1
|
Acuña AM, Nagy EK, Legg JL, Rodarte SE, Olive MF. Characterization of serum and brain cytokine levels following prolonged binge-like methamphetamine self-administration and cued methamphetamine seeking. J Neuroimmunol 2025; 400:578530. [PMID: 39854936 PMCID: PMC11875959 DOI: 10.1016/j.jneuroim.2025.578530] [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: 08/27/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Methamphetamine (METH) use is associated with peripheral and brain inflammation that can contribute to METH-associated toxicity and heightened cue reactivity. However, the persistence of these phenomena, especially with regards to changes in brain proinflammatory cytokine levels, is not yet clear. In this study, we determined the effects of repeated binge-like METH self-administration (96-h/week for 3 weeks) followed by cued drug seeking for up to 60 days into abstinence in male and female rats. Serum cytokine levels were assessed prior to cued drug seeking tests on days 21 and 60 of abstinence, and cytokine levels in the prefrontal cortex (PFC) and dorsal striatum (DStr) were assessed on the day following that last cued seeking test. We observed robust levels of METH intake in both sexes as well as a gradual increase in magnitude of METH seeking across abstinence that did not differ between sexes. Magnitude of METH seeking on days 10 and 60 were positively correlated with prior total drug intake. Sex- and region-dependent changes in various chemokines and interleukins were observed in the PFC and DStr, as were sex- and time-dependent changes in serum cytokine levels, with the largest number of cytokines altered on day 60 in male animals. Serum levels of IL-6 were positively correlated with brain levels of this cytokine, but serum levels of this and other cytokines did not correlate with the magnitude of METH seeking. These findings suggest that binge-like METH intake produces persistent yet divergent central and peripheral immune responses that extend well into abstinence.
Collapse
Affiliation(s)
- Amanda M Acuña
- Department of Psychology, Arizona State University, Tempe, AZ 85257, USA; Interdisciplinary Graduate Program in Neuroscience, School of Life Sciences, Arizona State University, Tempe, AZ 85257, USA
| | - Erin K Nagy
- Department of Psychology, Arizona State University, Tempe, AZ 85257, USA
| | - Justin L Legg
- Department of Psychology, Arizona State University, Tempe, AZ 85257, USA
| | - Serena E Rodarte
- Department of Psychology, Arizona State University, Tempe, AZ 85257, USA
| | - M Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ 85257, USA; Interdisciplinary Graduate Program in Neuroscience, School of Life Sciences, Arizona State University, Tempe, AZ 85257, USA.
| |
Collapse
|
2
|
Raymond JS, Athanasopoulos AG, Badolato CJ, Doolan TJ, Scicluna RL, Everett NA, Bowen MT, James MH. Emerging medications and pharmacological treatment approaches for substance use disorders. Pharmacol Biochem Behav 2025; 248:173952. [PMID: 39719161 PMCID: PMC12078786 DOI: 10.1016/j.pbb.2024.173952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Medications to treat substance use disorders (SUDs) remain suboptimal or, in the case of stimulants and cannabis, non-existent. Many factors have contributed to this paucity, including the biological complexity of addiction, regulatory challenges, and a historical lack of enthusiasm among pharmaceutical companies to commit resources to this disease space. Despite these headwinds, the recent opioid crisis has highlighted the devastating consequences of SUDs for both individuals and society, stimulating urgent efforts to identify novel treatment approaches. In addition, several neurobiological systems have been recently implicated in unique aspects of drug reward, opening the door to candidate medications with novel mechanisms of action. Here, we provide an overview of efforts to target several of these new systems, with a focus on those that are the subject of ongoing clinical trials as well as being areas of interest among the authors' research groups (MHJ, MTB, NAE). Specifically, we discuss new classes of medications targeting the serotonin 2A receptor (i.e., psychedelics), glucagon-like peptide 1 receptor, cannabidiol, dynorphin/kappa opioid receptor, orexin/hypocretin, and oxytocin receptor systems, as well as emergent approaches for modulating the more canonical dopaminergic system via agonist therapies for stimulant use disorders. Collectively, innovations in this space give reason for optimism for an improved therapeutic landscape for substance use disorders in the near future.
Collapse
Affiliation(s)
- Joel S Raymond
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA; Rutgers Addiction Research Center, Brain Health Institute, Rutgers Health, Piscataway, NJ, USA
| | - Alexander G Athanasopoulos
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Connie J Badolato
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Tylah J Doolan
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Rhianne L Scicluna
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas A Everett
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Michael T Bowen
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Morgan H James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA; Rutgers Addiction Research Center, Brain Health Institute, Rutgers Health, Piscataway, NJ, USA; School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
3
|
Chow JJ, Pitts KM, Negishi K, Madangopal R, Dong Y, Wolf ME, Shaham Y. Neurobiology of the incubation of drug craving: An update. Pharmacol Rev 2025; 77:100022. [PMID: 40148031 PMCID: PMC11964951 DOI: 10.1016/j.pharmr.2024.100022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/21/2024] [Indexed: 03/29/2025] Open
Abstract
Relapse to drug use is often preceded by drug craving. Clinical observations in the 1980s led clinical investigators to postulate that cue-induced cocaine craving may increase during abstinence. Over 2 decades ago, investigators identified an analogous phenomenon in rats of time-dependent increases in drug-seeking behavior during homecage abstinence and termed it incubation of cocaine craving. In 2011, we reviewed the first decade of studies on brain mechanisms of incubation of drug craving. In this review, we provide an update on incubation-related brain mechanisms from studies published since 2011. We first review studies using the standard method of incubation after homecage-forced abstinence from cocaine, methamphetamine, opioid drugs, and nicotine. Next, we review studies using newer methods to study incubation after voluntary abstinence in the drug environment. In these studies, abstinence is achieved by either providing rats alternative nondrug rewards in a choice setting or introducing rats to adverse consequences to drug seeking or taking. We then discuss translational human studies on incubation of cue-induced drug craving. We conclude by discussing several emerging topics, including sex differences in incubation of drug craving, role of sleep patterns, and similarities and differences in mechanisms of incubation of craving across drug classes and abstinence conditions. Our 2 main conclusions are as follows: (1) across drug classes, there are both similarities and differences in mechanisms of incubation of drug craving after forced abstinence, and (2) the method used to achieve abstinence (forced or voluntary) can influence the mechanisms controlling incubation of drug craving or its expression. SIGNIFICANCE STATEMENT: This article reviews results from preclinical and clinical studies published since 2011 on neurobiological mechanisms of incubation of drug craving after homecage-forced abstinence or voluntary abstinence in the drug environment. This article also reviews translational human studies on incubation of cue-induced subjective drug craving and brain response during abstinence. The results of the studies reviewed indicate that multiple brain mechanisms control incubation of drug craving after homecage-forced abstinence or voluntary abstinence.
Collapse
Affiliation(s)
- Jonathan J Chow
- Behavioral Neuroscience Research Branch, IRP/NIDA/NIH, Baltimore, Maryland.
| | - Kayla M Pitts
- Behavioral Neuroscience Research Branch, IRP/NIDA/NIH, Baltimore, Maryland
| | - Kenichiro Negishi
- Behavioral Neuroscience Research Branch, IRP/NIDA/NIH, Baltimore, Maryland
| | | | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Yavin Shaham
- Behavioral Neuroscience Research Branch, IRP/NIDA/NIH, Baltimore, Maryland.
| |
Collapse
|
4
|
Ramli FF, Rejeki PS, Ibrahim N'I, Abdullayeva G, Halim S. A Mechanistic Review on Toxicity Effects of Methamphetamine. Int J Med Sci 2025; 22:482-507. [PMID: 39898237 PMCID: PMC11783064 DOI: 10.7150/ijms.99159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 11/26/2024] [Indexed: 02/04/2025] Open
Abstract
Persistent methamphetamine use causes many toxic effects in various organs, including the brain, heart, liver, kidney and eyes. The extent of its toxicity depends on numerous pharmacological factors, including route of administration, dose, genetic polymorphism related to drug metabolism and polysubstance abuse. Several molecular pathways have been proposed to activate oxidative stress, inflammation and apoptosis: B-cell lymphoma protein 2 (Bcl-2)-associated X (Bax)/Bcl2/caspase-3, nuclear factor erythroid 2-related factor (Nrf2)/heme oxygenase-1 (HO-1), protein kinase B (Akt)/mammalian target of rapamycin (mTOR)/p70S6K, trace amine-associated receptor 1 (TAAR1)/cAMP/lysyl oxidase, Sigmar1/ cAMP response element-binding protein (CREB)/mitochondrial fission-1 protein (Fis1), NADPH-Oxidase-2 (NOX-2), renal autophagy pathway, vascular endothelial growth factor (VEGF)/phosphatidylinositol-3-kinase (PI3K)/ protein kinase B (Akt)/endothelial nitric oxide synthase (eNOS), Nupr1/Chop/P53/PUMA/Beclin1 and Toll-like receptor (TLR)4/MyD88/TRAF6 pathways. The activation promotes pathological changes, including the disruption of the blood-brain barrier, myocardial infarction, cardiomyopathy, acute liver failure, acute kidney injury, chronic kidney disease, keratitis, retinopathy and vision loss. This review revisits the pharmacological profiles of methamphetamine and its effects on the brain, heart, liver, eyes, kidneys and endothelium. Understanding the mechanisms of methamphetamine toxicity is essential in developing treatment strategies to reverse or attenuate the progress of methamphetamine-associated organ damage.
Collapse
Affiliation(s)
- Fitri Fareez Ramli
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Kuala Lumpur, Malaysia
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Purwo Sri Rejeki
- Physiology Division, Department of Medical Physiology and Biochemistry, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Nurul 'Izzah Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Kuala Lumpur, Malaysia
| | - Gulnar Abdullayeva
- Old Road Campus Research Building, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
- Institute of Molecular Biology & Biotechnologies, Ministry of Science and Education of the Republic of Azerbaijan, 11 Izzat Nabiyev Str., AZ1073, Baku, Azerbaijan
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, OX3 7TY, UK
| | - Shariff Halim
- Faculty of Health Sciences, Universiti Teknologi MARA Cawangan Pulau Pinang, Kampus Bertam, 13200 Pulau Pinang, Malaysia
| |
Collapse
|
5
|
Song SH, Kim S, Jang WJ, Ryu IS, Jeong CH, Lee S. Exploring the progression of drug dependence in a methamphetamine self-administration rat model through targeted and non-targeted metabolomics analyses. Sci Rep 2024; 14:22543. [PMID: 39343795 PMCID: PMC11439939 DOI: 10.1038/s41598-024-73247-5] [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: 06/14/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
Persistent neurochemical and biological disturbances resulting from repeated cycles of drug reward, withdrawal, and relapse contribute to drug dependence. Methamphetamine (MA) is a psychostimulant with substantial abuse potential and neurotoxic effects, primarily affecting monoamine neurotransmitter systems in the brain. In this study, we aimed to explore the progression of drug dependence in rat models of MA self-administration, extinction, and reinstatement through targeted and non-targeted metabolomics analyses. Metabolic profiles were examined in rat plasma during the following phases: after 16 days of MA self-administration (Group M); after 16 days of self-administration followed by 14 days of extinction (Group MS); and after self-administration and extinction followed by a reinstatement injection of MA (Group MSM). Each group of MA self-administration, extinction, and reinstatement induces distinct changes in the metabolic pathways, particularly those related to the TCA cycle, arginine and proline metabolism, and arginine biosynthesis. Additionally, the downregulation of glycerophospholipids and sphingomyelins in Group MSM suggests their potential role in MA reinstatement. These alterations may signify the progressive deterioration of these metabolic pathways, possibly contributing to drug dependence following repeated cycles of drug reward, withdrawal, and relapse. These results provide valuable insights into the metabolic changes associated with MA use at various stages, potentially facilitating the discovery of early diagnostic biomarkers and therapeutic targets for MA use disorders.
Collapse
Affiliation(s)
- Sang-Hoon Song
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - Suji Kim
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - Won-Jun Jang
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - In Soo Ryu
- Biorchestra Co., Ltd, Techno4-ro 17, Daejeon, 34013, Republic of Korea
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu, 42601, Republic of Korea.
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu, 42601, Republic of Korea.
| |
Collapse
|
6
|
Yates JR. Pharmacological Treatments for Methamphetamine Use Disorder: Current Status and Future Targets. Subst Abuse Rehabil 2024; 15:125-161. [PMID: 39228432 PMCID: PMC11370775 DOI: 10.2147/sar.s431273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024] Open
Abstract
The illicit use of the psychostimulant methamphetamine (METH) is a major concern, with overdose deaths increasing substantially since the mid-2010s. One challenge to treating METH use disorder (MUD), as with other psychostimulant use disorders, is that there are no available pharmacotherapies that can reduce cravings and help individuals achieve abstinence. The purpose of the current review is to discuss the molecular targets that have been tested in assays measuring the physiological, the cognitive, and the reinforcing effects of METH in both animals and humans. Several drugs show promise as potential pharmacotherapies for MUD when tested in animals, but fail to produce long-term changes in METH use in dependent individuals (eg, modafinil, antipsychotic medications, baclofen). However, these drugs, plus medications like atomoxetine and varenicline, may be better served as treatments to ameliorate the psychotomimetic effects of METH or to reverse METH-induced cognitive deficits. Preclinical studies show that vesicular monoamine transporter 2 inhibitors, metabotropic glutamate receptor ligands, and trace amine-associated receptor agonists are efficacious in attenuating the reinforcing effects of METH; however, clinical studies are needed to determine if these drugs effectively treat MUD. In addition to screening these compounds in individuals with MUD, potential future directions include increased emphasis on sex differences in preclinical studies and utilization of pharmacogenetic approaches to determine if genetic variances are predictive of treatment outcomes. These future directions can help lead to better interventions for treating MUD.
Collapse
Affiliation(s)
- Justin R Yates
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY, USA
| |
Collapse
|
7
|
Lin H, Olaniran A, Garmchi S, Firlie J, Rincon N, Li X. The estrous cycle has no effect on incubation of methamphetamine craving and associated Fos expression in dorsomedial striatum and anterior intralaminar nucleus of thalamus. ADDICTION NEUROSCIENCE 2024; 11:100158. [PMID: 38938268 PMCID: PMC11210321 DOI: 10.1016/j.addicn.2024.100158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Relapse is a major challenge in treating drug addiction, and drug seeking progressively increases after abstinence, a phenomenon termed "incubation of drug craving". Previous studies demonstrated both sex differences and an effect of estrous cycle in female rats in incubation of cocaine craving. In contrast, while incubation of methamphetamine craving is similar across sexes, whether estrous cycle plays a role in this incubation has yet to be fully addressed. Moreover, whether neural mechanisms underlying incubation of methamphetamine craving differ across estrous cycles is largely unknown. To address these gaps, we first compared methamphetamine self-administration, and methamphetamine seeking on both abstinence days 1 and 28 between male rats and female rats across the estrous cycle. Next, we examined neuronal activation associated with incubated methamphetamine seeking in dorsomedial striatum (DMS) and lateral portion of the anterior intralaminar nucleus of thalamus (AIT-L), two brain areas previously implicated in incubation of methamphetamine craving. We found no effect of sex or estrous cycle on methamphetamine self-administration and methamphetamine seeking on abstinence days 1 and 28. We also found no effect of sex or estrous cycle on the number of Fos-expressing cells in DMS or AIT-L following methamphetamine seeking test. Taken together, our results showed that methamphetamine self-administration and incubation of methamphetamine craving was not dependent on sex or estrous cycles under our experimental condition, and the role of DMS and AIT-L in incubation of methamphetamine craving may be similar across sexes and across estrous cycles in female rats.
Collapse
Affiliation(s)
- Hongyu Lin
- Department of Psychology, University of Maryland College Park, College Park, MD 20742, USA
| | - Adedayo Olaniran
- Department of Psychology, University of Maryland College Park, College Park, MD 20742, USA
| | - Sara Garmchi
- Department of Psychology, University of Maryland College Park, College Park, MD 20742, USA
| | - Julia Firlie
- Department of Psychology, University of Maryland College Park, College Park, MD 20742, USA
| | - Natalia Rincon
- Department of Psychology, University of Maryland College Park, College Park, MD 20742, USA
| | - Xuan Li
- Department of Psychology, University of Maryland College Park, College Park, MD 20742, USA
- Program in Neuroscience and Cognitive Science, University of Maryland College Park, College Park, MD 20742, USA
| |
Collapse
|
8
|
Lin CW, Cheng MH, Fan CH, Chen HH, Yeh CK. Focused ultrasound stimulation of infralimbic cortex attenuates reinstatement of methamphetamine-induced conditioned place preference in rats. Neurotherapeutics 2024; 21:e00328. [PMID: 38355360 PMCID: PMC10937235 DOI: 10.1016/j.neurot.2024.e00328] [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] [Received: 10/24/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Methamphetamine (MA) use disorder poses significant challenges to both the affected individuals and society. Current non-drug therapies like transcranial direct-current stimulation and transcranial magnetic stimulation have limitations due to their invasive nature and limited reach to deeper brain areas. Transcranial focused ultrasound (FUS) is gaining attention as a noninvasive option with precise spatial targeting, able to affect deeper areas of the brain. This research focused on assessing the effectiveness of FUS in influencing the infralimbic cortex (IL) to prevent the recurrence of MA-seeking behavior, using the conditioned place preference (CPP) method in rats. The study involved twenty male Sprague-Dawley rats. Neuronal activation by FUS was first examined via electromyography (EMG). Rats received alternately with MA or saline, and confined to one of two distinctive compartments in a three compartment apparatus over a 4-day period. After CPP test, extinction, the first reinstatement, and extinction again, FUS was applied to IL prior to the second MA priming-induced reinstatement. Safety assessments were conducted through locomotor and histological function examinations. EMG data confirmed the effectiveness of FUS in activating neurons. Significant attenuation of reinstatement of MA CPP was found, along with successful targeting of the IL region, confirmed through acoustic field scanning, c-Fos immunohistochemistry, and Evans blue dye staining. No damage to brain tissue or impaired locomotor activity was observed. The results of the study indicate that applying FUS to the IL markedly reduced the recurrence of MA seeking behavior, without harming brain tissue or impairing motor skills. This suggests that FUS could be a promising method for treating MA use disorder, with the infralimbic cortex being an effective target for FUS in preventing MA relapse.
Collapse
Affiliation(s)
- Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Min-Hsuan Cheng
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Hwei-Hsien Chen
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan; Institute of Neuroscience, National Chengchi University, Taipei, Taiwan; Graduate Program for Aging, China Medical University, Taichung, Taiwan.
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
9
|
Daiwile AP, Cadet JL. Modeling methamphetamine use disorder in mammals: Sex differences in behavioral, biochemical, and transcriptional consequences. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 99:145-168. [PMID: 38467480 PMCID: PMC11474929 DOI: 10.1016/bs.apha.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Methamphetamine (METH) is the most commonly misused amphetamine-type stimulant throughout the globe. METH is very rewarding, and its misuse can lead to a diagnosis of METH use disorder (MUD). Although METH use is observed in both sexes, there are, however, reported differences in the clinical manifestations of METH use and its consequences. These observations indicate the need for more research on the long-term sex-dependent consequences of METH taking in both preclinical and clinical settings. In effect, sex is a biological variable that can impact conclusions drawn from various basic and clinical studies. Thus, the present chapter provides a succinct review of the current state of the research on METH and its sex-associated consequences. In addition to behavioral and cognitive aspects of METH use, we discuss METH-induced changes in neurotransmitter systems and structures in the brain. Thus, the book chapter serves to highlight the significance of sex as a critical element that needs to be considered during discussions of novel therapeutic approaches to MUD.
Collapse
Affiliation(s)
- Atul P Daiwile
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD, United States
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD, United States.
| |
Collapse
|
10
|
Elhadi K, Daiwile AP, Cadet JL. Modeling methamphetamine use disorder and relapse in animals: short- and long-term epigenetic, transcriptional., and biochemical consequences in the rat brain. Neurosci Biobehav Rev 2023; 155:105440. [PMID: 38707245 PMCID: PMC11068368 DOI: 10.1016/j.neubiorev.2023.105440] [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: 07/24/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 05/07/2024]
Abstract
Methamphetamine use disorder (MUD) is a neuropsychiatric disorder characterized by binge drug taking episodes, intervals of abstinence, and relapses to drug use even during treatment. MUD has been modeled in rodents and investigators are attempting to identify its molecular bases. Preclinical experiments have shown that different schedules of methamphetamine self-administration can cause diverse transcriptional changes in the dorsal striatum of Sprague-Dawley rats. In the present review, we present data on differentially expressed genes (DEGs) identified in the rat striatum following methamphetamine intake. These include genes involved in transcription regulation, potassium channel function, and neuroinflammation. We then use the striatal data to discuss the potential significance of the molecular changes induced by methamphetamine by reviewing concordant or discordant data from the literature. This review identified potential molecular targets for pharmacological interventions. Nevertheless, there is a need for more research on methamphetamine-induced transcriptional consequences in various brain regions. These data should provide a more detailed neuroanatomical map of methamphetamine-induced changes and should better inform therapeutic interventions against MUD.
Collapse
Affiliation(s)
- Khalid Elhadi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| | - Atul P. Daiwile
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| |
Collapse
|
11
|
Hersey M, Bacon AK, Bailey LG, Lee MR, Chen AY, Leggio L, Tanda G. Oxytocin receptors mediate oxytocin potentiation of methylphenidate-induced stimulation of accumbens dopamine in rats. J Neurochem 2023; 164:613-623. [PMID: 36420597 PMCID: PMC10766115 DOI: 10.1111/jnc.15730] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022]
Abstract
While the illicit use and misuse of stimulants like cocaine and methylphenidate (MP) has increased, there remains no FDA-approved treatments for psychostimulant use disorders (PSUD). Oxytocin (OT) has shown promise as a potential pharmacotherapy for PSUD. Dopamine (DA) neurotransmission plays a significant role in PSUD. We have recently shown that OT blunts the reinforcing effects of MP but, surprisingly, enhanced MP-induced stimulation of DA levels. Such effects have been suggested as a result of activation of OT receptors or, alternatively, could be mediated by direct actions of OT on MP blockade of the DA transporter. Here, we employed fast scan cyclic voltammetry (FSCV) to investigate the effects of systemic OT on MP-induced changes in the dynamics of DA, phasic release and uptake, in the nucleus accumbens shell (NAS) of Sprague-Dawley rats. We also tested the systemic effects of an antagonist of OT receptors, atosiban, to counteract the OT enhancement of dopaminergic effects of MP under microdialysis procedures in the NAS in rats. Administration of OT alone (2 mg/kg; i.p.) did not significantly modify evoked NAS DA dynamics measured by FSCV, and when administered 10 min before MP (0.1, 0.3, 1.0 mg/kg; i.v.), OT did not potentiate MP-induced increases in phasic DA release and did not alter DA clearance rate, suggesting no direct interactions of OT with the MP-induced blockade of DA uptake. Also, OT alone did not elicit significant changes in tonic, extracellular NAS DA levels measured by microdialysis. However, consistent with previous studies, we observed that OT pretreatments (2 mg/kg; i.p.) potentiated MP-induced (0.1, 0.3, 1.0 mg/kg; i.v.) efflux of extracellular NAS DA levels. This effect was abolished when rats were pretreated with atosiban (2 mg/kg; i.p.), suggesting that OT receptors mediate this OT action. Overall, our results suggest that OT receptors mediated OT potentiation of MP-induced stimulation of extracellular NAS DA levels, likely driven by modulation of DA receptor signaling pathways, without affecting MP blockade of DAT.
Collapse
Affiliation(s)
| | | | | | - Mary R. Lee
- Veterans Affairs Medical Center, Washington, DC
| | - Andy Y. Chen
- Medication Development Program, NIDA IRP, Baltimore, MD
| | - Lorenzo Leggio
- Medication Development Program, NIDA IRP, Baltimore, MD
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, NIDA/NIAAA IRP, Baltimore, MD
| | | |
Collapse
|
12
|
Funke JR, Hwang EK, Wunsch AM, Baker R, Engeln KA, Murray CH, Milovanovic M, Caccamise AJ, Wolf ME. Persistent Neuroadaptations in the Nucleus Accumbens Core Accompany Incubation of Methamphetamine Craving in Male and Female Rats. eNeuro 2023; 10:ENEURO.0480-22.2023. [PMID: 36792361 PMCID: PMC10016192 DOI: 10.1523/eneuro.0480-22.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 02/17/2023] Open
Abstract
Relapse is a major problem in treating methamphetamine use disorder. "Incubation of craving" during abstinence is a rat model for persistence of vulnerability to craving and relapse. While methamphetamine incubation has previously been demonstrated in male and female rats, it has not been demonstrated after withdrawal periods greater than 51 d and most mechanistic work used males. Here, we address both gaps. First, although methamphetamine intake was higher in males during self-administration training (6 h/d × 10 d), incubation was similar in males and females, with "incubated" craving persisting through withdrawal day (WD)100. Second, using whole-cell patch-clamp recordings in medium spiny neurons (MSNs) of the nucleus accumbens (NAc) core, we assessed synaptic levels of calcium-permeable AMPA receptors (CP-AMPARs), as their elevation is required for expression of incubation in males. In both sexes, compared with saline-self-administering controls, CP-AMPAR levels were significantly higher in methamphetamine rats across withdrawal, although this was less pronounced in WD100-135 rats than WD15-35 or WD40-75 methamphetamine rats. We also examined membrane properties and NMDA receptor (NMDAR) transmission. In saline controls, MSNs from males exhibited lower excitability than females. This difference was eliminated after incubation because of increased excitability of MSNs from males. NMDAR transmission did not differ between sexes and was not altered after incubation. In conclusion, incubation persists for longer than previously described and equally persistent CP-AMPAR plasticity in NAc core occurs in both sexes. Thus, abstinence-related synaptic plasticity in NAc is similar in males and females although other methamphetamine-related behaviors and neuroadaptations show differences.
Collapse
Affiliation(s)
- Jonathan R Funke
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
| | - Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
| | - Amanda M Wunsch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Raines Baker
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
| | - Kimberley A Engeln
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
| | - Conor H Murray
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Mike Milovanovic
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Aaron J Caccamise
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97212
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| |
Collapse
|
13
|
Wang Y, Wang M, Xie B, Wen D, Li W, Zhou M, Wang X, Lu Y, Cong B, Ni Z, Ma C. Effects of molecular hydrogen intervention on the gut microbiome in methamphetamine abusers with mental disorder. Brain Res Bull 2023; 193:47-58. [PMID: 36516898 DOI: 10.1016/j.brainresbull.2022.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022]
Abstract
Methamphetamine (METH) is a potent and highly addictive psychostimulant and one of the most widely used illicit drugs, the abuse of which has become a severe public health problem worldwide. A growing amount of evidence has indicated potential connections between gut microbiota and mental disorders induced by METH and associations with neural and metabolic pathways. The present study aimed to explore the relationship between fecal microbial alterations and neuropsychiatric diseases in METH addictions. Thus, mental disorders and gut microbial alterations were analyzed by self-rating depression (SDS) and anxiety (SAS) scales and 16 S rRNA gene sequencing, respectively. Our results showed that increased SDS and SAS indices and decreased alpha diversity indicated more serious mental disorders and lower bacterial diversity in METH users than in the age-matched healthy control group. The gut microbial composition in female METH users was also significantly altered, with reductions in hydrogen-producing bacteria, including Bacteroides and Roseburia. Molecular hydrogen (H2) is spontaneously produced by intestinal bacteria in the process of anaerobic metabolism, which is the main pathway for H2 production in vivo. Numerous studies have shown that hydrogen intervention can significantly improve neuropsychiatric diseases, including Alzheimer's disease and Parkinson's disease. Our results showed that hydrogen intervention, including drinking and inhaling, significantly alleviated mental disorders induced by METH abuse, and the inhalation of hydrogen also altered gut microbiota profiles in the METH abusers. These results suggest that hydrogen intervention has potential therapeutic applicability in the treatment of mental disorders in METH abusers.
Collapse
Affiliation(s)
- Yong Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Mengmeng Wang
- Affiliated Hospital of Hebei University, College of Clinical Medicine, Hebei University, Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Baoding, Hebei Province 071000, PR China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Wenbo Li
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Meiqi Zhou
- College of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, Hebei Province 050017, PR China
| | - Xintao Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Yun Lu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China
| | - Zhiyu Ni
- Affiliated Hospital of Hebei University, College of Clinical Medicine, Hebei University, Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Baoding, Hebei Province 071000, PR China.
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei Province 050017, PR China.
| |
Collapse
|
14
|
Bachtell RK, Larson TA, Winkler MC. Adenosine receptor stimulation inhibits methamphetamine-associated cue seeking. J Psychopharmacol 2023; 37:192-203. [PMID: 36629009 DOI: 10.1177/02698811221147157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Methamphetamine (METH) is a psychostimulant drug that remains a popular and threatening drug of abuse with high abuse liability. There is no established pharmacotherapy to treat METH dependence, but evidence suggests that stimulation of adenosine receptors reduces the reinforcing properties of METH and could be a potential pharmacological target. This study examines the effects of adenosine receptor subtype stimulation on METH seeking using both a cue-induced reinstatement and cue-craving model of relapse. METHODS Male and female rats were trained to self-administer METH during daily 2-h sessions. Cue-induced reinstatement of METH seeking was evaluated after extinction training. A systemic pretreatment of an adenosine A1 receptor (A1R) or A2A receptor (A2AR) agonist was administered prior to an extinction or cue session to evaluate the effects of adenosine receptor subtype stimulation on METH seeking. The effects of a systemic pretreatment of A1R or A2AR agonists were also evaluated in a cue-craving model where the cued-seeking test was conducted after 21 days of forced home-cage abstinence without extinction training. RESULTS Cue-induced reinstatement was reduced in both male and female rats that received A1R or A2AR agonist pretreatments. Similarly, an A1R or A2AR agonist pretreatment also inhibited cue craving in both male and female rats. CONCLUSION Stimulation of either adenosine A1R or A2AR subtypes inhibits METH-seeking behavior elicited by METH-associated cues. These effects may be attributed to the ability of A1R and A2AR stimulation to disrupt cue-induced dopamine and glutamate signaling throughout the brain.
Collapse
Affiliation(s)
- Ryan K Bachtell
- Department of Psychology and Neuroscience and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Tracey A Larson
- Department of Psychology and Neuroscience and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Madeline C Winkler
- Department of Psychology and Neuroscience and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
15
|
Specific Inhibition of Interpeduncular Nucleus GABAergic Neurons Alleviates Anxiety-Like Behaviors in Male Mice after Prolonged Abstinence from Methamphetamine. J Neurosci 2023; 43:803-811. [PMID: 36564185 PMCID: PMC9899084 DOI: 10.1523/jneurosci.1767-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Anxiety is one of the most common withdrawal symptoms of methamphetamine (METH) abuse, which further drives relapse to drugs. Interpeduncular nucleus (IPN) has been implicated in anxiety-like behaviors and addiction, yet its role in METH-abstinence-induced anxiety remains unknown. Here, we found that prolonged abstinence from METH enhanced anxiety-like behaviors in male mice, accompanied by more excited IPN GABAergic neurons, as indicated by the increased c-fos expression and the enhanced neuronal excitability by electrophysiological recording in the GABAergic neurons. Using the designer receptors exclusively activated by designer drugs method, specific inhibition of IPN GABAergic neurons rescued the aberrant neuronal excitation of IPN GABAergic neurons and efficiently reduced anxiety-like behaviors, whereas it did not induce depression-like behaviors in male mice after prolonged abstinence from METH. These findings reveal that IPN GABAergic neurons should be a promising brain target to alleviate late withdrawal symptoms from METH with few side effects.SIGNIFICANCE STATEMENT Prolonged abstinence from METH triggers IPN GABAergic neurons and ultimately increases anxiety in male mice. Suppressing IPN GABAergic neurons rescues METH abstinence-induced aberrant neuronal excitation of IPN GABAergic neurons and efficiently reduces anxiety in mice.
Collapse
|
16
|
Kuebler IRK, Jolton JA, Hermreck C, Hubbard NA, Wakabayashi KT. Contrasting dose-dependent effects of acute intravenous methamphetamine on lateral hypothalamic extracellular glucose dynamics in male and female rats. J Neurophysiol 2022; 128:819-836. [PMID: 36043803 PMCID: PMC9529272 DOI: 10.1152/jn.00257.2022] [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: 06/13/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
Glucose is the brain's primary energetic resource. The brain's use of glucose is dynamic, balancing delivery from the neurovasculature with local metabolism. Although glucose metabolism is known to differ in humans with and without methamphetamine use disorder (MUD), it is unknown how central glucose regulation changes with acute methamphetamine experience. Here, we determined how intravenous methamphetamine regulates extracellular glucose levels in a brain region implicated in MUD-like behavior, the lateral hypothalamus (LH). We measured extracellular LH glucose in awake adult male and female drug-naive Wistar rats using enzyme-linked amperometric glucose biosensors. Changes in LH glucose were monitored during a single session after: 1) natural nondrug stimuli (novel object presentation and a tail-touch), 2) increasing cumulative doses of intravenous methamphetamine (0.025, 0.05, 0.1, and 0.2 mg/kg), and 3) an injection of 60 mg of glucose. We found second-scale fluctuations in LH glucose in response to natural stimuli that differed by both stimulus type and sex. Although rapid, second-scale changes in LH glucose during methamphetamine injections were variable, slow, minute-scale changes following most injections were robust and resulted in a reduction in LH glucose levels. Dose and sex differences at this timescale indicated that female rats may be more sensitive to the impact of methamphetamine on central glucose regulation. These findings suggest that the effects of MUD on healthy brain function may be linked to how methamphetamine alters extracellular glucose regulation in the LH and point to possible mechanisms by which methamphetamine influences central glucose metabolism more broadly.NEW & NOTEWORTHY Enzyme-linked glucose biosensors were used to monitor lateral hypothalamic (LH) extracellular fluctuations during nondrug stimuli and intravenous methamphetamine injections in drug-naive awake male and female rats. Second-scale glucose changes occurred after nondrug stimuli, differing by modality and sex. Robust minute-scale decreases followed most methamphetamine injections. Sex differences at the minute-scale indicate female central glucose regulation is more sensitive to methamphetamine effects. We discuss likely mechanisms underlying these fluctuations, and their implications in methamphetamine use disorder.
Collapse
Affiliation(s)
- Isabel R K Kuebler
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Joshua A Jolton
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Chase Hermreck
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Nicholas A Hubbard
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Ken T Wakabayashi
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| |
Collapse
|
17
|
Mayberry HL, Bavley CC, Karbalaei R, Peterson DR, Bongiovanni AR, Ellis AS, Downey SH, Toussaint AB, Wimmer ME. Transcriptomics in the nucleus accumbens shell reveal sex- and reinforcer-specific signatures associated with morphine and sucrose craving. Neuropsychopharmacology 2022; 47:1764-1775. [PMID: 35190706 PMCID: PMC9372067 DOI: 10.1038/s41386-022-01289-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/16/2022]
Abstract
Incubation of craving is a well-documented phenomenon referring to the intensification of drug craving over extended abstinence. The neural adaptations that occur during forced abstinence following chronic drug taking have been a topic of intense study. However, little is known about the transcriptomic changes occurring throughout this window of time. To define gene expression changes associated with morphine consumption and extended abstinence, male and female rats underwent 10 days of morphine self-administration. Separate drug-naive rats self-administered sucrose in order to compare opioid-induced changes from those associated with natural, non-drug rewards. After one or 30 days of forced abstinence, rats were tested for craving, or nucleus accumbens shell tissue was dissected for RNA sequencing. Morphine consumption was predictive of drug seeking after extended (30 days) but not brief (1 day) abstinence in both sexes. Extended abstinence was also associated with robust sex- and reinforcer-specific changes in gene expression, suggesting sex differences underlying incubation of morphine and sucrose seeking respectively. Importantly, these changes in gene expression occurred without re-exposure to drug-paired cues, indicating that chronic morphine causes long-lasting changes in gene expression that prime the system for increased craving. These findings lay the groundwork for identifying specific therapeutic targets for curbing opioid craving without impacting the natural reward system in males and females.
Collapse
Affiliation(s)
- Hannah L Mayberry
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Charlotte C Bavley
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Reza Karbalaei
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Drew R Peterson
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Angela R Bongiovanni
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Alexandra S Ellis
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Sara H Downey
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Andre B Toussaint
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Mathieu E Wimmer
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Baracz SJ, Robinson KJ, Wright AL, Turner AJ, McGregor IS, Cornish JL, Everett NA. Oxytocin as an adolescent treatment for methamphetamine addiction after early life stress in male and female rats. Neuropsychopharmacology 2022; 47:1561-1573. [PMID: 35581382 PMCID: PMC9206013 DOI: 10.1038/s41386-022-01336-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/02/2022] [Accepted: 04/27/2022] [Indexed: 11/08/2022]
Abstract
Early life stress (ELS) is associated with perturbed neural development and augmented vulnerability to mental health disorders, including addiction. How ELS changes the brain to increase addiction risk is poorly understood, and there are no therapies which target this ELS-induced vulnerability. ELS disrupts the oxytocin system, which can modulate addiction susceptibility, suggesting that targeting the oxytocin system may be therapeutic in this ELS-addiction comorbidity. Therefore, we determined whether adolescent oxytocin treatment after ELS could: (1) reduce vulnerability to anxiety, social deficits, and methamphetamine-taking and reinstatement; and (2) restore hypothalamic oxytocin and corticotropin-releasing factor expressing neurons and peripheral oxytocin and corticosterone levels. Long Evans pups underwent maternal separation (MS) for either 15 min or 360 min on postnatal days (PND) 1-21. During adolescence (PNDs 28-42), rats received a daily injection of either oxytocin or saline. In Experiment 1, adult rats were assessed using the elevated plus-maze, social interaction procedure, and methamphetamine self-administration procedure, including extinction, and cue-, methamphetamine- and yohimbine-induced reinstatement. In Experiment 2, plasma for enzyme immunoassays and brain tissue for immunofluorescence were collected from adult rats after acute stress exposure. Adolescent oxytocin treatment ameliorated ELS-induced anxiety and reduced methamphetamine- and yohimbine-induced reinstatement in both sexes, and suppressed methamphetamine intake and facilitated extinction in males only. Additionally, adolescent oxytocin treatment after ELS restored oxytocin-immunoreactive cells and stress-induced oxytocin levels in males, and attenuated stress-induced corticosterone levels in both sexes. Adolescent oxytocin treatment reverses some of the ELS effects on later-life psychopathology and vulnerability to addiction.
Collapse
Affiliation(s)
- Sarah J Baracz
- School of Psychological Sciences, Macquarie University, North Ryde, NSW, 2109, Australia.
- School of Psychology, University of Sydney, Camperdown, NSW, 2006, Australia.
- Centre for Emotional Health, Macquarie University, North Ryde, NSW, 2109, Australia.
| | - Katherine J Robinson
- School of Psychological Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Amanda L Wright
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Anita J Turner
- School of Psychological Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Iain S McGregor
- School of Psychology, University of Sydney, Camperdown, NSW, 2006, Australia
- Lambert Initiative of Cannabinoid Therapeutics, Brain and Mind Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Jennifer L Cornish
- School of Psychological Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
- Centre for Emotional Health, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Nicholas A Everett
- School of Psychological Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
- School of Psychology, University of Sydney, Camperdown, NSW, 2006, Australia
| |
Collapse
|
19
|
Daiwile AP, Jayanthi S, Cadet JL. Sex differences in methamphetamine use disorder perused from pre-clinical and clinical studies: Potential therapeutic impacts. Neurosci Biobehav Rev 2022; 137:104674. [PMID: 35452744 PMCID: PMC9119944 DOI: 10.1016/j.neubiorev.2022.104674] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 01/31/2023]
Abstract
Methamphetamine (METH) use, and misuse are associated with severe socioeconomic consequences. METH users develop tolerance, lose control over drug taking behaviors, and suffer frequent relapses even during treatment. The clinical course of METH use disorder is influenced by multifactorial METH-induced effects on the central and peripheral nervous systems. Although these METH-induced consequences are observed in humans of all ages, races, and sexes, sexual dimorphism in these outcomes have been observed in both pre-clinical and clinical settings. In this review, we have provided a detailed presentation of the sex differences reported in human and animal studies. We have therefore presented data that identified the influences of sex on METH pharmacokinetics, METH-induced changes in behaviors, cognitive processes, structural changes in the brain, and the effects of the drug on neurotransmitter systems and molecular mechanisms. Finally, we highlighted the potential significance of sex as a critical variable that should be considered when planning the development of new pharmacotherapeutic approaches against MEH use disorder in humans.
Collapse
Affiliation(s)
- Atul P Daiwile
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, USA.
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, USA.
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, USA.
| |
Collapse
|
20
|
Oxytocin, a Novel Treatment for Methamphetamine Use Disorder. Neurol Int 2022; 14:186-198. [PMID: 35225885 PMCID: PMC8883935 DOI: 10.3390/neurolint14010015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 01/27/2023] Open
Abstract
The treatment of substance abuse with oxytocin is a novel approach to a challenging public health issue that continues to contribute to a growing economic cost for societies worldwide. Methamphetamine addiction is one of the leading causes of mortality worldwide, and despite advances in understanding the neurobiology of methamphetamine addiction, treatment options are limited. There are no medications that the Food and Drug Administration currently approves for stimulant use disorder. Off-label use of therapies for stimulant misuse include antidepressants, anxiolytics, and milder stimulants as replacement agents. Due to the shortcomings of these attempts to treat a complicated psychiatric disorder, recent attention to oxytocin therapy (OT) has gained momentum in clinical studies as a possible therapy in the context of social stress, social anxiety, social cognition, and psychosis. Oxytocin produces enhanced connectivity between cortical regions. The results from studies in rodents with OT suggest that central neuromodulation of oxytocin may be beneficial across transition states of stimulant dependence and may alleviate intense withdrawal symptoms. Studies of oxytocin in the context of other drugs of abuse, including cocaine, cannabis, and alcohol, also support the potential of oxytocin to treat stimulant use disorder, methamphetamine type. Methamphetamine abuse continues to be a significant cause of distress and dysfunction throughout the world. The effects of oxytocin on methamphetamine use outlined in this review should act as a catalyst for further investigation into the efficacy of treating stimulant use disorder, methamphetamine type with oxytocin in humans. More human-based research should initiate studies involving the long-term efficacy, side effects, and patient selection.
Collapse
|
21
|
Nicolas C, Zlebnik NE, Farokhnia M, Leggio L, Ikemoto S, Shaham Y. Sex Differences in Opioid and Psychostimulant Craving and Relapse: A Critical Review. Pharmacol Rev 2022; 74:119-140. [PMID: 34987089 PMCID: PMC11060335 DOI: 10.1124/pharmrev.121.000367] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 08/15/2021] [Indexed: 01/11/2023] Open
Abstract
A widely held dogma in the preclinical addiction field is that females are more vulnerable than males to drug craving and relapse. Here, we first review clinical studies on sex differences in psychostimulant and opioid craving and relapse. Next, we review preclinical studies on sex differences in psychostimulant and opioid reinstatement of drug seeking after extinction of drug self-administration, and incubation of drug craving (time-dependent increase in drug seeking during abstinence). We also discuss ovarian hormones' role in relapse and craving in humans and animal models and speculate on brain mechanisms underlying their role in cocaine craving and relapse in rodent models. Finally, we discuss imaging studies on brain responses to cocaine cues and stress in men and women.The results of the clinical studies reviewed do not appear to support the notion that women are more vulnerable to psychostimulant and opioid craving and relapse. However, this conclusion is tentative because most of the studies reviewed were correlational, not sufficiently powered, and not a priori designed to detect sex differences. Additionally, imaging studies suggest sex differences in brain responses to cocaine cues and stress. The results of the preclinical studies reviewed provide evidence for sex differences in stress-induced reinstatement and incubation of cocaine craving but not cue- or cocaine-induced reinstatement of cocaine seeking. These sex differences are modulated in part by ovarian hormones. In contrast, the available data do not support the notion of sex differences in craving and relapse/reinstatement for methamphetamine or opioids in rodent models. SIGNIFICANCE STATEMENT: This systematic review summarizes clinical and preclinical studies on sex differences in psychostimulant and opioid craving and relapse. Results of the clinical studies reviewed do not appear to support the notion that women are more vulnerable to psychostimulant and opioid craving and relapse. Results of preclinical studies reviewed provide evidence for sex differences in reinstatement and incubation of cocaine seeking but not for reinstatement or incubation of methamphetamine or opioid seeking.
Collapse
Affiliation(s)
- Céline Nicolas
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| | - Natalie E Zlebnik
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| | - Mehdi Farokhnia
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| | - Lorenzo Leggio
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| | - Satoshi Ikemoto
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| | - Yavin Shaham
- Neurocentre Magendie, University of Bordeaux, Bordeaux, France (C.N.); Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, Present address: Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, CA (N.E.Z.); Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD (M.F., L.L., S.I., Y.S.); and Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD (M.F., L.L.)
| |
Collapse
|
22
|
Qian H, Shang Q, Liang M, Gao B, Xiao J, Wang J, Li A, Yang C, Yin J, Chen G, Li T, Liu X. MicroRNA-31-3p/RhoA signaling in the dorsal hippocampus modulates methamphetamine-induced conditioned place preference in mice. Psychopharmacology (Berl) 2021; 238:3207-3219. [PMID: 34313802 DOI: 10.1007/s00213-021-05936-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/12/2021] [Indexed: 12/18/2022]
Abstract
RATIONALE MicroRNAs (miRNAs) regulate neuroplasticity-related proteins and are implicated in methamphetamine (METH) addiction. RhoA is a small Rho GTPase that regulates synaptic plasticity and addictive behaviors. Nevertheless, the functional relationship between RhoA and upstream miRNAs of METH addiction remains unclear. OBJECTIVE To explore the molecular biology and epigenetic mechanisms of the miR-31-3p/RhoA pathway in METH addiction. METHODS RhoA protein and its potential upstream regulator, miR-31-3p, were detected. A dual luciferase reporter was employed to determine whether RhoA constituted a specific target of miR-31-3p. Following adeno-associated virus (AAV)-mediated knockdown or overexpression of miR-31-3p or RhoA in the dorsal hippocampus (dHIP), mice were subjected to conditioned place preference (CPP) to investigate the effects of miR-31-3p and RhoA on METH-induced addictive behaviors. RESULTS RhoA protein was significantly decreased in the dHIP of CPP mice with a concomitant increase in miR-31-3p. RhoA was identified as a direct target of miR-31-3p. Knockdown of miR-31-3p in the dHIP was associated with increased RhoA protein and attenuation of METH-induced CPP. Conversely, overexpression of miR-31-3p was associated with decreased RhoA protein and enhancement of METH effects. Similarly, knockdown of RhoA in the dHIP enhanced METH-induced CPP, whereas RhoA overexpression attenuated the effects of METH. Parallel experiments using sucrose preference revealed that the effects of miR-31-3p/RhoA pathway modulation were specific to METH. CONCLUSIONS Our findings indicate that the miR-31-3p/RhoA pathway in the dHIP modulates METH-induced CPP in mice. Our results highlight the potential role of epigenetics represented by non-coding RNAs in the treatment of METH addiction.
Collapse
Affiliation(s)
- Hongyan Qian
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China
| | - Qing Shang
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China
| | - Min Liang
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China
| | - Baoyao Gao
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jing Xiao
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jing Wang
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China
| | - Axiang Li
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China
| | - Canyu Yang
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jianmin Yin
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China
| | - Gang Chen
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China.,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China
| | - Tao Li
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China. .,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China.
| | - Xinshe Liu
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, People's Republic of China. .,College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Yanta Road W.76 , Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
23
|
Cai J, Che X, Xu T, Luo Y, Yin M, Lu X, Wu C, Yang J. Repeated oxytocin treatment during abstinence inhibited context- or restraint stress-induced reinstatement of methamphetamine-conditioned place preference and promoted adult hippocampal neurogenesis in mice. Exp Neurol 2021; 347:113907. [PMID: 34715133 DOI: 10.1016/j.expneurol.2021.113907] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/06/2021] [Accepted: 10/23/2021] [Indexed: 12/28/2022]
Abstract
Propensity to relapse, even after long-term abstinence, is a crucial feature of methamphetamine (METH) abuse. We and other laboratories have reported that acute treatment of oxytocin (OXT), a hormone and neuropeptide, could inhibit reinstatement of METH seeking in animal studies. However, the effects of repeated OXT treatment on METH reinstatement as well as underlying mechanisms are still unclear. In the present study, the effects of repeated OXT treatment during abstinence on context- or restraint stress-induced reinstatement were investigated using the mice conditioned place preference (CPP) paradigm. After three intermittent injections of METH (2 mg/kg, i.p.) to induce CPP, mice received a daily bilateral intra-hippocampus injection of OXT (0.625, 1.25 or 2.5 μg) for 8 consecutive days before the context- or restraint stress-induced reinstatement test. Meanwhile, adult hippocampal neurogenesis (AHN) level was detected using immunostaining. To further clarify the role of AHN underlying OXT's effects on METH-CPP reinstatement, temozolomide (TMZ, 25 mg/kg, i.p.) was employed to deplete AHN prior to OXT treatment. The data showed that repeated OXT treatment (1.25 and 2.5 μg, intra-hippocampus) significantly inhibited both context- and restraint stress-induced METH-CPP reinstatement and concomitantly promoted AHN in a dose-dependent manner. Notably, TMZ pre-treatment markedly abolished all the above-mentioned effects of OXT, suggesting that AHN was closely involved in OXT's inhibition on reinstatement induced by both triggers. Taken together, the present study indicated that repeated OXT treatment during abstinence could inhibit both context- and restraint stress-induced METH-CPP reinstatement possibly by promoting AHN in mice, which provided a better understanding for OXT's beneficial effects on METH addiction.
Collapse
Affiliation(s)
- Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiaohang Che
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Tianyu Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yuanchao Luo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Meixue Yin
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xianda Lu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
24
|
Li C, Wang H, Wang M, Chen C, Bai F, Ban M, Wu C. Oxytocin Attenuates Methamphetamine-Induced Apoptosis via Oxytocin Receptor in Rat Hippocampal Neurons. Front Pharmacol 2021; 12:639571. [PMID: 34483895 PMCID: PMC8415150 DOI: 10.3389/fphar.2021.639571] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 07/29/2021] [Indexed: 11/15/2022] Open
Abstract
Methamphetamine (METH) is a highly neurotoxic psychoactive substance that can directly damage the central nervous system through prolonged use. Oxytocin (OT) has attracted much attention because of its neuroprotective effect. The purpose of this study was to investigate whether OT is neuroprotective against METH-induced damage in rat hippocampal neurons. Our results revealed that pre-incubation with OT significantly prevented the damage of METH to hippocampal neurons, including the decrease of mitochondrial membrane potential and the increase of ROS (reactive oxygen species). OT pre-incubation attenuated the up-regulation of Cleaved-Caspase-3 expression and the down-regulation of Bcl-2/Bax expression induced by METH. Pre-incubation with OT prevented the decrease in oxytocin receptor density and P-CREB (phosphorylation of cAMP-response element binding) expression induced by METH in rat hippocampal neurons. Moreover, Pre-incubation of atosiban (ATO) significantly prevented these changes. In conclusion, our study proved that pre-administration of OT could significantly attenuate hippocampal neuron apoptosis induced by METH. Oxytocin receptor activation is involved in the preventive effect of OT on METH-induced apoptosis in rat hippocampal neurons.
Collapse
Affiliation(s)
- Chunli Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Haipeng Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Min Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Chunyun Chen
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Fei Bai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Mengqi Ban
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
25
|
Thornton JL, Everett NA, Webb P, Turner AJ, Cornish JL, Baracz SJ. Adolescent oxytocin administration reduces depression-like behaviour induced by early life stress in adult male and female rats. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110279. [PMID: 33567331 DOI: 10.1016/j.pnpbp.2021.110279] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/29/2021] [Accepted: 02/05/2021] [Indexed: 01/06/2023]
Abstract
Early life stress (ELS) exposure alters brain development, increasing vulnerability for mental illness in adulthood, including depression. Despite this association, there are no approved pharmacotherapies to protect against the emergence of mental illness resulting from ELS. Recent preclinical work showed that oxytocin (OT) administration in adulthood reduced depressive-like behaviour in male rats with a history of ELS. However, the ability of an OT treatment regime in adolescence, a critical developmental window for the OT system, to prevent the expression of depressive-like behaviours following ELS has not been investigated. Therefore, the present study aimed to determine whether chronic OT administration can ameliorate the enduring effects of ELS on depressive-like behaviours in both male and female rats. Following birth, Long Evans rat pups (N = 107) underwent maternal separation (MS) for either 15 min (MS15) or 6 h (MS360) on postnatal days (PND) 1-21. During adolescence (PND 28-42), rats received a daily injection of either OT (1 mg/kg) or saline. During adulthood (PND 57 onwards), effort-related motivation was measured using a model of effortful choice (EC), while behavioural despair was measured using the forced swim test (FST). Lastly, body and organ weights were measured to examine the physiological impacts of ELS and chronic OT administration. Overall, in both sexes, MS360 increased behavioural despair yet had no impact on effort-related motivation. Importantly, adolescent OT administration prevented the MS360-induced increase in behavioural despair in both males and females. Additionally, MS360 resulted in persistent reductions in body weight in both sexes post-weaning and increased spleen weight in males and adrenal weight in females. OT treatment had no impact on body weight in either sex, but prevented the MS-induced increase in adrenal gland weight in females. Overall, these findings have important implications for using oxytocin as a preventative pharmacotherapy after ELS.
Collapse
Affiliation(s)
- Jade L Thornton
- Department of Psychology, Macquarie University, North Ryde, NSW 2109, Australia
| | - Nicholas A Everett
- School of Psychology, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Paige Webb
- Department of Psychology, Macquarie University, North Ryde, NSW 2109, Australia
| | - Anita J Turner
- Department of Psychology, Macquarie University, North Ryde, NSW 2109, Australia
| | - Jennifer L Cornish
- Department of Psychology, Macquarie University, North Ryde, NSW 2109, Australia; Centre for Emotional Health, Macquarie University, North Ryde, NSW 2109, Australia
| | - Sarah J Baracz
- Department of Psychology, Macquarie University, North Ryde, NSW 2109, Australia; Centre for Emotional Health, Macquarie University, North Ryde, NSW 2109, Australia; School of Psychology, University of New South Wales, Randwick, NSW, 2052, Australia.
| |
Collapse
|
26
|
Martin EL, Doncheck EM, Reichel CM, McRae-Clark AL. Consideration of sex as a biological variable in the translation of pharmacotherapy for stress-associated drug seeking. Neurobiol Stress 2021; 15:100364. [PMID: 34345636 PMCID: PMC8319013 DOI: 10.1016/j.ynstr.2021.100364] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/23/2021] [Accepted: 07/08/2021] [Indexed: 12/01/2022] Open
Abstract
Stress is a frequent precipitant of relapse to drug use. Pharmacotherapies targeting a diverse array of neural systems have been assayed for efficacy in attenuating stress-induced drug-seeking in both rodents and in humans, but none have shown enough evidence of utility to warrant routine use in the clinic. We posit that a critical barrier in effective translation is inattention to sex as a biological variable at all phases of the research process. In this review, we detail the neurobiological systems implicated in stress-induced relapse to cocaine, opioids, methamphetamine, and cannabis, as well as the pharmacotherapies that have been used to target these systems in rodent models, the human laboratory, and in clinical trials. In each of these areas we additionally describe the potential influences of biological sex on outcomes, and how inattention to fundamental sex differences can lead to biases during drug development that contribute to the limited success of large clinical trials. Based on these observations, we determine that of the pharmacotherapies discussed only α2-adrenergic receptor agonists and oxytocin have a body of research with sufficient consideration of biological sex to warrant further clinical evaluation. Pharmacotherapies that target β-adrenergic receptors, other neuroactive peptides, the hypothalamic-pituitary-adrenal axis, neuroactive steroids, and the endogenous opioid and cannabinoid systems require further assessment in females at the preclinical and human laboratory levels before progression to clinical trials can be recommended.
Collapse
Affiliation(s)
- Erin L Martin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Elizabeth M Doncheck
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Carmela M Reichel
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Aimee L McRae-Clark
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA.,Department of Psychiatry, Medical University of South Carolina, Charleston, SC, 29425, USA
| |
Collapse
|
27
|
Bardo MT, Hammerslag LR, Malone SG. Effect of early life social adversity on drug abuse vulnerability: Focus on corticotropin-releasing factor and oxytocin. Neuropharmacology 2021; 191:108567. [PMID: 33862030 DOI: 10.1016/j.neuropharm.2021.108567] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/16/2021] [Accepted: 04/09/2021] [Indexed: 12/20/2022]
Abstract
Early life adversity can set the trajectory for later psychiatric disorders, including substance use disorders. There are a host of neurobiological factors that may play a role in the negative trajectory. The current review examines preclinical evidence suggesting that early life adversity specifically involving social factors (maternal separation, adolescent social isolation and adolescent social defeat) may influence drug abuse vulnerability by strengthening corticotropin-releasing factor (CRF) systems and weakening oxytocin (OT) systems. In adulthood, pharmacological and genetic evidence indicates that both CRF and OT systems are directly involved in drug reward processes. With early life adversity, numerous studies show an increase in drug abuse vulnerability measured in adulthood, along a concomitant strengthening of CRF systems and a weakening of OT systems. Mechanistic studies, while relatively few in number, are generally consistent with the theme that strengthened CRF systems and weakened OT systems mediate, at least in part, the link between early life adversity and drug abuse vulnerability. Establishing a direct role of CRF and OT in mediating the relation between early life social stressors and drug abuse vulnerability will inform clinical researchers and practitioners toward the development of intervention strategies to reduce risk among those suffering from early life adversities. This article is part of the special issue on 'Vulnerabilities to Substance Abuse'.
Collapse
Affiliation(s)
- Michael T Bardo
- Department of Psychology, University of Kentucky, Lexington, KY, 40536-0509, USA.
| | - Lindsey R Hammerslag
- Department of Psychology, University of Kentucky, Lexington, KY, 40536-0509, USA
| | - Samantha G Malone
- Department of Psychology, University of Kentucky, Lexington, KY, 40536-0509, USA
| |
Collapse
|
28
|
Sanna F, De Luca MA. The potential role of oxytocin in addiction: What is the target process? Curr Opin Pharmacol 2021; 58:8-20. [PMID: 33845377 DOI: 10.1016/j.coph.2021.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/26/2021] [Accepted: 03/05/2021] [Indexed: 01/27/2023]
Abstract
Oxytocin regulates a variety of centrally-mediated functions, ranging from socio-sexual behavior, maternal care, and affiliation to fear, stress, anxiety. In the past years, both clinical and preclinical studies characterized oxytocin for its modulatory role on reward-related neural substrates mainly involving the interplay with the mesolimbic and mesocortical dopaminergic pathways. This suggests a role of this nonapeptide on the neurobiology of addiction raising the possibility of its therapeutic use. Although far from a precise knowledge of the underlying mechanisms, the putative role of the bed nucleus of the stria terminalis as a key structure where oxytocin may rebalance altered neurochemical processes and neuroplasticity involved in dependence and relapse has been highlighted. This view opens new opportunities to address the health problems related to drug misuse.
Collapse
Affiliation(s)
- Fabrizio Sanna
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Cagliari 09042, Italy
| | | |
Collapse
|
29
|
Altshuler RD, Yang ES, Garcia KT, Davis IR, Olaniran A, Haile M, Razavi S, Li X. Role of orbitofrontal cortex in incubation of oxycodone craving in male rats. Addict Biol 2021; 26:e12927. [PMID: 32570285 DOI: 10.1111/adb.12927] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/06/2020] [Accepted: 05/14/2020] [Indexed: 12/25/2022]
Abstract
One of the main challenges in treating opioid-use disorders is relapse during abstinence, triggered by re-exposure to drug-associated cues. Previous studies have demonstrated that drug-seeking in rats progressively increases over time during withdrawal (incubation of drug craving). Here, we used male rats and examined neural mechanisms underlying incubation of craving to oxycodone, a commonly abused prescription opioid, and we focused on orbitofrontal cortex (OFC), a brain region previously implicated in incubation of heroin craving. We first used neuronal activity marker Fos and measured neuronal activation in OFC (ventral and lateral OFC) associated with day-1 and day-15 relapse tests. Next, we determined the effect of pharmacological reversible inactivation of OFC on incubated oxycodone seeking on withdrawal day 15. Finally, we determined the effect of reversible inactivation of OFC on nonincubated oxycodone seeking on withdrawal day 1. We found that lever presses during relapse tests were higher on withdrawal day 15 than on withdrawal day 1 (incubation of oxycodone craving). Incubation of oxycodone craving is accompanied with a time-dependent increase of Fos protein expression in both ventral and lateral OFC. Lastly, OFC inactivation decreased oxycodone seeking on withdrawal day 15 but had no effect on withdrawal day 1. Together with the previous heroin study, results here show that OFC plays a critical role in incubation of opioid craving.
Collapse
Affiliation(s)
- Rachel D. Altshuler
- Department of Psychology University of Maryland College Park College Park MD USA
| | - Eddy S. Yang
- Department of Psychology University of Maryland College Park College Park MD USA
| | - Kristine T. Garcia
- Department of Psychology University of Maryland College Park College Park MD USA
| | - Ian R. Davis
- Department of Psychology University of Maryland College Park College Park MD USA
| | - Adedayo Olaniran
- Department of Psychology University of Maryland College Park College Park MD USA
| | - Meron Haile
- Department of Psychology University of Maryland College Park College Park MD USA
| | - Syrus Razavi
- Department of Psychology University of Maryland College Park College Park MD USA
| | - Xuan Li
- Department of Psychology University of Maryland College Park College Park MD USA
| |
Collapse
|
30
|
Koriem KM, Selim AY, Mazen RA. N-acetylcysteine-amide improves tissue oxidative stress, DNA damage, and proteins disappearance in methamphetamine toxicity more efficiently than N-acetyl-L-cysteine. TOXICOLOGIE ANALYTIQUE ET CLINIQUE 2021. [DOI: 10.1016/j.toxac.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
31
|
Walker LC. A balancing act: the role of pro- and anti-stress peptides within the central amygdala in anxiety and alcohol use disorders. J Neurochem 2021; 157:1615-1643. [PMID: 33450069 DOI: 10.1111/jnc.15301] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/18/2020] [Accepted: 01/06/2021] [Indexed: 12/21/2022]
Abstract
The central nucleus of the amygdala (CeA) is widely implicated as a structure that integrates both appetitive and aversive stimuli. While intrinsic CeA microcircuits primarily consist of GABAergic neurons that regulate amygdala output, a notable feature of the CeA is the heterogeneity of neuropeptides and neuropeptide/neuromodulator receptors that it expresses. There is growing interest in the role of the CeA in mediating psychopathologies, including stress and anxiety states and their interactions with alcohol use disorders. Within the CeA, neuropeptides and neuromodulators often exert pro- or anti- stress actions, which can influence anxiety and alcohol associated behaviours. In turn, alcohol use can cause adaptions within the CeA, which may render an individual more vulnerable to stress which is a major trigger of relapse to alcohol seeking. This review examines the neurocircuitry, neurochemical phenotypes and how pro- and anti-stress peptide systems act within the CeA to regulate anxiety and alcohol seeking, focusing on preclinical observations from animal models. Furthermore, literature exploring the targeting of genetically defined populations or neuronal ensembles and the role of the CeA in mediating sex differences in stress x alcohol interactions are explored.
Collapse
Affiliation(s)
- Leigh C Walker
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Vic, Australia
| |
Collapse
|
32
|
Everett NA, Carey HA, Cornish JL, Baracz SJ. Sign tracking predicts cue-induced but not drug-primed reinstatement to methamphetamine seeking in rats: Effects of oxytocin treatment. J Psychopharmacol 2020; 34:1271-1279. [PMID: 33081558 DOI: 10.1177/0269881120954052] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND The incentive sensitisation theory of addiction posits that drug-associated stimuli become imbued with incentive motivational properties, driving pathological drug seeking. However, pre-existing variability in the incentive salience to non-drug reward cues ('sign trackers' (STs); 'goal trackers' (GTs)) is also predictive of the desire for and relapse to cocaine and opioids. Here, we asked whether variation in propensity to attribute incentive salience to a food cue is predictive of reinstatement to the highly addictive psychostimulant methamphetamine (METH), and whether treatment with the promising anti-addiction therapy oxytocin differentially reduces METH behaviour between STs and GTs. METHODS Rats were trained to associate a Pavlovian cue with delivery of a sucrose pellet over 8 days. They then received jugular vein catheters for intravenous METH self-administration, followed by behavioural extinction, and cue-induced and METH-primed reinstatement to METH-seeking behaviours. Oxytocin was administered prior to self-administration and reinstatement tests. RESULTS Despite the self-administration of similar amounts of METH, STs reinstated more to METH cues than did GTs, yet METH-priming reinstated STs and GTs similarly. Furthermore, oxytocin attenuated cue-induced reinstatement more so in STs than in GTs, and reduced METH-primed reinstatement to a greater extent in the top quartile of reinstaters, indicating that oxytocin treatment may be most effective for those at highest risk of addiction. CONCLUSIONS This pre-existing bias towards reward cues presents a possible tool to screen for METH addiction susceptibility and may be useful for understanding the neurobiology of addiction and for pharmacotherapeutic discovery.
Collapse
Affiliation(s)
| | - Harry A Carey
- Department of Psychology, Macquarie University, North Ryde, Australia
| | - Jennifer L Cornish
- Centre for Emotional Health, Department of Psychology, Macquarie University, North Ryde, Australia
| | - Sarah J Baracz
- Department of Psychology, Macquarie University, North Ryde, Australia.,Centre for Emotional Health, Department of Psychology, Macquarie University, North Ryde, Australia
| |
Collapse
|
33
|
Altshuler RD, Lin H, Li X. Neural mechanisms underlying incubation of methamphetamine craving: A mini-review. Pharmacol Biochem Behav 2020; 199:173058. [PMID: 33250444 DOI: 10.1016/j.pbb.2020.173058] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/27/2020] [Accepted: 10/09/2020] [Indexed: 12/20/2022]
Abstract
Cue-induced drug craving and seeking progressively increases during abstinence. This "incubation of drug craving" phenomenon has been observed in both laboratory animals and humans. Preclinical studies identified several neural mechanisms underlying incubation of drug craving after forced abstinence, primarily focusing on cocaine. Recently, studies started focusing on another powerful psychostimulant, methamphetamine (Meth), and developed new incubation procedures (choice-induced and punishment-induced abstinence). Here, we review mechanistic studies at the circuit, synaptic and molecular levels on incubation of Meth craving. First, we provide an overview of neural adaptations associated with prolonged forced abstinence after extended-access Meth self-administration. Next, we review studies examining the causal roles of discrete brain regions and associated circuits, glutamate transmission, histone deacetylase 5 and oxytocin in incubation of Meth craving after forced abstinence. Lastly, we review causal and correlational studies examining the mechanisms underlying incubation of Meth craving after choice-induced voluntary abstinence and punishment-induced abstinence, respectively. We conclude by discussing the translational potential of these mechanistic studies in Meth relapse prevention in human drug users.
Collapse
Affiliation(s)
- Rachel D Altshuler
- Department of Psychology, University of Maryland College Park, College Park, MD 20742, United States of America
| | - Hongyu Lin
- Department of Psychology, University of Maryland College Park, College Park, MD 20742, United States of America
| | - Xuan Li
- Department of Psychology, University of Maryland College Park, College Park, MD 20742, United States of America.
| |
Collapse
|
34
|
Westbrook SR, Carrica LK, Banks A, Gulley JM. AMPed-up adolescents: The role of age in the abuse of amphetamines and its consequences on cognition and prefrontal cortex development. Pharmacol Biochem Behav 2020; 198:173016. [PMID: 32828971 DOI: 10.1016/j.pbb.2020.173016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 01/24/2023]
Abstract
Adolescent use of amphetamine and its closely related, methylated version methamphetamine, is alarmingly high in those who use drugs for nonmedical purposes. This raises serious concerns about the potential for this drug use to have a long-lasting, detrimental impact on the normal development of the brain and behavior that is ongoing during adolescence. In this review, we explore recent findings from both human and laboratory animal studies that investigate the consequences of amphetamine and methamphetamine exposure during this stage of life. We highlight studies that assess sex differences in adolescence, as well as those that are designed specifically to address the potential unique effects of adolescent exposure by including groups at other life stages (typically young adulthood). We consider epidemiological studies on age and sex as vulnerability factors for developing problems with the use of amphetamines, as well as human and animal laboratory studies that tap into age differences in use, its short-term effects on behavior, and the long-lasting consequences of this exposure on cognition. We also focus on studies of drug effects in the prefrontal cortex, which is known to be critically important for cognition and is among the later maturing brain regions. Finally, we discuss important issues that should be addressed in future studies so that the field can further our understanding of the mechanisms underlying adolescent use of amphetamines and its outcomes on the developing brain and behavior.
Collapse
Affiliation(s)
- Sara R Westbrook
- Department of Psychology, University of Illinois at Urbana-Champaign, USA
| | - Lauren K Carrica
- Department of Psychology, University of Illinois at Urbana-Champaign, USA
| | - Asia Banks
- Department of Psychology, University of Illinois at Urbana-Champaign, USA
| | - Joshua M Gulley
- Department of Psychology, University of Illinois at Urbana-Champaign, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, USA.
| |
Collapse
|
35
|
Stauffer CS, Moschetto JM, McKernan S, Meinzer N, Chiang C, Rapier R, Hsiang E, Norona J, Borsari B, Woolley JD. Oxytocin-enhanced group therapy for methamphetamine use disorder: Randomized controlled trial. J Subst Abuse Treat 2020; 116:108059. [PMID: 32741502 DOI: 10.1016/j.jsat.2020.108059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/11/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Methamphetamine (METH) use is a public health crisis that disproportionately affects men who have sex with men (MSM). There are currently no FDA-approved pharmacological interventions to treat methamphetamine use disorder (MUD). MUD is associated with social impairments and extremely high treatment attrition rates. Administration of oxytocin, a neuropeptide involved in social attachment, may be a novel approach to addressing these issues. Moreover, oxytocin administration has shown promise for reducing METH-related addictive behavior in animal models, but has not yet been investigated in clinical trials for MUD. Last, oxytocin is known to modulate stress responsivity via regulation of the autonomic nervous system, which is dysregulated in METH users. We hypothesize that oxytocin, in combination with group psychotherapy, will increase treatment engagement, reduce addiction behavior, and mitigate stress hyperreactivity. METHODS This is a randomized, double blind trial of oxytocin 40-IU (n = 24) or placebo (n = 24) administered intranasally prior to each of six weekly motivational interviewing group therapy (MIGT) sessions for MUD in MSM. PRIMARY OUTCOME (a) session attendance. SECONDARY OUTCOMES (b) group cohesion, (c) anxiety, (d) METH craving, (e) METH use, and (f) in-session cardiac physiology. RESULTS Participants receiving oxytocin had significantly higher group therapy attendance than those receiving placebo, OR 3.26, 95% CI [1.27-8.41], p = .014. There was a small effect of oxytocin on group cohension, but not anxiety or craving. METH use did not change over the six-week MIGT course in either treatment arm. Participants receiving oxytocin had lower average heart rates during MIGT sessions and higher heart rate variability. There were positive main effects of MIGT over Time regardless of study drug. CONCLUSIONS This evidence, and the lack of any serious adverse events, suggests that oxytocin may safely increase treatment attendance. One possible mechanism by which it may do so is its modulation of the autonomic nervous system. Further investigation is warranted.
Collapse
Affiliation(s)
- Christopher S Stauffer
- San Francisco Veterans Affairs Medical Center, United States of America; University of California, San Francisco, United States of America; Portland Veterans Affairs Medical Center & Oregon Health and Science University, United States of America.
| | - Jenna M Moschetto
- San Francisco Veterans Affairs Medical Center, United States of America; University of California, San Francisco, United States of America
| | - Scott McKernan
- San Francisco Veterans Affairs Medical Center, United States of America; University of California, San Francisco, United States of America
| | | | - Chavy Chiang
- San Francisco Veterans Affairs Medical Center, United States of America
| | - Rachel Rapier
- San Francisco Veterans Affairs Medical Center, United States of America
| | - Elaine Hsiang
- University of California, San Francisco, United States of America
| | - Jerika Norona
- San Francisco Veterans Affairs Medical Center, United States of America
| | - Brian Borsari
- San Francisco Veterans Affairs Medical Center, United States of America; University of California, San Francisco, United States of America
| | - Joshua D Woolley
- San Francisco Veterans Affairs Medical Center, United States of America; University of California, San Francisco, United States of America
| |
Collapse
|