1
|
Smiley CE, Pate BS, Bouknight SJ, Wood SK. Individual differences in behavioral responses to predator odor predict subsequent stress reactivity in female rats. Stress 2025; 28:2479739. [PMID: 40181610 DOI: 10.1080/10253890.2025.2479739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Stress-induced neuropsychiatric disorders are among the most prevalent medical conditions and have widespread effects on both patients and society. Females experience over twice the rates of stress-related anxiety and depression when compared to males and often exhibit worse symptomatology and treatment outcomes. However, preclinical experiments exploring the neurobiological mechanisms of stress susceptibility in females have been traditionally understudied. Previous data from our lab has determined that females are selectively vulnerable to the consequences of vicarious witness stress, and these experiments were designed to determine specific behavioral and physiological factors that could predict which groups would be more susceptible to the effects of stress. Adult, female, Sprague-Dawley rats were first exposed to a ferret predator odor to determine baseline individual differences in behavioral responses. Rats were stratified by the duration of freezing behavior exhibited in response to the ferret odor and equally balanced into non-stressed controls and vicarious witness stress exposed groups. These female rats were then assessed on a battery of behavioral tasks including sucrose preference, elevated plus maze, acoustic startle, and the ferret odor and witness stress cue exposures to determine if baseline differences in stress responding can predict the behavioral response to future stress and stress cues. High freezing in response to the ferret odor was associated with behavioral sensitization to witness stress and hypervigilant responses to stress cues that was accompanied by exaggerated neuroimmune responses. These experiments establish a powerful behavioral predictor of stress susceptibility in females and begin to address neurobiological correlates that underlie this response.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
- WJB Dorn Veterans Administration Medical Center, Columbia, SC, USA
| | - Brittany S Pate
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
- Department of Exercise Science, University of South Carolina, Columbia, SC, USA
| | - Samantha J Bouknight
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
- WJB Dorn Veterans Administration Medical Center, Columbia, SC, USA
- USC Institute for Cardiovascular Disease Research, Columbia, SC, USA
| |
Collapse
|
2
|
Smiley CE, Pate BS, Bouknight SJ, Harrington EN, Jasnow AM, Wood SK. The functional role of locus coeruleus microglia in the female stress response. Mol Psychiatry 2025:10.1038/s41380-025-02971-9. [PMID: 40188312 DOI: 10.1038/s41380-025-02971-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/07/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025]
Abstract
Neuropsychiatric disorders that result from stress exposure are highly associated with central inflammation. Our previous work established that females selectively exhibit heightened proinflammatory cytokine production within the noradrenergic locus coeruleus (LC) along with a hypervigilant behavioral phenotype in response to witnessing social stress. Notably, ablation of microglia using pharmacological techniques prevents this behavioral response. These studies were designed to further investigate the impact of stress-induced neuroimmune signaling on the long-term behavioral and neuronal consequences of social stress exposure in females using chemogenetics. We first characterized the use of an AAV-CD68-Gi-DREADD virus targeted to microglia within the LC and confirmed viral transduction, selectivity, and efficacy. Clozapine-n-oxide (CNO) was used for the suppression of microglial reactivity during acute and chronic exposure to vicarious/witness social defeat in female rats. Chemogenetic-mediated inhibition of microglial reactivity during stress blunted the neuroimmune response to stress and prevented both acute and long-term hypervigilant behavioral responses. Further, a history of microglial suppression during stress prevented the heightened LC activity typically observed in response to stress cues. These studies are among the first to use a chemogenetic approach to inhibit central microglia in vivo and establish LC microglia as a key driver of the behavioral and neuronal responses to social stress in females.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC, 29209, US
- WJB Dorn Veterans Administration Medical Center, Columbia, SC, 29209, US
| | - Brittany S Pate
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC, 29209, US
- University of South Carolina, Department of Exercise Science, Columbia, SC, 29209, US
| | - Samantha J Bouknight
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC, 29209, US
| | - Evelynn N Harrington
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC, 29209, US
- WJB Dorn Veterans Administration Medical Center, Columbia, SC, 29209, US
| | - Aaron M Jasnow
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC, 29209, US
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC, 29209, US.
- WJB Dorn Veterans Administration Medical Center, Columbia, SC, 29209, US.
- USC Institute for Cardiovascular Disease Research, Columbia, SC, 29209, US.
| |
Collapse
|
3
|
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 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] [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
|
4
|
Shi S, Sun Y, Zan G, Zhao M. The interaction between central and peripheral immune systems in methamphetamine use disorder: current status and future directions. J Neuroinflammation 2025; 22:40. [PMID: 39955589 PMCID: PMC11829452 DOI: 10.1186/s12974-025-03372-z] [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: 12/13/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
Methamphetamine (METH) use disorder (MUD) is characterized by compulsive drug-seeking behavior and substantial neurotoxicity, posing a considerable burden on individuals and society. Traditionally perceived as a localized central nervous system disorder, recent preclinical and clinical studies have elucidated that MUD is a multifaceted disorder influenced by various biological systems, particularly the immune system. Emerging evidence suggests that both central and peripheral immune responses play a crucial role in the initiation and persistence of MUD. Conceptualizing it as a systemic immune process prompts significant inquiries regarding the mechanisms of communication between peripheral and central compartments. Also, whether this intercommunication could serve as diagnostic biomarkers or therapeutic targets. This review begins by offering an overview of mechanistic studies pertaining to the neuroimmune and peripheral immune systems. Finally, future directions are suggested through the integration of innovative technologies and multidimensional data to promote the translation of basic mechanistic research into clinical diagnostic and therapeutic interventions.
Collapse
Affiliation(s)
- Sai Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwen Sun
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guiying Zan
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Min Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences, Shanghai, China.
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wan Ping Road, Shanghai, 200030, China.
| |
Collapse
|
5
|
Smiley CE, Pate BS, Bouknight SJ, Harrington EN, Jasnow AM, Wood SK. The functional role of locus coeruleus microglia in the female stress response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575076. [PMID: 38260568 PMCID: PMC10802589 DOI: 10.1101/2024.01.10.575076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Neuropsychiatric disorders that result from stress exposure are highly associated with central inflammation. Our previous work established that females selectively exhibit heightened proinflammatory cytokine production within the noradrenergic locus coeruleus (LC) along with a hypervigilant behavioral phenotype in response to witnessing social stress, and ablation of microglia using pharmacological techniques prevents this behavioral response. These studies were designed to further investigate the impact of stress-induced neuroimmune signaling on the long-term behavioral and neuronal consequences of social stress exposure in females using chemogenetics. We first characterized the use of an AAV-CD68-Gi-DREADD virus targeted to microglia within the LC and confirmed viral transduction, selectivity, and efficacy. Clozapine-n-oxide (CNO) was used for the suppression of microglial reactivity during acute and chronic exposure to vicarious/witness social defeat in female rats. Chemogenetic-mediated inhibition of microglial reactivity during stress blunted the neuroimmune response to stress and prevented both acute and long-term hypervigilant behavioral responses. Further, a history of microglial suppression during stress prevented the heightened LC activity typically observed in response to stress cues. These studies are among the first to use a chemogenetic approach to inhibit microglia within the female brain in vivo and establish LC inflammation as a key mechanism underlying the behavioral and neuronal responses to social stress in females.
Collapse
Affiliation(s)
- Cora E. Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209
| | - Brittany S. Pate
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- University of South Carolina, Department of Exercise Science, Columbia, SC 29209
| | - Samantha J. Bouknight
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
| | - Evelynn N. Harrington
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209
| | - Aaron M. Jasnow
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
| | - Susan K. Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209
- WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209
| |
Collapse
|
6
|
Pate BS, Smiley CE, Harrington EN, Bielicki BH, Davis JM, Reagan LP, Grillo CA, Wood SK. Voluntary wheel running as a promising strategy to promote autonomic resilience to social stress in females: Vagal tone lies at the heart of the matter. Auton Neurosci 2024; 253:103175. [PMID: 38677130 PMCID: PMC11173375 DOI: 10.1016/j.autneu.2024.103175] [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: 12/13/2023] [Revised: 03/06/2024] [Accepted: 04/09/2024] [Indexed: 04/29/2024]
Abstract
Social stress is a major risk factor for comorbid conditions including cardiovascular disease and depression. While women exhibit 2-3× the risk for these stress-related disorders compared to men, the mechanisms underlying heightened stress susceptibility among females remain largely unknown. Due to a lack in understanding of the pathophysiology underlying stress-induced comorbidities among women, there has been a significant challenge in developing effective therapeutics. Recently, a causal role for inflammation has been established in the onset and progression of comorbid cardiovascular disease/depression, with women exhibiting increased sensitivity to stress-induced immune signaling. Importantly, reduced vagal tone is also implicated in stress susceptibility, through a reduction in the vagus nerve's well-recognized anti-inflammatory properties. Thus, examining therapeutic strategies that stabilize vagal tone during stress may shed light on novel targets for promoting stress resilience among women. Recently, accumulating evidence has demonstrated that physical activity exerts cardio- and neuro-protective effects by enhancing vagal tone. Based on this evidence, this mini review provides an overview of comorbid cardiovascular and behavioral dysfunction in females, the role of inflammation in these disorders, how stress may impart its negative effects on the vagus nerve, and how exercise may act as a preventative. Further, we highlight a critical gap in the literature with regard to the study of females in this field. This review also presents novel data that are the first to demonstrate a protective role for voluntary wheel running over vagal tone and biomarkers of cardiac dysfunction in the face of social stress exposure in female rats.
Collapse
Affiliation(s)
- Brittany S Pate
- Department of Exercise Science, University of South Carolina, Columbia, SC, United States of America; Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America
| | - Evelynn N Harrington
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America
| | - B Hunter Bielicki
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America
| | - J Mark Davis
- Department of Exercise Science, University of South Carolina, Columbia, SC, United States of America
| | - Lawrence P Reagan
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America
| | - Claudia A Grillo
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States of America; Columbia VA Health Care System, Columbia, SC, United States of America; USC Institute for Cardiovascular Disease Research, Columbia, SC, United States of America.
| |
Collapse
|
7
|
Acuña AM, Park C, Leyrer-Jackson JM, Olive MF. Promising immunomodulators for management of substance and alcohol use disorders. Expert Opin Pharmacother 2024; 25:867-884. [PMID: 38803314 PMCID: PMC11216154 DOI: 10.1080/14656566.2024.2360653] [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: 03/26/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION The neuroimmune system has emerged as a novel target for the treatment of substance use disorders (SUDs), with immunomodulation producing encouraging therapeutic benefits in both preclinical and clinical settings. AREAS COVERED In this review, we describe the mechanism of action and immune response to methamphetamine, opioids, cocaine, and alcohol. We then discuss off-label use of immunomodulators as adjunctive therapeutics in the treatment of neuropsychiatric disorders, demonstrating their potential efficacy in affective and behavioral disorders. We then discuss in detail the mechanism of action and recent findings regarding the use of ibudilast, minocycline, probenecid, dexmedetomidine, pioglitazone, and cannabidiol to treat (SUDs). These immunomodulators are currently being investigated in clinical trials described herein, specifically for their potential to decrease substance use, withdrawal severity, central and peripheral inflammation, comorbid neuropsychiatric disorder symptomology, as well as their ability to improve cognitive outcomes. EXPERT OPINION We argue that although mixed, findings from recent preclinical and clinical studies underscore the potential benefit of immunomodulation in the treatment of the behavioral, cognitive, and inflammatory processes that underlie compulsive substance use.
Collapse
Affiliation(s)
- Amanda M. Acuña
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, Arizona, USA
| | - Connor Park
- Department of Biomedical Sciences, Creighton University School of Medicine – Phoenix, Phoenix, Arizona, USA
| | - Jonna M. Leyrer-Jackson
- Department of Biomedical Sciences, Creighton University School of Medicine – Phoenix, Phoenix, Arizona, USA
| | - M. Foster Olive
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
8
|
Cui X, Li J, Wang C, Ishaq HM, Zhang R, Yang F. Relationship between sphingolipids-mediated neuroinflammation and alcohol use disorder. Pharmacol Biochem Behav 2024; 235:173695. [PMID: 38128765 DOI: 10.1016/j.pbb.2023.173695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Alcohol use disorder is a chronic recurrent encephalopathy, and its pathogenesis has not been fully understood. Among possible explanations, neuroinflammation caused by the disorders of brain central immune signaling has been identified as one possible mechanism of alcohol use disorder. As the basic components of cells and important bioactive molecules, sphingolipids are essential in regulating many cellular activities. Recent studies have shown that sphingolipids-mediated neuroinflammation may be involved in the development of alcohol use disorder. METHODS PubMed databases were searched for literature on sphingolipids and alcohol use disorder (alcohol abuse, alcohol addiction, alcohol dependence, and alcohol misuse) including evidence of the relationship between sphingolipids-mediated neuroinflammation and alcohol use disorder (formation, withdrawal, treatment). RESULTS Disorders of sphingolipid metabolism, including the different types of sphingolipids and regulatory enzyme activity, have been found in patients with alcohol use disorder as well as animal models, which in turn cause neuro-inflammation in the central nervous system. Thus, these disorders may also be an important mechanism in the development of alcohol use disorder in patients. In addition, different sphingolipids may have different or even reverse effects on alcohol use disorder. CONCLUSIONS The sphingolipids-mediated neuroinflammation plays an important role in the development of alcohol use disorder. This review proposes a potential approach to prevent and treat alcohol use disorders by manipulating sphingolipid metabolism.
Collapse
Affiliation(s)
- XiaoJian Cui
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China; Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - JiaZhen Li
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - ChuanSheng Wang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Hafiz Muhammad Ishaq
- Faculty of Veterinary and Animal Sciences, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - RuiLin Zhang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China.
| | - Fan Yang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China; Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
9
|
Grantham EK, Tiwari GR, Ponomareva O, Harris RA, Lopez MF, Becker HC, Mayfield RD. Transcriptome changes in the nucleus of the solitary tract induced by repeated stress, alcohol dependence, or stress-induced drinking in dependent mice. Neuropharmacology 2024; 242:109768. [PMID: 37865137 PMCID: PMC10688594 DOI: 10.1016/j.neuropharm.2023.109768] [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: 06/13/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/23/2023]
Abstract
Stress increases alcohol consumption in dependent animals and contributes to the development of alcohol use disorder. The nucleus of the solitary tract (NTS) is a critical brainstem region for integrating and relaying central and peripheral signals to regulate stress responses, but it is not known if it plays a role in alcohol dependence- or in stress-induced escalations in alcohol drinking in dependent mice. Here, we used RNA-sequencing and bioinformatics analyses to study molecular adaptations in the NTS of C57BL/6J male mice that underwent an ethanol drinking procedure that uses exposure to chronic intermittent ethanol (CIE) vapor, forced swim stress (FSS), or both conditions (CIE + FSS). Transcriptome profiling was performed at three different times after the last vapor cycle (0-hr, 72-hr, and 168-hr) to identify changes in gene expression associated with different stages of ethanol intoxication and withdrawal. In the CIE and CIE + FSS groups at 0-hr, there was upregulation of genes enriched for cellular response to type I interferon (IFN) and type I IFN- and cytokine-mediated signaling pathways, while the FSS group showed upregulation of neuronal genes. IFN signaling was the top gene network positively correlated with ethanol consumption levels in the CIE and CIE + FSS groups. Results from different analyses (differential gene expression, weighted gene coexpression network analysis, and rank-rank hypergeometric overlap) indicated that activation of type I IFN signaling would be expected to increase ethanol consumption. The CIE and CIE + FSS groups also shared an immune signature in the NTS as has been demonstrated in other brain regions after chronic ethanol exposure. A temporal-based clustering analysis revealed a unique expression pattern in the CIE + FSS group that suggests the interaction of these two stressors produces adaptations in synaptic and glial functions that may drive stress-induced drinking.
Collapse
Affiliation(s)
- Emily K Grantham
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Gayatri R Tiwari
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Olga Ponomareva
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Marcello F Lopez
- Department of Psychiatry & Behavioral Sciences and Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Howard C Becker
- Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, 28425, USA; Department of Psychiatry & Behavioral Sciences and Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Veterans Affairs Medical Center, Charleston, SC, 20401, USA
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA; Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
10
|
Qeadan F, Ross S, Barbeau WA, Madden EF, Venner KL, English K. Psychostimulant Misuse Among American Indian, Alaskan Native, or Native Hawaiian College Students in the U.S. From 2015 to 2019. Subst Abuse 2023; 17:11782218231209667. [PMID: 38025909 PMCID: PMC10655647 DOI: 10.1177/11782218231209667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/04/2023] [Indexed: 12/01/2023]
Abstract
Introduction This study examines factors associated with psychostimulant misuse, including polysubstance use and social factors, among the understudied American Indian/Alaska Native/Native Hawaiian (AI/AN/NH) college student population. Methods Data were from the 2015 to 2019 American College Health Association-National College Health Assessment IIc (ACHA-NCHA IIc) survey. Multivariable logistic regression models and odds ratios were used to estimate associations between psychostimulant misuse and potential risk and protective factors among AI/AN/NH college students, including licit and illicit substance use, social support, relationship factors, exposure to violence or abuse, mental health symptoms, drug and alcohol education, and sample demographics. Results Opioid misuse among AI/AN/NH college students significantly increased the odds of using psychostimulants. Specifically, for cocaine use, the adjusted odds ratio (aOR) was 3.17 with a 95% confidence interval (C.I.) of 2.17 to 4.63; for methamphetamine use, the aOR was 38.87 (95% C.I. 19.24-78.52). For amphetamine misuse among non-Tobacco users, the aOR was 5.47 (95% C.I. 3.49-8.55), while among Tobacco users, the aOR was 2.65 (95% C.I. 2.07-3.41). For cocaine and other stimulant misuse, the aOR was 3.64 (95% C.I. 2.30-5.67). Additionally, the use of other types of licit and illicit substances was associated with greater odds of psychostimulant use and misuse. Conversely, factors such as age, living on campus, and residing in parental/guardian housing were linked with lower odds of psychostimulant use and misuse. Conclusion Substance use prevention and treatment interventions targeting AI/AN/NH college students should address polysubstance use, including the combined use of opioids and psychostimulants. Substance use interventions should not be siloed to focus narrowly on single substances but rather should leverage potential protective factors against substance use, such as promoting supportive campus and family living conditions and other social support networks, in broad efforts to reduce multiple forms of substance use among AI/AN/NH students.
Collapse
Affiliation(s)
- Fares Qeadan
- Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
| | - Sydney Ross
- Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
| | - William A Barbeau
- Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
| | - Erin F Madden
- Department of Family Medicine and Public Health Sciences, Wayne State University, Detroit, MI, USA
| | - Kamilla L Venner
- Department of Psychology, Center on Alcohol, Substance Use and Addictions (CASAA), University of New Mexico, Albuquerque, NM, USA
| | - Kevin English
- Albuquerque Area Southwest Tribal Epidemiology Center, Albuquerque, NM, USA
| |
Collapse
|
11
|
Cheng J, He Z, Chen Q, Lin J, Peng Y, Zhang J, Yan X, Yan J, Niu S. Histone modifications in cocaine, methamphetamine and opioids. Heliyon 2023; 9:e16407. [PMID: 37265630 PMCID: PMC10230207 DOI: 10.1016/j.heliyon.2023.e16407] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Cocaine, methamphetamine and opioids are leading causes of drug abuse-related deaths worldwide. In recent decades, several studies revealed the connection between and epigenetics. Neural cells acquire epigenetic alterations that drive the onset and progress of the SUD by modifying the histone residues in brain reward circuitry. Histone modifications, especially acetylation and methylation, participate in the regulation of gene expression. These alterations, as well as other host and microenvironment factors, are associated with a serious of negative neurocognitive disfunctions in various patient populations. In this review, we highlight the evidence that substantially increase the field's ability to understand the molecular actions underlying SUD and summarize the potential approaches for SUD pharmacotherapy.
Collapse
Affiliation(s)
- Junzhe Cheng
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ziping He
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qianqian Chen
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Jiang Lin
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Yilin Peng
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Jinlong Zhang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, 830001, China
| | - Xisheng Yan
- Department of Cardiovascular Medicine, Wuhan Third Hospital & Tongren Hospital of Wuhan University, Wuhan, Hubei Province, 430074, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, 830001, China
| | - Shuliang Niu
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, 830001, China
| |
Collapse
|
12
|
Sodium Butyrate Supplementation Modulates Neuroinflammatory Response Aggravated by Antibiotic Treatment in a Mouse Model of Binge-like Ethanol Drinking. Int J Mol Sci 2022; 23:ijms232415688. [PMID: 36555338 PMCID: PMC9778941 DOI: 10.3390/ijms232415688] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Growing evidence supports the pivotal role of the bidirectional interplay between the gut microbiota and the central nervous system during the progression of alcohol use disorder (AUD). In our previous study, supplementation with sodium butyrate (SB) in C57BL/6J mice prevented increased ethanol consumption in a binge-like drinking paradigm (DID) as a result of treatment with a non-absorbable antibiotic cocktail (ABX). In this study, we tested the hypothesis that SB protection against enhanced ABX-induced ethanol consumption in mice is partially due to modulation of neuroinflammatory responses. Pro- and anti-inflammatory cytokines, as well as changes in microglia and astrocytes were analyzed in hippocampus tissues from ABX-, SB-, ABX+SB-treated mice subjected to 4-week DID. We found that ethanol without or with ABX treatment increased mRNA levels of key brain cytokines (MCP-1, TNF-α, IL-1β, IL-6 and IL-10) while SB supplementation prevented these changes. Additionally, SB supplementation prevented changes in microglia, i.e., increase in Iba-1 positive cell number and morphology, and in astrocytes, i.e., decrease in GFAP-positive cell number, induced by combination of ethanol and ABX treatments. Our results suggest that gut microbiota metabolites can influence drinking behavior by modulation of neuroinflammation, highlighting the potential for microbiome-targeting strategies for treatment or prevention of AUD.
Collapse
|
13
|
Wang H, Dong X, Awan MUN, Bai J. Epigenetic mechanisms involved in methamphetamine addiction. Front Pharmacol 2022; 13:984997. [PMID: 36091781 PMCID: PMC9458865 DOI: 10.3389/fphar.2022.984997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Methamphetamine (METH) is an illicit psychostimulant that is widely abused. The molecular mechanism of METH addiction is complicated and still unknown. METH causes the release of the neurotransmitters including dopamine, glutamate, norepinephrine and serotonin, which activate various brain areas in the central nervous system. METH also induces synaptic plasticity and pathological memory enhancement. Epigenetics plays the important roles in regulating METH addiction. This review will briefly summarize the studies on epigenetics involved in METH addiction.
Collapse
|