Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Physiol ; 601(15): 3151-3171, 2023 08.
Article in English | MEDLINE | ID: mdl-36223200

ABSTRACT

Electrophysiological recordings can provide detailed information of single neurons' dynamical features and shed light on their response to stimuli. Unfortunately, rapidly modelling electrophysiological data for inferring network-level behaviours remains challenging. Here, we investigate how modelled single neuron dynamics leads to network-level responses in the paraventricular nucleus of the hypothalamus (PVN), a critical nucleus for the mammalian stress response. Recordings of corticotropin releasing hormone neurons from the PVN (CRHPVN ) were performed using whole-cell current-clamp. These, neurons, which initiate the endocrine response to stress, were rapidly and automatically fit to a modified adaptive exponential integrate-and-fire model (AdEx) with particle swarm optimization (PSO). All CRHPVN neurons were accurately fit by the AdEx model with PSO. Multiple sets of parameters were found that reliably reproduced current-clamp traces for any single neuron. Despite multiple solutions, the dynamical features of the models such as the rheobase, fixed points, and bifurcations, were shown to be stable across fits. We found that CRHPVN neurons can be divided into two subtypes according to their bifurcation at the onset of firing: CRHPVN -integrators and CRHPVN -resonators. The existence of CRHPVN -resonators was then directly confirmed in a follow-up patch-clamp hyperpolarization protocol which readily induced post-inhibitory rebound spiking in 33% of patched neurons. We constructed networks of CRHPVN model neurons to investigate the network level responses of CRHPVN neurons. We found that CRHPVN -resonators maintain baseline firing in networks even when all inputs are inhibitory. The dynamics of a small subset of CRHPVN neurons may be critical to maintaining a baseline firing tone in the PVN. KEY POINTS: Corticotropin-releasing hormone neurons (CRHPVN ) in the paraventricular nucleus of the hypothalamus act as the final neural controllers of the stress response. We developed a computational modelling platform that uses particle swarm optimization to rapidly and accurately fit biophysical neuron models to patched CRHPVN neurons. A model was fitted to each patched neuron without the use of dynamic clamping, or other procedures requiring sophisticated inputs and fitting algorithms. Any neuron undergoing standard current clamp step protocols for a few minutes can be fitted by this procedure The dynamical analysis of the modelled neurons shows that CRHPVN neurons come in two specific 'flavours': CRHPVN -resonators and CRHPVN -integrators. We directly confirmed the existence of these two classes of CRHPVN neurons in subsequent experiments. Network simulations show that CRHPVN -resonators are critical to retaining the baseline firing rate of the entire network of CRHPVN neurons as these cells can fire rebound spikes and bursts in the presence of strong inhibitory synaptic input.


Subject(s)
Corticotropin-Releasing Hormone , Paraventricular Hypothalamic Nucleus , Corticotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Neurons/physiology
2.
J Neurosci ; 40(46): 8842-8852, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33051356

ABSTRACT

In many species, social networks provide benefit for both the individual and the collective. In addition to transmitting information to others, social networks provide an emotional buffer for distressed individuals. Our understanding about the cellular mechanisms that contribute to buffering is poor. Stress has consequences for the entire organism, including a robust change in synaptic plasticity at glutamate synapses onto corticotropin-releasing hormone (CRH) neurons in the paraventricular nucleus of the hypothalamus (PVN). In females, however, this stress-induced metaplasticity is buffered by the presence of a naive partner. This buffering may be because of discrete behavioral interactions, signals in the context in which the interaction occurs (i.e., olfactory cues), or it may be influenced by local signaling events in the PVN. Here, we show that local vasopressin (VP) signaling in PVN buffers the short-term potentiation (STP) at glutamate synapses after stress. This social buffering of metaplasticity, which requires the presence of another individual, was prevented by pharmacological inhibition of the VP 1a receptor (V1aR) in female mice. Exogenous VP mimicked the effects of social buffering and reduced STP in CRHPVN neurons from females but not males. These findings implicate VP as a potential mediator of social buffering in female mice.SIGNIFICANCE STATEMENT In many organisms, including rodents and humans, social groups are beneficial to overall health and well-being. Moreover, it is through these social interactions that the harmful effects of stress can be mitigated, a phenomenon known as social buffering. In the present study, we describe a critical role for the neuropeptide vasopressin (VP) in social buffering of synaptic metaplasticity in stress-responsive corticotropin-releasing hormone (CRH) neurons in female mice. These effects of VP do not extend to social buffering of stress behaviors, suggesting this is a very precise and local form of sex-specific neuropeptide signaling.


Subject(s)
Stress, Psychological/physiopathology , Synapses , Vasopressins , Animals , Corticotropin-Releasing Hormone , Cues , Female , Grooming , In Vitro Techniques , Male , Mice , Neuronal Plasticity , Neurons , Paraventricular Hypothalamic Nucleus/physiopathology , Receptors, N-Methyl-D-Aspartate , Receptors, Vasopressin/drug effects , Sex Characteristics , Smell , Social Environment , Stress, Psychological/psychology
3.
J Neurosci ; 35(38): 13160-70, 2015 Sep 23.
Article in English | MEDLINE | ID: mdl-26400945

ABSTRACT

Somatodendritically released peptides alter synaptic function through a variety of mechanisms, including autocrine actions that liberate retrograde transmitters. Cholecystokinin (CCK) is a neuropeptide expressed in neurons in the dorsomedial hypothalamic nucleus (DMH), a region implicated in satiety and stress. There are clear demonstrations that exogenous CCK modulates food intake and neuropeptide expression in the DMH, but there is no information on how endogenous CCK alters synaptic properties. Here, we provide the first report of somatodendritic release of CCK in the brain in male Sprague Dawley rats. CCK is released from DMH neurons in response to repeated postsynaptic depolarizations, and acts in an autocrine fashion on CCK2 receptors to enhance postsynaptic NMDA receptor function and liberate the retrograde transmitter, nitric oxide (NO). NO subsequently acts presynaptically to enhance GABA release through a soluble guanylate cyclase-mediated pathway. These data provide the first demonstration of synaptic actions of somatodendritically released CCK in the hypothalamus and reveal a new form of retrograde plasticity, depolarization-induced potentiation of inhibition. Significance statement: Somatodendritic signaling using endocannabinoids or nitric oxide to alter the efficacy of afferent transmission is well established. Despite early convincing evidence for somatodendritic release of neurohypophysial peptides in the hypothalamus, there is only limited evidence for this mode of release for other peptides. Here, we provide the first evidence for somatodendritic release of the satiety peptide cholecystokinin (CCK) in the brain. We also reveal a new form of synaptic plasticity in which postsynaptic depolarization results in enhancement of inhibition through the somatodendritic release of CCK.


Subject(s)
Cholecystokinin/metabolism , Dorsomedial Hypothalamic Nucleus/cytology , Inhibitory Postsynaptic Potentials/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Cholecystokinin/pharmacology , GABA Agents/pharmacology , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/pharmacology , In Vitro Techniques , Male , Patch-Clamp Techniques , Peptides/pharmacology , Proglumide/analogs & derivatives , Proglumide/pharmacology , Quinazolinones/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptor, Cholecystokinin B/metabolism , Signal Transduction/drug effects , Synaptosomal-Associated Protein 25/antagonists & inhibitors , Synaptosomal-Associated Protein 25/metabolism , Thionucleotides/pharmacology , gamma-Aminobutyric Acid/pharmacology
4.
Nat Neurosci ; 16(5): 605-12, 2013 May.
Article in English | MEDLINE | ID: mdl-23563580

ABSTRACT

Exposure to a stressor sensitizes behavioral and hormonal responses to future stressors. Stress-associated release of noradrenaline enhances the capacity of central synapses to show plasticity (metaplasticity). We found noradrenaline-dependent metaplasticity at GABA synapses in the paraventricular nucleus of the hypothalamus in rat and mouse that controls the hypothalamic-pituitary-adrenal axis. In vivo stress exposure was required for these synapses to undergo activity-dependent long-term potentiation (LTPGABA). The activation of ß-adrenergic receptors during stress functionally upregulated metabotropic glutamate receptor 1 (mGluR1), allowing for mGluR1-dependent LTPGABA during afferent bursts. LTPGABA was expressed postsynaptically and manifested as the emergence of new functional synapses. Our findings provide, to the best of our knowledge, the first demonstration that noradrenaline release during an in vivo challenge alters information storage capacity at GABA synapses. Because these GABA synapses become excitatory following acute stress, this metaplasticity may contribute to neuroendocrine sensitization to stress.


Subject(s)
Neuronal Plasticity/physiology , Norepinephrine/metabolism , Stress, Psychological/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Channelrhodopsins , Chelating Agents/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hypothalamus/cytology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Light , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/drug effects , Neurotransmitter Agents/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stress, Psychological/chemically induced , Stress, Psychological/pathology , Synapses/drug effects
5.
J Neurosci ; 31(33): 12011-20, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849561

ABSTRACT

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.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Excitatory Postsynaptic Potentials/physiology , Synaptic Transmission/physiology , Animals , Cannabinoid Receptor Modulators/metabolism , Excitatory Postsynaptic Potentials/drug effects , Male , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Synaptic Transmission/drug effects
6.
J Physiol ; 589(17): 4259-70, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21727221

ABSTRACT

Glutamatergic synaptic inputs onto parvocellular neurosecretory cells (PNCs) in the paraventricular nucleus of the hypothalamus (PVN) regulate the hypothalamic-pituitary-adrenal (HPA) axis responses to stress and undergo stress-dependent changes in their capacity to transmit information. In spite of their pivotal role in regulating PNCs, relatively little is known about the fundamental rules that govern transmission at these synapses. Furthermore, since salient information in the nervous system is often transmitted in bursts, it is also important to understand the short-term dynamics of glutamate transmission under basal conditions. To characterize these properties, we obtained whole-cell patch clamp recordings from PNCs in brain slices from postnatal day 21-35 male Sprague-Dawley rats and examined EPSCs. EPSCs were elicited by electrically stimulating glutamatergic afferents along the periventricular aspect. In response to a paired-pulse stimulation protocol, EPSCs generally displayed a robust short-term depression that recovered within 5 s. Similarly, trains of synaptic stimuli (5-50 Hz) resulted in a frequency-dependent depression until a near steady state was achieved. Application of inhibitors of AMPA receptor (AMPAR) desensitization or the low-affinity, competitive AMPAR antagonist failed to affect the depression due to paired-pulse and trains of synaptic stimulation indicating that this use-dependent short-term synaptic depression has a presynaptic locus of expression. We used cumulative amplitude profiles during trains of stimulation and variance-mean analysis to estimate synaptic parameters. Finally, we report that these properties contribute to hamper the efficiency with which high frequency synaptic inputs generate spikes in PNCs, indicating that these synapses operate as effective low-pass filters in basal conditions.


Subject(s)
Glutamic Acid , Paraventricular Hypothalamic Nucleus , Animals , Electric Stimulation , Excitatory Postsynaptic Potentials , Glutamic Acid/metabolism , Hypothalamus , In Vitro Techniques , Neuroendocrine Cells , Rats, Sprague-Dawley , Synapses
7.
Nat Neurosci ; 13(10): 1257-64, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20818385

ABSTRACT

Exposure to a stressor sensitizes or 'primes' the hypothalamic-pituitary-adrenal axis to a subsequent novel stressor. The synaptic mechanisms underlying this priming, however, are not known. We found that exposing a rat to a single stressor primed glutamate synapses in the paraventricular nucleus of the hypothalamus and allowed them to undergo a short-term potentiation (STP) following a burst of high-frequency afferent activity. This transient potentiation requires a corticotrophin-releasing hormone-dependent depression of postsynaptic NMDA receptors (NMDARs). The long-term depression of NMDAR function after stress prevented the vesicular release of an inhibitory retrograde messenger that, in control conditions, arrests STP. Following stress, STP manifested as an increase in the release probability of glutamate that was sufficient to induce multivesicular release. Our findings indicate that the priming of synapses to express STP is a synaptic correlate to stress-induced behavioral and neuroendocrine sensitization.


Subject(s)
Glutamic Acid/metabolism , Neuronal Plasticity/physiology , Paraventricular Hypothalamic Nucleus/cytology , Stress, Physiological/physiology , Synapses/physiology , Animals , Biophysical Phenomena/drug effects , Biophysics/methods , Chelating Agents/pharmacology , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Dizocilpine Maleate/pharmacology , Drug Interactions , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Exocytosis/drug effects , Freezing Reaction, Cataleptic/physiology , In Vitro Techniques , Male , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Patch-Clamp Techniques/methods , Pyrimidines/pharmacology , Pyrroles/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , SNARE Proteins/chemistry , SNARE Proteins/metabolism , SNARE Proteins/pharmacology , Synapses/drug effects , Time Factors
8.
J Neurosci ; 27(6): 1325-33, 2007 Feb 07.
Article in English | MEDLINE | ID: mdl-17287507

ABSTRACT

The probability of neurotransmitter release at the nerve terminal is an important determinant of synaptic efficacy. At some central synapses, the postsynaptic, or target, neuron determines neurotransmitter release probability (P(r)) at the presynaptic terminal. The mechanisms responsible for this target-cell dependent control of P(r) have not been elucidated. Using whole-cell patch-clamp recordings from magnocellular neurosecretory cells in the paraventricular and supraoptic nuclei of the hypothalamus, we demonstrate that inhibitory, GABA synapses specifically onto oxytocin (OT)-producing neurosecretory cells exhibit a low P(r) that is relatively uniform at multiple synapses onto the same cell. This low P(r) results from a two-step process that requires the tonic release of OT from the postsynaptic cell. The ambient extracellular levels of neuropeptide are sufficient to activate postsynaptic OT receptors and trigger the Ca2+-dependent production of endocannabinoids, which act in a retrograde manner at presynaptic cannabinoid CB1 receptors to decrease GABA release. The functional consequence of this tonic inhibition of GABA release is that all inhibitory inputs facilitate uniformly when activated at high rates of activity. This causes inhibition in the postsynaptic cell that is sufficiently powerful to disrupt firing. Blockade of CB1 receptors increases P(r) at these synapses, resulting in a rapid depression of IPSCs at high rates of activity, thereby eliminating the ability of afferent inputs to inhibit postsynaptic firing. By playing a deterministic role in GABA release at the afferent nerve terminal, the postsynaptic OT neuron effectively filters synaptic signals and thereby modulates its own activity patterns.


Subject(s)
Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Oxytocin/metabolism , gamma-Aminobutyric Acid/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Benzoxazines , Calcium/physiology , Egtazic Acid/pharmacology , Extracellular Fluid/physiology , Female , Hypothalamus, Anterior/drug effects , Hypothalamus, Anterior/physiology , Male , Morpholines/pharmacology , Naphthalenes/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiology , Patch-Clamp Techniques , Piperidines/pharmacology , Presynaptic Terminals/physiology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB1/physiology , gamma-Aminobutyric Acid/metabolism
9.
Nat Neurosci ; 8(8): 1078-86, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15995701

ABSTRACT

Glial cells actively participate in synaptic transmission. They clear molecules from the synaptic cleft, receive signals from neurons and, in turn, release molecules that can modulate signaling between neuronal elements. Whether glial-derived transmitters can contribute to enduring changes in postsynaptic efficacy, however, remains to be established. In rat hypothalamic paraventricular nucleus, we demonstrate an increase in the amplitude of miniature excitatory postsynaptic currents in response to norepinephrine that requires the release of ATP from glial cells. The increase in quantal efficacy, which likely results from an insertion of AMPA receptors, is secondary to the activation of P2X(7) receptors, an increase in postsynaptic calcium and the activation of phosphatidylinositol 3-kinase. The gliotransmitter ATP, therefore, contributes directly to the regulation of postsynaptic efficacy at glutamatergic synapses in the CNS.


Subject(s)
Adenosine Triphosphate/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Norepinephrine/pharmacology , Synapses/physiology , Animals , Biomarkers/metabolism , Calcium/metabolism , Enzyme Activation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , In Vitro Techniques , Male , Neuroglia/physiology , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiology , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X7 , Synapses/metabolism , Tissue Distribution
10.
J Neurosci ; 24(22): 5162-71, 2004 Jun 02.
Article in English | MEDLINE | ID: mdl-15175385

ABSTRACT

The release of the hormones oxytocin (OT) and vasopressin (VP) into the circulation is dictated by the electrical activity of hypothalamic magnocellular neurosecretory cells (MNCs). In the paraventricular nucleus of the hypothalamus (PVN), MNC neuronal activity is exquisitely sensitive to changes in input from inhibitory GABAergic synapses. To explore the hypothesis that efficacy at these synapses is dictated by the rate at which a given synapse is activated, we obtained whole-cell recordings from MNCs in postnatal day 21-27 male Sprague Dawley rat brain slices. IPSCs were elicited by electrically stimulating GABAergic projections from either the suprachiasmatic nucleus or putative interneuron populations immediately ventral to the fornix at 5, 10, 20, and 50 Hz. Short-term plasticity was observed at 88% of the synapses tested. Of this group, synaptic depression was observed in 58%, and synaptic facilitation was observed in 41%. Identification of cells using a combined electrophysiological and immunohistochemical approach revealed a strong correlation between cell phenotype and the nature of the plasticity. Short-term facilitation was observed preferentially in OT cells (86%), whereas short-term depression was predominant in VP neurons (69%). We next examined the effects of dopamine, which increases MNC excitability, on short-term plasticity. Activation of presynaptic D(4) receptors decreased the frequency of miniature IPSCs and prevented the development of synaptic depression at higher rates of activity. Synaptic facilitation, however, was unaffected by dopamine. These findings demonstrate that, by lowering GABA release probability, dopamine confers high-pass filtering properties to the majority of inhibitory synapses onto MNCs in PVN.


Subject(s)
Dopamine/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Dopamine/pharmacology , Electric Stimulation/methods , Hypothalamus/physiology , In Vitro Techniques , Interneurons/physiology , Male , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/physiology , Neurosecretory Systems/cytology , Neurosecretory Systems/physiology , Paraventricular Hypothalamic Nucleus/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D4 , Suprachiasmatic Nucleus/physiology , Synapses/drug effects , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL