Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 628(8009): 826-834, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38538787

RESUMEN

Empirical evidence suggests that heat exposure reduces food intake. However, the neurocircuit architecture and the signalling mechanisms that form an associative interface between sensory and metabolic modalities remain unknown, despite primary thermoceptive neurons in the pontine parabrachial nucleus becoming well characterized1. Tanycytes are a specialized cell type along the wall of the third ventricle2 that bidirectionally transport hormones and signalling molecules between the brain's parenchyma and ventricular system3-8. Here we show that tanycytes are activated upon acute thermal challenge and are necessary to reduce food intake afterwards. Virus-mediated gene manipulation and circuit mapping showed that thermosensing glutamatergic neurons of the parabrachial nucleus innervate tanycytes either directly or through second-order hypothalamic neurons. Heat-dependent Fos expression in tanycytes suggested their ability to produce signalling molecules, including vascular endothelial growth factor A (VEGFA). Instead of discharging VEGFA into the cerebrospinal fluid for a systemic effect, VEGFA was released along the parenchymal processes of tanycytes in the arcuate nucleus. VEGFA then increased the spike threshold of Flt1-expressing dopamine and agouti-related peptide (Agrp)-containing neurons, thus priming net anorexigenic output. Indeed, both acute heat and the chemogenetic activation of glutamatergic parabrachial neurons at thermoneutrality reduced food intake for hours, in a manner that is sensitive to both Vegfa loss-of-function and blockage of vesicle-associated membrane protein 2 (VAMP2)-dependent exocytosis from tanycytes. Overall, we define a multimodal neurocircuit in which tanycytes link parabrachial sensory relay to the long-term enforcement of a metabolic code.


Asunto(s)
Tronco Encefálico , Células Ependimogliales , Conducta Alimentaria , Calor , Hipotálamo , Vías Nerviosas , Neuronas , Animales , Femenino , Masculino , Ratones , Proteína Relacionada con Agouti/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/citología , Tronco Encefálico/citología , Tronco Encefálico/fisiología , Dopamina/metabolismo , Ingestión de Alimentos/fisiología , Células Ependimogliales/citología , Células Ependimogliales/fisiología , Conducta Alimentaria/fisiología , Ácido Glutámico/metabolismo , Hipotálamo/citología , Hipotálamo/fisiología , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/fisiología , Sensación Térmica/fisiología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/líquido cefalorraquídeo , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Neurosci Lett ; 750: 135794, 2021 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-33667599

RESUMEN

A subset of glutamatergic interneurons in the neonatal spinal superficial dorsal horn (SDH) exhibits intrinsic burst-firing (i.e. 'pacemaker' activity), which is tightly regulated by persistent, voltage-gated Na+ channels and classic inward-rectifying K+ (Kir2) channels and downregulated over the course of postnatal development. Ascending lamina I projection neurons targeting the parabrachial nucleus (PB) or periaqueductal gray (PAG) can also display pacemaker activity during early life. However, the degree to which the ionic mechanisms driving pacemaker activity are conserved across different cell types in the spinal dorsal horn, as well as whether the intrinsic bursting is restricted to newborn projection neurons, remains to be elucidated. Using in vitro patch clamp recordings from identified lamina I spinoparabrachial neurons in rat spinal cord slices, here we demonstrate that adolescent projection neurons retain their ability to generate pacemaker activity. In contrast to previous findings in lamina I interneurons, pacemaker projection neurons possessed higher membrane capacitance, lower membrane resistance, and a greater Kir-mediated conductance compared to adjacent spinoparabrachial neurons that lacked intrinsic burst-firing. Nonetheless, as previously seen in interneurons, the bath application of riluzole to block persistent Na+ channels significantly dampened pacemaker activity in projection neurons. Collectively, these results suggest that intrinsic burst-firing in the developing dorsal horn can be generated by multiple combinations of ionic conductances, and highlight the need for further investigation into the mechanisms governing pacemaker activity within the major output neurons of the SDH network.


Asunto(s)
Potenciales de Acción , Neuronas/fisiología , Núcleos Parabraquiales/fisiología , Asta Dorsal de la Médula Espinal/fisiología , Animales , Relojes Biológicos , Femenino , Masculino , Neuronas/metabolismo , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/crecimiento & desarrollo , Canales de Potasio de Rectificación Interna/metabolismo , Ratas , Ratas Sprague-Dawley , Canales de Sodio/metabolismo , Asta Dorsal de la Médula Espinal/citología , Asta Dorsal de la Médula Espinal/crecimiento & desarrollo
3.
Nat Commun ; 11(1): 5974, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33239627

RESUMEN

The lateral parabrachial nucleus (LPBN) is known to relay noxious information to the amygdala for processing affective responses. However, it is unclear whether the LPBN actively processes neuropathic pain characterized by persistent hyperalgesia with aversive emotional responses. Here we report that neuropathic pain-like hypersensitivity induced by common peroneal nerve (CPN) ligation increases nociceptive stimulation-induced responses in glutamatergic LPBN neurons. Optogenetic activation of GABAergic LPBN neurons does not affect basal nociception, but alleviates neuropathic pain-like behavior. Optogenetic activation of glutamatergic or inhibition of GABAergic LPBN neurons induces neuropathic pain-like behavior in naïve mice. Inhibition of glutamatergic LPBN neurons alleviates both basal nociception and neuropathic pain-like hypersensitivity. Repetitive pharmacogenetic activation of glutamatergic or GABAergic LPBN neurons respectively mimics or prevents the development of CPN ligation-induced neuropathic pain-like hypersensitivity. These findings indicate that a delicate balance between excitatory and inhibitory LPBN neuronal activity governs the development and maintenance of neuropathic pain.


Asunto(s)
Hiperalgesia/fisiopatología , Neuralgia/fisiopatología , Neuronas/metabolismo , Nocicepción/fisiología , Núcleos Parabraquiales/fisiología , Animales , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Agonistas del GABA/farmacología , Ácido Glutámico/metabolismo , Humanos , Hiperalgesia/etiología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Ratones , Ratones Transgénicos , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Neuralgia/etiología , Neuronas/efectos de los fármacos , Optogenética , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/efectos de los fármacos , Nervio Peroneo/lesiones , Nervio Peroneo/fisiopatología , Técnicas Estereotáxicas , Ácido gamma-Aminobutírico/metabolismo
4.
Elife ; 92020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32856589

RESUMEN

Parabrachial CGRP neurons receive diverse threat-related signals and contribute to multiple phases of adaptive threat responses in mice, with their inactivation attenuating both unconditioned behavioral responses to somatic pain and fear-memory formation. Because CGRPPBN neurons respond broadly to multi-modal threats, it remains unknown how these distinct adaptive processes are individually engaged. We show that while three partially separable subsets of CGRPPBN neurons broadly collateralize to their respective downstream partners, individual projections accomplish distinct functions: hypothalamic and extended amygdalar projections elicit assorted unconditioned threat responses including autonomic arousal, anxiety, and freezing behavior, while thalamic and basal forebrain projections generate freezing behavior and, unexpectedly, contribute to associative fear learning. Moreover, the unconditioned responses generated by individual projections are complementary, with simultaneous activation of multiple sites driving profound freezing behavior and bradycardia that are not elicited by any individual projection. This semi-parallel, scalable connectivity schema likely contributes to flexible control of threat responses in unpredictable environments.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Aprendizaje/fisiología , Núcleos Parabraquiales/citología , Animales , Conducta Animal/fisiología , Femenino , Masculino , Ratones , Neuronas/citología , Neuronas/metabolismo
5.
Neuron ; 107(5): 909-923.e6, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32649865

RESUMEN

The parabrachial nucleus (PBN) is one of the major targets of spinal projection neurons and plays important roles in pain. However, the architecture of the spinoparabrachial pathway underlying its functional role in nociceptive information processing remains elusive. Here, we report that the PBN directly relays nociceptive signals from the spinal cord to the intralaminar thalamic nuclei (ILN). We demonstrate that the spinal cord connects with the PBN in a bilateral manner and that the ipsilateral spinoparabrachial pathway is critical for nocifensive behavior. We identify Tacr1-expressing neurons as the major neuronal subtype in the PBN that receives direct spinal input and show that these neurons are critical for processing nociceptive information. Furthermore, PBN neurons receiving spinal input form functional monosynaptic excitatory connections with neurons in the ILN, but not the amygdala. Together, our results delineate the neural circuit underlying nocifensive behavior, providing crucial insight into the circuit mechanism underlying nociceptive information processing.


Asunto(s)
Vías Aferentes , Lateralidad Funcional/fisiología , Núcleos Talámicos Intralaminares , Nocicepción/fisiología , Núcleos Parabraquiales , Vías Aferentes/citología , Vías Aferentes/fisiología , Amígdala del Cerebelo , Animales , Núcleos Talámicos Intralaminares/citología , Núcleos Talámicos Intralaminares/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/fisiología , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/fisiología , Médula Espinal/citología , Médula Espinal/fisiología
6.
Cereb Cortex ; 30(9): 4811-4833, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32383444

RESUMEN

The parabrachial nucleus (PB) in the upper brain stem tegmentum includes several neuronal subpopulations with a wide variety of connections and functions. A subpopulation of PB neurons projects axons directly to the cerebral cortex, and limbic areas of the cerebral cortex send a return projection directly to the PB. We used retrograde and Cre-dependent anterograde tracing to identify genetic markers and characterize this PB-cortical interconnectivity in mice. Cortical projections originate from glutamatergic PB neurons that contain Lmx1b (81%), estrogen receptor alpha (26%), and Satb2 (20%), plus mRNA for the neuropeptides cholecystokinin (Cck, 48%) and calcitonin gene-related peptide (Calca, 13%), with minimal contribution from FoxP2+ PB neurons (2%). Axons from the PB produce an extensive terminal field in an unmyelinated region of the insular cortex, extending caudally into the entorhinal cortex, and arcing rostrally through the dorsolateral prefrontal cortex, with a secondary terminal field in the medial prefrontal cortex. In return, layer 5 neurons in the insular cortex and other prefrontal areas, along with a dense cluster of cells dorsal to the claustrum, send a descending projection to subregions of the PB that contain cortically projecting neurons. This information forms the neuroanatomical basis for testing PB-cortical interconnectivity in arousal and interoception.


Asunto(s)
Corteza Cerebral/citología , Vías Nerviosas/citología , Núcleos Parabraquiales/citología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
7.
Nat Commun ; 11(1): 1729, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32265438

RESUMEN

The TrkB receptor is critical for the control of energy balance, as mutations in its gene (NTRK2) lead to hyperphagia and severe obesity. The main neural substrate mediating the appetite-suppressing activity of TrkB, however, remains unknown. Here, we demonstrate that selective Ntrk2 deletion within paraventricular hypothalamus (PVH) leads to severe hyperphagic obesity. Furthermore, chemogenetic activation or inhibition of TrkB-expressing PVH (PVHTrkB) neurons suppresses or increases food intake, respectively. PVHTrkB neurons project to multiple brain regions, including ventromedial hypothalamus (VMH) and lateral parabrachial nucleus (LPBN). We find that PVHTrkB neurons projecting to LPBN are distinct from those to VMH, yet Ntrk2 deletion in PVH neurons projecting to either VMH or LPBN results in hyperphagia and obesity. Additionally, TrkB activation with BDNF increases firing of these PVH neurons. Therefore, TrkB signaling is a key regulator of a previously uncharacterized neuronal population within the PVH that impinges upon multiple circuits to govern appetite.


Asunto(s)
Hiperfagia/metabolismo , Glicoproteínas de Membrana/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Apetito/genética , Conducta Alimentaria/fisiología , Femenino , Hiperfagia/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/genética , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/fisiopatología , Proteínas Tirosina Quinasas/genética , Núcleo Hipotalámico Ventromedial/citología , Núcleo Hipotalámico Ventromedial/metabolismo
8.
J Neurosci ; 40(3): 632-647, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31744862

RESUMEN

The central nucleus of the amygdala plays a significant role in alcohol use and other affective disorders; however, the genetically-defined neuronal subtypes and projections that govern these behaviors are not well known. Here we show that neurotensin neurons in the central nucleus of the amygdala of male mice are activated by in vivo ethanol consumption and that genetic ablation of these neurons decreases ethanol consumption and preference in non-ethanol-dependent animals. This ablation did not impact preference for sucrose, saccharin, or quinine. We found that the most robust projection of the central amygdala neurotensin neurons was to the parabrachial nucleus, a brain region known to be important in feeding behaviors, conditioned taste aversion, and alarm. Optogenetic stimulation of projections from these neurons to the parabrachial nucleus is reinforcing, and increases ethanol drinking as well as consumption of sucrose and saccharin solutions. These data suggest that this central amygdala to parabrachial nucleus projection influences the expression of reward-related phenotypes and is a novel circuit promoting consumption of ethanol and palatable fluids.SIGNIFICANCE STATEMENT Alcohol use disorder (AUD) is a major health burden worldwide. Although ethanol consumption is required for the development of AUD, much remains unknown regarding the underlying neural circuits that govern initial ethanol intake. Here we show that ablation of a population of neurotensin-expressing neurons in the central amygdala decreases intake of and preference for ethanol in non-dependent animals, whereas the projection of these neurons to the parabrachial nucleus promotes consumption of ethanol as well as other palatable fluids.


Asunto(s)
Consumo de Bebidas Alcohólicas/psicología , Núcleo Amigdalino Central/fisiología , Preferencias Alimentarias/fisiología , Neuronas/fisiología , Neurotensina/fisiología , Animales , Ansiedad/psicología , Núcleo Amigdalino Central/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Optogenética , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/fisiología , Técnicas de Placa-Clamp , Recompensa , Edulcorantes , Gusto/fisiología
9.
eNeuro ; 6(6)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31662323

RESUMEN

Food consumption is necessary for organisms to maintain metabolic homeostasis. Both extrinsic and intrinsic processes, relayed via intricate neural circuitry, orchestrate the initiation and termination of food intake. More specifically, there are functionally distinct neural circuits that mediate either homeostatic or hedonic suppression of feeding. Notably, being satiated is a positive feeling whereas food aversion is a negative feeling. While significant progress has been made toward elucidating neural circuitry underlying aversive appetite suppression in mice, the circuitry underlying homeostatic satiety is not fully understood. The lateral parabrachial nucleus (PBL) is known as a node that regulates various sensory and visceral processes. Here, we identified and selectively labeled neurons in the caudal lateral region of PBL (PBcl) that are activated by consumption of condensed milk, chocolate Ensure, or peanut butter, which we refer to as PBcl-palatable-food activated neurons (PANs). Specific optogenetic activation of PANs induced positive place preference but decreased the consumption of high-caloric foods such as condensed milk, whereas silencing these cells significantly increased condensed milk consumption in feeding assays. Thus, the PBcl PANs revealed here represent a novel neural substrate regulating caloric-sufficiency mediated satiation.


Asunto(s)
Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Neuronas/citología , Núcleos Parabraquiales/citología , Saciedad/fisiología , Animales , Femenino , Alimentos , Masculino , Ratones , Vías Nerviosas/fisiología , Neuronas/fisiología , Núcleos Parabraquiales/fisiología
10.
Cell Rep ; 28(6): 1429-1438.e4, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31390558

RESUMEN

The dorsal horn of the spinal cord is the first integration site of somatosensory inputs from the periphery. In the superficial layers of the dorsal horn, nociceptive inputs are processed by a complex network of excitatory and inhibitory interneurons whose function and connectivity remain poorly understood. We examined the role of calretinin-expressing interneurons (CR neurons) in such processing and show that they receive direct inputs from nociceptive fibers and polysynaptic inputs from touch-sensitive Aß fibers. Their activation by chemogenetic or optogenetic stimulation produces mechanical allodynia and nocifensive responses. Furthermore, they monosynaptically engage spinoparabrachial (SPb) neurons in lamina I, suggesting CR neurons modulate one of the major ascending pain pathways of the dorsal horn. In conclusion, we propose a neuronal pathway in which CR neurons are positioned at the junction between nociceptive and innocuous circuits and directly control SPb neurons in lamina I.


Asunto(s)
Calbindina 2/fisiología , Interneuronas/fisiología , Células del Asta Posterior/fisiología , Asta Dorsal de la Médula Espinal/citología , Animales , Capsaicina , Hiperalgesia , Masculino , Memoria , Ratones Endogámicos C57BL , Vías Nerviosas , Nocicepción/fisiología , Optogenética , Núcleos Parabraquiales/citología , Reclutamiento Neurofisiológico
11.
J Therm Biol ; 83: 87-94, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31331530

RESUMEN

Both warm- and cold-sensitive neurons are found in the lateral parabrachial nucleus (LPB), a crucial relay for skin temperature information from the spinal cord to the preoptic area. The aims of this study were to investigate the electrophysiological properties of temperature-sensitive and -insensitive neurons in brain slices, and elucidate the basic mechanisms underlying the thermosensitivity of rat LPB neurons. In warm-sensitive neurons, temperature exerted no significant effects on resting membrane potential (RMP), threshold potential, and amplitude of the afterhyperpolarizing potential. However, warming significantly increased the prepotential rates of depolarization and the inactivation rates of potassium A current (IA) in warm-sensitive neurons, which in turn shortened their interspike interval and elevated the firing rate. In contrast, temperature had no significant effects on the depolarizing prepotentials and inactivation rate of IA in temperature-insensitive neurons. Besides, in cold-sensitive neurons, cooling and warming produced membrane depolarization and hyperpolarization, respectively, and there was a strong correlation between firing rate and membrane potential thermosensitivity. Nevertheless, temperature exhibited no significant effect on the depolarizing prepotential of cold-sensitive neurons. These results suggest that LPB neuronal warm sensitivity may reside in the temperature-dependent prepotentials and IA, while neuronal cold sensitivity might be mainly due to heat-induced changes in RMP.


Asunto(s)
Frío , Potenciales de la Membrana , Neuronas/fisiología , Núcleos Parabraquiales/fisiología , Animales , Masculino , Núcleos Parabraquiales/citología , Potasio/metabolismo , Ratas , Ratas Sprague-Dawley
12.
Neuron ; 102(3): 653-667.e6, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-30879785

RESUMEN

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


Asunto(s)
Conducta Alimentaria/fisiología , Neuronas/citología , Obesidad/fisiopatología , Núcleo Hipotalámico Paraventricular/citología , Respuesta de Saciedad/fisiología , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Metabolismo Energético , Encefalinas/metabolismo , Locus Coeruleus/citología , Locus Coeruleus/metabolismo , Locus Coeruleus/fisiología , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/fisiología , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/fisiología , Precursores de Proteínas/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Proteínas Represoras/metabolismo
13.
Neuroendocrinology ; 109(4): 310-321, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30889580

RESUMEN

Interleukin (IL)-6 in the hypothalamus and hindbrain is an important downstream mediator of suppression of body weight and food intake by glucagon-like peptide-1 (GLP-1) receptor stimulation. CNS GLP-1 is produced almost exclusively in prepro-glucagon neurons in the nucleus of the solitary tract. These neurons innervate energy balance-regulating areas, such as the external lateral parabrachial nucleus (PBNel); essential for induction of anorexia. Using a validated novel IL-6-reporter mouse strain, we investigated the interactions in PBNel between GLP-1, IL-6, and calcitonin gene-related peptide (CGRP, a well-known mediator of anorexia). We show that PBNel GLP-1R-containing cells highly (to about 80%) overlap with IL-6-containing cells on both protein and mRNA level. Intraperitoneal administration of a GLP-1 analogue exendin-4 to mice increased the proportion of IL-6-containing cells in PBNel 3-fold, while there was no effect in the rest of the lateral parabrachial nucleus. In contrast, injections of an anorexigenic peptide growth and differentiation factor 15 (GDF15) markedly increased the proportion of CGRP-containing cells, while IL-6-containing cells were not affected. In summary, GLP-1R are found on IL-6-producing cells in PBNel, and GLP-1R stimulation leads to an increase in the proportion of cells with IL-6-reporter fluorescence, supporting IL-6 mediation of GLP-1 effects on energy balance.


Asunto(s)
Proteínas Portadoras/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Interleucina-6/biosíntesis , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/metabolismo , Animales , Regulación del Apetito , Péptido Relacionado con Gen de Calcitonina/biosíntesis , Proteínas Portadoras/agonistas , Metabolismo Energético/efectos de los fármacos , Exenatida/administración & dosificación , Exenatida/farmacología , Genes Reporteros/efectos de los fármacos , Inmunohistoquímica , Inyecciones Intraperitoneales , Péptidos y Proteínas de Señalización Intracelular , Ratones , Núcleos Parabraquiales/efectos de los fármacos
14.
J Neurosci ; 39(9): 1631-1648, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30606758

RESUMEN

Taste and somatosensation both mediate protective behaviors. Bitter taste guides avoidance of ingestion of toxins while pain sensations, such as noxious heat, signal adverse conditions to ward off harm. Although brain pathways for taste and somatosensation are typically studied independently, prior data suggest that they intersect, potentially reflecting their common protective role. To investigate this, we applied electrophysiologic and optogenetic techniques in anesthetized mice of both sexes to evaluate relationships between oral somatosensory and taste activity in the parabrachial nucleus (PbN), implicated for roles in gustation and pain. Spikes were recorded from taste-active PbN neurons tested with oral delivery of thermal and chemesthetic stimuli, including agonists of nocisensitive transient receptor potential (TRP) ion channels on somatosensory fibers. Gustatory neurons were also tested to follow electrical pulse stimulation of an oral somatosensory region of the spinal trigeminal subnucleus caudalis (Vc), which projects to the PbN. Neurons composed classic taste groups, including sodium, electrolyte, appetitive, or bitter cells. Across groups, most neurons spiked to Vc pulse stimulation, implying that trigeminal projections reach PbN gustatory neurons. Among such cells, a subpopulation responsive to the bitter taste stimuli quinine and cycloheximide, and aversive concentrations of sodium, cofired to agonists of nocisensitive TRP channels, including capsaicin, mustard oil, and noxious heat. Such neurons populated the lateral PbN. Further, nociceptive activity in PbN bitter taste neurons was suppressed during optogenetic-assisted inhibition of the Vc, implying convergent trigeminal input contributed to such activity. Our results reveal a novel role for PbN gustatory cells in cross-system signaling related to protection.SIGNIFICANCE STATEMENT Prior data suggest that gustatory and trigeminal neural pathways intersect and overlap in the parabrachial area. However, no study has directly examined such overlap and why it may exist. Here we found that parabrachial gustatory neurons can receive afferent projections from trigeminal nuclei and fire to oral nociceptive stimuli that excite somatosensory receptors and fibers. Activation to aversive nociceptive stimuli in gustatory cells was associated with responding to behaviorally avoided bitter tastants. We were further able to show that silencing trigeminal projections inhibited nociceptive activity in parabrachial bitter taste neurons. Our results imply that in the parabrachial area, there is predictable overlap between taste and somatosensory processing related to protective coding and that classically defined taste neurons contribute to this process.


Asunto(s)
Nocicepción , Núcleos Parabraquiales/fisiología , Células Receptoras Sensoriales/metabolismo , Percepción del Gusto , Potenciales de Acción , Animales , Capsaicina/farmacología , Cicloheximida/farmacología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Planta de la Mostaza , Núcleos Parabraquiales/citología , Aceites de Plantas/farmacología , Quinina/farmacología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/fisiología , Gusto , Canales de Potencial de Receptor Transitorio/metabolismo
15.
Nature ; 565(7737): 86-90, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30532001

RESUMEN

Animals and humans display two types of response to noxious stimuli. The first includes reflexive defensive responses that prevent or limit injury; a well-known example of these responses is the quick withdrawal of one's hand upon touching a hot object. When the first-line response fails to prevent tissue damage (for example, a finger is burnt), the resulting pain invokes a second-line coping response-such as licking the injured area to soothe suffering. However, the underlying neural circuits that drive these two strings of behaviour remain poorly understood. Here we show in mice that spinal neurons marked by coexpression of TAC1Cre and LBX1Flpo drive coping responses associated with pain. Ablation of these spinal neurons led to the loss of both persistent licking and conditioned aversion evoked by stimuli (including skin pinching and burn injury) that-in humans-produce sustained pain, without affecting any of the reflexive defensive reactions that we tested. This selective indifference to sustained pain resembles the phenotype seen in humans with lesions of medial thalamic nuclei1-3. Consistently, spinal TAC1-lineage neurons are connected to medial thalamic nuclei by direct projections and via indirect routes through the superior lateral parabrachial nuclei. Furthermore, the anatomical and functional segregation observed at the spinal level also applies to primary sensory neurons. For example, in response to noxious mechanical stimuli, MRGPRD- and TRPV1-positive nociceptors are required to elicit reflexive and coping responses, respectively. Our study therefore reveals a fundamental subdivision within the cutaneous somatosensory system, and challenges the validity of using reflexive defensive responses to measure sustained pain.


Asunto(s)
Adaptación Psicológica/fisiología , Dolor Crónico/fisiopatología , Dolor Crónico/psicología , Vías Nerviosas/fisiología , Animales , Reacción de Prevención , Condicionamiento Clásico , Femenino , Humanos , Masculino , Núcleo Talámico Mediodorsal/citología , Núcleo Talámico Mediodorsal/fisiología , Ratones , Neuronas Aferentes/fisiología , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/fisiología , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Canales Catiónicos TRPV/metabolismo , Taquicininas/genética , Taquicininas/metabolismo
16.
Anat Rec (Hoboken) ; 302(7): 1178-1186, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30332715

RESUMEN

Lateral parabrachial nucleus (LPB) is a critical region in the integration and transmission of peripheral nociceptive information. The parabrachio-amygdaloid (P-Amy) pathway and parabrachio-ventral tegmental area (P-VTA) pathway is thought to be significant in regulation of pain-related negative emotions. In present study, retrograde tract tracers Fluoro-gold (FG) and tetramethylrhodramine-dextran (TMR) were stereotaxically injected into the right central amygdaloid nucleus (CeA) and right VTA, respectively. Then, part of these rats were performed with the spare nerve injury (SNI) in the controlateral side of FG and TMR injection. Afterwards, double- or triple-immunofluorescent histochemistry was used to examine FG/TMR double- and FG/TMR/FOS or FG/TMR/CGRP triple-labeled neurons in the LPB. The results showed that all of FG, TMR single- and FG/TMR double-labeled neurons were distributed in the LPB bilaterally with an ipsilateral predominance. The proportion of FG/TMR double-labeled neurons to the total number of FG- and TMR-labeled neurons was 10.78% and 13.07%, respectively. Nearly all of the FG/TMR double-labeled neurons (92.67%) showed calcitonin gene-related peptide (CGRP) immunopositive. On the other hand, in the SNI rats, about 89.49% and 77.87% of FG- and TMR-labeled neurons were FG/FOS- and TMR/FOS-positive neurons; about 93.33% of the FG/TMR double-labeled neurons were FOS-LI. Our results suggest that the part of CGRP immunopositive neurons in the LPB send projection fibers to both the CeA and VTA by the way of axon collaterals, which are activated by the nociceptive stimulation in the SNI condition, and may play an important role in the transmission of peripheral nociceptive information. Anat Rec, 302:1178-1186, 2019. © 2018 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.


Asunto(s)
Núcleo Amigdalino Central/fisiología , Neuralgia/fisiopatología , Nocicepción/fisiología , Núcleos Parabraquiales/fisiología , Área Tegmental Ventral/fisiología , Animales , Núcleo Amigdalino Central/citología , Modelos Animales de Enfermedad , Humanos , Masculino , Vías Nerviosas/fisiología , Neuralgia/etiología , Neuronas/fisiología , Núcleos Parabraquiales/citología , Ratas , Ratas Sprague-Dawley , Nervio Ciático/lesiones , Técnicas Estereotáxicas , Área Tegmental Ventral/citología
17.
Pain ; 159(9): 1719-1730, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29746349

RESUMEN

Spinal projection neurons convey nociceptive signals to multiple brain regions including the parabrachial (PB) nucleus, which contributes to the emotional valence of pain perception. Despite the clear importance of projection neurons to pain processing, our understanding of the factors that shape their intrinsic membrane excitability remains limited. Here, we investigate a potential role for the Na leak channel NALCN in regulating the activity of spino-PB neurons in the developing rodent. Pharmacological reduction of NALCN current (INALCN), or the genetic deletion of NALCN channels, significantly reduced the intrinsic excitability of lamina I spino-PB neurons. In addition, substance P (SP) activated INALCN in ascending projection neurons through downstream Src kinase signaling, and the knockout of NALCN prevented SP-evoked action potential discharge in this neuronal population. These results identify, for the first time, NALCN as a strong regulator of neuronal activity within central pain circuits and also elucidate an additional ionic mechanism by which SP can modulate spinal nociceptive processing. Collectively, these findings indicate that the level of NALCN conductance within spino-PB neurons tightly governs ascending nociceptive transmission to the brain and thereby potentially influences pain perception.


Asunto(s)
Potenciales de Acción/fisiología , Canales Iónicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Núcleos Parabraquiales/metabolismo , Células del Asta Posterior/metabolismo , Médula Espinal/metabolismo , Animales , Animales Recién Nacidos , Canales Iónicos/genética , Proteínas de la Membrana , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Núcleos Parabraquiales/citología , Células del Asta Posterior/citología , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología
18.
Trends Neurosci ; 41(5): 280-293, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29703377

RESUMEN

The parabrachial nucleus (PBN), which is located in the pons and is dissected by one of the major cerebellar output tracks, is known to relay sensory information (visceral malaise, taste, temperature, pain, itch) to forebrain structures including the thalamus, hypothalamus, and extended amygdala. The availability of mouse lines expressing Cre recombinase selectively in subsets of PBN neurons and viruses for Cre-dependent gene expression is beginning to reveal the connectivity and functions of PBN component neurons. This review focuses on PBN neurons expressing calcitonin gene-related peptide (CGRPPBN) that play a major role in regulating appetite and transmitting real or potential threat signals to the extended amygdala. The functions of other specific PBN neuronal populations are also discussed. This review aims to encourage investigation of the numerous unanswered questions that are becoming accessible.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Neuronas/citología , Neuronas/fisiología , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/fisiología , Animales , Humanos
19.
Nature ; 555(7698): 617-622, 2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29562230

RESUMEN

Animals must respond to various threats to survive. Neurons that express calcitonin gene-related peptide in the parabrachial nucleus (CGRPPBN neurons) relay sensory signals that contribute to satiation and pain-induced fear behaviour, but it is unclear how they encode these distinct processes. Here, by recording calcium transients in vivo from individual neurons in mice, we show that most CGRPPBN neurons are activated by noxious cutaneous (shock, heat, itch) and visceral stimuli (lipopolysaccharide). The same neurons are inhibited during feeding, but become activated during satiation, consistent with evidence that CGRPPBN neurons prevent overeating. CGRPPBN neurons are also activated during consumption of novel foods or by an auditory cue that has previously been paired with electrical footshocks. Correspondingly, silencing of CGRPPBN neurons attenuates the expression of food neophobia and conditioned fear responses. Therefore, in addition to transducing primary sensory danger signals, CGRPPBN neurons promote affective-behavioural states that limit harm in response to potential threats.


Asunto(s)
Reacción de Prevención/fisiología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Miedo/fisiología , Neuronas/metabolismo , Núcleos Parabraquiales/citología , Animales , Señalización del Calcio , Condicionamiento Clásico/fisiología , Dieta Alta en Grasa , Electrochoque , Miedo/psicología , Respuesta al Choque Térmico , Lipopolisacáridos/farmacología , Masculino , Recuerdo Mental/fisiología , Ratones , Dolor/psicología , Núcleos Parabraquiales/fisiología , Prurito , Respuesta de Saciedad/fisiología
20.
Artículo en Inglés | MEDLINE | ID: mdl-29467628

RESUMEN

Cerebellar malformations cause changes to the sleep-wake cycle, resulting in sleep disturbance. However, it is unclear how the cerebellum contributes to the sleep-wake cycle. To examine the neural connections between the cerebellum and the nuclei involved in the sleep-wake cycle, we investigated the axonal projections of Purkinje cells in the mouse posterior vermis by using an adeno-associated virus (AAV) vector (serotype rh10) as an anterograde tracer. When an AAV vector expressing humanized renilla green fluorescent protein was injected into the cerebellar lobule IX, hrGFP and synaptophysin double-positive axonal terminals were observed in the region of medial parabrachial nucleus (MPB). The MPB is involved in the phase transition from rapid eye movement (REM) sleep to Non-REM sleep and vice versa, and the cardiovascular and respiratory responses. The hrGFP-positive axons from lobule IX went through the ventral spinocerebellar tract and finally reached the MPB. By contrast, when the AAV vector was injected into cerebellar lobule VI, no hrGFP-positive axons were observed in the MPB. To examine neurons projecting to the MPB, we unilaterally injected Fast Blue and AAV vector (retrograde serotype, rAAV2-retro) as retrograde tracers into the MPB. The cerebellar Purkinje cells in lobules VIII-X on the ipsilateral side of the Fast Blue-injected MPB were retrogradely labeled by Fast Blue and AAV vector (retrograde serotype), but no retrograde-labeled Purkinje cells were observed in lobules VI-VII and the cerebellar hemispheres. These results indicated that Purkinje cells in lobules VIII-X directly project their axons to the ipsilateral MPB but not lobules VI-VII. The direct connection between lobules VIII-X and the MPB suggests that the cerebellum participates in the neural network controlling the sleep-wake cycle, and cardiovascular and respiratory responses, by modulating the physiological function of the MPB.


Asunto(s)
Vermis Cerebeloso/citología , Núcleos Parabraquiales/citología , Células de Purkinje/citología , Amidinas , Animales , Dependovirus/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones Endogámicos ICR , Vías Nerviosas/citología , Técnicas de Trazados de Vías Neuroanatómicas , Trazadores del Tracto Neuronal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...