Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 2081, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38453902

RESUMEN

The presence of a companion can reduce fear, but the neural mechanisms underlying this social buffering of fear are incompletely known. We studied social buffering of fear in male and female, and its encoding in the amygdala of male, auditory fear-conditioned rats. Pharmacological, opto,- and/or chemogenetic interventions showed that oxytocin signaling from hypothalamus-to-central amygdala projections underlied fear reduction acutely with a companion and social buffering retention 24 h later without a companion. Single-unit recordings with optetrodes in the central amygdala revealed fear-encoding neurons (showing increased conditioned stimulus-responses after fear conditioning) inhibited by social buffering and blue light-stimulated oxytocinergic hypothalamic projections. Other central amygdala neurons showed baseline activity enhanced by blue light and companion exposure, with increased conditioned stimulus responses that persisted without the companion. Social buffering of fear thus switches the conditioned stimulus from encoding "fear" to "safety" by oxytocin-mediated recruitment of a distinct group of central amygdala "buffer neurons".


Asunto(s)
Núcleo Amigdalino Central , Condicionamiento Psicológico , Ratas , Masculino , Femenino , Animales , Condicionamiento Psicológico/fisiología , Oxitocina , Ratas Wistar , Miedo/fisiología , Neuronas
2.
Mol Psychiatry ; 2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34035479

RESUMEN

The neuropeptide oxytocin (OXT) has generated considerable interest as potential treatment for psychiatric disorders, including anxiety and autism spectrum disorders. However, the behavioral and molecular consequences associated with chronic OXT treatment and chronic receptor (OXTR) activation have scarcely been studied, despite the potential therapeutic long-term use of intranasal OXT. Here, we reveal that chronic OXT treatment over two weeks increased anxiety-like behavior in rats, with higher sensitivity in females, contrasting the well-known anxiolytic effect of acute OXT. The increase in anxiety was transient and waned 5 days after the infusion has ended. The behavioral effects of chronic OXT were paralleled by activation of an intracellular signaling pathway, which ultimately led to alternative splicing of hypothalamic corticotropin-releasing factor receptor 2α (Crfr2α), an important modulator of anxiety. In detail, chronic OXT shifted the splicing ratio from the anxiolytic membrane-bound (mCRFR2α) form of CRFR2α towards the soluble CRFR2α (sCRFR2α) form. Experimental induction of alternative splicing mimicked the anxiogenic effects of chronic OXT, while sCRFR2α-knock down reduced anxiety-related behavior of male rats. Furthermore, chronic OXT treatment triggered the release of sCRFR2α into the cerebrospinal fluid with sCRFR2α levels positively correlating with anxiety-like behavior. In summary, we revealed that the shifted splicing ratio towards expression of the anxiogenic sCRFR2α underlies the adverse effects of chronic OXT treatment on anxiety.

3.
Neuropharmacology ; 173: 108130, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32389750

RESUMEN

Many studies in preclinical animal models have described fear-reducing effects of the neuropeptide oxytocin in the central nucleus of the amygdala. However, recent studies have refined the role of oxytocin in the central amygdala, which may extend to the selection of an active defensive coping style in the face of immediate threat, and also fear-enhancing effects have been reported. On top of this, oxytocin enables the discrimination of unfamiliar conspecifics on the basis of their emotional state, which could allow for the selection of an appropriate coping style. This is in line with many observations that support the hypothesis that the precise outcome of oxytocin signaling in the central amygdala or other brain regions depends on the emotional or physiological state of an animal. In this review, we highlight a number of studies to exemplify the diverse effects oxytocin exerts on fear in the central amygdala of rodents. These are discussed in the context of the organization of the neural network within the central amygdala and in relation to the oxytocin-synthesizing neurons in the hypothalamus.


Asunto(s)
Conducta/fisiología , Núcleo Amigdalino Central/metabolismo , Miedo/fisiología , Oxitocina/metabolismo , Animales , Bombesina/farmacología , Encéfalo/metabolismo , Humanos , Modelos Animales , Transducción de Señal
4.
Biol Psychiatry ; 85(10): 802-811, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30826070

RESUMEN

BACKGROUND: The neuropeptide oxytocin (OXT) mediates its actions, including anxiolysis, via its G protein-coupled OXT receptor. Within the paraventricular nucleus of the hypothalamus (PVN), OXT-induced anxiolysis is mediated, at least in part, via activation of the mitogen-activated protein kinase pathway following calcium influx through transient receptor potential cation channel subfamily V member 2 channels. In the periphery, OXT activates eukaryotic elongation factor 2 (eEF2), an essential mediator of protein synthesis. METHODS: In order to study whether OXT activates eEF2 also in neurons to exert its anxiolytic properties in the PVN, we performed in vivo and cell culture experiments. RESULTS: We demonstrate that OXT, in a protein kinase C-dependent manner, activates eEF2 both in a hypothalamic cell line and in vivo within the PVN. Next, we reveal that OXT stimulates de novo protein synthesis, while inhibition of protein synthesis within the PVN prevents the anxiolytic effect of OXT in male rats. Moreover, activation of eEF2 within the PVN conveyed an anxiolytic effect supporting a role of OXT-induced eEF2 activation and protein synthesis for its anxiolysis. Finally, we show that one of the proteins that is upregulated by OXT is the neuropeptide Y receptor 5. Infusion of a specific neuropeptide Y receptor 5 agonist into the PVN consequently led to decreased anxiety-related behavior, while pretreatment with a neuropeptide Y receptor 5 antagonist prevented the anxiolytic effect of OXT. CONCLUSIONS: Taken together, these results show that OXT recruits several intracellular signaling cascades to induce protein synthesis, which mediates the anxiolytic effects of OXT within the PVN and suggests that eEF2 represents a novel target for anxiety-related disorders.


Asunto(s)
Ansiolíticos/metabolismo , Ansiedad/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Animales , Ansiolíticos/administración & dosificación , Células Cultivadas , Regulación hacia Abajo , Sistema de Señalización de MAP Quinasas , Masculino , Oxitocina/administración & dosificación , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Proteína Quinasa C/metabolismo , Ratas Wistar , Receptores de Neuropéptido Y/metabolismo , Regulación hacia Arriba
5.
Cell Tissue Res ; 375(1): 93-101, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29948174

RESUMEN

The central amygdala has a rich repertoire of neuropeptides and neuropeptide receptors. The diverse ways in which they modulate neuronal activity and influence synaptic activity are discussed here mostly in the context of fear and anxiety-related behaviour but also with respect to nociception, hunger and satiety and chronic alcohol exposure that often come together with anxiety. It appears that neuropeptides exert rather specific effects on behaviour and physiology that can be quite different from the effects evoked by opto- or chemogenetical stimulation of the central amygdala neurons that synthesise them or express their receptors. Also, neuropeptides might work synergistically or antagonistically to fine-tune the final outcome of sensory processing in the central amygdala and bring about appropriate physiological and behavioural responses to threat. Taken together, we propose that neuropeptide signalling in the central amygdala mainly serves to establish or maintain emotional homeostasis in response to threatening and other sensory stimuli.


Asunto(s)
Núcleo Amigdalino Central/metabolismo , Neuropéptidos/metabolismo , Transducción de Señal , Animales , Miedo , Humanos , Memoria , Red Nerviosa/metabolismo
6.
Cell ; 175(3): 723-735.e16, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30340041

RESUMEN

Rodent research delineates how the basolateral amygdala (BLA) and central amygdala (CeA) control defensive behaviors, but translation of these findings to humans is needed. Here, we compare humans with natural-selective bilateral BLA lesions to rats with a chemogenetically silenced BLA. We find, across species, an essential role for the BLA in the selection of active escape over passive freezing during exposure to imminent yet escapable threat (Timm). In response to Timm, BLA-damaged humans showed increased startle potentiation and BLA-silenced rats demonstrated increased startle potentiation, freezing, and reduced escape behavior as compared to controls. Neuroimaging in humans suggested that the BLA reduces passive defensive responses by inhibiting the brainstem via the CeA. Indeed, Timm conditioning potentiated BLA projections onto an inhibitory CeA pathway, and pharmacological activation of this pathway rescued deficient Timm responses in BLA-silenced rats. Our data reveal how the BLA, via the CeA, adaptively regulates escape behavior from imminent threat and that this mechanism is evolutionary conserved across rodents and humans.


Asunto(s)
Complejo Nuclear Basolateral/fisiología , Reacción de Fuga , Adulto , Animales , Miedo , Femenino , Reacción Cataléptica de Congelación , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto , Especificidad de la Especie
7.
Horm Behav ; 79: 18-27, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26747375

RESUMEN

Recent studies using V1b receptor (V1bR) knockout mice or central pharmacological manipulations in lactating rats highlighted the influence of this receptor for maternal behavior. However, its role in specific brain sites known to be important for maternal behavior has not been investigated to date. In the present study, we reveal that V1bR mRNA (qPCR) and protein levels (Western blot) within either the medial preoptic area (MPOA) or the medial-posterior part of the bed nucleus of the stria terminalis (mpBNST) did not differ between virgin and lactating rats. Furthermore, we characterized the effects of V1bR blockade via bilateral injections of the receptor subtype-specific antagonist SSR149415 within the MPOA or the mpBNST on maternal behavior (maternal care under non-stress and stress conditions, maternal motivation to retrieve pups in a novel environment, maternal aggression) and anxiety-related behavior in lactating rats. Blocking V1bR within the MPOA increased pup retrieval, whereas within the mpBNST it decreased pup-directed behavior, specifically licking/grooming the pups, during the maternal defense test. In addition, immediately after termination of the maternal defense test, V1bR antagonism in both brain regions reduced nursing, particularly arched back nursing. Anxiety-related behavior was not affected by V1bR antagonism in either brain region. In conclusion our data indicate that V1bR antagonism significantly modulates different aspects of maternal behavior in a brain region-dependent manner.


Asunto(s)
Agresión/efectos de los fármacos , Antagonistas de los Receptores de Hormonas Antidiuréticas/farmacología , Indoles/farmacología , Conducta Materna/efectos de los fármacos , Motivación/efectos de los fármacos , Área Preóptica/efectos de los fármacos , Pirrolidinas/farmacología , Núcleos Septales/efectos de los fármacos , Agresión/psicología , Animales , Conducta Animal/efectos de los fármacos , Femenino , Aseo Animal/efectos de los fármacos , Lactancia/efectos de los fármacos , Masculino , Conducta Materna/psicología , Comportamiento de Nidificación/efectos de los fármacos , Embarazo , Área Preóptica/metabolismo , Ratas , Ratas Wistar , Receptores de Vasopresinas/metabolismo , Núcleos Septales/metabolismo
8.
J Neurosci ; 35(35): 12248-60, 2015 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-26338335

RESUMEN

The major regulator of the neuroendocrine stress response in the brain is corticotropin releasing factor (CRF), whose transcription is controlled by CREB and its cofactors CRTC2/3 (TORC2/3). Phosphorylated CRTCs are sequestered in the cytoplasm, but rapidly dephosphorylated and translocated into the nucleus following a stressful stimulus. As the stress response is attenuated by oxytocin (OT), we tested whether OT interferes with CRTC translocation and, thereby, Crf expression. OT (1 nmol, i.c.v.) delayed the stress-induced increase of nuclear CRTC3 and Crf hnRNA levels in the paraventricular nucleus of male rats and mice, but did not affect either parameter in the absence of the stressor. The increase in Crf hnRNA levels at later time points was parallel to elevated nuclear CRTC2/3 levels. A direct effect of Thr(4) Gly(7)-OT (TGOT) on CRTC3 translocation and Crf expression was found in rat primary hypothalamic neurons, amygdaloid (Ar-5), hypothalamic (H32), and human neuroblastoma (Be(2)M17) cell lines. CRTC3, but not CRCT2, knockdown using siRNA in Be(2)M17 cells prevented the effect of TGOT on Crf hnRNA levels. Chromatin-immunoprecipitation demonstrated that TGOT reduced CRTC3, but not CRTC2, binding to the Crf promoter after 10 min of forskolin stimulation. Together, the results indicate that OT modulates CRTC3 translocation, the binding of CRTC3 to the Crf promoter and, ultimately, transcription of the Crf gene. SIGNIFICANCE STATEMENT: The neuropeptide oxytocin has been proposed to reduce hypothalamic-pituitary-adrenal (HPA) axis activation during stress. The underlying mechanisms are, however, elusive. In this study we show that activation of the oxytocin receptor in the paraventricular nucleus delays transcription of the gene encoding corticotropin releasing factor (Crf), the main regulator of the stress response. It does so by sequestering the coactivator of the transcription factor CREB, CRTC3, in the cytosol, resulting in reduced binding of CRTC3 to the Crf gene promoter and subsequent Crf gene expression. This novel oxytocin receptor-mediated intracellular mechanism might provide a basis for the treatment of exaggerated stress responses in the future.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Regulación de la Expresión Génica , Oxitocina/farmacología , Estrés Psicológico/metabolismo , Tromboplastina/metabolismo , Animales , Células Cultivadas , Colforsina/farmacología , Hormona Liberadora de Corticotropina/genética , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/fisiología , Oxitócicos/farmacología , Oxitócicos/uso terapéutico , Oxitocina/análogos & derivados , Oxitocina/uso terapéutico , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Ratas , Ratas Wistar , Receptores de Oxitocina/metabolismo , Transducción de Señal/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/patología
9.
Neuropsychopharmacology ; 40(13): 2938-47, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26013963

RESUMEN

There is growing interest in anxiolytic and pro-social effects of the neuropeptide oxytocin (OXT), but the underlying intraneuronal mechanisms are largely unknown. Here we examined OXT-mediated anxiolysis in the hypothalamic paraventricular nucleus (PVN) of rats and effects of OXT administration on signaling events in hypothalamic primary and immortalized cells. In vivo, the application of SKF96365 prevented the anxiolytic activity of OXT in the PVN, suggesting that changes in intracellular Ca(2+) mediate the acute OXT behavioral effects. In vitro, mainly in the neurons with autonomous Ca(2+) oscillations, OXT increased intracellular Ca(2+) concentration and oscillation amplitude. Pharmacological intervention revealed OXT-dependent changes in Ca(2+) signaling that required activation of transient receptor potential vanilloid type-2 channel (TRPV2), mediated by phosphoinositide 3-kinase. TRPV2 induced the activation of the anxiolytic mitogen-activated protein kinase kinase (MEK1/2). In situ, immunohistochemistry revealed co-localization of TRPV2 and OXT in the PVN. Thus, functional and pharmacological analyses identified TRPV2 as a mediator of anxiolytic effects of OXT, conveying the OXT signal to MEK1/2 via modulation of intracellular Ca(2+).


Asunto(s)
Ansiolíticos/farmacología , Calcio/metabolismo , Espacio Extracelular/efectos de los fármacos , Oxitocina/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Cationes Bivalentes/metabolismo , Células Cultivadas , Evaluación Preclínica de Medicamentos , Espacio Extracelular/metabolismo , Imidazoles/farmacología , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas Wistar , Receptores de Oxitocina/antagonistas & inhibidores , Receptores de Oxitocina/metabolismo
10.
PLoS One ; 7(5): e37060, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615888

RESUMEN

The c-Raf - MEK1/2 - ERK1/2 mitogen-activated protein kinase (MAPK) intracellular signalling cascade in neurons plays important roles in the control of a variety of behaviours, including social behaviours and anxiety. These roles partially overlap with those described for oxytocin (OXT), and it has been shown that OXT activates the MAPK pathway in the hypothalamus (of male), and hippocampus (of female) rats. Here, by combining behavioural (light/dark box) and biochemical analyses (western blotting), we tested two hypotheses: (i) that OXT is anxiolytic within the hypothalamus of females, and (ii) that this effect, as well as that of lactation-associated anxiolysis, depends on the recruitment of the MAPK pathway. We found that, when injected bilaterally into the hypothalamic paraventricular nucleus (PVN), OXT decreased anxiety-like behaviour in virgins, and that this effect depended on phosphorylation of MEK1/2. MAPK pathway activation in lactation was evident by high phosphorylated (p) MEK1/2 levels, and nuclear translocation of ERK1. The high pMEK1/2 levels were necessary for the anxiolytic phenotype typically observed during lactation. Interestingly, exogenous OXT in lactating rats reduced pMEK1/2 levels without a concomitant effect on anxiety, indicating that OXT receptor activation can lead to recruitment of additional intracellular pathways to modulate MEK activity. Still other pathways could include MEK, but without subsequent activation of ERK, as we did not observe any increase in OXT-induced ERK phosphorylation. Together the results demonstrate that the MAPK pathway, especially MEK1/2, is critically involved in the regulation of anxiety-like behaviour in female rats.


Asunto(s)
Ansiedad/enzimología , Lactancia/metabolismo , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Núcleo Hipotalámico Paraventricular/enzimología , Animales , Ansiedad/psicología , Femenino , Sistema de Señalización de MAP Quinasas/fisiología , Oxitocina/farmacología , Fosforilación/fisiología , Embarazo , Ratas , Ratas Wistar , Transducción de Señal , Conducta Social
11.
J Mol Neurosci ; 43(2): 200-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20865346

RESUMEN

Neuropeptides of the brain are important neuromodulators, controlling behaviour and physiology. They signal through G protein-coupled receptors (GPCR) that couple to complex intracellular signalling pathways. These signalling networks integrate information from multiple sources, resulting in appropriate physiological and behavioural responses to environmental and internal cues. This paper will focus on the neuropeptides oxytocin and prolactin with respect to (1) the regulation of neuroendocrine stress responses and anxiety, and (2) the receptor-mediated molecular mechanisms underlying these actions of the neuropeptides. Besides its significant reproductive functions when released into the bloodstream, brain oxytocin reduces the activity of the hypothalamo-pituitary-adrenal (HPA) axis as well as anxiety-related behaviour in male and female rats. Oxytocin mediates its anxiolytic effect, at least in part, via binding to its GPCR in the hypothalamic paraventricular nucleus, followed by transactivation of the epidermal growth factor receptor, and subsequent activation of a MEK-extracellular signal-regulated kinase (ERK) MAP kinase pathway. Prolactin, by binding to its GPCR receptors, of which there are short and long forms, also activates ERK, and this is necessary for the control of the expression of corticotrophin-releasing hormone-an important regulator of the HPA axis. Liganded oxytocin and prolactin may also recruit other signalling pathways, but how these pathways contribute to the observed behavioural and physiological effects remains to be established. GPCR-mediated oxytocin and prolactin neuronal signalling are illustrative of the complexity of GPCR-activated regulation of appropriate neuroendocrine and behavioural responses to environmental and physiological demands.


Asunto(s)
Hipotálamo/metabolismo , Oxitocina/metabolismo , Prolactina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistemas de Mensajero Secundario/fisiología , Animales , Ansiolíticos/metabolismo , Ansiedad/fisiopatología , Hipotálamo/anatomía & histología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estrés Fisiológico/fisiología
12.
J Neurophysiol ; 97(3): 2373-84, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17202233

RESUMEN

Anesthetics may induce specific changes that alter the balance of activity within neural networks. Here we describe the effects of the GABA(A) receptor potentiating anesthetic etomidate on sensory processing, studied in a cerebellum-like structure, the electrosensory lateral line lobe (ELL) of mormyrid fish, in vitro. Previous studies have shown that the ELL integrates sensory input and removes predictable features by comparing reafferent sensory signals with a descending electromotor command-driven corollary signal that arrives in part through parallel fiber synapses with the apical dendrites of GABAergic interneurons. These synapses show spike timing-dependent depression when presynaptic activation is associated with postsynaptic backpropagating dendritic action potentials. Under etomidate, almost all neurons become tonically hyperpolarized. The threshold for action potential initiation increased for both synaptic activation and direct intracellular depolarization. Synaptically evoked inhibitory postsynaptic potentials (IPSPs) were also strongly potentiated and prolonged. Current source density analysis showed that backpropagation of action potentials through the apical dendritic arborization in the molecular layer was reduced but could be restored by increasing stimulus strength. These effects of etomidate were blocked by bicuculline or picrotoxin. It is concluded that etomidate affects both tonic and phasic inhibitory conductances at GABA(A) receptors and that increased shunting inhibition at the level of the proximal dendrites also contributes to increasing the threshold for action potential backpropagation. When stimulus strength is sufficient to evoke backpropagation, repetitive association of synaptic excitation with postsynaptic action potential initiation still results in synaptic depression, showing that etomidate does not interfere with the molecular mechanism underlying plastic modulation.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Vías Aferentes/efectos de los fármacos , Dendritas/efectos de los fármacos , Etomidato/farmacología , Hipnóticos y Sedantes/farmacología , Plasticidad Neuronal/efectos de los fármacos , Vías Aferentes/citología , Vías Aferentes/fisiología , Anestesia , Animales , Bicuculina/farmacología , Relación Dosis-Respuesta en la Radiación , Interacciones Farmacológicas , Pez Eléctrico/anatomía & histología , Pez Eléctrico/fisiología , Órgano Eléctrico/citología , Estimulación Eléctrica/métodos , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas del GABA/farmacología , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Plasticidad Neuronal/fisiología , Neuronas Aferentes/citología , Neuronas Aferentes/efectos de los fármacos
13.
Gen Comp Endocrinol ; 146(1): 36-44, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16403502

RESUMEN

The endocrine stress response is pivotal in vertebrate physiology. The stress hormone cortisol-the end product of the endocrine stress axis-(re-)directs energy flows for optimal performance under conditions where homeostasis may be or become at risk. Key players in the continuous adaptation process are corticotropin-releasing factor (CRF) from the hypothalamic nucleus preopticus (NPO), pituitary adrenocorticotropic hormone (ACTH) and cortisol produced by the interrenal cells in the headkidney (adrenal equivalent of fish). CRF is a member of a large family of related peptides that signals through CRF-receptor subtypes specific for central and peripheral actions of the peptide. CRF is "chaperoned" by a unique and phylogenetically very well-conserved binding protein (CRFBP); the functions of the CRFBP can only be speculated on so far, but its mRNA and protein abundance are important indicators of the central CRF-system activity, and indeed its mRNA levels are altered by restraint stress. Moreover, the unique structure and size of the CRFBP provide good tools in phylogenetic studies, that date the CRF-system to at least one billion years old. Pro-opiomelanocortin is produced and processed to ACTH and endorphin in the hypothalamic NPO and pituitary pars distalis ACTH-cells, to MSH and acetylated endorphins in the pituitary pars intermedia MSH-cells. ACTH is the prime corticotrope in acute stress conditions. In carp, MSH, considered a mild corticotrope in chronic stress responses in other fish, lacks corticotropic effects (in line with the absence of the melanocortin-5 receptor in headkidney); yet, an unknown corticotropic signal substance in the pars intermedia of carp awaits elucidation. Interesting observations were made on the CRF control of pituitary cells. CRF stimulates ACTH-cells, but only when these cells experience a mild dopaminergic block. Endorphin, produced in the NPO and transported via axons to the pituitary gland in vivo, reverses the stimulatory CRF action on MSH-cells to a differential inhibition of N-acetyl beta-endorphin release in vitro (MSH release is not affected). We speculate that the consistently observed elevation of plasma MSH during chronic stress may exert central actions related to feeding and leptin regulated processes. A BOLD-fMRI study revealed the functional anatomy of the stress response at work in a paradigm, where carp were exposed to a sudden water temperature drop. In carp (and other fish), the endocrine stress axis is already operational in very early life stages, viz., around hatching and comprises hypothalamic, pituitary, and interrenal signaling to adjust the physiology of the hatchling to its dynamically changing environment. Understanding of stress during early life stages is critical as the consequent rises in cortisol may have long lasting effects on survival and fish quality.


Asunto(s)
Carpas/fisiología , Hormona Liberadora de Corticotropina/fisiología , Estrés Fisiológico/fisiopatología , Animales , Carpas/embriología , Endorfinas/fisiología , Hipotálamo Anterior/fisiología , Imagen por Resonancia Magnética , Hormonas Estimuladoras de los Melanocitos/fisiología , Proopiomelanocortina/fisiología
14.
Am J Physiol Regul Integr Comp Physiol ; 289(3): R814-26, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15890786

RESUMEN

Cortisol release from fish head kidney during the acute phase of the stress response is controlled by the adrenocorticotropic hormone (ACTH) from the pituitary pars distalis (PD). Alpha-melanocyte-stimulating hormone (alpha-MSH) and beta-endorphin, from the pars intermedia (PI), have been implicated in cortisol release during the chronic phase. The present study addresses the regulation of cortisol release by ACTH and alpha-MSH in common carp (Cyprinus carpio) and includes characterization of their receptors, namely, the melanocortin-2 and melanocortin-5 receptors (MC2R and MC5R). We could not demonstrate corticotropic activity of alpha-MSH, beta-endorphin, and combinations of these. We do show a corticotrope in the PI, but its identity is as yet uncertain. Carp restrained for 1 and 7 days showed elevated plasma cortisol and alpha-MSH levels; cortisol is still elevated but lower at day 7 than day 1 of restraint. Interrenal response capacity is unaffected, as estimated by stimulation with a maximum dose ACTH in a superfusion setup. MC2R and MC5R appear phylogenetically well conserved. MC2R is predominantly expressed in head kidney; a low abundance was found in spleen and kidney. MC5R is expressed in brain, pituitary PD, kidney, and skin. Quantitative PCR analysis of MC2R and MC5R expression in the head kidney of restrained fish reveals MC2R mRNA downregulation after 7 days restraint, in line with lower plasma cortisol levels seen. We discuss regulation of corticosteroid production from a phylogenetic perspective. We propose that increased levels of alpha-MSH exert a positive feedback on hypothalamic corticotropin-releasing hormone release to sustain a mild stress axis activity.


Asunto(s)
Hormona Adrenocorticotrópica/metabolismo , Hidrocortisona/metabolismo , Receptor de Melanocortina Tipo 2/metabolismo , Receptores de Corticotropina/metabolismo , Estrés Fisiológico/metabolismo , alfa-MSH/metabolismo , Hormona Adrenocorticotrópica/farmacología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Carpas , Clonación Molecular , Hidrocortisona/sangre , Riñón/metabolismo , Datos de Secuencia Molecular , Filogenia , Receptor de Melanocortina Tipo 2/genética , Receptores de Corticotropina/genética , Receptores de Melanocortina , Restricción Física , Estrés Fisiológico/etiología , Distribución Tisular , alfa-MSH/sangre , alfa-MSH/farmacología
15.
J Neurophysiol ; 93(5): 2849-55, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15615828

RESUMEN

Spatial and temporal ambient temperature variations directly influence cellular biochemistry and thus the physiology of ectotherms. However, many aquatic ectothermic species maintain coordinated sensorimotor function during large acute body-temperature changes, which points to a compensatory mechanism within the neural system. Here we used high-resolution functional magnetic resonance imaging to study brain responses to a drop of 10 degrees C of ambient water temperature in common carp. We observed a strong drainage of blood out of the brain as of 90 s after the onset of the temperature drop, which would be expected to reduce entry of cold blood arriving from the gills so that the change in brain temperature would be slower. Although oxygen content in the brain thus decreased, we still found specific activation in the preoptic area (involved in temperature detection and stress responses), the pituitary pars distalis (stress response), and inactivation of the anterior part of the midbrain tegmentum and the pituitary pars intermedia. We propose that the blood drainage from the brain slows down the cooling of the brain during an acute temperature drop. This could help to maintain proper brain functioning including sensorimotor activity, initiation of the stress response, and the subsequent behavioral responses.


Asunto(s)
Volumen Sanguíneo/fisiología , Encéfalo/fisiopatología , Estrés Fisiológico/fisiopatología , Temperatura , Animales , Encéfalo/irrigación sanguínea , Mapeo Encefálico , Carpas , Imagen por Resonancia Magnética/métodos , Oxígeno/sangre , Hipófisis/irrigación sanguínea , Hipófisis/fisiología , Tiempo de Reacción , Sensación Térmica , Factores de Tiempo
16.
J Exp Biol ; 206(Pt 13): 2273-80, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12771175

RESUMEN

Isogenic carp Cyprinus carpio L. were acclimated to water temperatures of 15, 22 and 29 degrees C for at least 8 weeks. The acclimations consistently resulted in slightly, but significantly, different plasma osmolality, sodium, potassium and chloride concentrations between the groups studied. Plasma total and ionic calcium levels were unaffected, indicating successful adaptation. The apparent changes in set point for plasma ion levels are explained by altered sodium pump activity and hormonal control of branchial permeability to water and ions. It appears that in 15 degrees C-acclimated fish, a lower apparent Na(+)/K(+)-ATPase activity is compensated by strongly enhanced Na(+)/K(+)-ATPase expression (determined biochemically and immunohistochemically). In 29 degrees C-acclimated fish, the higher ambient temperature activates the enzyme. Arrhenius plots for branchial Na(+)/K(+)-ATPase preparations of the three groups of fish suggest the occurrence of different enzyme isoforms or protein (in)stability as explanations for differences in apparent enzyme activities, rather than temperature-dependent changes in membrane fluidity. As for hormonal control over permeability, prolactin mRNA expression (and anticipated production and release) is lower in fish kept at 29 degrees C, suggesting that control over branchial permeability to water and ions needs to be downregulated at higher temperatures. In so doing, enhanced sodium pump activity is balanced by a controlled passive ion loss to fine-tune plasma sodium levels. Basal plasma cortisol levels did not correlate positively with Na(+)/K(+)-ATPase expression, but doubling plasma cortisol levels in control fish by administering exogenous cortisol (for 7 days, using implanted minipumps and thus stress-free) enhanced Na(+)/K(+)-ATPase expression. This effect must be the result of a glucocorticoid action of the steroid: in fish, mineralocorticoid receptors have higher affinity for cortisol than glucocorticoid receptors. At a lower ambient temperature, branchial Na(+)/K(+)-ATPase expression is upregulated to counteract the temperature-inhibited activity of the sodium pump, perhaps via a mineralocorticoid receptor.


Asunto(s)
Aclimatación/fisiología , Carpas/metabolismo , Branquias/enzimología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Calcio/sangre , Carpas/fisiología , Cloro/sangre , Activación Enzimática , Regulación de la Expresión Génica , Hidrocortisona/farmacología , Concentración Osmolar , Potasio/sangre , Prolactina/genética , Sodio/sangre , ATPasa Intercambiadora de Sodio-Potasio/efectos de los fármacos , Temperatura , Regulación hacia Arriba/fisiología
17.
Neuroendocrinology ; 77(1): 15-23, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12624537

RESUMEN

This study investigates whether thyrotropin-releasing hormone (TRH), alpha-melanocyte-stimulating hormone (alpha-MSH) and N-acetyl beta-endorphin (NAc beta-END), or the thyroid hormones thyroxine (T4) and 3,5,3'-triiodothyronine (T3) are involved in the physiological response to temperature changes in the poikilotherm common carp (CYPRINUS CARPIO). Carps were either subjected to a rapid cold exposure or acclimated over time to three different temperatures. Acute cold exposure did not influence blood plasma alpha-MSH concentrations. Acclimation to 15, 22 or 29 degrees C led to a temperature-dependent increase of both alpha-MSH and NAc beta-END plasma concentrations. Moreover, the in vitro sensitivity to TRH of melanotrope cells (that synthesise these peptides) also correlated positively with ambient temperature. Increased TRH activation stimulated processing of the precursor of alpha-MSH and NAc beta-END, resulting in increased release of both peptides and storage of a surplus of NAc beta-END within melanotropes. Plasma T4 levels were highest in carps acclimated to the intermediate temperature tested, and correlated strongly with hypothalamic TRH content. Plasma T3 levels were unaffected by ambient water temperature. We conclude that ambient water temperature influences the sensitivity of melanotrope cells to TRH in carps. This effect, however, is not due to acute temperature change, but evolves during the acclimation process of carps to a new temperature.


Asunto(s)
Carpas/fisiología , Hipófisis/fisiología , Temperatura , Hormona Liberadora de Tirotropina/metabolismo , Tiroxina/sangre , Triyodotironina/sangre , alfa-MSH/metabolismo , betaendorfina/análogos & derivados , betaendorfina/metabolismo , Adaptación Fisiológica/fisiología , Animales , Hidrocortisona/sangre , Masculino , Hipófisis/citología , alfa-MSH/análisis , betaendorfina/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA