1
|
Gómez LCG, Medina NB, Blasco SS, Gravielle MC. Diazepam-Induced Down-Regulation of The Gaba a Receptor α1 Subunit, as Mediated by the Activation of L-Type Voltage-Gated Calcium Channel/Ca 2+/Protein Kinase A Signaling Cascade. Neurosci Lett 2023:137358. [PMID: 37356564 DOI: 10.1016/j.neulet.2023.137358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
Benzodiazepines are among the most prescribed drug class worldwide to treat disorders such as anxiety, insomnia, muscle spasticity, and convulsive disorders, and to induce presurgical sedation. Although benzodiazepines exhibit a high therapeutic index and low toxicity in short-term treatments, prolonged administration induces tolerance to most of their therapeutic actions. The mechanism of this tolerance remains unclear. The central actions of benzodiazepines are mediated by binding to GABAA receptors, which mediate most fast inhibitory transmission in the brain. The majority of GABAA receptors are composed of two α-(1-6), two β-(1-3) and one γ-subunits (1-3). In a previous report, we demonstrated that the prolonged exposure of cerebrocortical neurons to diazepam produces a transcriptional repression of the GABAA receptor α1 subunit gene via a mechanism dependent on the activation of L-type voltage-gated calcium channels (L-VGCCs). The results reported here confirm that the diazepam-induced downregulation of the α1 subunit is contingent upon calcium influx from extracellular space. In addition, this regulatory mechanism involves the activation of protein kinase A (PKA) and is accompanied by the activation of two transcription factors, the cAMP-response element-binding protein (CREB) and the inducible cAMP early repressor (ICER). Together, our results suggest that diazepam's activation of an L-VGCC/Ca2+/PKA/CREB-ICER signaling pathway is responsible for the regulation of GABAA receptors. This elucidation of the intracellular signaling cascade activated by a prolonged benzodiazepine exposure, itself potentially involved in the development of tolerance, may contribute to locating molecular targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Leydi Carolina González Gómez
- Instituto de Investigaciones Farmacológicas (ININFA). Facultad de Farmacia y Bioquímica. Universidad de Buenos Aires. CONICET. Buenos Aires, Argentina
| | - Nelsy Beatriz Medina
- Instituto de Investigaciones Farmacológicas (ININFA). Facultad de Farmacia y Bioquímica. Universidad de Buenos Aires. CONICET. Buenos Aires, Argentina
| | - Sara Sanz Blasco
- Instituto de Investigaciones Farmacológicas (ININFA). Facultad de Farmacia y Bioquímica. Universidad de Buenos Aires. CONICET. Buenos Aires, Argentina
| | - María Clara Gravielle
- Instituto de Investigaciones Farmacológicas (ININFA). Facultad de Farmacia y Bioquímica. Universidad de Buenos Aires. CONICET. Buenos Aires, Argentina.
| |
Collapse
|
2
|
Diazepam induces retrograde facilitation of object recognition and object location memory in male mice. Neuroreport 2023; 34:137-143. [PMID: 36574287 DOI: 10.1097/wnr.0000000000001869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Benzodiazepines are widely prescribed for patients suffering from anxiety and insomnia. Although amnesic effects of benzodiazepines are commonly known as side effects, it has also been reported that these drugs improve memory for information learned before drug intake, a phenomenon called retrograde facilitation. However, the retrograde effects of benzodiazepines on cognitive performances in rodents remain controversial. It should be considered that studies on diazepam-induced retrograde facilitation in humans have been conducted using a recall paradigm focused on short-term memory, whereas these studies in rodents have been conducted using memory tasks that mainly target long-term memory and/or require negative or positive reinforcers. In the current study, we investigated whether diazepam, a benzodiazepine, induces retrograde facilitation for object recognition memory and spatial memory in mice, using a novel object recognition test and an object location test, respectively. These tests are available for short-term memory and do not require any reinforcer. The mice treated with diazepam retained object recognition memory for at least 180 min and spatial memory for at least 150 min. In contrast, vehicle-treated control mice retained object recognition memory for 120 min but not 150 min or longer, and spatial memory for 90 min but not 120 min or longer. These data clearly demonstrated diazepam-induced retrograde facilitation for both object recognition and spatial memories in mice. The present study is expected to contribute to the elucidation of the neural basis of retrograde facilitation.
Collapse
|
3
|
Engin E. GABA A receptor subtypes and benzodiazepine use, misuse, and abuse. Front Psychiatry 2023; 13:1060949. [PMID: 36713896 PMCID: PMC9879605 DOI: 10.3389/fpsyt.2022.1060949] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/29/2022] [Indexed: 01/14/2023] Open
Abstract
Benzodiazepines have been in use for over half a century. While they remain highly prescribed, their unfavorable side-effect profile and abuse liability motivated a search for alternatives. Most of these efforts focused on the development of benzodiazepine-like drugs that are selective for specific GABAA receptor subtypes. While there is ample evidence that subtype-selective GABAA receptor ligands have great potential for providing symptom relief without typical benzodiazepine side-effects, it is less clear whether subtype-selective targeting strategies can also reduce misuse and abuse potential. This review focuses on the three benzodiazepine properties that are relevant to the DSM-5-TR criteria for Sedative, Hypnotic, or Anxiolytic Use Disorder, namely, reinforcing properties of benzodiazepines, maladaptive behaviors related to benzodiazepine use, and benzodiazepine tolerance and dependence. We review existing evidence regarding the involvement of different GABAA receptor subtypes in each of these areas. The reviewed studies suggest that α1-containing GABAA receptors play an integral role in benzodiazepine-induced plasticity in reward-related brain areas and might be involved in the development of tolerance and dependence to benzodiazepines. However, a systematic comparison of the contributions of all benzodiazepine-sensitive GABAA receptors to these processes, a mechanistic understanding of how the positive modulation of each receptor subtype might contribute to the brain mechanisms underlying each of these processes, and a definitive answer to the question of whether specific chronic modulation of any given subtype would result in some or all of the benzodiazepine effects are currently lacking from the literature. Moreover, how non-selective benzodiazepines might lead to the maladaptive behaviors listed in DSM and how different GABAA receptor subtypes might be involved in the development of these behaviors remains unexplored. Considering the increasing burden of benzodiazepine abuse, the common practice of benzodiazepine misuse that leads to severe dependence, and the current efforts to generate side-effect free benzodiazepine alternatives, there is an urgent need for systematic, mechanistic research that provides a better understanding of the brain mechanisms of benzodiazepine misuse and abuse, including the involvement of specific GABAA receptor subtypes in these processes, to establish an informed foundation for preclinical and clinical efforts.
Collapse
Affiliation(s)
- Elif Engin
- Stress Neurobiology Laboratory, Division of Basic Neuroscience, McLean Hospital, Belmont, MA, United States
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Cornett EM, Amarasinghe SN, Angelette A, Abubakar T, Kaye AM, Kaye AD, Neuchat EE, Urits I, Viswanath O. VALTOCO ® (Diazepam Nasal Spray) for the Acute Treatment of Intermittent Stereotypic Episodes of Frequent Seizure Activity. Neurol Int 2021; 13:64-78. [PMID: 33670456 PMCID: PMC7931041 DOI: 10.3390/neurolint13010007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 11/17/2022] Open
Abstract
Valtoco® is a new FDA-approved nasal spray version of diazepam indicated for the treatment of acute, intermittent, and stereotypic episodes of frequent seizure activity in epilepsy patients six years of age and older. Although IV and rectal diazepam are already used to treat seizure clusters, Valtoco® has less variability in plasma concentration compared to rectal diazepam. Furthermore, the intranasal administration of Valtoco® is more convenient and less invasive than rectal or IV diazepam, making it ideal for self-administration outside of a hospital setting. Multiple clinical trials have taken place comparing Valtoco® to the oral, rectal, and IV forms of diazepam. Aside from mild nasal irritation and lacrimation, Valtoco® was found to have no increased safety risk in comparison to traditional forms of diazepam. This review of Valtoco® will include a history of diazepam prescribing and withdrawal treatment, Valtoco® drug information, its mechanism of action, pharmacokinetics and pharmacodynamics, and a comprehensive review of clinical studies.
Collapse
Affiliation(s)
- Elyse M. Cornett
- Department of Anesthesiology, LSU Health Shreveport, Shreveport, LA 71103, USA; (S.N.A.); (A.A.); (T.A.); (A.D.K.); (I.U.); (O.V.)
| | - Sam N. Amarasinghe
- Department of Anesthesiology, LSU Health Shreveport, Shreveport, LA 71103, USA; (S.N.A.); (A.A.); (T.A.); (A.D.K.); (I.U.); (O.V.)
| | - Alexis Angelette
- Department of Anesthesiology, LSU Health Shreveport, Shreveport, LA 71103, USA; (S.N.A.); (A.A.); (T.A.); (A.D.K.); (I.U.); (O.V.)
| | - Tunde Abubakar
- Department of Anesthesiology, LSU Health Shreveport, Shreveport, LA 71103, USA; (S.N.A.); (A.A.); (T.A.); (A.D.K.); (I.U.); (O.V.)
| | - Adam M. Kaye
- Department of Pharmacy Practice, Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA 95211, USA;
| | - Alan David Kaye
- Department of Anesthesiology, LSU Health Shreveport, Shreveport, LA 71103, USA; (S.N.A.); (A.A.); (T.A.); (A.D.K.); (I.U.); (O.V.)
- LSU School of Medicine, LSUHSC New Orleans, New Orleans, LA 70112, USA
| | - Elisa E. Neuchat
- School of Medicine, Florida International University, Miami, FL 33199, USA;
| | - Ivan Urits
- Department of Anesthesiology, LSU Health Shreveport, Shreveport, LA 71103, USA; (S.N.A.); (A.A.); (T.A.); (A.D.K.); (I.U.); (O.V.)
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Critical Care, and Pain Medicine, Boston, MA 02215, USA
| | - Omar Viswanath
- Department of Anesthesiology, LSU Health Shreveport, Shreveport, LA 71103, USA; (S.N.A.); (A.A.); (T.A.); (A.D.K.); (I.U.); (O.V.)
- Valley Anesthesiology and Pain Consultants–Envision Physician Services, Phoenix, AZ 85004, USA
- Department of Anesthesiology, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85724, USA
- Department of Anesthesiology, Creighton University School of Medicine, Omaha, NE 68124, USA
| |
Collapse
|
5
|
Foitzick MF, Medina NB, Iglesias García LC, Gravielle MC. Benzodiazepine exposure induces transcriptional down-regulation of GABA A receptor α1 subunit gene via L-type voltage-gated calcium channel activation in rat cerebrocortical neurons. Neurosci Lett 2020; 721:134801. [PMID: 32007495 DOI: 10.1016/j.neulet.2020.134801] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/24/2020] [Accepted: 01/30/2020] [Indexed: 01/10/2023]
Abstract
GABAA receptors are targets of different pharmacologically relevant drugs, such as barbiturates, benzodiazepines, and anesthetics. In particular, benzodiazepines are prescribed for the treatment of anxiety, sleep disorders, and seizure disorders. Benzodiazepines potentiate GABA responses by binding to GABAA receptors, which are mainly composed of α (1-3, 5), β2, and γ2 subunits. Prolonged activation of GABAA receptors by endogenous and exogenous modulators induces adaptive changes that lead to tolerance. For example, chronic administration of benzodiazepines produces tolerance to most of their pharmacological actions, limiting their usefulness. The mechanism of benzodiazepine tolerance is still unknown. To investigate the molecular basis of tolerance, we studied the effect of sustained exposure of rat cerebral cortical neurons to diazepam on the GABAA receptor. Flunitrazepam binding experiments showed that diazepam treatment induced uncoupling between GABA and benzodiazepine sites, which was blocked by co-incubation with flumazenil, picrotoxin, or nifedipine. Diazepam also produced selective transcriptional down-regulation of GABAA receptor α1 subunit gene through a mechanism dependent on the activation of L-type voltage-gated calcium channels. These findings suggest benzodiazepine-induced stimulation of calcium influx through L-type voltage-gated calcium channels triggers the activation of a signaling pathway that leads to uncoupling and an alteration of receptor subunit expression. Insights into the mechanism of benzodiazepine tolerance will contribute to the design of new drugs that can maintain their efficacies after long-term treatments.
Collapse
Affiliation(s)
- María Florencia Foitzick
- Instituto de Investigaciones Farmacológicas (ININFA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires. CONICET, Buenos Aires, Argentina
| | - Nelsy Beatriz Medina
- Instituto de Investigaciones Farmacológicas (ININFA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires. CONICET, Buenos Aires, Argentina
| | - Lucía Candela Iglesias García
- Instituto de Investigaciones Farmacológicas (ININFA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires. CONICET, Buenos Aires, Argentina
| | - María Clara Gravielle
- Instituto de Investigaciones Farmacológicas (ININFA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires. CONICET, Buenos Aires, Argentina.
| |
Collapse
|
6
|
Levitt JG, Kalender G, O’Neill J, Diaz JP, Cook IA, Ginder N, Krantz D, Minzenberg MJ, Vince-Cruz N, Nguyen LD, Alger JR, Leuchter AF. Dorsolateral prefrontal γ-aminobutyric acid in patients with treatment-resistant depression after transcranial magnetic stimulation measured with magnetic resonance spectroscopy. J Psychiatry Neurosci 2019; 44:386-394. [PMID: 31199104 PMCID: PMC6821508 DOI: 10.1503/jpn.180230] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The therapeutic mechanism of repetitive transcranial magnetic stimulation (rTMS) for treatment-resistant depression (TRD) may involve modulation of γ-aminobutyric acid (GABA) levels. We used proton magnetic resonance spectroscopy (MRS) to assess changes in GABA levels at the site of rTMS in the left dorsolateral prefrontal cortex (DLPFC). METHODS In 26 adults with TRD, we used Mescher–Garwood point-resolved spectroscopy (MEGA-PRESS) spectral-editing MRS to measure GABA in the left DLPFC before and after standard clinical treatment with rTMS. All participants but 1 were medicated, including 12 patients on GABA agonist agents. RESULTS Mean GABA in the DLPFC increased 10.0% (p = 0.017) post-rTMS in the overall sample. As well, GABA increased significantly in rTMS responders (n = 12; 23.6%, p = 0.015) but not in nonresponders (n = 14; 4.1%, p = not significant). Changes in GABA were not significantly affected by GABAergic agonists, but clinical response was less frequent (p = 0.005) and weaker (p = 0.035) in the 12 participants who were receiving GABA agonists concomitant with rTMS treatment. LIMITATIONS This study had an open-label design in a population receiving naturalistic treatment. CONCLUSION Treatment using rTMS was associated with increases in GABA levels at the stimulation site in the left DLPFC, and the degree of GABA change was related to clinical improvement. Participants receiving concomitant treatment with a GABA agonist were less likely to respond to rTMS. These findings were consistent with earlier studies showing the effects of rTMS on GABA levels and support a GABAergic model of depression.
Collapse
Affiliation(s)
- Jennifer G. Levitt
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - Guldamla Kalender
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - Joseph O’Neill
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - Joel P. Diaz
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - Ian A. Cook
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - Nathaniel Ginder
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - David Krantz
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - Michael J. Minzenberg
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - Nikita Vince-Cruz
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - Lydia D. Nguyen
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - Jeffry R. Alger
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| | - Andrew F. Leuchter
- From the Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, Diaz, Cook, Ginder, Krantz, Minzenberg, Vince-Cruz, Nguyen, Leuchter); the Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles (Levitt, Kalender, O’Neill, Cook, Krantz, Minzenberg, Leuchter); the Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles (Kalender); the Division of Child and Adolescent Psychiatry, Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles (Levitt, O’Neill); the Department of Bioengineering, Henry Samueli School of Engineering at Applied Science at UCLA, Los Angeles (Cook); the Department of Neurology, UCLA David Geffen School of Medicine at UCLA, Los Angeles (Alger); the Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas (Alger); and the NeuroSpectroScopics, LCC, Sherman Oaks, California (Alger)
| |
Collapse
|
7
|
Calvo-Flores Guzmán B, Vinnakota C, Govindpani K, Waldvogel HJ, Faull RL, Kwakowsky A. The GABAergic system as a therapeutic target for Alzheimer's disease. J Neurochem 2018; 146:649-669. [DOI: 10.1111/jnc.14345] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/21/2018] [Accepted: 03/14/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Beatriz Calvo-Flores Guzmán
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| | - Chitra Vinnakota
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| | - Karan Govindpani
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| | - Henry J. Waldvogel
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| | - Richard L.M. Faull
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research; Faculty of Medical and Health Sciences; Department of Anatomy and Medical Imaging; University of Auckland; Auckland New Zealand
| |
Collapse
|
8
|
Roohbakhsh A, Shamsizadeh A, Arababadi MK, Ayoobi F, Fatemi I, Allahtavakoli M, Mohammad-Zadeh M. Tactile learning in rodents: Neurobiology and neuropharmacology. Life Sci 2016; 147:1-8. [DOI: 10.1016/j.lfs.2016.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 12/28/2015] [Accepted: 01/19/2016] [Indexed: 12/20/2022]
|
9
|
Gravielle MC. Activation-induced regulation of GABAA receptors: Is there a link with the molecular basis of benzodiazepine tolerance? Pharmacol Res 2015; 109:92-100. [PMID: 26733466 DOI: 10.1016/j.phrs.2015.12.030] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 12/01/2022]
Abstract
Benzodiazepines have been used clinically for more than 50 years to treat disorders such as insomnia, anxiety, and epilepsy, as well as to aid muscle relaxation and anesthesia. The therapeutic index for benzodiazepines if very high and the toxicity is low. However, their usefulness is limited by the development of either or both tolerance to most of their pharmacological actions and dependence. Tolerance develops at different rates depending on the pharmacological action, suggesting the existence of distinct mechanisms for each behavioral parameter. Alternatively, multiple mechanisms could coexist depending on the subtype of GABAA receptor expressed and the brain region involved. Because most of the pharmacological actions of benzodiazepines are mediated through GABAA receptor binding, adaptive alterations in the number, structure, and/or functions of these receptors may play an important role in the development of tolerance. This review is focused on the regulation of GABAA receptors induced by long-term benzodiazepine exposure and its relationship with the development of tolerance. Understanding the mechanisms behind benzodiazepine tolerance is critical for designing drugs that could maintain their efficacy during long-term treatments.
Collapse
Affiliation(s)
- María Clara Gravielle
- Instituto de Investigaciones Farmacológicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Junín 956, C1113AAD Buenos Aires, Argentina.
| |
Collapse
|
10
|
Orzelska J, Talarek S, Listos J, Fidecka S. Divergent effects of L-arginine-NO pathway modulators on diazepam and flunitrazepam responses in NOR task performance. Behav Brain Res 2015; 284:179-86. [PMID: 25698597 DOI: 10.1016/j.bbr.2015.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 02/02/2015] [Accepted: 02/06/2015] [Indexed: 01/09/2023]
Abstract
The goal of the study was an evaluation of the degree, in which nitric oxide (NO) is involved in the benzodiazepines (BZs)-induced recognition memory impairment in rats. The novel object recognition (NOR) test was used to examine recognition memory. The current research focused on the object memory impairing effects of diazepam (DZ; 0.5 and 1mg/kg, sc) and flunitrazepam (FNZ; 0.1 and 0.2mg/kg; sc) in 1-hour delay periods in rats. It was found that acute ip injection of L-arginine (L-arg; 250 and 500 mg/kg; ip), 5 min before DZ administration (0.5mg/kg, sc) prevented DZ-induced memory deficits. On the other hand, it was also proven that L-arg (125, 250 and 500 mg/kg; ip) did not change the behaviour of rats in the NOR test, following a combined administration with FNZ at a threshold dose (0.05 mg/kg; sc). It was also found that 7-nitroindazole (7-NI; 10, 20 and 40 mg/kg; ip) induced amnesic effects in DZ in rats, submitted to the NOR test, following a combined administration of 7-NI with a threshold dose of DZ (0.25mg/kg; sc). However, following a combined administration of 7-NI (10, 20 and 40 mg/kg; ip) with FNZ (0.1mg/kg; sc), it was observed that 7-NI inhibited the amnesic effects of FNZ on rats in the NOR test. Those findings led us to hypothesize that NO synthesis suppression may induce amnesic effects of DZ, while preventing FNZ memory impairment in rats, submitted to NOR tasks.
Collapse
Affiliation(s)
- Jolanta Orzelska
- Chair and Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4A, 20-093 Lublin, Poland.
| | - Sylwia Talarek
- Chair and Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4A, 20-093 Lublin, Poland
| | - Joanna Listos
- Chair and Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4A, 20-093 Lublin, Poland
| | - Sylwia Fidecka
- Chair and Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4A, 20-093 Lublin, Poland
| |
Collapse
|
11
|
Wright BT, Gluszek CF, Heldt SA. The effects of repeated zolpidem treatment on tolerance, withdrawal-like symptoms, and GABAA receptor mRNAs profile expression in mice: comparison with diazepam. Psychopharmacology (Berl) 2014; 231:2967-79. [PMID: 24531568 DOI: 10.1007/s00213-014-3473-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 01/26/2014] [Indexed: 10/25/2022]
Abstract
RATIONALE Zolpidem is a short-acting, non-benzodiazepine hypnotic that acts as a full agonist at α1-containing GABAA receptors. Overall, zolpidem purportedly has fewer instances of abuse and dependence than traditionally used benzodiazepines. However, several studies have shown that zolpidem may be more similar to benzodiazepines in terms of behavioral tolerance and withdrawal symptoms. OBJECTIVES In the current study, we examined whether subchronic zolpidem or diazepam administration produced deficits in zolpidem's locomotor-impairing effects, anxiety-like behaviors, and changes in GABAAR subunit messenger RNA (mRNA). METHODS Mice were given subchronic injections of either zolpidem (10 mg/kg), diazepam (20 mg/kg), or vehicle twice daily for 7 days. On day 8, mice were given a challenge dose of zolpidem (2 mg/kg) or vehicle before open field testing. Another set of mice underwent the same injection regimen but were sacrificed on day 8 for qRT-PCR analysis. RESULTS We found that subchronic zolpidem and diazepam administration produced deficits in the acute locomotor-impairing effects of zolpidem and increased anxiety-like behaviors 1 day after drug termination. In addition, we found that subchronic treatment of zolpidem and diazepam induced distinct but overlapping GABAAR subunit mRNA changes in the cortex but few changes in the hippocampus, amygdala, or prefrontal cortex. Levels of mRNA measured in separate mice after a single injection of either zolpidem or diazepam revealed no mRNA changes. CONCLUSIONS In mice, subchronic treatment of zolpidem and diazepam can produce deficits in the locomotor-impairing effects of zolpidem, anxiety-like withdrawal symptoms, and subunit-specific mRNA changes.
Collapse
Affiliation(s)
- Brittany T Wright
- The Department of Anatomy and Neurobiology, Neuroscience Institute, University of Tennessee Health Science Center, 855 Monroe Ave, Memphis, TN, 38163, USA
| | | | | |
Collapse
|
12
|
Calcaterra NE, Barrow JC. Classics in chemical neuroscience: diazepam (valium). ACS Chem Neurosci 2014; 5:253-60. [PMID: 24552479 DOI: 10.1021/cn5000056] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Diazepam (Valium) is among the most successful drugs from the onset of the psychopharmacological revolution that began during the 1950s. Efficacious in treating a wide-spectrum of CNS disorders, including anxiety and epilepsy, it set the standard for pharmacotherapy in terms of potency, onset of action, and safety. In this Review, the legacy of diazepam to chemical neuroscience will be considered along with its synthesis, pharmacology, drug metabolism, adverse events and dependence, clinical use, and regulatory issues.
Collapse
Affiliation(s)
- Nicholas E. Calcaterra
- Department
of Pharmacology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - James C. Barrow
- Department
of Pharmacology, Johns Hopkins University, Baltimore, Maryland 21205, United States
- Lieber Institute for Brain Development, Baltimore, Maryland 21205, United States
| |
Collapse
|
13
|
Dong C, Hu A, Ni Y, Zuo Y, Li GH. Effects of midazolam, pentobarbital and ketamine on the mRNA expression of ion channels in a model organism Daphnia pulex. BMC Anesthesiol 2013; 13:32. [PMID: 24134334 PMCID: PMC3879215 DOI: 10.1186/1471-2253-13-32] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/07/2013] [Indexed: 02/05/2023] Open
Abstract
Background Over the last few decades intensive studies have been carried out on the molecular targets mediating general anesthesia as well as the effects of general anesthetics. The γ-aminobutyric acid type A receptor (GABAAR) has been indicated as the primary target of general anaesthetics such as propofol, etomidate and isoflurane, and sedating drugs including benzodiazepines and barbiturates. The GABAAR is also involved in drug tolerance and dependence. However, the involvement of other ion channels is possible. Methods Using reverse transcription and quantitative PCR techniques, we systematically investigated changes in the mRNA levels of ion channel genes in response to exposure to midazolam, pentobarbital and ketamine in a freshwater model animal, Daphnia pulex. To retrieve the sequences of Daphnia ion channel genes, Blast searches were performed based on known human or Drosophila ion channel genes. Retrieved sequences were clustered with the maximum-likelihood method. To quantify changes in gene expression after the drug treatments for 4 hours, total RNA was extracted and reverse transcribed into cDNA and then amplified using quantitative PCR. Results A total of 108 ion channel transcripts were examined, and 19, 11 and 11 of them are affected by midazolam (100 μM), pentobarbital (200 μM) and ketamine (100 μM), respectively, covering a wide variety of ion channel types. There is some degree of overlap with midazolam- and pentobarbital-induced changes in the mRNA expression profiles, but ketamine causes distinct changes in gene expression pattern. In addition, flumazenil (10 μM) eliminates the effect of midazolam on the mRNA expression of the GABAA receptor subunit Rdl, suggesting a direct interaction between midazolam and GABAA receptors. Conclusions Recent research using high throughput technology suggests that changes in mRNA expression correlate with delayed protein expression. Therefore, the mRNA profile changes in our study may reflect the molecular targets not only in drug actions, but also in chronic drug addiction. Our data also suggest the possibility that hypnotic/anesthetic drugs are capable of altering the functions of the nervous system, as well as those non-nerve tissues with abundant ion channel expressions.
Collapse
Affiliation(s)
| | | | | | | | - Guo Hua Li
- Laboratory of Anesthesiology and Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
14
|
PWZ-029, an inverse agonist selective for α₅ GABAA receptors, improves object recognition, but not water-maze memory in normal and scopolamine-treated rats. Behav Brain Res 2012; 241:206-13. [PMID: 23261875 DOI: 10.1016/j.bbr.2012.12.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 01/03/2023]
Abstract
Inverse agonism at the benzodiazepine site of α(5) subunit-containing GABA(A) receptors is an attractive approach for the development of putative cognition-enhancing compounds, which are still far from clinical application. Several ligands with binding and/or functional selectivity for α(5) GABA(A) receptors have been synthesized and tested in a few animal models. PWZ-029 is an α(5) GABA(A) selective inverse agonist whose memory enhancing effects were demonstrated in the passive avoidance task in rats and in Pavlovian fear conditioning in mice. In the present study we investigated the effects of PWZ-029 administration in novel object recognition test and Morris water maze, in normal and scopolamine-treated rats. All the three doses of PWZ-029 (2, 5 and 10 mg/kg) improved object recognition after the 24-h delay period, as shown by significant differences between the exploration times of the novel and old object, and the respective discrimination indices. PWZ-029 (2 mg/kg) also successfully reversed the 0.3 mg/kg scopolamine-induced deficit in recognition memory after the 1-h delay. In the Morris water maze test, PWZ-029 (5, 10 and 15 mg/kg) did not significantly influence swim patterns, either during five acquisition days or during the treatment-free probe trial. PWZ-029 (2, 5 and 10 mg/kg) also proved to be ineffective in the reversal of the 1mg/kg scopolamine-induced memory impairment in the water maze. The present mixed results encourage use of a variety of tests and experimental conditions in order to increase the predictability of preclinical testing of selective α(5) GABA(A) inverse agonists.
Collapse
|
15
|
Guidotti A, Grayson DR. A neurochemical basis for an epigenetic vision of psychiatric disorders (1994-2009). Pharmacol Res 2011; 64:344-9. [PMID: 21699980 DOI: 10.1016/j.phrs.2011.05.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In 1996, Dr. Costa was invited by Prof. Boris Astrachan, Chairman of the Department of Psychiatry at the University of Illinois at Chicago, to direct the research of the "Psychiatric Institute, Department of Psychiatry, School of Medicine, at the University of Illinois at Chicago." He was asked to develop a seminal research program on psychiatric disorders. Viewed in retrospect, Dr. Costa met and surpassed the challenge, as was usual for him. To elucidate the molecular mechanisms whereby nurture (epigenetic factors) and nature (genetic factors) interact to cause major psychiatric disorders was at the center of Dr. Costa's mission for the last 15 years of his research at the Psychiatric Institute. The challenge for Dr. Costa and his colleagues (Auta, Caruncho, Davis, Grayson, Guidotti, Impagnatiello, Kiedrowski, Larson, Manev, Pappas, Pesold, Pinna, Sharma, Smalheiser, Sugaya, Tueting, Veldic [1-111]) had always been to find new ways to prevent and treat psychiatric disorders with pharmacological agents that failed to have major unwanted side effects. In this list, we have quoted the first authors of the papers pertaining to the field of research highlighted in the title. As you know, Dr. Costa was an eclectic scientist and in his 15 years of studies at UIC, he touched many other aspects of neuroscience research that are not discussed in this overview.
Collapse
Affiliation(s)
- Alessandro Guidotti
- The Psychiatric Institute, Department of Psychiatry, University of Illinois at Chicago, 1601 Taylor, Chicago, IL 60612, United States.
| | | |
Collapse
|
16
|
Kealy J, Commins S. The rat perirhinal cortex: A review of anatomy, physiology, plasticity, and function. Prog Neurobiol 2011; 93:522-48. [DOI: 10.1016/j.pneurobio.2011.03.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 01/28/2011] [Accepted: 03/10/2011] [Indexed: 11/26/2022]
|
17
|
Enna SJ. A legacy of discovery: from monoamines to GABA. Neuropharmacology 2010; 60:1050-7. [PMID: 20951150 DOI: 10.1016/j.neuropharm.2010.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 10/06/2010] [Indexed: 10/18/2022]
Abstract
Seldom does a single individual have such a profound effect on the development of a scientific discipline as Erminio Costa had on neuropharmacology. During nearly sixty years of research, Costa and his collaborators helped established many of the basic principles of the pharmacodynamic actions of psychotherapeutics. His contributions range from defining basic neurochemical, physiological and behavioral properties of neurotransmitters and their receptors, to the development of novel theories for drug discovery. Outlined in this report is a portion of his work relating to the involvement of monoamines and GABA in mediating the symptoms of neuropsychiatric disorders and as targets for drug therapies. These studies were selected for review because of their influence on my own work and as an illustration of his logical and insightful approach to research and his clever use of techniques and technologies. Given the significance of his work, the legions of scientist who collaborated with him, and those inspired by his reports, his research will continue to have an impact as long as there is a search for new therapeutics to alleviate the pain and suffering associated with neurological and psychiatric disorders. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.
Collapse
Affiliation(s)
- S J Enna
- Departments of Molecular and Integrative Physiology, and of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA.
| |
Collapse
|
18
|
Uusi-Oukari M, Korpi ER. Regulation of GABA(A) receptor subunit expression by pharmacological agents. Pharmacol Rev 2010; 62:97-135. [PMID: 20123953 DOI: 10.1124/pr.109.002063] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The gamma-aminobutyric acid (GABA) type A receptor system, the main fast-acting inhibitory neurotransmitter system in the brain, is the pharmacological target for many drugs used clinically to treat, for example, anxiety disorders and epilepsy, and to induce and maintain sedation, sleep, and anesthesia. These drugs facilitate the function of pentameric GABA(A) receptors that exhibit widespread expression in all brain regions and large structural and pharmacological heterogeneity as a result of composition from a repertoire of 19 subunit variants. One of the main problems in clinical use of GABA(A) receptor agonists is the development of tolerance. Most drugs, in long-term use and during withdrawal, have been associated with important modulations of the receptor subunit expression in brain-region-specific manner, participating in the mechanisms of tolerance and dependence. In most cases, the molecular mechanisms of regulation of subunit expression are poorly known, partly as a result of neurobiological adaptation to altered neuronal function. More knowledge has been obtained on the mechanisms of GABA(A) receptor trafficking and cell surface expression and the processes that may contribute to tolerance, although their possible pharmacological regulation is not known. Drug development for neuropsychiatric disorders, including epilepsy, alcoholism, schizophrenia, and anxiety, has been ongoing for several years. One key step to extend drug development related to GABA(A) receptors is likely to require deeper understanding of the adaptational mechanisms of neurons, receptors themselves with interacting proteins, and finally receptor subunits during drug action and in neuropsychiatric disease processes.
Collapse
Affiliation(s)
- Mikko Uusi-Oukari
- Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Itainen Pitkakatu 4, 20014 Turku, Finland.
| | | |
Collapse
|
19
|
Auta J, Impagnatiello F, Kadriu B, Guidotti A, Costa E. Imidazenil: a low efficacy agonist at alpha1- but high efficacy at alpha5-GABAA receptors fail to show anticonvulsant cross tolerance to diazepam or zolpidem. Neuropharmacology 2008; 55:148-53. [PMID: 18555494 PMCID: PMC2601598 DOI: 10.1016/j.neuropharm.2008.05.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 04/18/2008] [Accepted: 05/01/2008] [Indexed: 02/03/2023]
Abstract
Whereas advances in the molecular biology of GABA(A) receptor complex using knock-out and knock-in mice have been valuable in unveiling the structure, composition, receptor assembly, and several functions of different GABA(A) receptor subtypes, the mechanism(s) underlying benzodiazepine (BZ) tolerance and withdrawal remain poorly understood. Studies using specific GABA(A) receptor subunit knock-in mice suggest that tolerance to sedative action of diazepam requires long-term activation of alpha1 and alpha5 GABA(A) receptor subunits. We investigated the role of long-term activation of these GABA(A) receptor subunits during anticonvulsant tolerance using high affinity and high intrinsic efficacy ligands for GABA(A) receptors expressing the alpha5 subunit (imidazenil) or alpha1 subunit (zolpidem), and a non-selective BZ recognition site ligand (diazepam). We report here that long-term activation of GABA(A) receptors by zolpidem and diazepam but not by imidazenil elicits anticonvulsant tolerance. Although anticonvulsant cross-tolerance occurs between diazepam and zolpidem, there is no cross-tolerance between imidazenil and diazepam or zolpidem. Furthermore, diazepam or zolpidem long-term treatment decreased the expression of mRNA encoding the alpha1 GABA(A) receptor subunit in prefrontal cortex by 43% and 20% respectively. In addition, diazepam but not zolpidem long-term treatment produced a 30% increase in the expression of the alpha5 GABA(A) receptor subunit mRNA in prefrontal cortex. In contrast, imidazenil which is devoid of anticonvulsant tolerance does not elicit significant changes in the expression of alpha1 or alpha5 GABA(A) receptor subunit. These findings suggest that long-term activation of GABA(A) receptors containing the alpha1 or other subunits but not the alpha5 receptor subunit is essential for the induction of anticonvulsant tolerance.
Collapse
Affiliation(s)
- James Auta
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois, 1601 W. Taylor St., Chicago, IL 60612, USA.
| | | | | | | | | |
Collapse
|
20
|
Licata SC, Rowlett JK. Abuse and dependence liability of benzodiazepine-type drugs: GABA(A) receptor modulation and beyond. Pharmacol Biochem Behav 2008; 90:74-89. [PMID: 18295321 PMCID: PMC2453238 DOI: 10.1016/j.pbb.2008.01.001] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Revised: 12/14/2007] [Accepted: 01/02/2008] [Indexed: 01/12/2023]
Abstract
Over the past several decades, benzodiazepines and the newer non-benzodiazepines have become the anxiolytic/hypnotics of choice over the more readily abused barbiturates. While all drugs from this class act at the GABA(A) receptor, benzodiazepine-type drugs offer the clear advantage of being safer and better tolerated. However, there is still potential for these drugs to be abused, and significant evidence exists to suggest that this is a growing problem. This review examines the behavioral determinants of the abuse and dependence liability of benzodiazepine-type drugs. Moreover, the pharmacological and putative biochemical basis of the abuse-related behavior is discussed.
Collapse
Affiliation(s)
- Stephanie C Licata
- McLean Hospital/Harvard Medical School, Behavioral Psychopharmacology Research Laboratory, 115 Mill Street, Belmont, MA 02478, United States.
| | | |
Collapse
|
21
|
Birzniece V, Bäckström T, Johansson IM, Lindblad C, Lundgren P, Löfgren M, Olsson T, Ragagnin G, Taube M, Turkmen S, Wahlström G, Wang MD, Wihlbäck AC, Zhu D. Neuroactive steroid effects on cognitive functions with a focus on the serotonin and GABA systems. BRAIN RESEARCH REVIEWS 2006; 51:212-239. [PMID: 16368148 DOI: 10.1016/j.brainresrev.2005.11.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Revised: 11/10/2005] [Accepted: 11/11/2005] [Indexed: 01/20/2023]
Abstract
This article will review neuroactive steroid effects on serotonin and GABA systems, along with the subsequent effects on cognitive functions. Neurosteroids (such as estrogen, progesterone, and allopregnanolone) are synthesized in the central and peripheral nervous system, in addition to other tissues. They are involved in the regulation of mood and memory, in premenstrual syndrome, and mood changes related to hormone replacement therapy, as well as postnatal and major depression, anxiety disorders, and Alzheimer's disease. Estrogen and progesterone have their respective hormone receptors, whereas allopregnanolone acts via the GABA(A) receptor. The action of estrogen and progesterone can be direct genomic, indirect genomic, or non-genomic, also influencing several neurotransmitter systems, such as the serotonin and GABA systems. Estrogen alone, or in combination with antidepressant drugs affecting the serotonin system, has been related to improved mood and well being. In contrast, progesterone can have negative effects on mood and memory. Estrogen alone, or in combination with progesterone, affects the brain serotonin system differently in different parts of the brain, which can at least partly explain the opposite effects on mood of those hormones. Many of the progesterone effects in the brain are mediated by its metabolite allopregnanolone. Allopregnanolone, by changing GABA(A) receptor expression or sensitivity, is involved in premenstrual mood changes; and it also induces cognitive deficits, such as spatial-learning impairment. We have shown that the 3beta-hydroxypregnane steroid UC1011 can inhibit allopregnanolone-induced learning impairment and chloride uptake potentiation in vitro and in vivo. It would be important to find a substance that antagonizes allopregnanolone-induced adverse effects.
Collapse
Affiliation(s)
- Vita Birzniece
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University Hospital, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Birzniece V, Türkmen S, Lindblad C, Zhu D, Johansson IM, Bäckström T, Wahlström G. GABA(A) receptor changes in acute allopregnanolone tolerance. Eur J Pharmacol 2006; 535:125-134. [PMID: 16513107 DOI: 10.1016/j.ejphar.2006.01.059] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2005] [Revised: 01/03/2006] [Accepted: 01/20/2006] [Indexed: 11/17/2022]
Abstract
To study acute tolerance, rats were anesthetized with interrupted i.v. allopregnanolone infusions where the "silent second" in the electroencephalogram (EEG) was the target. Animals were killed either directly at the first silent second or at the silent second level after 30 or 90 min of anaesthesia. Acute tolerance was demonstrated at 90 min of anaesthesia as earlier shown. In situ hybridization showed a decreased expression of the gamma-aminobutyric acid(A) (GABA(A)) receptor subunit alpha4mRNA amount in the thalamus ventral-posteriomedial nucleus of the tolerant rats. A parallel change in the abundance of the alpha4 subunit was detected with immunohistochemistry. The increase in maintenance dose rate (MDR) was significantly negatively correlated with the alpha4mRNA in the thalamus ventral-posteriomedial nucleus, and positively correlated with alpha2mRNA in different hippocampal subregions. There was also a positive relationship between the alpha1mRNA amounts in the different hippocampal subregions, with significant differences between groups. These changes in GABA(A) receptor subunits mRNA expression and protein (alpha4) might be of importance for the development of acute tolerance to allopregnanolone.
Collapse
Affiliation(s)
- Vita Birzniece
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University Hospital, SE 901 85, Sweden
| | | | | | | | | | | | | |
Collapse
|
23
|
Raol YH, Zhang G, Budreck EC, Brooks-Kayal AR. Long-term effects of diazepam and phenobarbital treatment during development on GABA receptors, transporters and glutamic acid decarboxylase. Neuroscience 2005; 132:399-407. [PMID: 15802192 DOI: 10.1016/j.neuroscience.2005.01.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2005] [Indexed: 11/26/2022]
Abstract
Diazepam (DZ) and phenobarbital (PH) are commonly used to treat early-life seizures and act on GABAA receptors (GABAR). The developing GABAergic system is highly plastic, and the long-term effects of postnatal treatment with these drugs on the GABAergic system has not been extensively examined. In the present study, we investigated the effects of prolonged DZ and PH treatment during postnatal development and then discontinuation on expression of a variety of genes involved in GABAergic neurotransmission during adulthood. Rat pups were treated with DZ, PH or vehicle from postnatal day (P) 10-P40 and then the dose was tapered for 2 weeks and terminated at P55. Expression of GABAR subunits, GABAB receptor subunits, GABA transporters (GAT) and GABA synthesizing enzymes (glutamic acid decarboxylase: GAD) mRNAs in hippocampal dentate granule neurons (DGNs) were analyzed using antisense RNA amplification at P90. Protein levels for the alpha1 subunit of GABAR, GAD67, GAT1 and 3 were also assessed using Western blotting. At P90, mRNA expression for GAT-1, 3, 4, GABAR subunits alpha4, alpha6, beta3, delta and theta and GABAB receptor subunit R1 was increased and mRNA expression for GAD65, GAD67 and GABAR subunits alpha1 and alpha3 were decreased in DGNs of rats treated with DZ and PH. The current data suggest that prolonged DZ and PH treatment during postnatal development causes permanent alterations in the expression of hippocampal GABA receptor subunits, GATs and GAD long after therapy has ended.
Collapse
Affiliation(s)
- Y H Raol
- Division of Neurology, Pediatric Regional Epilepsy Program, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
24
|
Guidotti A, Auta J, Davis JM, Dong E, Grayson DR, Veldic M, Zhang X, Costa E. GABAergic dysfunction in schizophrenia: new treatment strategies on the horizon. Psychopharmacology (Berl) 2005; 180:191-205. [PMID: 15864560 DOI: 10.1007/s00213-005-2212-8] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2004] [Accepted: 02/11/2005] [Indexed: 01/15/2023]
Abstract
RATIONALE Cortical gamma-aminobutyric acid (GABA)ergic neurons contribute to the orchestration of pyramidal neuron population firing as follows: (1) by releasing GABA on GABA(A) and GABA(B) receptors, (2) by releasing reelin in the proximity of integrin receptors located on cortical pyramidal neuron dendritic spines, and (3) through reelin contributing to the regulation of dendritic spine plasticity by modulating dendritic resident mRNA translation. In schizophrenia (SZ) and bipolar (BP) postmortem brains, the downregulation of mRNAs encoding glutamic acid decarboxylase 67 (GAD(67)) and reelin decreases the cognate proteins coexpressed in prefrontal cortex (PFC) GABAergic neurons. This finding has been replicated in several laboratories. Such downregulation suggests that the neuropil hypoplasticity found in the PFC of SZ and BP disorder patients may depend on a downregulation of GABAergic function, which is associated with a decrease in reelin secretion from GABAergic neuron axon terminals on dendrites, somata, or axon initial segments of pyramidal neurons. Indirectly, this GABAergic neuron downregulation may play a key role in the expression of positive and negative symptoms of SZ and BP disorders. OBJECTIVES The above described GABAergic dysfunction may be addressed by pharmacological interventions to treat SZ and BP disorders using specific benzodiazepines (BZs), which are devoid of intrinsic activity at GABA(A) receptors including alpha(1) subunits but that act as full positive allosteric modulators of GABA action at GABA(A) receptors containing alpha(2), alpha(3), or alpha(5) subunits. These drugs are expected to enhance GABAergic signal transduction without eliciting sedation, amnesia, and tolerance or dependence liabilities. RESULTS AND CONCLUSIONS BZs, such as diazepam, although they are efficient in equilibrating GABA(A) receptor signal transduction in a manner beneficial in the treatment of positive and negative symptoms of SZ, may not be ideal drugs, because by mediating a full positive allosteric modulation of GABA(A) receptors containing the alpha(1) subunit, they contribute to sedation and to the development of tolerance after even a brief period of treatment. In contrast, other BZ-binding site ligands, such as 6-(2bromophenyl)-8-fluoro-4H-imidazo [1,5-a][1,4] benzodiazepine-3-carboxamide (imidazenil), which fail to allosterically and positively modulate the action of GABA at GABA(A) receptors with alpha(1) subunits but that selectively allosterically modulate cortical GABA(A) receptors containing alpha(5) subunits, contribute to the anxiolytic, antipanic, and anticonvulsant actions of these ligands without producing sedation, amnesia, or tolerance. Strong support for the use of imidazenil in psychosis emerges from experiments with reeler mice or with methionine-treated mice, which express a pronounced reelin and GAD(67) downregulation that is also operative in SZ and BP disorders. In mice that model SZ symptoms, imidazenil increases signal transduction at GABA(A) receptors containing alpha(5) subunits and contributes to the reduction of behavioral deficits without producing sedation or tolerance liability. Hence, we suggest that imidazenil may be considered a prototype for a new generation of positive allosteric modulators of GABA(A) receptors, which, either alone or in combination with neuroleptics, should be evaluated in GABAergic dysfunction operative in the treatment of SZ and BP disorders with psychosis.
Collapse
Affiliation(s)
- Alessandro Guidotti
- Psychiatric Institute, Department of Psychiatry, University of Illinois at Chicago, 1601 Taylor St, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Huopaniemi L, Keist R, Randolph A, Certa U, Rudolph U. Diazepam-induced adaptive plasticity revealed by α1 GABAA receptor-specific expression profiling. J Neurochem 2004; 88:1059-67. [PMID: 15009662 DOI: 10.1046/j.1471-4159.2003.02216.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Benzodiazepines are in wide clinical use for their sedative and tranquilizing actions, the former being mediated via alpha1-containing GABAA receptors. The signal transduction pathways elicited beyond the receptor are only poorly understood. Changes of transcript levels in cerebral cortex induced by acute diazepam administration were therefore compared by microarray analysis between wild-type and point mutated alpha1(H101R) mice, in which the alpha1 GABAA receptor subunit had been rendered insensitive to diazepam. In wild-type animals, diazepam reduced the expression levels of the alpha subunit of the calcium/calmodulin-dependent protein kinase II, as well as brain-derived neurotrophic factor, MAP kinase phosphatase, transcription factor GIF, c-fos and nerve growth factor induced gene-A. None of these transcripts was changed in the alpha1(H101R) mice after treatment with diazepam. Thus, the sedative action of diazepam is correlated with a selective down-regulation of transcripts involved in the regulation of neuronal plasticity and neurotrophic responses. Most transcript changes were transient except for the decrease of the CaMKIIalpha transcript which persisted even 40 h after the single dose of diazepam. This long-term alteration is likely to contribute to the resetting of the neuronal responsiveness, which may be involved in rebound phenomena and, under chronic treatment, in the development of tolerance and dependence.
Collapse
Affiliation(s)
- Laura Huopaniemi
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | | | | | | | | |
Collapse
|
26
|
Abstract
BACKGROUND A converging body of evidence implicates the gamma-aminobutyric acid (GABA) neurotransmitter system in the pathogenesis of schizophrenia. METHODS The authors review neuroscience literature and clinical studies investigating the role of the GABA system in the pathophysiology of schizophrenia. First, a background on the GABA system is provided, including GABA pharmacology and neuroanatomy of GABAergic neurons. Results from basic science schizophrenia animal models and human studies are reviewed. The role of GABA in cognitive dysfunction in schizophrenia is then presented, followed by a discussion of GABAergic compounds used in monotherapy or adjunctively in clinical schizophrenia studies. RESULTS In basic studies, reductions in GABAergic neuronal density and abnormalities in receptors and reuptake sites have been identified in several cortical and subcortical GABA systems. A model has been developed suggesting GABA's role (including GABA-dopamine interactions) in schizophrenia. In several clinical studies, the use of adjunctive GABA agonists was associated with greater improvement in core schizophrenia symptoms. CONCLUSIONS Alterations in the GABA neurotransmitter system are found in clinical and basic neuroscience schizophrenia studies as well as animal models and may be involved in the pathophysiology of schizophrenia. The interaction of GABA with other well-characterized neurotransmitter abnormalities remains to be understood. Future studies should elucidate the potential therapeutic role for GABA ligands in schizophrenia treatment.
Collapse
Affiliation(s)
- Adel Wassef
- University of Texas Health Sciences Center, Room 2C-07, Houston-Harris County Psychiatric Center, 2800 South MacGregor Way, Houston, TX 77021, USA.
| | | | | |
Collapse
|
27
|
Bonavita C, Ferrero A, Cereseto M, Velardez M, Rubio M, Wikinski S. Adaptive changes in the rat hippocampal glutamatergic neurotransmission are observed during long-term treatment with lorazepam. Psychopharmacology (Berl) 2003; 166:163-7. [PMID: 12545333 DOI: 10.1007/s00213-002-1373-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2002] [Accepted: 11/21/2002] [Indexed: 11/30/2022]
Abstract
RATIONALE Chronic treatment with benzodiazepines induces tolerance to most of their pharmacological effects. The best-studied neurochemical correlation to this phenomenon involves GABAergic adaptive changes. However, some compensation by excitatory neurotransmission could also be postulated. OBJECTIVE The aim of this work was to investigate the effect of chronic treatment with benzodiazepines on several parameters of hippocampal glutamatergic neurotransmission. METHODS Rats were injected (IP) with a single dose or daily doses (21 days) of 1 mg/kg lorazepam (LZ) or vehicle. Thirty minutes after the last dose, animals were killed and parameters were measured in the dissected hippocampi. We determined one presynaptic parameter, in vitro glutamate release induced by a 60 mM K(+) stimulus. [(3)H]MK-801 binding to postsynaptic NMDA receptors and the NMDA-stimulated efflux of cGMP were also evaluated. RESULTS While no changes were observed in any of the parameters after a single dose of the drug, we found an increase of 206% in in vitro glutamate release in chronically treated animals [two-way ANOVA: F(1,16)=6.22], together with an increment of 103% in the NMDA-stimulated cGMP efflux [two-way ANOVA: F(1,18)=14.05]. No changes either in K(D) or in B(max) values for [(3)H]MK-801 binding to hippocampal membranes were observed. CONCLUSIONS Taken together, these changes strongly suggest that a compensatory increase in the glutamatergic response develops in the hippocampus during chronic treatment with LZ. Our findings might indicate a contribution of glutamatergic mechanisms to the tolerance to hippocampal-mediated effects of LZ, such as amnesic and anticonvulsant activities.
Collapse
Affiliation(s)
- C Bonavita
- Instituto de Investigaciones Farmacológicas (ININFA), CONICET, Junín 956 Piso 5 (CP 1113), Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
28
|
Costa E, Auta J, Grayson DR, Matsumoto K, Pappas GD, Zhang X, Guidotti A. GABAA receptors and benzodiazepines: a role for dendritic resident subunit mRNAs. Neuropharmacology 2002; 43:925-37. [PMID: 12423662 DOI: 10.1016/s0028-3908(02)00199-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
This review is designed to describe the evolution of the seminal observation made simultaneously in 1975 by Dr. W. Haefely's laboratory (Hoffman La Roche, Basel, Switzerland) and in the Laboratory of Preclinical Pharmacology (NIH, St. Elizabeths Hospital, Washington DC), that benzodiazepine action was mediated by a modulation of GABA action at GABA(A) receptors. In fact, our suggestion was that the benzodiazepine receptor was "a receptor on a receptor" and that this receptor was GABA(A). Needless to say, this suggestion created opposition, but we did not abandon the original idea, in fact, as shown in this review, there is now universal agreement with our hypothesis on the mode of action of benzodiazepines. Hence, this review deals with the allosteric modulation of GABA(A) receptors by benzodiazepines, the role of GABA(A) receptors and benzodiazepine structure diversities in this modulation, and describes the results of our attempts to establish a benzodiazepine (imidazenil) devoid of tolerance, withdrawal symptoms, and changes in the expression of GABA(A) receptor subunits during tolerance. It also deals with the idea that the synthesis of GABA(A) receptor subunits triggered by tolerance resides in dendrites and spines where mRNAs and the apparatus for this translation is located. New analytic procedures may foster progress in the understanding of tolerance to and withdrawal from benzodiazepines.
Collapse
Affiliation(s)
- E Costa
- University of Illinois at Chicago, Department of Psychiatry, Psychiatric Institute, 1601 W Taylor St, Chicago, IL 60612, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Follesa P, Cagetti E, Mancuso L, Biggio F, Manca A, Maciocco E, Massa F, Desole MS, Carta M, Busonero F, Sanna E, Biggio G. Increase in expression of the GABA(A) receptor alpha(4) subunit gene induced by withdrawal of, but not by long-term treatment with, benzodiazepine full or partial agonists. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 92:138-48. [PMID: 11483250 DOI: 10.1016/s0169-328x(01)00164-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effects of long-term exposure to, and subsequent withdrawal of, diazepam or imidazenil (full and partial agonists of the benzodiazepine receptor, respectively) on the abundance of GABA(A) receptor subunit mRNAs and peptides were investigated in rat cerebellar granule cells in culture. Exposure of cells to 10 microM diazepam for 5 days significantly reduced the amounts of alpha(1) and gamma(2) subunit mRNAs, and had no effect on the amount of alpha(4) mRNA. These effects were accompanied by a decrease in the levels of alpha(1) and gamma(2) protein and by a reduction in the efficacy of diazepam with regard to potentiation of GABA-evoked Cl- current. Similar long-term treatment with 10 microM imidazenil significantly reduced the abundance of only the gamma(2)S subunit mRNA and had no effect on GABA(A) receptor function. Withdrawal of diazepam or imidazenil induced a marked increase in the amount of alpha(4) mRNA; withdrawal of imidazenil also reduced the amounts of alpha(1) and gamma(2) mRNAs. In addition, withdrawal of diazepam or imidazenil was associated with a reduced ability of diazepam to potentiate GABA action. These data give new insights into the different molecular events related to GABA(A) receptor gene expression and function produced by chronic treatment and withdrawal of benzodiazepines with full or partial agonist properties.
Collapse
Affiliation(s)
- P Follesa
- Department of Experimental Biology Bernardo Loddo, CNR, Center of Neuropharmacology, University of Cagliari, 09123 Cagliari, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Izzo E, Auta J, Impagnatiello F, Pesold C, Guidotti A, Costa E. Glutamic acid decarboxylase and glutamate receptor changes during tolerance and dependence to benzodiazepines. Proc Natl Acad Sci U S A 2001; 98:3483-8. [PMID: 11248104 PMCID: PMC30679 DOI: 10.1073/pnas.051628698] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Protracted administration of diazepam elicits tolerance, whereas discontinuation of treatment results in signs of dependence. Tolerance to the anticonvulsant action of diazepam is present in an early phase (6, 24, and 36 h) but disappears in a late phase (72-96 h) of withdrawal. In contrast, signs of dependence such as decrease in open-arm entries on an elevated plus-maze and increased susceptibility to pentylenetetrazol-induced seizures were apparent 96 h (but not 12, 24, or 48 h) after diazepam withdrawal. During the first 72 h of withdrawal, tolerance is associated with changes in the expression of GABA(A) (gamma-aminobutyric acid type A) receptor subunits (decrease in gamma(2) and alpha(1); increase in alpha(5)) and with an increase of mRNA expression of the most abundant form of glutamic acid decarboxylase (GAD), GAD(67). In contrast, dl-alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor GluR1 subunit mRNA and cognate protein, which are normal during the early phase of diazepam withdrawal, increase by approximately 30% in cortex and hippocampus in association with the appearance of signs of dependence 96 h after diazepam withdrawal. Immunohistochemical studies of GluR1 subunit expression with gold-immunolabeling technique reveal that the increase of GluR1 subunit protein is localized to layer V pyramidal neurons and their apical dendrites in the cortex, and to pyramidal neurons and in their dendritic fields in hippocampus. The results suggest an involvement of GABA-mediated processes in the development and maintenance of tolerance to diazepam, whereas excitatory amino acid-related processes (presumably via AMPA receptors) may be involved in the expression of signs of dependence after withdrawal.
Collapse
Affiliation(s)
- E Izzo
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
31
|
Suzuki T, Abe S, Yamaguchi M, Baba A, Hori T, Shiraishi H, Ito T. Effects of cocaine administration on receptor binding and subunits mRNA of GABA(A)-benzodiazepine receptor complexes. Synapse 2000; 38:198-215. [PMID: 11018794 DOI: 10.1002/1098-2396(200011)38:2<198::aid-syn11>3.0.co;2-k] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The effects of intermittent intraperitoneal (i.p.) administration of cocaine (20 mg/kg) on GABA(A)-benzodiazepine (BZD) receptors labeled by t-[(35)S]butylbicyclophosphorothionate (TBPS), and on several types of mRNA subunits were investigated in rat brain by in vitro quantitative receptor autoradiography and in situ hybridization. Phosphor screen imaging with high sensitivity and a wide linear range of response was utilized for imaging analysis. There was a significant decrease in the level of alpha 1, alpha 6, beta 2, beta 3, and gamma 2 subunits mRNA, with no alteration of [(35)S]TBPS binding in any regions in the brain of rats at 1 h following a single injection of cocaine. In chronically treated animals, the mean scores of stereotyped behavior were increased with the number of injections. The level of beta 3 subunit mRNA was decreased in the cortices and caudate putamen, at 24 h after a final injection of chronic administrations for 14 days. In the withdrawal from cocaine, the frontal cortex and hippocampal complexes showed a significant increase in [(35)S]TBPS binding and alpha1 and beta 3 subunit mRNA in the rats 1 week after a cessation of chronic administration of cocaine. These findings suggest that the disruption of GABA(A)-BZD receptor formation is closely involved in the development of cocaine-related behavioral disturbances. Further studies on the physiological functions on GABA(A)-BZD receptor complex will be necessary for an explanation of the precise mechanisms underlying the acute effects, development of hypersensitization, and withdrawal state of cocaine.
Collapse
Affiliation(s)
- T Suzuki
- Department of Psychiatry, Institute of Clinical Medicine, University of Tsukuba, Tennodai, Tsukuba, Japan.
| | | | | | | | | | | | | |
Collapse
|
32
|
Auta J, Guidotti A, Costa E. Imidazenil prevention of alprazolam-induced acquisition deficit in patas monkeys is devoid of tolerance. Proc Natl Acad Sci U S A 2000; 97:2314-9. [PMID: 10696114 PMCID: PMC15798 DOI: 10.1073/pnas.97.5.2314] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The partial allosteric modulators (PAMs) of gamma-aminobutyric acid-gated Cl(-) current intensities at gamma-aminobutyric acid type A receptors have high affinity but low intrinsic efficacy on benzodiazepine recognition sites. Unlike the full allosteric modulators (FAM), like alprazolam, triazolam, and diazepam, PAMs are virtually devoid of unwanted side effects, including tolerance. Imidazenil (IMD) is a PAM that elicits potent anxiolytic and anticonvulsant actions in rodents and nonhuman primates and retains its anticonvulsant and anxiolytic effects, even in rodents that are tolerant to FAMs. IMD antagonizes the side effects of FAMs in rodents and nonhuman primates. Using patas monkeys and a multiple schedule with repeated acquisition and performance of chain responses, we report that IMD administration for 17 days antagonized without showing tolerance ALP-induced disruption of acquisition.
Collapse
Affiliation(s)
- J Auta
- Department of Psychiatry, The Psychiatric Institute, University of Illinois, M/C 912, 1601 West Taylor Street, Chicago, IL 60612, USA
| | | | | |
Collapse
|
33
|
Abstract
Between 1987 and 1989, the different protein subunits that make up the receptor for the inhibitory neurotransmitter gamma-aminobutyric acid (GABA) were identified. These make up the alpha, beta, gamma and delta families, for each of which exist several subtypes. This receptor is the molecular target of modern hypnotic drugs (i.e. benzodiazepines, zopiclone, zolpidem and zaleplon). In the 10 years that have followed this milestone, significant progress has been made in exploring the molecular mechanisms of hypnotic drug action. Receptor subtype specificity of hypnotics has been explained in terms of differential affinity for receptors containing different alpha subunits, which are expressed in different brain regions. Zolpidem and zaleplon bind preferentially to alpha1-containing receptors, whereas benzodiazepines and zopiclone are aspecific. Different sets of subunits are encoded in contiguous 'cassettes' on the genome, and the transcription of each set appears to be regulated coherently. The predominant GABA(A) receptor composition found in the brain is alpha1beta2gamma2, which are all encoded on human chromosome 5. Targeted gene disruption has provided clues to the physiological functions served by GABA(A) receptors containing different subunits. Receptors containing gamma2 appear to have a vital role in maintaining appropriate central inhibition, beta3-containing receptors may also be important determinants of excitability in certain brain regions, whereas a clear role for alpha5-, alpha6- and gamma3-containing receptors has not yet been established by these techniques. Site-directed mutagenesis has indicated that benzodiazepines bind to a cleft on the GABA(A) receptor surface at the interface between the alpha and gamma subunits. Other drugs (flumazenil, zopiclone, zolpidem) also bind to the a subunit, but interact with amino acids in different binding domains to the benzodiazepines. The molecular mechanism of hypnotic dependence has been explored, and seems to involve downregulation of transcription of the normally prevalent alpha1, beta2 and gamma2 subunits, and the reciprocal upregulation of the expression of rarer subunits. Chronic treatment with hypnotic drugs that may have less dependence potential, such as zopiclone and zolpidem, appears to produce more limited change in GABA(A) receptor subunit expression. These ideas will be important both for designing new hypnotic drugs with a better safety/efficacy profile, and for evaluating more appropriate ways of using the drugs available today.
Collapse
Affiliation(s)
- A Doble
- Neuroscience and Endocrinology Department, Rhône-Poulenc Rorer SA, Antony, France.
| |
Collapse
|
34
|
Fujita M, Woods SW, Verhoeff NP, Abi-Dargham A, Baldwin RM, Zoghbi SS, Soares JC, Jatlow PA, Krystal JH, Rajeevan N, Charney DS, Seibyl JP, Innis RB. Changes of benzodiazepine receptors during chronic benzodiazepine administration in humans. Eur J Pharmacol 1999; 368:161-72. [PMID: 10193652 DOI: 10.1016/s0014-2999(99)00013-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Changes of central type GABA(A)/benzodiazepine receptors during 24-day per-oral administration of alprazolam (2 mg/day) were measured with single photon emission computed tomography (SPECT) in nine healthy human subjects. Receptor densities were measured on days -4 (baseline), 3, 10, 17 and 24. Comparison of baseline and day 3 SPECT images was used to assess receptor occupancy; comparisons of the four scans on medication were used to assess alterations in receptor levels. Clinical effects were evaluated by subjective ratings of mood and the Hopkins verbal learning test. Alprazolam induced sedation associated with a 16% receptor occupancy. Unoccupied receptor levels decreased 10% from day 3 to day 10 but then normalized to baseline values by day 17. Clinical effects showed corresponding changes 1-2 weeks after the changes in the receptor. Thus, the decrease of benzodiazepine receptor densities may be one of the major mechanisms for tolerance development in humans.
Collapse
Affiliation(s)
- M Fujita
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT 06516, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Transmitter receptor diversity often indicates differences in transmitter receptor transduction mechanisms. This is not the case for gamma-aminobutyric acid subtype A (GABAA) receptor subtypes despite the presence of 16 genes to encode the 5 families of native GABAA receptor subtypes. Similar considerations apply to GABAC receptors and GABAB receptors. Both GABAA and GABAB receptors cause hyperpolarization of neuronal membranes and inhibition of neuronal excitability, but their mechanisms differ. GABAB receptors involve an efflux of K+ rather than an influx of Cl-, as in the case of GABAA and GABAC receptors. The stimulation of GABAA receptors can sometimes cause depolarization by Cl- efflux; this efflux is not the result of a transduction mechanism modification, but of Cl(-)-concentration gradient modification. Presumably, GABAA receptor diversity is directly linked to the inhibitory activity of basket cells and other interneuron axons, each innervating several postsynaptic neurons (cortical and hippocampal pyramidal cells for instance). Since the role of this inhibition is to entrain hippocampal and cortical pyramidal neurons into columnary activity, the GABAA receptor diversification may be a mechanism expressed by these postsynaptic neuron populations that uses different GABA potencies to synchronize pyramidal neurons into columnary activity. Thus, GABA potency variability, which emerges from GABAA receptor diversity, plays a unifying role in the intrinsic functional mechanism of laminated structures. GABAA receptor structural differences also play a role in diazepam tolerance, which is a mechanism operative in neuronal circuit adaptation to the extreme amplification of GABA-gated Cl- current intensities. Partial agonists (such as imidazenil), which modestly amplify GABA action at many GABAA receptor subtypes, fail to cause tolerance, dependence, ataxia, or ethanol and barbiturate potentiation. Partial agonists might become a new class of anxiolytic and anticonvulsant drugs that are virtually devoid of the side effects that cause serious concerns in the clinical use of full allosteric positive modulators of GABA action, such as diazepam, alprazolam, triazolam, and others. None of the above can be used as anticonvulsants because of an extremely high tolerance liability. When there is tolerance to diazepam, signs of sensitization to proconvulsive action are exhibited simultaneously. After tolerance, associated changes in GABAA recepter subtype expression are virtually reversed in 72 h. Also, 96 h after termination of long-term diazepam treatment, rats exhibit anxiety and are more sensitive to kainic acid-elicited convulsions. At the same time, these rats have an increase in brain expression of GLuR1, R2, and R3. It is believed that the supersensitivity to kainic acid, convulsions and anxiety, and the increased expression of GLuR1, R2, and R3 may be parts of the mechanism of diazepam dependence.
Collapse
Affiliation(s)
- E Costa
- Department of Psychiatry, University of Illinois at Chicago 60612, USA.
| |
Collapse
|
36
|
Pesold C, Caruncho HJ, Impagnatiello F, Berg MJ, Fritschy JM, Guidotti A, Costa E. Tolerance to diazepam and changes in GABA(A) receptor subunit expression in rat neocortical areas. Neuroscience 1997; 79:477-87. [PMID: 9200730 DOI: 10.1016/s0306-4522(96)00609-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Long-term treatment with diazepam, a full allosteric modulator of the GABA(A) receptor, results in tolerance to its anticonvulsant effects, whereas an equipotent treatment with the partial allosteric modulator imidazenil does not produce tolerance. Use of subunit-specific antibodies linked to gold particles allowed an immunocytochemical estimation of the expression density of the alpha1, alpha2, alpha3, alpha5, gamma(2L&S) and beta(2/3) subunits of the GABA(A) receptor in the frontoparietal motor and frontoparietal somatosensory cortices of rats that received long-term treatment with vehicle, diazepam (three times daily for 14 days, doses increasing from 17.6 to 70.4 micromol/kg), or imidazenil (three times daily for 14 days, doses increasing from 2.5 to 10.0 micromol/kg). In this study, tolerance to diazepam was associated with a selective decrease (37%) in the expression of the alpha1 subunit in layers III-IV of the frontoparietal motor cortex, and a concomitant increase in the expression of the alpha5 (150%), gamma(2L&S) and beta(2/3) subunits (48%); an increase in alpha5 subunits was measured in all cortical layers. In the frontoparietal somatosensory cortex, diazepam-tolerant rats had a 221% increase in the expression of alpha5 subunits in all cortical layers, as well as a 35% increase in the expression of alpha3 subunits restricted to layers V-VI. Western blot analysis substantiated that these diazepam-induced changes reflected the expression of full subunit molecules. Rats that received equipotent treatment with imidazenil did not become tolerant to its anticonvulsant properties, and did not show significant changes in the expression of any of the GABA(A) receptor subunits studied, with the exception of a small decrease in alpha2 subunits in cortical layers V-VI of the frontoparietal somatosensory cortex. The results of this study suggest that tolerance to benzodiazepines may be associated with select changes in subunit abundance, leading to the expression of different GABA(A) receptor subtypes in specific brain areas. These changes might be mediated by a unique homeostatic mechanism regulating the expression of GABA(A) receptor subtypes that maintain specific functional features of GABAergic function in cortical cell layers.
Collapse
Affiliation(s)
- C Pesold
- University of Illinois at Chicago, The Psychiatric Institute, 60612, U.S.A
| | | | | | | | | | | | | |
Collapse
|