1
|
Guida CR, Maia JM, Ferreira LFR, Rahdar A, Branco LGS, Soriano RN. Advancements in addressing drug dependence: A review of promising therapeutic strategies and interventions. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111070. [PMID: 38908501 DOI: 10.1016/j.pnpbp.2024.111070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Substance dependence represents a pervasive global concern within the realm of public health. Presently, it is delineated as a persistent and recurrent neurological disorder stemming from drug-triggered neuroadaptations in the brain's reward circuitry. Despite the availability of various therapeutic modalities, there has been a steady escalation in the mortality rate attributed to drug overdoses. Substantial endeavors have been directed towards the exploration of innovative interventions aimed at mitigating cravings and drug-induced repetitive behaviors. Within this review, we encapsulate the most auspicious contemporary treatment methodologies, accentuating meta-analyses of efficacious pharmacological and non-pharmacological approaches: including gabapentin, topiramate, prazosin, physical exercise regimens, and cerebral stimulation techniques.
Collapse
Affiliation(s)
- Clara Rodrigues Guida
- Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | - Juliana Marino Maia
- Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | | | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol 538-98615, Iran
| | - Luiz G S Branco
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-904, Brazil; Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil.
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares, MG 35020-360, Brazil.
| |
Collapse
|
2
|
Sicher AR, Starnes WD, Griffith KR, Dao NC, Smith GC, Brockway DF, Crowley NA. Adolescent binge drinking leads to long-lasting changes in cortical microcircuits in mice. Neuropharmacology 2023; 234:109561. [PMID: 37137354 PMCID: PMC10386078 DOI: 10.1016/j.neuropharm.2023.109561] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023]
Abstract
Adolescent drug consumption has increased risks to the individual compared to consumption in adulthood, due to the likelihood of long-term and permanent behavioral and neurological adaptations. However, little is known about how adolescent alcohol consumption influences the maturation and trajectory of cortical circuit development. Here, we explore the consequences of adolescent binge drinking on somatostatin (SST) neuronal function in superficial layers of the prelimbic (PL) cortex in male and female SST-Ai9 mice. We find that adolescent drinking-in-the-dark (DID) produces sex-dependent increases in intrinsic excitability of SST neurons, with no change in overall SST cell number, persisting well into adulthood. While we did not find evidence of altered GABA release from SST neurons onto other neurons within the circuit, we found a complementary reduction in layer II/III pyramidal neuron excitability immediately after binge drinking; however, this hypoexcitability rebounded towards increased pyramidal neuron activity in adulthood in females, suggesting long-term homeostatic adaptations in this circuit. Together, this suggests that binge drinking during key developmental timepoints leads to permanent changes in PL microcircuitry function, which may have broad behavioral implications.
Collapse
Affiliation(s)
- Avery R Sicher
- Neuroscience Graduate Program, The Huck Institutes of the Life Sciences, University Park, PA, 16802, USA; Department of Biology, Penn State University, University Park, PA, 16802, USA
| | - William D Starnes
- Department of Biology, Penn State University, University Park, PA, 16802, USA
| | - Keith R Griffith
- Department of Biology, Penn State University, University Park, PA, 16802, USA
| | - Nigel C Dao
- Department of Biology, Penn State University, University Park, PA, 16802, USA
| | - Grace C Smith
- Department of Biology, Penn State University, University Park, PA, 16802, USA; Department of Biomedical Engineering, Penn State University, University Park, PA, 16802, USA
| | - Dakota F Brockway
- Neuroscience Graduate Program, The Huck Institutes of the Life Sciences, University Park, PA, 16802, USA; Department of Biology, Penn State University, University Park, PA, 16802, USA
| | - Nicole A Crowley
- Neuroscience Graduate Program, The Huck Institutes of the Life Sciences, University Park, PA, 16802, USA; Department of Biology, Penn State University, University Park, PA, 16802, USA; Department of Biomedical Engineering, Penn State University, University Park, PA, 16802, USA; Center for Neural Engineering, Penn State University, University Park, PA, 16802, USA.
| |
Collapse
|
3
|
Konar-Nié M, Guzman-Castillo A, Armijo-Weingart L, Aguayo LG. Aging in nucleus accumbens and its impact on alcohol use disorders. Alcohol 2023; 107:73-90. [PMID: 36087859 DOI: 10.1016/j.alcohol.2022.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 02/06/2023]
Abstract
Ethanol is one of the most widely consumed drugs in the world and prolonged excessive ethanol intake might lead to alcohol use disorders (AUDs), which are characterized by neuroadaptations in different brain regions, such as in the reward circuitry. In addition, the global population is aging, and it appears that they are increasing their ethanol consumption. Although research involving the effects of alcohol in aging subjects is limited, differential effects have been described. For example, studies in human subjects show that older adults perform worse in tests assessing working memory, attention, and cognition as compared to younger adults. Interestingly, in the field of the neurobiological basis of ethanol actions, there is a significant dichotomy between what we know about the effects of ethanol on neurochemical targets in young animals and how it might affect them in the aging brain. To be able to understand the distinct effects of ethanol in the aging brain, the following questions need to be answered: (1) How does physiological aging impact the function of an ethanol-relevant region (e.g., the nucleus accumbens)? and (2) How does ethanol affect these neurobiological systems in the aged brain? This review discusses the available data to try to understand how aging affects the nucleus accumbens (nAc) and its neurochemical response to alcohol. The data show that there is little information on the effects of ethanol in aged mice and rats, and that many studies had considered 2-3-month-old mice as adults, which needs to be reconsidered since more recent literature defines 6 months as young adults and >18 months as an older mouse. Considering the actual relevance of an aged worldwide population and that this segment is drinking more frequently, it appears at least reasonable to explore how ethanol affects the brain in adult and aged models.
Collapse
Affiliation(s)
- Macarena Konar-Nié
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepcion, Concepcion, Chile.
| | - Alejandra Guzman-Castillo
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepcion, Concepcion, Chile; Programa en Neurociencia, Psiquiatría y Salud Mental, Universidad de Concepción, Concepcion, Chile.
| | - Lorena Armijo-Weingart
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepcion, Concepcion, Chile; Programa en Neurociencia, Psiquiatría y Salud Mental, Universidad de Concepción, Concepcion, Chile.
| | - Luis Gerardo Aguayo
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepcion, Concepcion, Chile; Programa en Neurociencia, Psiquiatría y Salud Mental, Universidad de Concepción, Concepcion, Chile.
| |
Collapse
|
4
|
Jin D, Chen H, Chen SR, Pan HL. α2δ-1 protein drives opioid-induced conditioned reward and synaptic NMDA receptor hyperactivity in the nucleus accumbens. J Neurochem 2023; 164:143-157. [PMID: 36222452 PMCID: PMC9892208 DOI: 10.1111/jnc.15706] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/19/2022] [Accepted: 10/08/2022] [Indexed: 02/04/2023]
Abstract
Glutamate NMDA receptors (NMDARs) in the nucleus accumbens (NAc) are critically involved in drug dependence and reward. α2δ-1 is a newly discovered NMDAR-interacting protein that promotes synaptic trafficking of NMDARs independently of its conventional role as a calcium channel subunit. However, it remains unclear how repeated opioid exposure affects synaptic NMDAR activity and α2δ-1-NMDAR interaction in the NAc. In this study, whole-cell patch-clamp recordings showed that repeated treatment with morphine in mice markedly increased the NMDAR-mediated frequency of miniature excitatory postsynaptic currents (mEPSCs) and amplitude of puff NMDAR currents in medium spiny neurons in the NAc core region. Morphine treatment significantly increased the physical interaction of α2δ-1 with GluN1 and their synaptic trafficking in the NAc. In Cacna2d1 knockout mice, repeated treatment with morphine failed to increase the frequency of mEPSCs and amplitude of puff NMDAR currents in the NAc core. Furthermore, inhibition of α2δ-1 with gabapentin or disruption of the α2δ-1-NMDAR interaction with the α2δ-1 C terminus-interfering peptide blocked the morphine-elevated frequency of mEPSCs and amplitude of puff NMDAR currents in the NAc core. Correspondingly, systemically administered gabapentin, Cacna2d1 ablation, or microinjection of the α2δ-1 C terminus-interfering peptide into the NAc core attenuated morphine-induced conditioned place preference and locomotor sensitization. Our study reveals that repeated opioid exposure strengthens presynaptic and postsynaptic NMDAR activity in the NAc via α2δ-1. The α2δ-1-bound NMDARs in the NAc have a key function in the rewarding effect of opioids and could be targeted for treating opioid use disorder and addiction.
Collapse
Affiliation(s)
- Daozhong Jin
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hong Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
5
|
Lee SM, Yeh PWL, Yeh HH. L-Type Calcium Channels Contribute to Ethanol-Induced Aberrant Tangential Migration of Primordial Cortical GABAergic Interneurons in the Embryonic Medial Prefrontal Cortex. eNeuro 2022; 9:ENEURO.0359-21.2021. [PMID: 34930830 PMCID: PMC8805770 DOI: 10.1523/eneuro.0359-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/17/2021] [Accepted: 12/07/2021] [Indexed: 11/21/2022] Open
Abstract
Exposure of the fetus to alcohol (ethanol) via maternal consumption during pregnancy can result in fetal alcohol spectrum disorders (FASD), hallmarked by long-term physical, behavioral, and intellectual abnormalities. In our preclinical mouse model of FASD, prenatal ethanol exposure disrupts tangential migration of corticopetal GABAergic interneurons (GINs) in the embryonic medial prefrontal cortex (mPFC). We postulated that ethanol perturbed the normal pattern of tangential migration via enhancing GABAA receptor-mediated membrane depolarization that prevails during embryonic development in GABAergic cortical interneurons. However, beyond this, our understanding of the underlying mechanisms is incomplete. Here, we tested the hypothesis that the ethanol-enhanced depolarization triggers downstream an increase in high-voltage-activated nifedipine-sensitive L-type calcium channel (LTCC) activity and provide evidence implicating calcium dynamics in the signaling scheme underlying the migration of embryonic GINs and its aberrance. Tangentially migrating Nkx2.1+ GINs expressed immunoreactivity to Cav1.2, the canonical neuronal isoform of the L-type calcium channel. Prenatal ethanol exposure did not alter its protein expression profile in the embryonic mPFC. However, exposing ethanol concomitantly with the LTCC blocker nifedipine prevented the ethanol-induced aberrant migration both in vitro and in vivo In addition, whole-cell patch clamp recording of LTCCs in GINs migrating in embryonic mPFC slices revealed that acutely applied ethanol potentiated LTCC activity in migrating GINs. Based on evidence reported in the present study, we conclude that calcium is an important intracellular intermediary downstream of GABAA receptor-mediated depolarization in the mechanistic scheme of an ethanol-induced aberrant tangential migration of embryonic GABAergic cortical interneurons.
Collapse
Affiliation(s)
- Stephanie M Lee
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Pamela W L Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Hermes H Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| |
Collapse
|
6
|
Chatterjee D, Mahabir S, Chatterjee D, Gerlai R. Lasting effects of mild embryonic ethanol exposure on voltage-gated ion channels in adult zebrafish brain. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110327. [PMID: 33864849 DOI: 10.1016/j.pnpbp.2021.110327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/28/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023]
Abstract
The zebrafish is increasingly well utilized in alcohol research, particularly in modeling human fetal alcohol spectrum disorders (FASD). FASD results from alcohol reaching the developing fetus intra utero, a completely preventable yet prevalent and devastating life-long disorder. The hope with animal models, including the zebrafish, is to discover the mechanisms underlying this disease, which may aid treatment and diagnosis. In the past, we developed an embryonic alcohol exposure regimen that is aimed at mimicking the milder, and most prevalent, forms of FASD in zebrafish. We have found numerous lasting alterations in behavior, neurochemistry, neuronal markers and glial cell phenotypes in this zebrafish FASD model. Using the same model (2 h long bath immersion of 24 h post-fertilization old zebrafish eggs into 1% vol/vol ethanol), here we conduct a proof of concept analysis of voltage-gated cation channels, investigating potential embryonic alcohol induced changes in L-, T- and N- type Ca++ and the SCN1A Na+ channels using Western blot followed by immunohistochemical analysis of the same channels in the pallium and cerebellum of the zebrafish brain. We report significant reduction of expression in all four channel proteins using both methods. We conclude that reduced voltage-gated cation channel expression induced by short and low dose exposure to alcohol during embryonic development of zebrafish may contribute to the previously demonstrated lasting behavioral and neurobiological changes.
Collapse
Affiliation(s)
| | - Samantha Mahabir
- Department of Psychology, University of Toronto, Mississauga, Ontario, Canada
| | | | - Robert Gerlai
- Department of Psychology, University of Toronto, Mississauga, Ontario, Canada.
| |
Collapse
|
7
|
Bosse KE, Ghoddoussi F, Eapen AT, Charlton JL, Susick LL, Desai K, Berkowitz BA, Perrine SA, Conti AC. Calcium/calmodulin-stimulated adenylyl cyclases 1 and 8 regulate reward-related brain activity and ethanol consumption. Brain Imaging Behav 2019; 13:396-407. [PMID: 29594872 PMCID: PMC6202255 DOI: 10.1007/s11682-018-9856-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Evidence suggests a predictive link between elevated basal activity within reward-related networks (e.g., cortico-basal ganglia-thalamic networks) and vulnerability for alcoholism. Both calcium channel function and cyclic adenosine monophosphate (cAMP)/protein kinase A-mediated signaling are critical modulators of reward neurocircuitry and reward-related behaviors. Calcium/calmodulin-stimulated adenylyl cyclases (AC) 1 and 8 are sensitive to activity-dependent increases in intracellular calcium and catalyze cAMP production. Therefore, we hypothesized AC1 and 8 regulate brain activity in reward regions of the cortico-basal ganglia-thalamic circuit and that this regulatory influence predicts voluntary ethanol drinking responses. This hypothesis was evaluated by manganese-enhanced magnetic resonance imaging and chronic, intermittent ethanol access procedures. Ethanol-naïve mice with genetic deletion of both AC1 and 8 (DKO mice) exhibited bilateral reductions in baseline activity within cortico-basal ganglia-thalamic regions associated with reward processing compared to wild-type controls (WT, C57BL/6 mice). Significant activity changes were not evident in regions either outside of the cortico-basal ganglia-thalamic network or within the network that are not associated with reward processing. Parallel studies demonstrated that reward network hypoactivity in DKO mice predicted a significant attenuation in consumption and preference levels to escalating ethanol concentrations (12, 20 and 30%) compared to WT mice, an effect that was maintained over extended access (14 sessions) to 20% ethanol. Summarizing, these data support a contribution of AC1 and 8 in cortico-basal ganglia-thalamic activity and the predictive value of this regulatory influence on ethanol drinking behavior, which merits the future evaluation of calcium-stimulated ACs in the neural processes that engender vulnerability to maladaptive alcohol drinking.
Collapse
Affiliation(s)
- Kelly E Bosse
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Farhad Ghoddoussi
- Department of Anesthesiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ajay T Eapen
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jennifer L Charlton
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Laura L Susick
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kirt Desai
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bruce A Berkowitz
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Ophthalmology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Alana C Conti
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA.
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.
- Department of Neurosurgery, Wayne State University, 4646 John R St., Detroit, MI, 48201, USA.
| |
Collapse
|
8
|
Ehlers CL, Sanchez-Alavez M, Wills D. Effect of gabapentin on sleep and delta and theta EEG power in adult rats exposed to chronic intermittent ethanol vapor and protracted withdrawal during adolescence. Psychopharmacology (Berl) 2018; 235:1783-1791. [PMID: 29589069 PMCID: PMC5949268 DOI: 10.1007/s00213-018-4888-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 03/16/2018] [Indexed: 01/22/2023]
Abstract
RATIONALE Adolescents and young adults with alcohol problems may also have sleep difficulties. However, whether these sleep problems are a result of a history of drinking or arise due to other comorbid disorders is difficult to disentangle in human studies. Additionally, the mechanisms underlying adolescent alcohol-induced sleep disturbances and potential targets for therapy also remain under-investigated. Recent clinical trials have demonstrated that the anticonvulsant and analgesic drug gabapentin may have therapeutic value in normalizing sleep quality in adult recovering alcoholics, yet its potential for the treatment of adolescent sleep disturbances has not been investigated. OBJECTIVES This study sought to evaluate the effects of a history of 5 weeks of chronic intermittent ethanol vapor exposure, administered during adolescence (AIE), on EEG sleep, in young adult rats (n = 29). The ability of two doses of gabapentin (30, 120 mg/kg) to modify sleep and slow wave activity were also investigated in these young adult rats exposed to alcohol vapor during adolescence. RESULTS Adolescent vapor exposure in the rat was found to result in deficits in delta (1-4 Hz) and theta (4-8 Hz) power during slow wave sleep. Administration of gabapentin caused a "normalization" of the delta power deficits but did not affect theta power. CONCLUSIONS This report suggests that the potential mechanisms and therapeutic targets for sleep disturbance associated with adolescent alcohol exposure can be studied in preclinical models and that gabapentin may show partial efficacy in ameliorating these sleep deficits.
Collapse
Affiliation(s)
- Cindy L Ehlers
- Department of Neurosciences, The Scripps Research Institute, 10550 North Torrey Pines Road, SP30-1501, La Jolla, CA, 92037, USA.
| | - Manuel Sanchez-Alavez
- Department of Neurosciences, The Scripps Research Institute, 10550 North Torrey Pines Road, SP30-1501, La Jolla, CA, 92037, USA
| | - Derek Wills
- Department of Neurosciences, The Scripps Research Institute, 10550 North Torrey Pines Road, SP30-1501, La Jolla, CA, 92037, USA
| |
Collapse
|
9
|
Newton J, Suman S, Akinfiresoye LR, Datta K, Lovinger DM, N'Gouemo P. Alcohol withdrawal upregulates mRNA encoding for Ca V2.1-α1 subunit in the rat inferior colliculus. Alcohol 2018; 66:21-26. [PMID: 29277284 DOI: 10.1016/j.alcohol.2017.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 10/18/2022]
Abstract
We previously reported increased current density through P-type voltage-gated Ca2+ channels in inferior colliculus (IC) neurons during alcohol withdrawal. However, the molecular correlate of this increased P-type channel current is currently unknown. Here, we probe changes in mRNA and protein expression of the pore-forming CaV2.1-α1 (P/Q-type) subunits in IC neurons during the course of alcohol withdrawal-induced seizures (AWSs). Rats received three daily doses of ethanol or the vehicle every 8 h for 4 consecutive days. The IC was dissected at various time intervals following alcohol withdrawal, and the mRNA and protein levels of the CaV2.1-α1 subunits were measured. In separate experiments, rats were tested for acoustically evoked seizure susceptibility 3, 24, and 48 h after alcohol withdrawal. AWSs were observed 24 h after withdrawal; no seizures were observed at 3 or 48 h or in the control-treated rats. Compared to control-treated rats, the mRNA levels of the CaV2.1-α1 subunit were increased 1.9-fold and 2.1-fold at 3 and 24 h, respectively; change in mRNA expression was nonsignificant at 48 h following alcohol withdrawal. Western blot analyses revealed that protein levels of the CaV2.1-α1 subunits were not altered in IC neurons following alcohol withdrawal. We conclude that expression of the Cacna1a mRNA increased before the onset of AWS susceptibility, suggesting that altered CaV2.1 channel expression may play a role in AWS pathogenesis.
Collapse
|
10
|
Sanchez-Alavez M, Wills DN, Amodeo L, Ehlers CL. Effect of Gabapentin on Sleep and Event-Related Oscillations (EROs) in Rats Exposed to Chronic Intermittent Ethanol Vapor and Protracted Withdrawal. Alcohol Clin Exp Res 2018; 42:624-633. [PMID: 29286538 DOI: 10.1111/acer.13588] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/20/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Disturbances in sleep architecture, especially reductions in slow-wave sleep (SWS), are symptoms commonly observed in individuals with alcohol use disorders. Recent clinical trials have demonstrated that the anticonvulsant and analgesic drug gabapentin may have therapeutic value in normalizing sleep quality in recovering alcoholics. However, the brain mechanisms underlying this improvement in sleep following gabapentin treatment remain unknown. METHODS In this study, adult Wistar rats were exposed to 8 weeks of chronic intermittent ethanol [EtOH] vapor (blood EtOH concentrations averaged 128.2 ± 17.4 mg/dl) or control conditions and then withdrawn. Sleep electroencephalograms [EEGs] and event-related oscillations (EROs) were evaluated at baseline prior to EtOH exposure and 24 hours following EtOH withdrawal. Four weeks following EtOH withdrawal the effects of saline and 2 doses of gabapentin (30, 120 mg/kg), on EROs and sleep EEGs, were evaluated. RESULTS As compared to baseline, 24 hours following alcohol withdrawal SWS became fragmented as indexed by a significant increase in the number and a decrease in the duration of SWS episodes. Compared to controls, the EtOH-exposed group had more ERO energy in the beta frequency band in the parietal cortex. Gabapentin induced a dose-dependent decrease in the latency to the first SWS episode, and a reduction in sleep fragmentation. Gabapentin also produced a dose-dependent increase in ERO energy in the control group that was significantly attenuated in the EtOH-exposed group in the theta, and beta frequency bands. CONCLUSIONS Taken together, these studies suggest that gabapentin can reverse some of the alcohol-induced sleep and EEG deficits but does not eliminate all of the enduring brain effects of EtOH exposure.
Collapse
Affiliation(s)
| | - Derek N Wills
- Department of Neurosciences, The Scripps Research Institute, La Jolla, California
| | - Leslie Amodeo
- Department of Neurosciences, The Scripps Research Institute, La Jolla, California
| | - Cindy L Ehlers
- Department of Neurosciences, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
11
|
Mason BJ, Quello S, Shadan F. Gabapentin for the treatment of alcohol use disorder. Expert Opin Investig Drugs 2017; 27:113-124. [PMID: 29241365 DOI: 10.1080/13543784.2018.1417383] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Alcohol misuse is the fifth leading risk factor for premature death and disability worldwide. Fewer than 10% of afflicted Americans receive pharmacological treatment for alcohol use disorder. Gabapentin is a calcium channel GABAergic modulator that is widely used for pain. Studies showing reduced drinking and decreased craving and alcohol-related disturbances in sleep and affect in the months following alcohol cessation suggest therapeutic potential for alcohol use disorder. Areas covered: Human laboratory and clinical studies assessing gabapentin for alcohol use disorder are reviewed. Data were obtained by searching for English peer-reviewed articles on PubMed, reference lists of identified articles, and trials registered on clinicaltrials.gov. Additionally, the mechanism of action of gabapentin specific to alcohol use disorder, and studies of gabapentin for alcohol withdrawal and non-alcohol substance use disorders are summarized. Expert opinion: Alcohol use disorder represents a challenge and large, unmet medical need. Evidence from single-site studies lend support to the safety and efficacy of gabapentin as a novel treatment for alcohol use disorder, with unique benefits for alcohol-related insomnia and negative affect, relative to available treatments. Proprietary gabapentin delivery systems may open a path to pivotal trials and registration of gabapentin as a novel treatment for alcohol use disorder.
Collapse
Affiliation(s)
- Barbara J Mason
- a Pearson Center for Alcoholism and Addiction Research , The Scripps Research Institute , La Jolla , CA , USA
| | - Susan Quello
- a Pearson Center for Alcoholism and Addiction Research , The Scripps Research Institute , La Jolla , CA , USA
| | - Farhad Shadan
- b Division of Hospital Medicine , Scripps Clinic and Scripps Green Hospital , La Jolla , CA , USA
| |
Collapse
|
12
|
D'Souza MS. Glutamatergic transmission in drug reward: implications for drug addiction. Front Neurosci 2015; 9:404. [PMID: 26594139 PMCID: PMC4633516 DOI: 10.3389/fnins.2015.00404] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/12/2015] [Indexed: 12/12/2022] Open
Abstract
Individuals addicted to drugs of abuse such as alcohol, nicotine, cocaine, and heroin are a significant burden on healthcare systems all over the world. The positive reinforcing (rewarding) effects of the above mentioned drugs play a major role in the initiation and maintenance of the drug-taking habit. Thus, understanding the neurochemical mechanisms underlying the reinforcing effects of drugs of abuse is critical to reducing the burden of drug addiction in society. Over the last two decades, there has been an increasing focus on the role of the excitatory neurotransmitter glutamate in drug addiction. In this review, pharmacological and genetic evidence supporting the role of glutamate in mediating the rewarding effects of the above described drugs of abuse will be discussed. Further, the review will discuss the role of glutamate transmission in two complex heterogeneous brain regions, namely the nucleus accumbens (NAcc) and the ventral tegmental area (VTA), which mediate the rewarding effects of drugs of abuse. In addition, several medications approved by the Food and Drug Administration that act by blocking glutamate transmission will be discussed in the context of drug reward. Finally, this review will discuss future studies needed to address currently unanswered gaps in knowledge, which will further elucidate the role of glutamate in the rewarding effects of drugs of abuse.
Collapse
Affiliation(s)
- Manoranjan S D'Souza
- Pharmaceutical and Biomedical Sciences, Raabe College of Pharmacy, Ohio Northern University Ada, OH, USA
| |
Collapse
|
13
|
Abstract
Relapse to cocaine seeking is associated with potentiated excitatory synapses in nucleus accumbens. α2δ-1 is an auxiliary subunit of voltage-gated calcium channels that affects calcium-channel trafficking and kinetics, initiates extracellular signaling cascades, and promotes excitatory synaptogenesis. Previous data demonstrate that repeated exposure to alcohol, nicotine, methamphetamine, and morphine upregulates α2δ-1 in reward-related brain regions, but it was unclear whether this alteration generalized to cocaine. Here, we show that α2δ-1 protein was increased in nucleus accumbens after cocaine self-administration and extinction compared with saline controls. Furthermore, the endogenous ligand thrombospondin-1, responsible for the synaptogenic properties of the α2δ-1 receptor, was likewise elevated. Using whole-cell patch-clamp recordings of EPSCs in nucleus accumbens, we demonstrated that gabapentin, a specific α2δ-1 antagonist, preferentially reduced the amplitude and increased the paired-pulse ratio of EPSCs evoked by electrical stimulation in slices from cocaine-experienced rats compared with controls. In vivo, gabapentin microinjected in the nucleus accumbens core attenuated cocaine-primed but not cue-induced reinstatement. Importantly, gabapentin's effects on drug seeking were not due to a general depression of spontaneous or cocaine-induced locomotor activity. Moreover, gabapentin had no effect on reinstatement of sucrose seeking. These data indicate that α2δ-1 contributes specifically to cocaine-reinstated drug seeking, and identifies this protein as a target for the development of cocaine relapse medications. These results also inform ongoing discussion in the literature regarding efficacy of gabapentin as a candidate addiction therapy.
Collapse
|
14
|
Oka M, Hirouchi M, Tamura M, Sugahara S, Oyama T. Acamprosate {monocalcium bis(3-acetamidopropane-1-sulfonate)} reduces ethanol-drinking behavior in rats and glutamate-induced toxicity in ethanol-exposed primary rat cortical neuronal cultures. Eur J Pharmacol 2013; 718:323-31. [PMID: 24012782 DOI: 10.1016/j.ejphar.2013.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 07/19/2013] [Accepted: 08/24/2013] [Indexed: 01/09/2023]
Abstract
Acamprosate, the calcium salt of bis(3-acetamidopropane-1-sulfonate), contributes to the maintenance of abstinence in alcohol-dependent patients, but its mechanism of action in the central nervous system is unclear. Here, we report the effect of acamprosate on ethanol-drinking behavior in standard laboratory Wistar rats, including voluntary ethanol consumption and the ethanol-deprivation effect. After forced ethanol consumption arranged by the provision of only one drinking bottle containing 10% ethanol, the rats were given a choice between two drinking bottles, one containing water and the other containing 10% ethanol. In rats selected for high ethanol preference, repeated oral administration of acamprosate diminished voluntary ethanol drinking. After three months of continuous access to two bottles, rats were deprived of ethanol for three days and then presented with two bottles again. After ethanol deprivation, ethanol preference was increased, and the increase was largely abolished by acamprosate. After exposure of primary neuronal cultures of rat cerebral cortex to ethanol for four days, neurotoxicity, as measured by the extracellular leakage of lactate dehydrogenase (LDH), was induced by incubation with glutamate for 1h followed by incubation in the absence of ethanol for 24h. The N-methyl-D-aspartate receptor blocker 5-methyl-10,11-dihydro-5H-dibenzo[a,d]-cyclohepten-5,10-imine, the metabotropic glutamate receptor subtype 5 antagonist 6-methyl-2-(phenylethynyl)pyridine and the voltage-gated calcium-channel blocker nifedipine all inhibited glutamate-induced LDH leakage from ethanol-exposed neurons. Acamprosate inhibited the glutamate-induced LDH leakage from ethanol-exposed neurons more strongly than that from intact neurons. In conclusion, acamprosate showed effective reduction of drinking behavior in rats and protected ethanol-exposed neurons by multiple blocking of glutamate signaling.
Collapse
Affiliation(s)
- Michiko Oka
- Research Laboratories, Nippon Shinyaku Co., Ltd., 14, Nishinosho-monguchi-cho, Kisshoin, Minami-ku, Kyoto 601-8550, Japan.
| | | | | | | | | |
Collapse
|
15
|
Kouzoukas DE, Li G, Takapoo M, Moninger T, Bhalla RC, Pantazis NJ. Intracellular calcium plays a critical role in the alcohol-mediated death of cerebellar granule neurons. J Neurochem 2012; 124:323-35. [PMID: 23121601 DOI: 10.1111/jnc.12076] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 09/30/2012] [Accepted: 10/10/2012] [Indexed: 11/28/2022]
Abstract
Alcohol is a potent neuroteratogen that can trigger neuronal death in the developing brain. However, the mechanism underlying this alcohol-induced neuronal death is not fully understood. Utilizing primary cultures of cerebellar granule neurons (CGN), we tested the hypothesis that the alcohol-induced increase in intracellular calcium [Ca(2+)](i) causes the death of CGN. Alcohol induced a dose-dependent (200-800 mg/dL) neuronal death within 24 h. Ratiometric Ca(2+) imaging with Fura-2 revealed that alcohol causes a rapid (1-2 min), dose-dependent increase in [Ca(2+)](i), which persisted for the duration of the experiment (5 or 7 min). The alcohol-induced increase in [Ca(2+)](i) was observed in Ca(2+) -free media, suggesting intracellular Ca(2+) release. Pre-treatment of CGN cultures with an inhibitor (2-APB) of the inositol-triphosphate receptor (IP(3) R), which regulates Ca(2+) release from the endoplasmic reticulum (ER), blocked both the alcohol-induced rise in [Ca(2+)](i) and the neuronal death caused by alcohol. Similarly, pre-treatment with BAPTA/AM, a Ca(2+) -chelator, also inhibited the alcohol-induced surge in [Ca(2+) ](i) and prevented neuronal death. In conclusion, alcohol disrupts [Ca(2+)](i) homeostasis in CGN by releasing Ca(2+) from intracellular stores, resulting in a sustained increase in [Ca(2+)](i). This sustained increase in [Ca(2+)](i) may be a key determinant in the mechanism underlying alcohol-induced neuronal death.
Collapse
Affiliation(s)
- Dimitrios E Kouzoukas
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | | | | | | | | | | |
Collapse
|
16
|
Earl DE, Tietz EI. Inhibition of recombinant L-type voltage-gated calcium channels by positive allosteric modulators of GABAA receptors. J Pharmacol Exp Ther 2011; 337:301-11. [PMID: 21262851 DOI: 10.1124/jpet.110.178244] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Benzodiazepines (BDZs) depress neuronal excitability via positive allosteric modulation of inhibitory GABA(A) receptors (GABA(A)R). BDZs and other positive GABA(A)R modulators, including barbiturates, ethanol, and neurosteroids, can also inhibit L-type voltage-gated calcium channels (L-VGCCs), which could contribute to reduced neuronal excitability. Because neuronal L-VGCC function is up-regulated after long-term GABA(A)R modulator exposure, an interaction with L-VGCCs may also play a role in physical dependence. The current studies assessed the effects of BDZs (diazepam, flurazepam, and desalkylflurazepam), allopregnanolone, pentobarbital, and ethanol on whole-cell Ba(2+) currents through recombinant neuronal Ca(v)1.2 and Ca(v)1.3 L-VGCCs expressed with β(3) and α(2)δ-1 in HEK293T cells. Allopregnanolone was the most potent inhibitor (IC(50), ∼10 μM), followed by BDZs (IC(50), ∼50 μM), pentobarbital (IC(50), 0.3-1 mM), and ethanol (IC(50), ∼300 mM). Ca(v)1.3 channels were less sensitive to pentobarbital inhibition than Ca(v)1.2 channels, similar to dihydropyridine (DHP) L-VGCC antagonists. All GABA(A)R modulators induced a negative shift in the steady-state inactivation curve of Ca(v)1.3 channels, but only BDZs and pentobarbital induced a negative shift in Ca(v)1.2 channel inactivation. Mutation of the high-affinity DHP binding site (T1039Y and Q1043M) in Ca(v)1.2 channels reduced pentobarbital potency. Despite the structural similarity between benzothiazepines and BDZs, mutation of an amino acid important for diltiazem potency (I1150A) did not affect diazepam potency. Although L-VGCC inhibition by BDZs occurred at concentrations that are possibly too high to be clinically relevant and is not likely to play a role in the up-regulation of L-VGCCs during long-term treatment, pentobarbital and ethanol inhibited L-VGCCs at clinically relevant concentrations.
Collapse
Affiliation(s)
- Damien E Earl
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Health Science Campus, Toledo, OH 43614, USA
| | | |
Collapse
|
17
|
Norrell S, Reyes-Vasquez C, Burau K, Dafny N. Alcohol usage and abrupt cessation modulate diurnal activity. Brain Res Bull 2010; 83:57-64. [PMID: 20615456 DOI: 10.1016/j.brainresbull.2010.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Revised: 06/18/2010] [Accepted: 06/29/2010] [Indexed: 11/15/2022]
Abstract
Alcohol has many effects throughout the body. The effect on circadian rhythms and the correlation of these effects to withdrawal effects of alcohol present interesting findings. By measuring 3 planes of activity of female Sprague-Dawley rats during alcohol usage and continuing study through the first 2 days following withdrawal of alcohol allow for the observation of a drastic modulation of the circadian pattern of activity.
Collapse
Affiliation(s)
- Stacy Norrell
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, United States
| | | | | | | |
Collapse
|
18
|
Baliño P, Pastor R, Aragon CMG. Participation of L-type calcium channels in ethanol-induced behavioral stimulation and motor incoordination: effects of diltiazem and verapamil. Behav Brain Res 2010; 209:196-204. [PMID: 20122967 DOI: 10.1016/j.bbr.2010.01.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 01/19/2010] [Accepted: 01/24/2010] [Indexed: 10/19/2022]
Abstract
Calcium flux through voltage gate calcium channels (VGCC) is involved in many neuronal processes such as membrane depolarization, gene expression, hormone secretion, and neurotransmitter release. Several studies have shown that either acute or chronic exposure to ethanol modifies calcium influx through high voltage activated channels. Of special relevance is the L-type VGCC. Pharmacological manipulation of L-type calcium channels affects ethanol intake, ethanol discrimination and manifestations of withdrawal syndrome. The present study investigates the role of L-type channels on the psychomotor effects (stimulation and sedation/ataxia) of ethanol by testing the effects of different L-type calcium channel blockers (CCB) on such behaviors. Mice were pretreated intraperitoneally with the CCB, diltiazem (0-40 mg/kg) or verapamil (0-30 mg/kg) 30 min before ethanol (0-3.5 g/kg). Locomotion was measured in an open field chamber for 20 min immediately after ethanol. The two CCB tested prevented locomotor stimulation, but not locomotor suppression produced by ethanol. Doses of the two CCB which reduced ethanol stimulation, did not alter spontaneous locomotion. The ataxic effects of ethanol (1.25 g/kg), measured with an accelerating rotarod task, were not affected by diltiazem (20mg/kg) or verapamil (15 mg/kg). In addition, our results indicated that ethanol is more sensitive to the antagonism of L-type calcium channels than other drugs with stimulant properties; doses of the two CCB that reduced ethanol stimulation did not reduce the psychomotor effects of amphetamine, caffeine or cocaine. In conclusion, these data provide further evidence of the important involvement of L-type calcium channels in the behavioral effects produced by ethanol.
Collapse
Affiliation(s)
- Pablo Baliño
- Area de Psicobiología, Universtitat Jaume I, Avda. Sos Baynat s/n, 12071 Castellón, Spain
| | | | | |
Collapse
|
19
|
Moonat S, Starkman BG, Sakharkar A, Pandey SC. Neuroscience of alcoholism: molecular and cellular mechanisms. Cell Mol Life Sci 2010; 67:73-88. [PMID: 19756388 PMCID: PMC3747955 DOI: 10.1007/s00018-009-0135-y] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 07/28/2009] [Accepted: 08/13/2009] [Indexed: 01/21/2023]
Abstract
Alcohol use and abuse appear to be related to neuroadaptive changes at functional, neurochemical, and structural levels. Acute and chronic ethanol exposure have been shown to modulate function of the activity-dependent gene transcription factor, cAMP-responsive element binding (CREB) protein in the brain, which may be associated with the development of alcoholism. Study of the downstream effectors of CREB have identified several important CREB-related genes, such as neuropeptide Y, brain-derived neurotrophic factor, activity-regulated cytoskeleton-associated protein, and corticotrophin-releasing factor, that may play a crucial role in the behavioral effects of ethanol and molecular changes in the specific neurocircuitry that underlie both alcohol addiction and a genetic predisposition to alcoholism. Brain chromatin remodeling due to histone covalent modifications may also be involved in mediating the behavioral effects and neuroadaptive changes that occur during ethanol exposure. This review outlines progressive neuroscience research into molecular and epigenetic mechanisms of alcoholism.
Collapse
Affiliation(s)
- Sachin Moonat
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL USA
- Jesse Brown VA Medical Center, Chicago, IL USA
| | - Bela G. Starkman
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL USA
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL USA
- Jesse Brown VA Medical Center, Chicago, IL USA
| | - Amul Sakharkar
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL USA
- Jesse Brown VA Medical Center, Chicago, IL USA
| | - Subhash C. Pandey
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL USA
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL USA
- Jesse Brown VA Medical Center, Chicago, IL USA
- Department of Psychiatry, University of Illinois at Chicago and Jesse Brown VA Medical Center, 820 S. Damen Avenue (M/C 151), Chicago, IL 60612 USA
| |
Collapse
|
20
|
Abstract
Despite the generally held view that alcohol is an unspecific pharmacological agent, recent molecular pharmacology studies demonstrated that alcohol has only a few known primary targets. These are the NMDA, GABA(A), glycine, 5-hydroxytryptamine 3 (serotonin) and nicotinic ACh receptors as well as L-type Ca(2+) channels and G-protein-activated inwardly rectifying K(+) channels. Following this first hit of alcohol on specific targets in the brain, a second wave of indirect effects on a variety of neurotransmitter/neuropeptide systems is initiated that leads subsequently to the typical acute behavioural effects of alcohol, ranging from disinhibition to sedation and even hypnosis, with increasing concentrations of alcohol. Besides these acute pharmacodynamic aspects of alcohol, we discuss the neurochemical substrates that are involved in the initiation and maintenance phase of an alcohol drinking behaviour. Finally, addictive behaviour towards alcohol as measured by alcohol-seeking and relapse behaviour is reviewed in the context of specific neurotransmitter/neuropeptide systems and their signalling pathways. The activity of the mesolimbic dopaminergic system plays a crucial role during the initiation phase of alcohol consumption. Following long-term, chronic alcohol consumption virtually all brain neurotransmission seems to be affected, making it difficult to define which of the systems contributes the most to the transition from controlled to compulsive alcohol use. However, compulsive alcohol drinking is characterized by a decrease in the function of the reward neurocircuitry and a recruitment of antireward/stress mechanisms comes into place, with a hypertrophic corticotropin-releasing factor system and a hyperfunctional glutamatergic system being the most important ones.
Collapse
|
21
|
Katsura M, Shibasaki M, Kurokawa K, Tsujimura A, Ohkuma S. Up-regulation of L-type high voltage-gated calcium channel subunits by sustained exposure to 1,4- and 1,5-benzodiazepines in cerebrocortical neurons. J Neurochem 2007; 103:2518-28. [PMID: 17949410 DOI: 10.1111/j.1471-4159.2007.04984.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The aim of this study is to examine how sustained exposure to two 1,4-benzodiazepines (BZDs) with different action period, diazepam and brotizolam, and a 1,5-BZD, clobazam, affects L-type high voltage-gated calcium channel (HVCC) functions and its mechanisms using primary cultures of mouse cerebral cortical neurons. The sustained exposure to these three BZDs increased [(45)Ca2+] influx, which was due to the enhanced [(45)Ca2+] entry through L-type HVCCs but not through of Cav2.1 and Cav2.2. Increase in [(3)H]diltiazem binding after the exposure to these three BZDs was due to the increase in the binding sites of [(3)H]diltiazem. Western blot analysis showed increase of Cav1.2 and Cav1.3 in association with the increased expression of alpha2/delta1 subunit. Similar changes in [(3)H]diltiazem binding and L-type HVCC subunit expression were found in the cerebral cortex from mouse with BZD physical dependence. These results indicate that BZDs examined here have the potential to increase L-type HVCC functions mediated via the enhanced expression of not only Cav1.2 and Cav1.3 but also alpha2/delta1 subunit after their sustained exposure, which may participate in the development of physical dependence by these BZDs.
Collapse
Affiliation(s)
- Masashi Katsura
- Department of Pharmacology, Kawasaki Medical University, Kurashiki, Japan
| | | | | | | | | |
Collapse
|
22
|
Shibasaki M, Katsura M, Kurokawa K, Torigoe F, Ohkuma S. Regional Differences of L-type High Voltage-Gated Calcium Channel Subunit Expression in the Mouse Brain After Chronic Morphine Treatment. J Pharmacol Sci 2007; 105:177-83. [DOI: 10.1254/jphs.fp0070885] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|