea/metabolismo , Neurônios/metabolismo , Animais , Proteína Semelhante a Receptor de Calcitonina/genética , Proteína Semelhante a Receptor de Calcitonina/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Camundongos , Camundongos Knockout
18.
Cell ; 181(3): 574-589.e14, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32259485

RESUMO

Sensory neurons initiate defensive reflexes that ensure airway integrity. Dysfunction of laryngeal neurons is life-threatening, causing pulmonary aspiration, dysphagia, and choking, yet relevant sensory pathways remain poorly understood. Here, we discover rare throat-innervating neurons (∼100 neurons/mouse) that guard the airways against assault. We used genetic tools that broadly cover a vagal/glossopharyngeal sensory neuron atlas to map, ablate, and control specific afferent populations. Optogenetic activation of vagal P2RY1 neurons evokes a coordinated airway defense program-apnea, vocal fold adduction, swallowing, and expiratory reflexes. Ablation of vagal P2RY1 neurons eliminates protective responses to laryngeal water and acid challenge. Anatomical mapping revealed numerous laryngeal terminal types, with P2RY1 neurons forming corpuscular endings that appose laryngeal taste buds. Epithelial cells are primary airway sentinels that communicate with second-order P2RY1 neurons through ATP. These findings provide mechanistic insights into airway defense and a general molecular/genetic roadmap for internal organ sensation by the vagus nerve.


Assuntos
Nervo Glossofaríngeo/fisiologia , Faringe/inervação , Nervo Vago/fisiologia , Vias Aferentes , Animais , Feminino , Regulação da Expressão Gênica/genética , Nervo Glossofaríngeo/metabolismo , Laringe/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Purinérgicos P2Y1/genética , Receptores Purinérgicos P2Y1/metabolismo , Células Receptoras Sensoriais/metabolismo , Nervo Vago/metabolismo
19.
Cell Rep ; 29(8): 2192-2201.e3, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31747594

RESUMO

Mechanosensory neurons across physiological systems sense force using diverse terminal morphologies. Arterial baroreceptors are sensory neurons that monitor blood pressure for real-time stabilization of cardiovascular output. Various aortic sensory terminals have been described, but those that sense blood pressure are unclear because of a lack of selective genetic tools. Here, we find that all baroreceptor neurons are marked in Piezo2-ires-Cre mice and then use genetic approaches to visualize the architecture of mechanosensory endings. Cre-guided ablation of vagal and glossopharyngeal PIEZO2 neurons eliminates the baroreceptor reflex and aortic depressor nerve effects on blood pressure and heart rate. Genetic mapping reveals that PIEZO2 neurons form a distinctive mechanosensory structure: macroscopic claws that surround the aortic arch and exude fine end-net endings. Other arterial sensory neurons that form flower-spray terminals are dispensable for baroreception. Together, these findings provide structural insights into how blood pressure is sensed in the aortic vessel wall.


Assuntos
Sistema Nervoso Autônomo/metabolismo , Pressão Sanguínea/fisiologia , Interocepção/fisiologia , Gânglio Nodoso/metabolismo , Pressorreceptores/metabolismo , Animais , Mecanotransdução Celular/fisiologia , Camundongos , Neurônios/metabolismo , Nervo Vago/metabolismo
20.
Cell Metab ; 29(3): 681-694.e5, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30472090

RESUMO

Understanding the neural framework behind appetite control is fundamental to developing effective therapies to combat the obesity epidemic. The paraventricular hypothalamus (PVH) is critical for appetite regulation, yet, the real-time, physiological response properties of PVH neurons to nutrients are unknown. Using a combination of fiber photometry, electrophysiology, immunohistochemistry, and neural manipulation strategies, we determined the population dynamics of four molecularly delineated PVH subsets implicated in feeding behavior: glucagon-like peptide 1 receptor (PVHGlp1r), melanocortin-4 receptor (PVHMc4r), oxytocin (PVHOxt), and corticotropin-releasing hormone (PVHCrh). We identified both calorie- and state-dependent sustained activity increases and decreases in PVHGlp1r and PVHCrh populations, respectively, while observing transient bulk changes of PVHMc4r, but no response in PVHOxt, neurons to food. Furthermore, we highlight the role of PVHGlp1r neurons in orchestrating acute feeding behavior, independent of the anti-obesity drug liraglutide, and demonstrate the indispensability of PVHGlp1r and PVHMc4r, but not PVHOxt or PVHCrh neurons, in body weight maintenance.


Assuntos
Regulação do Apetite/fisiologia , Hormônio Liberador da Corticotropina/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Animais , Ingestão de Energia/fisiologia , Metabolismo Energético , Feminino , Masculino , Camundongos , Neurônios/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...