Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 16(7)2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-39066248

RESUMEN

Rotavirus is infamous for being extremely contagious and for causing diarrhea and vomiting in infants. However, the symptomology is far more complex than what could be expected from a pathogen restricted to the boundaries of the small intestines. Other rotavirus sickness symptoms like fever, fatigue, sleepiness, stress, and loss of appetite have been clinically established for decades but remain poorly studied. A growing body of evidence in recent years has strengthened the idea that the evolutionarily preserved defensive responses that cause rotavirus sickness symptoms are more than just passive consequences of illness and rather likely to be coordinated events from the central nervous system (CNS), with the aim of maximizing the survival of the individual as well as the collective group. In this review, we discuss both established and plausible mechanisms of different rotavirus sickness symptoms as a series of CNS responses coordinated from the brain. We also consider the protective and the harmful nature of these events and highlight the need for further and deeper studies on rotavirus etiology.


Asunto(s)
Encéfalo , Infecciones por Rotavirus , Rotavirus , Humanos , Infecciones por Rotavirus/virología , Encéfalo/virología , Rotavirus/fisiología , Animales , Diarrea/virología
2.
J Virol ; 97(11): e0152623, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37905839

RESUMEN

IMPORTANCE: Alterations of the gut microbiome can have significant effects on gastrointestinal homeostasis leading to various diseases and symptoms. Increased understanding of rotavirus infection in relation to the microbiota can provide better understanding on how microbiota can be used for clinical prevention as well as treatment strategies. Our volumetric 3D imaging data show that antibiotic treatment and its consequent reduction of the microbial load does not alter the extent of rotavirus infection of enterocytes in the small intestine and that restriction factors other than bacteria limit the infection of colonocytes.


Asunto(s)
Colon , Microbioma Gastrointestinal , Infecciones por Rotavirus , Animales , Humanos , Colon/virología , Tracto Gastrointestinal , Intestino Delgado/virología , Rotavirus , Ratones
3.
mBio ; 14(2): e0356722, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36976000

RESUMEN

Viral infections have a major impact on physiology and behavior. The clinical symptoms of human rotavirus and norovirus infection are primarily diarrhea, fever, and vomiting, but several other sickness symptoms, such as nausea, loss of appetite, and stress response are never or rarely discussed. These physiological and behavioral changes can be considered as having evolved to reduce the spread of the pathogen and increase the chances of survival of the individual as well as the collective. The mechanisms underlying several sickness symptoms have been shown to be orchestrated by the brain, specifically, the hypothalamus. In this perspective, we have described how the central nervous system contributes to the mechanisms underlying the sickness symptoms and behaviors of these infections. Based on published findings, we propose a mechanistic model depicting the role of the brain in fever, nausea, vomiting, cortisol-induced stress, and loss of appetite.


Asunto(s)
Infecciones por Caliciviridae , Enteritis , Infecciones por Enterovirus , Gastroenteritis , Norovirus , Infecciones por Rotavirus , Rotavirus , Humanos , Lactante , Vómitos/etiología , Náusea/etiología , Heces
4.
mBio ; 13(5): e0138722, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36094089

RESUMEN

While rotavirus diarrhea has been considered to occur only due to intrinsic intestinal effects within the enteric nervous system, we provide evidence for central nervous system control underlying the clinical symptomology. Our data visualize infection by large-scale three-dimensional (3D) volumetric tissue imaging of a mouse model and demonstrate that rotavirus infection disrupts the homeostasis of the autonomous system by downregulating tyrosine hydroxylase in the noradrenergic sympathetic nervous system in ileum, concomitant with increased intestinal transit. Interestingly, the nervous response was found to occur before the onset of clinical symptoms. In adult infected animals, we found increased pS6 immunoreactivity in the area postrema of the brain stem and decreased phosphorylated STAT5-immunoreactive neurons in the bed nucleus of the stria terminalis, which has been associated with autonomic control, including stress response. Our observations contribute to knowledge of how rotavirus infection induces gut-nerve-brain interaction early in the disease. IMPORTANCE Previous studies have investigated the mechanisms of rotavirus diarrhea mainly by focusing on intrinsic intestinal signaling. Although these observations are compelling and have provided important mechanistic information on rotavirus diarrhea, no information is available on how the gut communicates with the central nervous system (CNS) during rotavirus infection or on how this communication initiates sickness symptoms. We show that rotavirus infection presymptomatically disrupts the autonomic balance by downregulating the noradrenergic sympathetic nervous system in ileum, concomitant with increased intestinal transit and altered CNS activity, particularly in regions associated with autonomic control and stress response. Altogether, these observations reveal that the rotavirus-infected gut communicates with the CNS before the onset of diarrhea, a surprising observation that brings a new understanding of how rotavirus gives rise to sickness symptoms.


Asunto(s)
Infecciones por Rotavirus , Rotavirus , Ratones , Animales , Rotavirus/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Factor de Transcripción STAT5 , Diarrea , Sistema Nervioso Simpático/metabolismo , Íleon , Encéfalo/metabolismo
5.
J Virol ; 95(15): e0075121, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-33980599

RESUMEN

Rotavirus infection is highly prevalent in children, and the most severe effects are diarrhea and vomiting. It is well accepted that the enteric nervous system (ENS) is activated and plays an important role, but knowledge of how rotavirus activates nerves within ENS and to the vomiting center is lacking. Serotonin is released during rotavirus infection, and antagonists to the serotonin receptor subtype 3 (5-HT3 receptor) can attenuate rotavirus-induced diarrhea. In this study, we used a 5-HT3 receptor knockout (KO) mouse model to investigate the role of this receptor in rotavirus-induced diarrhea, motility, electrolyte secretion, inflammatory response, and vomiting reflex. The number of diarrhea days (P = 0.03) and the number of mice with diarrhea were lower in infected 5-HT3 receptor KO than wild-type pups. In vivo investigation of fluorescein isothiocyanate (FITC)-dextran transit time showed that intestinal motility was lower in the infected 5-HT3 receptor KO compared to wild-type mice (P = 0.0023). Ex vivo Ussing chamber measurements of potential difference across the intestinal epithelia showed no significant difference in electrolyte secretion between the two groups. Immediate early gene cFos expression level showed no difference in activation of the vomiting center in the brain. Cytokine analysis of the intestine indicated a low effect of inflammatory response in rotavirus-infected mice lacking the 5-HT3 receptor. Our findings indicate that the 5-HT3 receptor is involved in rotavirus-induced diarrhea via its effect on intestinal motility and that the vagus nerve signaling to the vomiting center occurs also in the absence of the 5-HT3 receptor. IMPORTANCE The mechanisms underlying rotavirus-induced diarrhea and vomiting are not yet fully understood. To better understand rotavirus pathophysiology, characterization of nerve signaling within the ENS and through vagal efferent nerves to the brain, which have been shown to be of great importance to the disease, is necessary. Serotonin (5-HT), a mediator of both diarrhea and vomiting, has been shown to be released from enterochromaffin cells in response to rotavirus infection and the rotavirus enterotoxin NSP4. Here, we investigated the role of the serotonin receptor 5-HT3, which is known to be involved in the nerve signals that regulate gut motility, intestinal secretion, and signal transduction through the vagus nerve to the brain. We show that the 5-HT3 receptor is involved in rotavirus-induced diarrhea by promoting intestinal motility. The findings shed light on new treatment possibilities for rotavirus diarrhea.


Asunto(s)
Diarrea/fisiopatología , Sistema Nervioso Entérico/fisiopatología , Motilidad Gastrointestinal/fisiología , Receptores de Serotonina 5-HT3/metabolismo , Infecciones por Rotavirus/patología , Vómitos/fisiopatología , Animales , Células Enterocromafines/metabolismo , Motilidad Gastrointestinal/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Serotonina 5-HT3/genética , Rotavirus/fisiología , Serotonina/metabolismo , Antagonistas del Receptor de Serotonina 5-HT3/farmacología
6.
Viruses ; 13(4)2021 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-33920421

RESUMEN

This qualitative review on rotavirus infection and its complications in the central nervous system (CNS) aims to understand the gut-brain mechanisms that give rise to CNS driven symptoms such as vomiting, fever, feelings of sickness, convulsions, encephalitis, and encephalopathy. There is substantial evidence to indicate the involvement of the gut-brain axis in symptoms such as vomiting and diarrhea. The underlying mechanisms are, however, not rotavirus specific, they represent evolutionarily conserved survival mechanisms for protection against pathogen entry and invasion. The reviewed studies show that rotavirus can exert effects on the CNS trough nervous gut-brain communication, via the release of mediators, such as the rotavirus enterotoxin NSP4, which stimulates neighboring enterochromaffin cells in the intestine to release serotonin and activate both enteric neurons and vagal afferents to the brain. Another route to CNS effects is presented through systemic spread via lymphatic pathways, and there are indications that rotavirus RNA can, in some cases where the blood brain barrier is weakened, enter the brain and have direct CNS effects. CNS effects can also be induced indirectly as a consequence of systemic elevation of toxins, cytokines, and/or other messenger molecules. Nevertheless, there is still no definitive or consistent evidence for the underlying mechanisms of rotavirus-induced CNS complications and more in-depth studies are required in the future.


Asunto(s)
Gastroenteritis/virología , Enfermedades del Sistema Nervioso/virología , Infecciones por Rotavirus/complicaciones , Rotavirus/patogenicidad , Animales , Barrera Hematoencefálica/virología , Encefalitis/virología , Tracto Gastrointestinal/virología , Humanos , Ratones , Infecciones por Rotavirus/virología , Toxinas Biológicas/biosíntesis , Toxinas Biológicas/fisiología , Proteínas no Estructurales Virales/biosíntesis , Proteínas no Estructurales Virales/fisiología
7.
J Neurosci ; 40(16): 3203-3216, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32209609

RESUMEN

Giving birth triggers a wide repertoire of physiological and behavioral changes in the mother to enable her to feed and care for her offspring. These changes require coordination and are often orchestrated from the CNS, through as of yet poorly understood mechanisms. A neuronal population with a central role in puerperal changes is the tuberoinfundibular dopamine (TIDA) neurons that control release of the pituitary hormone, prolactin, which triggers key maternal adaptations, including lactation and maternal care. Here, we used Ca2+ imaging on mice from both sexes and whole-cell recordings on female mouse TIDA neurons in vitro to examine whether they adapt their cellular and network activity according to reproductive state. In the high-prolactin state of lactation, TIDA neurons shift to faster membrane potential oscillations, a reconfiguration that reverses upon weaning. During the estrous cycle, however, which includes a brief, but pronounced, prolactin peak, oscillation frequency remains stable. An increase in the hyperpolarization-activated mixed cation current, Ih, possibly through unmasking as dopamine release drops during nursing, may partially explain the reconfiguration of TIDA rhythms. These findings identify a reversible plasticity in hypothalamic network activity that can serve to adapt the dam for motherhood.SIGNIFICANCE STATEMENT Motherhood requires profound behavioral and physiological adaptations to enable caring for offspring, but the underlying CNS changes are poorly understood. Here, we show that, during lactation, neuroendocrine dopamine neurons, the "TIDA" cells that control prolactin secretion, reorganize their trademark oscillations to discharge in faster frequencies. Unlike previous studies, which typically have focused on structural and transcriptional changes during pregnancy and lactation, we demonstrate a functional switch in activity and one that, distinct from previously described puerperal modifications, reverses fully on weaning. We further provide evidence that a specific conductance (Ih) contributes to the altered network rhythm. These findings identify a new facet of maternal brain plasticity at the level of membrane properties and consequent ensemble activity.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiología , Neuronas Dopaminérgicas/fisiología , Lactancia/fisiología , Red Nerviosa/fisiología , Plasticidad Neuronal/fisiología , Potenciales de Acción/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Transgénicos
8.
Elife ; 72018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29722649

RESUMEN

Electrical junctions are widespread within the mammalian CNS. Yet, their role in organizing neuronal ensemble activity remains incompletely understood. Here, in a functionally well-characterized system - neuroendocrine tuberoinfundibular dopamine (TIDA) neurons - we demonstrate a striking species difference in network behavior: rat TIDA cells discharge in highly stereotyped, robust, synchronized slow oscillations, whereas mouse oscillations are faster, flexible and show substantial cell-to-cell variability. We show that these distinct operational modes are explained by the presence of strong TIDA-TIDA gap junction coupling in the rat, and its complete absence in the mouse. Both species, however, encompass a similar heterogeneous range of intrinsic resonance frequencies, suggesting similar network building blocks. We demonstrate that gap junctions select and impose the slow network rhythm. These data identify a role for electrical junctions in determining oscillation frequency and show how related species can rely on distinct network strategies to accomplish adaptive control of hormone release.


Asunto(s)
Potenciales de Acción , Relojes Biológicos , Uniones Comunicantes/metabolismo , Red Nerviosa/fisiología , Neuronas/fisiología , Animales , Ratones , Ratas
9.
Sci Rep ; 7: 41535, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28145492

RESUMEN

Hypocretin/Orexin (H/O) neurons of the lateral hypothalamus are compelling modulator candidates for the chronobiology of neuroendocrine output and, as a consequence, hormone release from the anterior pituitary. Here we investigate the effects of H/O peptides upon tuberoinfundibular dopamine (TIDA) neurons - cells which control, via inhibition, the pituitary secretion of prolactin. In whole cell recordings performed in male rat hypothalamic slices, application of H/O-A, as well as H/O-B, excited oscillating TIDA neurons, inducing a reversible depolarising switch from phasic to tonic discharge. The H/O-induced inward current underpinning this effect was post-synaptic (as it endured in the presence of tetrodotoxin), appeared to be carried by a Na+-dependent transient receptor potential-like channel (as it was blocked by 2-APB and was diminished by removal of extracellular Na+), and was a consequence of OX2R receptor activation (as it was blocked by the OX2R receptor antagonist TCS OX2 29, but not the OX1R receptor antagonist SB 334867). Application of the hormone, melatonin, failed to alter TIDA membrane potential or oscillatory activity. This first description of the electrophysiological effects of H/Os upon the TIDA network identifies cellular mechanisms that may contribute to the circadian rhythmicity of prolactin secretion.


Asunto(s)
Cationes/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/fisiología , Receptores de Orexina/metabolismo , Orexinas/farmacología , Potenciales Sinápticos/efectos de los fármacos , Animales , Compuestos de Boro/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Melatonina/metabolismo , Melatonina/farmacología , Sistemas Neurosecretores/efectos de los fármacos , Sistemas Neurosecretores/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas , Sodio/metabolismo , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores
10.
Nat Neurosci ; 20(2): 176-188, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27991900

RESUMEN

The hypothalamus contains the highest diversity of neurons in the brain. Many of these neurons can co-release neurotransmitters and neuropeptides in a use-dependent manner. Investigators have hitherto relied on candidate protein-based tools to correlate behavioral, endocrine and gender traits with hypothalamic neuron identity. Here we map neuronal identities in the hypothalamus by single-cell RNA sequencing. We distinguished 62 neuronal subtypes producing glutamatergic, dopaminergic or GABAergic markers for synaptic neurotransmission and harboring the ability to engage in task-dependent neurotransmitter switching. We identified dopamine neurons that uniquely coexpress the Onecut3 and Nmur2 genes, and placed these in the periventricular nucleus with many synaptic afferents arising from neuromedin S+ neurons of the suprachiasmatic nucleus. These neuroendocrine dopamine cells may contribute to the dopaminergic inhibition of prolactin secretion diurnally, as their neuromedin S+ inputs originate from neurons expressing Per2 and Per3 and their tyrosine hydroxylase phosphorylation is regulated in a circadian fashion. Overall, our catalog of neuronal subclasses provides new understanding of hypothalamic organization and function.


Asunto(s)
Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Hipotálamo/metabolismo , Neuropéptidos/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Animales , Inmunohistoquímica/métodos , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurotransmisores/fisiología , Núcleo Supraquiasmático/metabolismo , Transmisión Sináptica/fisiología
11.
J Neurosci ; 36(28): 7392-406, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27413150

RESUMEN

UNLABELLED: Selective serotonin reuptake inhibitors (SSRIs) are commonly prescribed for depression, but sexual side effects often compromise compliance. These reproductive dysfunctions are likely mediated by elevations of the hormone prolactin. Yet, how serotonin (5-HT) and SSRIs cause changes in prolactin secretion is not known. Here, using in vitro whole-cell patch-clamp recordings, we show that 5-HT hyperpolarizes and abolishes phasic discharge in rat neuroendocrine tuberoinfundibular dopamine (TIDA) neurons, the main inhibitor of prolactin secretion. This process is underpinned by 5-HT1A receptor-mediated activation of G-protein-coupled inwardly rectifying K(+)-like currents. We further demonstrate that the SSRIs, fluoxetine and sertraline, directly suppress TIDA neuron activity through parallel effects, independent of 5-HT transmission. This inhibition involves decreased intrinsic excitability and a slowing of TIDA network rhythms. These findings indicate that SSRIs may inhibit neuroendocrine dopamine release through both 5-HT-dependent and -independent actions, providing a mechanistic explanation for, and potential molecular targets for the amelioration of, the hyperprolactinemia and sexual dysfunction associated with these drugs. SIGNIFICANCE STATEMENT: Depression affects approximately one-tenth of the population and is commonly treated with selective serotonin reuptake inhibitors (SSRIs; e.g., Prozac). Yet, many patients withdraw from SSRI therapy due to sexual side effects (e.g., infertility, menstrual disturbances, and impotence). Although it is generally accepted that sexual side effects are due to the ability of these drugs to elevate blood levels of the hormone prolactin, the mechanism for this hormonal imbalance is not known. Here, we show that SSRIs can inhibit hypothalamic dopamine neurons that normally suppress the secretion of prolactin. Intriguingly this inhibition can be explained both by increased serotonin activity and also by parallel serotonin-independent actions.


Asunto(s)
Antidepresivos/farmacología , Núcleo Arqueado del Hipotálamo/citología , Neuronas Dopaminérgicas/efectos de los fármacos , Lactotrofos/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Serotonina/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Estimulación Eléctrica , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Técnicas In Vitro , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Antagonistas de la Serotonina/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Tetrahidronaftalenos/farmacología , Tetrodotoxina/farmacología , Tirosina 3-Monooxigenasa/metabolismo
12.
Neuropharmacology ; 107: 89-99, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26947946

RESUMEN

Phosphorylation of histone H3 (H3) on serine 28 (S28) at genomic regions marked by trimethylation of lysine 27 (H3K27me3) often correlates with increased expression of genes normally repressed by Polycomb group proteins (PcG). We show that amphetamine, an addictive psychostimulant, and haloperidol, a typical antipsychotic drug, increase the phosphorylation of H3 at S28 and that this effect occurs in the context of H3K27me3. The increases in H3K27me3S28p occur in distinct populations of projection neurons located in the striatum, the major component of the basal ganglia. Genetic inactivation of the protein phosphatase-1 inhibitor, dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32), reduces the phosphorylation of H3K27me3S28 produced by amphetamine and haloperidol. In contrast, knockout of the mitogen- and stress activated kinase 1 (MSK1), which is implicated in the phosphorylation of histone H3, decreases the effect of amphetamine, but not that of haloperidol. Chromatin immunoprecipitation analysis shows that amphetamine and haloperidol increase the phosphorylation of H3K27me3S28 at the promoter regions of Atf3, Npas4 and Lipg, three genes repressed by PcG. These results identify H3K27me3S28p as a potential mediator of the effects exerted by amphetamine and haloperidol, and suggest that these drugs may act by re-activating PcG repressed target genes.


Asunto(s)
Cuerpo Estriado/metabolismo , Histonas/metabolismo , Neuronas/metabolismo , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Anfetamina/farmacología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Fármacos del Sistema Nervioso Central/farmacología , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/fisiología , Haloperidol/farmacología , Histonas/genética , Lipasa/genética , Lipasa/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/citología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/fisiología , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
13.
J Chem Neuroanat ; 61-62: 20-32, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25014433

RESUMEN

Calcium binding proteins (CaBPs) form a diverse group of molecules that function as signal transducers or as intracellular buffers of Ca(2+) concentration. They have been extensively used to histochemically categorize cell types throughout the brain. One region which has not yet been characterized with regard to CaBP expression is the hypothalamic arcuate nucleus, which plays a vital role in neuroendocrine control and the central regulation of energy metabolism. Using in situ hybridization and immunofluorescence, we have investigated the cellular distribution of the three CaBPs, calbindin-D28k (CB), calretinin (CR) and parvalbumin (PV) in the rat arcuate nucleus. Both mRNA and immunoreactivity was detected in the arcuate nucleus for CB - located in the medial aspects - and CR - located ventrolaterally. No PV mRNA was detected in the arcuate nucleus. Immunofluorescence results for PV were ambiguous; while one antibody detected a group of cell somata, a different antibody failed to visualize any arcuate nucleus cell profiles. Using double-labeling, neither of the examined CaBPs were observed in cells immunoreactive for the signaling molecules agouti gene-related protein, tyrosine hydroxylase, neurotensin, growth hormone-releasing hormone, somatostatin, enkephalin, dynorphin or galanin. We did, however, observe CB- and CR-immunoreactivity, in two distinct populations of neurons immunoreactive for the melanocortin peptide α-melanocyte-stimulating hormone. These data identify distinct subpopulations of arcuate neurons defined by their expression of CaBPs and provide further support for differentiation between subpopulations of anorexigenic melanocortin neurons.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Calbindina 1/biosíntesis , Calbindina 2/biosíntesis , Neuronas/metabolismo , Parvalbúminas/biosíntesis , Animales , Calbindina 1/análisis , Calbindina 2/análisis , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Masculino , Parvalbúminas/análisis , Ratas , Ratas Sprague-Dawley
14.
J Neurosci ; 32(23): 8074-83, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22674282

RESUMEN

Balance in the body's hormonal axes depends on feedback onto neuroendocrine hypothalamic neurons. This phenomenon involves transcriptional and biosynthetic effects, yet less is known about the potential rapid modulation of electrical properties. Here, we investigated this issue in the lactotrophic axis, in which the pituitary hormone prolactin is tonically inhibited by tuberoinfundibular dopamine (TIDA) neurons located in the hypothalamic arcuate nucleus. Whole-cell recordings were performed on slices of the rat hypothalamus. In the presence of prolactin, spontaneously oscillating TIDA cells depolarized, switched from phasic to tonic discharge, and exhibited broadened action potentials. The underlying prolactin-induced current is composed of separate low- and high-voltage components that include the activation of a transient receptor potential-like current and the inhibition of a Ca(2+)-dependent BK-type K(+) current, respectively, as revealed by ion substitution experiments and pharmacological manipulation. The two components of the prolactin-induced current appear to be mediated through distinct signaling pathways as the high-voltage component is abolished by the phosphoinositide 3-kinase blocker wortmannin, whereas the low-voltage component is not. This first description of the central electrophysiological actions of prolactin suggests a novel feedback mechanism. By simultaneously enhancing the discharge and spike duration of TIDA cells, increased serum prolactin can promote dopamine release to limit its own secretion with implications for the control of lactation, sexual libido, fertility, and body weight.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiología , Dopamina/fisiología , Retroalimentación Fisiológica/fisiología , Lactancia/fisiología , Neuronas/fisiología , Prolactina/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Compuestos de Boro/farmacología , Calcio/farmacología , Interpretación Estadística de Datos , Fenómenos Electrofisiológicos , Retroalimentación Fisiológica/efectos de los fármacos , Femenino , Indicadores y Reactivos , Canales Iónicos/fisiología , Lactancia/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Potenciales de la Membrana/fisiología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Prolactina/farmacología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA