Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39005420

ABSTRACT

Background: The nucleus accumbens (NAc) mediates reward learning and motivation. Despite an abundance of neuropeptides, peptidergic neurotransmission from the NAc has not been integrated into current models of reward learning. The existence of a sparse population of neurons containing corticotropin releasing factor (CRF) has been previously documented. Here we provide a comprehensive analysis of their identity and functional role in shaping reward learning. Methods: To do this, we took a multidisciplinary approach that included florescent in situ hybridization (N mice ≥ 3), tract tracing (N mice = 5), ex vivo electrophysiology (N cells ≥ 30), in vivo calcium imaging with fiber photometry (N mice ≥ 4) and use of viral strategies in transgenic lines to selectively delete CRF peptide from NAc neurons (N mice ≥ 4). Behaviors used were instrumental learning, sucrose preference and spontaneous exploration in an open field. Results: Here we show that the vast majority of NAc CRF-containing (NAc CRF ) neurons are spiny projection neurons (SPNs) comprised of dopamine D1-, D2- or D1/D2-containing SPNs that primarily project and connect to the ventral pallidum and to a lesser extent the ventral midbrain. As a population, they display mature and immature SPN firing properties. We demonstrate that NAc CRF neurons track reward outcomes during operant reward learning and that CRF release from these neurons acts to constrain initial acquisition of action-outcome learning, and at the same time facilitates flexibility in the face of changing contingencies. Conclusion: We conclude that CRF release from this sparse population of SPNs is critical for reward learning under normal conditions.

2.
bioRxiv ; 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38659848

ABSTRACT

Cholinergic interneurons (ChIs) act as master regulators of striatal output, finely tuning neurotransmission to control motivated behaviors. ChIs are a cellular target of many peptide and hormonal neuromodulators, including corticotropin releasing factor, opioids, insulin and leptin, which can influence an animal's behavior by signaling stress, pleasure, pain and nutritional status. However, little is known about how sex hormones via estrogen receptors influence the function of these other neuromodulators. Here, we performed in situ hybridization on mouse striatal tissue to characterize the effect of sex and sex hormones on choline acetyltransferase ( Chat ), estrogen receptor alpha ( Esr1 ), and corticotropin releasing factor type 1 receptor ( Crhr1 ) expression. Although we did not detect sex differences in ChAT protein levels in the striatum, we found that female mice have more Chat mRNA-expressing neurons than males. At the population level, we observed a sexually dimorphic distribution of Esr1 - and Crhr1 -expressing ChIs in the ventral striatum that demonstrates an antagonistic correlational relationship, which is abolished by ovariectomy. Only in the NAc did we find a significant population of ChIs that co-express Crhr1 and Esr1 . At the cellular level, Crhr1 and Esr1 transcript levels were negatively correlated only during estrus, indicating that changes in sex hormones levels can modulate the interaction between Crhr1 and Esr1 mRNA levels. Together, these data provide evidence for the unique expression and interaction of Esr1 and Crhr1 in ventral striatal ChIs, warranting further investigation into how these transcriptomic patterns might underlie important functions for ChIs at the intersection of stress and reproductive behaviors.

3.
bioRxiv ; 2023 Sep 17.
Article in English | MEDLINE | ID: mdl-37745598

ABSTRACT

Cholinergic interneurons (ChIs) provide the main source of acetylcholine in the striatum and have emerged as a critical modulator of behavioral flexibility, motivation, and associative learning. In the dorsal striatum, ChIs display heterogeneous firing patterns. Here, we investigated the spontaneous firing patterns of ChIs in the nucleus accumbens (NAc) shell, a region of the ventral striatum. Using male and female mice, we performed cell-attached patch clamp electrophysiology recordings from ChIs. We identified four distinct ChI firing signatures: regular single-spiking, irregular single-spiking, rhythmic bursting followed by pauses or low activity, and a mixed-mode pattern composed of bursting activity and regular single spiking. ChIs from females had lower firing rates compared to males and had both a higher proportion of mixed-mode firing patterns and a lower proportion of regular single-spiking neurons compared to males. We further observed that across the estrous cycle, the estrus phase was characterized by higher proportions of mixed-mode, rhythmic bursting, and irregular ChI firing patterns compared to other phases. ChI firing mode was not driven by glutamatergic synaptic transmission in the slice preparation. Using pooled data from males and females, we examined the how the stress-associated neuropeptide corticotropin releasing factor (CRF) impacts these firing patterns. ChI firing patterns showed differential sensitivity to CRF. Furthermore, CRF shifted the proportion of ChI firing patterns toward more regular spiking activity over bursting patterns. These findings highlight the heterogeneous nature of ChI firing patterns, which may have implications for accumbal-dependent motivated behaviors. New and Noteworthy: ChIs within the dorsal and ventral striatum can exert a huge influence on network output and motivated behaviors. However, the firing patterns and neuromodulation of ChIs within the ventral striatum,specifically the NAc shell, are understudied. Here we report that NAc shell ChIs have a heterogenous distribution of ChI firing patterns that are labile and can be modulated by the stress-linked neuropeptide CRF and by estrous cycle.

4.
J Neurosci ; 2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35896424

ABSTRACT

There are five cloned muscarinic acetylcholine receptors (M1-M5). Of these, the muscarinic type 5 receptor (M5) is the only one localized to dopamine neurons in the ventral tegmental area and substantia nigra. Unlike M1-M4, the M5 receptor has relatively restricted expression in the brain, making it an attractive therapeutic target. Here we performed an in-depth characterization of M5-dependent potentiation of dopamine transmission in the nucleus accumbens and accompanying exploratory behaviors in male and female mice. We show that M5 receptors potentiate dopamine transmission by acting directly on the terminals within the nucleus accumbens. Using the muscarinic agonist oxotremorine, we revealed a unique concentration-response curve and a sensitivity to repeated forced swim stress or restraint stress exposure. We found that constitutive deletion of M5 receptors reduced exploration of the center of an open field while at the same time impairing normal habituation only in male mice. In addition, M5 deletion reduced exploration of salient stimuli, especially under conditions of high novelty, yet had no effect on hedonia assayed using the sucrose preference test or on stress coping strategy assayed using the forced swim test. We conclude that M5 receptors are critical for both engaging with the environment and updating behavioral output in response to environment cues, specifically in male mice. A cardinal feature of mood and anxiety disorders is withdrawal from the environment. These data indicate that boosting M5 receptor activity may be a useful therapeutic target for ameliorating these symptoms of depression and anxiety.Significance Statement:The basic physiological and behavioral functions of the muscarinic M5 receptor remain understudied. Furthermore, its presence on dopamine neurons, relatively restricted expression in the brain, and recent crystallization make it an attractive target for therapeutic development. Yet, most preclinical studies of M5 receptor function have primarily focused on substance use disorders in male rodents. Here we characterized the role of M5 receptors in potentiating dopamine transmission in the nucleus accumbens, finding impaired functioning after stress exposure. Furthermore, we show that M5 receptors can modulate exploratory behavior in a sex-specific manner, without impacting hedonic behavior. These findings further illustrate the therapeutic potential of the M5 receptor, warranting further research in the context of treating mood disorders.

6.
Nat Neurosci ; 24(10): 1414-1428, 2021 10.
Article in English | MEDLINE | ID: mdl-34385700

ABSTRACT

The long-range GABAergic input from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) is relatively understudied, and therefore its role in reward processing has remained unknown. In the present study, we show, in both male and female mice, that long-range GABAergic projections from the VTA to the ventral NAc shell, but not to the dorsal NAc shell or NAc core, are engaged in reward and reinforcement behavior. We show that this GABAergic projection exclusively synapses on to cholinergic interneurons (CINs) in the ventral NAc shell, thereby serving a specialized function in modulating reinforced reward behavior through the inhibition of ventral NAc shell CINs. These findings highlight the diversity in the structural and functional topography of VTA GABAergic projections, and their neuromodulatory interactions across the dorsoventral gradient of the NAc shell. They also further our understanding of neuronal circuits that are directly implicated in neuropsychiatric conditions such as depression and addiction.


Subject(s)
Cholinergic Neurons/drug effects , Nucleus Accumbens/drug effects , Reinforcement, Psychology , Ventral Tegmental Area/physiopathology , gamma-Aminobutyric Acid/physiology , Animals , Brain Mapping , Conditioning, Operant/drug effects , Electrophysiological Phenomena , Female , Interneurons/drug effects , Male , Mice , Mice, Inbred C57BL , Reward , Self Stimulation
8.
Front Behav Neurosci ; 14: 564054, 2020.
Article in English | MEDLINE | ID: mdl-33132859

ABSTRACT

Stress is highly pervasive in humans, impacting motivated behaviors with an enormous toll on life quality. Many of the effects of stress are orchestrated by neuropeptides such as corticotropin-releasing factor (CRF). It has previously been shown that in stress-naïve male mice, CRF acts in the core of the nucleus accumbens (NAc) to produce appetitive effects and to increase dopamine release; yet in stress-exposed male mice, CRF loses its capacity to modulate NAc dopamine release and is aversive. In the current research, we tested whether this effect is comparable in females to males and whether the neuroadaptation is susceptible to social transmission. We found that, like in males, CRF increased dopamine release in stress-naïve but not stress-exposed female mice. Importantly, this persistent physiological change was not accompanied by overt behavioral changes that would be indicative of depression- or anxiety-like phenotype. Nonetheless, when these mice were housed for 7 days with stress-naïve conspecifics, the cage mates also exhibited a loss of dopamine potentiation by CRF. These data demonstrate the asymptomatic, yet pervasive transmission of stress-related neuroadaptations in the population.

9.
J Neurosci ; 40(39): 7510-7522, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32859717

ABSTRACT

Dopamine (DA) signals in the striatum are critical for a variety of vital processes, including motivation, motor learning, and reinforcement learning. Striatal DA signals can be evoked by direct activation of inputs from midbrain DA neurons (DANs) as well as cortical and thalamic inputs to the striatum. In this study, we show that in vivo optogenetic stimulation of prelimbic (PrL) and infralimbic (IL) cortical afferents to the striatum triggers an increase in extracellular DA concentration, which coincides with elevation of striatal acetylcholine (ACh) levels. This increase is blocked by a nicotinic ACh receptor (nAChR) antagonist. Using single or dual optogenetic stimulation in brain slices from male and female mice, we compared the properties of these PrL/IL-evoked DA signals with those evoked by stimulation from midbrain DAN axonal projections. PrL/IL-evoked DA signals are undistinguishable from DAN evoked DA signals in their amplitudes and electrochemical properties. However, PrL/IL-evoked DA signals are spatially restricted and preferentially recorded in the dorsomedial striatum. PrL/IL-evoked DA signals also differ in their pharmacological properties, requiring activation of glutamate and nicotinic ACh receptors. Thus, both in vivo and in vitro results indicate that cortical evoked DA signals rely on recruitment of cholinergic interneurons, which renders DA signals less able to summate during trains of stimulation and more sensitive to both cholinergic drugs and temperature. In conclusion, cortical and midbrain inputs to the striatum evoke DA signals with unique spatial and pharmacological properties that likely shape their functional roles and behavioral relevance.SIGNIFICANCE STATEMENT Dopamine signals in the striatum play a critical role in basal ganglia function, such as reinforcement and motor learning. Different afferents to the striatum can trigger dopamine signals, but their release properties are not well understood. Further, these input-specific dopamine signals have only been studied in separate animals. Here we show that optogenetic stimulation of cortical glutamatergic afferents to the striatum triggers dopamine signals both in vivo and in vitro These afferents engage cholinergic interneurons, which drive dopamine release from dopamine neuron axons by activation of nicotinic acetylcholine receptors. We also show that cortically evoked dopamine signals have other unique properties, including spatial restriction and sensitivity to temperature changes than dopamine signals evoked by stimulation of midbrain dopamine neuron axons.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Prefrontal Cortex/metabolism , Acetylcholine/metabolism , Animals , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Corpus Striatum/cytology , Corpus Striatum/physiology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Evoked Potentials , Female , Interneurons/metabolism , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology
11.
J Neurosci ; 39(29): 5647-5661, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31109960

ABSTRACT

Cholinergic interneurons (CINs) are critical regulators of striatal network activity and output. Changes in CIN activity are thought to encode salient changes in the environment and stimulus-response-outcome associations. Here we report that the stress-associated neuropeptide corticotropin releasing factor (CRF) produces a profound and reliable increase in the spontaneous firing of CINs in both dorsal striatum and nucleus accumbens (NAc) through activation of CRF type 1 receptors, production of cAMP and reduction in spike accommodation in male mice. The increase of CIN firing by CRF results in the activation muscarinic acetylcholine receptors type 5, which mediate potentiation of dopamine transmission in the striatum. This study provides critical mechanistic insight into how CRF modulates striatal activity and dopamine transmission in the NAc to likely account for CRF facilitation of appetitive behaviors.SIGNIFICANCE STATEMENT Although the presence of CRF receptors in the dorsal and ventral striatum has been acknowledged, the cellular identity and the functional consequences of receptor activation is unknown. Here we report that striatal cholinergic interneurons express CRF-R1 receptors and are acutely activated by the neuropeptide CRF that is released in response to salient environmental stimuli. Cholinergic interneurons make <1% of the cells in the striatum but are critical regulators of the striatal circuitry and its output. CRF's fast and potent activation of cholinergic interneurons could have far reaching behavioral implications across motivated behaviors controlled by the striatum.


Subject(s)
Corpus Striatum/metabolism , Corticotropin-Releasing Hormone/administration & dosage , Interneurons/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Corpus Striatum/drug effects , Interneurons/chemistry , Interneurons/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Receptors, Corticotropin-Releasing Hormone/agonists
12.
Neuron ; 90(5): 1100-13, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27181061

ABSTRACT

Striatal medium spiny neurons (MSNs) form inhibitory synapses on neighboring striatal neurons through axon collaterals. The functional relevance of this lateral inhibition and its regulation by dopamine remains elusive. We show that synchronized stimulation of collateral transmission from multiple indirect-pathway MSNs (iMSNs) potently inhibits action potentials in direct-pathway MSNs (dMSNs) in the nucleus accumbens. Dopamine D2 receptors (D2Rs) suppress lateral inhibition from iMSNs to disinhibit dMSNs, which are known to facilitate locomotion. Surprisingly, D2R inhibition of synaptic transmission was larger at axon collaterals from iMSNs than their projections to the ventral pallidum. Targeted deletion of D2Rs from iMSNs impaired cocaine's ability to suppress lateral inhibition and increase locomotion. These impairments were rescued by chemogenetic activation of Gi-signaling in iMSNs. These findings shed light on the functional significance of lateral inhibition between MSNs and offer a novel synaptic mechanism by which dopamine gates locomotion and cocaine exerts its canonical stimulant response. VIDEO ABSTRACT.


Subject(s)
Cocaine/pharmacology , Corpus Striatum/cytology , Corpus Striatum/drug effects , Dopamine/metabolism , Locomotion/drug effects , Neural Inhibition/drug effects , Neural Inhibition/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Central Nervous System Sensitization/drug effects , Central Nervous System Sensitization/physiology , Corpus Striatum/physiology , Dose-Response Relationship, Drug , Mice , Mice, Knockout , Mice, Transgenic , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Receptors, Dopamine D2/physiology
13.
Neuron ; 90(4): 824-38, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27196975

ABSTRACT

Bradykinesia is a prominent phenotype of Parkinson's disease, depression, and other neurological conditions. Disruption of dopamine (DA) transmission plays an important role, but progress in understanding the exact mechanisms driving slowness of movement has been impeded due to the heterogeneity of DA receptor distribution on multiple cell types within the striatum. Here we show that selective deletion of DA D2 receptors (D2Rs) from indirect-pathway medium spiny neurons (iMSNs) is sufficient to impair locomotor activity, phenocopying DA depletion models of Parkinson's disease, despite this mouse model having intact DA transmission. There was a robust enhancement of GABAergic transmission and a reduction of in vivo firing in striatal and pallidal neurons. Mimicking D2R signaling in iMSNs with Gi-DREADDs restored the level of tonic GABAergic transmission and rescued the motor deficit. These findings indicate that DA, through D2R activation in iMSNs, regulates motor output by constraining the strength of GABAergic transmission.


Subject(s)
Corpus Striatum/metabolism , Hypokinesia/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Dopamine/metabolism , Globus Pallidus/metabolism , Mice, Transgenic , Neurons/metabolism , Parkinson Disease/metabolism , Substantia Nigra/metabolism
14.
Ann Neurol ; 73(3): 355-69, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23225132

ABSTRACT

OBJECTIVE: Prenatal cocaine exposure (PCE) can cause persistent neuropsychological and motor abnormalities in affected children, but the physiological consequences of PCE remain unclear. Conclusions drawn from clinical studies can sometimes be confounded by polysubstance abuse and nutritional deprivation. However, existing observations suggest that cocaine exposure in utero, as in adults, increases synaptic dopamine and promotes enduring dopamine-dependent plasticity at striatal synapses, altering behaviors and basal ganglia function. METHODS: We used a combination of behavioral measures, electrophysiology, optical imaging, and biochemical and electrochemical recordings to examine corticostriatal activity in adolescent mice exposed to cocaine in utero. RESULTS: We show that PCE caused abnormal dopamine-dependent behaviors, including heightened excitation following stress and blunted locomotor augmentation after repeated treatment with amphetamine. These abnormal behaviors were consistent with abnormal γ-aminobutyric acid (GABA) interneuron function, which promoted a reversible depression in corticostriatal activity. PCE hyperpolarized and reduced tonic GABA currents in both fast-spiking and persistent low-threshold spiking type GABA interneurons to increase tonic inhibition at GABAB receptors on presynaptic corticostriatal terminals. Although D2 receptors paradoxically increased glutamate release following PCE, normal corticostriatal modulation by dopamine was reestablished with a GABAA receptor antagonist. INTERPRETATION: The dynamic alterations at corticostriatal synapses that occur in response to PCE parallel the reported effects of repeated psychostimulants in mature animals, but differ in being specifically generated through GABAergic mechanisms. Our results indicate approaches that normalize GABA and D2 receptor-dependent synaptic plasticity may be useful for treating the behavioral effects of PCE and other developmental disorders that are generated through abnormal GABAergic signaling.


Subject(s)
Cerebral Cortex/pathology , Cocaine/toxicity , Corpus Striatum/pathology , Dopamine Uptake Inhibitors/toxicity , Neural Inhibition/drug effects , Prenatal Exposure Delayed Effects , Age Factors , Analysis of Variance , Anesthetics, Local/pharmacology , Animals , Biophysics , Dopamine/metabolism , Dopamine Agents/pharmacology , Drug Interactions , Electric Stimulation/adverse effects , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Exploratory Behavior/drug effects , Female , GABA Agents/pharmacology , Green Fluorescent Proteins/genetics , Hindlimb Suspension/methods , In Vitro Techniques , Interneurons/drug effects , Interneurons/physiology , Lidocaine/analogs & derivatives , Lidocaine/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neural Inhibition/physiology , Neuronal Plasticity/drug effects , Patch-Clamp Techniques , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Quinoxalines/pharmacology , Quinpirole/pharmacology , Receptors, GABA-A/metabolism , Rotarod Performance Test , Sodium Channel Blockers/pharmacology , Statistics, Nonparametric , Tetrodotoxin/pharmacology
15.
J Neurosci ; 32(49): 17582-96, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23223282

ABSTRACT

Activation of the dynorphin/κ-opioid receptor (KOR) system by repeated stress exposure or agonist treatment produces place aversion, social avoidance, and reinstatement of extinguished cocaine place preference behaviors by stimulation of p38α MAPK, which subsequently causes the translocation of the serotonin transporter (SERT, SLC6A4) to the synaptic terminals of serotonergic neurons. In the present study we extend those findings by showing that stress-induced potentiation of cocaine conditioned place preference occurred by a similar mechanism. In addition, SERT knock-out mice did not show KOR-mediated aversion, and selective reexpression of SERT by lentiviral injection into the dorsal raphe restored the prodepressive effects of KOR activation. Kinetic analysis of several neurotransporters demonstrated that repeated swim stress exposure selectively increased the V(max) but not K(m) of SERT without affecting dopamine transport or the high-capacity, low-affinity transporters. Although the serotonergic neurons in the dorsal raphe project throughout the forebrain, a significant stress-induced increase in cell-surface SERT expression was only evident in the ventral striatum, and not in the dorsal striatum, hippocampus, prefrontal cortex, amygdala, or dorsal raphe. Stereotaxic microinjections of the long-lasting KOR antagonist norbinaltorphimine demonstrated that local KOR activation in the nucleus accumbens, but not dorsal raphe, mediated this stress-induced increase in ventral striatal surface SERT expression. Together, these results support the hypothesis that stress-induced activation of the dynorphin/KOR system produces a transient increase in serotonin transport locally in the ventral striatum that may underlie some of the adverse consequences of stress exposure, including the potentiation of the rewarding effects of cocaine.


Subject(s)
Avoidance Learning/physiology , Cocaine/pharmacology , Corpus Striatum/metabolism , Dynorphins/physiology , Reward , Serotonin Plasma Membrane Transport Proteins/metabolism , Stress, Psychological/metabolism , Stress, Psychological/psychology , Animals , Avoidance Learning/drug effects , Brain/metabolism , Dopamine/metabolism , Dynorphins/metabolism , G-Protein-Coupled Receptor Kinase 3/genetics , G-Protein-Coupled Receptor Kinase 3/physiology , Male , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microinjections/methods , Naltrexone/administration & dosage , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacokinetics , Nicotine/adverse effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Raphe Nuclei/physiology , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/physiology , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Substance Withdrawal Syndrome/metabolism , Synaptosomes/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/physiology
16.
J Neurosci ; 32(36): 12325-36, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-22956823

ABSTRACT

Repeated stress releases dynorphins and causes subsequent activation of κ-opioid receptors (KORs) in limbic brain regions. The serotonergic dorsal raphe nucleus (DRN) has previously been found to be an important site of action for the dysphoric effects of dynorphin-κ-opioid receptor system activation during stress-evoked behaviors, and KOR-induced activation of p38α mitogen-activated protein kinase (MAPK) in serotonergic neurons was found to be a critical mediator of the aversive properties of stress. Yet, how dynorphins and KORs functionally regulate the excitability of serotonergic DRN neurons both in adaptive and pathological stress states is poorly understood. Here we report that acute KOR activation by the selective agonist U69,593 [(+)-(5α,7α,8ß)-N-methyl-N-[7-(1-pyrrolidinyl)-1-oxaspiro[4.5]dec-8-yl]benzeneacetamide] inhibits serotonergic neuronal excitability within the DRN through both presynaptic inhibition of excitatory synaptic transmission and postsynaptic activation of G-protein-gated inwardly rectifying potassium channels (GIRKs) electrophysiologically recorded in brain slices. C57BL/6 mice subjected to repeated swim, stress sessions had significantly reduced KOR-mediated GIRK currents recorded in serotonergic neurons in DRN postsynaptically, without significantly affecting presynaptic KOR-mediated regulation of excitatory transmission. This effect was blocked by genetic excision of p38α MAPK selectively from serotonergic neurons. An increase in phospho-immunoreactivity suggests that this functional dysregulation may be a consequence of tyrosine phosphorylation of GIRK (K(IR)3.1) channels. These data elucidate a mechanism for stress-induced dysregulation of the excitability of neurons in the DRN and identify a functional target of stress-induced p38α MAPK activation that may underlie some of the negative effects of pathological stress exposure.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Mitogen-Activated Protein Kinase 14/physiology , Raphe Nuclei/enzymology , Signal Transduction/physiology , Stress, Psychological/enzymology , Animals , Benzeneacetamides/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Phosphorylation , Pyrrolidines/pharmacology , Raphe Nuclei/drug effects , Reaction Time/physiology , Serotonin/physiology , Signal Transduction/drug effects , Time Factors , Tyrosine/metabolism
17.
Nature ; 490(7420): 402-6, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-22992525

ABSTRACT

Stressors motivate an array of adaptive responses ranging from 'fight or flight' to an internal urgency signal facilitating long-term goals. However, traumatic or chronic uncontrollable stress promotes the onset of major depressive disorder, in which acute stressors lose their motivational properties and are perceived as insurmountable impediments. Consequently, stress-induced depression is a debilitating human condition characterized by an affective shift from engagement of the environment to withdrawal. An emerging neurobiological substrate of depression and associated pathology is the nucleus accumbens, a region with the capacity to mediate a diverse range of stress responses by interfacing limbic, cognitive and motor circuitry. Here we report that corticotropin-releasing factor (CRF), a neuropeptide released in response to acute stressors and other arousing environmental stimuli, acts in the nucleus accumbens of naive mice to increase dopamine release through coactivation of the receptors CRFR1 and CRFR2. Remarkably, severe-stress exposure completely abolished this effect without recovery for at least 90 days. This loss of CRF's capacity to regulate dopamine release in the nucleus accumbens is accompanied by a switch in the reaction to CRF from appetitive to aversive, indicating a diametric change in the emotional response to acute stressors. Thus, the current findings offer a biological substrate for the switch in affect which is central to stress-induced depressive disorders.


Subject(s)
Appetitive Behavior/physiology , Avoidance Learning/physiology , Corticotropin-Releasing Hormone/metabolism , Nucleus Accumbens/metabolism , Stress, Psychological/metabolism , Animals , Appetitive Behavior/drug effects , Avoidance Learning/drug effects , Corticotropin-Releasing Hormone/pharmacology , Dopamine/metabolism , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/physiopathology , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/deficiency , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction/drug effects , Stress, Psychological/physiopathology
18.
Neuron ; 71(3): 498-511, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21835346

ABSTRACT

Maladaptive responses to stress adversely affect human behavior, yet the signaling mechanisms underlying stress-responsive behaviors remain poorly understood. Using a conditional gene knockout approach, the α isoform of p38 mitogen-activated protein kinase (MAPK) was selectively inactivated by AAV1-Cre-recombinase infection in specific brain regions or by promoter-driven excision of p38α MAPK in serotonergic neurons (by Slc6a4-Cre or ePet1-Cre) or astrocytes (by Gfap-CreERT2). Social defeat stress produced social avoidance (a model of depression-like behaviors) and reinstatement of cocaine preference (a measure of addiction risk) in wild-type mice, but not in mice having p38α MAPK selectively deleted in serotonin-producing neurons of the dorsal raphe nucleus. Stress-induced activation of p38α MAPK translocated the serotonin transporter to the plasma membrane and increased the rate of transmitter uptake at serotonergic nerve terminals. These findings suggest that stress initiates a cascade of molecular and cellular events in which p38α MAPK induces a hyposerotonergic state underlying depression-like and drug-seeking behaviors.


Subject(s)
Cocaine-Related Disorders/genetics , Depression/genetics , Mitogen-Activated Protein Kinase 14/physiology , Neurons/physiology , Serotonin/physiology , Stress, Psychological/psychology , Animals , Avoidance Learning/physiology , Choice Behavior/physiology , Cocaine-Related Disorders/psychology , Conditioning, Psychological/physiology , Depression/psychology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Raphe Nuclei/metabolism , Raphe Nuclei/physiology , Raphe Nuclei/physiopathology , Receptors, Opioid/physiology , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Stress, Psychological/physiopathology , Nociceptin Receptor
19.
Neuropharmacology ; 61(3): 524-43, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21530552

ABSTRACT

The median (MR) and dorsal raphe (DR) nuclei contain the majority of the 5-hydroxytryptamine (5-HT, serotonin) neurons that project to limbic forebrain regions, are important in regulating homeostatic functions and are implicated in the etiology and treatment of mood disorders and schizophrenia. The primary synaptic inputs within and to the raphe are glutamatergic and GABAergic. The DR is divided into three subfields, i.e., ventromedial (vmDR), lateral wings (lwDR) and dorsomedial (dmDR). Our previous work shows that cell characteristics of 5-HT neurons and the magnitude of the 5-HT(1A) and 5-HT(1B) receptor-mediated responses in the vmDR and MR are not the same. We extend these observations to examine the electrophysiological properties across all four raphe subfields in both 5-HT and non-5-HT neurons. The neurochemical topography of glutamatergic and GABAergic cell bodies and nerve terminals were identified using immunohistochemistry and the morphology of the 5-HT neurons was measured. Although 5-HT neurons possessed similar physiological properties, important differences existed between subfields. Non-5-HT neurons were indistinguishable from 5-HT neurons. GABA neurons were distributed throughout the raphe, usually in areas devoid of 5-HT neurons. Although GABAergic synaptic innervation was dense throughout the raphe (immunohistochemical analysis of the GABA transporters GAT1 and GAT3), their distributions differed. Glutamate neurons, as defined by vGlut3 anti-bodies, were intermixed and co-localized with 5-HT neurons within all raphe subfields. Finally, the dendritic arbor of the 5-HT neurons was distinct between subfields. Previous studies regard 5-HT neurons as a homogenous population. Our data support a model of the raphe as an area composed of functionally distinct subpopulations of 5-HT and non-5-HT neurons, in part delineated by subfield. Understanding the interaction of the cell properties of the neurons in concert with their morphology, local distribution of GABA and glutamate neurons and their synaptic input, reveals a more complicated and heterogeneous raphe. These results provide an important foundation for understanding how specific subfields modulate behavior and for defining which aspects of the circuitry are altered during the etiology of psychological disorders.


Subject(s)
Brain Chemistry , Neurons/physiology , Neurons/ultrastructure , Raphe Nuclei/physiology , Raphe Nuclei/ultrastructure , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1B/metabolism , Synaptic Transmission , Animals , Dendrites/physiology , Dendrites/ultrastructure , Electric Impedance , GABA Plasma Membrane Transport Proteins/metabolism , Glucose Transporter Type 3/metabolism , Glutamate Decarboxylase/metabolism , Immunohistochemistry , Male , Membrane Potentials , Patch-Clamp Techniques , Raphe Nuclei/chemistry , Rats , Rats, Sprague-Dawley , Tryptophan Hydroxylase/metabolism
20.
Methods Mol Biol ; 717: 197-219, 2011.
Article in English | MEDLINE | ID: mdl-21370032

ABSTRACT

Psychiatric disorders including anxiety, depression, and addiction are both precipitated and exacerbated by severe or chronic stress exposure. While acutely, stress responses are adaptive, repeated exposure to stress can dysregulate the brain in such a way as to predispose the organism to both physiological and mental illness. Understanding the neuronal chemicals, cell types, and circuits involved in both normal and pathological stress responses are essential in developing new therapeutics for psychiatric diseases. Varying degrees of stressor exposure cause the release of a constellation of chemicals, including neuropeptides such as dynorphin. Neuropeptidergic release can be very difficult to directly measure with adequate spatial and temporal resolution. Moreover, the downstream consequences following release and receptor binding are numerous and also difficult to measure with cellular resolution. Following repeated stressor exposure, dynorphin is released, binds to the kappa opioid receptor (KOR), and causes activation of KOR. Agonist-activated KOR becomes a substrate for G protein receptor kinase (GRK), which phosphorylates the Ser369 residue at the C-terminal tail of the receptor in the first step in the ß-Arrestin-dependent desensitization cascade. Through the use of phospho--selective antibodies developed and validated in the laboratory, we have the tools, to assess with fine cellular resolution, the strength of behavioral stimulus required for release, time course of the release, and regional location of release. We have gone on to show that following KOR activation, both ERK 1/2 and p38 MAP kinase phosphorylation are increased through use of commercially available phospho-selective antibodies. Finally, we have identified that one effector of KOR/p38MAP kinase is K(IR) 3.1 and have developed a phospho-selective antibody against the Y12 motif of this channel. Much like KOR and p38 MAP kinase, phosphorylation of this potassium channel increases following repeated stress. The following chapter discusses immunohistochemical and quantification methods used for phospho-selective antibodies used in various brain regions following behavioral manipulations.


Subject(s)
Antibodies, Phospho-Specific/analysis , Immunohistochemistry/methods , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antibodies, Phospho-Specific/immunology , Antibodies, Phospho-Specific/isolation & purification , Brain/metabolism , Brain/ultrastructure , Cell Line , Chromatography, Affinity/methods , Humans , Mice , Microscopy/methods , Mitogen-Activated Protein Kinase 1/analysis , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/analysis , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , Potassium Channels, Inwardly Rectifying/analysis , Potassium Channels, Inwardly Rectifying/immunology , Rats , Receptors, Opioid, kappa/analysis , Receptors, Opioid, kappa/immunology , p38 Mitogen-Activated Protein Kinases/analysis , p38 Mitogen-Activated Protein Kinases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...