Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21.588
Filtrar
Mais filtros

Coleção CLAP
Intervalo de ano de publicação
1.
Cell ; 185(23): 4298-4316.e21, 2022 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-36323317

RESUMO

After ingestion of toxin-contaminated food, the brain initiates a series of defensive responses (e.g., nausea, retching, and vomiting). How the brain detects ingested toxin and coordinates diverse defensive responses remains poorly understood. Here, we developed a mouse-based paradigm to study defensive responses induced by bacterial toxins. Using this paradigm, we identified a set of molecularly defined gut-to-brain and brain circuits that jointly mediate toxin-induced defensive responses. The gut-to-brain circuit consists of a subset of Htr3a+ vagal sensory neurons that transmit toxin-related signals from intestinal enterochromaffin cells to Tac1+ neurons in the dorsal vagal complex (DVC). Tac1+ DVC neurons drive retching-like behavior and conditioned flavor avoidance via divergent projections to the rostral ventral respiratory group and lateral parabrachial nucleus, respectively. Manipulating these circuits also interferes with defensive responses induced by the chemotherapeutic drug doxorubicin. These results suggest that food poisoning and chemotherapy recruit similar circuit modules to initiate defensive responses.


Assuntos
Eixo Encéfalo-Intestino , Núcleos Parabraquiais , Nervo Vago , Animais , Camundongos , Neurônios/fisiologia , Neurônios Aferentes/fisiologia , Nervo Vago/fisiologia
2.
Cell ; 181(3): 509-511, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32359432

RESUMO

Recent events bring the importance of respiratory health to the forefront of our collective attention. In this issue of Cell, a new study by Prescott and Umans et al. reveals how a dedicated laryngeal sensory motor reflex circuit protects our airways from aspirated foods or liquids.


Assuntos
Laringe , Nervo Vago , Reflexo
3.
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
4.
Cell ; 179(5): 1129-1143.e23, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31730854

RESUMO

Energy homeostasis requires precise measurement of the quantity and quality of ingested food. The vagus nerve innervates the gut and can detect diverse interoceptive cues, but the identity of the key sensory neurons and corresponding signals that regulate food intake remains unknown. Here, we use an approach for target-specific, single-cell RNA sequencing to generate a map of the vagal cell types that innervate the gastrointestinal tract. We show that unique molecular markers identify vagal neurons with distinct innervation patterns, sensory endings, and function. Surprisingly, we find that food intake is most sensitive to stimulation of mechanoreceptors in the intestine, whereas nutrient-activated mucosal afferents have no effect. Peripheral manipulations combined with central recordings reveal that intestinal mechanoreceptors, but not other cell types, potently and durably inhibit hunger-promoting AgRP neurons in the hypothalamus. These findings identify a key role for intestinal mechanoreceptors in the regulation of feeding.


Assuntos
Comportamento Alimentar/fisiologia , Fenômenos Genéticos , Células Receptoras Sensoriais/fisiologia , Nervo Vago/fisiologia , Proteína Relacionada com Agouti/metabolismo , Animais , Encéfalo/fisiologia , Trato Gastrointestinal/inervação , Marcadores Genéticos , Mecanorreceptores/metabolismo , Camundongos , Nervo Vago/anatomia & histologia , Vísceras/inervação
5.
Cell ; 175(3): 665-678.e23, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30245012

RESUMO

The gut is now recognized as a major regulator of motivational and emotional states. However, the relevant gut-brain neuronal circuitry remains unknown. We show that optical activation of gut-innervating vagal sensory neurons recapitulates the hallmark effects of stimulating brain reward neurons. Specifically, right, but not left, vagal sensory ganglion activation sustained self-stimulation behavior, conditioned both flavor and place preferences, and induced dopamine release from Substantia nigra. Cell-specific transneuronal tracing revealed asymmetric ascending pathways of vagal origin throughout the CNS. In particular, transneuronal labeling identified the glutamatergic neurons of the dorsolateral parabrachial region as the obligatory relay linking the right vagal sensory ganglion to dopamine cells in Substantia nigra. Consistently, optical activation of parabrachio-nigral projections replicated the rewarding effects of right vagus excitation. Our findings establish the vagal gut-to-brain axis as an integral component of the neuronal reward pathway. They also suggest novel vagal stimulation approaches to affective disorders.


Assuntos
Intestinos/fisiologia , Recompensa , Substância Negra/fisiologia , Nervo Vago/fisiologia , Vias Aferentes/metabolismo , Vias Aferentes/fisiologia , Animais , Dopamina/metabolismo , Neurônios Dopaminérgicos/fisiologia , Ácido Glutâmico/metabolismo , Intestinos/inervação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Optogenética
6.
Cell ; 166(1): 209-21, 2016 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-27238020

RESUMO

Neural inputs from internal organs are essential for normal autonomic function. The vagus nerve is a key body-brain connection that monitors the digestive, cardiovascular, and respiratory systems. Within the gastrointestinal tract, vagal sensory neurons detect gut hormones and organ distension. Here, we investigate the molecular diversity of vagal sensory neurons and their roles in sensing gastrointestinal inputs. Genetic approaches allowed targeted investigation of gut-to-brain afferents involved in homeostatic responses to ingested nutrients (GPR65 neurons) and mechanical distension of the stomach and intestine (GLP1R neurons). Optogenetics, in vivo ganglion imaging, and genetically guided anatomical mapping provide direct links between neuron identity, peripheral anatomy, central anatomy, conduction velocity, response properties in vitro and in vivo, and physiological function. These studies clarify the roles of vagal afferents in mediating particular gut hormone responses. Moreover, genetic control over gut-to-brain neurons provides a molecular framework for understanding neural control of gastrointestinal physiology.


Assuntos
Vias Neurais , Neurônios/metabolismo , Células Receptoras Sensoriais/metabolismo , Nervo Vago/metabolismo , Animais , Gânglios/metabolismo , Motilidade Gastrointestinal , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Camundongos , Optogenética , Receptores Acoplados a Proteínas G/metabolismo , Serotonina/metabolismo , Estômago/inervação
7.
Cell ; 161(3): 622-633, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25892222

RESUMO

Breathing is essential for survival and under precise neural control. The vagus nerve is a major conduit between lung and brain required for normal respiration. Here, we identify two populations of mouse vagus nerve afferents (P2ry1, Npy2r), each a few hundred neurons, that exert powerful and opposing effects on breathing. Genetically guided anatomical mapping revealed that these neurons densely innervate the lung and send long-range projections to different brainstem targets. Npy2r neurons are largely slow-conducting C fibers, while P2ry1 neurons are largely fast-conducting A fibers that contact pulmonary endocrine cells (neuroepithelial bodies). Optogenetic stimulation of P2ry1 neurons acutely silences respiration, trapping animals in exhalation, while stimulating Npy2r neurons causes rapid, shallow breathing. Activating P2ry1 neurons did not impact heart rate or gastric pressure, other autonomic functions under vagal control. Thus, the vagus nerve contains intermingled sensory neurons constituting genetically definable labeled lines with different anatomical connections and physiological roles.


Assuntos
Respiração , Células Receptoras Sensoriais/fisiologia , Nervo Vago/citologia , Animais , Tronco Encefálico/fisiologia , Pulmão/inervação , Camundongos , Receptores Acoplados a Proteínas G/metabolismo , Células Receptoras Sensoriais/citologia , Nervo Vago/fisiologia
8.
Nature ; 630(8017): 695-703, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38692285

RESUMO

The body-brain axis is emerging as a principal conductor of organismal physiology. It senses and controls organ function1,2, metabolism3 and nutritional state4-6. Here we show that a peripheral immune insult strongly activates the body-brain axis to regulate immune responses. We demonstrate that pro-inflammatory and anti-inflammatory cytokines communicate with distinct populations of vagal neurons to inform the brain of an emerging inflammatory response. In turn, the brain tightly modulates the course of the peripheral immune response. Genetic silencing of this body-brain circuit produced unregulated and out-of-control inflammatory responses. By contrast, activating, rather than silencing, this circuit affords neural control of immune responses. We used single-cell RNA sequencing, combined with functional imaging, to identify the circuit components of this neuroimmune axis, and showed that its selective manipulation can effectively suppress the pro-inflammatory response while enhancing an anti-inflammatory state. The brain-evoked transformation of the course of an immune response offers new possibilities in the modulation of a wide range of immune disorders, from autoimmune diseases to cytokine storm and shock.


Assuntos
Encéfalo , Citocinas , Inflamação , Neuroimunomodulação , Animais , Feminino , Masculino , Camundongos , Encéfalo/citologia , Encéfalo/imunologia , Encéfalo/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Neuroimunomodulação/imunologia , Neuroimunomodulação/fisiologia , Neurônios/fisiologia , Nervo Vago/citologia , Nervo Vago/fisiologia , Análise da Expressão Gênica de Célula Única
9.
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
10.
Nature ; 623(7986): 387-396, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37914931

RESUMO

Visceral sensory pathways mediate homeostatic reflexes, the dysfunction of which leads to many neurological disorders1. The Bezold-Jarisch reflex (BJR), first described2,3 in 1867, is a cardioinhibitory reflex that is speculated to be mediated by vagal sensory neurons (VSNs) that also triggers syncope. However, the molecular identity, anatomical organization, physiological characteristics and behavioural influence of cardiac VSNs remain mostly unknown. Here we leveraged single-cell RNA-sequencing data and HYBRiD tissue clearing4 to show that VSNs that express neuropeptide Y receptor Y2 (NPY2R) predominately connect the heart ventricular wall to the area postrema. Optogenetic activation of NPY2R VSNs elicits the classic triad of BJR responses-hypotension, bradycardia and suppressed respiration-and causes an animal to faint. Photostimulation during high-resolution echocardiography and laser Doppler flowmetry with behavioural observation revealed a range of phenotypes reflected in clinical syncope, including reduced cardiac output, cerebral hypoperfusion, pupil dilation and eye-roll. Large-scale Neuropixels brain recordings and machine-learning-based modelling showed that this manipulation causes the suppression of activity across a large distributed neuronal population that is not explained by changes in spontaneous behavioural movements. Additionally, bidirectional manipulation of the periventricular zone had a push-pull effect, with inhibition leading to longer syncope periods and activation inducing arousal. Finally, ablating NPY2R VSNs specifically abolished the BJR. Combined, these results demonstrate a genetically defined cardiac reflex that recapitulates characteristics of human syncope at physiological, behavioural and neural network levels.


Assuntos
Coração , Reflexo , Células Receptoras Sensoriais , Síncope , Nervo Vago , Humanos , Área Postrema , Bradicardia/complicações , Bradicardia/fisiopatologia , Baixo Débito Cardíaco/complicações , Baixo Débito Cardíaco/fisiopatologia , Ecocardiografia , Coração/fisiologia , Frequência Cardíaca , Hipotensão/complicações , Hipotensão/fisiopatologia , Fluxometria por Laser-Doppler , Rede Nervosa , Reflexo/fisiologia , Células Receptoras Sensoriais/fisiologia , Análise da Expressão Gênica de Célula Única , Síncope/complicações , Síncope/etiologia , Nervo Vago/citologia , Nervo Vago/fisiologia
11.
Annu Rev Neurosci ; 43: 337-353, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32101483

RESUMO

Guided by sight, scent, texture, and taste, animals ingest food. Once ingested, it is up to the gut to make sense of the food's nutritional value. Classic sensory systems rely on neuroepithelial circuits to convert stimuli into signals that guide behavior. However, sensation of the gut milieu was thought to be mediated only by the passive release of hormones until the discovery of synapses in enteroendocrine cells. These are gut sensory epithelial cells, and those that form synapses are referred to as neuropod cells. Neuropod cells provide the foundation for the gut to transduce sensory signals from the intestinal milieu to the brain through fast neurotransmission onto neurons, including those of the vagus nerve. These findings have sparked a new field of exploration in sensory neurobiology-that of gut-brain sensory transduction.


Assuntos
Encéfalo/fisiologia , Células Enteroendócrinas/fisiologia , Sinapses/fisiologia , Nervo Vago/fisiologia , Animais , Humanos , Neurônios/fisiologia , Transdução de Sinais/fisiologia
12.
Nature ; 602(7897): 468-474, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35082448

RESUMO

Ingested food and water stimulate sensory systems in the oropharyngeal and gastrointestinal areas before absorption1,2. These sensory signals modulate brain appetite circuits in a feed-forward manner3-5. Emerging evidence suggests that osmolality sensing in the gut rapidly inhibits thirst neurons upon water intake. Nevertheless, it remains unclear how peripheral sensory neurons detect visceral osmolality changes, and how they modulate thirst. Here we use optical and electrical recording combined with genetic approaches to visualize osmolality responses from sensory ganglion neurons. Gut hypotonic stimuli activate a dedicated vagal population distinct from mechanical-, hypertonic- or nutrient-sensitive neurons. We demonstrate that hypotonic responses are mediated by vagal afferents innervating the hepatic portal area (HPA), through which most water and nutrients are absorbed. Eliminating sensory inputs from this area selectively abolished hypotonic but not mechanical responses in vagal neurons. Recording from forebrain thirst neurons and behavioural analyses show that HPA-derived osmolality signals are required for feed-forward thirst satiation and drinking termination. Notably, HPA-innervating vagal afferents do not sense osmolality itself. Instead, these responses are mediated partly by vasoactive intestinal peptide secreted after water ingestion. Together, our results reveal visceral hypoosmolality as an important vagal sensory modality, and that intestinal osmolality change is translated into hormonal signals to regulate thirst circuit activity through the HPA pathway.


Assuntos
Intestinos , Saciação , Células Receptoras Sensoriais , Sede , Gânglios Sensitivos/citologia , Intestinos/citologia , Intestinos/inervação , Concentração Osmolar , Pressão Osmótica , Saciação/fisiologia , Células Receptoras Sensoriais/citologia , Sede/fisiologia , Nervo Vago/citologia , Nervo Vago/fisiologia , Água/metabolismo
13.
Nature ; 610(7933): 722-730, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36070796

RESUMO

The perception of fat evokes strong appetitive and consummatory responses1. Here we show that fat stimuli can induce behavioural attraction even in the absence of a functional taste system2,3. We demonstrate that fat acts after ingestion via the gut-brain axis to drive preference for fat. Using single-cell data, we identified the vagal neurons responding to intestinal delivery of fat, and showed that genetic silencing of this gut-to-brain circuit abolished the development of fat preference. Next, we compared the gut-to-brain pathways driving preference for fat versus sugar4, and uncovered two parallel systems, one functioning as a general sensor of essential nutrients, responding to intestinal stimulation with sugar, fat and amino acids, whereas the other is activated only by fat stimuli. Finally, we engineered mice lacking candidate receptors to detect the presence of intestinal fat, and validated their role as the mediators of gut-to-brain fat-evoked responses. Together, these findings reveal distinct cells and receptors that use the gut-brain axis as a fundamental conduit for the development of fat preference.


Assuntos
Eixo Encéfalo-Intestino , Encéfalo , Preferências Alimentares , Intestinos , Neurônios , Animais , Camundongos , Aminoácidos/metabolismo , Encéfalo/citologia , Encéfalo/fisiologia , Neurônios/metabolismo , Açúcares/metabolismo , Nervo Vago/citologia , Nervo Vago/fisiologia , Preferências Alimentares/fisiologia , Análise de Célula Única , Eixo Encéfalo-Intestino/genética , Eixo Encéfalo-Intestino/fisiologia , Intestinos/inervação , Intestinos/metabolismo
14.
Nature ; 603(7903): 878-884, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35296859

RESUMO

Interoception, the ability to timely and precisely sense changes inside the body, is critical for survival1-4. Vagal sensory neurons (VSNs) form an important body-to-brain connection, navigating visceral organs along the rostral-caudal axis of the body and crossing the surface-lumen axis of organs into appropriate tissue layers5,6. The brain can discriminate numerous body signals through VSNs, but the underlying coding strategy remains poorly understood. Here we show that VSNs code visceral organ, tissue layer and stimulus modality-three key features of an interoceptive signal-in different dimensions. Large-scale single-cell profiling of VSNs from seven major organs in mice using multiplexed projection barcodes reveals a 'visceral organ' dimension composed of differentially expressed gene modules that code organs along the body's rostral-caudal axis. We discover another 'tissue layer' dimension with gene modules that code the locations of VSN endings along the surface-lumen axis of organs. Using calcium-imaging-guided spatial transcriptomics, we show that VSNs are organized into functional units to sense similar stimuli across organs and tissue layers; this constitutes a third 'stimulus modality' dimension. The three independent feature-coding dimensions together specify many parallel VSN pathways in a combinatorial manner and facilitate the complex projection of VSNs in the brainstem. Our study highlights a multidimensional coding architecture of the mammalian vagal interoceptive system for effective signal communication.


Assuntos
Percepção , Psicofisiologia , Nervo Vago , Órgão Vomeronasal , Animais , Encéfalo/metabolismo , Cálcio/metabolismo , Mamíferos/metabolismo , Camundongos , Células Receptoras Sensoriais/metabolismo
15.
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
16.
Nature ; 598(7882): 641-645, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34646018

RESUMO

Somatosensory autonomic reflexes allow electroacupuncture stimulation (ES) to modulate body physiology at distant sites1-6 (for example, suppressing severe systemic inflammation6-9). Since the 1970s, an emerging organizational rule about these reflexes has been the presence of body-region specificity1-6. For example, ES at the hindlimb ST36 acupoint but not the abdominal ST25 acupoint can drive the vagal-adrenal anti-inflammatory axis in mice10,11. The neuroanatomical basis of this somatotopic organization is, however, unknown. Here we show that PROKR2Cre-marked sensory neurons, which innervate the deep hindlimb fascia (for example, the periosteum) but not abdominal fascia (for example, the peritoneum), are crucial for driving the vagal-adrenal axis. Low-intensity ES at the ST36 site in mice with ablated PROKR2Cre-marked sensory neurons failed to activate hindbrain vagal efferent neurons or to drive catecholamine release from adrenal glands. As a result, ES no longer suppressed systemic inflammation induced by bacterial endotoxins. By contrast, spinal sympathetic reflexes evoked by high-intensity ES at both ST25 and ST36 sites were unaffected. We also show that optogenetic stimulation of PROKR2Cre-marked nerve terminals through the ST36 site is sufficient to drive the vagal-adrenal axis but not sympathetic reflexes. Furthermore, the distribution patterns of PROKR2Cre nerve fibres can retrospectively predict body regions at which low-intensity ES will or will not effectively produce anti-inflammatory effects. Our studies provide a neuroanatomical basis for the selectivity and specificity of acupoints in driving specific autonomic pathways.


Assuntos
Glândulas Suprarrenais/fisiologia , Sistema Nervoso Autônomo , Eletroacupuntura , Nervo Vago/fisiologia , Pontos de Acupuntura , Animais , Membro Posterior/inervação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reflexo
17.
Annu Rev Physiol ; 85: 71-91, 2023 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-36170660

RESUMO

Coughing is a dynamic physiological process resulting from input of vagal sensory neurons innervating the airways and perceived airway irritation. Although cough serves to protect and clear the airways, it can also be exploited by respiratory pathogens to facilitate disease transmission. Microbial components or infection-induced inflammatory mediators can directly interact with sensory nerve receptors to induce a cough response. Analysis of cough-generated aerosols and transmission studies have further demonstrated how infectious disease is spread through coughing. This review summarizes the neurophysiology of cough, cough induction by respiratory pathogens and inflammation, and cough-mediated disease transmission.


Assuntos
Doenças Transmissíveis , Tosse , Humanos , Sistema Respiratório/inervação , Nervo Vago/fisiologia , Células Receptoras Sensoriais
18.
Semin Cell Dev Biol ; 156: 210-218, 2024 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-37507330

RESUMO

The vagus nerve vitally connects the brain and body to coordinate digestive, cardiorespiratory, and immune functions. Its efferent neurons, which project their axons from the brainstem to the viscera, are thought to comprise "functional units" - neuron populations dedicated to the control of specific vagal reflexes or organ functions. Previous research indicates that these functional units differ from one another anatomically, neurochemically, and physiologically but have yet to define their identity in an experimentally tractable way. However, recent work with genetic technology and single-cell genomics suggests that genetically distinct subtypes of neurons may be the functional units of the efferent vagus. Here we review how these approaches are revealing the organizational principles of the efferent vagus in unprecedented detail.


Assuntos
Neurônios Eferentes , Nervo Vago , Nervo Vago/metabolismo , Neurônios/fisiologia
19.
Semin Cell Dev Biol ; 156: 244-252, 2024 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-37500301

RESUMO

Maintaining blood glucose at an appropriate physiological level requires precise coordination of multiple organs and tissues. The vagus nerve bidirectionally connects the central nervous system with peripheral organs crucial to glucose mobilization, nutrient storage, and food absorption, thereby presenting a key pathway for the central control of blood glucose levels. However, the precise mechanisms by which vagal populations that target discrete tissues participate in glucoregulation are much less clear. Here we review recent advances unraveling the cellular identity, neuroanatomical organization, and functional contributions of both vagal efferents and vagal afferents in the control of systemic glucose metabolism. We focus on their involvement in relaying glucoregulatory cues from the brain to peripheral tissues, particularly the pancreatic islet, and by sensing and transmitting incoming signals from ingested food to the brain. These recent findings - largely driven by advances in viral approaches, RNA sequencing, and cell-type selective manipulations and tracings - have begun to clarify the precise vagal neuron populations involved in the central coordination of glucose levels, and raise interesting new possibilities for the treatment of glucose metabolism disorders such as diabetes.


Assuntos
Glicemia , Nervo Vago , Glicemia/metabolismo , Nervo Vago/metabolismo , Glucose/metabolismo
20.
Semin Cell Dev Biol ; 156: 228-243, 2024 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-37558522

RESUMO

The communication between the gut and brain is crucial for regulating various essential physiological functions, such as energy balance, fluid homeostasis, immune response, and emotion. The vagal sensory pathway plays an indispensable role in connecting the gut to the brain. Recently, our knowledge of the vagal gut-brain axis has significantly advanced through molecular genetic studies, revealing a diverse range of vagal sensory cell types with distinct peripheral innervations, response profiles, and physiological functions. Here, we review the current understanding of how vagal sensory neurons contribute to gut-brain communication. First, we highlight recent transcriptomic and genetic approaches that have characterized different vagal sensory cell types. Then, we focus on discussing how different subtypes encode numerous gut-derived signals and how their activities are translated into physiological and behavioral regulations. The emerging insights into the diverse cell types and functional properties of vagal sensory neurons have paved the way for exciting future directions, which may provide valuable insights into potential therapeutic targets for disorders involving gut-brain communication.


Assuntos
Encéfalo , Nervo Vago , Vias Aferentes/fisiologia , Encéfalo/fisiologia , Nervo Vago/fisiologia , Células Receptoras Sensoriais , Perfilação da Expressão Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA