Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 905
Filtrar
1.
Nature ; 623(7986): 387-396, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37914931

RESUMEN

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.


Asunto(s)
Corazón , Reflejo , Células Receptoras Sensoriales , Síncope , Nervio Vago , Humanos , Área Postrema , Bradicardia/complicaciones , Bradicardia/fisiopatología , Gasto Cardíaco Bajo/complicaciones , Gasto Cardíaco Bajo/fisiopatología , Ecocardiografía , Corazón/fisiología , Frecuencia Cardíaca , Hipotensión/complicaciones , Hipotensión/fisiopatología , Flujometría por Láser-Doppler , Red Nerviosa , Reflejo/fisiología , Células Receptoras Sensoriales/fisiología , Análisis de Expresión Génica de una Sola Célula , Síncope/complicaciones , Síncope/etiología , Nervio Vago/citología , Nervio Vago/fisiología
2.
J Physiol ; 601(10): 1881-1896, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36975145

RESUMEN

Circadian regulation of autonomic reflex pathways pairs physiological function with the daily light cycle. The brainstem nucleus of the solitary tract (NTS) is a key candidate for rhythmic control of the autonomic nervous system. Here we investigated circadian regulation of NTS neurotransmission and synaptic throughput using patch-clamp electrophysiology in brainstem slices from mice. We found that spontaneous quantal glutamate release onto NTS neurons showed strong circadian rhythmicity, with the highest rate of release during the light phase and the lowest in the dark, that were sufficient to drive day/night differences in constitutive postsynaptic action potential firing. In contrast, afferent evoked action potential throughput was enhanced during the dark and diminished in the light. Afferent-driven synchronous release pathways showed a similar decrease in release probability that did not explain the enhanced synaptic throughput during the night. However, analysis of postsynaptic membrane properties revealed diurnal changes in conductance, which, when coupled with the circadian changes in glutamate release pathways, tuned synaptic throughput between the light and dark phases. These coordinated pre-/postsynaptic changes encode nuanced control over synaptic performance and pair NTS action potential firing and vagal throughput with time of day. KEY POINTS: Vagal afferent neurons relay information from peripheral organs to the brainstem nucleus of the solitary tract (NTS) to initiate autonomic reflex pathways as well as providing important controls of food intake, digestive function and energy balance. Vagally mediated reflexes and behaviours are under strong circadian regulation. Diurnal fluctuations in presynaptic vesicle release pathways and postsynaptic membrane conductances provide nuanced control over NTS action potential firing and vagal synaptic throughput. Coordinated pre-/postsynaptic changes represent a fundamental mechanism mediating daily changes in vagal afferent signalling and autonomic function.


Asunto(s)
Ritmo Circadiano , Ácido Glutámico , Núcleo Solitario , Sinapsis , Ritmo Circadiano/fisiología , Ácido Glutámico/metabolismo , Núcleo Solitario/citología , Núcleo Solitario/fisiología , Sinapsis/metabolismo , Neuronas Aferentes/metabolismo , Nervio Vago/citología , Nervio Vago/fisiología , Potenciales de Acción , Masculino , Animales , Ratones , Ganglio Nudoso/metabolismo , Transducción de Señal , Conductividad Eléctrica , Técnicas de Placa-Clamp
3.
Nature ; 610(7933): 722-730, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36070796

RESUMEN

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.


Asunto(s)
Eje Cerebro-Intestino , Encéfalo , Preferencias Alimentarias , Intestinos , Neuronas , Animales , Ratones , Aminoácidos/metabolismo , Encéfalo/citología , Encéfalo/fisiología , Neuronas/metabolismo , Azúcares/metabolismo , Nervio Vago/citología , Nervio Vago/fisiología , Preferencias Alimentarias/fisiología , Análisis de la Célula Individual , Eje Cerebro-Intestino/genética , Eje Cerebro-Intestino/fisiología , Intestinos/inervación , Intestinos/metabolismo
4.
Nature ; 602(7897): 468-474, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35082448

RESUMEN

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.


Asunto(s)
Intestinos , Saciedad , Células Receptoras Sensoriales , Sed , Ganglios Sensoriales/citología , Intestinos/citología , Intestinos/inervación , Concentración Osmolar , Presión Osmótica , Saciedad/fisiología , Células Receptoras Sensoriales/citología , Sed/fisiología , Nervio Vago/citología , Nervio Vago/fisiología , Agua/metabolismo
5.
Physiol Rep ; 9(18): e15056, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34582125

RESUMEN

The intrinsic cardiac nervous system represents the final site of signal integration for neurotransmission to the myocardium to enable local control of cardiac performance. The electrophysiological characteristics and ganglionic transmission of adult mouse intrinsic cardiac ganglion (ICG) neurons were investigated using a whole-mount ganglion preparation of the excised right atrial ganglion plexus and intracellular microelectrode recording techniques. The passive and active electrical properties of ICG neurons and synaptic transmission including synaptic response strength and efficacy as a function of stimulation frequency were examined. The resting membrane potential and input resistance of ICG neurons were -47.9 ± 4.0 mV and 197.2 ± 81.5 MΩ, respectively. All neurons had somatic action potentials with overshoots of >+15 mV and after-hyperpolarizations having an average of 10 mV amplitude and ~45 ms half duration. Phasic discharge activities were recorded from the majority of neurons studied and several types of excitatory synaptic responses were recorded following inputs from the vagus or interganglionic nerve trunk(s). Most postganglionic neurons (>75%) received a strong, suprathreshold synaptic input and reliably followed high-frequency repetitive nerve stimulation up to at least 50 Hz. Nerve-evoked synaptic transmission was blocked by extracellular Cd2+ , ω-conotoxin CVIE, or α-conotoxin RegIIA, a selective α3-containing nicotinic acetylcholine receptor antagonist. Synaptic transmission and the electrical properties of murine ICG neurons contribute to the pattern of discharge which regulates chronotropic, dromotropic, and inotropic elements of cardiac function.


Asunto(s)
Potenciales de Acción , Corazón/inervación , Neuronas/fisiología , Transmisión Sináptica , Nervio Vago/fisiología , Animales , Cadmio/farmacología , Conotoxinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Antagonistas Nicotínicos/farmacología , Nervio Vago/citología , Nervio Vago/efectos de los fármacos
6.
Brain Res ; 1769: 147625, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34416255

RESUMEN

The nucleus of the solitary tract (NTS) receives viscerosensory information from the vagus nerve to regulate diverse homeostatic reflex functions. The NTS projects to a wide network of other brain regions, including the paraventricular nucleus of the hypothalamus (PVN). Here we examined the synaptic characteristics of primary afferent pathways to PVN-projecting NTS neurons in rat brainstem slices.Expression of the Transient Receptor Potential Vanilloid receptor (TRPV1+ ) distinguishes C-fiber afferents within the solitary tract (ST) from A-fibers (TRPV1-). We used resiniferatoxin (RTX), a TRPV1 agonist, to differentiate the two. The variability in the latency (jitter) of evoked excitatory postsynaptic currents (ST-EPSCs) distinguished monosynaptic from polysynaptic ST-EPSCs. Rhodamine injected into PVN was retrogradely transported to identify PVN-projecting NTS neurons within brainstem slices. Graded shocks to the ST elicited all-or-none EPSCs in rhodamine-positive NTS neurons with latencies that had either low jitter (<200 µs - monosynaptic), high jitter (>200 µs - polysynaptic inputs) or both. RTX blocked ST-evoked TRPV1 + EPSCs whether mono- or polysynaptic. Most PVN-projecting NTS neurons (17/21 neurons) had at least one input polysynaptically connected to the ST. Compared to unlabeled NTS neurons, PVN-projecting NTS neurons were more likely to receive indirect inputs and be higher order. Surprisingly, sEPSC rates for PVN-projecting neurons were double that of unlabeled NTS neurons. The ST synaptic responses for PVN-projecting NTS neurons were either all TRPV1+ or all TRPV1-, including neurons that received both direct and indirect inputs. Overall, PVN-projecting NTS neurons received direct and indirect vagal afferent information with strict segregation regarding TRPV1 expression.


Asunto(s)
Vías Aferentes/fisiología , Fibras Nerviosas Amielínicas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Nervio Vago/fisiología , Animales , Diterpenos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Masculino , Núcleo Hipotalámico Paraventricular/citología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Núcleo Solitario/metabolismo , Sinapsis/efectos de los fármacos , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/metabolismo , Nervio Vago/citología
7.
Nat Commun ; 12(1): 157, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33420038

RESUMEN

The vagus nerve supports diverse autonomic functions and behaviors important for health and survival. To understand how specific components of the vagus contribute to behaviors and long-term physiological effects, it is critical to modulate their activity with anatomical specificity in awake, freely behaving conditions using reliable methods. Here, we introduce an organ-specific scalable, multimodal, wireless optoelectronic device for precise and chronic optogenetic manipulations in vivo. When combined with an advanced, coil-antenna system and a multiplexing strategy for powering 8 individual homecages using a single RF transmitter, the proposed wireless telemetry enables low cost, high-throughput, and precise functional mapping of peripheral neural circuits, including long-term behavioral and physiological measurements. Deployment of these technologies reveals an unexpected role for stomach, non-stretch vagal sensory fibers in suppressing appetite and demonstrates the durability of the miniature wireless device inside harsh gastric conditions.


Asunto(s)
Apetito/fisiología , Ensayos Analíticos de Alto Rendimiento/instrumentación , Optogenética/instrumentación , Estómago/fisiología , Nervio Vago/fisiología , Animales , Técnicas de Observación Conductual/instrumentación , Péptido Relacionado con Gen de Calcitonina/genética , Células Quimiorreceptoras/fisiología , Diseño de Equipo , Femenino , Masculino , Ratones Transgénicos , Modelos Animales , Vías Nerviosas/fisiología , Tecnología de Sensores Remotos/instrumentación , Estómago/citología , Estómago/inervación , Nervio Vago/citología , Tecnología Inalámbrica/instrumentación
8.
PLoS One ; 16(1): e0245785, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33481922

RESUMEN

Evocalcet is a novel calcimimetic agent with fewer gastrointestinal (GI) adverse effects compared to cinacalcet. Although it is thought that cinacalcet induces GI side effects through the direct stimulation of the calcium receptor (CaR) expressed in the GI tract, the differences in the direct stimulatory effects of these two drugs on the GI tract have not been reported. In this study, we analyzed the difference in the GI effects of these two calcimimetic agents using miniature pigs by detecting vagus nerve stimulation after oral administration of the agents. Although cinacalcet induced vomiting in miniature pigs, evocalcet never induced emetic symptoms. A significant increase in the vagus nerve action potentials was observed after the administration of cinacalcet. Although the increase of that after the administration of evocalcet was mild and not significant in comparison to that in the vehicle group, it was not significantly different from the vagus nerve action potentials after cinacalcet treatment.


Asunto(s)
Tracto Gastrointestinal/inervación , Naftalenos/efectos adversos , Pirrolidinas/efectos adversos , Nervio Vago/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Masculino , Porcinos , Porcinos Enanos , Nervio Vago/citología , Nervio Vago/fisiología , Vómitos/inducido químicamente
9.
J Neurophysiol ; 125(1): 199-210, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33296617

RESUMEN

Vagal afferent fibers contact neurons in the nucleus of the solitary tract (NTS) and release glutamate via three distinct release pathways: synchronous, asynchronous, and spontaneous. The presence of TRPV1 in vagal afferents is predictive of activity-dependent asynchronous glutamate release along with temperature-sensitive spontaneous vesicle fusion. However, pharmacological blockade or genetic deletion of TRPV1 does not eliminate the asynchronous profile and only attenuates the temperature-dependent spontaneous release at high temperatures (>40°C), indicating additional temperature-sensitive calcium conductance(s) contributing to these release pathways. The transient receptor potential cation channel melastatin subtype 3 (TRPM3) is a calcium-selective channel that functions as a thermosensor (30-37°C) in somatic primary afferent neurons. We predict that TRPM3 is expressed in vagal afferent neurons and contributes to asynchronous and spontaneous glutamate release pathways. We investigated these hypotheses via measurements on cultured nodose neurons and in brainstem slice preparations containing vagal afferent to NTS synaptic contacts. We found histological and genetic evidence that TRPM3 is highly expressed in vagal afferent neurons. The TRPM3-selective agonist, pregnenolone sulfate, rapidly and reversibly activated the majority (∼70%) of nodose neurons; most of which also contained TRPV1. We confirmed the role of TRPM3 with pharmacological blockade and genetic deletion. In the brain, TRPM3 signaling strongly controlled both basal and temperature-driven spontaneous glutamate release. Surprisingly, genetic deletion of TRPM3 did not alter synchronous or asynchronous glutamate release. These results provide convergent evidence that vagal afferents express functional TRPM3 that serves as an additional temperature-sensitive calcium conductance involved in controlling spontaneous glutamate release onto neurons in the NTS.NEW & NOTEWORTHY Vagal afferent signaling coordinates autonomic reflex function and informs associated behaviors. Thermosensitive transient receptor potential (TRP) channels detect temperature and nociceptive stimuli in somatosensory afferent neurons, however their role in vagal signaling remains less well understood. We report that the TRPM3 ion channel provides a major thermosensitive point of control over vagal signaling and synaptic transmission. We conclude that TRPM3 translates physiological changes in temperature to neurophysiological outputs and can serve as a cellular integrator in vagal afferent signaling.


Asunto(s)
Ácido Glutámico/metabolismo , Neuronas Aferentes/metabolismo , Canales Catiónicos TRPM/metabolismo , Nervio Vago/metabolismo , Potenciales de Acción , Animales , Potenciales Postsinápticos Excitadores , Exocitosis , Calor , Masculino , Neuronas Aferentes/fisiología , Pregnenolona/farmacología , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPM/agonistas , Canales Catiónicos TRPM/genética , Nervio Vago/citología , Nervio Vago/fisiología
10.
J Neurophysiol ; 125(1): 86-104, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33085556

RESUMEN

Biophysically based computational models of nerve fibers are important tools for designing electrical stimulation therapies, investigating drugs that affect ion channels, and studying diseases that affect neurons. Although peripheral nerves are primarily composed of unmyelinated axons (i.e., C-fibers), most modeling efforts focused on myelinated axons. We implemented the single-compartment model of vagal afferents from Schild et al. (1994) (Schild JH, Clark JW, Hay M, Mendelowitz D, Andresen MC, Kunze DL. J Neurophysiol 71: 2338-2358, 1994) and extended the model into a multicompartment axon, presenting the first cable model of a C-fiber vagal afferent. We also implemented the updated parameters from the Schild and Kunze (1997) model (Schild JH, Kunze DL. J Neurophysiol 78: 3198-3209, 1997). We compared the responses of these novel models with those of three published models of unmyelinated axons (Rattay F, Aberham M. IEEE Trans Biomed Eng 40: 1201-1209, 1993; Sundt D, Gamper N, Jaffe DB. J Neurophysiol 114: 3140-3153, 2015; Tigerholm J, Petersson ME, Obreja O, Lampert A, Carr R, Schmelz M, Fransén E. J Neurophysiol 111: 1721-1735, 2014) and with experimental data from single-fiber recordings. Comparing the two models by Schild et al. (1994, 1997) revealed that differences in rest potential and action potential shape were driven by changes in maximum conductances rather than changes in sodium channel dynamics. Comparing the five model axons, the conduction speeds and strength-duration responses were largely within expected ranges, but none of the models captured the experimental threshold recovery cycle-including a complete absence of late subnormality in the models-and their action potential shapes varied dramatically. The Tigerholm et al. (2014) model best reproduced the experimental data, but these modeling efforts make clear that additional data are needed to parameterize and validate future models of autonomic C-fibers.NEW & NOTEWORTHY Peripheral nerves are primarily composed of unmyelinated axons, and there is growing interest in electrical stimulation of the autonomic nervous system to treat various diseases. We present the first cable model of an unmyelinated vagal nerve fiber and compare its ion channel isoforms and conduction responses with other published models of unmyelinated axons, establishing important tools for advancing modeling of autonomic nerves.


Asunto(s)
Potenciales de Acción , Axones/fisiología , Modelos Neurológicos , Fibras Nerviosas Amielínicas/fisiología , Animales , Neuronas Aferentes/fisiología , Nervio Vago/citología , Nervio Vago/fisiología
11.
Nat Commun ; 11(1): 6295, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33293550

RESUMEN

The central melanocortin system plays a fundamental role in the control of feeding and body weight. Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC) also regulate overall glucose homeostasis via insulin-dependent and -independent pathways. Here, we report that a subset of ARC POMC neurons innervate the liver via preganglionic parasympathetic acetylcholine (ACh) neurons in the dorsal motor nucleus of the vagus (DMV). Optogenetic stimulation of this liver-projecting melanocortinergic pathway elevates blood glucose levels that is associated with increased expression of hepatic gluconeogenic enzymes in female and male mice. Pharmacological blockade and knockdown of the melanocortin-4 receptor gene in the DMV abolish this stimulation-induced effect. Activation of melanocortin-4 receptors inhibits DMV cholinergic neurons and optogenetic inhibition of liver-projecting parasympathetic cholinergic fibers increases blood glucose levels. This elevated blood glucose is not due to altered pancreatic hormone release. Interestingly, insulin-induced hypoglycemia increases ARC POMC neuron activity. Hence, this liver-projecting melanocortinergic circuit that we identified may play a critical role in the counterregulatory response to hypoglycemia.


Asunto(s)
Glucemia/metabolismo , Hipoglucemia/etiología , Hígado/inervación , Proopiomelanocortina/metabolismo , Nervio Vago/metabolismo , Acetilcolina/metabolismo , Potenciales de Acción/fisiología , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Glucemia/análisis , Neuronas Colinérgicas/metabolismo , Corticosterona/sangre , Corticosterona/metabolismo , Modelos Animales de Enfermedad , Vías Eferentes/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Glucagón/sangre , Glucagón/metabolismo , Gluconeogénesis/genética , Humanos , Hipoglucemia/sangre , Hipoglucemia/diagnóstico , Insulina/sangre , Insulina/metabolismo , Hígado/enzimología , Masculino , Ratones , Optogenética , ARN Mensajero/metabolismo , Receptor de Melanocortina Tipo 4/antagonistas & inhibidores , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Regulación hacia Arriba , Nervio Vago/citología
12.
STAR Protoc ; 1(1): 100030, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-33111081

RESUMEN

Vagal sensory neurons relay viscero- and somatosensory information from within the body and play a key role in maintaining physiological homeostasis. We recently characterized the diversity of vagal sensory neurons in the mouse using a single-cell transcriptomics approach. Here, we provide an in-depth protocol for the extraction of mouse vagal ganglia and the production of high-quality single-cell suspensions from this tissue. This effective protocol can also be applied for use with other peripheral and central neuron populations with few modifications. For complete details on the use and execution of this protocol, please refer to Kupari et al. (2019).


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Perfilación de la Expresión Génica/métodos , Células Receptoras Sensoriales/citología , Análisis de la Célula Individual/métodos , Nervio Vago/citología , Animales , Células Cultivadas , Ratones , Células Receptoras Sensoriales/metabolismo , Transcriptoma , Nervio Vago/metabolismo
13.
Stem Cell Reports ; 15(3): 557-565, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32857978

RESUMEN

The enteric nervous system (ENS) is derived primarily from the vagal neural crest, a migratory multipotent cell population emerging from the dorsal neural tube between somites 1 and 7. Defects in the development and function of the ENS cause a range of enteric neuropathies, including Hirschsprung disease. Little is known about the signals that specify early ENS progenitors, limiting progress in the generation of enteric neurons from human pluripotent stem cells (hPSCs) to provide tools for disease modeling and regenerative medicine for enteric neuropathies. We describe the efficient and accelerated generation of ENS progenitors from hPSCs, revealing that retinoic acid is critical for the acquisition of vagal axial identity and early ENS progenitor specification. These ENS progenitors generate enteric neurons in vitro and, following in vivo transplantation, achieved long-term colonization of the ENS in adult mice. Thus, hPSC-derived ENS progenitors may provide the basis for cell therapy for defects in the ENS.


Asunto(s)
Sistema Nervioso Entérico/citología , Cresta Neural/citología , Células-Madre Neurales/citología , Tretinoina/farmacología , Animales , Línea Celular , Humanos , Ratones , Células-Madre Neurales/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Nervio Vago/citología
14.
J Neurosci ; 40(37): 7054-7064, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32817248

RESUMEN

Leptin signaling within the nucleus of the solitary tract (NTS) contributes to the control of food intake, and injections of leptin into the NTS reduce meal size and increase the efficacy of vagus-mediated satiation signals. Leptin receptors (LepRs) are expressed by vagal afferents as well as by a population of NTS neurons. However, the electrophysiological properties of LepR-expressing NTS neurons have not been well characterized, and it is unclear how leptin might act on these neurons to reduce food intake. To address this question, we recorded from LepR-expressing neurons in horizontal brain slices containing the NTS from male and female LepR-Cre X Rosa-tdTomato mice. We found that the vast majority of NTS LepR neurons received monosynaptic innervation from vagal afferent fibers and LepR neurons exhibited large synaptic NMDA receptor (NMDAR)-mediated currents compared with non-LepR neurons. During high-frequency stimulation of vagal afferents, leptin increased the size of NMDAR-mediated currents, but not AMPAR-mediated currents. Leptin also increased the size of evoked EPSPs and the ability of low-intensity solitary tract stimulation to evoke action potentials in LepR neurons. These effects of leptin were blocked by bath applying a competitive NMDAR antagonist (DCPP-ene) or by an NMDAR channel blocker applied through the recording pipette (MK-801). Last, feeding studies using male rats demonstrate that intra-NTS injections of DCPP-ene attenuate reduction of overnight food intake following intra-NTS leptin injection. Our results suggest that leptin acts in the NTS to reduce food intake by increasing NMDAR-mediated currents, thus enhancing NTS sensitivity to vagal inputs.SIGNIFICANCE STATEMENT Leptin is a hormone that critically impacts food intake and energy homeostasis. The nucleus of the solitary tract (NTS) is activated by vagal afferents from the gastrointestinal tract, which promotes termination of a meal. Injection of leptin into the NTS inhibits food intake, while knockdown of leptin receptors (LepRs) in NTS neurons increases food intake. However, little was known about how leptin acts in the NTS neurons to inhibit food intake. We found that leptin increases the sensitivity of LepR-expressing neurons to vagal inputs by increasing NMDA receptor-mediated synaptic currents and that NTS NMDAR activation contributes to leptin-induced reduction of food intake. These findings suggest a novel mechanism by which leptin, acting in the NTS, could potentiate gastrointestinal satiation signals.


Asunto(s)
Potenciales Postsinápticos Excitadores , Leptina/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Núcleo Solitario/metabolismo , Nervio Vago/metabolismo , Animales , Maleato de Dizocilpina/farmacología , Ingestión de Alimentos , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/fisiología , Prolina/análogos & derivados , Prolina/farmacología , Piridinas/farmacología , Ratas , Núcleo Solitario/citología , Núcleo Solitario/fisiología , Sinapsis/metabolismo , Sinapsis/fisiología , Nervio Vago/citología , Nervio Vago/fisiología
15.
Tissue Cell ; 64: 101344, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32473709

RESUMEN

The jugular ganglion (JG) contains sensory neurons of the vagus nerve which innervate somatic and visceral structures in cranial and cervical regions. In this study, the number of sensory neurons in the human JG was investigated. And, the morphology of sensory neurons in the human JG and nodose ganglion (NG) was compared. The estimated number of JG neurons was 2721.8-9301.1 (average number of sensory neurons ±â€¯S.D. = 7975.1 ±â€¯3312.8). There was no significant difference in sizes of the neuronal cell body and nucleus within the JG (cell body, 1128.8 ±â€¯99.7 µâ€¯m2; nucleus, 127.7 ±â€¯20.8 µâ€¯m2) and NG (cell body, 963.8 ±â€¯225.7 µâ€¯m2; nucleus, 123.2 ±â€¯32.3 µâ€¯m2). These findings indicate that most of sensory neurons show the similar morphology in the JG and NG. Our immunohistochemical method also demonstrated the distribution of ion channels, neurotransmitter agents and calcium-binding proteins in the human JG. Numerous JG neurons were immunoreactive for transient receptor potential cation channel subfamily V member 1 (TRPV1, mean ±â€¯SD = 19.9 ±â€¯11.5 %) and calcitonin gene-related peptide (CGRP, 28.4 ±â€¯6.7 %). A moderate number of JG neurons contained TRPV2 (12.0 ±â€¯4.7 %), substance P (SP, 15.7 ±â€¯6.9 %) and secreted protein, acidic and rich in cysteine-like 1 (SPARCL1, 14.6 ±â€¯7.4 %). A few JG neurons had vesicular glutamate transporter 2 (VGLUT2, 5.6 ±â€¯2.9 %) and parvalbumin (PV, 2.3 ±â€¯1.4 %). SP- and TRPV2-containing JG neurons had mainly small and medium-sized cell bodies, respectively. TRPV1- and VGLUT2- containing JG neurons were small to medium-sized. CGRP- and SPARCL1-containing JG neurons were of various cell body sizes. Sensory neurons in the human JG were mostly free of vasoactive intestinal polypeptide (VIP), tyrosine hydroxylase (TH) and neuropeptide Y (NPY). In the external auditory canal skin, subepithelial nerve fibers contained TRPV1, TRPV2, SP, CGRP and VGLUT2. Perivascular nerve fibers also had TRPV1, TRPV2, SP, CGRP, VIP, NPY and TH. However, PV- and SPARCL1-containing nerve endings could not be seen in the external auditory canal. It is likely that sensory neurons in the human JG can transduce nociceptive and mechanoreceptive information from the external auditory canal. Theses neurons may be also associated with neurogenic inflammation in the external auditory canal and ear-cough reflex through the vagus nerve.


Asunto(s)
Ganglios , Neuropéptidos/metabolismo , Canales Catiónicos TRPV/metabolismo , Anciano , Autopsia , Péptido Relacionado con Gen de Calcitonina/metabolismo , Conducto Auditivo Externo/citología , Conducto Auditivo Externo/metabolismo , Femenino , Ganglios/citología , Ganglios/metabolismo , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neurotransmisores/metabolismo , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/metabolismo , Sustancia P/metabolismo , Nervio Vago/citología , Nervio Vago/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
16.
Transplant Proc ; 52(1): 61-66, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31837820

RESUMEN

BACKGROUND: Cardiac ganglia are rechargeable batteries of the heart. The essential role of cardiac ganglia on cardiac life expectancy has not been examined following brain death. The aim of this study was to determine cardiac ganglia numbers and neuron density following subarachnoid hemorrhage (SAH). METHODS: Twenty-five hybrid rabbits were grouped as control (n = 5), sham (n = 5), and SAH (n = 15). The SAH groups' animals were subjected to injections of lethal dose of 2.00 cc autologous blood into their cisterna magna until linear EEG was obtained. The hearts of all animals were extracted following intracardiac formalin injection and examined. Cardiac ganglia and normal/degenerated neuron densities of cardiac neurons were recorded. RESULTS: The mean volume of normal neuron density of ganglia was 6.980 ± 830/mm3, and the degenerated neuron density of ganglia was 3 ± 1/mm3 in the control group, 6134 ± 712/mm3; 23 ± 9/mm3 in the sham group, 3456 ± 589; 1161 ± 72/mm3 in the surviving group; and 1734 ± 341/mm3, 4259 ± 865/mm3 in the dead animals in the SAH group. The algebraic results of heart work capacity (Wh) were estimated as 1375 ± 210 Wh in the control group, 1036 ± 225 in the sham group, 800 ± 110 Wh in the surviving group, and < 100 ± 20 in the dead animals in the SAH group. Degenerated cardiac neuron density/Wh correlation is statistically meaningful between the dead in the SAH group versus the SAH-surviving, sham, and control groups (P < .0005). CONCLUSIONS: Normal cardiac ganglia numbers and/or cardiac ganglia neuron density may be related to cardiac survival following brain death after subarachnoid hemorrhage.


Asunto(s)
Corazón/inervación , Neuronas/citología , Hemorragia Subaracnoidea/complicaciones , Nervio Vago/citología , Animales , Muerte Encefálica/patología , Muerte , Modelos Animales de Enfermedad , Masculino , Conejos , Hemorragia Subaracnoidea/patología
17.
J Comp Neurol ; 528(5): 816-839, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31618460

RESUMEN

Vagal afferents innervating the small intestinal mucosa regulate feeding, gastrointestinal (GI) digestive, and immune functions. Their anatomical-functional characterization has been impeded by the inability to selectively label and manipulate them. Nav 1.8-Cre-tdTomato mice label 80% of nodose and dorsal root ganglia neurons. Here, the origin of these neuron's terminals and their distribution in the small intestinal mucosa were examined by quantitatively comparing tdTomato-labeled innervation in nonoperated (control), subdiaphragmatic vagotomy (VAGX), and sham-operated mice. Control mice exhibited a large proximal-to-distal decrease and a moderate mesentery-to-antimesentery decrease in villus innervation. VAGX reduced this innervation to a greater degree proximally (91-93%) than distally (65-72%), resulting in flat proximal-distal distributions. Therefore, estimates of vagal villus afferent distributions (control minus VAGX) paralleled control distributions, but were slightly reduced in magnitude. Compared with villus afferents, crypt innervation exhibited a muted proximal-to-distal decrease in control mice and a smaller loss after VAGX (45-48%). Sham-operated mice exhibited similar distributions of villus and crypt afferents as control mice, suggesting surgery did not contribute to the effects of VAGX. Most crypt and villus afferent terminals along the entire proximal-distal small intestinal axis had similar morphology to those previously reported in the proximal duodenum, but the density of terminal branches varied. Our findings suggest the majority of small intestinal mucosal innervation labeled in Nav 1.8-Cre-tdTomato mice is vagal in origin. Therefore, these mice will be valuable for studying vagal mucosal afferent morphology, interactions with other GI elements, plasticity, and function.


Asunto(s)
Mucosa Intestinal/inervación , Intestino Delgado/inervación , Neuronas Aferentes/citología , Nervio Vago/citología , Animales , Ratones , Ratones Endogámicos C57BL , Vagotomía
18.
Cell Rep ; 27(8): 2508-2523.e4, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31116992

RESUMEN

Sensory functions of the vagus nerve are critical for conscious perceptions and for monitoring visceral functions in the cardio-pulmonary and gastrointestinal systems. Here, we present a comprehensive identification, classification, and validation of the neuron types in the neural crest (jugular) and placode (nodose) derived vagal ganglia by single-cell RNA sequencing (scRNA-seq) transcriptomic analysis. Our results reveal major differences between neurons derived from different embryonic origins. Jugular neurons exhibit fundamental similarities to the somatosensory spinal neurons, including major types, such as C-low threshold mechanoreceptors (C-LTMRs), A-LTMRs, Aδ-nociceptors, and cold-, and mechano-heat C-nociceptors. In contrast, the nodose ganglion contains 18 distinct types dedicated to surveying the physiological state of the internal body. Our results reveal a vast diversity of vagal neuron types, including many previously unanticipated types, as well as proposed types that are consistent with chemoreceptors, nutrient detectors, baroreceptors, and stretch and volume mechanoreceptors of the respiratory, gastrointestinal, and cardiovascular systems.


Asunto(s)
Ganglio Nudoso/metabolismo , Nervio Vago/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Ganglio Nudoso/citología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcriptoma , Nervio Vago/citología
19.
Brain Res ; 1715: 94-105, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30914247

RESUMEN

Inflammation causes activation of nociceptive sensory nerves, resulting in debilitating sensations and reflexes. Inflammation also induces mitochondrial dysfunction through multiple mechanisms. Sensory nerve terminals are densely packed with mitochondria, suggesting that mitochondrial signaling may play a role in inflammation-induced nociception. We have previously shown that agents that induce mitochondrial dysfunction, such as antimycin A, activate a subset of nociceptive vagal sensory nerves that express transient receptor potential (TRP) channels ankyrin 1 (A1) and vanilloid 1 (V1). However, the mechanisms underlying these responses are incompletely understood. Here, we studied the contribution of TRPA1, TRPV1 and reactive oxygen species (ROS) to antimycin A-induced vagal sensory nerve activation in dissociated neurons and at the sensory terminals of bronchopulmonary C-fibers. Nociceptive neurons were defined chemically and genetically. Antimycin A-evoked activation of vagal nociceptors in a Fura2 Ca2+ assay correlated with TRPV1 responses compared to TRPA1 responses. Nociceptor activation was dependent on both TRP channels, with TRPV1 predominating in a majority of responding nociceptors and TRPA1 predominating only in nociceptors with the greatest responses. Surprisingly, both TRPA1 and TRPV1 were activated by H2O2 when expressed in HEK293. Nevertheless, targeting ROS had no effect of antimycin A-evoked TRPV1 activation in either HEK293 or vagal neurons. In contrast, targeting ROS inhibited antimycin A-evoked TRPA1 activation in HEK293, vagal neurons and bronchopulmonary C-fibers, and a ROS-insensitive TRPA1 mutant was completely insensitive to antimycin A. We therefore conclude that mitochondrial dysfunction activates vagal nociceptors by ROS-dependent (TRPA1) and ROS-independent (TRPV1) mechanisms.


Asunto(s)
Antimicina A/farmacología , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canal Catiónico TRPA1/metabolismo , Canales Catiónicos TRPV/metabolismo , Nervio Vago/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Calcio/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Nociceptores/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Nervio Vago/citología , Nervio Vago/efectos de los fármacos
20.
Endocrinology ; 160(5): 1307-1322, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30907928

RESUMEN

Leptin has been proposed to modulate viscerosensory information directly at the level of vagal afferents. In support of this view, broad expression for the leptin receptor (Lepr) has previously been reported in vagal afferents. However, the exact identity and distribution of leptin-sensitive vagal afferents has not been elucidated. Using quantitative PCR, we found that the whole mouse nodose ganglion was predominantly enriched in the short form of Lepr, rather than its long form. Consistent with this observation, the acute administration of leptin did not stimulate JAK-STAT signaling in the nodose ganglion. Using chromogenic in situ hybridization in wild-type mice and several reporter mouse models, we demonstrated that Lepr mRNA was restricted to nonneuronal cells in the epineurium and parenchyma of the nodose ganglion and a subset of vagal afferents, which accounted for only 3% of all neuronal profiles. Double labeling studies further established that Lepr-expressing vagal afferents were Nav1.8-negative fibers that did not supply the peritoneal cavity. Finally, double chromogenic in situ hybridization revealed that many Lepr-expressing neurons coexpressed the angiotensin 1a receptor (At1ar), which is a gene expressed in baroreceptors. Taken together, our data challenge the commonly held view that Lepr is broadly expressed in vagal afferents. Instead, our data suggest that leptin may exert a previously unrecognized role, mainly via its short form, as a direct modulator of a very small group of At1ar-positive vagal fibers.


Asunto(s)
Expresión Génica/efectos de los fármacos , Leptina/farmacología , Neuronas/efectos de los fármacos , Ganglio Nudoso/efectos de los fármacos , Receptores de Leptina/genética , Animales , Células Cultivadas , Hibridación in Situ/métodos , Leptina/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.8/genética , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Neuronas/metabolismo , Neuronas Aferentes/metabolismo , Ganglio Nudoso/citología , Ganglio Nudoso/metabolismo , Receptores de Leptina/metabolismo , Nervio Vago/citología , Nervio Vago/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...