151
|
Bienkowski MS, Rinaman L. Immune challenge activates neural inputs to the ventrolateral bed nucleus of the stria terminalis. Physiol Behav 2011; 104:257-65. [PMID: 21402087 PMCID: PMC3118915 DOI: 10.1016/j.physbeh.2011.03.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 02/15/2011] [Accepted: 03/08/2011] [Indexed: 01/16/2023]
Abstract
Hypothalamo-pituitary-adrenal (HPA) axis activation in response to infection is an important mechanism by which the nervous system can suppress inflammation. HPA output is controlled by the hypothalamic paraventricular nucleus (PVN). Previously, we determined that noradrenergic inputs to the PVN contribute to, but do not entirely account for, the ability of bacterial endotoxin (i.e., lipopolysacharide, LPS) to activate the HPA axis. The present study investigated LPS-induced recruitment of neural inputs to the ventrolateral bed nucleus of the stria terminalis (vlBNST). GABAergic projections from the vlBNST inhibit PVN neurons at the apex of the HPA axis; thus, we hypothesize that LPS treatment activates inhibitory inputs to the vlBNST to thereby "disinhibit" the PVN and increase HPA output. To test this hypothesis, retrograde neural tracer was iontophoretically delivered into the vlBNST of adult male rats to retrogradely label central sources of axonal input. After one week, rats were injected i.p. with either LPS (200 μg/kg BW) or saline vehicle, and then perfused with fixative 2.5h later. Brains were processed for immunohistochemical localization of retrograde tracer and the immediate-early gene product, Fos (a marker of neural activation). Brain regions that provide inhibitory input to the vlBNST (e.g., caudal nucleus of the solitary tract, central amygdala, dorsolateral BNST) were preferentially activated by LPS, whereas sources of excitatory input (e.g., paraventricular thalamus, medial prefrontal cortex) were not activated or were activated less robustly. These results suggest that LPS treatment recruits central neural systems that actively suppress vlBNST neural activity, thereby removing a potent source of inhibitory control over the HPA axis.
Collapse
|
152
|
McGinty VB, Hayden BY, Heilbronner SR, Dumont EC, Graves SM, Mirrione MM, du Hoffmann J, Sartor GC, España RA, Millan EZ, Difeliceantonio AG, Marchant NJ, Napier TC, Root DH, Borgland SL, Treadway MT, Floresco SB, McGinty JF, Haber S. Emerging, reemerging, and forgotten brain areas of the reward circuit: Notes from the 2010 Motivational Neural Networks conference. Behav Brain Res 2011; 225:348-57. [PMID: 21816177 DOI: 10.1016/j.bbr.2011.07.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 07/18/2011] [Indexed: 10/17/2022]
Abstract
On April 24-27, 2010, the Motivational Neuronal Networks meeting took place in Wrightsville Beach, North Carolina. The conference was devoted to "Emerging, re-emerging, and forgotten brain areas" of the reward circuit. A central feature of the conference was four scholarly discussions of cutting-edge topics related to the conference's theme. These discussions form the basis of the present review, which summarizes areas of consensus and controversy, and serves as a roadmap for the next several years of research.
Collapse
Affiliation(s)
- Vincent B McGinty
- Department of Neurobiology, Stanford University, Stanford, CA 94305-5125, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Longoni R, Spina L, Vinci S, Acquas E. The MEK inhibitor SL327 blocks acquisition but not expression of lithium-induced conditioned place aversion: a behavioral and immunohistochemical study. Psychopharmacology (Berl) 2011; 216:63-73. [PMID: 21312031 DOI: 10.1007/s00213-011-2192-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 01/17/2011] [Indexed: 11/27/2022]
Abstract
RATIONALE Recent evidence involves extracellular signal-regulated kinase (ERK) in positive motivational properties of drugs as determined by conditioned place preference but, to date, its role in conditioned place aversion (CPA) still awaits to be fully characterized. OBJECTIVES The aim of this study was to assess whether activated ERK (pERK) plays a role in the acquisition and/or expression of lithium-induced CPA. METHODS C57BL/6J mice were subjected to lithium (150 mg/kg)-induced CPA. The role of pERK was determined by administering the mitogen-activating extracellular kinase inhibitor, SL327, (a) 25 and 50 mg/kg, before each exposure to the lithium-associated compartment (acquisition), and (b) 25, 50, and 100 mg/kg, before post-conditioning test (expression). To assess whether ERK is activated by acute lithium and, in distinct experiments, during CPA expression, mice were sacrificed, 30 min after lithium, and immediately after post-conditioning test, respectively, for pERK immunohistochemistry. RESULTS Lithium increased pERK-positive neurons in bed nucleus of stria termialis, in central and basolateral amygdala and elicited significant CPA. SL327 (50 mg/kg) significantly prevented its acquisition. In addition, the post-conditioning test of lithium-conditioned mice determined a significant increase of pERK-positive neurons in the dorsal striatum and SL327 (50 mg/kg), administered before post-conditioning test, while failing at the doses of 25, 50, and 100 mg/kg, to affect lithium-induced CPA expression, completely prevented it. CONCLUSIONS These results indicate that pERK is critical for acquisition, but not expression, of lithium-induced CPA and that its activation in the dorsal striatum, during expression, is not critical for retrieval of the aversive memory.
Collapse
Affiliation(s)
- Rosanna Longoni
- Department of Toxicology, University of Cagliari, Via Ospedale, 72, 09124, Cagliari, Italy
| | | | | | | |
Collapse
|
154
|
Stone EA, Lin Y, Sarfraz Y, Quartermain D. The role of the central noradrenergic system in behavioral inhibition. BRAIN RESEARCH REVIEWS 2011; 67:193-208. [PMID: 21315760 PMCID: PMC3101301 DOI: 10.1016/j.brainresrev.2011.02.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Revised: 01/30/2011] [Accepted: 02/03/2011] [Indexed: 02/06/2023]
Abstract
Although the central noradrenergic system has been shown to be involved in a number of behavioral and neurophysiological processes, the relation of these to its role in depressive illness has been difficult to define. The present review discusses the hypothesis that one of its chief functions that may be related to affective illness is the inhibition of behavioral activation, a prominent symptom of the disorder. This hypothesis is found to be consistent with most previous neuropsychopharmacological and immunohistochemical experiments on active behavior in rodents in a variety of experimental conditions using manipulation of neurotransmission at both locus coeruleus and forebrain adrenergic receptors. The findings support a mechanism in which high rates of noradrenergic neural activity suppress the neural activity of principal neurons in forebrain regions mediating active behavior. The suppression may be mediated through postsynaptic galaninergic and adrenergic receptors, and via the release of corticotrophin-releasing hormone. The hypothesis is consistent with clinical evidence for central noradrenergic system hyperactivity in depressives and with the view that this hyperactivity is a contributing etiological factor in the disorder. A similar mechanism may underlie the ability of the noradrenergic system to suppress seizure activity suggesting that inhibition of the spread of neural activation may be a unifying function.
Collapse
Affiliation(s)
- Eric A Stone
- Department of Psychiatry, New York University Langone School of Medicine, 550 First Ave., New York, NY 10016, USA.
| | | | | | | |
Collapse
|
155
|
Nobis WP, Kash TL, Silberman Y, Winder DG. β-Adrenergic receptors enhance excitatory transmission in the bed nucleus of the stria terminalis through a corticotrophin-releasing factor receptor-dependent and cocaine-regulated mechanism. Biol Psychiatry 2011; 69:1083-90. [PMID: 21334600 PMCID: PMC3090515 DOI: 10.1016/j.biopsych.2010.12.030] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 12/29/2010] [Accepted: 12/29/2010] [Indexed: 02/05/2023]
Abstract
BACKGROUND Evidence suggests that the noradrenergic and corticotrophin-releasing factor (CRF) systems play critical roles in relapse and stress-related behaviors. In particular, behavioral studies point to a serial signaling process initiated by β-adrenergic receptors that requires CRF receptor (CRFR)-dependent signaling in the bed nucleus of the stria terminalis (BNST) to produce stress-induced relapse to cocaine seeking. METHODS We used whole cell patch clamp recordings from acutely prepared mouse brain slices to examine the actions of β-adrenergic receptors and CRFR1 on excitatory transmission in BNST. We examined the effects of agonists of these receptors in slices prepared from naive, sham, and cocaine-conditioned mice. RESULTS β(1)-adrenergic receptor activation within the BNST produces an enhancement of excitatory synaptic transmission that requires CRFR1-dependent signaling. We show that chronic cocaine administration transiently disrupts β(1)-adrenergic- and CRFR1-dependent enhancement of glutamatergic transmission, that this disruption wanes with time, and that it can be reintroduced with a cocaine challenge. CONCLUSIONS In total, these studies identify a circuit mechanism within the BNST that may play an important role in CRF- and norepinephrine-regulated behaviors.
Collapse
Affiliation(s)
- William P. Nobis
- Neuroscience Graduate Program, Center Molecular Neuroscience, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Thomas L. Kash
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yuval Silberman
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Danny G. Winder
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Kennedy Center for Research on Human Development, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
156
|
du Hoffmann J, Kim JJ, Nicola SM. An inexpensive drivable cannulated microelectrode array for simultaneous unit recording and drug infusion in the same brain nucleus of behaving rats. J Neurophysiol 2011; 106:1054-64. [PMID: 21613588 DOI: 10.1152/jn.00349.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neurons are functionally segregated into discrete populations that perform specific computations. These computations, mediated by neuron-neuron electrochemical signaling, form the neural basis of behavior. Thus fundamental to a brain-based understanding of behavior is the precise determination of the contribution made by specific neurotransmitters to behaviorally relevant neural activity. To facilitate this understanding, we have developed a cannulated microelectrode array for use in behaving rats that enables simultaneous neural ensemble recordings and local infusion of drugs in the same brain nucleus. The system is inexpensive, easy to use, and produces robust and quantitatively reproducible drug effects on recorded neurons.
Collapse
Affiliation(s)
- Johann du Hoffmann
- Department of Psychiatry and Behavioral Science, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | |
Collapse
|
157
|
Navarro-Zaragoza J, Núñez C, Ruiz-Medina J, Laorden ML, Valverde O, Milanés MV. CRF₂ mediates the increased noradrenergic activity in the hypothalamic paraventricular nucleus and the negative state of morphine withdrawal in rats. Br J Pharmacol 2011; 162:851-62. [PMID: 20973778 DOI: 10.1111/j.1476-5381.2010.01090.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Recent evidence suggests that corticotropin-releasing factor (CRF) receptor signalling is involved in modulating the negative symptoms of opiate withdrawal. In this study, a series of experiments were performed to further characterize the role of CRF-type 2 receptor (CRF₂) signalling in opiate withdrawal-induced physical signs of dependence, hypothalamus-pituitary-adrenal (HPA) axis activation, enhanced noradrenaline (NA) turnover in the hypothalamic paraventricular nucleus (PVN) and tyrosine hydroxylase (TH) phosphorylation (activation), as well as CRF₂ expression in the nucleus of the solitary tract-A₂ noradrenergic cell group (NTS-A₂). EXPERIMENTAL APPROACH The contribution of CRF₂ signalling in opiate withdrawal was assessed by i.c.v. infusion of the selective CRF₂ antagonist, antisauvagine-30 (AS-30). Rats were implanted with two morphine (or placebo) pellets. Six days later, rats were pretreated with AS-30 or saline 10 min before naloxone and the physical signs of abstinence, the HPA axis activity, NA turnover, TH activation and CRF₂ expression were measured using immunoblotting, RIA, HPLC and immunohistochemistry. KEY RESULTS Rats pretreated with AS-30 showed decreased levels of somatic signs of naloxone-induced opiate withdrawal, but the corticosterone response was not modified. AS-30 attenuated the increased production of the NA metabolite, 3-methoxy-4-hydroxyphenylglycol, as well as the enhanced NA turnover observed in morphine-withdrawn rats. Finally, AS-30 antagonized the TH phosphorylation at Serine40 induced by morphine withdrawal. CONCLUSIONS AND IMPLICATIONS These results suggest that physical signs of opiate withdrawal, TH activation and stimulation of noradrenergic pathways innervating the PVN are modulated by CRF₂ signalling. Furthermore, they indicate a marginal role for the HPA axis in CRF₂-mediation of opiate withdrawal.
Collapse
|
158
|
Deyama S, Ide S, Kondoh N, Yamaguchi T, Yoshioka M, Minami M. Inhibition of noradrenaline release by clonidine in the ventral bed nucleus of the stria terminalis attenuates pain-induced aversion in rats. Neuropharmacology 2011; 61:156-60. [PMID: 21497164 DOI: 10.1016/j.neuropharm.2011.03.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 03/28/2011] [Accepted: 03/30/2011] [Indexed: 10/18/2022]
Abstract
Pain is an unpleasant sensory and emotional experience. The neural systems underlying the sensory component of pain have been studied extensively, but we are only beginning to understand those underlying the affective component of pain. Previously, we showed the pivotal role of noradrenergic transmission in the ventral part of the bed nucleus of the stria terminalis (vBNST) in the negative affective component of pain using a conditioned place paradigm. In this study, we examined the effect of local administration of clonidine, an α(2)-adrenoceptor agonist, into the vBNST on noradrenaline release and on conditioned place aversion (CPA) induced by an intraplantar formalin injection in male Sprague-Dawley rats. In vivo microdialysis showed that the formalin-induced increase in the extracellular noradrenaline level within the vBNST was significantly suppressed by clonidine (100 μM) perfusion through a microdialysis probe. Bilateral intra-vBNST injections of clonidine (1 and 10 nmol/side) dose-dependently attenuated formalin-induced CPA without reducing nociceptive behaviors. These results suggest that clonidine inhibits noradrenaline release by acting on α(2)-adrenoceptors located in the vBNST and thereby attenuates pain-induced aversion. α(2)-adrenoceptors in the vBNST play a pivotal role in the regulation of negative affective, but not the sensory, component of pain.
Collapse
Affiliation(s)
- Satoshi Deyama
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | | | | | | | | | | |
Collapse
|
159
|
Mena JD, Sadeghian K, Baldo BA. Induction of hyperphagia and carbohydrate intake by μ-opioid receptor stimulation in circumscribed regions of frontal cortex. J Neurosci 2011; 31:3249-60. [PMID: 21368037 PMCID: PMC3131113 DOI: 10.1523/jneurosci.2050-10.2011] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 12/09/2010] [Accepted: 01/02/2011] [Indexed: 12/22/2022] Open
Abstract
Frontal cortical regions are activated by food-associated stimuli, and this activation appears to be dysregulated in individuals with eating disorders. Nevertheless, frontal control of basic unconditioned feeding responses remains poorly understood. Here we show that hyperphagia can be driven by μ-opioid receptor stimulation in restricted regions of ventral medial prefrontal cortex (vmPFC) and orbitofrontal cortex. In both ad libitum-fed and food-restricted male Sprague Dawley rats, bilateral infusions of the μ-opioid agonist [d-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO) markedly increased intake of standard rat chow. When given a choice between palatable fat-enriched versus carbohydrate-enriched test diets, intra-vmPFC DAMGO infusions selectively increased carbohydrate intake, even in rats with a baseline fat preference. Rats also exhibited motor hyperactivity characterized by rapid switching between brief bouts of investigatory and ingestive behaviors. Intra-vmPFC DAMGO affected neither water intake nor nonspecific oral behavior. Similar DAMGO infusions into neighboring areas of lateral orbital or anterior motor cortex had minimal effects on feeding. Neither stimulation of vmPFC-localized δ-opioid, κ-opioid, dopaminergic, serotonergic, or noradrenergic receptors, nor antagonism of D1, 5HT1A, or α- or β-adrenoceptors, reproduced the profile of DAMGO effects. Muscimol-mediated inactivation of the vmPFC, and intra-vmPFC stimulation of κ-opioid receptors or blockade of 5-HT2A (5-hydroxytryptamine receptor 2A) receptors, suppressed motor activity and increased feeding bout duration-a profile opposite to that seen with DAMGO. Hence, μ-opioid-induced hyperphagia and carbohydrate intake can be elicited with remarkable pharmacological and behavioral specificity from discrete subterritories of the frontal cortex. These findings may have implications for understanding affect-driven feeding and loss of restraint in eating disorders.
Collapse
Affiliation(s)
| | - Ken Sadeghian
- Department of Psychiatry, University of Wisconsin-Madison, Madison, Wisconsin 53719
| | - Brian A. Baldo
- Neuroscience Training Program and
- Department of Psychiatry, University of Wisconsin-Madison, Madison, Wisconsin 53719
| |
Collapse
|
160
|
Brown ZJ, Nobrega JN, Erb S. Central injections of noradrenaline induce reinstatement of cocaine seeking and increase c-fos mRNA expression in the extended amygdala. Behav Brain Res 2011; 217:472-6. [DOI: 10.1016/j.bbr.2010.09.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 09/21/2010] [Accepted: 09/22/2010] [Indexed: 02/08/2023]
|
161
|
Krawczyk M, Georges F, Sharma R, Mason X, Berthet A, Bézard E, Dumont EC. Double-dissociation of the catecholaminergic modulation of synaptic transmission in the oval bed nucleus of the stria terminalis. J Neurophysiol 2011; 105:145-53. [PMID: 21047935 PMCID: PMC4011827 DOI: 10.1152/jn.00710.2010] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The bed nucleus of the stria terminalis (BST) is a cluster of nuclei within the extended amygdala, a forebrain macrostructure with extensive projection to motor nuclei of the hindbrain. The subnuclei of the BST coordinate autonomic, neuroendocrine, and somato-motor functions and receive robust neuromodulatory monoaminergic afferents, including 5-HT-, noradrenaline (NA)-, and dopamine (DA)-containing terminals. In contrast to 5-HT and NA, little is known about how DA modulates neuronal activity or synaptic transmission in the BST. DA-containing afferents to the BST originate in the ventral tegmental area, the periaqueducal gray, and the retrorubral field. They form a fairly diffuse input to the dorsolateral BST with dense terminal fields in the oval (ovBST) and juxtacapsular (jxBST) nuclei. The efferent-afferent connectivity of the BST suggests that it may play a key role in motivated behaviors, consistent with recent evidence that the dorsolateral BST is a target for drugs of abuse. This study describes the effects of DA on synaptic transmission in the ovBST. Whole cell voltage clamp recordings were performed on ovBST neurons in brain slices from adult rats in the presence or absence of exogenous DA and receptor-targeted agonists and antagonists. The results showed that DA selectively and exclusively reduced inhibitory synaptic transmission in the ovBST in a dose-dependent and D2-like dopamine receptor-dependent manner. DA also modulated excitatory synaptic transmission in a dose-dependent dependent manner. However, this effect was mediated by α2-noradrenergic receptors. Thus these data reveal a double dissociation in catecholaminergic regulation of excitatory and inhibitory synaptic transmission in the ovBST and may shed light on the mechanisms involved in neuropathological behaviors such as stress-induced relapse to consumption of drugs of abuse.
Collapse
Affiliation(s)
- Michal Krawczyk
- Department of Anesthesiology and Perioperative Medicine and Center for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | | | | | | | | | | | | |
Collapse
|
162
|
Buffalari DM, See RE. Inactivation of the bed nucleus of the stria terminalis in an animal model of relapse: effects on conditioned cue-induced reinstatement and its enhancement by yohimbine. Psychopharmacology (Berl) 2011; 213:19-27. [PMID: 20827461 PMCID: PMC3132192 DOI: 10.1007/s00213-010-2008-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 08/21/2010] [Indexed: 02/03/2023]
Abstract
RATIONALE Drug-associated cues and stress increase craving and lead to greater risk of relapse in abstinent drug users. Animal models of reinstatement of drug seeking have been utilized to study the neural circuitry by which either drug-associated cues or stress exposure elicit drug seeking. Recent evidence has shown a strong enhancing effect of yohimbine stress on subsequent cue-elicited reinstatement; however, there has been no examination of the neural substrates of this interactive effect. OBJECTIVES The current study examined whether inactivation of the bed nucleus of the stria terminalis (BNST), an area previously implicated in stress activation of drug seeking, would affect reinstatement of cocaine seeking caused by conditioned cues, yohimbine stress, or the combination of these factors. METHODS Male rats experienced daily IV cocaine self-administration, followed by extinction of lever responding in the absence of cocaine-paired cues. Reinstatement of responding was measured during presentation of cocaine-paired cues, following pretreatment with the pharmacological stressor, yohimbine (2.5 mg/kg, IP), or the combination of cues and yohimbine. RESULTS All three conditions led to reinstatement of cocaine seeking, with the highest responding seen after the combination of cues and yohimbine. Reversible inactivation of the BNST using the gamma-aminobutyric acid receptor agonists, baclofen + muscimol, significantly reduced all three forms of reinstatement. CONCLUSION These results demonstrate a role for the BNST in cocaine seeking elicited by cocaine-paired cues, and suggest the BNST as a key mediator for the interaction of stress and cues for the reinstatement of cocaine seeking.
Collapse
Affiliation(s)
- Deanne M. Buffalari
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ronald E. See
- Department of Neurosciences, Medical University of South Carolina, BSB416B, 173 Ashley Avenue, Charleston, SC 29425, USA
| |
Collapse
|
163
|
Bruijnzeel AW, Bishnoi M, van Tuijl IA, Keijzers KFM, Yavarovich KR, Pasek TM, Ford J, Alexander JC, Yamada H. Effects of prazosin, clonidine, and propranolol on the elevations in brain reward thresholds and somatic signs associated with nicotine withdrawal in rats. Psychopharmacology (Berl) 2010; 212:485-99. [PMID: 20697697 PMCID: PMC3042243 DOI: 10.1007/s00213-010-1970-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 07/20/2010] [Indexed: 11/26/2022]
Abstract
RATIONALE Tobacco withdrawal is characterized by a negative mood state and relatively mild somatic symptoms. Increased noradrenergic transmission has been reported to play an important role in opioid withdrawal, but little is known about the role of noradrenergic transmission in nicotine withdrawal. OBJECTIVES The aim of these experiments was to investigate the effects of prazosin, clonidine, and propranolol on the negative mood state and somatic signs associated with nicotine withdrawal in rats. METHODS A discrete-trial intracranial self-stimulation procedure was used to assess the negative affective state of nicotine withdrawal. Elevations in brain reward thresholds are indicative of a deficit in brain reward function. RESULTS In all the experiments, the nicotinic acetylcholine receptor antagonist mecamylamine (3 mg/kg) elevated the brain reward thresholds of the nicotine-treated rats and did not affect those of the control rats. The α1-adrenergic receptor antagonist prazosin (0.0625 and 0.125 mg/kg) dose-dependently attenuated the elevations in brain reward thresholds associated with precipitated nicotine withdrawal. The α2-adrenergic receptor agonist clonidine (10-40 μg/kg) and the nonselective β-adrenergic receptor antagonist propranolol (2.5-10 mg/kg) did not attenuate the elevations in brain reward thresholds associated with nicotine withdrawal. Furthermore, mecamylamine (2 mg/kg) induced more somatic signs in the nicotine-treated rats than in the control rats. Clonidine and propranolol, but not prazosin, decreased the total number of somatic signs associated with nicotine withdrawal. CONCLUSION Blockade of α1-adrenergic receptors attenuates the deficit in brain reward function associated with nicotine withdrawal. Antagonism of β-adrenergic receptors or stimulation of α2-adrenergic receptors attenuates the somatic symptoms of nicotine withdrawal.
Collapse
Affiliation(s)
- Adrie W Bruijnzeel
- Department of Psychiatry, McKnight Brain Institute, University of Florida, 100 S. Newell Dr., P.O. Box 100256, Gainesville, FL 32610, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Rinaman L. Hindbrain noradrenergic A2 neurons: diverse roles in autonomic, endocrine, cognitive, and behavioral functions. Am J Physiol Regul Integr Comp Physiol 2010; 300:R222-35. [PMID: 20962208 DOI: 10.1152/ajpregu.00556.2010] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Central noradrenergic (NA) signaling is broadly implicated in behavioral and physiological processes related to attention, arousal, motivation, learning and memory, and homeostasis. This review focuses on the A2 cell group of NA neurons, located within the hindbrain dorsal vagal complex (DVC). The intra-DVC location of A2 neurons supports their role in vagal sensory-motor reflex arcs and visceral motor outflow. A2 neurons also are reciprocally connected with multiple brain stem, hypothalamic, and limbic forebrain regions. The extra-DVC connections of A2 neurons provide a route through which emotional and cognitive events can modulate visceral motor outflow and also a route through which interoceptive feedback from the body can impact hypothalamic functions as well as emotional and cognitive processing. This review considers some of the hallmark anatomical and chemical features of A2 neurons, followed by presentation of evidence supporting a role for A2 neurons in modulating food intake, affective behavior, behavioral and physiological stress responses, emotional learning, and drug dependence. Increased knowledge about the organization and function of the A2 cell group and the neural circuits in which A2 neurons participate should contribute to a better understanding of how the brain orchestrates adaptive responses to the various threats and opportunities of life and should further reveal the central underpinnings of stress-related physiological and emotional dysregulation.
Collapse
Affiliation(s)
- Linda Rinaman
- Dept. of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA.
| |
Collapse
|
165
|
Hogle JM, Kaye JT, Curtin JJ. Nicotine withdrawal increases threat-induced anxiety but not fear: neuroadaptation in human addiction. Biol Psychiatry 2010; 68:719-25. [PMID: 20673878 PMCID: PMC2949532 DOI: 10.1016/j.biopsych.2010.06.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 05/17/2010] [Accepted: 06/08/2010] [Indexed: 11/16/2022]
Abstract
BACKGROUND Stress response neuroadaptation has been repeatedly implicated in animal addiction models for many drugs, including nicotine. Programmatic laboratory research that examines the stress response of nicotine-deprived humans is necessary to confirm that stress neuroadaptations observed in animal models generalize to humans. METHODS Two experiments tested the prediction that nicotine deprivation selectively increases startle response associated with anxiety during unpredictable threat but not fear during imminent, predictable threat. Dependent smokers (n = 117) were randomly assigned to 24-hour nicotine-deprived or nondeprived groups and participated in one of two experiments wherein electric shock was administered either unpredictably (noncontingent shock; Experiment 1) or predictably (cue-contingent shock; Experiment 2). RESULTS Nicotine deprivation increased overall startle response in Experiment 1, which involved unpredictable administration of shock. Age of first cigarette and years of daily smoking were significant moderators of this deprivation effect. Self-reported withdrawal symptoms also predicted startle response during unpredictable shock. In contrast, nicotine deprivation did not alter overall or fear-potentiated startle in Experiment 2, which involved predictable administration of shock. CONCLUSIONS These results provide evidence that startle response during unpredictable threat may be a biomarker of stress neuroadaptations among smokers in nicotine withdrawal. Contrast of results across unpredictable versus predictable shock experiments provides preliminary evidence that these stress neuroadaptations manifest selectively as anxiety during unpredictable threat rather than in every stressful context. Individual differences in unpredictable threat startle response associated with withdrawal symptoms, age of first cigarette, and years daily smoking link this laboratory biomarker to clinically relevant indexes of addiction risk and relapse.
Collapse
Affiliation(s)
- Joanne M Hogle
- Department of Psychology, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
166
|
Orexins in the midline thalamus are involved in the expression of conditioned place aversion to morphine withdrawal. Physiol Behav 2010; 102:42-50. [PMID: 20951152 DOI: 10.1016/j.physbeh.2010.10.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 10/01/2010] [Accepted: 10/08/2010] [Indexed: 01/07/2023]
Abstract
Previous studies have implicated the bed nucleus of the stria terminalis, central nucleus of the amygdala and the shell of the nucleus accumbens (collectively called the extended amygdala) as playing an important role in mediating the aversive emotion associated with opioid withdrawal. The paraventricular nucleus of the thalamus (PVT) provides a very dense input to the extended amygdala, and the PVT is densely innervated by orexin neurons, which appear to be involved in producing some of the physical and emotional effects associated with morphine withdrawal. In the present study, we confirm that the PVT is densely innervated by orexin fibers, whereas the regions of the extended amygdala associated with the effects of morphine withdrawal are poorly innervated. Microinjections of the orexin-1 receptor (OX1R) antagonist SB334867 or the orexin-2 receptor (OX2R) antagonist TCSOX229 at doses of 5.0 or 15.0 microg into the PVT region did not affect the acquisition of the conditioned place aversion (CPA) nor the physical effects produced by naloxone-precipitated morphine withdrawal. In contrast, microinjections of TCSOX229 (15.0 microg) in the PVT region significantly attenuated the expression of naloxone-induced CPA while microinjections of SB334867 at the same dose had no effect. The results from these experiments indicate a role for OX2R in the PVT on the expression of CPA associated with morphine withdrawal. Orexins may mediate the aversive effects of morphine withdrawal by engaging the extended amygdala indirectly through the action of orexins on the PVT.
Collapse
|
167
|
Effects of β-adrenoceptor antagonists on alcohol drinking by alcohol-dependent rats. Psychopharmacology (Berl) 2010; 212:431-9. [PMID: 20676608 PMCID: PMC2966867 DOI: 10.1007/s00213-010-1967-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 07/18/2010] [Indexed: 12/28/2022]
Abstract
RATIONALE Alcohol-dependent animals display enhanced stress responsivity, reward thresholds, and alcohol self-administration during alcohol withdrawal, and some of these aspects of alcohol dependence may be mediated by activation of brain norepinephrine (NE) systems. OBJECTIVES This study examined the effects of propranolol, a β-adrenoceptor antagonist, on operant alcohol-reinforced responding by alcohol-dependent and non-dependent rats. METHODS Adult male Wistar rats were trained to respond for alcohol in an operant conditioning paradigm on fixed-ratio-1 (FR-1) and progressive ratio (PR) reinforcement schedules. Rats were either made dependent on alcohol via chronic intermittent (14 h ON/10 h OFF) alcohol vapor inhalation or were not exposed to alcohol vapor. Rats were tested for the effects of propranolol (0-10 mg/kg) or nadolol (0-20 mg/kg) on operant alcohol-reinforced responding at the time point corresponding to 6-8 h withdrawal in dependent animals. RESULTS All doses of propranolol suppressed FR-1 operant alcohol-reinforced responding in alcohol-dependent rats, but only the highest dose suppressed FR-1 responding by controls. No dose of propranolol affected water responding. Nadolol did not affect operant behavior. Propranolol suppressed PR operant alcohol-reinforced responding across groups, an effect attributable to significant suppression of alcohol responding at the highest dose. CONCLUSIONS Following development of alcohol dependence, rats exhibit hypersensitivity to the suppressive effects of propranolol on operant alcohol-reinforced responding. This effect is mediated by central actions of the drug, is not attributable to motor effects, and may reflect activation of brain NE systems that contributes to withdrawal-induced negative emotional states and drives alcohol drinking in the dependent organism.
Collapse
|
168
|
Carvalho AF, Reyes ARS, Sterling RC, Unterwald E, Van Bockstaele EJ. Contribution of limbic norepinephrine to cannabinoid-induced aversion. Psychopharmacology (Berl) 2010; 211:479-91. [PMID: 20602088 PMCID: PMC3272334 DOI: 10.1007/s00213-010-1923-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 06/15/2010] [Indexed: 11/26/2022]
Abstract
RATIONALE The cannabinoid system has risen to the forefront in the development of novel treatments for a number of pathophysiological processes. However, significant side effects have been observed in clinical trials raising concerns regarding the potential clinical utility of cannabinoid-based agents. Understanding the neural circuits and neurochemical substrates impacted by cannabinoids will provide a better means of gaging their actions within the central nervous system that may contribute to the expression of unwanted side effects. OBJECTIVES In the present study, we investigated whether norepinephrine (NE) in the limbic forebrain is a critical determinant of cannabinoid receptor agonist-induced aversion and anxiety in rats. METHODS An immunotoxin lesion approach was combined with behavioral analysis using a place conditioning paradigm and the elevated zero maze. RESULTS Our results show that the non-selective CB1/CB2 receptor agonist, WIN 55,212-2, produced a significant place aversion in rats. Further, NE in the nucleus accumbens was critical for WIN 55,212-2-induced aversion but did not affect anxiety-like behaviors. Depletion of NE from the bed nucleus of the stria terminalis was ineffective in altering WIN 55,212-2-induced aversion and anxiety. CONCLUSIONS These results indicate that limbic, specifically accumbal, NE is required for cannabinoid-induced aversion but is not essential to cannabinoid-induced anxiety.
Collapse
Affiliation(s)
- Ana Franky Carvalho
- Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | | | | | |
Collapse
|
169
|
Parlato R, Cruz H, Otto C, Murtra P, Parkitna JR, Martin M, Bura SA, Begus-Nahrmann Y, von Bohlen und Halbach O, Maldonado R, Schütz G, Lüscher C. Effects of the cell type-specific ablation of the cAMP-responsive transcription factor in noradrenergic neurons on locus coeruleus firing and withdrawal behavior after chronic exposure to morphine. J Neurochem 2010; 115:563-73. [PMID: 20367754 DOI: 10.1111/j.1471-4159.2010.06709.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Repeated exposure to opiates leads to cellular and molecular changes and behavioral alterations reflecting a state of dependence. In noradrenergic neurons, cyclic AMP (cAMP)-dependent pathways are activated during opiate withdrawal, but their contribution to the activity of locus coeruleus noradrenergic neurons and behavioral manifestations remains controversial. Here, we test whether the cAMP-dependent transcription factors cAMP responsive element binding protein (CREB) and cAMP-responsive element modulator (CREM) in noradrenergic neurons control the cellular markers and the physical signs of morphine withdrawal in mice. Using the Cre/loxP system we ablated the Creb1 gene in noradrenergic neurons. To avoid adaptive effects because of compensatory up-regulation of CREM, we crossed the conditional Creb1 mutant mice with a Crem-/- line. We found that the enhanced expression of tyrosine hydroxylase normally observed during withdrawal was attenuated in CREB/CREM mutants. Moreover, the withdrawal-associated cellular hyperactivity and c-fos expression was blunted. In contrast, naloxone-precipitated withdrawal signs, such as jumping, paw tremor, tremor and mastication were preserved. We conclude by a specific genetic approach that the withdrawal-associated hyperexcitability of noradrenergic neurons depends on CREB/CREM activity in these neurons, but does not mediate several behavioral signs of morphine withdrawal.
Collapse
Affiliation(s)
- Rosanna Parlato
- Department of Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Erb S. Evaluation of the relationship between anxiety during withdrawal and stress-induced reinstatement of cocaine seeking. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:798-807. [PMID: 19969038 DOI: 10.1016/j.pnpbp.2009.11.025] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 11/13/2009] [Accepted: 11/30/2009] [Indexed: 01/30/2023]
Abstract
The initial termination of cocaine consumption in human addicts is associated with heightened anxiety states and low levels of craving. Craving, however, tends to increase progressively over time, remains high for extended periods of time, and can be exacerbated by stressors, leading to relapse. Laboratory rats, likewise, exhibit heightened states of anxiety after withdrawal from drug, and follow a time course of cocaine seeking that parallels the time course of craving reported in humans. In addition, laboratory rats show heightened susceptibility to relapse when exposed to stressors after extended periods of withdrawal, and exhibit persistent and heightened expressions of stress-induced anxiety. The general objective of this paper is to consider the relationship between anxiety states after withdrawal from cocaine and stress-induced reinstatement of cocaine seeking in laboratory rats, and to identify the neural substrates involved. The focus of the review is on studies addressing the roles of corticotropin-releasing factor (CRF) and noradrenaline pathways of the extended amygdala circuitry, and their direct or indirect interactions with the mesocorticolimbic dopamine system, in anxiety after withdrawal from cocaine and stress-induced reinstatement of cocaine seeking. Furthermore, the effects of time after withdrawal from cocaine and amount of cocaine exposure during self-administration on the activity of CRF, noradrenaline, and dopamine pathways of the extended amygdala and mesocorticolimbic systems will be considered. The review will highlight how changing levels of activity within these systems may serve to alter the nature of the relationship between anxiety and stress-induced reinstatement of cocaine seeking at different times after withdrawal from cocaine.
Collapse
Affiliation(s)
- Suzanne Erb
- Center for Neurobiology of Stress, Department of Psychology, University of Toronto Scarborough, Toronto, Ontario, Canada M1C 1A4.
| |
Collapse
|
171
|
Abstract
The locus coeruleus (LC) is regarded as a part of the central 'stress circuitry' because robust activation of the LC has been reported after stressful stimuli in experimental animals. A considerable amount of clinical evidence also suggests the relationship between the central noradrenergic (NAergic) system and fear/anxiety states or depression. However, previous animal studies have not been able to demonstrate unequivocally the involvement of the NAergic system in mediating fear or anxiety. The forebrain structures, including the hypothalamus, receive massive inputs from the medullary NAergic nuclei via the ventral NAergic bundle (VNAB). The VNAB has been implicated in the neuroendocrine stress axis mainly through its action on the corticotrophin-releasing factor neurones in the paraventricular nucleus of the hypothalamus. Novel tools were introduced that are capable of disrupting the NAergic system more selectively and/or thoroughly than the neurotoxins employed in previous studies: the anti-dopamine-beta hydroxylase (DBH)-saporin is an immunotoxin that is taken up from nerve endings and disrupt the NAergic neurones in a retrograde manner. The genetically DBH-depleted mice were also introduced, which lack endogenous noradrenaline. Owing to the rapid development of functional imaging technique, visualisation of the emotional phenomena has become possible in human subjects. Along with the advent of these technologies, endeavors have been continued to unravel the functional relevance of the central NAergic system to stress, anxiety and depression.
Collapse
Affiliation(s)
- K Itoi
- Division of Neuroendocrinology, Graduate School of Medicine, Tohoku University, Sendai 980-8579, Japan.
| | | |
Collapse
|
172
|
Abstract
The progression from recreational drug use to drug addiction impacts multiple neurobiological processes and can be conceptualized as a transition from positive to negative reinforcement mechanisms driving both drug-taking and drug-seeking behaviors. Neurobiological mechanisms for negative reinforcement, defined as drug taking that alleviates a negative emotional state, involve changes in the brain reward system and recruitment of brain stress (or antireward) systems within forebrain structures, including the extended amygdala. These systems are hypothesized to be dysregulated by excessive drug intake and to contribute to allostatic changes in reinforcement mechanisms associated with addiction. Points of intersection between positive and negative motivational circuitry may further drive the compulsivity of drug addiction but also provide a rich neurobiological substrate for therapeutic intervention.
Collapse
Affiliation(s)
- Scott Edwards
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 North Torrey Pines Road, SP30–2400, La Jolla, CA 92037, USA
| | - George F Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 North Torrey Pines Road, SP30–2400, La Jolla, CA 92037, USA
| |
Collapse
|
173
|
Núñez C, Martín F, Földes A, Luisa Laorden M, Kovács KJ, Victoria Milanés M. Induction of FosB/DeltaFosB in the brain stress system-related structures during morphine dependence and withdrawal. J Neurochem 2010; 114:475-87. [PMID: 20438612 DOI: 10.1111/j.1471-4159.2010.06765.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The transcription factor DeltaFosB is induced in the nucleus accumbens (NAc) by drugs of abuse. This study was designed to evaluate the possible modifications in FosB/DeltaFosB expression in both hypothalamic and extrahypothalamic brain stress system during morphine dependence and withdrawal. Rats were made dependent on morphine and, on day 8, were injected with saline or naloxone. Using immunohistochemistry and western blot, the expression of FosB/DeltaFosB, tyrosine hydroxylase (TH), corticotropin-releasing factor (CRF) and pro-dynorphin (DYN) was measured in different nuclei from the brain stress system in morphine-dependent rats and after morphine withdrawal. Additionally, we studied the expression of FosB/DeltaFosB in CRF-, TH- and DYN-positive neurons. FosB/DeltaFosB was induced after chronic morphine administration in the parvocellular part of the hypothalamic paraventricular nucleus (PVN), NAc-shell, bed nucleus of the stria terminalis, central amygdala and A(2) noradrenergic part of the nucleus tractus solitarius (NTS-A(2)). Morphine dependence and withdrawal evoked an increase in FosB/DeltaFosB-TH and FosB/DeltaFosB-CRF double labelling in NTS-A(2) and PVN, respectively, besides an increase in TH levels in NTS-A(2) and CRF expression in PVN. These data indicate that neuroadaptation to addictive substances, observed as accumulation of FosB/DeltaFosB, is not limited to the reward circuits but may also manifest in other brain regions, such as the brain stress system, which have been proposed to be directly related to addiction.
Collapse
Affiliation(s)
- Cristina Núñez
- Department of Pharmacology, University School of Medicine, Murcia, Spain.
| | | | | | | | | | | |
Collapse
|
174
|
Gallego X, Murtra P, Zamalloa T, Canals JM, Pineda J, Amador-Arjona A, Maldonado R, Dierssen M. Increased opioid dependence in a mouse model of panic disorder. Front Behav Neurosci 2010; 3:60. [PMID: 20204153 PMCID: PMC2831706 DOI: 10.3389/neuro.08.060.2009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Accepted: 12/16/2009] [Indexed: 11/17/2022] Open
Abstract
Panic disorder is a highly prevalent neuropsychiatric disorder that shows co-occurrence with substance abuse. Here, we demonstrate that TrkC, the high-affinity receptor for neurotrophin-3, is a key molecule involved in panic disorder and opiate dependence, using a transgenic mouse model (TgNTRK3). Constitutive TrkC overexpression in TgNTRK3 mice dramatically alters spontaneous firing rates of locus coeruleus (LC) neurons and the response of the noradrenergic system to chronic opiate exposure, possibly related to the altered regulation of neurotrophic peptides observed. Notably, TgNTRK3 LC neurons showed an increased firing rate in saline-treated conditions and profound abnormalities in their response to met5-enkephalin. Behaviorally, chronic morphine administration induced a significantly increased withdrawal syndrome in TgNTRK3 mice. In conclusion, we show here that the NT-3/TrkC system is an important regulator of neuronal firing in LC and could contribute to the adaptations of the noradrenergic system in response to chronic opiate exposure. Moreover, our results indicate that TrkC is involved in the molecular and cellular changes in noradrenergic neurons underlying both panic attacks and opiate dependence and support a functional endogenous opioid deficit in panic disorder patients.
Collapse
Affiliation(s)
- Xavier Gallego
- Genes and Disease Program, Genomic Regulation Center-CRG, Barcelona Biomedical Research Park-PRBB and CIBER de Enfermedades Raras Barcelona, Catalonia, Spain
| | | | | | | | | | | | | | | |
Collapse
|
175
|
Koob GF. The role of CRF and CRF-related peptides in the dark side of addiction. Brain Res 2010; 1314:3-14. [PMID: 19912996 PMCID: PMC2819562 DOI: 10.1016/j.brainres.2009.11.008] [Citation(s) in RCA: 198] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 11/02/2009] [Accepted: 11/03/2009] [Indexed: 12/13/2022]
Abstract
Drug addiction is a chronically relapsing disorder characterized by a compulsion to seek and take drugs, the development of dependence, and the manifestation of a negative emotional state when the drug is removed. Activation of brain stress systems is hypothesized to be a key element of the negative emotional state produced by dependence that drives drug-seeking through negative reinforcement mechanisms, defined as the "dark side" of addiction. The focus of the present review is on the role of corticotropin-releasing factor (CRF) and CRF-related peptides in the dark side of addiction. CRF is a key mediator of the hormonal, autonomic, and behavior responses to stressors. Emphasis is placed on the role of CRF in extrahypothalamic systems in the extended amygdala, including the central nucleus of the amygdala, bed nucleus of the stria terminalis, and a transition area in the shell of the nucleus accumbens, in the dark side of addiction. The urocortin/CRF(2) systems have been less explored, but results suggest their role in the neuroadaptation associated with chronic drug use, sometimes in opposition to the effects produced by the CRF(1) receptor. Compelling evidence argues that the CRF stress system, including its activation of the hypothalamic-pituitary-adrenal axis, plays a key role in engaging the transition to dependence and maintaining dependence once it is initiated. Understanding the role of the CRF systems in addiction not only provides insight into the neurobiology of the dark side of addiction, but also provides novel targets for identifying vulnerability to addiction and the treatment of addiction.
Collapse
Affiliation(s)
- George F Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 North Torrey Pines Road, SP30-2400 La Jolla, CA 92037, USA.
| |
Collapse
|
176
|
Forebrain PENK and PDYN gene expression levels in three inbred strains of mice and their relationship to genotype-dependent morphine reward sensitivity. Psychopharmacology (Berl) 2010; 208:291-300. [PMID: 19997907 DOI: 10.1007/s00213-009-1730-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 11/10/2009] [Indexed: 12/18/2022]
Abstract
RATIONALE Vulnerability to drug abuse disorders is determined not only by environmental but also by genetic factors. A body of evidence suggests that endogenous opioid peptide systems may influence rewarding effects of addictive substances, and thus, their individual expression levels may contribute to drug abuse liability. OBJECTIVES The aim of our study was to assess whether basal genotype-dependent brain expression of opioid propeptides genes can influence sensitivity to morphine reward. METHODS Experiments were performed on inbred mouse strains C57BL/6J, DBA/2J, and SWR/J, which differ markedly in responses to morphine administration: DBA/2J and SWR/J show low and C57BL/6J high sensitivity to opioid reward. Proenkephalin (PENK) and prodynorphin (PDYN) gene expression was measured by in situ hybridization in brain regions implicated in addiction. The influence of the kappa opioid receptor antagonist nor-binaltorphimine (nor-BNI), which attenuates effects of endogenous PDYN-derived peptides, on rewarding actions of morphine was studied using the conditioned place preference (CPP) paradigm. RESULTS DBA/2J and SWR/J mice showed higher levels of PDYN and lower levels of PENK messenger RNA in the nucleus accumbens than the C57BL/6J strain. Pretreatment with nor-BNI enhanced morphine-induced CPP in the opioid-insensitive DBA/2J and SWR/J strains. CONCLUSIONS Our results demonstrate that inter-strain differences in PENK and PDYN genes expression in the nucleus accumbens parallel sensitivity of the selected mouse strains to rewarding effects of morphine. They suggest that high expression of PDYN may protect against drug abuse by limiting drug-produced reward, which may be due to dynorphin-mediated modulation of dopamine release in the nucleus accumbens.
Collapse
|
177
|
Puente N, Elezgarai I, Lafourcade M, Reguero L, Marsicano G, Georges F, Manzoni OJ, Grandes P. Localization and function of the cannabinoid CB1 receptor in the anterolateral bed nucleus of the stria terminalis. PLoS One 2010; 5:e8869. [PMID: 20111610 PMCID: PMC2810340 DOI: 10.1371/journal.pone.0008869] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Accepted: 01/04/2010] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The bed nucleus of the stria terminalis (BNST) is involved in behaviors related to natural reward, drug addiction and stress. In spite of the emerging role of the endogenous cannabinoid (eCB) system in these behaviors, little is known about the anatomy and function of this system in the anterolateral BNST (alBNST). The aim of this study was to provide a detailed morphological characterization of the localization of the cannabinoid 1 (CB1) receptor a necessary step toward a better understanding of the physiological roles of the eCB system in this region of the brain. METHODOLOGY/PRINCIPAL FINDINGS We have combined anatomical approaches at the confocal and electron microscopy level to ex-vivo electrophysiological techniques. Here, we report that CB1 is localized on presynaptic membranes of about 55% of immunopositive synaptic terminals for the vesicular glutamate transporter 1 (vGluT1), which contain abundant spherical, clear synaptic vesicles and make asymmetrical synapses with alBNST neurons. About 64% of vGluT1 immunonegative synaptic terminals show CB1 immunolabeling. Furthermore, 30% and 35% of presynaptic boutons localize CB1 in alBNST of conditional mutant mice lacking CB1 mainly from GABAergic neurons (GABA-CB1-KO mice) and mainly from cortical glutamatergic neurons (Glu-CB1-KO mice), respectively. Extracellular field recordings and whole cell patch clamp in the alBNST rat brain slice preparation revealed that activation of CB1 strongly inhibits excitatory and inhibitory synaptic transmission. CONCLUSIONS/SIGNIFICANCE This study supports the anterolateral BNST as a potential neuronal substrate of the effects of cannabinoids on stress-related behaviors.
Collapse
Affiliation(s)
- Nagore Puente
- Department of Neurosciences, Faculty of Medicine and Dentistry, Basque Country University, Bilbao, Spain
| | - Izaskun Elezgarai
- Department of Neurosciences, Faculty of Medicine and Dentistry, Basque Country University, Bilbao, Spain
| | - Mathieu Lafourcade
- INSERM U862 Equipe “Physiopathologie de la Transmission et de la Plasticité Synaptique”, Bordeaux, France
| | - Leire Reguero
- Department of Neurosciences, Faculty of Medicine and Dentistry, Basque Country University, Bilbao, Spain
| | - Giovanni Marsicano
- “Endocannabinoids and Neuroadaptation”, INSERM U862 NeuroCentre Magendie, Université Bordeaux 2, Bordeaux, France
| | - François Georges
- INSERM U862 Equipe “Physiopathologie de la Transmission et de la Plasticité Synaptique”, Bordeaux, France
| | - Olivier J. Manzoni
- INSERM U862 Equipe “Physiopathologie de la Transmission et de la Plasticité Synaptique”, Bordeaux, France
| | - Pedro Grandes
- Department of Neurosciences, Faculty of Medicine and Dentistry, Basque Country University, Bilbao, Spain
| |
Collapse
|
178
|
Koob GF, Volkow ND. Neurocircuitry of addiction. Neuropsychopharmacology 2010; 35:217-38. [PMID: 19710631 PMCID: PMC2805560 DOI: 10.1038/npp.2009.110] [Citation(s) in RCA: 3648] [Impact Index Per Article: 243.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2009] [Revised: 07/13/2009] [Accepted: 07/14/2009] [Indexed: 12/29/2022]
Abstract
Drug addiction is a chronically relapsing disorder that has been characterized by (1) compulsion to seek and take the drug, (2) loss of control in limiting intake, and (3) emergence of a negative emotional state (eg, dysphoria, anxiety, irritability) reflecting a motivational withdrawal syndrome when access to the drug is prevented. Drug addiction has been conceptualized as a disorder that involves elements of both impulsivity and compulsivity that yield a composite addiction cycle composed of three stages: 'binge/intoxication', 'withdrawal/negative affect', and 'preoccupation/anticipation' (craving). Animal and human imaging studies have revealed discrete circuits that mediate the three stages of the addiction cycle with key elements of the ventral tegmental area and ventral striatum as a focal point for the binge/intoxication stage, a key role for the extended amygdala in the withdrawal/negative affect stage, and a key role in the preoccupation/anticipation stage for a widely distributed network involving the orbitofrontal cortex-dorsal striatum, prefrontal cortex, basolateral amygdala, hippocampus, and insula involved in craving and the cingulate gyrus, dorsolateral prefrontal, and inferior frontal cortices in disrupted inhibitory control. The transition to addiction involves neuroplasticity in all of these structures that may begin with changes in the mesolimbic dopamine system and a cascade of neuroadaptations from the ventral striatum to dorsal striatum and orbitofrontal cortex and eventually dysregulation of the prefrontal cortex, cingulate gyrus, and extended amygdala. The delineation of the neurocircuitry of the evolving stages of the addiction syndrome forms a heuristic basis for the search for the molecular, genetic, and neuropharmacological neuroadaptations that are key to vulnerability for developing and maintaining addiction.
Collapse
Affiliation(s)
- George F Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | |
Collapse
|
179
|
Glass MJ. The role of functional postsynaptic NMDA receptors in the central nucleus of the amygdala in opioid dependence. VITAMINS AND HORMONES 2010; 82:145-66. [PMID: 20472137 DOI: 10.1016/s0083-6729(10)82008-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Activation of ionotropic N-methyl-D-aspartate (NMDA)-type glutamate receptors in limbic system nuclei, such as the central nucleus of the amygdala (CeA), plays an essential role in autonomic, behavioral, and affective processes that are profoundly impacted by exposure to opioids. However, the heterogeneous ultrastructural distribution of the NMDA receptor, its complex pharmacology, and the paucity of genetic models have hampered the development of linkages between functional amygdala NMDA receptors and opioid dependence. To overcome these shortcomings, high-resolution imaging and molecular pharmacology were used to (1) Identify the ultrastructural localization of the essential NMDA-NR1 receptor (NR1) subunit and its relationship to the mu-opioid receptor (microOR), the major cellular target of abused opioids like morphine, in the CeA and (2) Determine the effect of CeA NR1 deletion on the physical, and particularly, psychological aspects of opioid dependence. Combined immunogold and immuoperoxidase electron microscopic analysis showed that NR1 was prominently expressed in postsynaptic (i.e., somata, dendrites) locations of CeA neurons, where they were also frequently colocalized with the microOR. A spatial-temporal deletion of NR1 in postsynaptic sites of CeA neurons was produced by local microinjection of a neurotropic recombinant adeno-associated virus (rAAV), expressing the green fluorescent protein (GFP) reporter and Cre recombinase (rAAV-GFP-Cre), in adult "floxed" NR1 (fNR1) mice. Mice with deletion of NR1 in the CeA showed no obvious impairments in sensory, motor, or nociceptive function. In addition, when administered chronic morphine, these mice also displayed an acute physical withdrawal syndrome precipitated by naloxone. However, opioid-dependent CeA NR1 knockout mice failed to exhibit a conditioned place aversion induced by naloxone-precipitated withdrawal. These results indicate that postsynaptic NMDA receptor activity in central amygdala neurons is required for the expression of a learned affective behavior associated with opioid withdrawal. The neurogenetic dissociation of physical and psychological properties of opioid dependence demonstrates the value of combined ultrastructural analysis and molecular pharmacology in clarifying the neurobiological mechanisms subserving opioid-mediated plasticity.
Collapse
Affiliation(s)
- Michael J Glass
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, USA
| |
Collapse
|
180
|
Bruijnzeel AW. kappa-Opioid receptor signaling and brain reward function. BRAIN RESEARCH REVIEWS 2009; 62:127-46. [PMID: 19804796 PMCID: PMC2787673 DOI: 10.1016/j.brainresrev.2009.09.008] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 09/28/2009] [Accepted: 09/28/2009] [Indexed: 02/06/2023]
Abstract
The dynorphin-like peptides have profound effects on the state of the brain reward system and human and animal behavior. The dynorphin-like peptides affect locomotor activity, food intake, sexual behavior, anxiety-like behavior, and drug intake. Stimulation of kappa-opioid receptors, the endogenous receptor for the dynorphin-like peptides, inhibits dopamine release in the striatum (nucleus accumbens and caudate putamen) and induces a negative mood state in humans and animals. The administration of drugs of abuse increases the release of dopamine in the striatum and mediates the concomitant release of dynorphin-like peptides in this brain region. The reviewed studies suggest that chronic drug intake leads to an upregulation of the brain dynorphin system in the striatum and in particular in the dorsal part of the striatum/caudate putamen. This might inhibit drug-induced dopamine release and provide protection against the neurotoxic effects of high dopamine levels. After the discontinuation of chronic drug intake these neuroadaptations remain unopposed which has been suggested to contribute to the negative emotional state associated with drug withdrawal and increased drug intake. kappa-Opioid receptor agonists have also been shown to inhibit calcium channels. Calcium channel inhibitors have antidepressant-like effects and inhibit the release of norepinephrine. This might explain that in some studies kappa-opioid receptor agonists attenuate nicotine and opioid withdrawal symptomatology. A better understanding of the role of dynorphins in the regulation of brain reward function might contribute to the development of novel treatments for mood disorders and other disorders that stem from a dysregulation of the brain reward system.
Collapse
Affiliation(s)
- Adrie W Bruijnzeel
- Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, 32610, USA.
| |
Collapse
|
181
|
Le Merrer J, Becker JAJ, Befort K, Kieffer BL. Reward processing by the opioid system in the brain. Physiol Rev 2009; 89:1379-412. [PMID: 19789384 DOI: 10.1152/physrev.00005.2009] [Citation(s) in RCA: 714] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The opioid system consists of three receptors, mu, delta, and kappa, which are activated by endogenous opioid peptides processed from three protein precursors, proopiomelanocortin, proenkephalin, and prodynorphin. Opioid receptors are recruited in response to natural rewarding stimuli and drugs of abuse, and both endogenous opioids and their receptors are modified as addiction develops. Mechanisms whereby aberrant activation and modifications of the opioid system contribute to drug craving and relapse remain to be clarified. This review summarizes our present knowledge on brain sites where the endogenous opioid system controls hedonic responses and is modified in response to drugs of abuse in the rodent brain. We review 1) the latest data on the anatomy of the opioid system, 2) the consequences of local intracerebral pharmacological manipulation of the opioid system on reinforced behaviors, 3) the consequences of gene knockout on reinforced behaviors and drug dependence, and 4) the consequences of chronic exposure to drugs of abuse on expression levels of opioid system genes. Future studies will establish key molecular actors of the system and neural sites where opioid peptides and receptors contribute to the onset of addictive disorders. Combined with data from human and nonhuman primate (not reviewed here), research in this extremely active field has implications both for our understanding of the biology of addiction and for therapeutic interventions to treat the disorder.
Collapse
Affiliation(s)
- Julie Le Merrer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Département Neurobiologie et Génétique, Illkirch, France
| | | | | | | |
Collapse
|
182
|
Park J, Kile BM, Wightman RM. In vivo voltammetric monitoring of norepinephrine release in the rat ventral bed nucleus of the stria terminalis and anteroventral thalamic nucleus. Eur J Neurosci 2009; 30:2121-33. [PMID: 20128849 DOI: 10.1111/j.1460-9568.2009.07005.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The role and contribution of the dense noradrenergic innervation in the ventral bed nucleus of the stria terminalis (vBNST) and anteroventral thalamic nucleus (AV) to biological function and animal behaviors is poorly understood due to the small size of these nuclei. The aim of this study was to compare norepinephrine release and uptake in the vBNST with that in the AV of anesthetized rats. Measurements were made in vivo with fast-scan cyclic voltammetry following electrical stimulation of noradrenergic projection pathways, either the dorsal noradrenergic bundle (DNB) or the ventral noradrenergic bundle (VNB). The substance detected was identified as norepinephrine based upon voltammetric, anatomical, neurochemical and pharmacological evidence. Fast-scan cyclic voltammetry enables the selective monitoring of local norepinephrine overflow in the vBNST evoked by the stimulation of either the DNB or the VNB while norepinephrine in the AV was only evoked by DNB stimulation. The alpha2-adrenoceptor antagonist yohimbine and the norepinephrine uptake inhibitor desipramine increased norepinephrine overflow and slowed its disappearance in both regions. However, control of extracellular norepinephrine by both autoreceptors and uptake was greater in the AV. The greater control exerted by autoreceptors and uptake in the AV resulted in reduced extracellular concentration compared with the v BNST when large numbers of stimulation pulses were employed. The differences in noradrenergic transmission observed in the terminal fields of the v BNST and the AV may differentially regulate activity in these two regions that both contain high densities of norepinephrine terminals.
Collapse
Affiliation(s)
- Jinwoo Park
- Department of Chemistry and Neuroscience Center, University of North Carolina , Chapel Hill, NC 27599-3290, USA
| | | | | |
Collapse
|
183
|
Jalabert M, Aston-Jones G, Herzog E, Manzoni O, Georges F. Role of the bed nucleus of the stria terminalis in the control of ventral tegmental area dopamine neurons. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:1336-46. [PMID: 19616054 PMCID: PMC3635540 DOI: 10.1016/j.pnpbp.2009.07.010] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 07/04/2009] [Accepted: 07/08/2009] [Indexed: 11/25/2022]
Abstract
Projections from neurons of the bed nucleus of the stria terminalis (BST) to the ventral tegmental area (VTA) are crucial to behaviors related to reward and motivation. Over the past few years, we have undertaken a series of studies to understand: 1) how excitatory inputs regulate in vivo excitable properties of BST neurons, and 2) how BST inputs in turn modulate neuronal activity of dopamine neurons in VTA. Using in vivo extracellular recording techniques in anesthetized rats and tract-tracing approaches, we have demonstrated that inputs from the infralimbic cortex and the ventral subiculum exert a strong excitatory influence on BST neurons projecting to the VTA. Thus, the BST is uniquely positioned to receive emotional and learning-associated informations and to integrate these into the reward/motivation circuitry. We will discuss how changes in the activity of BST neurons projecting to the VTA could participate in the development or exacerbation of psychiatric conditions such as drug addiction.
Collapse
Affiliation(s)
- Marion Jalabert
- INSERM, U862, Neurocentre Magendie, Pathophysiology of synaptic plasticity group, Bordeaux, F-33000, France,Université de Bordeaux, Bordeaux, F-33000, France
| | - Gary Aston-Jones
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Ave., Suite 403 BSB, MSC 510, Charleston, SC 29425-5100, USA
| | - Etienne Herzog
- INSERM U952, 9 Quai St Bernard, 75005, Paris, France,CNRS UMR 7224, 9 Quai St Bernard, 75005, Paris, France,Université Pierre et Marie Curie (UPMC), Paris 06, Paris, France
| | - Olivier Manzoni
- INSERM, U862, Neurocentre Magendie, Pathophysiology of synaptic plasticity group, Bordeaux, F-33000, France,Université de Bordeaux, Bordeaux, F-33000, France
| | - François Georges
- INSERM, U862, Neurocentre Magendie, Pathophysiology of synaptic plasticity group, Bordeaux, F-33000, France,Université de Bordeaux, Bordeaux, F-33000, France,Authors for correspondence at above address: Phone: +33 557-57-40-99, Fax: +33 557-57-37-76,
| |
Collapse
|
184
|
Poulin JF, Arbour D, Laforest S, Drolet G. Neuroanatomical characterization of endogenous opioids in the bed nucleus of the stria terminalis. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:1356-65. [PMID: 19583989 DOI: 10.1016/j.pnpbp.2009.06.021] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2009] [Revised: 06/26/2009] [Accepted: 06/29/2009] [Indexed: 12/21/2022]
Abstract
Numerous neuroanatomical data indicate that the bed nucleus of the stria terminalis (BST) provides an interface between cortical and amygdaloid neurons, and effector neurons modulating motor, autonomic and neuroendocrine responses. Distinct divisions of the BST may be involved in stress response, homeostatic regulation, nociception, and motivated behaviors. Endogenous opioid peptides and receptors are expressed in the BST, but their exact distribution is poorly characterized. The present study used in situ hybridization in order to characterize the endogenous opioid system of the BST, focusing on both enkephalin and dynorphin neuropeptides, as well as their respective receptors (mu, delta, and kappa opioid receptors). We report that preprodynorphin mRNA is observed in distinct nuclei of the BST, namely the fusiform, oval and anterior lateral nuclei. In contrast, there is a widespread expression of preproenkephalin mRNA in both anterior and posterior divisions of the BST. Similarly, mu and kappa opioid receptors are broadly expressed in the BST, whereas delta opioid receptor mRNA was observed only in the principal nucleus. For further characterization of enkephalin-expressing neurons of the BST, we performed a double fluorescent in situ hybridization in order to reveal the coexpression of enkephalin peptides and markers of GABAergic and glutamatergic neurons. Although most neurons of the BST are GABAergic, there is also a modest population of glutamatergic cells expressing vesicular glutamate transporter 2 (VGLUT2) in specific nuclei of the BST. Finally, we identified a previously unreported population of enkephalinergic neurons expressing VGLUT2, which is principally located in the posterior BST.
Collapse
Affiliation(s)
- Jean-François Poulin
- Centre de recherche du CHUQ (CHUL), Neurosciences, Université Laval, Québec, QC, Canada.
| | | | | | | |
Collapse
|
185
|
Walker DL, Miles LA, Davis M. Selective participation of the bed nucleus of the stria terminalis and CRF in sustained anxiety-like versus phasic fear-like responses. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:1291-308. [PMID: 19595731 PMCID: PMC2783512 DOI: 10.1016/j.pnpbp.2009.06.022] [Citation(s) in RCA: 268] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Revised: 06/26/2009] [Accepted: 06/29/2009] [Indexed: 01/09/2023]
Abstract
The medial division of the central nucleus of the amygdala (CeA(M)) and the lateral division of the bed nucleus of the stria terminalis (BNST(L)) are closely related. Both receive projections from the basolateral amygdala (BLA) and both project to brain areas that mediate fear-influenced behaviors. In contrast to CeA(M) however, initial attempts to implicate the BNST in conditioned fear responses were largely unsuccessful. More recent studies have shown that the BNST does participate in some types of anxiety and stress responses. Here, we review evidence suggesting that the CeA(M) and BNST(L) are functionally complementary, with CeA(M) mediating short- but not long-duration threat responses (i.e., phasic fear) and BNST(L) mediating long- but not short-duration responses (sustained fear or 'anxiety'). We also review findings implicating the stress-related peptide corticotropin-releasing factor (CRF) in sustained but not phasic threat responses, and attempt to integrate these findings into a neural circuit model which accounts for these and related observations.
Collapse
Affiliation(s)
- D L Walker
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA 30329, USA.
| | | | | |
Collapse
|
186
|
McElligott ZA, Winder DG. Modulation of glutamatergic synaptic transmission in the bed nucleus of the stria terminalis. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:1329-35. [PMID: 19524008 PMCID: PMC2783684 DOI: 10.1016/j.pnpbp.2009.05.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 05/12/2009] [Accepted: 05/14/2009] [Indexed: 11/27/2022]
Abstract
Glutamate, catecholamine and neuropeptide signaling within the bed nucleus of the stria terminalis (BNST) have all been identified as key participants in anxiety-like behaviors and behaviors related to withdrawal from exposure to substances of abuse. The BNST is thought to serve as a key relay between limbic cognitive centers and reward, stress and anxiety nuclei. Human studies and animal models have demonstrated that stressors and drugs of abuse can result in long term behavioral modifications that can culminate in psychological diseases such as addiction and post-traumatic stress disorder. The ability of catecholamines and neuropeptides to influence synaptic glutamatergic transmission (stemming from cognitive centers) within the BNST may have profound consequences over these behaviors. In this review we highlight studies examining synaptic plasticity and modulation of excitatory transmission within the BNST, emphasizing how such modulation may result in alterations in anxiety and reward related behavior.
Collapse
Affiliation(s)
| | - Danny G. Winder
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
- Kennedy Center For Human Development, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
187
|
Francesconi W, Berton F, Koob GF, Sanna PP. Intrinsic neuronal plasticity in the juxtacapsular nucleus of the bed nuclei of the stria terminalis (jcBNST). Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:1347-55. [PMID: 19683025 PMCID: PMC2935256 DOI: 10.1016/j.pnpbp.2009.08.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 08/04/2009] [Accepted: 08/04/2009] [Indexed: 11/30/2022]
Abstract
The juxtacapsular nucleus of the anterior division of the BNST (jcBNST) receives robust glutamatergic projections from the basolateral nucleus of the amygdala (BLA), the postpiriform transition area, and the insular cortex as well as dopamine (DA) inputs from the midbrain. In turn the jcBNST sends GABAergic projections to the medial division of the central nucleus of the amygdala (CEAm) as well as other brain regions. We recently described a form of long-term potentiation of the intrinsic excitability (LTP-IE) of neurons of the juxtacapsular nucleus of BNST (jcBNST) in response to high-frequency stimulation (HFS) of the stria terminalis that was impaired during protracted withdrawal from alcohol, cocaine, and heroin and in rats chronically treated with corticotropin-releasing factor (CRF) intracerebroventricularly. Here we show that DAergic neurotransmission is required for the induction of LTP-IE of jcBNST neurons through dopamine (DA) D1 receptors. Thus, activation of the central CRF stress system and altered DAergic neurotransmission during protracted withdrawal from alcohol and drugs of abuse may contribute to the disruption of LTP-IE in the jcBNST. Impairment of this form of intrinsic neuronal plasticity in the jcBNST could result in inadequate neuronal integration and reduced inhibition of the CEA, contributing to the negative affective state that characterizes protracted abstinence in post-dependent individuals. These results provide a novel neurobiological target for vulnerability to alcohol and drug dependence.
Collapse
Affiliation(s)
- Walter Francesconi
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, California 92037, USA.
| | - Fulvia Berton
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, California 92037, USA
| | - George F. Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, California 92037, USA
| | - Pietro Paolo Sanna
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, California 92037, USA,Correspondence: W. Francesconi () or P.P. Sanna ()
| |
Collapse
|
188
|
Dumont EC. What is the bed nucleus of the stria terminalis? Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:1289-90. [PMID: 19602427 PMCID: PMC4011829 DOI: 10.1016/j.pnpbp.2009.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 07/08/2009] [Indexed: 10/20/2022]
|
189
|
Involvement of actin rearrangements within the amygdala and the dorsal hippocampus in aversive memories of drug withdrawal in acute morphine-dependent rats. J Neurosci 2009; 29:12244-54. [PMID: 19793983 DOI: 10.1523/jneurosci.1970-09.2009] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aversive memories of drug withdrawal can generate a motivational state leading to compulsive drug taking. Changes in synaptic plasticity may be involved in the formation of aversive memories. Dynamic rearrangement of the cytoskeletal actin, a major structural component of the dendritic spine, regulates synaptic plasticity. Here, the potential involvement of actin rearrangements in the induction of aversive memories of morphine withdrawal was examined. We found that lesions of the amygdala or dorsal hippocampus (DH) but not nucleus accumbens (NAc) impaired conditioned place aversion (CPA) of acute morphine-dependent rats. Accordingly, conditioned morphine withdrawal induced actin rearrangements in the amygdala and the DH but not in the NAc. In addition, we found that conditioned morphine withdrawal also increased activity-regulated cytoskeletal-associated protein (Arc) expression in the amygdala but not in the DH, although actin rearrangements were observed in both areas. We further found that inhibition of actin rearrangements by intra-amygdala or intra-DH injections of latrunculin A, an inhibitor of actin polymerization, significantly attenuated CPA. Furthermore, we found that manipulation of amygdala beta-adrenoceptor activity by its antagonist propranolol and agonist clenbuterol differentially altered actin rearrangements in the DH. Therefore, our findings reveal that actin rearrangements in the amygdala and the DH are required for the acquisition and consolidation of the aversive memories of drug withdrawal and that the beta-noradrenergic system within the amygdala modulates aversive memory consolidation by regulating actin rearrangements but not Arc protein expression in the DH, which is distinct from its role in modulation of inhibitory avoidance memory.
Collapse
|
190
|
Abstract
Dysregulation of the brain emotional systems that mediate arousal and stress is a key component of the pathophysiology of drug addiction. Drug addiction is a chronically relapsing disorder characterized by a compulsion to seek and take drugs and the development of dependence and manifestation of a negative emotional state when the drug is removed. Activation of brain stress systems is hypothesized to be a key element of the negative emotional state produced by dependence that drives drug-seeking through negative reinforcement mechanisms. The focus of the present review is on the role of two key brain arousal/stress systems in the development of dependence. Emphasis is placed on the neuropharmacological actions of corticotropin-releasing factor (CRF) and norepinephrine in extrahypothalamic systems in the extended amygdala, including the central nucleus of the amygdala, bed nucleus of the stria terminalis, and a transition area in the shell of the nucleus accumbens. Compelling evidence argues that these brain stress systems, a heretofore largely neglected component of dependence and addiction, play a key role in engaging the transition to dependence and maintaining dependence once it is initiated. Understanding the role of the brain stress and anti-stress systems in addiction not only provides insight into the neurobiology of the "dark side" of addiction but also provides insight into the organization and function of basic brain emotional circuitry that guides motivated behavior.
Collapse
Affiliation(s)
- George F Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 North Torrey Pines Road, SP30-2400, La Jolla, CA 92037, USA.
| |
Collapse
|
191
|
Lateral hypothalamic orexin/hypocretin neurons: A role in reward-seeking and addiction. Brain Res 2009; 1314:74-90. [PMID: 19815001 DOI: 10.1016/j.brainres.2009.09.106] [Citation(s) in RCA: 301] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 09/27/2009] [Accepted: 09/29/2009] [Indexed: 12/30/2022]
Abstract
Orexins (synonymous with hypocretins) are recently discovered neuropeptides made exclusively in hypothalamus. Behavioral, anatomical, and neurophysiological studies show that a subset of these cells, specifically those in lateral hypothalamus (LH), are involved in reward processing and addictive behaviors. Fos expression in LH orexin neurons varied in proportion to conditioned place preference (CPP) for morphine, cocaine, or food. This relationship occurred both in drug-naïve rats and in animals during protracted morphine withdrawal, when drug preference was elevated but food preference was decreased. Inputs to the LH orexin cell field from lateral septum and bed nucleus of the stria terminalis were Fos-activated during cocaine CPP in proportion to the preference expressed in each animal. This implies that these inputs may be involved in driving the conditioned responses in LH orexin neurons. Related studies showed that LH orexin neurons that project to ventral tegmental area (VTA) had greater Fos induction in association with elevated morphine preference during protracted withdrawal than non-VTA-projecting orexin neurons, indicating that the VTA is an important site of action for orexin's role in reward processing. In addition, stimulation of LH orexin neurons, or microinjection of orexin into VTA, reinstated an extinguished morphine preference. In self-administration studies, the orexin 1 receptor antagonist SB-334867 (SB) blocked cocaine-seeking induced by discrete or contextual cues previously associated with cocaine, but not by a priming injection of cocaine. There was no effect of SB on cocaine self-administration itself, indicating that it did not interfere with the drug's reinforcing properties. Neurophysiological studies revealed that locally applied orexin often augmented responses of VTA dopamine (DA) neurons to activation of the medial prefrontal cortex (mPFC), consistent with the view that orexin facilitates activation of VTA DA neurons by stimulus-reward associations. This LH-to-VTA orexin pathway was found to be necessary for learning a morphine place preference. These findings are consistent with results showing that orexin facilitates glutamate-mediated responses, and is necessary for glutamate-dependent long-term potentiation in VTA DA neurons. We surmise from these studies that LH orexin neurons play an important role in reward processing and addiction and that LH orexin cells are an important input to VTA for behavioral effects associated with reward-paired stimuli.
Collapse
|
192
|
Shields AD, Wang Q, Winder DG. alpha2A-adrenergic receptors heterosynaptically regulate glutamatergic transmission in the bed nucleus of the stria terminalis. Neuroscience 2009; 163:339-51. [PMID: 19527774 PMCID: PMC2744292 DOI: 10.1016/j.neuroscience.2009.06.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 06/06/2009] [Accepted: 06/09/2009] [Indexed: 10/20/2022]
Abstract
Stress is a major driving force in reinstatement of drug-seeking behavior. The bed nucleus of the stria terminalis (BNST) has been identified as a key brain region in this behavior, and receives a dense input of the stress-neurotransmitter norepinephrine through the ventral noradrenergic bundle. Activation of alpha(2)-adrenergic receptors (alpha(2)-ARs) in the BNST blocks stress-induced reinstatement of drug-seeking, indicating a potentially important role for these receptors. Currently, it is unclear how alpha(2)-AR agonists elicit this behavioral action, or through which alpha(2)-AR subtype. Activation of alpha(2)-ARs decreases glutamatergic transmission in the BNST, an effect which is nearly absent in the alpha(2A)-AR knockout mouse. Here, we take advantage of a knock-in mouse in which a hemagglutinin-tagged alpha(2A)-AR was inserted into the endogenous locus, along with the alpha(2A)-AR selective agonist guanfacine, to further study the role of the alpha(2A)-AR subtype in modulation of neurotransmission in the BNST. Using immunohistochemistry, we find that alpha(2A)-ARs are highly expressed in the BNST, and that this expression is more similar in distribution to the vesicular glutamate transporters than to either norepinephrine transporter or tyrosine hydroxylase positive terminals. Using whole cell patch-clamp recordings, we show that guanfacine causes a depression of evoked excitatory and, to a more limited extent, inhibitory fast synaptic transmission. In total, these data support a prominent heterosynaptic role for alpha(2A)-ARs in modulating fast synaptic transmission in the BNST.
Collapse
MESH Headings
- Adrenergic alpha-Agonists/pharmacology
- Animals
- Excitatory Postsynaptic Potentials/drug effects
- Excitatory Postsynaptic Potentials/physiology
- Gene Knock-In Techniques
- Glutamic Acid/metabolism
- Guanfacine/pharmacology
- Inhibitory Postsynaptic Potentials/drug effects
- Inhibitory Postsynaptic Potentials/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neurons/cytology
- Neurons/drug effects
- Neurons/metabolism
- Norepinephrine/metabolism
- Organ Culture Techniques
- Patch-Clamp Techniques
- Receptors, Adrenergic, alpha-2/drug effects
- Receptors, Adrenergic, alpha-2/genetics
- Receptors, Adrenergic, alpha-2/metabolism
- Septal Nuclei/cytology
- Septal Nuclei/drug effects
- Septal Nuclei/metabolism
- Stress, Psychological/metabolism
- Stress, Psychological/physiopathology
- Substance-Related Disorders/metabolism
- Substance-Related Disorders/physiopathology
- Synaptic Transmission/drug effects
- Synaptic Transmission/physiology
- Time Factors
Collapse
Affiliation(s)
- Angela D. Shields
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville TN 37232-0615
| | - Qin Wang
- Department of Physiology and Biophysics, University of Alabama, Nashville TN 37232-0615
| | - Danny G. Winder
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville TN 37232-0615
- Center for Molecular Neuroscience, Vanderbilt University School of Medicine, Nashville TN 37232-0615
- J.F. Kennedy Center for Research on Human Development, Vanderbilt University School of Medicine, Nashville TN 37232-0615
| |
Collapse
|
193
|
Minami M. Neuronal mechanisms for pain-induced aversion behavioral studies using a conditioned place aversion test. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 85:135-44. [PMID: 19607966 DOI: 10.1016/s0074-7742(09)85010-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Pain consists of sensory discriminative and negative affective components. Although the neural systems responsible for the sensory component of pain have been studied extensively, the neural basis of the affective component is not well understood. Recently, behavioral studies using conditioned place aversion (CPA) tests have successfully elucidated the neural circuits and mechanisms underlying the negative affective component of pain. Excitotoxic lesions of the anterior cingulate cortex (ACC), central amygdaloid nucleus, basolateral amygdaloid nucleus (BLA), or bed nucleus of the stria terminalis (BNST) suppressed intraplantar formalin-induced aversive responses. Glutamatergic transmission within the ACC and BLA via NMDA receptors was shown to play a critical role in the affective component of pain. In the BNST, especially its ventral part, noradrenergic transmission via beta-adrenergic receptors was demonstrated as important for pain-induced aversion. Because persistent pain is frequently associated with psychological and emotional dysfunction, studies of the neural circuits and the molecular mechanisms involved in the affective component of pain may have considerable clinical importance in the treatment of chronic pain.
Collapse
Affiliation(s)
- Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| |
Collapse
|
194
|
Roth-Deri I, Friedman A, Abraham L, Lax E, Flaumenhaft Y, Dikshtein Y, Yadid G. Antidepressant treatment facilitates dopamine release and drug seeking behavior in a genetic animal model of depression. Eur J Neurosci 2009; 30:485-92. [PMID: 19614746 DOI: 10.1111/j.1460-9568.2009.06840.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Anhedonia and lack of motivation are core symptoms of depression. In contrast, hyper-motivation and euphoria characterize intoxicated states. In order to explore the relationship between these two behavioral states we examined cocaine self-administration tasks in an animal model of depression [Flinders Sensitive Line (FSL) rats]. We found that FSL rats exhibit sub-sensitivity in their cocaine-seeking behavior, which was normalized following a chronic treatment with the antidepressant desipramine. However, when the cocaine dosage was increased, FSL rats demonstrated a similar cocaine-seeking behavior to that of controls. In light of dopamine's central role in modulating cocaine reinforcement, we examined dopaminergic neurotransmission in the nucleus accumbens, a brain region implicated in the rewarding and hedonic effects of substances of misuse. FSL rats exhibited low but dose-dependent increases in extracellular levels of dopamine in the nucleus accumbens after acute intravenous cocaine injection. Furthermore, by using the dopamine transporter blocker GBR-12909 we were able to demonstrate that the low extracellular dopamine levels, observed in FSL rats, were a consequence of low dopamine release in the nucleus accumbens, as opposed to the possibility of increased uptake. Treatment of FSL rats with the antidepressant desipramine raised cocaine- and GBR-12909-induced dopamine release to the level of controls. This treatment also resulted in increased cocaine-seeking behavior.
Collapse
Affiliation(s)
- Ilana Roth-Deri
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | | | | | | | | | | |
Collapse
|
195
|
Núñez C, Földes A, Pérez-Flores D, García-Borrón JC, Laorden ML, Kovács KJ, Milanés MV. Elevated glucocorticoid levels are responsible for induction of tyrosine hydroxylase mRNA expression, phosphorylation, and enzyme activity in the nucleus of the solitary tract during morphine withdrawal. Endocrinology 2009; 150:3118-27. [PMID: 19179436 PMCID: PMC2703550 DOI: 10.1210/en.2008-1732] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Chronic opiate exposure induces neurochemical adaptations in the noradrenergic system. Enhanced responsiveness of the hypothalamo-pituitary-adrenal axis after morphine withdrawal has been associated with hyperactivity of ascending noradrenergic input from the nucleus of the solitary tract (NTS-A(2)) cell group to the hypothalamic paraventricular nucleus (PVN). This study addressed the role of morphine withdrawal-induced corticosterone (CORT) release in regulation of tyrosine hydroxylase (TH), the rate-limiting enzyme of catecholamine biosynthesis in adrenalectomized (ADX) rats supplemented with low CORT pellet (ADX plus CORT). Present results show that in sham-ADX rats, noradrenergic neurons in the NTS-A(2) became activated during morphine withdrawal, as indicated by increased TH mRNA expression. However, this induction of TH expression is not detected in ADX plus CORT rats that are unable to mount CORT secretory response to morphine withdrawal. Total TH protein levels were elevated in the NTS-A(2) from sham-operated rats during morphine dependence and withdrawal, whereas we did not find any alteration in ADX plus CORT animals. Furthermore, high levels of TH phosphorylated (activated) at Ser31 (but not at Ser40) were found in the A(2) area from sham-morphine withdrawn rats. Consistent with these effects, we observed an increase in the enzyme activity of TH in the PVN. However, induction of morphine withdrawal to ADX plus CORT animals did not alter the phosphorylation (activation) of TH in NTS-A(2) and decreased TH activity in the PVN. These results suggest the existence of a positive reverberating circle in which elevated glucocorticoids during morphine abstinence play a permissive role in morphine withdrawal-induced activation of noradrenergic pathway innervating the PVN.
Collapse
Affiliation(s)
- Cristina Núñez
- Department of Pharmacology, University School of Medicine, Campus de Espinardo, 30100 Murcia, Spain
| | | | | | | | | | | | | |
Collapse
|
196
|
Marcinkiewcz CA, Prado MM, Isaac SK, Marshall A, Rylkova D, Bruijnzeel AW. Corticotropin-releasing factor within the central nucleus of the amygdala and the nucleus accumbens shell mediates the negative affective state of nicotine withdrawal in rats. Neuropsychopharmacology 2009; 34:1743-52. [PMID: 19145226 PMCID: PMC2680924 DOI: 10.1038/npp.2008.231] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tobacco addiction is a chronic disorder that is characterized by a negative affective state upon smoking cessation and relapse after periods of abstinence. Previous research has shown that an increased central release of corticotropin-releasing factor (CRF) at least partly mediates the deficit in brain reward function associated with nicotine withdrawal in rats. The aim of these studies was to investigate the role of CRF in the central nucleus of the amygdala (CeA), the lateral bed nucleus of the stria terminalis (BNST), and the nucleus accumbens shell (Nacc shell) in the deficit in brain reward function associated with precipitated nicotine withdrawal. The intracranial self-stimulation procedure was used to assess the negative affective aspects of nicotine withdrawal. Elevations in brain reward thresholds are indicative of a deficit in brain reward function. In all experiments, the nicotinic receptor antagonist mecamylamine (3 mg/kg) elevated the brain reward thresholds of the nicotine-dependent rats (9 mg/kg per day of nicotine salt) and did not affect the brain reward thresholds of the saline-treated control rats. The administration of the nonspecific CRF1/2 receptor antagonist D-Phe CRF((12-41)) into the CeA and the Nacc shell prevented the mecamylamine-induced elevations in brain reward thresholds in the nicotine-dependent rats. Blockade of CRF1/2 receptors in the lateral BNST did not prevent the mecamylamine-induced elevations in brain reward thresholds in the nicotine-dependent rats. These studies indicate that the negative emotional state associated with precipitated nicotine withdrawal is at least partly mediated by an increased release of CRF in the CeA and the Nacc shell.
Collapse
Affiliation(s)
- Catherine A Marcinkiewcz
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610 , USA
| | | | | | | | | | | |
Collapse
|
197
|
Brown RM, Lawrence AJ. Neurochemistry underlying relapse to opiate seeking behaviour. Neurochem Res 2009; 34:1876-87. [PMID: 19418222 DOI: 10.1007/s11064-009-9967-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Accepted: 03/27/2009] [Indexed: 11/24/2022]
Abstract
Relapse is a major clinical problem and remains a major challenge in the treatment of addictions. A goal of current research is to gain a greater understanding of the neurochemistry underlying relapse to opiate use. Factors which trigger relapse in humans such as stress, exposure to opiates and/or drug-associated cues, can also trigger opiate-seeking in animals. This review will overview preclinical studies relating to the neurochemistry of opiate-seeking with a focus on studies published from 2005 to present.
Collapse
Affiliation(s)
- Robyn M Brown
- Howard Florey Institute, University of Melbourne, Parkville, VIC 3010, Australia
| | | |
Collapse
|
198
|
Moberg CA, Curtin JJ. Alcohol selectively reduces anxiety but not fear: startle response during unpredictable versus predictable threat. JOURNAL OF ABNORMAL PSYCHOLOGY 2009; 118:335-47. [PMID: 19413408 PMCID: PMC2756160 DOI: 10.1037/a0015636] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recent theory and empirical research have suggested that fear and anxiety are distinct processes with separable neurobiological substrates. Furthermore, a laboratory procedure has been developed to manipulate fear versus anxiety independently via administration of predictable or unpredictable electric shock, respectively. Benzodiazepines appear to selectively reduce anxiety but not fear in this procedure. The primary aim of this experiment was to determine if alcohol produced a similar selective reduction in anxiety. Intoxicated (target blood alcohol concentration of .08%) and nonintoxicated participants viewed a series of colored squares separated by variable intertrial intervals (ITIs) in 3 conditions. In the predictable shock condition, shocks were administered contingently during every square. In the unpredictable shock condition, shocks were administered noncontingently during both squares and ITIs. In the no-shock condition, no shocks were administered at any time. Alcohol significantly reduced startle potentiation during cues signaling unpredictable but not predictable shock, consistent with the thesis that alcohol selectively reduces anxiety but not fear. In addition, alcohol's effect on startle potentiation during unpredictable shock was mediated by vigilance. This anxiolytic effect may clarify the nature of alcohol's reinforcing effects in social and problem drinkers.
Collapse
Affiliation(s)
- Christine A Moberg
- Department of Psychology, University of Wisconsin-Madison,1202 West Johnson Street, Madison, WI 53706, USA.
| | | |
Collapse
|
199
|
Vargas-Perez H, Ting-A-Kee R, van der Kooy D. Different neural systems mediate morphine reward and its spontaneous withdrawal aversion. Eur J Neurosci 2009; 29:2029-34. [DOI: 10.1111/j.1460-9568.2009.06749.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
200
|
Francesconi W, Berton F, Repunte-Canonigo V, Hagihara K, Thurbon D, Lekic D, Specio SE, Greenwell TN, Chen SA, Rice KC, Richardson HN, O'Dell LE, Zorrilla EP, Morales M, Koob GF, Sanna PP. Protracted withdrawal from alcohol and drugs of abuse impairs long-term potentiation of intrinsic excitability in the juxtacapsular bed nucleus of the stria terminalis. J Neurosci 2009; 29:5389-401. [PMID: 19403807 PMCID: PMC2938175 DOI: 10.1523/jneurosci.5129-08.2009] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 01/26/2009] [Accepted: 01/27/2009] [Indexed: 11/21/2022] Open
Abstract
The juxtacapsular bed nucleus of the stria terminalis (jcBNST) is activated in response to basolateral amygdala (BLA) inputs through the stria terminalis and projects back to the anterior BLA and to the central nucleus of the amygdala. Here we show a form of long-term potentiation of the intrinsic excitability (LTP-IE) of jcBNST neurons in response to high-frequency stimulation of the stria terminalis. This LTP-IE, which was characterized by a decrease in the firing threshold and increased temporal fidelity of firing, was impaired during protracted withdrawal from self-administration of alcohol, cocaine, and heroin. Such impairment was graded and was more pronounced in rats that self-administered amounts of the drugs sufficient to maintain dependence. Dysregulation of the corticotropin-releasing factor (CRF) system has been implicated in manifestation of protracted withdrawal from dependent drug use. Administration of the selective corticotropin-releasing factor receptor 1 (CRF(1)) antagonist R121919 [2,5-dimethyl-3-(6-dimethyl-4-methylpyridin-3-yl)-7-dipropylamino-pyrazolo[1,5-a]pyrimidine)], but not of the CRF(2) antagonist astressin(2)-B, normalized jcBNST LTP-IE in animals with a history of alcohol dependence; repeated, but not acute, administration of CRF itself produced a decreased jcBNST LTP-IE. Thus, changes in the intrinsic properties of jcBNST neurons mediated by chronic activation of the CRF system may contribute to the persistent emotional dysregulation associated with protracted withdrawal.
Collapse
Affiliation(s)
- Walter Francesconi
- Molecular and Integrative Neurosciences Department and
- Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - Fulvia Berton
- Molecular and Integrative Neurosciences Department and
| | | | | | - David Thurbon
- Molecular and Integrative Neurosciences Department and
| | - Dusan Lekic
- Molecular and Integrative Neurosciences Department and
| | - Sheila E. Specio
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037
| | - Thomas N. Greenwell
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037
| | - Scott A. Chen
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037
| | - Kenner C. Rice
- National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20892
| | - Heather N. Richardson
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037
| | - Laura E. O'Dell
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037
| | - Eric P. Zorrilla
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037
| | - Marisela Morales
- Laboratory of Cellular Neurophysiology, National Institutes on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, and
| | - George F. Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037
| | | |
Collapse
|