Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Exp Biol ; 226(5)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36805713

RESUMEN

Corticotropin-releasing hormone (CRH) neurons are the primary neural population controlling the hypothalamic-pituitary-adrenal (HPA) axis and the secretion of adrenal stress hormones. Previous work has demonstrated that stress hormone secretion can be regulated by circulating levels of estradiol. However, the effect of estradiol on CRH neuron excitability is less clear. Here, we show that chronic estradiol replacement following ovariectomy increases two types of potassium channel currents in CRH neurons: fast inactivating voltage-gated A-type K+ channel currents (IA) and non-inactivating M-type K+ channel currents (IM). Despite the increase in K+ currents following estradiol replacement, there was no overall change in CRH neuron spiking excitability assessed with either frequency-current curves or current ramps. Together, these data reveal a complex picture whereby ovariectomy and estradiol replacement differentially modulate distinct aspects of CRH neuron and HPA axis function.


Asunto(s)
Hormona Liberadora de Corticotropina , Sistema Hipotálamo-Hipofisario , Femenino , Humanos , Hormona Liberadora de Corticotropina/farmacología , Sistema Hipotálamo-Hipofisario/metabolismo , Estradiol/farmacología , Estradiol/fisiología , Sistema Hipófiso-Suprarrenal/metabolismo , Neuronas/fisiología
2.
J Physiol ; 600(19): 4347-4359, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36040213

RESUMEN

Noradrenaline is a neurotransmitter released in response to homeostatic challenge and activates the hypothalamic-pituitary-adrenal axis via stimulation of corticotropin-releasing hormone (CRH) neurons. Here we investigated the mechanism through which noradrenaline regulates activity within the CRH neuronal network. Using a combination of in vitro GCaMP6f Ca2+ imaging and electrophysiology, we show that noradrenaline induces a robust increase in excitability in a proportion of CRH neurons with many neurons displaying a bursting mode of activity. Noradrenaline-induced activation required α1 -adrenoceptors and L-type voltage-gated Ca2+ channels, but not GABA/glutamate synaptic transmission or sodium action potentials. Exposure of mice to elevated corticosterone levels was able to suppress noradrenaline-induced activation. These results provide further insight into the mechanisms by which noradrenaline regulates CRH neural network activity and hence stress responses. KEY POINTS: GCaMP6f Ca2+ imaging and on-cell patch-clamp recordings reveal that corticotropin-releasing hormone neurons are activated by noradrenaline with many neurons displaying a bursting mode of activity. Noradrenaline-induced activation requires α1 -adrenoceptors. Noradrenaline-induced Ca2+ elevations persist after blocking GABAA , AMPA, NMDA receptors and voltage-gated Na+ channels. Noradrenaline-induced Ca2+ elevations require L-type voltage-gated Ca2+ channels. Corticosterone suppresses noradrenaline-induced excitation.


Asunto(s)
Hormona Liberadora de Corticotropina , Sistema Hipotálamo-Hipofisario , Animales , Corticosterona/farmacología , Hormona Liberadora de Corticotropina/metabolismo , Glutamatos , Sistema Hipotálamo-Hipofisario/fisiología , Ratones , Neuronas/fisiología , Norepinefrina/farmacología , Sistema Hipófiso-Suprarrenal/fisiología , Receptores Adrenérgicos alfa 1 , Receptores de N-Metil-D-Aspartato , Sodio , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico
3.
J Physiol ; 600(7): 1753-1770, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35045190

RESUMEN

The hormone, oxytocin, is synthesised by magnocellular neurones of the supraoptic and paraventricular nuclei and is released from the posterior pituitary gland into the circulation to trigger uterine contractions during parturition. Kisspeptin fibre density increases around the supraoptic nucleus over pregnancy and intracerebroventricular kisspeptin excites oxytocin neurones only in late pregnancy. However, the mechanism of this excitation is unknown. Here, we found that microdialysis administration of kisspeptin into the supraoptic nucleus consistently increased the action potential (spike) firing rate of oxytocin neurones in urethane-anaesthetised late-pregnant rats (gestation day 18-21) but not in non-pregnant rats. Hazard analysis of action potential firing showed that kisspeptin specifically increased the probability of another action potential firing immediately after each action potential (post-spike excitability) in late-pregnant rats. Patch-clamp electrophysiology in hypothalamic slices showed that bath application of kisspeptin did not affect action potential frequency or baseline membrane potential in supraoptic nucleus neurones. Moreover, kisspeptin superfusion did not affect the frequency or amplitude of excitatory postsynaptic currents or inhibitory postsynaptic currents in supraoptic nucleus neurones. Taken together, these studies suggest that kisspeptin directly activates oxytocin neurones in late pregnancy, at least in part, via increased post-spike excitability. KEY POINTS: Oxytocin secretion is triggered by action potential firing in magnocellular neurones of the hypothalamic supraoptic and paraventricular nuclei to induce uterine contractions during birth. In late pregnancy, kisspeptin expression increases in rat periventricular nucleus neurones that project to the oxytocin system. Here, we show that intra-supraoptic nucleus administration of kisspeptin increases the action potential firing rate of oxytocin neurones in anaesthetised late-pregnant rats, and that the increased firing rate is associated with increased oxytocin neurone excitability immediately after each action potential. By contrast, kisspeptin superfusion of hypothalamic slices did not affect the activity of supraoptic nucleus neurones or the strength of local synaptic inputs to supraoptic nucleus neurones. Hence, kisspeptin might activate oxytocin neurons in late pregnancy by transiently increasing oxytocin neuron excitability after each action potential.


Asunto(s)
Kisspeptinas , Oxitocina , Potenciales de Acción/fisiología , Animales , Femenino , Kisspeptinas/metabolismo , Kisspeptinas/farmacología , Neuronas/fisiología , Oxitocina/metabolismo , Embarazo , Ratas , Núcleo Supraóptico/fisiología , Vasopresinas/metabolismo
4.
Int J Mol Sci ; 23(14)2022 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-35887077

RESUMEN

Oxytocin is secreted by hypothalamic supraoptic nucleus (SON) and paraventricular nucleus (PVN) oxytocin neurons to induce uterine contractions during parturition. Increased activation of oxytocin neurons at parturition involves a network of afferent inputs that increase oxytocin neuron excitability. Kisspeptin fibre density increases around oxytocin neurons during pregnancy, and central kisspeptin administration excites oxytocin neurons only in late pregnancy. Kisspeptin signals via extracellular regulated kinase 1/2 (ERK1/2) and p38. Therefore, to determine whether kisspeptin excites oxytocin neurons via ERK1/2-p38 signalling in late-pregnant rats, we performed immunohistochemistry for phosphorylated ERK1/2 (pERK1/2) and phosphorylated p38 (p-p38) in oxytocin neurons of non-pregnant and late-pregnant rats. Intracerebroventricular (ICV) kisspeptin administration (2 µg) did not affect pERK1/2 or p-p38 expression in SON and PVN oxytocin neurons of non-pregnant or late-pregnant rats. Furthermore, ICV kisspeptin did not affect pERK1/2 or p-p38 expression in brain areas with major projections to the SON and PVN: the nucleus tractus solitarius, rostral ventrolateral medulla, locus coeruleus, dorsal raphe nucleus, organum vasculosum of the lamina terminalis, median preoptic nucleus, subfornical organ, anteroventral periventricular nucleus, periventricular nucleus and arcuate nucleus. Hence, kisspeptin-induced excitation of oxytocin neurons in late pregnancy does not appear to involve ERK1/2 or p38 activation in oxytocin neurons or their afferent inputs.


Asunto(s)
Kisspeptinas , Oxitocina , Animales , Femenino , Kisspeptinas/metabolismo , Kisspeptinas/farmacología , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/metabolismo , Oxitocina/metabolismo , Fosforilación , Embarazo , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Proc Natl Acad Sci U S A ; 115(5): E1041-E1050, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29339520

RESUMEN

Emotional responses, such as fear and anxiety, are fundamentally important behavioral phenomena with strong fitness components in most animal species. Anxiety-related disorders continue to represent a major unmet medical need in our society, mostly because we still do not fully understand the mechanisms of these diseases. Animal models may speed up discovery of these mechanisms. The zebrafish is a highly promising model organism in this field. Here, we report the identification of a chemokine-like gene family, samdori (sam), and present functional characterization of one of its members, sam2 We show exclusive mRNA expression of sam2 in the CNS, predominantly in the dorsal habenula, telencephalon, and hypothalamus. We found knockout (KO) zebrafish to exhibit altered anxiety-related responses in the tank, scototaxis and shoaling assays, and increased crh mRNA expression in their hypothalamus compared with wild-type fish. To investigate generalizability of our findings to mammals, we developed a Sam2 KO mouse and compared it to wild-type littermates. Consistent with zebrafish findings, homozygous KO mice exhibited signs of elevated anxiety. We also found bath application of purified SAM2 protein to increase inhibitory postsynaptic transmission onto CRH neurons of the paraventricular nucleus. Finally, we identified a human homolog of SAM2, and were able to refine a candidate gene region encompassing SAM2, among 21 annotated genes, which is associated with intellectual disability and autism spectrum disorder in the 12q14.1 deletion syndrome. Taken together, these results suggest a crucial and evolutionarily conserved role of sam2 in regulating mechanisms associated with anxiety.


Asunto(s)
Ansiedad/genética , Trastorno del Espectro Autista/genética , Quimiocinas/genética , Miedo , Mutación , Animales , Trastornos de Ansiedad , Conducta Animal , Condicionamiento Psicológico/fisiología , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Variación Genética , Proteínas Fluorescentes Verdes/metabolismo , Homocigoto , Humanos , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Conducta Social , Pez Cebra
6.
Proc Natl Acad Sci U S A ; 114(47): E10216-E10223, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109258

RESUMEN

The pulsatile release of luteinizing hormone (LH) is critical for mammalian fertility. However, despite several decades of investigation, the identity of the neuronal network generating pulsatile reproductive hormone secretion remains unproven. We use here a variety of optogenetic approaches in freely behaving mice to evaluate the role of the arcuate nucleus kisspeptin (ARNKISS) neurons in LH pulse generation. Using GCaMP6 fiber photometry, we find that the ARNKISS neuron population exhibits brief (∼1 min) synchronized episodes of calcium activity occurring as frequently as every 9 min in gonadectomized mice. These ARNKISS population events were found to be near-perfectly correlated with pulsatile LH secretion. The selective optogenetic activation of ARNKISS neurons for 1 min generated pulses of LH in freely behaving mice, whereas inhibition with archaerhodopsin for 30 min suppressed LH pulsatility. Experiments aimed at resetting the activity of the ARNKISS neuron population with halorhodopsin were found to reset ongoing LH pulsatility. These observations indicate the ARNKISS neurons as the long-elusive hypothalamic pulse generator driving fertility.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante/metabolismo , Red Nerviosa/metabolismo , Neuronas/metabolismo , Potenciales de Acción , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/diagnóstico por imagen , Femenino , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Optogenética/métodos , Periodicidad , Fotometría/métodos , Imagen de Colorante Sensible al Voltaje
7.
J Neurosci ; 37(12): 3342-3351, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28235895

RESUMEN

Fast cell-to-cell communication in the brain is achieved by action potential-dependent synaptic release of neurotransmitters. The fast kinetics of transmitter release are determined by transient Ca2+ elevations in presynaptic nerve terminals. Neuromodulators have previously been shown to regulate transmitter release by inhibiting presynaptic Ca2+ influx. Few studies to date have demonstrated the opposite, that is, neuromodulators directly driving presynaptic Ca2+ rises and increases in nerve terminal excitability. Here we use GCaMP Ca2+ imaging in brain slices from mice to address how nerve terminal Ca2+ is controlled in gonadotropin-releasing hormone (GnRH) neurons via action potentials and neuromodulators. Single spikes and bursts of action potentials evoked fast, voltage-gated Ca2+ channel-dependent Ca2+ elevations. In contrast, brief exposure to the neuropeptide kisspeptin-evoked long-lasting Ca2+ plateaus that persisted for tens of minutes. Neuropeptide-mediated Ca2+ elevations were independent of action potentials, requiring Ca2+ entry via voltage-gated Ca2+ channels and transient receptor potential channels in addition to release from intracellular store mechanisms. Together, these data reveal that neuromodulators can exert powerful and long-lasting regulation of nerve terminal Ca2+ independently from actions at the soma. Thus, GnRH nerve terminal function is controlled over disparate timescales via both classical spike-dependent and nonclassical neuropeptide-dependent mechanisms.SIGNIFICANCE STATEMENT Nerve terminals are highly specialized regions of a neuron where neurotransmitters and neurohormones are released. Many neuroendocrine neurons release neurohormones in long-duration bursts of secretion. To understand how this is achieved, we have performed live Ca2+ imaging in the nerve terminals of gonadotropin-releasing hormone neurons. We find that bursts of action potentials and local neuropeptide signals are both capable of evoking large increases in nerve terminal Ca2+ Increases in Ca2+ driven by spike bursts last seconds; however, the increases in nerve terminal Ca2+ driven by neuropeptides can persist for tens of minutes. These findings reveal new mechanisms by which neuroendocrine nerve terminal Ca2+ can be controlled in the brain.


Asunto(s)
Potenciales de Acción/fisiología , Señalización del Calcio/fisiología , Calcio/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Neuropéptidos/metabolismo , Terminales Presinápticos/fisiología , Animales , Células Cultivadas , Femenino , Masculino , Ratones , Plasticidad Neuronal/fisiología , Factores de Tiempo
8.
J Physiol ; 594(4): 1005-15, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26662615

RESUMEN

KEY POINTS: Many excitatory synapses in the brain release glutamate with both synchronous and asynchronous components. Immediately following an action potential, neurons display a reduced excitability due to the post-spike afterhyperpolarization (AHP). This gives rise to a relative refractory period. When an action potential is evoked by glutamate synaptic input possessing asynchronous release, the delayed glutamate release events act to depolarize the neuron during the AHP and overcome the relative refractory period. These results demonstrate a new role for asynchronous release in regulating post-spike excitability and the relative refractory period in central neurons. ABSTRACT: Post-spike afterhyperpolarizations (AHPs) functionally inhibit neuronal excitability for tens to hundreds of milliseconds following each action potential. This imposes a relative refractory period during which synaptic excitation is less effective at evoking spikes. Here we asked whether some synapses have mechanisms in place that allow them to overcome the AHP and drive spiking in target cells during this period of reduced excitability. We examined glutamate synapses onto oxytocin and vasopressin neurons in the paraventricular nucleus of the hypothalamus. These synapses can display pronounced asynchronous glutamate release following a single presynaptic spike, with the time course of release being similar to that of the post-spike AHP. To test whether asynchronous release is more effective at overcoming the relative refractory period, we evoked a single action potential with either a brief synchronous depolarization or an asynchronous potential and then assessed excitability at multiple time points following the spike. Neurons receiving asynchronous depolarizing synaptic inputs had a shorter relative refractory period than those receiving synchronous depolarizations. Our data demonstrate that synapses releasing glutamate in an asynchronous and delayed manner are ideally adapted to counter the AHP. By effectively overcoming the relative refractory period, the kinetics of excitatory synaptic input can play an important role in controlling post-spike excitability.


Asunto(s)
Potenciales Postsinápticos Excitadores , Ácido Glutámico/metabolismo , Terminales Presinápticos/metabolismo , Periodo Refractario Electrofisiológico , Animales , Exocitosis , Masculino , Neuronas/metabolismo , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/fisiología , Terminales Presinápticos/fisiología , Ratas , Ratas Sprague-Dawley
9.
Neuroendocrinology ; 102(1-2): 1-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25300776

RESUMEN

Gonadotropin-releasing hormone (GnRH) neurons integrate synaptic information in their dendrites in order to precisely control GnRH secretion and hence fertility. Recent discoveries concerning the structure and function of GnRH neuron dendrites have shed new light on the control of GnRH neuron excitability and GnRH secretion. This work suggests that GnRH neurons have a unique projection to the median eminence that possesses both dendritic and axonal properties. We propose that this 'dendron' projection allows GnRH neurons to multitask and integrate information in ways that would not be possible in a classically envisioned axon projection.


Asunto(s)
Dendritas/fisiología , Hormona Liberadora de Gonadotropina/fisiología , Neuronas/fisiología , Animales , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Eminencia Media/citología , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Neuronas/citología
10.
J Neurosci ; 33(22): 9394-401, 2013 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-23719807

RESUMEN

The gonadotropin-releasing hormone (GnRH) neurons are the key cells regulating fertility in all mammalian species. The scattered distribution of these neurons has made investigation of their properties extremely difficult and the key goal of recording their electrical activity in vivo near impossible. The caudal-most extension of the GnRH neuron continuum brings some cells very close to the base of the brain at the level of the anterior hypothalamic area. Taking insight from this, we developed an experimental procedure in anesthetized GnRH-GFP mice that allows the electrical activity of these GnRH neurons to be recorded in vivo. On-cell recordings revealed that the majority of GnRH neurons (86%) were spontaneously active, exhibiting a range of firing patterns, although only a minority (15%) exhibited burst firing. Mean firing frequencies ranged from 0.06 to 3.65 Hz, with the most common interspike interval being ~500 ms. All GnRH neurons tested were activated by AMPA and kisspeptin. Whereas the GABAA receptor agonist muscimol evoked excitatory, inhibitory, or mixed effects on GnRH neuron firing, the GABAA receptor antagonist picrotoxin resulted in a consistent suppression of firing. These observations represent the first electrical recordings of GnRH neurons in vivo. They reveal that GnRH neurons in vivo exhibit considerable heterogeneity in their firing patterns with both similarities and differences to firing in vitro. These variable patterns of firing in vivo are found to be critically dependent upon ongoing GABAA receptor signaling.


Asunto(s)
Hormona Liberadora de Gonadotropina/fisiología , Neuronas/fisiología , Receptores de GABA-A/fisiología , Transducción de Señal/fisiología , Animales , Electrodos Implantados , Fenómenos Electrofisiológicos/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Kisspeptinas/farmacología , Masculino , Ratones , Muscimol/farmacología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Pentobarbital/farmacología , Faringe/inervación , Faringe/fisiología , Picrotoxina/farmacología , Receptores de GABA-A/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tetrodotoxina/farmacología , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
11.
J Neurosci ; 33(31): 12689-97, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23904605

RESUMEN

Information processing by neurons has been traditionally envisioned to occur in discrete neuronal compartments. Specifically, dendrites receive and integrate synaptic inputs while axons initiate and conduct spikes to distal neuronal targets. We report here in mice, using morphological reconstructions and electrophysiology, that the gonadotropin-releasing hormone (GnRH) neurons that control mammalian fertility do not conform to this stereotype and instead possess a single projection structure that functions simultaneously as an axon and dendrite. Specifically, we show that the GnRH neuron projection to the median eminence to control pituitary hormone secretion possesses a spike initiation site and conducts action potentials while also exhibiting spines and synaptic appositions along its entire length. Classical axonal or dendritic markers are not detectable in the projection process. Activation of ionotropic glutamate and/or GABA receptors along the GnRH neuron projection is capable of depolarizing the membrane potential and initiating action potentials. In addition, focal glutamate application to the projection is able to regulate the width of propagating spikes. These data demonstrate that GnRH neurons elaborate a previously uncharacterized neuronal projection that functions simultaneously as an axon and dendrite. This structure, termed a "dendron," greatly expands the dynamic control of GnRH secretion into the pituitary portal system to regulate fertility.


Asunto(s)
Axones/fisiología , Dendritas/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/citología , Neuronas/metabolismo , Animales , Biotina/metabolismo , Vasos Sanguíneos/metabolismo , Channelrhodopsins , GABAérgicos/farmacología , Galectina 1/metabolismo , Ácido Glutámico/farmacología , Hormona Liberadora de Gonadotropina/genética , Proteínas Fluorescentes Verdes/genética , Técnicas In Vitro , Masculino , Eminencia Media/citología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Estimulación Luminosa , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , Proteínas tau/metabolismo
12.
J Neurosci ; 32(1): 151-8, 2012 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-22219278

RESUMEN

Gonadotropin-releasing hormone (GnRH) neurons are the final output neurons in a complex neuronal network that regulates fertility. The morphology of GnRH neuron dendrites is very different to other central neurons in that they are very long, thin, and unbranched. To study the function of these dendrites, we have used Na(+) and Ca(2+) imaging in combination with dual soma and dendrite electrical recordings in brain slices from GnRH-GFP mice. Here, we show that GnRH neurons actively propagate Na(+) spikes throughout their dendrites. Multisite dendritic recordings confirmed that these spikes were observed in one of the dendrites before the soma in the great majority of neurons tested. Na(+) imaging experiments revealed that the initial 150 µm of dendrite has a higher density of functional Na(+) channels than more distal regions, suggesting that this region of dendrite is highly excitable and may be the site of spike initiation. Finally, we show that the depolarization from dendritic spikes opens voltage-gated Ca(2+) channels giving rise to dendritic Ca(2+) transients. Together, these findings suggest that the proximal dendrites of GnRH neurons are highly excitable and are likely to be the site of action potential initiation in these neurons.


Asunto(s)
Potenciales de Acción/fisiología , Dendritas/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/fisiología , Área Preóptica/fisiología , Animales , Canales de Calcio/fisiología , Señalización del Calcio/fisiología , Dendritas/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/metabolismo , Técnicas de Cultivo de Órganos , Área Preóptica/citología , Área Preóptica/metabolismo , Canales de Sodio/fisiología
13.
J Neurosci ; 31(33): 12011-20, 2011 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-21849561

RESUMEN

Endocannabinoids (eCBs) are feedback messengers in the nervous system that act at the presynaptic nerve terminal to inhibit transmitter release. Here we report that in brain slices from rat, eCBs are released from vasopressin (VP) neurons in the paraventricular nucleus of the hypothalamus following coincident bursts of presynaptic and postsynaptic spiking. eCBs transiently depress glutamate release from excitatory terminals and, in doing so, prevent the synapses from undergoing long-term depression (LTD). Specifically, we show that blockade of CB1 receptors unmasks LTD following coincident presynaptic and postsynaptic activity. This LTD is presynaptic in nature, but requires the release of the opioid peptide dynorphin from the postsynaptic neuron. Dynorphin release and subsequent LTD require the activation of postsynaptic metabotropic glutamate receptors (mGluRs). Our findings indicate that eCBs, by transiently depressing glutamate release, limit mGluR activation and indirectly gate release of dynorphin from the postsynaptic neuron. We propose that eCBs, in addition to their well described role in the rapid modulation of transmitter release from the nerve terminal, also regulate the release of other retrograde transmitters and thus encode for multiple temporal windows of synaptic plasticity.


Asunto(s)
Moduladores de Receptores de Cannabinoides/fisiología , Endocannabinoides , Potenciales Postsinápticos Excitadores/fisiología , Transmisión Sináptica/fisiología , Animales , Moduladores de Receptores de Cannabinoides/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Masculino , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Piperidinas/farmacología , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Transmisión Sináptica/efectos de los fármacos
14.
J Neuroendocrinol ; 34(4): e13084, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35034400

RESUMEN

Activity-dependent release of retrograde signaling molecules form micro-feedback loops to regulate synaptic function in neural circuits. Single neurons can release multiple forms of these signaling molecules, including endocannabinoids and endovanilloids, which act via cannabinoid (CB) receptors and transient receptor potential vanilloid 1 (TRPV1) receptors. In hypothalamic corticotrophin-releasing hormone (CRH) neurons, endocannabinoids acting via CB1 receptors have been shown to play an important role in regulating excitability and hence stress hormone secretion. However, the importance of endovanilloid signaling in CRH neurons is currently unclear. Here, we show that, in response to postsynaptic depolarization, CRH neurons release endocannabinoid/endovanilloid molecules that can activate CB1 and TRPV1 receptors. Activation of CB1 receptors suppresses glutamate neurotransmission whereas activation of TRPV1 enhances spontaneous glutamate transmission. However, the excitatory effects of TRPV1 are normally masked by the inhibitory effects of CB1. When the degradation of the endocannabinoid 2-arachidonoylglycerol (2-AG) was inhibited, this revealed tonic activation of CB1 receptors, suggesting tonic endocannabinoid release. However, we found no evidence for tonic activation of TRPV1 receptors under similar conditions. These findings show that activation of CRH neurons can drive the release of signaling molecules that activate parallel endocannabinoid and endovanilloid receptor pathways to mediate opposing forms of synaptic plasticity.


Asunto(s)
Cannabinoides , Endocannabinoides , Cannabinoides/farmacología , Hormona Liberadora de Corticotropina/metabolismo , Ácido Glutámico/metabolismo , Plasticidad Neuronal , Neuronas/metabolismo , Canales Catiónicos TRPV/metabolismo
15.
Front Cell Neurosci ; 16: 958876, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36090787

RESUMEN

Neuronal hyperexcitability in Alzheimer's disease (AD) models is thought to either contribute to the formation of amyloid beta plaques or result from their formation. Neuronal hyperexcitability has been shown in the cerebral cortex of the widely used young APPswe/PS1dE9 mice, which have accelerated plaque formation. However, it is currently unclear if hyperexcitability also occurs in CA1 hippocampal neurons of aged animals in this model. In the present work, we have compared intrinsic excitability and spontaneous synaptic inputs from CA1 pyramidal cells of 8-month-old APPswe/PS1dE9 and wildtype control mice. We find no change in intrinsic excitability or spontaneous postsynaptic currents (PSCs) between groups. We did, however, find a reduced input resistance and an increase in hyperpolarization-activated sag current. These results are consistent with findings from other aged AD model mice, including the widely used 5xFAD and 3xTg. Together these results suggest that neuronal hyperexcitability is not a consistent feature of all AD mouse models, particularly at advanced ages.

16.
Front Neuroendocrinol ; 31(3): 296-306, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20347860

RESUMEN

Glutamate synapses drive the output of neuroendocrine cells in the hypothalamus, but until recently, relatively little was known about the fundamental properties of transmission at these synapses. Here we review recent advances in the understanding of glutamate signals in magnocellular neurosecretory cells (MNCs) in the paraventricular (PVN) and supraoptic nuclei (SON) of the hypothalamus that serve as the last step in synaptic integration before neurohormone release. While these synapses exhibit many similarities with other glutamate synapses described throughout the brain, they also exhibit a number of unique properties that are particularly well suited to the physiology of this system and will be discussed here. In addition, a number of recent studies begin to provide insights into new forms of synaptic plasticity that may be common in other brain regions, but in these cells, may serve important adaptive roles.


Asunto(s)
Ácido Glutámico/metabolismo , Células Neuroendocrinas/metabolismo , Células Neuroendocrinas/fisiología , Plasticidad Neuronal/fisiología , Transmisión Sináptica/fisiología , Potenciales de Acción/fisiología , Animales , Humanos , Modelos Biológicos , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/fisiología
17.
Sci Rep ; 11(1): 16700, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34404890

RESUMEN

Stress responses are highly plastic and vary across physiological states. The female estrous cycle is associated with a number of physiological changes including changes in stress responses, however, the mechanisms driving these changes are poorly understood. Corticotropin-releasing hormone (CRH) neurons are the primary neural population controlling the hypothalamic-pituitary-adrenal (HPA) axis and stress-evoked corticosterone secretion. Here we show that CRH neuron intrinsic excitability is regulated over the estrous cycle with a peak in proestrus and a nadir in estrus. Fast inactivating voltage-gated potassium channel (IA) currents showed the opposite relationship, with current density being lowest in proestrus compared to other cycle stages. Blocking IA currents equalized excitability across cycle stages revealing a role for IA in mediating plasticity in stress circuit function over the female estrous cycle.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Ciclo Estral , Hipotálamo/fisiología , Neuronas/metabolismo , Animales , Femenino , Hipotálamo/citología , Ratones , Plasticidad Neuronal , Neuronas/citología , Sistema Hipófiso-Suprarrenal/fisiología , Estrés Fisiológico
18.
J Neuroendocrinol ; 33(11): e13012, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34289195

RESUMEN

During parturition and lactation, oxytocin neurones in the supraoptic and paraventricular nuclei fire high-frequency bursts of action potentials that are coordinated across the entire population. Each burst generates a large pulse of oxytocin release into the circulation to induce uterine contraction for parturition and mammary duct contraction for milk ejection. Bursts are stimulated by cervical stretch during parturition and by suckling during lactation. However, the mechanisms by which these stimuli are translated into episodic bursts are poorly understood, as are the mechanisms that coordinate bursts across the oxytocin neurone population. An elegant series of experiments conducted in the 1980s and 1990s used serial paired recordings to show that oxytocin neurones do not act as a syncytium during bursts; rather, they start each burst within a few hundred milliseconds of each other but with no distinct "leaders" or "followers". In addition to afferent noradrenergic inputs that relay the systemic stimuli to oxytocin neurones, bursts depend on somato-dendritic oxytocin release within the hypothalamus. Hence, bursts are considered to be an emergent property of oxytocin neurones that is bootstrapped by appropriate afferent stimulation. Although much progress was made using traditional electrophysiological recordings in head-fixed anaesthetised animals, research has effectively stalled in the last few decades. However, the emergence of new technologies to monitor neuronal activity in freely-behaving animals has reinvigorated efforts to understand the biology underpinning burst firing in oxytocin neurones. Here, we report the use of fibre photometry to monitor the dynamics of milk ejection bursts in the oxytocin neurone population of freely-behaving mice. This approach will shed light on the neural mechanisms that control the oxytocin bursts underpinning parturition and lactation.


Asunto(s)
Eyección Láctea , Oxitocina , Potenciales de Acción , Animales , Femenino , Lactancia/fisiología , Ratones , Oxitocina/fisiología , Parto , Embarazo , Núcleo Supraóptico/fisiología
19.
J Neuroendocrinol ; 33(5): e12972, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33896057

RESUMEN

Chronic stress exerts multiple negative effects on the physiology and health of an individual. In the present study, we examined hypothalamic, pituitary and endocrine responses to 14 days of chronic variable stress (CVS) in male and female C57BL/6J mice. In both sexes, CVS induced a significant decrease in body weight and enhanced the acute corticosterone stress response, which was accompanied by a reduction in thymus weight only in females. However, single-point blood measurements of basal prolactin, thyroid-stimulating hormone, luteinising hormone, growth hormone and corticosterone levels taken at the end of the CVS were not different from those of controls. Similarly, pituitary mRNA expression of Fshb, Lhb, Prl and Gh was unchanged by CVS, although Pomc and Tsh were significantly elevated. Within the adrenal medulla, mRNA for Th, Vip and Gal were elevated following CVS. Avp transcript levels within the paraventricular nucleus of the hypothalamus were increased by CVS; however, levels of Gnrh1, Crh, Oxt, Sst, Trh, Ghrh, Th and Kiss1 remained unchanged. Oestrous cycles were lengthened slightly by CVS and ovarian histology revealed a reduction in the number of preovulatory follicles and corpora lutea. Taken together, these observations indicate that 14 days of CVS induces an up-regulation of the neuroendocrine stress axis and creates a mild disruption of female reproductive function. However, the lack of changes in other neuroendocrine axes controlling anterior and posterior pituitary secretion suggest that most neuroendocrine axes are relatively resilient to CVS.


Asunto(s)
Hipotálamo/metabolismo , Folículo Ovárico/metabolismo , Hipófisis/metabolismo , Proopiomelanocortina/metabolismo , Estrés Psicológico/metabolismo , Animales , Cuerpo Lúteo/metabolismo , Corticosterona/metabolismo , Femenino , Hormona del Crecimiento/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Prolactina/metabolismo , Tirotropina/metabolismo
20.
J Neurosci ; 29(22): 7349-58, 2009 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-19494156

RESUMEN

Opioid signaling in the CNS is critical for controlling cellular excitability, yet the conditions under which endogenous opioid peptides are released and the precise mechanisms by which they affect synaptic transmission remain poorly understood. The opioid peptide dynorphin is present in the soma and dendrites of vasopressin neurons in the hypothalamus and dynamically controls the excitability of these cells in vivo. Here, we show that dynorphin is released from dendritic vesicles in response to postsynaptic activity and acts in a retrograde manner to inhibit excitatory synaptic transmission. This inhibition, which requires the activation of kappa-opioid receptors, results from a reduction in presynaptic release of glutamate vesicles. The opioid inhibition is downstream of Ca(2+) entry and is likely mediated by a direct modulation of presynaptic fusion machinery. These findings demonstrate that neurons may self-regulate their excitability through the dendritic release of opioids to inhibit excitatory synaptic transmission.


Asunto(s)
Calcio/metabolismo , Dinorfinas/metabolismo , Ácido Glutámico/metabolismo , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/citología , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología , 4-Aminopiridina/farmacología , Análisis de Varianza , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Bencenoacetamidas/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Dendritas/efectos de los fármacos , Dendritas/fisiología , Dinorfinas/genética , Estimulación Eléctrica/métodos , Inhibidores Enzimáticos/farmacología , Proteínas Fluorescentes Verdes/genética , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Masculino , Antagonistas de Narcóticos , Inhibición Neural/efectos de los fármacos , Vías Nerviosas/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Bloqueadores de los Canales de Potasio/farmacología , Pirrolidinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Opioides/agonistas , Receptores Opioides/fisiología , Transducción de Señal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , omega-Conotoxina GVIA/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA