1
|
Solecki WB, Kielbinski M, Wilczkowski M, Zajda K, Karwowska K, Joanna B, Rajfur Z, Przewłocki R. Regulation of cocaine seeking behavior by locus coeruleus noradrenergic activity in the ventral tegmental area is time- and contingency-dependent. Front Neurosci 2022; 16:967969. [PMID: 35992934 PMCID: PMC9388848 DOI: 10.3389/fnins.2022.967969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Substance use disorder is linked to impairments in the ventral tegmental area (VTA) dopamine (DA) reward system. Noradrenergic (NA) inputs from locus coeruleus (LC) into VTA have been shown to modulate VTA neuronal activity, and are implicated in psychostimulant effects. Phasic LC activity controls time- and context-sensitive processes: decision making, cognitive flexibility, motivation and attention. However, it is not yet known how such temporally-distinct LC activity contributes to cocaine seeking. In a previous study we demonstrated that pharmacological inhibition of NA signaling in VTA specifically attenuates cocaine-seeking. Here, we used virally-delivered opsins to target LC neurons for inhibition or excitation, delivered onto afferents in VTA of male rats seeking cocaine under extinction conditions. Optogenetic stimulation or inhibition was delivered in distinct conditions: upon active lever press, contingently with discreet cues; or non-contingently, i.e., throughout the cocaine seeking session. Non-contingent inhibition of LC noradrenergic terminals in VTA attenuated cocaine seeking under extinction conditions. In contrast, contingent inhibition increased, while contingent stimulation reduced cocaine seeking. These findings were specific for cocaine, but not natural reward (food) seeking. Our results show that NA release in VTA drives behavior depending on timing and contingency between stimuli – context, discreet conditioned cues and reinforcer availability. We show that, depending on those factors, noradrenergic signaling in VTA has opposing roles, either driving CS-induced drug seeking, or contributing to behavioral flexibility and thus extinction.
Collapse
Affiliation(s)
- Wojciech B. Solecki
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
- *Correspondence: Wojciech B. Solecki,
| | - Michał Kielbinski
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Michał Wilczkowski
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
- Department of Brain Biochemistry, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Katarzyna Zajda
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Karolina Karwowska
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Bernacka Joanna
- Laboratory of Pharmacology and Brain Biostructure, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Zenon Rajfur
- Department of Biosystems Physics, Institute of Physics, Jagiellonian University, Kraków, Poland
| | - Ryszard Przewłocki
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
2
|
Carnovale C, Perrotta C, Baldelli S, Cattaneo D, Montrasio C, Barbieri SS, Pompilio G, Vantaggiato C, Clementi E, Pozzi M. Antihypertensive drugs and brain function: mechanisms underlying therapeutically beneficial and harmful neuropsychiatric effects. Cardiovasc Res 2022; 119:647-667. [PMID: 35895876 PMCID: PMC10153433 DOI: 10.1093/cvr/cvac110] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 11/14/2022] Open
Abstract
A bidirectional relationship exists between hypertension and psychiatric disorders, including unipolar and bipolar depression, anxiety, post-traumatic stress disorder (PTSD), psychosis, schizophrenia, mania, and dementia/cognitive decline. Repurposing of antihypertensive drugs to treat mental disorders is thus being explored. A systematic knowledge of the mechanisms of action and clinical consequences of the use of antihypertensive agents on neuropsychiatric functions has not been achieved yet. In this article, we review the putative role of antihypertensive agents in psychiatric disorders, discuss the targets and mechanisms of action, and examine how and to what extent specific drug classes/molecules may trigger, worsen, or mitigate psychiatric symptoms. In addition, we review pharmacokinetics (brain penetration of drugs) and pharmacogenetics data that add important information to assess risks and benefits of antihypertensive drugs in neuropsychiatric settings. The scientific literature shows robust evidence of a positive effect of α1 blockers on PTSD symptoms, nightmares and sleep quality, α2 agonists on core symptoms, executive function and quality of life in Attention-Deficit/Hyperactivity Disorder, PTSD, Tourette's syndrome, and β blockers on anxiety, aggression, working memory, and social communication. Renin-angiotensin system modulators exert protective effects on cognition, depression, and anxiety, and the loop diuretic bumetanide reduced the core symptoms of autism in a subset of patients. There is no evidence of clear benefits of calcium channel blockers in mood disorders in the scientific literature. These findings are mainly from preclinical studies; clinical data are still insufficient or of anecdotal nature, and seldom systematic. The information herewith provided can support a better therapeutic approach to hypertension, tailored to patients with, or with high susceptibility to, psychiatric illness. It may prompt clinical studies exploring the potential benefit of antihypertensive drugs in selected patients with neuropsychiatric comorbidities that include outcomes of neuropsychiatric interest and specifically assess undesirable effects or interactions.
Collapse
Affiliation(s)
- Carla Carnovale
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, 20157 Milano, Italy
| | - Cristiana Perrotta
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, 20157 Milano, Italy
| | - Sara Baldelli
- Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, 20157 Milano, Italy
| | - Dario Cattaneo
- Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, 20157 Milano, Italy
| | - Cristina Montrasio
- Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, 20157 Milano, Italy
| | - Silvia S Barbieri
- Unit of Brain-Heart axis: cellular and molecular mechanisms - Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine - Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
| | | | - Emilio Clementi
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, 20157 Milano, Italy.,Scientific Institute IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Marco Pozzi
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| |
Collapse
|
3
|
Tijani AO, Garg J, Frempong D, Verana G, Kaur J, Joga R, Sabanis CD, Kumar S, Kumar N, Puri A. Sustained drug delivery strategies for treatment of common substance use disorders: Promises and challenges. J Control Release 2022; 348:970-1003. [PMID: 35752256 DOI: 10.1016/j.jconrel.2022.06.034] [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: 04/06/2022] [Revised: 06/16/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
Substance use disorders (SUDs) are a leading cause of death and other ill health effects in the United States and other countries in the world. Several approaches ranging from detoxification, behavioral therapy, and the use of antagonists or drugs with counter effects are currently being applied for its management. Amongst these, drug therapy is the mainstay for some drug abuse incidences, as is in place specifically for opioid abuse or alcohol dependence. The severity of the havocs observed with the SUDs has triggered constant interest in the discovery and development of novel medications as well as suitable or most appropriate methods for the delivery of these agents. The chronic need of such drugs in users warrants the need for their prolonged or sustained systemic availability. Further, the need to improve patient tolerance to medication, limit invasive drug use and overall treatment outcome are pertinent considerations for embracing sustained release designs for medications used in managing SUDs. This review aims to provide an overview on up-to-date advances made with regards to sustained delivery systems for the drugs for treatment of different types of SUDs such as opioid, alcohol, tobacco, cocaine, and cannabis use disorders. The clinical relevance, promises and the limitations of deployed sustained release approaches along with future opportunities are discussed.
Collapse
Affiliation(s)
- Akeemat O Tijani
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614, USA.
| | - Jivesh Garg
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Dorcas Frempong
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614, USA.
| | - Gabrielle Verana
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614, USA.
| | - Jagroop Kaur
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614, USA.
| | - Ramesh Joga
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, Telangana, India.
| | - Chetan D Sabanis
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, Telangana, India.
| | - Sandeep Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, Telangana, India.
| | - Neeraj Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, Telangana, India.
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614, USA.
| |
Collapse
|
4
|
Hadizadeh H, Flores J, Nunes E, Mayerson T, Potenza MN, Angarita GA. Novel Pharmacological Agents for the Treatment of Cocaine Use Disorder. Curr Behav Neurosci Rep 2022. [DOI: 10.1007/s40473-022-00246-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
5
|
Eddie D, Bates ME, Buckman JF. Closing the brain-heart loop: Towards more holistic models of addiction and addiction recovery. Addict Biol 2022; 27:e12958. [PMID: 32783345 PMCID: PMC7878572 DOI: 10.1111/adb.12958] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/09/2020] [Accepted: 07/26/2020] [Indexed: 01/03/2023]
Abstract
Much research seeks to articulate the brain structures and pathways implicated in addiction and addiction recovery. Prominent neurobiological models emphasize the interplay between cortical and limbic brain regions as a main driver of addictive processes, but largely do not take into consideration sensory and visceral information streams that link context and state to the brain and behavior. Yet these brain-body information streams would seem to be necessary elements of a comprehensive model of addiction. As a starting point, we describe the overlap between one current model of addiction circuitry and the neural network that not only regulates cardiovascular system activity but also receives feedback from peripheral cardiovascular processes through the baroreflex loop. We highlight the need for neurobiological, molecular, and behavioral studies of neural and peripheral cardiovascular signal integration during the experience of internal states and environmental contexts that drive alcohol and other drug use behaviors. We end with a call for systematic, mechanistic research on the promising, yet largely unexamined benefits to addiction treatment of neuroscience-informed, adjunctive interventions that target the malleability of the cardiovascular system to alter brain processes.
Collapse
Affiliation(s)
- David Eddie
- Recovery Research Institute, Center for Addiction Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA,Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Marsha E. Bates
- Department of Kinesiology and Health, Rutgers University, New Brunswick, New Jersey, USA,Center of Alcohol and Substance Use Studies, Rutgers University, New Brunswick, New Jersey, USA
| | - Jennifer F. Buckman
- Department of Kinesiology and Health, Rutgers University, New Brunswick, New Jersey, USA,Center of Alcohol and Substance Use Studies, Rutgers University, New Brunswick, New Jersey, USA
| |
Collapse
|
6
|
Solecki WB, Kielbinski M, Bernacka J, Gralec K, Klasa A, Pradel K, Rojek-Sito K, Przewłocki R. Alpha 1-adrenergic receptor blockade in the ventral tegmental area attenuates acquisition of cocaine-induced pavlovian associative learning. Front Behav Neurosci 2022; 16:969104. [PMID: 35990723 PMCID: PMC9386374 DOI: 10.3389/fnbeh.2022.969104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/18/2022] [Indexed: 11/15/2022] Open
Abstract
Activity of the alpha1-adrenergic receptor (α1-AR) in the ventral tegmental area (VTA) modulates dopaminergic activity, implying its modulatory role in the behavioral functions of the dopamine (DA) system. Indeed, intra-VTA α1-AR blockade attenuates conditioned stimulus dependent behaviors such as drug seeking responses signifying a role of the noradrenergic signaling in the VTA in conditioned behaviors. Importantly, the role of the VTA α1-AR activity in Pavlovian associative learning with positive outcomes remains unknown. Here, we aimed to examine how intra-VTA α1-AR blockade affects acquisition of cocaine-induced Pavlovian associative learning in the conditioned place preference (CPP) paradigm. The impact of α1-AR blockade on cocaine-reinforced operant responding and cocaine-evoked ultrasonic vocalizations (USVs) was also studied. In addition, both α1-AR immunoreactivity in the VTA and its role in phasic DA release in the nucleus accumbens (NAc) were assessed. We demonstrated cellular localization of α1-AR expression in the VTA, providing a neuroanatomical substrate for the α1-AR mechanism. We showed that prazosin (α1-AR selective antagonist; 1 μg/0.5 μl) microinfusion attenuated electrically evoked DA transients in the NAc and dose-dependently (0.1-1 μg/0.5 μl) prevented the acquisition of cocaine CPP but did not affect cocaine-reinforced operant responding nor cocaine-induced positive affective state (measured as USVs). We propose that the VTA α1-AR signaling is necessary for the acquisition of Pavlovian associative learning but does not encode hedonic value. Thus, α1-AR signaling in the VTA might underlie salience encoding of environmental stimuli and reflect an ability of alerting/orienting functions, originating from bottom-up information processing to guide behaviors.
Collapse
Affiliation(s)
- Wojciech B Solecki
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Michał Kielbinski
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Joanna Bernacka
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland.,Laboratory of Pharmacology and Brain Biostructure, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Katarzyna Gralec
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Adam Klasa
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Karolina Rojek-Sito
- Department of Behavioral Neuroscience and Drug Development, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Ryszard Przewłocki
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
7
|
The Structural Determinants for α 1-Adrenergic/Serotonin Receptors Activity among Phenylpiperazine-Hydantoin Derivatives. Molecules 2021; 26:molecules26227025. [PMID: 34834117 PMCID: PMC8623851 DOI: 10.3390/molecules26227025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Several studies confirmed the reciprocal interactions between adrenergic and serotoninergic systems and the influence of these phenomena on the pathogenesis of anxiety. Hence, searching for chemical agents with a multifunctional pharmacodynamic profile may bring highly effective therapy for CNS disorders. This study presents a deep structural insight into the hydantoin-arylpiperazine group and their serotonin/α-adrenergic activity. The newly synthesized compounds were tested in the radioligand binding assay and the intrinsic activity was evaluated for the selected derivatives. The computer-aided SAR analysis enabled us to answer questions about the influence of particular structural fragments on selective vs. multifunctional activity. As a result of the performed investigations, there were two leading structures: (a) compound 12 with multifunctional adrenergic-serotonin activity, which is a promising candidate to be an effective anxiolytic agent; (b) compound 14 with high α1A/α1D affinity and selectivity towards α1B, which is recommended due to the elimination of probable cardiotoxic effect. The structural conclusions of this work provide significant support for future lead optimization in order to achieve the desired pharmacodynamic profile in searching for new CNS-modulating agents.
Collapse
|
8
|
Substance Use and Addiction Affect More Than the Brain: the Promise of Neurocardiac Interventions. CURRENT ADDICTION REPORTS 2021; 8:431-439. [DOI: 10.1007/s40429-021-00379-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
9
|
Velasquez-Martinez MC, Santos-Vera B, Velez-Hernandez ME, Vazquez-Torres R, Jimenez-Rivera CA. Alpha-1 Adrenergic Receptors Modulate Glutamate and GABA Neurotransmission onto Ventral Tegmental Dopamine Neurons during Cocaine Sensitization. Int J Mol Sci 2020; 21:E790. [PMID: 31991781 PMCID: PMC7036981 DOI: 10.3390/ijms21030790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/25/2022] Open
Abstract
The ventral tegmental area (VTA) plays an important role in the reward and motivational processes that facilitate the development of drug addiction. Presynaptic α1-AR activation modulates glutamate and Gamma-aminobutyric acid (GABA) release. This work elucidates the role of VTA presynaptic α1-ARs and their modulation on glutamatergic and GABAergic neurotransmission during cocaine sensitization. Excitatory and inhibitory currents (EPSCs and IPSCs) measured by a whole cell voltage clamp show that α1-ARs activation increases EPSCs amplitude after 1 day of cocaine treatment but not after 5 days of cocaine injections. The absence of a pharmacological response to an α1-ARs agonist highlights the desensitization of the receptor after repeated cocaine administration. The desensitization of α1-ARs persists after a 7-day withdrawal period. In contrast, the modulation of α1-ARs on GABA neurotransmission, shown by decreases in IPSCs' amplitude, is not affected by acute or chronic cocaine injections. Taken together, these data suggest that α1-ARs may enhance DA neuronal excitability after repeated cocaine administration through the reduction of GABA inhibition onto VTA dopamine (DA) neurons even in the absence of α1-ARs' function on glutamate release and protein kinase C (PKC) activation. α1-AR modulatory changes in cocaine sensitization increase our knowledge of the role of the noradrenergic system in cocaine addiction and may provide possible avenues for therapeutics.
Collapse
Affiliation(s)
- Maria Carolina Velasquez-Martinez
- Grupo de Neurociencias y Comportamiento, Departamento de Ciencias Básicas, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga 680006, Colombia;
| | - Bermary Santos-Vera
- Department of Biology, Cayey Campus, University of Puerto Rico, Cayey, PR 00737, USA;
| | - Maria E. Velez-Hernandez
- Department of Biological and Health Sciences, Texas A&M University-Kingsville, Kingsville, TX 78363, USA;
| | - Rafael Vazquez-Torres
- Department of Physiology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00925, USA;
| | - Carlos A. Jimenez-Rivera
- Department of Physiology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00925, USA;
| |
Collapse
|
10
|
Shorter DI, Zhang X, Domingo CB, Nielsen EM, Kosten TR, Nielsen DA. Doxazosin treatment in cocaine use disorder: pharmacogenetic response based on an alpha-1 adrenoreceptor subtype D genetic variant. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2020; 46:184-193. [PMID: 31914324 DOI: 10.1080/00952990.2019.1674864] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Background: The α1 antagonist doxazosin reduces cocaine use in individuals with cocaine use disorder (CUD) through a functional polymorphism of the α1 adrenoreceptor. The regulatory role of the α1 adrenoreceptor subtype D (ADRA1D) gene polymorphism in CUD is uncharacterized.Objectives: To study how the genetic variant of ADRA1D gene (T1848A, rs2236554) may affect the treatment efficacy of doxazosin in reducing cocaine use.Methods: This 12-week pilot trial included 76 participants with CUD with ADRA1D (T1848A, rs2236554) AA (N = 40) or AT/TT genotype (N = 36). Participants were randomized to doxazosin (8 mg/day, N = 47) or placebo (N = 29), and followed with thrice weekly urine toxicology and once weekly cognitive behavioral psychotherapy.Results: The AA and the AT/TT groups had comparable baseline rates of cocaine positive urines at weeks 1-2 (~ 76%). In the placebo group, an increase of cocaine positive urines in the AT/TT group was found as compared to the AA group (24% vs. 9%). In the doxazosin group, a greater decrease in cocaine positive urines was found in the AT/TT group relative to the AA group. The difference between the doxazosin and placebo groups in cocaine negative urines became evident at weeks 5-6 and peaked at weeks 9-10 (~35% difference). The AT/TT group demonstrated a significant medication and time by medication effect (p < .001), whereas the AA group did not.Conclusion: The T-allele carriers showed a greater reduction of cocaine use after treatment with doxazosin in participants with the ADRA1D gene polymorphism (T1848A), suggesting that this SNP may serve as a pharmacogenetic marker in pharmacotherapy of CUD.
Collapse
Affiliation(s)
- Daryl I Shorter
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Xuefeng Zhang
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Coreen B Domingo
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Ellen M Nielsen
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Thomas R Kosten
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - David A Nielsen
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
11
|
Alpha1-adrenergic receptor blockade in the ventral tegmental area modulates conditional stimulus-induced cocaine seeking. Neuropharmacology 2019; 158:107680. [DOI: 10.1016/j.neuropharm.2019.107680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 05/31/2019] [Accepted: 06/20/2019] [Indexed: 11/24/2022]
|
12
|
Zhang X, Nielsen DA, Domingo CB, Shorter DI, Nielsen EM, Kosten TR. Pharmacogenetics of Dopamine β-Hydroxylase in cocaine dependence therapy with doxazosin. Addict Biol 2019; 24:531-538. [PMID: 29498170 DOI: 10.1111/adb.12611] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 10/17/2017] [Accepted: 01/20/2018] [Indexed: 02/02/2023]
Abstract
The α1 -adrenergic antagonist, doxazosin, has improved cocaine use disorder (CUD) presumably by blocking norepinephrine (NE) stimulation and reward from cocaine-induced NE increases. If the NE levels for release were lower, then doxazosin might more readily block this NE stimulation and be more effective. The NE available for release can be lower through a genetic polymorphism in dopamine β-hydroxylase (DBH) (C-1021T, rs1611115), which reduces DβH's conversion of dopamine to NE. We hypothesize that doxazosin would be more effective in CUD patients who have these genetically lower DβH levels. This 12-week, double-blind, randomized, placebo-controlled trial included 76 CUD patients: 49 with higher DβH levels from the DBH CC genotype and 27 with lower DβH levels from T-allele carriers (CT or TT). Patients were randomized to doxazosin (8 mg/day, N = 47) or placebo (N = 29) and followed with thrice weekly urine toxicology and once weekly cognitive behavioral psychotherapy. Cocaine use was reduced at a higher rate among patients in the doxazosin than in the placebo arm. We found significantly lower cocaine use rates among patients carrying the T-allele (CT/TT) than the CC genotype. The percentage of cocaine positive urines was reduced by 41 percent from baseline in the CT/TT group with low DβH and NE levels, as compared with no net reduction in the CC genotype group with normal DβH and NE levels. The DBH polymorphism appears play an important role in CUD patients' response to doxazosin treatment, supporting a pharmacogenetic association and potential application for personalized medicine.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Michael E. DeBakey Veterans Affairs Medical Center Houston TX USA
- Menninger Department of Psychiatry and Behavioral SciencesBaylor College of Medicine Houston TX USA
| | - David A. Nielsen
- Michael E. DeBakey Veterans Affairs Medical Center Houston TX USA
- Menninger Department of Psychiatry and Behavioral SciencesBaylor College of Medicine Houston TX USA
| | - Coreen B. Domingo
- Michael E. DeBakey Veterans Affairs Medical Center Houston TX USA
- Menninger Department of Psychiatry and Behavioral SciencesBaylor College of Medicine Houston TX USA
| | - Daryl I. Shorter
- Michael E. DeBakey Veterans Affairs Medical Center Houston TX USA
- Menninger Department of Psychiatry and Behavioral SciencesBaylor College of Medicine Houston TX USA
| | - Ellen M. Nielsen
- Michael E. DeBakey Veterans Affairs Medical Center Houston TX USA
- Menninger Department of Psychiatry and Behavioral SciencesBaylor College of Medicine Houston TX USA
| | - Thomas R. Kosten
- Michael E. DeBakey Veterans Affairs Medical Center Houston TX USA
- Menninger Department of Psychiatry and Behavioral SciencesBaylor College of Medicine Houston TX USA
| |
Collapse
|
13
|
Kaag AM, Reneman L, Homberg J, van den Brink W, van Wingen GA. Enhanced Amygdala-Striatal Functional Connectivity during the Processing of Cocaine Cues in Male Cocaine Users with a History of Childhood Trauma. Front Psychiatry 2018; 9:70. [PMID: 29593581 PMCID: PMC5857536 DOI: 10.3389/fpsyt.2018.00070] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 02/21/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND AIMS Childhood trauma is associated with increased levels of anxiety later in life, an increased risk for the development of substance use disorders, and neurodevelopmental abnormalities in the amygdala and frontostriatal circuitry. The aim of this study was to investigate the (neurobiological) link among childhood trauma, state anxiety, and amygdala-frontostriatal activity in response to cocaine cues in regular cocaine users. METHODS In this study, we included 59 non-treatment seeking regular cocaine users and 58 non-drug using controls. Blood oxygenation level-dependent responses were measured using functional magnetic resonance imaging while subjects performed a cue reactivity paradigm with cocaine and neutral cues. Psychophysiological interaction analyses were applied to assess functional connectivity between the amygdala and other regions in the brain. Self-report questionnaires were used to measure childhood trauma, state anxiety, drug use, drug use severity, and craving. RESULTS Neural activation was increased during the presentation of cocaine cues, in a widespread network including the frontostriatal circuit and amygdala in cocaine users but not in controls. Functional coupling between the amygdala and medial prefrontal cortex was reduced in response to cocaine cues, in both cocaine users and controls, which was further diminished with increasing state anxiety. Importantly, amygdala-striatal connectivity was positively associated with childhood trauma in regular cocaine users, while there was a negative association in controls. At the behavioral level, state anxiety was positively associated with cocaine use severity and craving related to negative reinforcement. CONCLUSION Childhood trauma is associated with enhanced amygdala-striatal connectivity during cocaine cue reactivity in regular cocaine users, which may contribute to increased habit behavior and poorer cognitive control. While we cannot draw conclusions on causality, this study provides novel information on how childhood trauma may contribute to the development and persistence of cocaine use disorder.
Collapse
Affiliation(s)
- Anne Marije Kaag
- Department of Developmental Psychology, University of Amsterdam, Amsterdam, Netherlands
- Departement of Psychiatry, Academic Medical Centre, Amsterdam, Netherlands
- Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, Netherlands
| | - Liesbeth Reneman
- Departement of Radiology and Nuclear Medicine, Academic Medical Centre, Amsterdam, Netherlands
| | - Judith Homberg
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University, Medical Centre, Nijmegen, Netherlands
| | - Wim van den Brink
- Departement of Psychiatry, Academic Medical Centre, Amsterdam, Netherlands
| | - Guido A. van Wingen
- Departement of Psychiatry, Academic Medical Centre, Amsterdam, Netherlands
- Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
14
|
Solecki WB, Szklarczyk K, Pradel K, Kwiatkowska K, Dobrzański G, Przewłocki R. Noradrenergic signaling in the VTA modulates cocaine craving. Addict Biol 2018. [PMID: 28635140 DOI: 10.1111/adb.12514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Exposure to drug-associated cues evokes drug-seeking behavior and is regarded as a major cause of relapse. Conditional stimulus upregulates noradrenaline (NA) system activity, but the drug-seeking behavior depends particularly on phasic dopamine signaling downstream from the ventral tegmental area (VTA). The VTA dopamine-ergic activity is regulated via the signaling of alpha1 -adrenergic and alpha2 -adrenergic receptors (α1 -ARs and α2 -ARs); thus, the impact of the conditional stimulus on drug-seeking behavior might involve NAergic signaling in the VTA. To date, the role of VTA ARs in regulating cocaine seeking was not studied. We found that cocaine seeking under extinction conditions in male Sprague-Dawley rats was attenuated by intra-VTA prazosin or terazosin-two selective α1 -AR antagonists. In contrast, cocaine seeking was facilitated by intra-VTA administration of the selective α1 -AR agonist phenylephrine as well as α2 -AR antagonist RX 821002, whereas the selective β-AR antagonist propranolol had no effects. In addition, blockade of α1 -AR in the VTA prevented α2 -AR antagonist-induced enhancement of cocaine seeking. Importantly, the potential non-specific effects of the VTA AR blockade on cocaine seeking could be excluded, because none of the AR antagonists influenced sucrose seeking under extinction conditions or locomotor activity in the open field test. These results demonstrate that NAergic signaling potently and selectively regulates cocaine seeking during early cocaine withdrawal via VTA α1 -AR and α2 -AR but not β-AR. Our findings provide new insight into the NAergic mechanisms that underlie cocaine craving.
Collapse
Affiliation(s)
- Wojciech Barnaba Solecki
- Department of Neurobiology and Neuropsychology; Institute of Applied Psychology, Jagiellonian University; Poland
- Department of Molecular Neuropharmacology; Institute of Pharmacology, Polish Academy of Sciences; Poland
| | - Klaudia Szklarczyk
- Department of Neurobiology and Neuropsychology; Institute of Applied Psychology, Jagiellonian University; Poland
| | - Kamil Pradel
- Department of Molecular Neuropharmacology; Institute of Pharmacology, Polish Academy of Sciences; Poland
| | - Krystyna Kwiatkowska
- Department of Molecular Neuropharmacology; Institute of Pharmacology, Polish Academy of Sciences; Poland
| | - Grzegorz Dobrzański
- Department of Molecular Neuropharmacology; Institute of Pharmacology, Polish Academy of Sciences; Poland
| | - Ryszard Przewłocki
- Department of Molecular Neuropharmacology; Institute of Pharmacology, Polish Academy of Sciences; Poland
| |
Collapse
|
15
|
The α-1 adrenoceptor (ADRA1A) genotype moderates the magnitude of acute cocaine-induced subjective effects in cocaine-dependent individuals. Pharmacogenet Genomics 2017; 26:428-35. [PMID: 27379509 DOI: 10.1097/fpc.0000000000000234] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES We examined whether a functional variant of the ADRA1A gene moderated cocaine-induced subjective effects in a group of cocaine-dependent individuals. METHODS This study was a within-participant, double-blind, placebo-controlled inpatient human laboratory evaluation of 65 nontreatment-seeking, cocaine-dependent [Diagnostic and Statistical Manual of Mental Disorders, 4th ed. (DSM-IV)] individuals aged 18-55 years. Participants received both placebo (saline, IV) and cocaine (40 mg, IV), and subjective responses were assessed 15 min before receiving an infusion and at 5 min intervals for the subsequent 20 min. The rs1048101 variant of the α1A-adrenoceptor (ADRA1A) gene was genotyped and it was evaluated whether the Cys to Arg substitution at codon 347 in exon 2 (Cys347Arg) moderated the magnitude of the subjective effects produced by cocaine. RESULTS Thirty (46%) participants were found to have the major allele CC genotype and 35 (44%) carried at least one minor T-allele of rs1048101 (TT or TC genotype). Individuals with the CC genotype showed greater responses for 'desire' (P<0.0001), 'high' (P<0.0001), 'any drug effect' (P<0.0001), 'like cocaine' (P<0.0001), and 'likely to use cocaine if given access' (P<0.05) with experiment-wise significance. CONCLUSION This study indicates that the ADRA1A genotype could be used to identify individuals for whom acute cocaine exposure may be more rewarding and by inference may result in greater difficulty in establishing and/or maintaining abstinence from cocaine.
Collapse
|
16
|
Synergistic interactions between mirtazapine and prazosin prevent the induction and expression of behavioral sensitization to cocaine in rats. Physiol Behav 2017; 180:137-145. [DOI: 10.1016/j.physbeh.2017.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/21/2017] [Accepted: 08/23/2017] [Indexed: 11/22/2022]
|
17
|
Mahoney JJ, Kalechstein AD, Newton TF, De La Garza R. The limited impact that cocaine use patterns have on neurocognitive functioning in individuals with cocaine use disorder. J Psychopharmacol 2017; 31:989-995. [PMID: 28695777 DOI: 10.1177/0269881117715606] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
It is well-documented in the literature that cocaine use is associated with neurocognitive impairment. The manner in which patterns of cocaine use, such as years of use, recent use over the past month, and daily amount of cocaine use, moderate neurocognition has been studied in a relatively piecemeal manner. Hence, the purpose of the study was to evaluate whether cocaine use patterns modulate neurocognition in individuals with cocaine use disorder. Cocaine users who were cocaine-negative ( n=125) were divided into tertiles based on cocaine use patterns and the performances of the highest and lowest groups were compared on the following cognitive measures: Continuous Performance Task-II, n-back, and Hopkins Verbal Learning Task-Revised. Participants with cocaine use disorder who used for more years (25.2±0.6 versus 10.1±0.6 years; mean±standard error of the mean) and who had more recent cocaine use over the past month (26.3±0.5 versus 6.0±0.6 days) did not differ significantly on any of the neurocognitive variables when compared to those with use patterns of shorter duration and less frequency (all p's >0.05). Lastly, participants reporting the greatest amount daily cocaine use (1.8±0.0 g) demonstrated better performance on an auditory working memory task when compared to those with the lowest daily use (0.7±0.0 g; p=0.04). While one might expect that individuals who used greater amounts of cocaine over longer periods of time would demonstrate relatively poorer performance on measures of neurocognition, particularly in the initial phase of abstinence, our findings did not confirm this. While speculative, a potential explanation for these findings is that after an individual uses cocaine for a certain number of years, or uses a specific amount over time, then the deleterious effects of cocaine on neurocognition stabilizes, and increased duration of cocaine use does not further exacerbate those impairments.
Collapse
Affiliation(s)
- James J Mahoney
- 1 Department of Behavioral Medicine and Psychiatry, West Virginia University School of Medicine, Morgantown, USA
| | - Ari D Kalechstein
- 2 Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, USA
| | - Thomas F Newton
- 2 Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, USA
| | - Richard De La Garza
- 2 Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, USA
| |
Collapse
|
18
|
Verplaetse TL, Weinberger AH, Oberleitner LM, Smith KM, Pittman BP, Shi JM, Tetrault JM, Lavery ME, Picciotto MR, McKee SA. Effect of doxazosin on stress reactivity and the ability to resist smoking. J Psychopharmacol 2017; 31:830-840. [PMID: 28440105 PMCID: PMC5823502 DOI: 10.1177/0269881117699603] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Preclinical findings support a role for α1-adrenergic antagonists in reducing nicotine-motivated behaviors, but these findings have yet to be translated to humans. The current study evaluated whether doxazosin would attenuate stress-precipitated smoking in the human laboratory. Using a well-validated laboratory analogue of smoking-lapse behavior, this pilot study evaluated whether doxazosin (4 and 8 mg/day) versus placebo attenuated the effect of stress (vs neutral imagery) on tobacco craving, the ability to resist smoking and subsequent ad-libitum smoking in nicotine-deprived smokers ( n=35). Cortisol, adrenocorticotropin, norepinephrine, epinephrine, and physiologic reactivity were assessed. Doxazosin (4 and 8 mg/day vs placebo) decreased cigarettes per day during the 21-day titration period. Following titration, doxazosin (4 and 8 mg/day vs placebo) decreased tobacco craving. During the laboratory session, doxazosin (8 mg/day vs placebo) further decreased tobacco craving following stress versus neutral imagery. Doxazosin increased the latency to start smoking following stress, and reduced the number of cigarettes smoked. Dosage of 8 mg/day doxazosin increased or normalized cortisol levels following stress imagery and decreased cortisol levels following neutral imagery. These preliminary findings support a role for the noradrenergic system in stress-precipitated smoking behavior, and support further development of doxazosin as a novel pharmacotherapeutic treatment strategy for smoking cessation.
Collapse
Affiliation(s)
| | - Andrea H. Weinberger
- Ferkauf Graduate School of Psychology, Yeshiva University, Bronx, NY
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY
| | | | - Kathryn M.Z. Smith
- Division on Substance Abuse, Department of Psychiatry, Columbia University Medical Center/New York State Psychiatric Institute, New York, NY
| | - Brian P. Pittman
- Departments of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Julia M. Shi
- Internal Medicine, Yale University School of Medicine, New Haven, CT
| | | | - Meaghan E. Lavery
- Departments of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Marina R. Picciotto
- Departments of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Sherry A. McKee
- Departments of Psychiatry, Yale University School of Medicine, New Haven, CT
- Correspondence to: Sherry A. McKee, PhD, 2 Church St South, Suite 109, Yale University School of Medicine, New Haven, CT 06519; Tele: 203.737-3529 Fax: 203.737-4243
| |
Collapse
|
19
|
Schmidt KT, Schroeder JP, Foster SL, Squires K, Smith BM, Pitts EG, Epstein MP, Weinshenker D. Norepinephrine regulates cocaine-primed reinstatement via α1-adrenergic receptors in the medial prefrontal cortex. Neuropharmacology 2017; 119:134-140. [PMID: 28392265 PMCID: PMC5495469 DOI: 10.1016/j.neuropharm.2017.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 03/15/2017] [Accepted: 04/05/2017] [Indexed: 01/02/2023]
Abstract
Drug-primed reinstatement of cocaine seeking in rats is thought to reflect relapse-like behavior and is mediated by the integration of signals from mesocorticolimbic dopaminergic projections and corticostriatal glutamatergic innervation. Cocaine-primed reinstatement can also be attenuated by systemic administration of dopamine β-hydroxylase (DBH) inhibitors, which prevent norepinephrine (NE) synthesis, or by α1-adrenergic receptor (α1AR) antagonists, indicating functional modulation by the noradrenergic system. In the present study, we sought to further discern the role of NE in cocaine-seeking behavior by determining whether α1AR activation can induce reinstatement on its own or is sufficient to permit cocaine-primed reinstatement in the absence of all other AR signaling, and identifying the neuroanatomical substrate within the mesocorticolimbic reward system harboring the critical α1ARs. We found that while intracerebroventricular infusion of the α1AR agonist phenylephrine did not induce reinstatement on its own, it did overcome the blockade of cocaine-primed reinstatement by the DBH inhibitor nepicastat. Furthermore, administration of the α1AR antagonist terazosin in the medial prefrontal cortex (mPFC), but not the ventral tegmental area (VTA) or nucleus accumbens (NAc) shell, attenuated cocaine-primed reinstatement. Combined, these data indicate that α1AR activation in the mPFC is required for cocaine-primed reinstatement, and suggest that α1AR antagonists merit further investigation as pharmacotherapies for cocaine dependence.
Collapse
Affiliation(s)
- Karl T Schmidt
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Jason P Schroeder
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | | | - Katherine Squires
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Brilee M Smith
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Elizabeth G Pitts
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Michael P Epstein
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
20
|
Mahoney JJ, Haile CN, De La Garza R, Thakkar H, Newton TF. Electrocardiographic characteristics in individuals with cocaine use disorder. Am J Addict 2017; 26:221-227. [DOI: 10.1111/ajad.12524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/09/2017] [Accepted: 02/12/2017] [Indexed: 11/27/2022] Open
Affiliation(s)
- James J. Mahoney
- Department of Behavioral Medicine and Psychiatry; West Virginia University School of Medicine; Morgantown West Virginia
| | - Colin N. Haile
- Menninger Department of Psychiatry and Behavioral Sciences; Baylor College of Medicine; Houston Texas
| | - Richard De La Garza
- Menninger Department of Psychiatry and Behavioral Sciences; Baylor College of Medicine; Houston Texas
| | - Harsh Thakkar
- Menninger Department of Psychiatry and Behavioral Sciences; Baylor College of Medicine; Houston Texas
| | - Thomas F. Newton
- Menninger Department of Psychiatry and Behavioral Sciences; Baylor College of Medicine; Houston Texas
| |
Collapse
|
21
|
Kenna GA, Haass-Koffler CL, Zywiak WH, Edwards SM, Brickley MB, Swift RM, Leggio L. Role of the α1 blocker doxazosin in alcoholism: a proof-of-concept randomized controlled trial. Addict Biol 2016; 21:904-14. [PMID: 26037245 DOI: 10.1111/adb.12275] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Evidence suggests that the norepinephrine system represents an important treatment target for alcohol dependence (AD) and the α1 -blocker prazosin may reduce alcohol drinking in rodents and alcoholic patients. The α1 -blocker doxazosin demonstrates a more favorable pharmacokinetic profile than prazosin, but has never been studied for AD. A double-blind placebo-controlled randomized clinical trial was conducted in AD individuals seeking outpatient treatment. Doxazosin or matched placebo was titrated to 16 mg/day (or maximum tolerable dose). Drinks per week (DPW) and heavy drinking days (HDD) per week were the primary outcomes. Family history density of alcoholism (FHDA), severity of AD and gender were a priori moderators. Forty-one AD individuals were randomized, 30 (doxazosin = 15) completed the treatment phase and 28 (doxazosin = 14) also completed the follow-up. There were no significant differences between groups on DPW and HDD per week. With FHDA as a moderator, there were significant FHDA × medication interactions for both DPW (pcorrected = 0.001, d = 1.18) and HDD (pcorrected = 0.00009, d = 1.30). Post hoc analyses revealed that doxazosin significantly reduced alcohol drinking in AD patients with high FHDA and by contrast increased drinking in those with low FHDA. Doxazosin may be effective selectively in AD patients with high FHDA. This study provides preliminary evidence for personalized medicine using α1 -blockade to treat AD. However, confirmatory studies are required.
Collapse
Affiliation(s)
- George A. Kenna
- Department of Psychiatry and Human Behavior; Center for Alcohol and Addiction Studies; Brown University; Providence RI USA
| | - Carolina L. Haass-Koffler
- Department of Behavioral and Social Sciences; Center for Alcohol and Addiction Studies; Brown University; Providence RI USA
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology; National Institute on Alcohol Abuse and Alcoholism; National Institute on Drug Abuse; National Institutes of Health; Bethesda MD USA
| | - William H. Zywiak
- Department of Psychiatry and Human Behavior; Center for Alcohol and Addiction Studies; Brown University; Providence RI USA
- Decision Sciences Institute; PIRE; Pawtucket RI USA
| | - Steven M. Edwards
- Department of Psychology; University of Nebraska-Lincoln; Lincoln NE USA
| | - Michael B. Brickley
- Department of Behavioral and Social Sciences; Center for Alcohol and Addiction Studies; Brown University; Providence RI USA
| | - Robert M. Swift
- Department of Psychiatry and Human Behavior; Center for Alcohol and Addiction Studies; Brown University; Providence RI USA
- Veterans Affairs Medical Center; Providence RI USA
| | - Lorenzo Leggio
- Department of Behavioral and Social Sciences; Center for Alcohol and Addiction Studies; Brown University; Providence RI USA
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology; National Institute on Alcohol Abuse and Alcoholism; National Institute on Drug Abuse; National Institutes of Health; Bethesda MD USA
| |
Collapse
|
22
|
Richards JR, Garber D, Laurin EG, Albertson TE, Derlet RW, Amsterdam EA, Olson KR, Ramoska EA, Lange RA. Treatment of cocaine cardiovascular toxicity: a systematic review. Clin Toxicol (Phila) 2016; 54:345-64. [PMID: 26919414 DOI: 10.3109/15563650.2016.1142090] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Cocaine abuse is a major worldwide health problem. Patients with acute cocaine toxicity presenting to the emergency department may require urgent treatment for tachycardia, dysrhythmia, hypertension, and coronary vasospasm, leading to pathological sequelae such as acute coronary syndrome, stroke, and death. OBJECTIVE The objective of this study is to review the current evidence for pharmacological treatment of cardiovascular toxicity resulting from cocaine abuse. METHODS MEDLINE, PsycINFO, Database of Abstracts of Reviews of Effects (DARE), OpenGrey, Google Scholar, and the Cochrane Library were searched from inception to November 2015. Articles on pharmacological treatment involving human subjects and cocaine were selected and reviewed. Evidence was graded using Oxford Centre for Evidence-Based Medicine guidelines. Treatment recommendations were compared to current American College of Cardiology/American Heart Association guidelines. Special attention was given to adverse drug events or treatment failure. The search resulted in 2376 articles with 120 eligible involving 2358 human subjects. Benzodiazepines and other GABA-active agents: There were five high-quality (CEBM Level I/II) studies, three retrospective (Level III), and 25 case series/reports (Level IV/V) supporting the use of benzodiazepines and other GABA-active agents in 234 subjects with eight treatment failures. Benzodiazepines may not always effectively mitigate tachycardia, hypertension, and vasospasm from cocaine toxicity. Calcium channel blockers: There were seven Level I/II, one Level III, and seven Level IV/V studies involving 107 subjects and one treatment failure. Calcium channel blockers may decrease hypertension and coronary vasospasm, but not necessarily tachycardia. Nitric oxide-mediated vasodilators: There were six Level I/II, one Level III, and 25 Level IV/V studies conducted in 246 subjects with 11 treatment failures and two adverse drug events. Nitroglycerin may lead to severe hypotension and reflex tachycardia. Alpha-adrenoceptor blocking drugs: There were two Level I studies and three case reports. Alpha-1 blockers may improve hypertension and vasospasm, but not tachycardia, although evidence is limited. Alpha-2-adrenoceptor agonists: There were two high-quality studies and one case report detailing the successful use of dexmedetomidine. Beta-blockers and β/α-blockers: There were nine Level I/II, seven Level III, and 34 Level IV/V studies of β-blockers, with 1744 subjects, seven adverse drug events, and three treatment failures. No adverse events were reported for use of combined β/α-blockers such as labetalol and carvedilol, which were effective in attenuating both hypertension and tachycardia. Antipsychotics: Seven Level I/II studies, three Level III studies, and seven Level IV/V case series and reports involving 168 subjects have been published. Antipsychotics may improve agitation and psychosis, but with inconsistent reduction in tachycardia and hypertension and risk of extrapyramidal adverse effects. Other agents: There was only one high level study of morphine, which reversed cocaine-induced coronary vasoconstriction but increased heart rate. Other agents reviewed included lidocaine, sodium bicarbonate, amiodarone, procainamide, propofol, intravenous lipid emulsion, propofol, and ketamine. CONCLUSIONS High-quality evidence for pharmacological treatment of cocaine cardiovascular toxicity is limited but can guide acute management of associated tachycardia, dysrhythmia, hypertension, and coronary vasospasm. Future randomized prospective trials are needed to evaluate new agents and further define optimal treatment of cocaine-toxic patients.
Collapse
Affiliation(s)
- John R Richards
- a Department of Emergency Medicine , University of California Davis Medical Center , Sacramento , CA , USA
| | - Dariush Garber
- a Department of Emergency Medicine , University of California Davis Medical Center , Sacramento , CA , USA
| | - Erik G Laurin
- a Department of Emergency Medicine , University of California Davis Medical Center , Sacramento , CA , USA
| | - Timothy E Albertson
- b Department of Internal Medicine, Divisions of Toxicology, Pulmonary and Critical Care , University of California Davis Medical Center , Sacramento , CA , USA
| | - Robert W Derlet
- a Department of Emergency Medicine , University of California Davis Medical Center , Sacramento , CA , USA
| | - Ezra A Amsterdam
- c Department of Internal Medicine, Division of Cardiology , University of California Davis Medical Center , Sacramento , CA , USA
| | - Kent R Olson
- d Departments of Medicine and Clinical Pharmacy , University of California, San Francisco, Medical Director, California Poison Control System, San Francisco Division , San Francisco , CA , USA
| | - Edward A Ramoska
- e Department of Emergency Medicine , Drexel University , Philadelphia , PA , USA
| | - Richard A Lange
- f Department of Internal Medicine, Division of Cardiology , Texas Tech University Health Sciences Center , El Paso , TX , USA
| |
Collapse
|
23
|
The Roles of Dopamine and α1-Adrenergic Receptors in Cocaine Preferences in Female and Male Rats. Neuropsychopharmacology 2015; 40:2696-704. [PMID: 25900120 PMCID: PMC4864645 DOI: 10.1038/npp.2015.116] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/13/2015] [Accepted: 04/16/2015] [Indexed: 01/16/2023]
Abstract
Cocaine dependence is characterized by compulsive drug taking and reduced involvement in social, occupational, or recreational activities. Unraveling the diverse mechanisms contributing to the loss-of-interest in these 'non-drug' pursuits is essential for understanding the neurobiology of addiction and could provide additional targets for treating addiction. The study objectives were to examine changes in cocaine-induced dopamine (DA) overflow in the nucleus accumbens (NAc) over the course of self-administration and determine the roles of α1- and β-adrenergic receptors (AR) in the loss-of-interest in food rewards following the development of an addicted phenotype in male and female rats. Subjects were given access to cocaine and palatable food pellets in a choice self-administration paradigm to identify 'addicted' cocaine-preferring (CP) individuals and resistant pellet-preferring (PP) individuals based on their patterns of self-administration over 7 weeks. Cocaine-induced DA overflow in the NAc was examined with microdialysis early and late during self-administration (weeks 2 and 7). Subjects were treated in counter-balanced order with propranolol (β-AR antagonist), terazosin (α1-AR antagonist), or vehicle for an additional 3 weeks of self-administration. CP rats displayed increased motivation for cocaine and attenuated motivation for pellets following the development of cocaine preferences. In females, the estrous cycle affected pellet, but not cocaine, self-administration. CP rats displayed attenuated cocaine-induced DA overflow in the NAc. Propranolol enhanced cocaine reinforcement and reduced pellet intake, whereas terazosin enhanced motivation for pellets and reversed preferences in a subset of CP rats. The implications of these results for the treatment of addiction are discussed.
Collapse
|
24
|
Dela Peña I, Gevorkiana R, Shi WX. Psychostimulants affect dopamine transmission through both dopamine transporter-dependent and independent mechanisms. Eur J Pharmacol 2015; 764:562-570. [PMID: 26209364 PMCID: PMC4600454 DOI: 10.1016/j.ejphar.2015.07.044] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 07/15/2015] [Accepted: 07/20/2015] [Indexed: 12/14/2022]
Abstract
The precise mechanisms by which cocaine and amphetamine-like psychostimulants exert their reinforcing effects are not yet fully defined. It is widely believed, however, that these drugs produce their effects by enhancing dopamine neurotransmission in the brain, especially in limbic areas such as the nucleus accumbens, by inducing dopamine transporter-mediated reverse transport and/or blocking dopamine reuptake though the dopamine transporter. Here, we present the evidence that aside from dopamine transporter, non-dopamine transporter-mediated mechanisms also participate in psychostimulant-induced dopamine release and contribute to the behavioral effects of these drugs, such as locomotor activation and reward. Accordingly, psychostimulants could increase norepinephrine release in the prefrontal cortex, the latter then alters the firing pattern of dopamine neurons resulting in changes in action potential-dependent dopamine release. These alterations would further affect the temporal pattern of dopamine release in the nucleus accumbens, thereby modifying information processing in that area. Hence, a synaptic input to a nucleus accumbens neuron may be enhanced or inhibited by dopamine depending on its temporal relationship to dopamine release. Specific temporal patterns of dopamine release may also be required for certain forms of synaptic plasticity in the nucleus accumbens. Together, these effects induced by psychostimulants, mediated through a non-dopamine transporter-mediated mechanism involving norepinephrine and the prefrontal cortex, may also contribute importantly to the reinforcing properties of these drugs.
Collapse
Affiliation(s)
- Ike Dela Peña
- Departments of Pharmaceutical and Administrative Sciences, Loma Linda University Schools of Pharmacy and Medicine, Loma Linda, CA 92350, USA.
| | - Ruzanna Gevorkiana
- Departments of Pharmaceutical and Administrative Sciences, Loma Linda University Schools of Pharmacy and Medicine, Loma Linda, CA 92350, USA
| | - Wei-Xing Shi
- Departments of Pharmaceutical and Administrative Sciences, Loma Linda University Schools of Pharmacy and Medicine, Loma Linda, CA 92350, USA; Basic Sciences, Loma Linda University Schools of Pharmacy and Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
25
|
Evaluation of the dopamine β-hydroxylase (DβH) inhibitor nepicastat in participants who meet criteria for cocaine use disorder. Prog Neuropsychopharmacol Biol Psychiatry 2015; 59:40-48. [PMID: 25602710 PMCID: PMC4777897 DOI: 10.1016/j.pnpbp.2015.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/15/2015] [Accepted: 01/15/2015] [Indexed: 11/22/2022]
Abstract
In the present study, we tested the hypothesis that the potent and selective dopamine-β-hydroxylase (DβH) inhibitor nepicastat would have minimal effects on cardiovascular and pharmacokinetic parameters associated with cocaine administration and would reduce the positive subjective effects produced by cocaine. We conducted a double-blind, placebo-controlled, inpatient study of oral nepicastat (0, 80 and 160mg) concurrent with intravenous (IV) cocaine (0, 10, 20 and 40mg) in non-treatment seeking participants who metcriteria for cocaine use disorder. Safety analyses revealed that nepicastat was well-tolerated and there were no differences in adverse events observed after nepicastat plus cocaine vs. cocaine alone. In addition, the pharmacokinetic properties of cocaine administration were not altered by nepicastat treatment. Cocaine-induced cardiovascular and subjective effects were evaluated for completers in the cohort randomized to nepicastat (n=13) using a within-subjects statistical analysis strategy. Specifically, the cardiovascular and subjective effects of cocaine were assessed in the presence of placebo (0mg), 80mg of nepicastat or 160mg of nepicastat on study Days 4, 8 and 12, respectively. Analyses revealed a main effect of nepicastat to reduce several cocaine-induced positive subjective effects. Taken together, these data indicate that nepicastat is safe when co-administered with cocaine and may suppress its positive subjective effects, and may be viable as a pharmacotherapy for treatment of cocaine use disorder.
Collapse
|
26
|
Verrico CD, Haile CN, Mahoney JJ, Thompson-Lake DGY, Newton TF, De La Garza R. Treatment with modafinil and escitalopram, alone and in combination, on cocaine-induced effects: a randomized, double blind, placebo-controlled human laboratory study. Drug Alcohol Depend 2014; 141:72-8. [PMID: 24928479 PMCID: PMC4120836 DOI: 10.1016/j.drugalcdep.2014.05.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 05/08/2014] [Accepted: 05/08/2014] [Indexed: 02/01/2023]
Abstract
BACKGROUND Concurrent administration of dopamine and serotonin reuptake inhibitors reduces cocaine self-administration in monkeys. Consonant with this, clinical trials assessing modafinil and selective serotonin reuptake inhibitors alone show some efficacy as potential pharmacotherapies for cocaine dependence. We hypothesized that combining modafinil with escitalopram would attenuate the euphoric effects of cocaine to a greater degree than modafinil alone. METHODS In a randomized, double blind, parallel groups design participants received either placebo (0mg/day; n=16), modafinil (200mg/day; n=16), escitalopram (20mg/day; n=17), or modafinil+escitalopram (200+20mg/day; n=15) for 5 days. On day 5, during separate sessions participants received an intravenous sample of cocaine (0 or 20mg; randomized) and five $1 bills. Participants rated the subjective effects of the infusions and subsequently made choices to either return $1 and receive another infusion or keep $1 and receive no infusion. RESULTS Compared to saline, cocaine (20mg) significantly (p≤0.008) increased most ratings, including "good effects", "stimulated", and "high". Relative to placebo, modafinil significantly (p≤0.007) attenuated subject-rated increases of "any drug effect", "high", "good effects", and "stimulated" produced by cocaine. Compared to saline, participants chose cocaine infusions significantly more; however, no treatment significantly reduced choices for cocaine infusions. Escitalopram did not enhance the efficacy of modafinil to reduce any measure. CONCLUSIONS Modafinil attenuated many positive subjective effects produced by cocaine; however, escitalopram combined with modafinil did not enhance the efficacy of modafinil to reduce cocaine effects.
Collapse
Affiliation(s)
- Christopher D. Verrico
- Menninger Department of Psychiatry and Behavioral Sciences,Department of Pharmacology,Corresponding Author: Christopher D. Verrico, Baylor College of Medicine, 1977 Butler Blvd., Suite E4.163, Houston, Texas 77030, , Phone: (713) 791-1414 x26849
| | - Colin N. Haile
- Menninger Department of Psychiatry and Behavioral Sciences
| | | | | | - Thomas F. Newton
- Menninger Department of Psychiatry and Behavioral Sciences,Department of Pharmacology
| | - Richard De La Garza
- Menninger Department of Psychiatry and Behavioral Sciences,Department of Pharmacology,Department of Neuroscience
| |
Collapse
|
27
|
Kim JH, Lawrence AJ. Drugs currently in Phase II clinical trials for cocaine addiction. Expert Opin Investig Drugs 2014; 23:1105-22. [PMID: 24773297 DOI: 10.1517/13543784.2014.915312] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION There are no FDA-approved pharmacotherapies for treating cocaine addiction; thus, developing drugs to treat cocaine dependence is an unmet critical need. Fortunately, there are a number of drugs that are currently in Phase II clinical trial/s. This is due in part to the advances from in vivo imaging in humans which provided a roadmap of the neurochemistry of the cocaine-dependent brain. Most drugs currently in Phase II clinical trials attempt to modulate the disturbed neurochemistry in cocaine dependents to resemble those of healthy individuals. These predominantly modulate dopamine, serotonin, glutamate, GABA or noradrenaline signalling. AREAS COVERED This review summarizes the therapeutic potential of each drug as evidenced by clinical and preclinical studies. It also discusses their utility in terms of bioavailability and half-life. EXPERT OPINION Amphetamine salts and topiramate clearly stand out in terms of their potential efficacy in treating cocaine addiction. The efficacy of topiramate was closely associated with regular cognitive-behavioural therapy (CBT), which highlights the importance of a combined effort to promote abstinence and enhance retention via CBT. Cognitive/psychological screening appears necessary for a more symptom-based approach with more reasonable outcomes other than abstinence (e.g., improved quality of life) in treating cocaine addiction.
Collapse
Affiliation(s)
- Jee Hyun Kim
- The Florey Institute of Neuroscience and Mental Health, Behavioural Neuroscience Division , Parkville, VIC 3052 , Australia
| | | |
Collapse
|
28
|
Psychostimulant addiction treatment. Neuropharmacology 2014; 87:150-60. [PMID: 24727297 DOI: 10.1016/j.neuropharm.2014.04.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/14/2014] [Accepted: 04/01/2014] [Indexed: 01/29/2023]
Abstract
Treatment of psychostimulant addiction has been a major, and not fully met, challenge. For opioid addiction, there is strong evidence for the effectiveness of several medications. For psychostimulants, there is no corresponding form of agonist maintenance that has met criteria for regulatory approval or generally accepted use. Stimulant-use disorders remain prevalent and can result in both short-term and long-term adverse consequences. The mainstay of treatment remains behavioral interventions. In this paper, we discuss those interventions and some promising candidates in the search for pharmacological interventions. This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
|
29
|
De La Garza R, Galloway GP, Newton T, Mendelson J, Haile C, Dib E, Hawkins RY, Chen CYA, Mahoney J, Mojsiak J, Lao G, Anderson A, Kahn R. Assessment of safety, cardiovascular and subjective effects after intravenous cocaine and lofexidine. Prog Neuropsychopharmacol Biol Psychiatry 2014; 50:44-52. [PMID: 24316175 PMCID: PMC4562471 DOI: 10.1016/j.pnpbp.2013.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 11/19/2013] [Accepted: 11/25/2013] [Indexed: 01/05/2023]
Abstract
The primary objective of this study was to determine the safety of lofexidine, an α2 receptor agonist, alone and concurrent with cocaine in non-treatment seeking cocaine-dependent or cocaine-abusing participants. After screening, eligible participants received double-blind, randomized infusions of saline and 20mg of cocaine on Day 1, and saline and 40mg of cocaine on Day 2. Subjects were randomized and started receiving daily administration of placebo (N=4) or lofexidine on Day 3 and continued on this schedule until Day 7. Two dosing regimens for lofexedine were investigated: 0.8 QID (N=3) and 0.2mg QID (N=11). On Days 6 and 7, subjects received double-blind infusions of saline and 20mg of cocaine on Day 6, and saline and 40mg of cocaine on Day 7. The data reveal a notable incidence of hemodynamic-related AEs over the course of the study. Two of the three participants at the 0.8mg dose level discontinued, and five of 11 participants at the 0.2mg dose level were withdrawn (or voluntarily discontinued) after hemodynamic AEs. Subjective effects and cardiovascular data were derived from all participants who were eligible to receive infusions (i.e., did not meet stopping criteria) on Days 6 and 7 (6 received lofexidine 0.2mg, QID and 4 received placebo, QID). As expected, cocaine significantly increased heart rate and blood pressure, as well as several positive subjective effects. There was a trend for lofexidine to decrease cocaine-induced cardiovascular changes and cocaine-induced ratings for "any drug effect", "good effects", and "desire cocaine", but sample size issues limit the conclusions that can be drawn. Despite the trends to reduce cocaine-induced subjective effects, cardiovascular AEs may limit future utility of lofexidine as a treatment for this population.
Collapse
Affiliation(s)
- R. De La Garza
- Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, Houston, TX
| | - G. P. Galloway
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - T.F. Newton
- Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, Houston, TX
| | - J. Mendelson
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - C.N. Haile
- Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, Houston, TX
| | - E. Dib
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - R. Y. Hawkins
- Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, Houston, TX
| | - C-Y A. Chen
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - J.J. Mahoney
- Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, Houston, TX
| | - J. Mojsiak
- National Institute on Drug Abuse, NIH, Bethesda, MD
| | - G. Lao
- National Institute on Drug Abuse, NIH, Bethesda, MD
| | - A. Anderson
- National Institute on Drug Abuse, NIH, Bethesda, MD
| | - R. Kahn
- National Institute on Drug Abuse, NIH, Bethesda, MD
| |
Collapse
|
30
|
Schmidt KT, Weinshenker D. Adrenaline rush: the role of adrenergic receptors in stimulant-induced behaviors. Mol Pharmacol 2014; 85:640-50. [PMID: 24499709 DOI: 10.1124/mol.113.090118] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Psychostimulants, such as cocaine and amphetamines, act primarily through the monoamine neurotransmitters dopamine (DA), norepinephrine, and serotonin. Although stimulant addiction research has largely focused on DA, medication development efforts targeting the dopaminergic system have thus far been unsuccessful, leading to alternative strategies aimed at abating stimulant abuse. Noradrenergic compounds have shown promise in altering the behavioral effects of stimulants in rodents, nonhuman primates, and humans. In this review, we discuss the contribution of each adrenergic receptor (AR) subtype (α1, α2, and β) to five stimulant-induced behaviors relevant to addiction: locomotor activity, conditioned place preference, anxiety, discrimination, and self-administration. AR manipulation has diverse effects on these behaviors; each subtype profoundly influences outcomes in some paradigms but is inconsequential in others. The functional neuroanatomy and intracellular signaling mechanisms underlying the impact of AR activation/blockade on these behaviors remain largely unknown, presenting a new frontier for research on psychostimulant-AR interactions.
Collapse
Affiliation(s)
- Karl T Schmidt
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia
| | | |
Collapse
|
31
|
Haile CN, De La Garza R, Mahoney JJ, Newton TF. Effects of methamphetamine on the noradrenergic activity biomarker salivary alpha-amylase. Drug Alcohol Depend 2013; 133:759-62. [PMID: 23968815 PMCID: PMC4432865 DOI: 10.1016/j.drugalcdep.2013.07.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/26/2013] [Accepted: 07/28/2013] [Indexed: 11/20/2022]
Abstract
BACKGROUND Methamphetamine (METH) potently activates the sympathetic nervous system (SNS) by increasing central and peripheral norepinephrine (NE). Salivary α-amylase (sAA) is a biomarker of SNS activation that correlates with plasma NE levels. The purpose of this study was to determine the impact of METH on sAA activity and whether changes in sAA activity were correlated with subjective effects ratings. METHODS Non-treatment seeking METH-dependent volunteers (N=8) participated in this within-subjects laboratory-based study. Volunteers received randomly administered intravenous METH (0mg, 30 mg) and sAA activity, cardiovascular measures and subjective ratings were assessed at baseline (-15 min) and five post-METH time points (10, 20, 30, 45, and 60 min). RESULTS METH (30 mg) increased sAA activity over time. sAA activity significantly correlated with diastolic blood pressure following 0mg METH and systolic blood pressure following 30 mg METH. Subjective ratings (ANY EFFECT, HIGH, GOOD, STIMULATED, LIKE, WLLING TO PAY) highly correlated with sAA over five post-METH time points (N=40; r's=0.543-0.684, p's<0.001). Age, body mass index and METH amount received on a mg/kg basis were significantly associated with sAA activity. Multiple linear regression analysis indicated sAA activity remained a significant predictor of subjective ratings following METH after controlling for these factors. CONCLUSIONS The NE peripheral biomarker sAA activity is associated with METH's subjective effects.
Collapse
Affiliation(s)
- Colin N Haile
- Baylor College of Medicine, Menninger Department of Psychiatry & Behavioral Sciences, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|
32
|
Abstract
Stimulant-related disorders (SRD) continue to be an important public health problem for which there are presently no approved pharmacotherapies. Although behavioral interventions provide some benefit response varies. The development of novel and effective pharmacotherapies continues to be a research priority. Understanding neural mechanisms critical to the action of stimulants has helped reveal several potential pharmacotherapies that have already shown promise in controlled clinical trials. Common to some of these medications is the ability to reverse neural deficits in individuals with SRD. Results from thoroughly conducted clinical trials continue to broaden our knowledge increasing the possibility of soon developing effective pharmacotherapies for SRD.
Collapse
|
33
|
α₁-Adrenergic receptors contribute to the acute effects of 3,4-methylenedioxymethamphetamine in humans. J Clin Psychopharmacol 2013; 33:658-66. [PMID: 23857311 DOI: 10.1097/jcp.0b013e3182979d32] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Preclinical studies implicate a role for α₁-noradrenergic receptors in the effects of psychostimulants, including 3,4-methylenedioxymethamphetamine (MDMA, "ecstasy"). The present study evaluated the effects of the α₁-noradrenergic receptor antagonist doxazosin on the acute pharmacodynamic and pharmacokinetic response to MDMA in 16 healthy subjects. Doxazosin (8 mg/d) or placebo was administered for 3 days before MDMA (125 mg) or placebo using a randomized, double-blind, placebo-controlled, 4-session, crossover design. Doxazosin reduced MDMA-induced elevations in blood pressure, body temperature, and moderately attenuated positive mood but enhanced tachycardia associated with MDMA. The results indicate that α₁-adrenergic receptors contribute to the acute cardiostimulant and to a minor extent possibly also to the thermogenic and euphoric effects of MDMA in humans.
Collapse
|
34
|
Verrico CD, Haile CN, Newton TF, Kosten TR, De La Garza R, De La Garza R. Pharmacotherapeutics for substance-use disorders: a focus on dopaminergic medications. Expert Opin Investig Drugs 2013; 22:1549-68. [PMID: 24033127 DOI: 10.1517/13543784.2013.836488] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Illicit substance-use is a substantial public health concern, contributing over $150 billion in costs annually to Americans. A complex disease, a substance-use disorder affects neural circuits involved in reinforcement, motivation, learning and memory, and inhibitory control. AREAS COVERED The modulatory influence of dopamine in mesocorticolimbic circuits contributes to encoding the primary reinforcing effects of substances and numerous studies suggest that aberrant signaling within these circuits contributes to the development of a substance-use disorder in some individuals. Decades of research focused on the clinical development of medications that directly target dopamine receptors has led to recent studies of agonist-like dopaminergic treatments for stimulant-use disorders and, more recently, cannabis-use disorder. Human studies evaluating the efficacy of dopaminergic agonist-like medications to reduce reinforcing effects and substance-use provide some insight into the design of future pharmacotherapy trials. A search of PubMed using specific brain regions, medications, and/or the terms 'dopamine', 'cognition', 'reinforcement', 'cocaine', 'methamphetamine', 'amphetamine', 'cannabis', 'treatment/pharmacotherapy', 'addiction/abuse/dependence' identified articles relevant to this review. EXPERT OPINION Conceptualization of substance-use disorders and their treatment continues to evolve. Current efforts increasingly focus on a strategy fostering combination pharmacotherapies that target multiple neurotransmitter systems.
Collapse
Affiliation(s)
- Christopher D Verrico
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine , One Baylor Plaza, Houston, TX 77030-3411 , USA
| | | | | | | | | | | |
Collapse
|
35
|
Shorter D, Lindsay JA, Kosten TR. The alpha-1 adrenergic antagonist doxazosin for treatment of cocaine dependence: A pilot study. Drug Alcohol Depend 2013; 131:66-70. [PMID: 23306096 PMCID: PMC3655111 DOI: 10.1016/j.drugalcdep.2012.11.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 11/26/2012] [Accepted: 11/27/2012] [Indexed: 11/16/2022]
Abstract
BACKGROUND Medications decreasing central noradrenergic activity have been associated with attenuation of cocaine effects. AIMS This pilot study examined the efficacy of doxazosin versus placebo for reducing cocaine use in treatment-seeking cocaine dependent persons. METHODS We screened 108 cocaine dependent subjects and assigned 35 participants to receive either doxazosin (8mg/day) or placebo for 13 weeks. Participants were titrated on the study medication according to two different schedules. During the initial phase of the study, patients were titrated onto the study medication over an 8-week period (DOX-slow). After reviewing data from our human laboratory study, a second phase was initiated, wherein titration was accelerated to a 4-week period (DOX-fast). All participants received weekly cognitive behavioral therapy. Urine toxicology was performed thrice weekly. RESULTS Baseline subject characteristics were comparable. Thirty subjects entered the study: 8 subjects in DOX-slow, 9 subjects in DOX-fast, and 13 subjects in placebo. Total number of cocaine-negative urines was significantly increased in the DOX-fast group; and percentage of total cocaine-negative urines by group were 10% for DOX-slow group, 35% for DOX-fast group, and 14% for placebo (χ(2)=36.3, df=2, p<0.0001). The percentage of participants achieving two or more consecutive weeks of abstinence by group was 0% for DOX-slow group, 44% for DOX-fast group, and 7% for placebo (χ(2)=7.35, df=2, p<0.023). CONCLUSIONS This pilot study suggests the potential efficacy of doxazosin when rapidly titrated in reducing cocaine use.
Collapse
Affiliation(s)
- D Shorter
- The Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, and Michael E. DeBakey V.A. Medical Center, Houston, TX, United States.
| | | | | |
Collapse
|
36
|
Simmler LD, Buser TA, Donzelli M, Schramm Y, Dieu LH, Huwyler J, Chaboz S, Hoener MC, Liechti ME. Pharmacological characterization of designer cathinones in vitro. Br J Pharmacol 2013; 168:458-70. [PMID: 22897747 PMCID: PMC3572571 DOI: 10.1111/j.1476-5381.2012.02145.x] [Citation(s) in RCA: 519] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 07/30/2012] [Accepted: 08/08/2012] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Designer β-keto amphetamines (e.g. cathinones, 'bath salts' and 'research chemicals') have become popular recreational drugs, but their pharmacology is poorly characterized. EXPERIMENTAL APPROACH We determined the potencies of cathinones to inhibit DA, NA and 5-HT transport into transporter-transfected HEK 293 cells, DA and 5-HT efflux from monoamine-preloaded cells, and monoamine receptor binding affinity. KEY RESULTS Mephedrone, methylone, ethylone, butylone and naphyrone acted as non-selective monoamine uptake inhibitors, similar to cocaine. Mephedrone, methylone, ethylone and butylone also induced the release of 5-HT, similar to 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) and other entactogens. Cathinone, methcathinone and flephedrone, similar to amphetamine and methamphetamine, acted as preferential DA and NA uptake inhibitors and induced the release of DA. Pyrovalerone and 3,4-methylenedioxypyrovalerone (MDPV) were highly potent and selective DA and NA transporter inhibitors but unlike amphetamines did not evoke the release of monoamines. The non-β-keto amphetamines are trace amine-associated receptor 1 ligands, whereas the cathinones are not. All the cathinones showed high blood-brain barrier permeability in an in vitro model; mephedrone and MDPV exhibited particularly high permeability. CONCLUSIONS AND IMPLICATIONS Cathinones have considerable pharmacological differences that form the basis of their suggested classification into three groups. The predominant action of all cathinones on the DA transporter is probably associated with a considerable risk of addiction.
Collapse
Affiliation(s)
- L D Simmler
- Division of Clinical Pharmacology and Toxicology, Departments of Biomedicine and Internal Medicine, University Hospital and University of Basel, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Leggio L, Kenna GA. Commentary: Doxazosin for alcoholism. Alcohol Clin Exp Res 2012; 37:191-3. [PMID: 23278505 DOI: 10.1111/acer.12064] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 11/07/2012] [Indexed: 11/26/2022]
Abstract
Recent preclinical and clinical evidence using prazosin indicates that α(1) -blockade may represent a new approach to treat alcohol dependence (AD). While most of the alcohol research on α(1) -blockade has been conducted testing prazosin, O'Neil and colleagues recently performed a set of preclinical experiments testing another α(1) -blocker, doxazosin, which has a longer half-life that may enhance clinical utility. Doxazosin and prazosin share the same chemical structure, in which the central element is a piperazine ring. O'Neil and colleagues' main results are that doxazosin significantly reduced alcohol intake without affecting locomotor activity. As such, O'Neil and colleagues provide the first preclinical evidence of the possible role of doxazosin in AD. Additional translational research is needed to further test this hypothesis.
Collapse
Affiliation(s)
- Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
38
|
The α1 Antagonist Doxazosin Alters the Behavioral Effects of Cocaine in Rats. Brain Sci 2012; 2:619-33. [PMID: 24961263 PMCID: PMC4061810 DOI: 10.3390/brainsci2040619] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 10/26/2012] [Accepted: 11/08/2012] [Indexed: 01/11/2023] Open
Abstract
Medications that target norepinephrine (NE) neurotransmission alter the behavioral effects of cocaine and may be beneficial for stimulant-use disorders. We showed previously that the short-acting, α1-adrenergic antagonist, prazosin, blocked drug-induced reinstatement of cocaine-seeking in rats and doxazosin (DOX), a longer-acting α1 antagonist blocked cocaine’s subjective effects in cocaine-dependent volunteers. To further characterize DOX as a possible pharmacotherapy for cocaine dependence, we assessed its impact on the development and expression of cocaine-induced locomotor sensitization in rats. Rats (n = 6–8) were administered saline, cocaine (COC, 10 mg/kg) or DOX (0.3 or 1.0 mg/kg) alone or in combination for 5 consecutive days (development). Following 10-days of drug withdrawal, all rats were administered COC and locomotor activity was again assessed (expression). COC increased locomotor activity across days indicative of sensitization. The high dose (1.0 mg/kg), but not the low dose (0.3 mg/kg) of DOX significantly decreased the development and expression of COC sensitization. DOX alone did not differ from saline. These results are consistent with studies showing that α1 receptors are essential for the development and expression of cocaine’s behavioral effects. Results also suggest that blockade of both the development and expression of locomotor sensitization may be important characteristics of possible pharmacotherapies for cocaine dependence in humans.
Collapse
|
39
|
Haile CN, De La Garza R, Mahoney JJ, Nielsen DA, Kosten TR, Newton TF. The impact of disulfiram treatment on the reinforcing effects of cocaine: a randomized clinical trial. PLoS One 2012; 7:e47702. [PMID: 23144826 PMCID: PMC3493584 DOI: 10.1371/journal.pone.0047702] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 09/13/2012] [Indexed: 12/17/2022] Open
Abstract
Background Clinical trials indicate that disulfiram (250 mg/d) reduces cocaine use, though one study found that treatment with lower doses of disulfiram (62.5 and 125 mg/d) increased cocaine use. We conducted the present study to better understand how disulfiram alters the reinforcing effects of cocaine in cocaine users. Methods Seventeen non-treatment seeking, cocaine-dependent volunteers participated in this double-blind, placebo-controlled, laboratory-based study. A cross-over design was utilized in which participants received placebo in one phase and disulfiram (250 mg/d) in the other. Following three days of study medication participants completed two choice sessions. In one they made 10 choices between receiving an intravenous infusion of saline or money that increased in value (US$ 0.05–16) and in the other cocaine (20 mg) or money. Results Participants chose cocaine more than saline under both disulfiram and placebo conditions (p<0.05). Unexpectedly, disulfiram increased both the number of cocaine and saline infusion choices (p<0.05). We next examined the relationship between disulfiram dose and cocaine choices. Disulfiram dose (mg/kg bodyweight) was negatively correlated with number of choices for cocaine (p<0.05). Disulfiram also enhanced cocaine-induced increases in cardiovascular measures (p's<0.05–0.01). Conclusions Disulfiram's impact on the reinforcing effects of cocaine depends on dose relative to body weight. Our results suggest that the use of weight-based medication doses would produce more reliable effects, consistent with weight-based dosing used in pediatrics and in preclinical research. Trial Registration Clinicaltrials.gov NCT00729300
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas F. Newton
- Baylor College of Medicine, Menninger Department of Psychiatry & Behavioral Sciences, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
40
|
O'Neil ML, Beckwith LE, Kincaid CL, Rasmussen DD. The α1-adrenergic receptor antagonist, doxazosin, reduces alcohol drinking in alcohol-preferring (P) Rats. Alcohol Clin Exp Res 2012; 37:202-12. [PMID: 22758213 DOI: 10.1111/j.1530-0277.2012.01884.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 05/02/2012] [Indexed: 11/28/2022]
Abstract
BACKGROUND Evidence supports a role for the noradrenergic system in alcohol drinking in animals and humans. Our previous studies demonstrated the efficacy of prazosin, an α1-adrenergic antagonist, in decreasing alcohol drinking in rat models of alcohol dependence. Prazosin has also been shown to decrease alcohol drinking in treatment-seeking alcohol-dependent men. Clinically, the use of prazosin is limited by the requirement for multiple daily administrations, whereas doxazosin, a structurally similar α1-adrenergic antagonist, requires only once-daily dosing. In this study, we tested the hypothesis that doxazosin, like prazosin, would decrease alcohol drinking in rats selectively bred for alcohol preference (P line). METHODS Adult male P rats were given 2 h/d scheduled access to a 2-bottle choice (15% v/v alcohol vs. water) session 5 d/wk (M-F), with food and water available ad libitum 24 h/d. Rats were injected with doxazosin (0 to 10 mg/kg, IP) 40 minutes prior to initiation of the alcohol access session in 3 trials (of 3, 5, and 5 consecutive days) each separated by 5 to 8 weeks. The third trial included 1 day without alcohol access (for locomotor testing), and 1 day of a single hour of alcohol access (for plasma alcohol determination). RESULTS Doxazosin significantly reduced alcohol intake in all 3 trials. The 5 mg/kg dose consistently reduced alcohol intake, increased water drinking, did not affect locomotor activity, and resulted in lower plasma alcohol concentrations, suggesting that the doxazosin-induced reduction in alcohol drinking was not dependent on a motor impairment or an alteration in alcohol clearance. CONCLUSIONS Doxazosin decreases voluntary alcohol consumption by male alcohol-preferring (P) rats, supporting a role for the noradrenergic system in alcohol drinking in P rats and suggesting that doxazosin could potentially be an effective once-daily pharmacotherapeutic agent for the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- Meghan L O'Neil
- VA Puget Sound Health Care System , Seattle, Washington; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98108, USA
| | | | | | | |
Collapse
|
41
|
Pharmacotherapeutics directed at deficiencies associated with cocaine dependence: focus on dopamine, norepinephrine and glutamate. Pharmacol Ther 2012; 134:260-77. [PMID: 22327234 DOI: 10.1016/j.pharmthera.2012.01.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 01/19/2012] [Indexed: 11/20/2022]
Abstract
Much effort has been devoted to research focused on pharmacotherapies for cocaine dependence yet there are no FDA-approved medications for this brain disease. Preclinical models have been essential to defining the central and peripheral effects produced by cocaine. Recent evidence suggests that cocaine exerts its reinforcing effects by acting on multiple neurotransmitter systems within mesocorticolimibic circuitry. Imaging studies in cocaine-dependent individuals have identified deficiencies in dopaminergic signaling primarily localized to corticolimbic areas. In addition to dysregulated striatal dopamine, norepinephrine and glutamate are also altered in cocaine dependence. In this review, we present these brain abnormalities as therapeutic targets for the treatment of cocaine dependence. We then survey promising medications that exert their therapeutic effects by presumably ameliorating these brain deficiencies. Correcting neurochemical deficits in cocaine-dependent individuals improves memory and impulse control, and reduces drug craving that may decrease cocaine use. We hypothesize that using medications aimed at reversing known neurochemical imbalances is likely to be more productive than current approaches. This view is also consistent with treatment paradigms used in neuropsychiatry and general medicine.
Collapse
|