1
|
van Staden C, Weinshenker D, Finger-Baier K, Botha TL, Brand L, Wolmarans DW. Posttraumatic anxiety-like behaviour in zebrafish is dose-dependently attenuated by the alpha-2A receptor agonist, guanfacine. Behav Pharmacol 2025; 36:47-59. [PMID: 39718044 DOI: 10.1097/fbp.0000000000000808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Traumatic stress exposure increases noradrenaline (NA) release, which contributes to anxiety and impaired risk-appraisal. Guanfacine, a selective alpha-2A adrenergic receptor agonist, has been used to treat stress-related disorders characterised by impaired prefrontal cortex function. By acting on both presynaptic inhibitory autoreceptors and postsynaptic heteroreceptors, guanfacine attenuates stress reactivity and enhances cognition. However, its effectiveness in treating trauma-related anxiety and risk-taking behaviour remains unclear. Leveraging the advantages of zebrafish (Danio rerio ) as a sensitive and efficient preclinical model which is ideal for stress research, we explored the impact of traumatic stress exposure combined with varying concentrations of guanfacine in adult zebrafish. Zebrafish were evaluated for trauma-related anxiety using both the novel tank test (NTT) and a novel version of the open-field test (nOFT), the latter which was also used to investigate risk-taking behaviour. We found that (1) traumatic stress exposure led to heightened risk-taking behaviour in the nOFT, and (2) low-to-moderate concentrations of guanfacine (3-20 µg/L) attenuated anxiety-like, but not risk-taking behaviour, with the highest concentration (40 µg/L), showing no effect. These results highlight the complex role of NA in modulating dysregulated behaviours during traumatic events and indicate the potential of guanfacine for improving trauma-related anxiety and risk-taking behaviour.
Collapse
Affiliation(s)
- Cailin van Staden
- Department of Pharmacology, Center of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Karin Finger-Baier
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Tarryn L Botha
- Department of Zoology, University of Johannesburg, Johannesburg, South Africa
| | - Linda Brand
- Department of Pharmacology, Center of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - De Wet Wolmarans
- Department of Pharmacology, Center of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
2
|
Wilson CA, Miller BW, Renton RM, Lominac KD, Szumlinski KK. Investigation into the biomolecular bases of blunted cocaine-induced glutamate release within the nucleus accumbens elicited by adolescent exposure to phenylpropanolamine. Drug Alcohol Depend 2024; 264:112465. [PMID: 39427535 DOI: 10.1016/j.drugalcdep.2024.112465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/17/2024] [Accepted: 10/03/2024] [Indexed: 10/22/2024]
Abstract
Globally, phenylpropanolamine (PPA) is a prevalent primary active ingredient in over-the-counter cough and cold, as well as weight-loss medications. Previously, we showed that a sensitization of cocaine-induced glutamate release within the nucleus accumbens (NAC) and the expression of cocaine-conditioned reward is not apparent in adult mice with a prior history of repeated PPA exposure during adolescence. As NAC glutamate is a purported driver of cocaine reward and reinforcement, the present study employed in vivo microdialysis and immunoblotting approaches to inform as to the receptor and transporter anomalies that might underpin the disrupted glutamate response to cocaine in adolescent PPA-exposed mice. For this, male and female C57BL/6J mice were pretreated, once daily, with either 0 or 40mg/kg PPA during post-natal days 35-44. Adolescent PPA pretreatment significantly altered the expression of mGlu2/3 and α2 receptors in the NAC, with less robust changes detected for EAAT2, D2 receptors, DAT and NET. However, we detected no overt change in the capacity of these receptors or transporters to affect extracellular glutamate levels in adolescent PPA-pretreated mice. The present findings contrast with the pronounced changes in the capacity of mGlu2/3 receptors, EAAT, DAT and NET to regulate NAC extracellular glutamate reported previously for juvenile PPA-pretreated mice, indicating further that the long-term biochemical consequences of PPA depend on the critical period of neurodevelopment during which an individual is PPA-exposed, although the specific biomolecular changes underpinning the cocaine phenotype produced by adolescent PPA remain to be elucidated.
Collapse
Affiliation(s)
- Casey A Wilson
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Bailey W Miller
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Rachel M Renton
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Kevin D Lominac
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA 93106, USA; Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA.
| |
Collapse
|
3
|
de Cássia Collaço R, Lammens M, Blevins C, Rodgers K, Gurau A, Yamauchi S, Kim C, Forrester J, Liu E, Ha J, Mei Y, Boehm C, Wohler E, Sobreira N, Rowe PC, Valle D, Brock MV, Bosmans F. Anxiety and dysautonomia symptoms in patients with a Na V1.7 mutation and the potential benefits of low-dose short-acting guanfacine. Clin Auton Res 2024; 34:191-201. [PMID: 38064009 PMCID: PMC11805752 DOI: 10.1007/s10286-023-01004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/15/2023] [Indexed: 03/17/2024]
Abstract
PURPOSE Guanfacine is an α2A-adrenergic receptor agonist, FDA-approved to treat attention-deficit hyperactivity disorder and high blood pressure, typically as an extended-release formulation up to 7 mg/day. In our dysautonomia clinic, we observed that off-label use of short-acting guanfacine at 1 mg/day facilitated symptom relief in two families with multiple members presenting with severe generalized anxiety. We also noted anecdotal improvements in associated dysautonomia symptoms such as hyperhidrosis, cognitive impairment, and palpitations. We postulated that a genetic deficit existed in these patients that might augment guanfacine susceptibility. METHODS We used whole-exome sequencing to identify mutations in patients with shared generalized anxiety and dysautonomia symptoms. Guanfacine-induced changes in the function of voltage-gated Na+ channels were investigated using voltage-clamp electrophysiology. RESULTS Whole-exome sequencing uncovered the p.I739V mutation in SCN9A in the proband of two nonrelated families. Moreover, guanfacine inhibited ionic currents evoked by wild-type and mutant NaV1.7 encoded by SCN9A, as well as other NaV channel subtypes to a varying degree. CONCLUSION Our study provides further evidence for a possible pathophysiological role of NaV1.7 in anxiety and dysautonomia. Combined with off-target effects on NaV channel function, daily administration of 1 mg short-acting guanfacine may be sufficient to normalize NaV channel mutation-induced changes in sympathetic activity, perhaps aided by partial inhibition of NaV1.7 or other channel subtypes. In a broader context, expanding genetic and functional data about ion channel aberrations may enable the prospect of stratifying patients in which mutation-induced increased sympathetic tone normalization by guanfacine can support treatment strategies for anxiety and dysautonomia symptoms.
Collapse
Affiliation(s)
- Rita de Cássia Collaço
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium
| | - Maxime Lammens
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium
| | - Carley Blevins
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kristen Rodgers
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Andrei Gurau
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Suguru Yamauchi
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Christine Kim
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jeannine Forrester
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Edward Liu
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jinny Ha
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yuping Mei
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Corrine Boehm
- McKusick-Nathans Department of Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Elizabeth Wohler
- McKusick-Nathans Department of Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nara Sobreira
- McKusick-Nathans Department of Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Rowe
- Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David Valle
- McKusick-Nathans Department of Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Malcolm V Brock
- Department of Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| | - Frank Bosmans
- Molecular Physiology and Neurophysics Group, Department of Basic and Applied Medical Sciences, University of Ghent, Ghent, Belgium.
| |
Collapse
|
4
|
Vines L, Sotelo D, Giddens N, Manza P, Volkow ND, Wang GJ. Neurological, Behavioral, and Pathophysiological Characterization of the Co-Occurrence of Substance Use and HIV: A Narrative Review. Brain Sci 2023; 13:1480. [PMID: 37891847 PMCID: PMC10605099 DOI: 10.3390/brainsci13101480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Combined antiretroviral therapy (cART) has greatly reduced the severity of HIV-associated neurocognitive disorders in people living with HIV (PLWH); however, PLWH are more likely than the general population to use drugs and suffer from substance use disorders (SUDs) and to exhibit risky behaviors that promote HIV transmission and other infections. Dopamine-boosting psychostimulants such as cocaine and methamphetamine are some of the most widely used substances among PLWH. Chronic use of these substances disrupts brain function, structure, and cognition. PLWH with SUD have poor health outcomes driven by complex interactions between biological, neurocognitive, and social factors. Here we review the effects of comorbid HIV and psychostimulant use disorders by discussing the distinct and common effects of HIV and chronic cocaine and methamphetamine use on behavioral and neurological impairments using evidence from rodent models of HIV-associated neurocognitive impairments (Tat or gp120 protein expression) and clinical studies. We also provide a biopsychosocial perspective by discussing behavioral impairment in differentially impacted social groups and proposing interventions at both patient and population levels.
Collapse
Affiliation(s)
- Leah Vines
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.V.); (D.S.); (P.M.); (N.D.V.)
| | - Diana Sotelo
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.V.); (D.S.); (P.M.); (N.D.V.)
| | - Natasha Giddens
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA;
| | - Peter Manza
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.V.); (D.S.); (P.M.); (N.D.V.)
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.V.); (D.S.); (P.M.); (N.D.V.)
| | - Gene-Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA; (L.V.); (D.S.); (P.M.); (N.D.V.)
| |
Collapse
|
5
|
Brown JA, Petersen N, Centanni SW, Jin AY, Yoon HJ, Cajigas SA, Bedenbaugh MN, Luchsinger JR, Patel S, Calipari ES, Simerly RB, Winder DG. An ensemble recruited by α 2a-adrenergic receptors is engaged in a stressor-specific manner in mice. Neuropsychopharmacology 2023; 48:1133-1143. [PMID: 36085168 PMCID: PMC10267140 DOI: 10.1038/s41386-022-01442-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/08/2022]
Abstract
α2a-adrenergic receptor (α2a-AR) agonists are candidate substance use disorder therapeutics due to their ability to recruit noradrenergic autoreceptors to dampen stress system engagement. However, we recently found that postsynaptic α2a-ARs are required for stress-induced reinstatement of cocaine-conditioned behavior. Understanding the ensembles recruited by these postsynaptic receptors (heteroceptors) is necessary to understand noradrenergic circuit control. We utilized a variety of approaches in FosTRAP (Targeted Recombination in Active Populations) mice to define an ensemble of cells activated by the α2a-AR partial agonist guanfacine ("Guansembles") in the bed nucleus of the stria terminalis (BST/BNST), a region key to stress-induced reinstatement of drug seeking. We define BNST "Guansembles" and show they differ from restraint stress-activated cells. Guanfacine produced inhibition of cAMP-dependent signaling in Guansembles, while chronic restraint stress increased cAMP-dependent signaling. Guanfacine both excited and inhibited aspects of Guansemble neuronal activity. Further, while some stressors produced overall reductions in Guansemble activity, active coping events during restraint stress and exposure to unexpected shocks were both associated with Guansemble recruitment. Using viral tracing, we define a BNST Guansemble afferent network that includes regions involved in the interplay of stress and homeostatic functions. Finally, we show that activation of Guansembles produces alterations in behavior on the elevated plus maze consistent with task-specific anxiety-like behavior. Overall, we define a population of BNST neurons recruited by α2a-AR signaling that opposes the behavioral action of canonical autoreceptor α2a-AR populations and which are differentially recruited by distinct stressors. Moreover, we demonstrate stressor-specific physiological responses in a specific neuronal population.
Collapse
Affiliation(s)
- Jordan A Brown
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Nicholas Petersen
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Samuel W Centanni
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Allie Y Jin
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Hye Jean Yoon
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Stephanie A Cajigas
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Michelle N Bedenbaugh
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Joseph R Luchsinger
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Sachin Patel
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Richard B Simerly
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Danny G Winder
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
6
|
Raby WN, Heller M, Milliaressis D, Choi CJ, Basaraba C, Levin FR, Church S, Pavlicova M, Nunes EV. Cocaine use disorder patients develop distinct patterns of regulation of acth secretion by a vasopressin agonist and oxytocin: Report on a laboratory study. DRUG AND ALCOHOL DEPENDENCE REPORTS 2023; 7:100158. [PMID: 37397438 PMCID: PMC10311151 DOI: 10.1016/j.dadr.2023.100158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/27/2023] [Accepted: 04/05/2023] [Indexed: 07/04/2023]
Abstract
Background : Oxytocin and Vasopressin systems in the brain sustain adaptation to stressors. Cocaine being a stressor, it may alter brain homeostatic function. This dysregulation may entrench cocaine use disorder. Method : This is a human laboratory study of the effects of intranasal desmopressin (a Vasopressin 1b receptor agonist) and oxytocin on ACTH secretion in cocaine use disorder patients versus a control group. It consisted of two endocrine challenges performed on consecutive days. On day 1, the effect of intranasal desmopressin (80 IU) on ACTH secretion was measured. On day 2, a pre-treatment with intranasal oxytocin (24 IU) preceded intranasal desmopressin to monitor its effect on desmopressin-induced ACTH secretion. We hypothesized that the effect of intranasal oxytocin in controls would differ from the effect in cocaine use disorder patients. Results : Forty-three patients were included in this study: 14 controls and 29 cocaine use disorder patients. Significant differences were noted in the direction of change of ACTH secretion between the two groups. In cocaine use disorder patients, overall ACTH secretion was on average 2.7 pg/ml/min higher after intranasal desmopressin than after intranasal oxytocin/desmopressin (t292 = 2.91, p = 0.004). The opposite was observed in controls: overall ACTH secretion averaged 3.3 pg/ml/min less after intranasal desmopressin than after intranasal oxytocin/desmopressin (t292 = -2.35, p = 0.02). Conclusion : Intranasal oxytocin and desmopressin revealed a pattern of ACTH secretion in cocaine use disorder patients that is distinct from a non-addicted control group. (ClinicalTrial.gov00255357, 10/2014).
Collapse
Affiliation(s)
- Wilfrid Noël Raby
- Division on Substance Abuse, Department of Psychiatry, Montefiore Medical Center, Albert Einstein College of Medicine, 1510 Waters Place, 2nd Floor, Bronx, NY, 10461
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, New York, NY, USA
| | - Matthew Heller
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, New York, NY, USA
| | - Demetrios Milliaressis
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, New York, NY, USA
| | - C. Jean Choi
- Division of Mental Health Data Science, New York State Psychiatric Institute, New York, NY, USA
| | - Cale Basaraba
- Division of Mental Health Data Science, New York State Psychiatric Institute, New York, NY, USA
| | - Frances R. Levin
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, New York, NY, USA
| | - Sarah Church
- Wholeview Wellness Centers, 369 Lexington Avenue, Suite 14A, New York City, NY, 10017, USA
| | - Martina Pavlicova
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Edward V. Nunes
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
7
|
Fox HC, Milivojevic V, Sinha R. Therapeutics for Substance-Using Women: The Need to Elucidate Sex-Specific Targets for Better-Tailored Treatments. Handb Exp Pharmacol 2023; 282:127-161. [PMID: 37592081 DOI: 10.1007/164_2023_687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
In the last decade, alcohol consumption in the US has risen by 84% in women compared with 35% in men. Furthermore, research has shown that sex- and gender-related differences may disadvantage women in terms of developing a range of psychological, cognitive, and medical problems considerably earlier in their drinking history than men, and despite consuming a similar quantity of substances. While this "telescoping" process has been acknowledged in the literature, a concomitant understanding of the underlying biobehavioral mechanisms, and an increase in the development of specific treatments tailored to women, has not occurred. In the current chapter we focus on understanding why the need for personalized, sex-specific medications is imperative, and highlight some of the potential sex-specific gonadal and stress-related adaptations underpinning the accelerated progress from controlled to compulsive drug and alcohol seeking in women. We additionally discuss the efficacy of these mechanisms as novel targets for medications development, using exogenous progesterone and guanfacine as examples. Finally, we assess some of the challenges faced and progress made in terms of developing innovative medications in women. We suggest that agents such as exogenous progesterone and adrenergic medications, such as guanfacine, may provide some efficacy in terms of attenuating stress-induced craving for several substances, as well as improving the ability to emotionally regulate in the face of stress, preferentially in women. However, to fully leverage the potential of these therapeutics in substance-using women, greater focus needs to the placed on reducing barriers to treatment and research by encouraging women into clinical trials.
Collapse
Affiliation(s)
- Helen C Fox
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| | - Verica Milivojevic
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Rajita Sinha
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
8
|
Voetterl H, van Wingen G, Michelini G, Griffiths KR, Gordon E, DeBeus R, Korgaonkar MS, Loo SK, Palmer D, Breteler R, Denys D, Arnold LE, du Jour P, van Ruth R, Jansen J, van Dijk H, Arns M. Brainmarker-I Differentially Predicts Remission to Various Attention-Deficit/Hyperactivity Disorder Treatments: A Discovery, Transfer, and Blinded Validation Study. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2023; 8:52-60. [PMID: 35240343 DOI: 10.1016/j.bpsc.2022.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/11/2022] [Accepted: 02/18/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Attention-deficit/hyperactivity disorder is characterized by neurobiological heterogeneity, possibly explaining why not all patients benefit from a given treatment. As a means to select the right treatment (stratification), biomarkers may aid in personalizing treatment prescription, thereby increasing remission rates. METHODS The biomarker in this study was developed in a heterogeneous clinical sample (N = 4249) and first applied to two large transfer datasets, a priori stratifying young males (<18 years) with a higher individual alpha peak frequency (iAPF) to methylphenidate (N = 336) and those with a lower iAPF to multimodal neurofeedback complemented with sleep coaching (N = 136). Blinded, out-of-sample validations were conducted in two independent samples. In addition, the association between iAPF and response to guanfacine and atomoxetine was explored. RESULTS Retrospective stratification in the transfer datasets resulted in a predicted gain in normalized remission of 17% to 30%. Blinded out-of-sample validations for methylphenidate (n = 41) and multimodal neurofeedback (n = 71) corroborated these findings, yielding a predicted gain in stratified normalized remission of 36% and 29%, respectively. CONCLUSIONS This study introduces a clinically interpretable and actionable biomarker based on the iAPF assessed during resting-state electroencephalography. Our findings suggest that acknowledging neurobiological heterogeneity can inform stratification of patients to their individual best treatment and enhance remission rates.
Collapse
Affiliation(s)
- Helena Voetterl
- Research Institute Brainclinics, Brainclinics Foundation, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Guido van Wingen
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Giorgia Michelini
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, California; Department of Biological & Experimental Psychology, Queen Mary University of London, London, United Kingdom
| | - Kristi R Griffiths
- Brain Dynamics Centre, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Roger DeBeus
- Department of Psychology, University of North Carolina at Asheville, Asheville, North Carolina
| | - Mayuresh S Korgaonkar
- Brain Dynamics Centre, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Sandra K Loo
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, California
| | | | - Rien Breteler
- Department of Clinical Psychology, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Damiaan Denys
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - L Eugene Arnold
- Department of Psychiatry & Behavioral Health, Nisonger Center, Ohio State University, Columbus, Ohio
| | | | | | - Jeanine Jansen
- Open Mind Neuroscience, Eindhoven, the Netherlands; Eindhovens Psychologisch Instituut, Eindhoven, the Netherlands
| | - Hanneke van Dijk
- Research Institute Brainclinics, Brainclinics Foundation, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Martijn Arns
- Research Institute Brainclinics, Brainclinics Foundation, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
9
|
Role of alpha-2 adrenergic and kappa opioid receptors in the effects of alcohol gavage-induced dependence on alcohol seeking. Behav Brain Res 2022; 434:114032. [DOI: 10.1016/j.bbr.2022.114032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022]
|
10
|
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
|
11
|
Noël Raby W, Heller M, Milliaressis D, Jean Choi C, Basaraba C, Pavlicova M, Alschuler DM, Levin FR, Church S, Nunes EV. Intranasal oxytocin may improve odds of abstinence in cocaine-dependent patients: results from a preliminary study. DRUG AND ALCOHOL DEPENDENCE REPORTS 2021; 2:100016. [PMID: 36845891 PMCID: PMC9948893 DOI: 10.1016/j.dadr.2021.100016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022]
Abstract
Background Oxytocin (OT) treatment in drug addiction studies have suggested potential therapeutic benefits. There is a paucity of clinical trial studies of oxytocin in cocaine use disorders. Method This was a 6-week randomized, double-blind, outpatient clinical trial study investigating the effect of daily Intranasal Oxytocin (24 IU) on cocaine use by cocaine use disorder patients. After a 7-day inpatient abstinence induction stage, patients were randomized to intranasal oxytocin or intranasal placebo. During the outpatient phase, cocaine use disorder patients were required to present themselves to the research staff 3 times a week for witnessed randomized medication administration, to provide a urine sample for qualitative toxicology, and complete mandatory assessments, including the Time-Line-Follow Back. For the interim days, patients were given an "at-home" bottle that was weighed at each clinic visit to monitor compliance. Results Neither administration of Intranasal placebo (n = 11) or Oxytocin (n = 15) induced at least 3 weeks of continuous abstinence. However, from week 3, the odds of weekly abstinence increased from 4.61 (95% CI = 1.05, 20.3) to 15.0 (CI = 1.18, 190.2) by week 6 for the Intranasal Oxytocin group (t = 2.12, p = 0.037), though there was no significant group difference overall in the odds of abstinence over time (F1,69 = 1.73, p = 0.19). More patients on Intranasal Oxytocin dropped out (p = 0.0005). Conclusions Intranasal Oxytocin increased the odds of weekly abstinence in Cocaine patients after 2 weeks compared to PBO, but was associated with a higher dropout rate. (ClinicalTrials.gov 02,255,357, 10/2014).
Collapse
Affiliation(s)
- Wilfrid Noël Raby
- Division on Substance Abuse, Department of Psychiatry, Montefiore Medical Center, Albert Einstein College of Medicine, 1510 Waters Place, 2nd Floor, Bronx, NY 10461, United States of America
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, New York, NY, United States of America
- Corresponding author at: Division on Substance Abuse, Department of Psychiatry, Montefiore Medical Center, Albert Einstein College of Medicine, 1510 Waters Place, 2nd Floor, Bronx, NY 10461, United States of America.
| | - Matthew Heller
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, New York, NY, United States of America
| | - Demetrios Milliaressis
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, New York, NY, United States of America
| | - C. Jean Choi
- Division of Mental Health Data Science, New York State Psychiatric Institute, New York, NY, United States of America
| | - Cale Basaraba
- Division of Mental Health Data Science, New York State Psychiatric Institute, New York, NY, United States of America
| | - Martina Pavlicova
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, United States of America
| | - Daniel M. Alschuler
- Division of Mental Health Data Science, New York State Psychiatric Institute, New York, NY, United States of America
| | - Frances R. Levin
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, New York, NY, United States of America
| | - Sarah Church
- Wholeview Wellness Centers, 369 Lexington Avenue, Suite 14A, New York City, NY 10017, United States of America
| | - Edward V. Nunes
- Division on Substance Use Disorders, Department of Psychiatry, Columbia University Irving Medical Center and the New York State Psychiatric Institute, New York, NY, United States of America
| |
Collapse
|
12
|
Guille C, King C, Ramakrishnan V, Baker N, Cortese B, Nunn L, Rogers T, McRae-Clark A, Brady K. The impact of lofexidine on stress-related opioid craving and relapse: Design and methodology of a randomized clinical trial. Contemp Clin Trials 2021; 111:106616. [PMID: 34737091 PMCID: PMC8761253 DOI: 10.1016/j.cct.2021.106616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/27/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022]
Abstract
Opioid Use Disorders (OUDs) and drug overdose deaths are increasing at alarmingly high rates in the United States. Stress and dysregulation in biologic stress response systems such as the hypothalamic-pituitary-adrenal axis and noradrenergic system appear to play an important role in the pathophysiology of substance use disorders and relapse to drug use, particularly for women. Alpha-2 adrenergic agonist medications effectively decrease noradrenergic activity and have demonstrated benefit in preventing relapse to substance use and decreasing stress-reactivity and craving in cocaine- and nicotine-dependent women, compared to men. Alpha-2 adrenergic agonists may help decrease stress reactivity in individuals with OUDs and prevent relapse to drug use, but gender differences have yet to be systematically explored. We describe the rationale, study design and methodology of a randomized, double-blind, placebo-controlled clinical trial examining gender differences in stress, craving and drug use among adult men and women with OUD taking methadone or buprenorphine and randomly assigned to an alpha-2 adrenergic agonist, lofexidine, compared to placebo. In addition, we describe methods for measuring daily stress, craving and drug use in participant's natural environment as well as participant's physiological (i.e., heart rate, cortisol) and psychological (i.e., stress, craving) response to laboratory social and drug cue stressors. Lastly, we detail methods adopted to sustain research activity while following guidelines for the COVID-19 pandemic. ClinicalTrials.gov Registration Number: NCT03718065.
Collapse
Affiliation(s)
- Constance Guille
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Courtney King
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Viswanathan Ramakrishnan
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Nathaniel Baker
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Bernadette Cortese
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Lisa Nunn
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Taylor Rogers
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Aimee McRae-Clark
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Kathleen Brady
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| |
Collapse
|
13
|
Venniro M, Reverte I, Ramsey LA, Papastrat KM, D'Ottavio G, Milella MS, Li X, Grimm JW, Caprioli D. Factors modulating the incubation of drug and non-drug craving and their clinical implications. Neurosci Biobehav Rev 2021; 131:847-864. [PMID: 34597716 PMCID: PMC8931548 DOI: 10.1016/j.neubiorev.2021.09.050] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/20/2021] [Accepted: 09/26/2021] [Indexed: 12/28/2022]
Abstract
It was suggested in 1986 that cue-induced cocaine craving increases progressively during early abstinence and remains high during extended periods of time. Clinical evidence now supports this hypothesis and that this increase is not specific to cocaine but rather generalize across several drugs of abuse. Investigators have identified an analogous incubation phenomenon in rodents, in which time-dependent increases in cue-induced drug seeking are observed after abstinence from intravenous drug or palatable food self-administration. Incubation of craving is susceptible to variation in magnitude as a function of biological and/or the environmental circumstances surrounding the individual. During the last decade, the neurobiological correlates of the modulatory role of biological (sex, age, genetic factors) and environmental factors (environmental enrichment and physical exercise, sleep architecture, acute and chronic stress, abstinence reinforcement procedures) on incubation of drug craving has been investigated. In this review, we summarized the behavioral procedures adopted, the key underlying neurobiological correlates and clinical implications of these studies.
Collapse
Affiliation(s)
- Marco Venniro
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, USA.
| | - Ingrid Reverte
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - Leslie A Ramsey
- Behavioral Neuroscience Research Branch, Intramural Research Program, Baltimore NIDA, NIH, USA
| | - Kimberly M Papastrat
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, USA
| | - Ginevra D'Ottavio
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | | | - Xuan Li
- Department of Psychology, University of Maryland College Park, College Park, USA.
| | - Jeffrey W Grimm
- Department of Psychology and Program in Behavioral Neuroscience, Western Washington University, Bellingham, USA.
| | - Daniele Caprioli
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy.
| |
Collapse
|
14
|
Perez RE, Basu A, Nabit BP, Harris NA, Folkes OM, Patel S, Gilsbach R, Hein L, Winder DG. α 2A-adrenergic heteroreceptors are required for stress-induced reinstatement of cocaine conditioned place preference. Neuropsychopharmacology 2020; 45:1473-1481. [PMID: 32074627 PMCID: PMC7360592 DOI: 10.1038/s41386-020-0641-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 02/08/2023]
Abstract
The α2a-adrenergic receptor (α2a-AR) agonist guanfacine has been investigated as a potential treatment for substance use disorders. While decreasing stress-induced reinstatement of cocaine seeking in animal models and stress-induced craving in human studies, guanfacine has not been reported to decrease relapse rates. Although guanfacine engages α2a-AR autoreceptors, it also activates excitatory Gi-coupled heteroreceptors in the bed nucleus of the stria terminalis (BNST), a key brain region in driving stress-induced relapse. Thus, BNST α2a-AR heteroreceptor signaling might decrease the beneficial efficacy of guanfacine. We aimed to determine the role of α2a-AR heteroreceptors and BNST Gi-GPCR signaling in stress-induced reinstatement of cocaine conditioned place preference (CPP) and the effects of low dose guanfacine on BNST activity and stress-induced reinstatement. We used a genetic deletion strategy and the cocaine CPP procedure to first define the contributions of α2a-AR heteroreceptors to stress-induced reinstatement. Next, we mimicked BNST Gi-coupled α2a-AR heteroreceptor signaling using a Gi-coupled designer receptor exclusively activated by designer drug (Gi-DREADD) approach. Finally, we evaluated the effects of low-dose guanfacine on BNST cFOS immunoreactivity and stress-induced reinstatement. We show that α2a-AR heteroreceptor deletion disrupts stress-induced reinstatement and that BNST Gi-DREADD activation is sufficient to induce reinstatement. Importantly, we found that low-dose guanfacine does not increase BNST activity, but prevents stress-induced reinstatement. Our findings demonstrate a role for α2a-AR heteroreceptors and BNST Gi-GPCR signaling in stress-induced reinstatement of cocaine CPP and provide insight into the impact of dose on the efficacy of guanfacine as a treatment for stress-induced relapse of cocaine use.
Collapse
Affiliation(s)
- Rafael E Perez
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
| | - Aakash Basu
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
| | - Bretton P Nabit
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
| | - Nicholas A Harris
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
| | - Oakleigh M Folkes
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| | - Sachin Patel
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Ralf Gilsbach
- Institute for Cardiovascular Physiology, University Hospital, Goethe University, Frankfurt, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Danny G Winder
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States.
- Department of Pharmacology, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States.
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Basic Sciences, Nashville, TN, United States.
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Vanderbilt University School of Medicine, Nashville, TN, United States.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States.
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University School of Medicine, Nashville, TN, United States.
| |
Collapse
|
15
|
Riga D, Schmitz LJ, Mourik Y, Hoogendijk WJ, De Vries TJ, Smit AB, Spijker S. Stress vulnerability promotes an alcohol-prone phenotype in a preclinical model of sustained depression. Addict Biol 2020; 25:e12701. [PMID: 30561063 PMCID: PMC6916303 DOI: 10.1111/adb.12701] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/28/2018] [Accepted: 11/05/2018] [Indexed: 12/15/2022]
Abstract
Major depression and alcohol‐related disorders frequently co‐occur. Depression severity weighs on the magnitude and persistence of comorbid alcohol use disorder (AUD), with severe implications for disease prognosis. Here, we investigated whether depression vulnerability drives propensity to AUD at the preclinical level. We used the social defeat–induced persistent stress (SDPS) model of chronic depression in combination with operant alcohol self‐administration (SA). Male Wistar rats were subjected to social defeat (five episodes) and prolonged social isolation (~12 weeks) and subsequently classified as SDPS‐prone or SDPS‐resilient based on their affective and cognitive performance. Using an operant alcohol SA paradigm, acquisition, motivation, extinction, and cue‐induced reinstatement of alcohol seeking were examined in the two subpopulations. SDPS‐prone animals showed increased alcohol SA, heightened motivation to acquire alcohol, persistent alcohol seeking despite alcohol unavailability, signs of extinction resistance, and increased cue‐induced relapse; the latter could be blocked by the α2 adrenoreceptor agonist guanfacine. In SDPS‐resilient rats, prior exposure to social defeat increased alcohol SA without affecting any other measures of alcohol seeking and alcohol taking. Our data revealed that depression proneness confers vulnerability to alcohol, emulating patterns of alcohol dependence seen in human addicts, and that depression resilience to a large extent protects from the development of AUD‐like phenotypes. Furthermore, our data suggest that stress exposure alone, independently of depressive symptoms, alters alcohol intake in the long‐term.
Collapse
Affiliation(s)
- Danai Riga
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam The Netherlands
| | - Leanne J.M. Schmitz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam The Netherlands
| | - Yvar Mourik
- Department of Anatomy and Neurosciences, Amsterdam NeuroscienceVrije Universiteit Medical Center Amsterdam The Netherlands
| | | | - Taco J. De Vries
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam The Netherlands
- Department of Anatomy and Neurosciences, Amsterdam NeuroscienceVrije Universiteit Medical Center Amsterdam The Netherlands
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam The Netherlands
| | - Sabine Spijker
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam The Netherlands
| |
Collapse
|
16
|
Regier PS, Kampman KM, Childress AR. Clinical Trials for Stimulant Use Disorders: Addressing Heterogeneities That May Undermine Treatment Outcomes. Handb Exp Pharmacol 2020; 258:299-322. [PMID: 32193666 DOI: 10.1007/164_2019_303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In recent years, use of cocaine and amphetamines and deaths associated with stimulants have been on the rise, and there are still no FDA-approved medications for stimulant use disorders. One contributing factor may involve heterogeneity. At the neurobiological level, dual dopamine dysfunction may be undermining medication efficacy, suggesting a need for combination pharmacotherapies. At the population level, individual variability is expressed in a number of ways and, if left unaddressed, may interfere with medication efficacy. This chapter reviews studies investigating medications to address dopamine dysfunction, and it also identifies several prominent heterogeneities associated with stimulant (and other substance) use disorders. The chapter has implications for improving interventions to treat stimulant use disorders, and the theme of individual heterogeneity may have broader application across substance use disorders.
Collapse
Affiliation(s)
- Paul S Regier
- Department of Psychiatry, Perelman School of Medicine, Center for Studies of Addiction, University of Pennsylvania, Philadelphia, PA, USA.
| | - Kyle M Kampman
- Department of Psychiatry, Perelman School of Medicine, Center for Studies of Addiction, University of Pennsylvania, Philadelphia, PA, USA
| | - Anna Rose Childress
- Department of Psychiatry, Perelman School of Medicine, Center for Studies of Addiction, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
17
|
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]
|
18
|
Wemm SE, Larkin C, Hermes G, Tennen H, Sinha R. A day-by-day prospective analysis of stress, craving and risk of next day alcohol intake during alcohol use disorder treatment. Drug Alcohol Depend 2019; 204:107569. [PMID: 31574406 PMCID: PMC6916671 DOI: 10.1016/j.drugalcdep.2019.107569] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/27/2019] [Accepted: 07/15/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Stress has been known to increase craving in individuals with Alcohol Use Disorders (AUD) and predict future alcohol relapse risk, but whether stress on a particular day affects craving on that day to impact prospective alcohol intake in the real world, particularly during early treatment and recovery, has not been studied thus far. METHOD The first study included 85 AUD individuals who reported their daily stress, craving, and alcohol intake in the first two weeks of early treatment. A second validation study included 28 AUD patients monitored daily during eight weeks of outpatient 12-Step based behavioral counseling treatment for AUD. Data were collected from telephone-based daily diaries for 903 days in Study 1 and 1488 in Study 2. Multilevel latent models tested if daily and person-averaged craving mediated the link between stressful events and next day drinking during treatment. RESULTS In both Study 1 and 2, exposure to a stressful event on a particular day predicted increased craving on that day (p's≤.002); and such increases in craving predicted the likelihood of drinking the next day (p's≤.014) and the drinking amount (p's< = 008). Individuals who experienced more stressful events reported higher craving (p's≤.012), and higher cravers reported greater next day drinking (p's<.001). CONCLUSIONS The results across two studies with separate samples are the first to establish that craving directly mediates the association between stress and next day alcohol intake in individuals with AUD. Findings suggest a need for novel treatment approaches to address stress-induced craving to improve alcohol use outcomes.
Collapse
Affiliation(s)
- Stephanie E Wemm
- Yale Stress Center, Yale School of Medicine, 2 Church St South Suite 209, New Haven, CT, 06379, USA
| | - Chloe Larkin
- Yale Stress Center, Yale School of Medicine, 2 Church St South Suite 209, New Haven, CT, 06379, USA
| | - Gretchen Hermes
- Yale Stress Center, Yale School of Medicine, 2 Church St South Suite 209, New Haven, CT, 06379, USA
| | - Howard Tennen
- Department of Community Medicine, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06032, USA
| | - Rajita Sinha
- Yale Stress Center, Yale School of Medicine, 2 Church St South Suite 209, New Haven, CT, 06379, USA.
| |
Collapse
|
19
|
Incubation of Cocaine Craving After Intermittent-Access Self-administration: Sex Differences and Estrous Cycle. Biol Psychiatry 2019; 85:915-924. [PMID: 30846301 PMCID: PMC6534474 DOI: 10.1016/j.biopsych.2019.01.015] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 11/20/2022]
Abstract
BACKGROUND Studies using continuous-access drug self-administration showed that cocaine seeking increases during abstinence (incubation of cocaine craving). Recently, studies using intermittent-access self-administration showed increased motivation to self-administer and seek cocaine. We examined whether intermittent cocaine self-administration would potentiate incubation of craving in male and female rats and examined the estrous cycle's role in this incubation. METHODS In experiment 1, male and female rats self-administered cocaine either continuously (8 hours/day) or intermittently (5 minutes ON, 25 minutes OFF × 16) for 12 days, followed by relapse tests after 2 or 29 days. In experiments 2 and 3, female rats self-administered cocaine intermittently for six, 12, or 18 sessions. In experiment 4, female rats self-administered cocaine continuously followed by relapse tests after 2 or 29 days. In experiments 3 and 4, the estrous cycle was measured using a vaginal smear test. RESULTS Incubation of cocaine craving was observed in both sexes after either intermittent or continuous drug self-administration. Independent of access condition and abstinence day, cocaine seeking was higher in female rats than in male rats. In both sexes, cocaine seeking on both abstinence days was higher after intermittent drug access than after continuous drug access. In female rats, incubation of craving after either intermittent or continuous drug access was significantly higher during estrus than during non-estrus; for intermittent drug access, this effect was independent of the training duration. CONCLUSIONS In both sexes, intermittent cocaine access caused time-independent increases in drug seeking during abstinence. In female rats, the time-dependent increase in drug seeking (incubation) is critically dependent on the estrous cycle phase.
Collapse
|
20
|
Wemm SE, Sinha R. Drug-induced stress responses and addiction risk and relapse. Neurobiol Stress 2019; 10:100148. [PMID: 30937354 PMCID: PMC6430516 DOI: 10.1016/j.ynstr.2019.100148] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/07/2018] [Accepted: 01/30/2019] [Indexed: 12/21/2022] Open
Abstract
A number of studies have assessed the effects of psychoactive drugs on stress biology, the neuroadaptations resulting from chronic drug use on stress biology, and their effects on addiction risk and relapse. This review mainly covers human research on the acute effects of different drugs of abuse (i.e., nicotine, cannabis, psychostimulants, alcohol, and opioids) on the hypothalamic-pituitary-adrenal (HPA) axis and the autonomic nervous system (ANS) responses. We review the literature on acute peripheral stress responses in naïve or light recreational users and binge/heavy or chronic drug users. We also discuss evidence of alterations in tonic levels, or tolerance, in the latter relative to the former and associated changes in the phasic stress responses. We discuss the impact of the stress system tolerance in heavy users on their response to drug- and stress-related cue responses and craving as compared to control subjects. A summary is provided of the effects of glucocorticoid responses and their adaptations on brain striatal and prefrontal cortices involved in the regulation of drug seeking and relapse risk. Finally, we summarize important considerations, including individual difference factors such as gender, co-occurring drug use, early trauma and adversity and drug use history and variation in methodologies, that may further influence the effects of these drugs on stress biology.
Collapse
Affiliation(s)
- Stephanie E. Wemm
- Yale Stress Center, Yale School of Medicine, 2 Church St South Suite 209, New Haven, CT, 06519, USA
| | | |
Collapse
|
21
|
Nawata Y, Yamaguchi T, Fukumori R, Yamamoto T. Inhibition of Monoacylglycerol Lipase Reduces the Reinstatement of Methamphetamine-Seeking and Anxiety-Like Behaviors in Methamphetamine Self-Administered Rats. Int J Neuropsychopharmacol 2018; 22:165-172. [PMID: 30481332 PMCID: PMC6368370 DOI: 10.1093/ijnp/pyy086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Methamphetamine is a highly addictive psychostimulant with reinforcing properties. Our laboratory previously found that Δ8-tetrahydrocannabinol, an exogenous cannabinoid, suppressed the reinstatement of methamphetamine-seeking behavior. The purpose of this study was to determine whether the elevation of endocannabinoids modulates the reinstatement of methamphetamine-seeking behavior and emotional changes in methamphetamine self-administered rats. METHODS Rats were tested for the reinstatement of methamphetamine-seeking behavior following methamphetamine self-administration and extinction. The elevated plus-maze test was performed in methamphetamine self-administered rats during withdrawal. We investigated the effects of JZL184 and URB597, 2 inhibitors of endocannabinoid hydrolysis, on the reinstatement of methamphetamine-seeking and anxiety-like behaviors. RESULTS JZL184 (32 and 40 mg/kg, i.p.), an inhibitor of monoacylglycerol lipase, significantly attenuated both the cue- and stress-induced reinstatement of methamphetamine-seeking behavior. Furthermore, URB597 (3.2 and 10 mg/kg, i.p.), an inhibitor of fatty acid amide hydrolase, attenuated only cue-induced reinstatement. AM251, a cannabinoid CB1 receptor antagonist, antagonized the attenuation of cue-induced reinstatement by JZL184 but not URB597. Neither JZL184 nor URB597 reinstated methamphetamine-seeking behavior when administered alone. In the elevated plus-maze test, rats that were in withdrawal from methamphetamine self-administration spent less time in the open arms. JZL184 ameliorated the decrease in time spent in the open arms. CONCLUSION We showed that JZL184 reduced both the cue- and stress-induced reinstatement of methamphetamine-seeking and anxiety-like behaviors in rats that had self-administered methamphetamine. It was suggested that a decrease in 2-arachidonoylglycerol in the brain could drive the reinstatement of methamphetamine-seeking and anxiety-like behaviors.
Collapse
Affiliation(s)
- Yoko Nawata
- Department of Pharmacology, Faculty of Pharmaceutical Science, Nagasaki International University, Nagasaki, Japan
| | - Taku Yamaguchi
- Department of Pharmacology, Faculty of Pharmaceutical Science, Nagasaki International University, Nagasaki, Japan
| | - Ryo Fukumori
- Department of Pharmacology, Faculty of Pharmaceutical Science, Nagasaki International University, Nagasaki, Japan
| | - Tsuneyuki Yamamoto
- Department of Pharmacology, Faculty of Pharmaceutical Science, Nagasaki International University, Nagasaki, Japan,Correspondence: Tsuneyuki Yamamoto, PhD, Department of Pharmacology, Faculty of Pharmaceutical Science, Nagasaki International University, 2825–7 Huis Ten Bosch Sasebo, Nagasaki 859–3298, Japan ()
| |
Collapse
|
22
|
Interaction between noradrenergic and cholinergic signaling in amygdala regulates anxiety- and depression-related behaviors in mice. Neuropsychopharmacology 2018; 43:2118-2125. [PMID: 29472646 PMCID: PMC6098039 DOI: 10.1038/s41386-018-0024-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 12/20/2022]
Abstract
Medications that target the noradrenergic system are important therapeutics for depression and anxiety disorders. More recently, clinical studies have shown that the α2-noradrenergic receptor (α2AR) agonist guanfacine can decrease stress-induced smoking relapse during acute abstinence, suggesting that targeting the noradrenergic system may aid in smoking cessation through effects on stress pathways in the brain. Acetylcholine (ACh), like the nicotine in tobacco, acts at nicotinic acetylcholine receptors (nAChRs) to regulate behaviors related to anxiety and depression. We therefore investigated interactions between guanfacine and ACh signaling in tests of anxiolytic and antidepressant efficacy in female and male C57BL/6J mice, focusing on the amygdala as a potential site of noradrenergic/cholinergic interaction. The antidepressant-like effects of guanfacine were blocked by shRNA-mediated knockdown of α2AR in amygdala. Knockdown of the high-affinity β2 nAChR subunit in amygdala also prevented antidepressant-like effects of guanfacine, suggesting that these behavioral effects require ACh signaling through β2-containing nAChRs in this brain area. Ablation of NE terminals prevented the anxiolytic- and antidepressant-like effects of the nicotinic partial agonist cytisine, whereas administration of the cholinesterase antagonist physostigmine induced a depression-like phenotype that was not altered by knocking down α2AR in the amygdala. These studies suggest that ACh and NE have opposing actions on behaviors related to anxiety and depression and that cholinergic signaling through β2-containing nAChRs and noradrenergic signaling through α2a receptors in neurons of the amygdala are critical for regulation of these behaviors.
Collapse
|
23
|
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
|
24
|
Ellis AS, Fosnocht AQ, Lucerne KE, Briand LA. Disruption of GluA2 phosphorylation potentiates stress responsivity. Behav Brain Res 2017; 333:83-89. [PMID: 28668281 DOI: 10.1016/j.bbr.2017.06.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 12/15/2022]
Abstract
Cocaine addiction is characterized by persistent craving and addicts frequently relapse even after long periods of abstinence. Exposure to stress can precipitate relapse in humans and rodents. Stress and drug use can lead to common alterations in synaptic plasticity and these commonalities may contribute to the ability of stress to elicit relapse. These common changes in synaptic plasticity are mediated, in part, by alterations in the trafficking and stabilization of AMPA receptors. Exposure to both cocaine and stress can lead to alterations in protein kinase C-mediated phosphorylation of GluA2 AMPA subunits and thus alter the trafficking of GluA2-containing AMPARs. However, it is not clear what role AMPAR trafficking plays in the interactions between stress and cocaine. The current study utilized a mouse with a point mutation within the GluA2 subunit c-terminus resulting in a disruption of PKC-mediated GluA2 phosphorylation to examine stress responsivity. Although no differences were seen in the response to a forced swim stress in naïve mice, GluA2 K882A knock-in mice exhibited an increased stress response following cocaine self-administration. Furthermore, we demonstrated that disrupting GluA2 phosphorylation increases vulnerability to stress-induced reinstatement of both cocaine seeking and cocaine-conditioned reward. Finally, GluA2 K882A knock-in mice exhibit an increased vulnerability to social defeat as indicated by increased social avoidance. Taken together these results indicate that disrupting GluA2 phosphorylation leads to increased responsivity to acute stress following cocaine exposure and increased vulnerability to chronic stress. These results highlight the GluA2 phosphorylation site as a novel target for the stress-related disorders.
Collapse
Affiliation(s)
- Alexandra S Ellis
- Department of Psychology, Temple University, United States; Neuroscience Program, Temple University, United States
| | | | | | - Lisa A Briand
- Department of Psychology, Temple University, United States; Neuroscience Program, Temple University, United States.
| |
Collapse
|
25
|
Vranjkovic O, Pina M, Kash TL, Winder DG. The bed nucleus of the stria terminalis in drug-associated behavior and affect: A circuit-based perspective. Neuropharmacology 2017; 122:100-106. [PMID: 28351600 DOI: 10.1016/j.neuropharm.2017.03.028] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 10/19/2022]
Abstract
The bed nucleus of the stria terminalis was first described nearly a century ago and has since emerged as a region central to motivated behavior and affective states. The last several decades have firmly established a role for the BNST in drug-associated behavior and implicated this region in addiction-related processes. Whereas past approaches used to characterize the BNST have focused on a more general role of this region and its subnuclei in behavior, more recent work has begun to reveal its elaborate circuitry and cellular components. Such recent developments are largely owed to methodological advances, which have made possible efforts previously deemed intractable, such as tracing of long-range cell-type specific projections and identifying functional efferent and afferent connections. In this review, we integrate earlier foundational work with more recent and advanced studies to construct a broad overview of the molecular neurocircuitry of the BNST in drug-associated behavior and affect. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Oliver Vranjkovic
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, USA
| | - Melanie Pina
- Bowles Center for Alcohol Studies, Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, USA
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, USA
| | - Danny G Winder
- Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, USA; Department of Psychiatry, Vanderbilt University School of Medicine, USA; Department of Pharmacology, Vanderbilt University School of Medicine, USA; Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
26
|
Havranek MM, Hulka LM, Tasiudi E, Eisenegger C, Vonmoos M, Preller KH, Mössner R, Baumgartner MR, Seifritz E, Grünblatt E, Quednow BB. α 2A -Adrenergic receptor polymorphisms and mRNA expression levels are associated with delay discounting in cocaine users. Addict Biol 2017; 22:561-569. [PMID: 26549422 DOI: 10.1111/adb.12324] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/19/2015] [Accepted: 10/02/2015] [Indexed: 12/01/2022]
Abstract
Cocaine users characteristically display preferences for smaller immediate rewards over larger delayed rewards, and this delay discounting (DD) has been proposed as an endophenotype of cocaine addiction. Recent evidence suggests that the norepinephrine system and more specifically the α2A -adrenergic receptor (ADRA2A) are impacted by chronic cocaine use while also being potentially involved in the neural mechanisms underlying DD. Hence, we investigated the effects of ADRA2A polymorphisms and ADRA2A mRNA expression levels on DD of cocaine users and stimulant-naïve controls. Two hundred and twenty-three participants (129 cocaine users and 94 stimulant-naïve healthy controls) completed a computerized DD paradigm and were genotyped for three single nucleotide polymorphisms (SNPs; rs1800544, rs521674 and rs602618) in the ADRA2A gene, while their peripheral ADRA2A mRNA expression was quantified in whole blood samples. The three SNPs were in near-perfect linkage disequilibrium. Accordingly, significant group*genotype interactions were found for all three ADRA2A variants revealing steeper DD in cocaine users (but not in controls) carrying the G-allele of SNP rs1800544, the T-allele of rs521674 and the C-allele of rs602618. Similarly, high ADRA2A mRNA expression levels were significantly associated with a reduced tendency to choose smaller more immediate rewards (over larger delayed rewards) in cocaine users but not in controls. As the relationship between DD and cocaine use was moderated by ADRA2A SNPs and by peripheral ADRA2A gene expression, we propose that the norepinephrine system is involved in DD deficits observed in cocaine using individuals. Consequently, pharmacological compounds targeting ADRA2As might be considered for the symptom-specific treatment of delay aversion in stimulant addiction.
Collapse
Affiliation(s)
- Michael M. Havranek
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital; University of Zurich; Switzerland
| | - Lea M. Hulka
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital; University of Zurich; Switzerland
| | - Eve Tasiudi
- University Clinic for Child and Adolescent Psychiatry; University of Zurich; Switzerland
| | - Christoph Eisenegger
- Neuropsychopharmacology and Biopsychology Unit, Institute of Basic Psychological Research and Research Methods; University of Vienna; Austria
| | - Matthias Vonmoos
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital; University of Zurich; Switzerland
| | - Katrin H. Preller
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital; University of Zurich; Switzerland
| | - Rainald Mössner
- Department of Psychiatry and Psychotherapy; University of Tübingen; Germany
| | - Markus R. Baumgartner
- Center of Forensic Hairanalytics, Institute of Legal Medicine; University of Zurich; Switzerland
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital; University of Zurich; Switzerland
- Neuroscience Center Zurich; University and ETH Zurich; Switzerland
| | - Edna Grünblatt
- University Clinic for Child and Adolescent Psychiatry; University of Zurich; Switzerland
- Neuroscience Center Zurich; University and ETH Zurich; Switzerland
| | - Boris B. Quednow
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital; University of Zurich; Switzerland
- Neuroscience Center Zurich; University and ETH Zurich; Switzerland
| |
Collapse
|
27
|
Passamonti L, Luijten M, Ziauddeen H, Coyle-Gilchrist ITS, Rittman T, Brain SAE, Regenthal R, Franken IHA, Sahakian BJ, Bullmore ET, Robbins TW, Ersche KD. Atomoxetine effects on attentional bias to drug-related cues in cocaine dependent individuals. Psychopharmacology (Berl) 2017; 234:2289-2297. [PMID: 28551713 PMCID: PMC5519645 DOI: 10.1007/s00213-017-4643-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 05/11/2017] [Indexed: 12/19/2022]
Abstract
RATIONALE Biased attention towards drug-related cues and reduced inhibitory control over the regulation of drug-intake characterize drug addiction. The noradrenaline system has been critically implicated in both attentional and response inhibitory processes and is directly affected by drugs such as cocaine. OBJECTIVES We examined the potentially beneficial effects of the noradrenaline reuptake inhibitor atomoxetine in improving cognitive control during two tasks that used cocaine- and non-cocaine-related stimuli. METHODS A double-blind, placebo-controlled, and cross-over psycho-pharmacological design was employed. A single oral dose of atomoxetine (40 mg) was administered to 28 cocaine-dependent individuals (CDIs) and 28 healthy controls. All participants performed a pictorial attentional bias task involving both cocaine- and non-cocaine-related pictures as well as a verbal go/no-go task composed of cocaine- and food-related words. RESULTS As expected, CDIs showed attentional bias to cocaine-related cues whilst controls did not. More importantly, however, atomoxetine, relative to placebo, significantly attenuated attentional bias in CDIs (F 26 = 6.73, P = 0.01). During the go/no-go task, there was a treatment × trial × group interaction, although this finding only showed a trend towards statistical significance (F 26 = 3.38, P = 0.07). CONCLUSIONS Our findings suggest that atomoxetine reduces attentional bias to drug-related cues in CDIs. This may result from atomoxetine's modulation of the balance between tonic/phasic activity in the locus coeruleus and the possibly parallel enhancement of noradrenergic neurotransmission within the prefrontal cortex. Studying how cognitive enhancers such as atomoxetine influence key neurocognitive indices in cocaine addiction may help to develop reliable biomarkers for patient stratification in future clinical trials.
Collapse
Affiliation(s)
- Luca Passamonti
- Departments of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0SZ, UK. .,Consiglio Nazionale delle Ricerche, Istituto di Bioimmagini e Fisiologia Molecolare, Catanzaro, Italy.
| | - M. Luijten
- Behavioural Science Institute, Radboud University, Nijmegen, Netherlands
| | - H. Ziauddeen
- Cambridgeshire and Peterborough Foundation Trust, Cambridge, UK
| | | | - T. Rittman
- Departments of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0SZ UK
| | - S. A. E. Brain
- Departments of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0SZ UK
| | - R. Regenthal
- Division of Clinical Pharmacology, Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - I. H. A. Franken
- Institute of Psychology, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - B. J. Sahakian
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - E. T. Bullmore
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK ,GlaxoSmithKline, Clinical Unit Cambridge, Cambridge, UK
| | - T. W. Robbins
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - K. D. Ersche
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
28
|
Fosnocht AQ, Briand LA. Substance use modulates stress reactivity: Behavioral and physiological outcomes. Physiol Behav 2016; 166:32-42. [PMID: 26907955 DOI: 10.1016/j.physbeh.2016.02.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 02/11/2016] [Accepted: 02/15/2016] [Indexed: 01/01/2023]
Abstract
Drug addiction is a major public health concern in the United States costing taxpayers billions in health care costs, lost productivity and law enforcement. However, the availability of effective treatment options remains limited. The development of novel therapeutics will not be possible without a better understanding of the addicted brain. Studies in both clinical and preclinical models indicate that chronic drug use leads to alterations in the body and brain's response to stress. Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis may shed light on the ability of stress to increase vulnerability to relapse. Further, within both the HPA axis and limbic brain regions, corticotropin-releasing factor (CRF) is critically involved in the brain's response to stress. Alterations in both central and peripheral CRF activity seen following chronic drug use provide a mechanism by which substance use can alter stress reactivity, thus mediating addictive phenotypes. While many reviews have focused on how stress alters drug-mediated changes in physiology and behavior, the goal of this review is to focus on how substance use alters responses to stress.
Collapse
Affiliation(s)
| | - Lisa A Briand
- Department of Psychology, Temple University, United States.
| |
Collapse
|
29
|
Mineur YS, Bentham MP, Zhou WL, Plantenga ME, McKee SA, Picciotto MR. Antidepressant-like effects of guanfacine and sex-specific differences in effects on c-fos immunoreactivity and paired-pulse ratio in male and female mice. Psychopharmacology (Berl) 2015; 232:3539-49. [PMID: 26146014 PMCID: PMC4561580 DOI: 10.1007/s00213-015-4001-3] [Citation(s) in RCA: 24] [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] [Received: 05/13/2015] [Accepted: 06/25/2015] [Indexed: 02/02/2023]
Abstract
RATIONALE The a2A-noradrenergic agonist guanfacine can decreases stress-induced smoking in female, but not male, human smokers. It is not known whether these effects are due to effects on mood regulation and/or result from nicotinic-cholinergic interactions. OBJECTIVES The objective of the study was to determine whether there are sex differences in the effect of guanfacine in tests of anxiolytic and antidepressant efficacy in mice at baseline and in a hypercholinergic model of depression induced by the acetylcholinesterase inhibitor physostigmine. METHODS The effects of guanfacine were measured in the light/dark box, tail suspension, and the forced swim test in female and male C57BL/6J mice. In parallel, electrophysiological properties were evaluated in the prefrontal cortex, a critical brain region involved in stress responses. c-fos immunoreactivity was measured in other brain regions known to regulate mood. RESULTS Despite a baseline sex difference in behavior in the forced swim test (female mice were more immobile), guanfacine had similar, dose-dependent, antidepressant-like effects in mice of both sexes (optimal dose, 0.15 mg/kg). An antidepressant-like effect of guanfacine was also observed following pre-treatment with physostigmine. A sex difference in the paired-pulse ratio in the prefrontal cortex (PFC) (male, 1.4; female, 2.1) was observed at baseline that was normalized by guanfacine. Other brain areas involved in cholinergic control of depression-like behaviors, including the basolateral amygdala and lateral septum, showed sex-specific changes in c-fos expression. CONCLUSIONS Guanfacine has a robust antidepressant-like effect and can reverse a depression-like state induced by increased acetylcholine (ACh) signaling. These data suggest that different brain areas are recruited in female and male mice, despite similar behavioral responses to guanfacine.
Collapse
Affiliation(s)
| | | | | | | | | | - Marina R. Picciotto
- Correspondence should be addressed to: Marina R. Picciotto, Dept.
of Psychiatry, Yale University School of Medicine, 34 Park Street – 3rd floor
research, New Haven, CT 06508, Phone: 203-737-2041; Fax: 203-737-2043;
| |
Collapse
|
30
|
Kowalczyk WJ, Phillips KA, Jobes ML, Kennedy AP, Ghitza UE, Agage DA, Schmittner JP, Epstein DH, Preston KL. Clonidine Maintenance Prolongs Opioid Abstinence and Decouples Stress From Craving in Daily Life: A Randomized Controlled Trial With Ecological Momentary Assessment. Am J Psychiatry 2015; 172:760-7. [PMID: 25783757 PMCID: PMC6233893 DOI: 10.1176/appi.ajp.2014.14081014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The authors tested whether clonidine blocks stress-induced seeking of heroin and cocaine. The study was also intended to confirm translational findings from a rat model of drug relapse by using ecological momentary assessment of patients' stress to test hypotheses about clonidine's behavioral mechanism of action. METHOD The authors conducted a randomized double-blind placebo-controlled clinical trial with 208 opioid-dependent patients at an outpatient buprenorphine clinic. The 118 participants (57%) who maintained abstinence during weeks 5-6 were continued on buprenorphine and randomly assigned to receive clonidine (N=61) or placebo (N=57) for 14 weeks. Urine was tested thrice weekly. Lapse was defined as any opioid-positive or missed urine test, and relapse as two or more consecutive lapses. Time to lapse and relapse were examined with Cox regressions; longest period of abstinence was examined with a t test, and ecological momentary assessment data were examined with generalized linear mixed models. RESULTS In an intent-to-treat analysis, clonidine produced the longest duration (in consecutive days) of abstinence from opioids during the intervention phase (34.8 days [SD=3.7] compared with 25.5 days [SD=2.7]; Cohen's d=0.38). There was no group difference in time to relapse, but the clonidine group took longer to lapse (hazard ratio=0.67, 95% CI=0.45-1.00). Ecological momentary assessment showed that daily-life stress was partly decoupled from opioid craving in the clonidine group, supporting the authors' hypothesized mechanism for clonidine's benefits. CONCLUSIONS Clonidine, a readily available medication, is useful in opioid dependence not just for reduction of withdrawal signs, but also as an adjunctive maintenance treatment that increases duration of abstinence. Even in the absence of physical withdrawal, it decouples stress from craving in everyday life.
Collapse
Affiliation(s)
| | | | - Michelle L. Jobes
- Clinical Pharmacology and Therapeutics Research Branch, NIDA, Baltimore
| | | | - Udi E. Ghitza
- Clinical Trials Operations and Biostatistics Branch, NIMH, Rockville, Md
| | - Daniel A. Agage
- Clinical Pharmacology and Therapeutics Research Branch, NIDA, Baltimore
| | - John P. Schmittner
- Advanced Heart Failure Program, Spectrum Health System, Grand Rapids, Mich
| | - David H. Epstein
- Clinical Pharmacology and Therapeutics Research Branch, NIDA, Baltimore
| | - Kenzie L. Preston
- Clinical Pharmacology and Therapeutics Research Branch, NIDA, Baltimore
| |
Collapse
|
31
|
Hains AB, Yabe Y, Arnsten AF. Chronic Stimulation of Alpha-2A-Adrenoceptors With Guanfacine Protects Rodent Prefrontal Cortex Dendritic Spines and Cognition From the Effects of Chronic Stress. Neurobiol Stress 2015; 2:1-9. [PMID: 25664335 PMCID: PMC4316374 DOI: 10.1016/j.ynstr.2015.01.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/18/2014] [Accepted: 01/07/2015] [Indexed: 01/01/2023] Open
Abstract
The prefrontal cortex (PFC) provides top-down regulation of behavior, cognition, and emotion, including spatial working memory. However, these PFC abilities are greatly impaired by exposure to acute or chronic stress. Chronic stress exposure in rats induces atrophy of PFC dendrites and spines that correlates with working memory impairment. As similar PFC grey matter loss appears to occur in mental illness, the mechanisms underlying these changes need to be better understood. Acute stress exposure impairs PFC cognition by activating feedforward cAMP-calcium-K+ channel signaling, which weakens synaptic inputs and reduces PFC neuronal firing. Spine loss with chronic stress has been shown to involve calcium-protein kinase C signaling, but it is not known if inhibiting cAMP signaling would similarly prevent the atrophy induced by repeated stress. The current study examined whether inhibiting cAMP signaling through alpha-2A-adrenoceptor stimulation with chronic guanfacine treatment would protect PFC spines and working memory performance during chronic stress exposure. Guanfacine was selected due to 1) its established effects on cAMP signaling at post-synaptic alpha-2A receptors on spines in PFC, and 2) its increasing clinical use for the treatment of pediatric stress disorders. Daily guanfacine treatment compared to vehicle control was found to prevent dendritic spine loss in layer II/III pyramidal neurons of prelimbic PFC in rats exposed to chronic restraint stress. Guanfacine also protected working memory performance; cognitive performance correlated with dendritic spine density. These findings suggest that chronic guanfacine use may have clinical utility by protecting PFC gray matter from the detrimental effects of stress.
Collapse
Affiliation(s)
| | | | - Amy F.T. Arnsten
- Department of Neurobiology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| |
Collapse
|
32
|
Ghitza UE. Needed Relapse-Prevention Research on Novel Framework (ASPIRE Model) for Substance Use Disorders Treatment. Front Psychiatry 2015; 6:37. [PMID: 25798112 PMCID: PMC4351566 DOI: 10.3389/fpsyt.2015.00037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/21/2015] [Indexed: 11/29/2022] Open
Affiliation(s)
- Udi E Ghitza
- Center for the Clinical Trials Network, National Institute on Drug Abuse, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
33
|
The Effects of Stress Exposure on Prefrontal Cortex: Translating Basic Research into Successful Treatments for Post-Traumatic Stress Disorder. Neurobiol Stress 2015; 1:89-99. [PMID: 25436222 PMCID: PMC4244027 DOI: 10.1016/j.ynstr.2014.10.002] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Research on the neurobiology of the stress response in animals has led to successful new treatments for Post-Traumatic Stress Disorder (PTSD) in humans. Basic research has found that high levels of catecholamine release during stress rapidly impair the top-down cognitive functions of the prefrontal cortex (PFC), while strengthening the emotional and habitual responses of the amygdala and basal ganglia. Chronic stress exposure leads to dendritic atrophy in PFC, dendritic extension in the amygdala, and strengthening of the noradrenergic (NE) system. High levels of NE release during stress engage low affinity alpha-1 adrenoceptors, (and likely beta-1 adrenoceptors), which rapidly reduce the firing of PFC neurons, but strengthen amygdala function. In contrast, moderate levels of NE release during nonstress conditions engage higher affinity alpha-2A receptors, which strengthen PFC, weaken amygdala, and regulate NE cell firing. Thus, either alpha-1 receptor blockade or alpha-2A receptor stimulation can protect PFC function during stress. Patients with PTSD have signs of PFC dysfunction. Clinical studies have found that blocking alpha-1 receptors with prazosin, or stimulating alpha-2A receptors with guanfacine or clonidine can be useful in reducing the symptoms of PTSD. Placebo-controlled trials have shown that prazosin is helpful in veterans, active duty soldiers and civilians with PTSD, including improvement of PFC symptoms such as impaired concentration and impulse control. Open label studies suggest that guanfacine may be especially helpful in treating children and adolescents who have experienced trauma. Thus, understanding the neurobiology of the stress response has begun to help patients with stress disorders. Research in animals has revealed how prefrontal cortex goes “off-line” during stress. Prefrontal cortical function is protected by α2A-, but impaired by α1-adrenoceptors. Based on this research, α1 blockers and α2A agonists are now in use to treat PTSD.
Collapse
|
34
|
α(2A)-adrenergic receptors filter parabrachial inputs to the bed nucleus of the stria terminalis. J Neurosci 2014; 34:9319-31. [PMID: 25009265 DOI: 10.1523/jneurosci.0822-14.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
α2-adrenergic receptors (AR) within the bed nucleus of the stria terminalis (BNST) reduce stress-reward interactions in rodent models. In addition to their roles as autoreceptors, BNST α(2A)-ARs suppress glutamatergic transmission. One prominent glutamatergic input to the BNST originates from the parabrachial nucleus (PBN) and consists of asymmetric axosomatic synapses containing calcitonin gene-related peptide (CGRP) and vGluT2. Here we provide immunoelectron microscopic data showing that many asymmetric axosomatic synapses in the BNST contain α(2A)-ARs. Further, we examined optically evoked glutamate release ex vivo in BNST from mice with virally delivered channelrhodopsin2 (ChR2) expression in PBN. In BNST from these animals, ChR2 partially colocalized with CGRP, and activation generated EPSCs in dorsal anterolateral BNST neurons that elicited two cell-type-specific outcomes: (1) feedforward inhibition or (2) an EPSP that elicited firing. We found that the α(2A)-AR agonist guanfacine selectively inhibited this PBN input to the BNST, preferentially reducing the excitatory response in ex vivo mouse brain slices. To begin to assess the overall impact of α(2A)-AR control of this PBN input on BNST excitatory transmission, we used a Thy1-COP4 mouse line with little postsynaptic ChR2 expression nor colocalization of ChR2 with CGRP in the BNST. In slices from these mice, we found that guanfacine enhanced, rather than suppressed, optogenetically initiated excitatory drive in BNST. Thus, our study reveals distinct actions of PBN afferents within the BNST and suggests that α(2A)-AR agonists may filter excitatory transmission in the BNST by inhibiting a component of the PBN input while enhancing the actions of other inputs.
Collapse
|
35
|
Fox HC, Morgan PT, Sinha R. Sex differences in guanfacine effects on drug craving and stress arousal in cocaine-dependent individuals. Neuropsychopharmacology 2014; 39:1527-37. [PMID: 24395021 PMCID: PMC3988558 DOI: 10.1038/npp.2014.1] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 11/08/2013] [Accepted: 11/11/2013] [Indexed: 12/12/2022]
Abstract
Currently, no FDA-approved medication exists for the treatment of cocaine use disorder. Furthermore, as women become increasingly more at risk for the consequences of cocaine addiction, the need to establish better-tailored treatment medications is paramount. We examine the effects of the alpha2 adrenergic agonist, guanfacine HCl, on responses to stress and drug cue in a group of cocaine-dependent men and women who also abuse alcohol and nicotine. Forty early abstinent treatment-seeking cocaine-dependent males and females were randomly assigned to receive either daily placebo (12 M/7 F) or guanfacine (2 or 3 mg) (15 M/6 F) for 3 weeks. In week 4, they participated in a laboratory experiment and were exposed to three 10-min guided imagery conditions (stress/stress, cue/cue, and stress/cue), one per day, consecutively in a random, counterbalanced order. Craving, negative emotion, anxiety, and cardiovascular function were assessed at baseline, immediately following imagery exposure, and at various recovery time points. Guanfacine significantly attenuated cocaine craving, alcohol craving, anxiety, and negative emotion following exposure to all three imagery conditions in females, but not males. Guanfacine did, however, reduce sympathetic tone as well as stress and cue-induced nicotine craving and systolic blood pressure (SBP) in both males and females. These findings highlight sex-specific effects of guanfacine on drug craving, anxiety, and negative mood with significant effects in women and not men. The findings suggest further evaluation of guanfacine in the treatment of cocaine use disorder with a specific focus on sex differences in treatment response.
Collapse
Affiliation(s)
- Helen C Fox
- Department of Psychiatry, The Connecticut Mental Health Center, Yale University School of Medicine, New Haven, CT, USA
| | - Peter T Morgan
- Department of Psychiatry, The Connecticut Mental Health Center, Yale University School of Medicine, New Haven, CT, USA
| | - Rajita Sinha
- Department of Psychiatry, The Yale Stress Center, Yale University School of Medicine, New Haven, CT, USA,Department of Psychiatry, The Yale Stress Center, Yale University School of Medicine, 2 Church Street South Suite 209, New Haven, CT 06519, USA, Tel: +1 203 737 5805, Fax: +1 203 974 7076, E-mail:
| |
Collapse
|
36
|
Ghitza UE. ASPIRE Model for Treating Cannabis and Other Substance Use Disorders: A Novel Personalized-Medicine Framework. Front Psychiatry 2014; 5:180. [PMID: 25538635 PMCID: PMC4258994 DOI: 10.3389/fpsyt.2014.00180] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 11/25/2014] [Indexed: 01/23/2023] Open
Affiliation(s)
- Udi E Ghitza
- Center for the Clinical Trials Network, National Institute on Drug Abuse, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|