1
|
Wood EK, Huang E, Sano ER, Loftis JM. Greater fatigue, disturbed sleep, persistent memory problems, and reduced CD4 + T cell and B cell percentages in adults with a history of methamphetamine dependence. J Neuroimmunol 2025; 402:578567. [PMID: 40088605 PMCID: PMC11974374 DOI: 10.1016/j.jneuroim.2025.578567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/31/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025]
Abstract
Methamphetamine (MA) dependence is associated with immunotoxicity and high rates of neuropsychiatric impairments that persist into remission. Although there are currently no FDA-approved pharmacotherapies for MA use disorders, preclinical and clinical studies are beginning to test interventions that directly impact immune signaling. This study was conducted to investigate the relative contribution of immune cell function to the neuropsychiatric sequelae associated with MA dependence and remission. Participants were enrolled into the following study groups: i) control (CTL) group (n = 62): adults with no lifetime history of dependence on any substance other than nicotine or caffeine; and ii) MA group (n = 98) [MA-remission group (n = 55): adults in remission ≥1 month and ≤ 6 months and MA-active group (n = 43): adults actively using MA and meeting criteria for MA dependence]. Participants completed a clinical interview, urine drug analysis, blood sample collection, and questionnaires. Peripheral blood mononuclear cells were analyzed by flow cytometry. Results suggest that early remission from MA dependence is associated with increased fatigue and persistent sleep and prospective and retrospective memory problems, along with reduced B and CD4+ T cell percentages, compared to the CTL group. Preliminary findings support the hypothesis that the immune system modulates the sleep impairments associated with drug actions and provide implications for future research studies and treatment approaches.
Collapse
Affiliation(s)
- Elizabeth K Wood
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; Center for Mental Health Innovation, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.
| | - Elaine Huang
- Cell, Developmental and Cancer Biology Department, Oregon Health & Science University, 3181 Sam Jackson Park Rd, Portland, OR 97239, USA.
| | - Emily R Sano
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd, Portland, OR 97239, USA; Mental Health and Clinical Neurosciences Division, Veterans Affairs Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd, Portland, OR 97239, USA.
| | - Jennifer M Loftis
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd, Portland, OR 97239, USA; Mental Health and Clinical Neurosciences Division, Veterans Affairs Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd, Portland, OR 97239, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.
| |
Collapse
|
2
|
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
|
3
|
Bernardo HT, Lodetti G, de Farias ACS, de Pieri Pickler K, Baldin SL, Dondossola ER, Rico EP. Naltrexone Alters Neurochemical and Behavioral Parameters in a Zebrafish Model of Repeated Alcohol Exposure. Neurochem Res 2025; 50:97. [PMID: 39920352 DOI: 10.1007/s11064-025-04349-3] [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: 10/01/2024] [Revised: 01/04/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
Between the neurotransmission systems modulated by alcohol, the opioid system has been receiving attention in studies that seek to understand its relationship to the effects of addictive substances and different neuropsychiatric disorders. The use of naltrexone stands out in determining the mechanisms of the opioid system, as it acts as an opioid antagonist and consequently generates neurochemical responses. This study aimed to evaluate the pharmacological modulation of opioids on behavioral and neurobiological aspects in adult zebrafish submitted to the protocol of repeated exposure to ethanol and treated with naltrexone. Opioid modulation using naltrexone has been shown to modulate anxiety-like behavior, presenting anxiolytic properties in isolation, in addition to reversing the anxiogenic effect of ethanol through the Novel tank and Light/dark test. Naltrexone increased serotonin and dopamine levels, while ethanol antagonized these effects. In contrast, the interaction between ethanol and naltrexone raised noradrenaline levels. Naltrexone altered glutamate levels, however, ethanol reversed it. Ethanol acted on glutamate transporters increasing their activities, while naltrexone treatment reduced activities. No significant results were found in the pro-oxidant parameters, however, ethanol reduced SOD activity while naltrexone reversed. The same occurred in CAT activity. Also, naltrexone up-regulated the expression of genes related to the dopaminergic, glutamatergic, and opioid systems. The genes used as markers of the inflammatory process and glial activity were modulated by ethanol and together with naltrexone, respectively. Taken together, our findings reinforce the importance of opioid signaling on biochemical and molecular bases related to neuropsychiatric behaviors and diseases, such as anxiety and substance dependence.
Collapse
Affiliation(s)
- Henrique Teza Bernardo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Guilherme Lodetti
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Ana Caroline Salvador de Farias
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Karolyne de Pieri Pickler
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Samira Leila Baldin
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Eduardo Ronconi Dondossola
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Eduardo Pacheco Rico
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
4
|
Castro EM, Lotfipour S, Leslie FM. Neuroglia in substance use disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:347-369. [PMID: 40148055 DOI: 10.1016/b978-0-443-19102-2.00014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Substance use disorders (SUD) remain a major public health concern in which individuals are unable to control their use of substances despite significant harm and negative consequences. Drugs of abuse dysregulate major brain and behavioral functions. Glial cells, primarily microglia and astrocytes, play a crucial role in these drug-induced molecular and behavioral changes. This review explores preclinical and clinical studies of how neuroglia and their associated neuroinflammatory responses contribute to SUD and reward-related properties. We evaluate preclinical and clinical evidence for targeting neuroglia as therapeutic interventions. In addition, we evaluate the literature on the gut microbiome and its role in SUD. Clinical treatments are most effective for reducing drug cravings, and some have yielded promising results in other measures of drug use. N-Acetylcysteine, through modulation of cysteine-glutamate antiporter of glial cells, shows encouraging results across a variety of drug classes. Neuroglia and gut microbiome interactions are important factors to consider with regard to SUD and could lead to novel therapeutic avenues. Age- and sex-dependent properties of neuroglia, gut microbiome, and drug use behaviors are important areas in need of further investigation.
Collapse
Affiliation(s)
- Emily M Castro
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States; Department of Emergency Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States; Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Frances M Leslie
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States.
| |
Collapse
|
5
|
Stoops WW, Shellenberg TP, Regnier SD, Cox DH, Adatorwovor R, Hays LR, Anderson DM, Lile JA, Schmitz JM, Havens JR, Segerstrom SC. Influence of cocaine use reduction on markers of immune function. J Neuroimmunol 2024; 397:578470. [PMID: 39504756 PMCID: PMC11620913 DOI: 10.1016/j.jneuroim.2024.578470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/28/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024]
Abstract
This study determined the effects of reduced cocaine use on immune function. Treatment seeking participants with Cocaine Use Disorder enrolled in a 12-week contingency management trial to reduce cocaine use. Participants were randomly assigned 1:1:1 to High Value Reinforcers (i.e., $55/negative urine sample) for cocaine abstinence (n = 41), Low Value Reinforcers (i.e., $13/negative urine sample) for cocaine abstinence (n = 33) or Non-Contingent Control (n = 33). Immune measures were collected at 6-week intervals. The High Value group had greatest use reductions, increased erythema and IL-6 and decreased IL-10 and CCL5, suggesting an activated immune response. Cocaine use reduction may promote changes in immune health.
Collapse
Affiliation(s)
- William W Stoops
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA; Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA; Department of Psychology, University of Kentucky College of Arts and Sciences, 171 Funkhouser Drive, Lexington, KY 40506-0044, USA; Center on Drug and Alcohol Research, University of Kentucky College of Medicine, 845 Angliana Ave, Lexington, KY 40508, USA.
| | - Thomas P Shellenberg
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA; Department of Psychology, University of Kentucky College of Arts and Sciences, 171 Funkhouser Drive, Lexington, KY 40506-0044, USA
| | - Sean D Regnier
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA
| | - David H Cox
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA
| | - Reuben Adatorwovor
- Department of Biostatistics, University of Kentucky, 725 Rose Street, Multi-disciplinary Science Building, MDS 208D, Lexington, KY 40536-0082, USA
| | - Lon R Hays
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA
| | - Danielle M Anderson
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA
| | - Joshua A Lile
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA; Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA; Department of Psychology, University of Kentucky College of Arts and Sciences, 171 Funkhouser Drive, Lexington, KY 40506-0044, USA
| | - Joy M Schmitz
- Center for Neurobehavioral Research on Addiction (CNRA), University of Texas Houston, 1941 East Road Houston, TX 77054, USA
| | - Jennifer R Havens
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA; Center on Drug and Alcohol Research, University of Kentucky College of Medicine, 845 Angliana Ave, Lexington, KY 40508, USA
| | - Suzanne C Segerstrom
- School of Human Development and Family Studies, Oregon State University, Waldo Hall 453, 2250 SW Jefferson Way, Corvallis, OR 97331, USA
| |
Collapse
|
6
|
Du Y, Zhang J, Cao D, Yang W, Li J, Li D, Song M, Yang Z, Zhang J, Jiang T, Liu J. Neuro-immune communication at the core of craving-associated brain structural network reconfiguration in methamphetamine users. Neuroimage 2024; 301:120883. [PMID: 39384079 DOI: 10.1016/j.neuroimage.2024.120883] [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: 06/17/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024] Open
Abstract
Methamphetamine (MA) use disorder is a chronic neurotoxic brain disease characterized by a high risk of relapse driven by intense cravings. However, the neurobiological signatures of cravings remain unclear, limiting the effectiveness of various treatment methods. Diffusion MRI (dMRI) scans from 62 MA users and 57 healthy controls (HC) were used in this study. The MA users were longitudinally followed up during their period of long-term abstinence (duration of long-term abstinence: 347.52±99.25 days). We systematically quantified the control ability of each brain region for craving-associated state transitions using network control theory from a causal perspective. Craving-associated structural alterations (CSA) were investigated through multivariate group comparisons and biological relevance analysis. The neural mechanisms underlying CSA were elucidated using transcriptomic and neurochemical analyses. We observed that long-term abstinence-induced structural alterations significantly influenced the state transition energy involved in the cognitive control response to external information, which correlated with changes in craving scores (r ∼ 0.35, P <0.01). Our causal network analysis further supported the crucial role of the prefrontal cortex (PFC) in craving mechanisms. Notably, while the PFC is central to the craving, the CSAs were distributed widely across multiple brain regions (PFDR<0.05), with strong alterations in somatomotor regions (PFDR<0.05) and moderate alterations in high-level association networks (PFDR<0.05). Additionally, transcriptomic, chemical compounds, cell-type analyses, and molecular imaging collectively highlight the influence of neuro-immune communication on human craving modulation. Our results offer an integrative, multi-scale perspective on unraveling the neural underpinnings of craving and suggest that neuro-immune signaling may be a promising target for future human addiction therapeutics.
Collapse
Affiliation(s)
- Yanyao Du
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China
| | - Jiaqi Zhang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Dan Cao
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Wenhan Yang
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China
| | - Jin Li
- School of Psychology, Capital Normal University, Beijing 100048, PR China
| | - Deying Li
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ming Song
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Zhengyi Yang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Jun Zhang
- Hunan Judicial Police Academy, Changsha, Hunan 410138, PR China
| | - Tianzi Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, PR China; Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, PR China; Xiaoxiang Institute for Brain Health and Yongzhou Central Hospital, Yongzhou 425000, Hunan Province, PR China.
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, PR China; Clinical Research Center for Medical Imaging in Hunan Province, Changsha, Hunan 410011, China; Department of Radiology Quality Control Center, Changsha, Hunan 410011, China.
| |
Collapse
|
7
|
Lucerne KE, Dean CR, Osman A, Meckel KR, Dave YA, Shipman AL, Cazarez DR, Cathomas F, Hofford RS, Kiraly DD. Colony-stimulating factor 2 (CSF2) as a gut microbiome dependent immune factor that alters molecular and behavioral responses to cocaine in male mice. Brain Behav Immun 2024; 122:137-149. [PMID: 39098439 PMCID: PMC11831408 DOI: 10.1016/j.bbi.2024.08.003] [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: 03/05/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Abstract
Cocaine use disorder is a condition that leads to tremendous morbidity and mortality for which there are currently no FDA-approved pharmacotherapies. Previous research has demonstrated an important role for the resident population of bacteria of the large intestine, collectively dubbed the gut microbiome, in modulating brain and behavior in models of cocaine and other substance use disorders. Importantly, previous work has repeatedly shown that depletion of the gut microbiome leads to increased cocaine taking and seeking behaviors in multiple models. While the precise mechanism of these gut-brain signaling pathways in models of cocaine use is not fully clear, and intriguing possibility is through gut microbiome influences on innate immune system function. In this manuscript we identify the cytokine colony stimulating factor 2 (CSF2) as an immune factor that is increased by cocaine in a gut microbiome dependent manner. Peripherally injected CSF2 crosses the blood-brain barrier into the nucleus accumbens, a brain region central to behavioral responses to cocaine. Treatment with peripheral CSF2 reduces acute and sensitized locomotor responses to cocaine as well as reducing cocaine place preference at high doses. On a molecular level, we find that peripheral injections of CSF2 alter the transcriptional response to both acute and repeated cocaine in the nucleus accumbens. Finally, treatment of microbiome depleted mice with CSF2 reverses the behavioral effects of microbiome depletion on the conditioned place preference assay. Taken together, this work identifies an innate immune factor that represents a novel gut-brain signaling cascade in models of cocaine use and lays the foundations for further translational work targeting this pathway.
Collapse
Affiliation(s)
- Kelsey E Lucerne
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Calista R Dean
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Atrium Wake Forest Baptist Health, Winston-Salem, NC 27101, United States
| | - Aya Osman
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Katherine R Meckel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Yesha A Dave
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Ava L Shipman
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Dannis R Cazarez
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Atrium Wake Forest Baptist Health, Winston-Salem, NC 27101, United States
| | - Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Rebecca S Hofford
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Translational Neuroscience, Wake Forest University School of Medicine, Atrium Wake Forest Baptist Health, Winston-Salem, NC 27101, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Translational Neuroscience, Wake Forest University School of Medicine, Atrium Wake Forest Baptist Health, Winston-Salem, NC 27101, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Psychiatry, Wake Forest University School of Medicine, Atrium Wake Forest Baptist Health, Winston-Salem, NC 27101, United States.
| |
Collapse
|
8
|
Mikhalitskaya EV, Vyalova NM, Bokhan NA, Ivanova SA. Alcohol-Induced Activation of Chemokine System and Neuroinflammation Development. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1889-1903. [PMID: 39647818 DOI: 10.1134/s0006297924110038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 12/10/2024]
Abstract
Chemokines are immunoregulatory proteins with pleiotropic functions involved in neuromodulation, neurogenesis, and neurotransmission. The way chemokines affect the CNS plays an important role in modulating various conditions that could have negative impact on CNS functions, including development of alcohol use disorders. In this review, we analyzed the literature data available on the problem of chemokine participation in pathogenesis, clinical presentation, and remission of alcohol use disorders both in animal models and in the study of patients with alcoholism. The presented information confirms the hypothesis that the alcohol-induced chemokine production could modulate chronic neuroinflammation. Thus, the data summarized and shown in this review are focused on the relevant direction of research in the field of psychiatry, which is in demand by both scientists and clinical specialists.
Collapse
Affiliation(s)
- Ekaterina V Mikhalitskaya
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia.
| | - Natalya M Vyalova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia
| | - Nikolay A Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia
| |
Collapse
|
9
|
Hoprekstad GE, Skrede S, Bartz-Johannessen C, Joa I, Reitan SK, Steen VM, Torsvik A, Johnsen E, Kroken RA, Rettenbacher M. Association between cytokines and suicidality in patients with psychosis: A multicentre longitudinal analysis. Brain Behav Immun Health 2024; 37:100756. [PMID: 38549611 PMCID: PMC10973600 DOI: 10.1016/j.bbih.2024.100756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 03/17/2024] [Indexed: 11/27/2024] Open
Abstract
Suicide is a common cause of death in all phases of schizophrenia spectrum disorder, particularly in the youngest patients. Clinical measures have demonstrated limited value in suicide prediction, spurring the search for potential biomarkers. The causes of suicidal behaviour are complex, but the immune system seems to be involved as it reflects or even causes mental suffering. We aimed to identify cytokines with associations to suicidality in a sample of patients with symptoms of active psychosis. Patients with schizophrenia spectrum disorder (N = 144) participating in a semi-randomized antipsychotic drug trial (the BeSt InTro study) were assessed with the Positive and Negative Syndrome Scale (PANSS) and the Calgary Depression Scale for Schizophrenia (CDSS) at eight visits across 12 months. The Clinical Global Impression for Severity of Suicidality scale (CGI-SS) was used for assessing suicidality. Serum concentrations of tumour necrosis factor (TNF)-alpha, interferon (IFN)-gamma, interleukin (IL)-1beta, IL-2, IL-4, IL-6, and IL-10 were measured using immunoassays. A logistic regression model was used to investigate the association between cytokine levels and suicidality. To enhance clinical significance, the CGI-SS scores were dichotomized into two groups before analyses: low (=1) and high (≥2) risk for suicidality. Both uni- and multi-variate analyses revealed an inverse correlation between IL-2 and IL-10 serum levels and suicidality, where lower cytokine concentrations of IL-2 and IL-10 were associated with higher suicidality scores. The results were consistent when adjusted for depression and substance use. These results indicate that inflammatory processes are linked to the risk of suicidality in patients with schizophrenia spectrum disorders.
Collapse
Affiliation(s)
- Gunnhild E. Hoprekstad
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Haukeland University Hospital, Bergen, Norway
| | - Silje Skrede
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Christoffer Bartz-Johannessen
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Haukeland University Hospital, Bergen, Norway
| | - Inge Joa
- TIPS, Network for Clinical Research in Psychosis, Stavanger University Hospital, Stavanger, Norway
- Department of Public Health, Faculty of Health Science, University of Stavanger, Stavanger, Norway
| | - Solveig K. Reitan
- St. Olav's University Hospital, Department of Mental Health, Nidelv DPS, Trondheim, Norway
- Norwegian University of Science and Technology, Department of Mental Health, Trondheim, Norway
| | - Vidar M. Steen
- Norwegian Centre for Mental Disorders Research (NORMENT), Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, 5020, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Anja Torsvik
- Norwegian Centre for Mental Disorders Research (NORMENT), Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, 5020, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Erik Johnsen
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Haukeland University Hospital, Bergen, Norway
| | - Rune A. Kroken
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Haukeland University Hospital, Bergen, Norway
| | | |
Collapse
|
10
|
Nusslock R, Alloy LB, Brody GH, Miller GE. Annual Research Review: Neuroimmune network model of depression: a developmental perspective. J Child Psychol Psychiatry 2024; 65:538-567. [PMID: 38426610 PMCID: PMC11090270 DOI: 10.1111/jcpp.13961] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2024] [Indexed: 03/02/2024]
Abstract
Depression is a serious public health problem, and adolescence is an 'age of risk' for the onset of Major Depressive Disorder. Recently, we and others have proposed neuroimmune network models that highlight bidirectional communication between the brain and the immune system in both mental and physical health, including depression. These models draw on research indicating that the cellular actors (particularly monocytes) and signaling molecules (particularly cytokines) that orchestrate inflammation in the periphery can directly modulate the structure and function of the brain. In the brain, inflammatory activity heightens sensitivity to threats in the cortico-amygdala circuit, lowers sensitivity to rewards in the cortico-striatal circuit, and alters executive control and emotion regulation in the prefrontal cortex. When dysregulated, and particularly under conditions of chronic stress, inflammation can generate feelings of dysphoria, distress, and anhedonia. This is proposed to initiate unhealthy, self-medicating behaviors (e.g. substance use, poor diet) to manage the dysphoria, which further heighten inflammation. Over time, dysregulation in these brain circuits and the inflammatory response may compound each other to form a positive feedback loop, whereby dysregulation in one organ system exacerbates the other. We and others suggest that this neuroimmune dysregulation is a dynamic joint vulnerability for depression, particularly during adolescence. We have three goals for the present paper. First, we extend neuroimmune network models of mental and physical health to generate a developmental framework of risk for the onset of depression during adolescence. Second, we examine how a neuroimmune network perspective can help explain the high rates of comorbidity between depression and other psychiatric disorders across development, and multimorbidity between depression and stress-related medical illnesses. Finally, we consider how identifying neuroimmune pathways to depression can facilitate a 'next generation' of behavioral and biological interventions that target neuroimmune signaling to treat, and ideally prevent, depression in youth and adolescents.
Collapse
Affiliation(s)
- Robin Nusslock
- Department of Psychology, Northwestern University, Evanston IL, USA
- Institute for Policy Research, Northwestern University, Evanston IL, USA
| | - Lauren B. Alloy
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA. USA
| | - Gene H. Brody
- Center for Family Research, University of Georgia, Athens GA, USA
| | - Gregory E. Miller
- Department of Psychology, Northwestern University, Evanston IL, USA
- Institute for Policy Research, Northwestern University, Evanston IL, USA
| |
Collapse
|
11
|
Steinfeld MR, Torregrossa MM. Consequences of adolescent drug use. Transl Psychiatry 2023; 13:313. [PMID: 37802983 PMCID: PMC10558564 DOI: 10.1038/s41398-023-02590-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/22/2023] [Accepted: 08/23/2023] [Indexed: 10/08/2023] Open
Abstract
Substance use in adolescence is a known risk factor for the development of neuropsychiatric and substance use disorders in adulthood. This is in part due to the fact that critical aspects of brain development occur during adolescence, which can be altered by drug use. Despite concerted efforts to educate youth about the potential negative consequences of substance use, initiation remains common amongst adolescents world-wide. Additionally, though there has been substantial research on the topic, many questions remain about the predictors and the consequences of adolescent drug use. In the following review, we will highlight some of the most recent literature on the neurobiological and behavioral effects of adolescent drug use in rodents, non-human primates, and humans, with a specific focus on alcohol, cannabis, nicotine, and the interactions between these substances. Overall, consumption of these substances during adolescence can produce long-lasting changes across a variety of structures and networks which can have enduring effects on behavior, emotion, and cognition.
Collapse
Affiliation(s)
- Michael R Steinfeld
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA.
| | - Mary M Torregrossa
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
- Center for Neuroscience, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15213, USA
| |
Collapse
|
12
|
Ibáñez C, Acuña T, Quintanilla ME, Pérez-Reytor D, Morales P, Karahanian E. Fenofibrate Decreases Ethanol-Induced Neuroinflammation and Oxidative Stress and Reduces Alcohol Relapse in Rats by a PPAR-α-Dependent Mechanism. Antioxidants (Basel) 2023; 12:1758. [PMID: 37760061 PMCID: PMC10525752 DOI: 10.3390/antiox12091758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
High ethanol consumption triggers neuroinflammation, implicated in sustaining chronic alcohol use. This inflammation boosts glutamate, prompting dopamine release in reward centers, driving prolonged drinking and relapse. Fibrate drugs, activating peroxisome proliferator-activated receptor alpha (PPAR-α), counteract neuroinflammation in other contexts, prompting investigation into their impact on ethanol-induced inflammation. Here, we studied, in UChB drinker rats, whether the administration of fenofibrate in the withdrawal stage after chronic ethanol consumption reduces voluntary intake when alcohol is offered again to the animals (relapse-type drinking). Furthermore, we determined if fenofibrate was able to decrease ethanol-induced neuroinflammation and oxidative stress in the brain. Animals treated with fenofibrate decreased alcohol consumption by 80% during post-abstinence relapse. Furthermore, fenofibrate decreased the expression of the proinflammatory cytokines tumor necrosis factor-alpha (TNF-α) and interleukins IL-1β and IL-6, and of an oxidative stress-induced gene (heme oxygenase-1), in the hippocampus, nucleus accumbens, and prefrontal cortex. Animals treated with fenofibrate showed an increase M2-type microglia (with anti-inflammatory proprieties) and a decrease in phagocytic microglia in the hippocampus. A PPAR-α antagonist (GW6471) abrogated the effects of fenofibrate, indicating that they are dependent on PPAR-α activation. These findings highlight the potential of fenofibrate, an FDA-approved dyslipidemia medication, as a supplementary approach to alleviating relapse severity in individuals with alcohol use disorder (AUD) during withdrawal.
Collapse
Affiliation(s)
- Cristina Ibáñez
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.I.); (D.P.-R.)
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 8910060, Chile;
| | - Tirso Acuña
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile;
| | - María Elena Quintanilla
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 8910060, Chile;
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile;
| | - Diliana Pérez-Reytor
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.I.); (D.P.-R.)
| | - Paola Morales
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 8910060, Chile;
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile;
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Eduardo Karahanian
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.I.); (D.P.-R.)
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 8910060, Chile;
| |
Collapse
|
13
|
Alloy LB, Chat IKY, Grehl MM, Stephenson AR, Adogli ZV, Olino TM, Ellman LM, Miller GE, Nusslock R. Reward and Immune Systems in Emotion (RISE) prospective longitudinal study: Protocol overview of an integrative reward-inflammation model of first onset of major depression in adolescence. Brain Behav Immun Health 2023; 30:100643. [PMID: 37304334 PMCID: PMC10250584 DOI: 10.1016/j.bbih.2023.100643] [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: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/13/2023] Open
Abstract
Background Depression is associated with a reduced sensitivity to rewards and low reward-related brain function in cortico-striatal circuitry. A separate literature documents elevated peripheral inflammation in depression. Recently, integrated reward-inflammation models of depression have been proposed. These models draw on work indicating that peripheral inflammatory proteins access the brain, where they lower reward responsiveness. This blunted reward responsiveness is proposed to initiate unhealthy behaviors (substance use, poor diet), as well as sleep disruption and stress generation, which further heighten inflammation. Over time, dysregulation in reward responsiveness and immune signaling may synergize in a positive feedback loop, whereby dysregulation in each system exacerbates dysregulation in the other. Project RISE (Reward and Immune Systems in Emotion) provides a first systematic test of reward-immune dysregulation as a synergistic and dynamic vulnerability for first onset of major depressive disorder and increases in depressive symptoms during adolescence. Methods This NIMH-funded R01 study is a 3-year prospective, longitudinal investigation of approximately 300 community adolescents from the broader Philadelphia area, United States of America. Eligible participants must be 13-16 years old, fluent in English, and without a prior major depressive disorder. They are being selected along the entire dimension of self-reported reward responsiveness, with oversampling at the low tail of the dimension in order to increase the likelihood of major depression onsets. At Time 1 (T1), T3, and T5, each a year apart, participants complete blood draws to quantify biomarkers of low-grade inflammation, self-report and behavioral measures of reward responsiveness, and fMRI scans of reward neural activity and functional connectivity. At T1-T5 (with T2 and T4 six months between the yearly sessions), participants also complete diagnostic interviews and measures of depressive symptoms, reward-relevant life events, and behaviors that increase inflammation. Adversity history is assessed at T1 only. Discussion This study is an innovative integration of research on multi-organ systems involved in reward and inflammatory signaling in understanding first onset of major depression in adolescence. It has the potential to facilitate novel neuroimmune and behavioral interventions to treat, and ideally prevent, depression.
Collapse
Affiliation(s)
- Lauren B. Alloy
- Department of Psychology and Neuroscience, Temple University, USA
| | - Iris K.-Y. Chat
- Department of Psychology and Neuroscience, Temple University, USA
| | - Mora M. Grehl
- Department of Psychology and Neuroscience, Temple University, USA
| | | | - Zoe V. Adogli
- Department of Psychology and Neuroscience, Temple University, USA
| | - Thomas M. Olino
- Department of Psychology and Neuroscience, Temple University, USA
| | - Lauren M. Ellman
- Department of Psychology and Neuroscience, Temple University, USA
| | | | | |
Collapse
|
14
|
Isaza C, Castaño-Ramírez OM, Vélez JP, Henao J, Beltrán-Angarita L, Sepúlveda-Arias JC. Influence of CYP2B6 Genotype on Methadone Dosage in Patients from the Methadone Maintenance Treatment (MMT) Program in Pereira, Colombia. Life (Basel) 2023; 13:life13041038. [PMID: 37109567 PMCID: PMC10145268 DOI: 10.3390/life13041038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Methadone treatment reduces the use of heroin and withdrawal symptoms; however, methadone is an expensive medication with a narrow safety margin. We compared the retention rates, persistence of heroin use, and quality of life of a group of patients undergoing conventional Methadone Maintenance Treatment (MMT) with a group for whom the CYP2B6 516G>T polymorphism was used in addition to the MMT to calculate the required methadone dose. Over 12 weeks, the retention rate, heroin usage, and quality of life of patients under conventional treatment (n = 34) were compared with those of patients for whom we used genetic markers to calculate methadone dosage (n = 38). At the end of the study, 26.4% of patients abandoned the program, and neither demographic nor clinical variables were associated with treatment adherence. Of the remaining patients, 16% of the control group and 8% of patients in the pharmacogenetic group reported heroin use, while both groups showed a 64% reduction in the use of cocaine/crack (no significant differences between the groups were found). Starting in the second week, the methadone dosage was lower among the patients for whom methadone was prescribed based on genotype. Although there were six individuals in the control group and three in the pharmacogenetic group with QTc intervals > 450 ms (a threshold that is considered dangerous), we did not find a relationship between the QTc interval and methadone dosage. There were no differences in the perception of quality of life between the two groups. The results of this pilot study suggest that concerning methadone therapy, the CYP2B6 genotype contributes to reduced effective doses and treatment costs.
Collapse
Affiliation(s)
- Carlos Isaza
- Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| | | | | | - Julieta Henao
- Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| | | | - Juan Carlos Sepúlveda-Arias
- Grupo Infección e Inmunidad, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira 660003, Colombia
| |
Collapse
|
15
|
ElShebiney S, Elgohary R, El-Shamarka M, Mowaad N, Abulseoud OA. Natural Polyphenols-Resveratrol, Quercetin, Magnolol, and β-Catechin-Block Certain Aspects of Heroin Addiction and Modulate Striatal IL-6 and TNF-α. TOXICS 2023; 11:379. [PMID: 37112606 PMCID: PMC10145039 DOI: 10.3390/toxics11040379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 06/19/2023]
Abstract
We have examined the effects of four different polyphenols in attenuating heroin addiction using a conditioned place preference (CPP) paradigm. Adult male Sprague Dawley rats received heroin (alternating with saline) in escalating doses starting from 10 mg/kg, i.p. up to 80 mg/kg/d for 14 consecutive days. The rats were treated with distilled water (1 mL), quercetin (50 mg/kg/d), β-catechin (100 mg/kg/d), resveratrol (30 mg/kg/d), or magnolol (50 mg/kg/d) through oral gavage for 7 consecutive days, 30 min before heroin administration, starting on day 8. Heroin withdrawal manifestations were assessed 24 h post last heroin administration following the administration of naloxone (1 mg/kg i.p). Heroin CPP reinstatement was tested following a single dose of heroin (10 mg/kg i.p.) administration. Striatal interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) were quantified (ELISA) after naloxone-precipitated heroin withdrawal. Compared to the vehicle, the heroin-administered rats spent significantly more time in the heroin-paired chamber (p < 0.0001). Concomitant administration of resveratrol and quercetin prevented the acquisition of heroin CPP, while resveratrol, quercetin, and magnolol blocked heroin-triggered reinstatement. Magnolol, quercetin, and β-catechin blocked naloxone-precipitated heroin withdrawal and increased striatal IL-6 concentration (p < 0.01). Resveratrol administration was associated with significantly higher withdrawal scores compared to those of the control animals (p < 0.0001). The results of this study show that different polyphenols target specific behavioral domains of heroin addiction in a CPP model and modulate the increase in striatal inflammatory cytokines TNF-α and IL-6 observed during naloxone-precipitated heroin withdrawal. Further research is needed to study the clinical utility of polyphenols and to investigate the intriguing finding that resveratrol enhances, rather than attenuates naloxone-precipitated heroin withdrawal.
Collapse
Affiliation(s)
- Shaimaa ElShebiney
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Rania Elgohary
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Marwa El-Shamarka
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Noha Mowaad
- Department of Narcotics, Ergogenics, and Poisons, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Osama A. Abulseoud
- Department of Psychiatry and Psychology, Mayo Clinic, Phoenix, AZ 85001, USA
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ 85001, USA
| |
Collapse
|
16
|
VanderZwaag J, Halvorson T, Dolhan K, Šimončičová E, Ben-Azu B, Tremblay MÈ. The Missing Piece? A Case for Microglia's Prominent Role in the Therapeutic Action of Anesthetics, Ketamine, and Psychedelics. Neurochem Res 2023; 48:1129-1166. [PMID: 36327017 DOI: 10.1007/s11064-022-03772-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
There is much excitement surrounding recent research of promising, mechanistically novel psychotherapeutics - psychedelic, anesthetic, and dissociative agents - as they have demonstrated surprising efficacy in treating central nervous system (CNS) disorders, such as mood disorders and addiction. However, the mechanisms by which these drugs provide such profound psychological benefits are still to be fully elucidated. Microglia, the CNS's resident innate immune cells, are emerging as a cellular target for psychiatric disorders because of their critical role in regulating neuroplasticity and the inflammatory environment of the brain. The following paper is a review of recent literature surrounding these neuropharmacological therapies and their demonstrated or hypothesized interactions with microglia. Through investigating the mechanism of action of psychedelics, such as psilocybin and lysergic acid diethylamide, ketamine, and propofol, we demonstrate a largely under-investigated role for microglia in much of the emerging research surrounding these pharmacological agents. Among others, we detail sigma-1 receptors, serotonergic and γ-aminobutyric acid signalling, and tryptophan metabolism as pathways through which these agents modulate microglial phagocytic activity and inflammatory mediator release, inducing their therapeutic effects. The current review includes a discussion on future directions in the field of microglial pharmacology and covers bidirectional implications of microglia and these novel pharmacological agents in aging and age-related disease, glial cell heterogeneity, and state-of-the-art methodologies in microglial research.
Collapse
Affiliation(s)
- Jared VanderZwaag
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Torin Halvorson
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Vancouver, BC, Canada
- Department of Biology, University of Victoria, Vancouver, BC, Canada
| | - Eva Šimončičová
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de médecine moléculaire, Université Laval, Québec City, QC, Canada.
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
17
|
Yen E, Gaddis N, Jantzie L, Davis JM. A review of the genomics of neonatal abstinence syndrome. Front Genet 2023; 14:1140400. [PMID: 36845389 PMCID: PMC9950123 DOI: 10.3389/fgene.2023.1140400] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Neonatal abstinence syndrome (NAS) is a constellation of signs of withdrawal occurring after birth following in utero exposure to licit or illicit opioids. Despite significant research and public health efforts, NAS remains challenging to diagnose, predict, and manage due to highly variable expression. Biomarker discovery in the field of NAS is crucial for stratifying risk, allocating resources, monitoring longitudinal outcomes, and identifying novel therapeutics. There is considerable interest in identifying important genetic and epigenetic markers of NAS severity and outcome that can guide medical decision making, research efforts, and public policy. A number of recent studies have suggested that genetic and epigenetic changes are associated with NAS severity, including evidence of neurodevelopmental instability. This review will provide an overview of the role of genetics and epigenetics in short and longer-term NAS outcomes. We will also describe novel research efforts using polygenic risk scores for NAS risk stratification and salivary gene expression to understand neurobehavioral modulation. Finally, emerging research focused on neuroinflammation from prenatal opioid exposure may elucidate novel mechanisms that could lead to development of future novel therapeutics.
Collapse
Affiliation(s)
- Elizabeth Yen
- Department of Pediatrics, Tufts Medical Center, Boston, MA, United States
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, United States
- Tufts University School of Medicine, Boston, MA, United States
| | - Nathan Gaddis
- Research Triangle Institute International, Research Triangle Park, Durham, NC, United States
| | - Lauren Jantzie
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan M. Davis
- Department of Pediatrics, Tufts Medical Center, Boston, MA, United States
- Tufts University School of Medicine, Boston, MA, United States
- Tufts Clinical and Translational Sciences Institute, Boston, MA, United States
| |
Collapse
|
18
|
De Simone U, Pignatti P, Villani L, Russo LA, Sargenti A, Bonetti S, Buscaglia E, Coccini T. Human Astrocyte Spheroids as Suitable In Vitro Screening Model to Evaluate Synthetic Cannabinoid MAM2201-Induced Effects on CNS. Int J Mol Sci 2023; 24:ijms24021421. [PMID: 36674936 PMCID: PMC9861655 DOI: 10.3390/ijms24021421] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
There is growing concern about the consumption of synthetic cannabinoids (SCs), one of the largest groups of new psychoactive substances, its consequence on human health (general population and workers), and the continuous placing of new SCs on the market. Although drug-induced alterations in neuronal function remain an essential component for theories of drug addiction, accumulating evidence indicates the important role of activated astrocytes, whose essential and pleiotropic role in brain physiology and pathology is well recognized. The study aims to clarify the mechanisms of neurotoxicity induced by one of the most potent SCs, named MAM-2201 (a naphthoyl-indole derivative), by applying a novel three-dimensional (3D) cell culture model, mimicking the physiological and biochemical properties of brain tissues better than traditional two-dimensional in vitro systems. Specifically, human astrocyte spheroids, generated from the D384 astrocyte cell line, were treated with different MAM-2201 concentrations (1-30 µM) and exposure times (24-48 h). MAM-2201 affected, in a concentration- and time-dependent manner, the cell growth and viability, size and morphological structure, E-cadherin and extracellular matrix, CB1-receptors, glial fibrillary acidic protein, and caspase-3/7 activity. The findings demonstrate MAM-2201-induced cytotoxicity to astrocyte spheroids, and support the use of this human 3D cell-based model as species-specific in vitro tool suitable for the evaluation of neurotoxicity induced by other SCs.
Collapse
Affiliation(s)
- Uliana De Simone
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Patrizia Pignatti
- Allergy and Immunology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Laura Villani
- Pathology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | | | | | - Simone Bonetti
- CNR-ISMN, Institute for Nanostructured Materials, 40129 Bologna, Italy
| | - Eleonora Buscaglia
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Teresa Coccini
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
- Correspondence: ; Tel.: +39-0382-592416
| |
Collapse
|
19
|
Tan H, Hubertus S, Thomas S, Lee AM, Gerhardt S, Gerchen MF, Sommer WH, Kiefer F, Schad L, Vollstädt-Klein S. Association between iron accumulation in the dorsal striatum and compulsive drinking in alcohol use disorder. Psychopharmacology (Berl) 2023; 240:249-257. [PMID: 36577866 PMCID: PMC9879829 DOI: 10.1007/s00213-022-06301-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022]
Abstract
RATIONALE Brain iron accumulation has been observed in neuropsychiatric disorders and shown to be related to neurodegeneration. OBJECTIVES In this study, we used quantitative susceptibility mapping (QSM), an emerging MRI technique developed for quantifying tissue magnetic susceptibility, to examine brain iron accumulation in individuals with alcohol use disorder (AUD) and its relation to compulsive drinking. METHODS Based on our previous projects, QSM was performed as a secondary analysis with gradient echo sequence images, in 186 individuals with AUD and 274 healthy participants. Whole-brain susceptibility values were calculated with morphology-enabled dipole inversion and referenced to the cerebrospinal fluid. Then, the susceptibility maps were compared between AUD individuals and healthy participants. The relationship between drinking patterns and susceptibility was explored. RESULTS Whole-brain analyses showed that the susceptibility in the dorsal striatum (putamen and caudate) among AUD individuals was higher than healthy participants and was positively related to the Obsessive Compulsive Drinking Scale (OCDS) scores and the amount of drinking in the past three months. CONCLUSIONS Increased susceptibility suggests higher iron accumulation in the dorsal striatum in AUD. This surrogate for the brain iron level was linearly associated with the compulsive drinking pattern and the recent amount of drinking, which provides us a new clinical perspective in relation to brain iron accumulation, and also might indicate an association of AUD with neuroinflammation as a consequence of brain iron accumulation. The iron accumulation in the striatum is further relevant for functional imaging studies in AUD by potentially producing signal dropout and artefacts in fMRI images.
Collapse
Affiliation(s)
- Haoye Tan
- grid.7700.00000 0001 2190 4373Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Medical Faculty of Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Simon Hubertus
- grid.7700.00000 0001 2190 4373Computer Assisted Clinical Medicine, Medical Faculty of Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Sebastian Thomas
- grid.7700.00000 0001 2190 4373Computer Assisted Clinical Medicine, Medical Faculty of Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Alycia M. Lee
- grid.7700.00000 0001 2190 4373Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Medical Faculty of Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Sarah Gerhardt
- grid.7700.00000 0001 2190 4373Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Medical Faculty of Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Martin Fungisai Gerchen
- grid.7700.00000 0001 2190 4373Department of Clinical Psychology, Central Institute of Mental Health, Medical Faculty of Mannheim, Heidelberg University, 68159 Mannheim, Germany ,grid.455092.fBernstein Center for Computational Neuroscience Heidelberg/Mannheim, 68159 Mannheim, Germany ,grid.7700.00000 0001 2190 4373Department of Psychology, Heidelberg University, 69117 Heidelberg, Germany
| | - Wolfgang H. Sommer
- grid.7700.00000 0001 2190 4373Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Medical Faculty of Mannheim, Heidelberg University, 68159 Mannheim, Germany ,grid.7700.00000 0001 2190 4373Institute of Psychopharmacology, Central Institute of Mental Health, Heidelberg University, 68159 Mannheim, Germany ,Bethania Hospital for Psychiatry, Psychosomatics, and Psychotherapy, Greifswald, Germany
| | - Falk Kiefer
- grid.7700.00000 0001 2190 4373Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Medical Faculty of Mannheim, Heidelberg University, 68159 Mannheim, Germany ,grid.7700.00000 0001 2190 4373Mannheim Center for Translational Neurosciences (MCTN), Medical Faculty of Mannheim, Heidelberg University, 68159 Mannheim, Germany ,grid.7700.00000 0001 2190 4373Feuerlein Center on Translational Addiction Medicine, Heidelberg University, 69117 Heidelberg, Germany
| | - Lothar Schad
- grid.7700.00000 0001 2190 4373Computer Assisted Clinical Medicine, Medical Faculty of Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Sabine Vollstädt-Klein
- Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Medical Faculty of Mannheim, Heidelberg University, 68159, Mannheim, Germany. .,Mannheim Center for Translational Neurosciences (MCTN), Medical Faculty of Mannheim, Heidelberg University, 68159, Mannheim, Germany.
| |
Collapse
|
20
|
Martinez SS, Stebliankin V, Hernandez J, Martin H, Tamargo J, Rodriguez JB, Teeman C, Johnson A, Seminario L, Campa A, Narasimhan G, Baum MK. Multiomic analysis reveals microbiome-related relationships between cocaine use and metabolites. AIDS 2022; 36:2089-2099. [PMID: 36382433 PMCID: PMC9673179 DOI: 10.1097/qad.0000000000003363] [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] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Over 19 million individuals globally have a cocaine use disorder, a significant public health crisis. Cocaine has also been associated with a pro-inflammatory state and recently with imbalances in the intestinal microbiota as compared to nonuse. The objective of this pilot study was to characterize the gut microbiota and plasma metabolites in people with HIV (PWH) who use cocaine compared with those who do not. DESIGN Cross-sectional study. METHODS A pilot study in PWH was conducted on 25 cocaine users and 25 cocaine nonusers from the Miami Adult Studies on HIV cohort. Stool samples and blood plasma were collected. Bacterial composition was characterized using 16S rRNA sequencing. Metabolomics in plasma were determined using gas and liquid chromatography/mass spectrometry. RESULTS The relative abundances of the Lachnopspira genus, Oscillospira genus, Bifidobacterium adolescentis species, and Euryarchaeota phylum were significantly higher in the cocaine- using PWH compared to cocaine-nonusing PWH. Cocaine-use was associated with higher levels of several metabolites: products of dopamine catabolism (3-methoxytyrosine and 3-methoxytyramine sulfate), phenylacetate, benzoate, butyrate, and butyrylglycine. CONCLUSIONS Cocaine use was associated with higher abundances of taxa and metabolites known to be associated with pathogenic states that include gastrointestinal conditions. Understanding key intestinal bacterial functional pathways that are altered due to cocaine use in PWH will provide a better understanding of the relationships between the host intestinal microbiome and potentially provide novel treatments to improve health.
Collapse
Affiliation(s)
| | - Vitalii Stebliankin
- Florida International University, Bioinformatics Research Group (BioRG), Miami, FL, USA
| | - Jacqueline Hernandez
- Florida International University, R. Stempel College of Public Health and Social Work
| | - Haley Martin
- Florida International University, R. Stempel College of Public Health and Social Work
| | - Javier Tamargo
- Florida International University, R. Stempel College of Public Health and Social Work
| | | | - Colby Teeman
- Florida International University, R. Stempel College of Public Health and Social Work
| | - Angelique Johnson
- Florida International University, R. Stempel College of Public Health and Social Work
| | - Leslie Seminario
- Florida International University, R. Stempel College of Public Health and Social Work
| | - Adriana Campa
- Florida International University, R. Stempel College of Public Health and Social Work
| | - Giri Narasimhan
- Florida International University, Bioinformatics Research Group (BioRG), Miami, FL, USA
| | - Marianna K Baum
- Florida International University, R. Stempel College of Public Health and Social Work
| |
Collapse
|
21
|
Smiley CE, Wood SK. Stress- and drug-induced neuroimmune signaling as a therapeutic target for comorbid anxiety and substance use disorders. Pharmacol Ther 2022; 239:108212. [PMID: 35580690 DOI: 10.1016/j.pharmthera.2022.108212] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
Stress and substance use disorders remain two of the most highly prevalent psychiatric conditions and are often comorbid. While individually these conditions have a debilitating impact on the patient and a high cost to society, the symptomology and treatment outcomes are further exacerbated when they occur together. As such, there are few effective treatment options for these patients, and recent investigation has sought to determine the neural processes underlying the co-occurrence of these disorders to identify novel treatment targets. One such mechanism that has been linked to stress- and addiction-related conditions is neuroimmune signaling. Increases in inflammatory factors across the brain have been heavily implicated in the etiology of these disorders, and this review seeks to determine the nature of this relationship. According to the "dual-hit" hypothesis, also referred to as neuroimmune priming, prior exposure to either stress or drugs of abuse can sensitize the neuroimmune system to be hyperresponsive when exposed to these insults in the future. This review completes an examination of the literature surrounding stress-induced increases in inflammation across clinical and preclinical studies along with a summarization of the evidence regarding drug-induced alterations in inflammatory factors. These changes in neuroimmune profiles are also discussed within the context of their impact on the neural circuitry responsible for stress responsiveness and addictive behaviors. Further, this review explores the connection between neuroimmune signaling and susceptibility to these conditions and highlights the anti-inflammatory pharmacotherapies that may be used for the treatment of stress and substance use disorders.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| |
Collapse
|
22
|
Jiménez-González A, Gómez-Acevedo C, Ochoa-Aguilar A, Chavarría A. The Role of Glia in Addiction: Dopamine as a Modulator of Glial Responses in Addiction. Cell Mol Neurobiol 2022; 42:2109-2120. [PMID: 34057683 PMCID: PMC11421599 DOI: 10.1007/s10571-021-01105-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
Addiction is a chronic and potentially deadly disease considered a global health problem. Nevertheless, there is still no ideal treatment for its management. The alterations in the reward system are the most known pathophysiological mechanisms. Dopamine is the pivotal neurotransmitter involved in neuronal drug reward mechanisms and its neuronal mechanisms have been intensely investigated in recent years. However, neuroglial interactions and their relation to drug addiction development and maintenance of drug addiction have been understudied. Many reports have found that most neuroglial cells express dopamine receptors and that dopamine activity may induce neuroimmunomodulatory effects. Furthermore, current research has also shown that pro- and anti-inflammatory molecules modulate dopaminergic neuron activity. Thus, studying the immune mechanisms of dopamine associated with drug abuse is vital in researching new pathophysiological mechanisms and new therapeutic targets for addiction management.
Collapse
Affiliation(s)
- Ariadna Jiménez-González
- Laboratorio de Biomembranas, Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Claudia Gómez-Acevedo
- Laboratorio de Biomembranas, Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Abraham Ochoa-Aguilar
- Plan de Estudios Combinados en Medicina, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Anahí Chavarría
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
23
|
Margiani G, Castelli MP, Pintori N, Frau R, Ennas MG, Pagano Zottola AC, Orrù V, Serra V, Fiorillo E, Fadda P, Marsicano G, De Luca MA. Adolescent self-administration of the synthetic cannabinoid receptor agonist JWH-018 induces neurobiological and behavioral alterations in adult male mice. Psychopharmacology (Berl) 2022; 239:3083-3102. [PMID: 35943523 PMCID: PMC9481487 DOI: 10.1007/s00213-022-06191-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 07/10/2022] [Indexed: 12/02/2022]
Abstract
RATIONALE The use of synthetic cannabinoid receptor agonists (SCRAs) is growing among adolescents, posing major medical and psychiatric risks. JWH-018 represents the reference compound of SCRA-containing products. OBJECTIVES This study was performed to evaluate the enduring consequences of adolescent voluntary consumption of JWH-018. METHODS The reinforcing properties of JWH-018 were characterized in male CD1 adolescent mice by intravenous self-administration (IVSA). Afterwards, behavioral, neurochemical, and molecular evaluations were performed at adulthood. RESULTS Adolescent mice acquired operant behavior (lever pressing, Fixed Ratio 1-3; 7.5 µg/kg/inf); this behavior was specifically directed at obtaining JWH-018 since it increased under Progressive Ratio schedule of reinforcement, and was absent in vehicle mice. JWH-018 IVSA was reduced by pretreatment of the CB1-antagonist/inverse agonist AM251. Adolescent exposure to JWH-018 by IVSA increased, at adulthood, both nestlet shredding and marble burying phenotypes, suggesting long-lasting repetitive/compulsive-like behavioral effects. JWH-018 did not affect risk proclivity in the wire-beam bridge task. In adult brains, there was an increase of ionized calcium binding adaptor molecule 1 (IBA-1) positive cells in the caudate-putamen (CPu) and nucleus accumbens (NAc), along with a decrease of glial fibrillary acidic protein (GFAP) immunoreactivity in the CPu. These glial alterations in adult brains were coupled with an increase of the chemokine RANTES and a decrease of the cytokines IL2 and IL13 in the cortex, and an increase of the chemokine MPC1 in the striatum. CONCLUSIONS This study suggests for the first time that male mice self-administer the prototypical SCRA JWH-018 during adolescence. The adolescent voluntary consumption of JWH-018 leads to long-lasting behavioral and neurochemical aberrations along with glia-mediated inflammatory responses in adult brains.
Collapse
Affiliation(s)
- Giulia Margiani
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | | | - Nicholas Pintori
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.,"Guy Everett" Laboratory, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Maria Grazia Ennas
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Antonio C Pagano Zottola
- INSERM, U1215 NeuroCentre Magendie, Bordeaux, France.,University of Bordeaux, Bordeaux, France.,Institut de Biochimie et Génétique Cellulaires, UMR 5095, Bordeaux, France
| | - Valeria Orrù
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Valentina Serra
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Edoardo Fiorillo
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Lanusei, Italy
| | - Paola Fadda
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.,Institute of Neuroscience-Cagliari, National Research Council (CNR), Cagliari, Italy
| | - Giovanni Marsicano
- INSERM, U1215 NeuroCentre Magendie, Bordeaux, France.,University of Bordeaux, Bordeaux, France
| | | |
Collapse
|
24
|
Wang TY, Lu RB, Lee SY, Chang YH, Chen SL, Tsai TY, Tseng HH, Chen PS, Chen KC, Yang YK, Hong JS. Association Between Inflammatory Cytokines, Executive Function, and Substance Use in Patients With Opioid Use Disorder and Amphetamine-Type Stimulants Use Disorder. Int J Neuropsychopharmacol 2022; 26:42-51. [PMID: 36181736 PMCID: PMC9850661 DOI: 10.1093/ijnp/pyac069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/25/2022] [Accepted: 09/30/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Long-term opioid and amphetamine-type stimulants (ATS) abuse may affect immunological function and impair executive function. We aimed to determine whether biomarkers of inflammation and executive function were associated with substance use in individuals with opioid use disorder (OUD) and ATS use disorder (ATSUD). The interactions between these biomarkers were also explored. METHODS We assessed plasma cytokines [tumor necrosis factor (TNF)-α, C-reactive protein (CRP), interleukin (IL)-8, IL-6, transforming growth factor (TGF)-β1, brain-derived neurotrophic factor (BDNF), and executive function in terms of the Wisconsin Card Sorting Test (WCST) and Continuous Performance Test (CPT) in OUD and ATSUD patients and healthy controls (HC). OUD and ATSUD patients were followed for 12 weeks, and their urine morphine and amphetamine tests, cytokine levels, and executive function were repeatedly measured. RESULTS We enrolled 483 patients and 145 HC. Plasma TNF-α, CRP, IL-8, IL-6, and BDNF levels and most subscale scores on the WCST and CPT significantly differed between OUD and ATSUD patients and HC. Increased TNF-α levels and more perseveration error on the WCST were significantly associated with more urine drug-positive results and less abstinence. Plasma IL-6 and CRP levels were significantly negatively correlated with WCST and CPT performance. CONCLUSION OUD and ATSUD patients had more inflammation and worse executive function than HC. Inflammatory markers and WCST performance were associated with their urinary drug results, and higher inflammation was associated with poor executive function. Studies on regulating the inflammatory process and enhancing executive function in OUD and ATSUD are warranted.
Collapse
Affiliation(s)
- Tzu-Yun Wang
- Correspondence: Tzu-Yun Wang, MD, Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70403, Taiwan ()
| | - Ru-Band Lu
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan,YiNing Hospital, Beijing, China
| | - Sheng-Yu Lee
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yun-Hsuan Chang
- Institute of Gerontology,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan,Institute of Genomics and Bioinformatics, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shiou-Lan Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsung-Yu Tsai
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Huai-Hsuan Tseng
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po See Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kao Chin Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen Kuang Yang
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan,Department of Psychiatry, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
| | - Jau-Shyong Hong
- Neurobiology Laboratory, NIH/NIEHS, Research Triangle Park, North Carolina, USA
| |
Collapse
|
25
|
Bongiovanni AR, Zhao P, Inan S, Wiah S, Shekarabi A, Farkas DJ, Watson MN, Wimmer ME, Ruff MR, Rawls SM. Multi-chemokine receptor antagonist RAP-103 inhibits opioid-derived respiratory depression, reduces opioid reinforcement and physical dependence, and normalizes opioid-induced dysregulation of mesolimbic chemokine receptors in rats. Drug Alcohol Depend 2022; 238:109556. [PMID: 35843139 PMCID: PMC9444981 DOI: 10.1016/j.drugalcdep.2022.109556] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/20/2022] [Accepted: 06/29/2022] [Indexed: 11/03/2022]
Abstract
Chemokine-opioid crosstalk is a physiological crossroads for influencing therapeutic and adverse effects of opioids. Activation of chemokine receptors, especially CCR2, CCR5 and CXCR4, reduces opioid-induced analgesia by desensitizing OPRM1 receptors. Chemokine receptor antagonists (CRAs) enhance opioid analgesia, but knowledge about how CRAs impact adverse opioid effects remains limited. We examined effects of RAP-103, a multi-CRA orally active peptide analog of "DAPTA", on opioid-derived dependence, reinforcement, and respiratory depression in male rats and on changes in chemokine and OPRM1 (µ opioid) receptor levels in mesolimbic substrates during opioid abstinence. In rats exposed to chronic morphine (75 mg pellet x 7 d), daily RAP-103 (1 mg/kg, IP) treatment reduced the severity of naloxone-precipitated withdrawal responses. For self-administration (SA) studies, RAP-103 (1 mg/kg, IP) reduced heroin acquisition (0.1 mg/kg/inf) and reinforcing efficacy (assessed by motivation on a progressive-ratio reinforcement schedule) but did not impact sucrose intake. RAP-103 (1-3 mg/kg, IP) also normalized the deficits in oxygen saturation and enhancement of respiratory rate caused by morphine (5 mg/kg, SC) exposure. Abstinence from chronic morphine elicited brain-region specific changes in chemokine receptor protein levels. CCR2 and CXCR4 were increased in the ventral tegmental area (VTA), whereas CCR2 and CCR5 were reduced in the nucleus accumbens (NAC). Effects of RAP-103 (1 mg/kg, IP) were focused in the NAC, where it normalized morphine-induced deficits in CCR2 and CCR5. These results identify CRAs as potential biphasic function opioid signaling modulators to enhance opioid analgesia and inhibit opioid-derived dependence and respiratory depression.
Collapse
Affiliation(s)
- Angela R Bongiovanni
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Pingwei Zhao
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Sonita Wiah
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Aryan Shekarabi
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Daniel J Farkas
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Mia N Watson
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Mathieu E Wimmer
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | | | - Scott M Rawls
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Cocaine Self-Administration Influences Central Nervous System Immune Responses in Male HIV-1 Transgenic Rats. Cells 2022; 11:cells11152405. [PMID: 35954251 PMCID: PMC9368446 DOI: 10.3390/cells11152405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/25/2022] [Accepted: 07/30/2022] [Indexed: 01/19/2023] Open
Abstract
Cocaine use increases the neurotoxic severity of human immunodeficiency virus-1 (HIV-1) infection and the development of HIV-associated neurocognitive disorders (HAND). Among the studied cellular mechanisms promoting neurotoxicity in HIV-1 and cocaine use, central nervous system (CNS) immunity, such as neuroimmune signaling and reduced antiviral activity, are risk determinants; however, concrete evidence remains elusive. In the present study, we tested the hypothesis that cocaine self-administration by transgenic HIV-1 (HIV-1Tg) rats promotes CNS inflammation. To test this hypothesis, we measured cytokine, chemokine, and growth factor protein levels in the frontal cortex (fCTX) and caudal striatum (cSTR). Our results demonstrated that cocaine self-administration significantly increased fCTX inflammation in HIV-1Tg rats, but not in the cSTR. Accordingly, we postulate that cocaine synergizes with HIV-1 proteins to increase neuroinflammation in a region-selective manner, including the fCTX. Given the fCTX role in cognition, this interaction may contribute to the hyperimmunity and reduced antiviral activity associated with cocaine-mediated enhancement of HAND.
Collapse
|
27
|
dos Santos ACM, dos Santos BRC, dos Santos BB, de Moura EL, Neto ABL, Pereira e Silva AC, de Farias KF, de Medeiros Alves V, Nardi AE, de Souza Figueiredo EVM. IL-10 (-819C/T), TNFA (-30G/A) and ENOS (-786T/C) Polymorphisms Modulating the Outcome Related to Mental Disorders in Crack Addicted Users. Clin Pract Epidemiol Ment Health 2022; 18:e174501792201140. [PMID: 37274848 PMCID: PMC10156023 DOI: 10.2174/17450179-v18-e2201140] [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: 06/27/2021] [Revised: 11/23/2021] [Accepted: 12/20/2021] [Indexed: 06/07/2023]
Abstract
Background Cocaine/crack use affects immune system molecules and development of mental disorders has been identified. Objective To investigate the relationship of polymorphisms in the TNFA (-308G/A), IL-10 (-819C/T) and ENOS (-786T/C) genes with mental disorders in cocaine and crack users. Methods A case-control study was carried out, which included 107 cocaine and crack users and 115 controls who never used healthy cocaine and crack. The SNPs in the TNFA (-308G/A), IL-10 (-819C/T) and ENOS (-786T/C) genes were genotyped by real time PCR. Results As for the individuals included in this study, the average age of 31.4 years (± 8.59). We identified that the G/A genotype to TNFA (-308) (OR = 0.24; p = 0.03) and the A allele (OR = 0.30; p = 0.03) were associated with reduced risk for dysthymic disorder. The T allele of the IL-10 (-819) polymorphism was associated with decreased risk of developing panic disorder (OR = 0.44; p = 0.01), while the C allele was correlated with an increased risk for alcohol dependence (OR = 1.97; p = 0.04), alcohol abuse (OR = 1.81; p = 0.04) and psychotic syndrome (OR = 2.23; p = 0.01). C/C genotype was correlated with increased chances of developing current psychotic syndrome (OR = 4.23; p = 0.01). Conclusion Our results suggest that genetic polymorphisms promote susceptibility or promote protection for clinical phenotypes of psychiatric comorbidities in cocaine and crack users and be considered as good prognostic markers.
Collapse
Affiliation(s)
- Ana Caroline Melo dos Santos
- Program in Health Sciences, Molecular Biology and Gene Expression Laboratory, Federal University of Alagoas, Maceio, Brazil
| | | | - Bruna Brandão dos Santos
- Program in Health Sciences, Molecular Biology and Gene Expression Laboratory, Federal University of Alagoas, Maceio, Brazil
| | - Edilson Leite de Moura
- Program in Health Sciences, Molecular Biology and Gene Expression Laboratory, Federal University of Alagoas, Maceio, Brazil
| | - Abel Barbosa Lira Neto
- Program in Health Sciences, Molecular Biology and Gene Expression Laboratory, Federal University of Alagoas, Maceio, Brazil
| | | | - Karol Fireman de Farias
- Program in Nursing, Molecular Biology and Gene Expression Laboratory, Federal University of Alagoas, Maceio, Brazil
| | | | - Antônio Egídio Nardi
- Institute of Psychiatry, Federal University of Rio de Janeiro, Porto Alegre, Brazil
| | | |
Collapse
|
28
|
Shi S, Chen T, Zhao M. The Crosstalk Between Neurons and Glia in Methamphetamine-Induced Neuroinflammation. Neurochem Res 2022; 47:872-884. [PMID: 34982394 DOI: 10.1007/s11064-021-03513-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/06/2023]
Abstract
Methamphetamine (METH), an illicit psycho-stimulant, is widely known as an addictive drug that may cause neurotoxic effects. Previous researches on METH abuse have mainly focused on neurotransmitters, such as dopamine and glutamate. However, there is growing evidence that neuroinflammation also plays an important role in the etiology and pathophysiology of brain dysfunction induced by METH abuse. This has cast a spotlight on the research of microglia and astrocyte, which are critical mediators of neuroimmune pathology in recent years. In the central nervous system (CNS) immunity, abnormalities of the microglia and astrocytes have been observed in METH abusers from both postmortem and preclinical studies. The bidirectional communication between neurons and glia is essential for the homeostasis and biological function of the CNS while activation of glia induces the release of cytokines and chemokines during pathological conditions, which will affect the neuron-glia interactions and lead to adverse behavioral consequences. However, the underlying mechanisms of interaction between neurons and glia in METH-induced neuroinflammation remain elusive. Notably, discovering and further understanding glial activity and functions, as well as the crosstalk between neurons and glia may help to explain the pathogenesis of METH abuse and behavioral changes in abusers. In this review, we will discuss the current understanding of the crosstalk between neurons and glia in METH-induced neuroinflammation. We also review the existing microglia-astrocyte interaction under METH exposure. We hope the present review will lead the way for more studies on the development of new therapeutic strategies for METH abuse in the near future.
Collapse
Affiliation(s)
- Sai Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wan Ping Road, Shanghai, 200030, China
| | - Tianzhen Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wan Ping Road, Shanghai, 200030, China
| | - Min Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wan Ping Road, Shanghai, 200030, 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.
| |
Collapse
|
29
|
Liu J, Li JX, Wu R. Toll-Like Receptor 4: A Novel Target to Tackle Drug Addiction? Handb Exp Pharmacol 2022; 276:275-290. [PMID: 35434747 PMCID: PMC9829382 DOI: 10.1007/164_2022_586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Drug addiction is a chronic brain disease characterized by compulsive drug-seeking and drug-taking behaviors despite the major negative consequences. Current well-established neuronal underpinnings of drug addiction have promoted the substantial progress in understanding this disorder. However, non-neuronal mechanisms of drug addiction have long been underestimated. Fortunately, increased evidence indicates that neuroimmune system, especially Toll-like receptor 4 (TLR4) signaling, plays an important role in the different stages of drug addiction. Drugs like opioids, psychostimulants, and alcohol activate TLR4 signaling and enhance the proinflammatory response, which is associated with drug reward-related behaviors. While extensive studies have shown that inhibition of TLR4 attenuated drug-related responses, there are conflicting findings implicating that TLR4 signaling may not be essential to drug addiction. In this chapter, preclinical and clinical studies will be discussed to further evaluate whether TLR4-based neuroimmune pharmacotherapy can be used to treat drug addiction. Furthermore, the possible mechanisms underlying the effects of TLR4 inhibition in modulating drug-related behaviors will also be discussed.
Collapse
Affiliation(s)
- Jianfeng Liu
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA,Corresponding authors: Dr. Jun-Xu Li, , Department of Pharmacology and Toxicology, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY 14214. Tel: +1 716 829 2482; Fax: +1 716 829 2801 And Dr. Ruyan Wu, , School of Medicine, Yangzhou University, Yangzhou 225000, China
| | - Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA,School of Medicine, Yangzhou University, Yangzhou, China,Corresponding authors: Dr. Jun-Xu Li, , Department of Pharmacology and Toxicology, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY 14214. Tel: +1 716 829 2482; Fax: +1 716 829 2801 And Dr. Ruyan Wu, , School of Medicine, Yangzhou University, Yangzhou 225000, China
| |
Collapse
|
30
|
Green JM, Sundman MH, Chou YH. Opioid-induced microglia reactivity modulates opioid reward, analgesia, and behavior. Neurosci Biobehav Rev 2022; 135:104544. [DOI: 10.1016/j.neubiorev.2022.104544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/31/2021] [Accepted: 01/16/2022] [Indexed: 12/21/2022]
|
31
|
Leroy C, Saba W. Contribution of TSPO imaging in the understanding of the state of gliosis in substance use disorders. Eur J Nucl Med Mol Imaging 2021; 49:186-200. [PMID: 34041563 DOI: 10.1007/s00259-021-05408-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Recent research in last years in substance use disorders (SUD) synthesized a proinflammatory hypothesis of SUD based on reported pieces of evidence of non-neuronal central immune signalling pathways modulated by drug of abuse and that contribute to their pharmacodynamic actions. Positron emission tomography has been shown to be a precious imaging technique to study in vivo neurochemical processes involved in SUD and to highlight the central immune signalling actions of drugs of abuse. METHODS In this review, we investigate the contribution of the central immune system, with a particular focus on translocator protein 18 kDa (TSPO) imaging, associated with a series of drugs involved in substance use disorders (SUD) specifically alcohol, opioids, tobacco, methamphetamine, cocaine, and cannabis. RESULTS The large majority of preclinical and clinical studies presented in this review converges towards SUD modulation of the neuroimmune responses and TSPO expression and speculated a pivotal positioning in the pathogenesis of SUD. However, some contradictions concerning the same drug or between preclinical and clinical studies make it difficult to draw a clear picture about the significance of glial state in SUD. DISCUSSION Significant disparities in clinical and biological characteristics are present between investigated populations among studies. Heterogeneity in genetic factors and other clinical co-morbidities, difficult to be reproduced in animal models, may affect findings. On the other hand, technical aspects including study designs, radioligand limitations, or PET imaging quantification methods could impact the study results and should be considered to explain discrepancies in outcomes. CONCLUSION The supposed neuroimmune component of SUD provides new therapeutic approaches in the prediction and treatment of SUD pointing to the central immune signalling.
Collapse
Affiliation(s)
- Claire Leroy
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4 place du général Leclerc, 91401, Orsay, France
| | - Wadad Saba
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 4 place du général Leclerc, 91401, Orsay, France.
| |
Collapse
|
32
|
Sivalingam K, Doke M, Khan MA, Samikkannu T. Influence of psychostimulants and opioids on epigenetic modification of class III histone deacetylase (HDAC)-sirtuins in glial cells. Sci Rep 2021; 11:21335. [PMID: 34716387 PMCID: PMC8556237 DOI: 10.1038/s41598-021-00836-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/24/2021] [Indexed: 01/29/2023] Open
Abstract
Substance abuse affects the central nervous system (CNS) and remains a global health problem. Psychostimulants, such as cocaine and methamphetamine (METH), and opioids affect neuronal function and lead to behavioral impairments via epigenetic modification. Epigenetic changes occur via classical pathways, especially the class III histone deacetylase (HDAC)-sirtuin (SIRT) family, that act as cellular sensors to regulate energy homeostasis and coordinate cellular responses to maintain genome integrity. However, SIRT family (1-7)-associated neurodegeneration has not been elucidated in the context of energy metabolism. The present study examined the effects of psychostimulants, such as cocaine and METH, and opioids, such as morphine, on SIRT family (1-7) [class I, II, III and IV] expression and cellular translocation-mediated dysfunction in astrocytes and microglial cells. The "nootropic" drug piracetam played a preventative role against psychostimulant- and opioid-induced SIRT (1-7) expression in astrocytes. These results indicate that cocaine, METH, and morphine affected deacetylation and cellular function, and these changes were prevented by piracetam in astrocytes.
Collapse
Affiliation(s)
- Kalaiselvi Sivalingam
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, 1010 W Avenue B, Kingsville, TX, 78363, USA
| | - Mayur Doke
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, 1010 W Avenue B, Kingsville, TX, 78363, USA
| | - Mansoor A Khan
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, 1010 W Avenue B, Kingsville, TX, 78363, USA
| | - Thangavel Samikkannu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, 1010 W Avenue B, Kingsville, TX, 78363, USA.
| |
Collapse
|
33
|
Montagud-Romero S, Miñarro J, Rodríguez-Arias M. Unravelling the Neuroinflammatory Mechanisms Underlying the Effects of Social Defeat Stress on Use of Drugs of Abuse. Curr Top Behav Neurosci 2021; 54:153-180. [PMID: 34628585 DOI: 10.1007/7854_2021_260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The immune system provides the first line of the organism's defenses, working to maintain homeostasis against external threats and respond also to internal danger signals. There is much evidence to suggest that modifications of inflammatory parameters are related to vulnerability to develop mental illnesses, such as depression, autism, schizophrenia, and substance use disorders. In addition, not only are inflammatory parameters related to these disorders, but stress also induces the activation of the immune system, as recent preclinical research demonstrates. Social stress activates the immune response in the central nervous system through a number of mechanisms; for example, by promoting microglial stimulation, modifying peripheral and brain cytokine levels, and altering the blood brain barrier, which allows monocytes to traffic into the brain. In this chapter, we will first deal with the most important short- and long-term consequences of social defeat (SD) stress on the neuroinflammatory response. SD experiences (brief episodes of social confrontations during adolescence and adulthood) induce functional modifications in the brain, which are accompanied by an increase in proinflammatory markers. Most importantly, inflammatory mechanisms play a significant role in mediating the process of adaptation in the face of adversity (resilience vs susceptibility), allowing us to understand individual differences in stress responses. Secondly, we will address the role of the immune system in the vulnerability and enhanced sensitivity to drugs of abuse after social stress. We will explore in depth the effects seen in the inflammatory system in response to social stress and how they enhance the rewarding effects of drugs such as alcohol or cocaine. To conclude, we will consider pharmacological and environmental interventions that seek to influence the inflammatory response to social stress and diminish increased drug intake, as well as the translational potential and future directions of this exciting new field of research.
Collapse
Affiliation(s)
- S Montagud-Romero
- Department of Psychology and Sociology, University of Zaragoza, Teruel, Spain
| | - J Miñarro
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - M Rodríguez-Arias
- Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Valencia, Spain. .,Red Temática de Investigación Cooperativa en Salud (RETICS-Trastornos Adictivos), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain.
| |
Collapse
|
34
|
Zhang X, Wang H, Wang Y, Li H, Wu S, Gao J, Zhang T, Xie J, Wang X. Nalmefene non-enantioselectively targets myeloid differentiation protein 2 and inhibits toll-like receptor 4 signaling: wet-lab techniques and in silico simulations. Phys Chem Chem Phys 2021; 23:12260-12269. [PMID: 34013938 DOI: 10.1039/d1cp00237f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nalmefene is an opiate derivative having a similar structure to naltrexone. Recent evidence suggests that nalmefene, acting as the innate immune protein toll-like receptor 4 (TLR4) antagonist, effectively reduces the injury of lung ischemia-reperfusion and prevents neuroinflammation. However, the molecular recognition mechanism, especially the enantioselectivity, of nalmefene by the innate immune receptor is not well understood. Herein in vitro assays and in silico simulations were performed to dissect the innate immune recognition of nalmefene at the atomic, molecular, and cellular levels. Biophysical binding experiments and molecular dynamic simulations provide direct evidence that (-)-nalmefene and (+)-nalmefene bind to the hydrophobic cavity of myeloid differentiation protein 2 (MD-2) and behave similarly, which is primarily driven by hydrophobic interactions. The inhibition activity and the calculated binding free energies show that no enantioselectivity was observed during the interaction of nalmefene with MD-2 as well as the inhibition of TLR4 signaling. Interestingly, nalmefene showed ∼6 times better TLR4 antagonisic activity than naltrexone, indicating that the bioisosteric replacement with the methylene group is critical for the molecular recognition of nalmefene by MD-2. In all, this study provides molecular insight into the innate immune recognition of nalmefene, which demonstrates that nalmefene is non-enantioselectively sensed by MD-2.
Collapse
Affiliation(s)
- Xiaozheng Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | - Yibo Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | - Siru Wu
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China. and Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China.
| | - Jingwei Gao
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China. and Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China.
| | - Tianshu Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China. and Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
35
|
Zhu C, Tao H, Rong S, Xiao L, Li X, Jiang S, Guo B, Wang L, Ding J, Gao C, Chang H, Sun T, Wang F. Glucagon-Like Peptide-1 Analog Exendin-4 Ameliorates Cocaine-Mediated Behavior by Inhibiting Toll-Like Receptor 4 Signaling in Mice. Front Pharmacol 2021; 12:694476. [PMID: 34349653 PMCID: PMC8327264 DOI: 10.3389/fphar.2021.694476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Exendin-4 (Ex4), a long-lasting glucagon-like peptide-1 analog, was reported to exert favourable actions on inhibiting cocaine-associated rewarding and reinforcing effects of drug in animal models of addiction. However, the therapeutic potential of different dose of GLP-1 receptor agonist Ex4 in different behavioral paradigms and the underlying pharmacological mechanisms of action are incompletely understood. Herein, we firstly investigated the effects of Ex4 on cocaine-induced condition place preference (CPP) as well as extinction and reinstatement in male C57BL/6J mice. Additionally, we sought to elucidate the underlying pharmacological mechanism of these actions of Ex4. The paradigm of cocaine-induced CPP was established using 20 mg/kg cocaine or saline alternately during conditioning, while the reinstatement paradigm was modeled using 10 mg/kg cocaine on the reinstatement day. Different dose of Ex4 was administrated intraperitoneally either during conditioning or during extinction state or only on the test day. To elucidate the molecular mechanism underlying the potential effects of Ex4 on maladaptive behaviors of cocaine, the TLR4-related inflammation within the hippocampus was observed by immunofluorescence staining, and the expression levels of toll-like receptor 4 (TLR4), tumor necrosis factor (TNF)-α, and interleukin (IL)-1β were detected by Western blotting. As a consequence, systemic administration of different dose of Ex4 was sufficient to inhibit the acquisition and expression of cocaine-induced CPP, facilitate the extinction of cocaine-associated reward and attenuate reinstatement of cocaine-induced behavior. Furthermore, Ex4 treatment diminished expression levels of TLR4, TNF-α, and IL-1β, which were up-regulated by cocaine exposure. Altogether, our results indicated that Ex4 effectively ameliorated cocaine-induced behaviors likely through neurobiological mechanisms partly attributable to the inhibition of TLR4, TNF-α and IL-1β in mice. Consequently, our findings improved our understanding of the efficacy of Ex4 for the amelioration of cocaine-induced behavior and suggested that Ex4 may be applied as a drug candidate for cocaine addiction.
Collapse
Affiliation(s)
- Changliang Zhu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Hong Tao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shikuo Rong
- Department of General Surgery, Chengdu Second Hospital, Chendu, China
| | - Lifei Xiao
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Xinxiao Li
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Shucai Jiang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Baorui Guo
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Lei Wang
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Jiangwei Ding
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Caibing Gao
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Haigang Chang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China.,Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Feng Wang
- Ningxia Key Laboratory of Cerebro Cranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China.,Department of Neurosurgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
36
|
Mahajan SD, Homish GG, Quisenberry A. Multifactorial Etiology of Adolescent Nicotine Addiction: A Review of the Neurobiology of Nicotine Addiction and Its Implications for Smoking Cessation Pharmacotherapy. Front Public Health 2021; 9:664748. [PMID: 34291026 PMCID: PMC8287334 DOI: 10.3389/fpubh.2021.664748] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Nicotine is the primary pharmacologic component of tobacco, and its highly addictive nature is responsible for its widespread use and significant withdrawal effects that result in challenges to smoking cessation therapeutics. Nicotine addiction often begins in adolescence and this is at least partially attributed to the fact that adolescent brain is most susceptible to the neuro-inflammatory effects of nicotine. There is increasing evidence for the involvement of microglial cells, which are the brain's primary homeostatic sensor, in drug dependence and its associated behavioral manifestations particularly in the adolescent brain. A hallmark of neuro-inflammation is microglial activation and activation of microglia by nicotine during adolescent development, which may result in long-term addiction to nicotine. This non-systematic review examines multifactorial etiology of adolescent nicotine addiction, neurobiology of nicotine addiction and the potential mechanisms that underlie the effects of nicotine on inflammatory signaling in the microglia, understanding how nicotine affects the adolescent brain. We speculate, that modulating homeostatic balance in microglia, could have promising therapeutic potential in withdrawal, tolerance, and abstinence-related neural adaptations in nicotine addiction, in the adolescent brain. Further, we discuss nicotine addiction in the context of the sensitization-homeostasis model which provides a theoretical framework for addressing the potential role of microglial homeostasis in neural adaptations underlying nicotine abuse.
Collapse
Affiliation(s)
- Supriya D. Mahajan
- Department of Community Health and Health Behavior, School of Public Health, University at Buffalo, Buffalo, NY, United States
| | - Gregory G. Homish
- Department of Community Health and Health Behavior, School of Public Health, University at Buffalo, Buffalo, NY, United States
| | - Amanda Quisenberry
- Department of Health Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
37
|
Galván ST, Flores-López M, Romero-Sanchiz P, Requena-Ocaña N, Porras-Perales O, Nogueira-Arjona R, Mayoral F, Araos P, Serrano A, Muga R, Pavón FJ, García-Marchena N, de Fonseca FR. Plasma concentrations of granulocyte colony-stimulating factor (G-CSF) in patients with substance use disorders and comorbid major depressive disorder. Sci Rep 2021; 11:13629. [PMID: 34211033 PMCID: PMC8249412 DOI: 10.1038/s41598-021-93075-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) has raised much interest because of its role in cocaine addiction in preclinical models. We explored the plasma concentrations of G-CSF in patients diagnosed with substance use disorder (SUD) and highly comorbid psychiatric disorders. In particular, we investigated the association between G-CSF concentrations and comorbid major depressive disorder (MDD) in patients with cocaine and alcohol use disorders (CUD and AUD, respectively). Additionally, patients with MDD but not SUD were included in the study. Three hundred and eleven participants were enrolled in this exploratory study: 136 control subjects, 125 patients with SUD (SUD group) from outpatient treatment programs for cocaine (N = 60, cocaine subgroup) and alcohol (N = 65, alcohol subgroup), and 50 patients with MDD but not SUD (MDD group) from primary-care settings. Participants were assessed based on DSM-IV-TR criteria, and a blood sample was collected to examine the plasma concentrations of G-CSF. G-CSF concentrations were negatively correlated with age in the entire sample (r = - 0.233, p < 0.001) but not in the patients with MDD. G-CSF concentrations were lower in patients with SUD than in controls (p < 0.05), specifically in the cocaine subgroup (p < 0.05). Patients with SUD and comorbid MDD had lower G-CSF concentrations than patients with SUD but not comorbid MDD or controls (p < 0.05). In contrast, patients with MDD but not SUD showed no differences compared with their controls. The negative association between G-CSF concentrations and age in the sample was not observed in patients with MDD. G-CSF concentrations were decreased in patients with SUD and comorbid MDD but not in patients with MDD. Therefore, G-CSF may be useful to improve the stratification of patients with dual diagnosis seeking treatment. Further investigation is needed to explore the impact of sex and type of drug on the expression of G-CSF.
Collapse
Affiliation(s)
- Sandra Torres Galván
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - María Flores-López
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| | - Pablo Romero-Sanchiz
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Department of Psychology, University of Roehampton, London, UK
| | - Nerea Requena-Ocaña
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Oscar Porras-Perales
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Facultad de Psicología, Universidad de Málaga, Málaga, Spain
- Unidad de Gestión Clínica del Corazón, Hospital Universitario Virgen de La Victoria, Málaga, Spain
| | - Raquel Nogueira-Arjona
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Department of Psychology, University of Roehampton, London, UK
| | - Fermín Mayoral
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Pedro Araos
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
- Facultad de Psicología, Universidad de Málaga, Málaga, Spain
| | - Antonia Serrano
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Roberto Muga
- Unidad de Adicciones- Servicio de Medicina Interna. Institut D'Investigació en Ciències de La Salut Germans Trias I Pujol (IGTP), Campus Can Ruti, Carrer del Canyet s/n, 08916, Badalona, Spain
- Departamento de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francisco Javier Pavón
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain.
- Unidad de Gestión Clínica del Corazón, Hospital Universitario Virgen de La Victoria, Málaga, Spain.
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| | - Nuria García-Marchena
- Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.
- Unidad de Adicciones- Servicio de Medicina Interna. Institut D'Investigació en Ciències de La Salut Germans Trias I Pujol (IGTP), Campus Can Ruti, Carrer del Canyet s/n, 08916, Badalona, Spain.
| | - Fernando Rodríguez de Fonseca
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Avda. Carlos Haya 82, sótano, 29010, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain.
- Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
38
|
Lucerne KE, Osman A, Meckel KR, Kiraly DD. Contributions of neuroimmune and gut-brain signaling to vulnerability of developing substance use disorders. Neuropharmacology 2021; 192:108598. [PMID: 33965398 PMCID: PMC8220934 DOI: 10.1016/j.neuropharm.2021.108598] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/19/2021] [Accepted: 05/03/2021] [Indexed: 02/06/2023]
Abstract
Epidemiology and clinical research indicate that only a subset of people who are exposed to drugs of abuse will go on to develop a substance use disorder. Numerous factors impact individual susceptibility to developing a substance use disorder, including intrinsic biological factors, environmental factors, and interpersonal/social factors. Given the extensive morbidity and mortality that is wrought as a consequence of substance use disorders, a substantial body of research has focused on understanding the risk factors that mediate the shift from initial drug use to pathological drug use. Understanding these risk factors provides a clear path for the development of risk mitigation strategies to help reduce the burden of substance use disorders in the population. Here we will review the rapidly growing body of literature that examines the importance of interactions between the peripheral immune system, the gut microbiome, and the central nervous system (CNS) in mediating the transition to pathological drug use. While these systems had long been viewed as distinct, there is growing evidence that there is bidirectional communication between both the immune system and the gut microbiome that drive changes in neural and behavioral plasticity relevant to substance use disorders. Further, both of these systems are highly sensitive to environmental perturbations and are implicated in numerous neuropsychiatric conditions. While the field of study examining these interactions in substance use disorders is in its relative infancy, clarifying the relationship between gut-immune-brain signaling and substance use disorders has potential to improve our understanding of individual propensity to developing addiction and yield important insight into potential treatment options.
Collapse
Affiliation(s)
- Kelsey E Lucerne
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aya Osman
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine R Meckel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
39
|
O'Sullivan SJ, Schwaber JS. Similarities in alcohol and opioid withdrawal syndromes suggest common negative reinforcement mechanisms involving the interoceptive antireward pathway. Neurosci Biobehav Rev 2021; 125:355-364. [PMID: 33647322 PMCID: PMC8555739 DOI: 10.1016/j.neubiorev.2021.02.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 12/19/2022]
Abstract
Alcohol and opioids are two major contributors to so-called deaths of despair. Though the effects of these substances on mammalian systems are distinct, commonalities in their withdrawal syndromes suggest a shared pathophysiology. For example, both are characterized by marked autonomic dysregulation and are treated with alpha-2 agonists. Moreover, alcohol and opioids rapidly induce dependence motivated by withdrawal avoidance. Resemblances observed in withdrawal syndromes and abuse behavior may indicate common addiction mechanisms. We argue that neurovisceral feedback influences autonomic and emotional circuits generating antireward similarly for both substances. Amygdala is central to this hypothesis as it is principally responsible for negative emotion, prominent in addiction and motivated behavior, and processes autonomic inputs while generating autonomic outputs. The solitary nucleus (NTS) has strong bidirectional connections to the amygdala and receives interoceptive inputs communicating visceral states via vagal afferents. These visceral-emotional hubs are strongly influenced by the periphery including gut microbiota. We propose that gut dysbiosis contributes to alcohol and opioid withdrawal syndromes by contributing to peripheral and neuroinflammation that stimulates these antireward pathways and motivates substance dependence.
Collapse
Affiliation(s)
- Sean J O'Sullivan
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - James S Schwaber
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Chat IKY, Nusslock R, Moriarity DP, Bart CP, Mac Giollabhui N, Damme KSF, Carroll AL, Miller GE, Alloy LB. Goal-striving tendencies moderate the relationship between reward-related brain function and peripheral inflammation. Brain Behav Immun 2021; 94:60-70. [PMID: 33705866 PMCID: PMC8075112 DOI: 10.1016/j.bbi.2021.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 01/19/2023] Open
Abstract
Inflammation is associated with both lower and higher activity in brain regions that process rewarding stimuli. How can both low and high sensitivity to rewards be associated with higher inflammation? We propose that one potential mechanism underlying these apparently conflicting findings pertains to how people pursue goals in their environment. This prediction is based on evidence that both an inability to disengage from unattainable goals and low interest in and pursuit of important life goals are associated with poor health outcomes, including inflammation. Accordingly, this study examined the relationship between reward-related brain function and peripheral inflammation among individuals with different levels of ambitious goal-striving tendencies. Eighty-three participants completed an ambitious goal-striving tendency measure, an fMRI Monetary Incentive Delay task assessing orbitofrontal cortex (OFC) and nucleus accumbens (NAc) activation during reward anticipation and outcome, and a venous blood draw to assess the inflammatory biomarkers interleukin (IL)-6, IL-8, tumor necrosis factor-alpha, and C-reactive protein, from which we computed an inflammation composite score. We observed a reward anticipation by goal-striving interaction on inflammation, such that high OFC and NAc activation to reward anticipation (but not outcome) were associated with more inflammation, among high goal-striving individuals. By contrast, low NAc activation during reward anticipation (but not outcome) was associated with more inflammation, among low goal-striving individuals. The current study provides further evidence that both blunted and elevated reward function can be associated with inflammation. It also highlights the role that goal-striving tendencies may play in moderating the relationship between neural reward anticipation and inflammation.
Collapse
Affiliation(s)
- Iris Ka-Yi Chat
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | - Robin Nusslock
- Department of Psychology, Northwestern University, Evanston, IL, USA; Institute for Policy Research, Northwestern University, Evanston, IL, USA
| | | | - Corinne P Bart
- Department of Psychology, Temple University, Philadelphia, PA, USA
| | | | - Katherine S F Damme
- Department of Psychology, Northwestern University, Evanston, IL, USA; Institute for Innovations in Developmental Sciences (DevSci), Northwestern University, Evanston and Chicago, IL, USA
| | - Ann L Carroll
- Department of Psychology, Northwestern University, Evanston, IL, USA
| | - Gregory E Miller
- Department of Psychology, Northwestern University, Evanston, IL, USA; Institute for Policy Research, Northwestern University, Evanston, IL, USA
| | - Lauren B Alloy
- Department of Psychology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
41
|
Villavicencio-Tejo F, Flores-Bastías O, Marambio-Ruiz L, Pérez-Reytor D, Karahanian E. Fenofibrate (a PPAR-α Agonist) Administered During Ethanol Withdrawal Reverts Ethanol-Induced Astrogliosis and Restores the Levels of Glutamate Transporter in Ethanol-Administered Adolescent Rats. Front Pharmacol 2021; 12:653175. [PMID: 33959021 PMCID: PMC8093785 DOI: 10.3389/fphar.2021.653175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/31/2021] [Indexed: 01/11/2023] Open
Abstract
High-ethanol intake induces a neuroinflammatory response, which has been proposed as responsible for the maintenance of chronic ethanol consumption. Neuroinflammation decreases glutamate transporter (GLT-1) expression, increasing levels of glutamate that trigger dopamine release at the corticolimbic reward areas, driving long-term drinking behavior. The activation of peroxisome proliferator-activated receptor alpha (PPARα) by fibrates inhibits neuroinflammation, in models other than ethanol consumption. However, the effect of fibrates on ethanol-induced neuroinflammation has not yet been studied. We previously reported that the administration of fenofibrate to ethanol-drinking rats decreased ethanol consumption. Here, we studied whether fenofibrate effects are related to a decrease in ethanol-induced neuroinflammation and to the normalization of the levels of GLT-1. Rats were administered ethanol on alternate days for 4 weeks (2 g/kg/day). After ethanol withdrawal, fenofibrate was administered for 14 days (50 mg/kg/day) and the levels of glial fibrillary acidic protein (GFAP), phosphorylated NF-κB-inhibitory protein (pIκBα) and GLT-1, were quantified in the prefrontal cortex, hippocampus, and hypothalamus. Ethanol treatment increased the levels of GFAP in the hippocampus and hypothalamus, indicating a clear astrocytic activation. Similarly, ethanol increased the levels of pIκBα in the three areas. The administration of fenofibrate decreased the expression of GFAP and pIκBα in the three areas. These results indicate that fenofibrate reverts both astrogliosis and NF-κB activation. Finally, ethanol decreased GLT-1 expression in the prefrontal cortex and hippocampus. Fenofibrate normalized the levels of GLT-1 in both areas, suggesting that its effect in reducing ethanol consumption could be due to the normalization of glutamatergic tone.
Collapse
Affiliation(s)
| | - Osvaldo Flores-Bastías
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Lucas Marambio-Ruiz
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Diliana Pérez-Reytor
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Eduardo Karahanian
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
42
|
Nwachukwu KN, Evans WA, Sides TR, Trevisani CP, Davis A, Marshall SA. Chemogenetic manipulation of astrocytic signaling in the basolateral amygdala reduces binge-like alcohol consumption in male mice. J Neurosci Res 2021; 99:1957-1972. [PMID: 33844860 DOI: 10.1002/jnr.24841] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/21/2021] [Indexed: 12/18/2022]
Abstract
Binge drinking is a common occurrence in the United States, but a high concentration of alcohol in the blood has been shown to have reinforcing and reciprocal effects on the neuroimmune system in both dependent and non-dependent scenarios. The first part of this study examined alcohol's effects on the astrocytic response in the central amygdala and basolateral amygdala (BLA) in a non-dependent model. C57BL/6J mice were given access to either ethanol, water, or sucrose during a "drinking in the dark" paradigm, and astrocyte number and astrogliosis were measured using immunohistochemistry. Results indicate that non-dependent consumption increased glial fibrillary acidic protein (GFAP) density but not the number of GFAP+ cells, suggesting that non-dependent ethanol is sufficient to elicit astrocyte activation. The second part of this study examined how astrocytes impacted behaviors and the neurochemistry related to alcohol using the chemogenetic tool, DREADDs (designer receptors exclusively activated by designer drugs). Transgenic GFAP-hM3Dq mice were administered clozapine N-oxide both peripherally, affecting the entire central nervous system (CNS), or directly into the BLA. In both instances, GFAP-Gq-signaling activation significantly reduced ethanol consumption and correlating blood ethanol concentrations. However, GFAP-Gq-DREADD activation throughout the CNS had more broad effects resulting in decreased locomotor activity and sucrose consumption. More targeted GFAP-Gq-signaling activation in the BLA only impacted ethanol consumption. Finally, a glutamate assay revealed that after GFAP-Gq-signaling activation glutamate concentrations in the amygdala were partially normalized to control levels. Altogether, these studies support the theory that astrocytes represent a viable target for alcohol use disorder therapies.
Collapse
Affiliation(s)
- Kala N Nwachukwu
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - William A Evans
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - Tori R Sides
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - Christopher P Trevisani
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - Ambryia Davis
- Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA
| | - S Alex Marshall
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, USA.,Department of Basic Pharmaceutical Sciences, Fred P. Wilson School of Pharmacy, High Point University, High Point, NC, USA.,Department of Psychology & Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
43
|
Chuang HG, Aziz NHA, Wong JH, Mustapha M, Abdullah JM, Idris Z, Abdullah Z, Alrafiah A, Muthuraju S. Role of toll-like receptor 4 antagonist Lipopolysaccharide-Rhodobacter sphaeroides on acute stress-induced voluntary ethanol preference and drinking behaviour: In vivo Swiss Albino mouse model. Eur Neuropsychopharmacol 2021; 45:59-72. [PMID: 32014377 DOI: 10.1016/j.euroneuro.2019.12.121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 06/06/2019] [Accepted: 12/24/2019] [Indexed: 12/14/2022]
Abstract
The present study focused on investigating the effect of toll-like receptor 4 (TLR4) antagonist Lipopolysaccharide-Rhodobacter sphaeroides(LPS-RS) on acute, stress-induced voluntary ethanol preference and drinking behaviour, neuronal components activation, and gene expression associated with stress and addictive behaviour. This study involved the exposure of restraint stress and social isolation using Swiss Albino mice. Two-bottle choice ethanol preference analysis was used in the evaluation of voluntary ethanol seeking and drinking behaviour. Several behavioural assessments were carried out to assess fear and anxiety-like behaviour, neuromuscular ability, motor coordination and locomotion. Morphological and immunoreactivity analysis and gene expression analysis were done after the completion of behavioural assessments. TLR4 antagonist LPS-RS treated stressed-mice showed a significant decrease in ethanol drinking compared with stressed mice. Behavioural results showed that stress exposure induced fear and anxiety-like behaviour; however; no significant deficit was found on motor coordination, neuromuscular ability, locomotion and exploratory behaviour among groups. Morphological analysis showed no significant change in the prefrontal cortex and hippocampus among all groups, while immunoreactivity analysis showed higher expression of c-Fos in prefrontal cortex and hippocampus, higher TLR4 expression in the prefrontal cortex and glial fibrillary acidic protein (GFAP) in hippocampus among stressed-animals. Stressed-mice also showed significant increase in TLR4, Nuclear Factor-Kappa B (NF-kB), inducible nitric oxide synthase (iNOS), dopamine receptor D2 (DRD2), cyclic adenosine monophosphate (cAMP) response element binding protein-1 (CREB-1) and opioid receptor MU-1 (OPRM-1) genes expression compared with control and LPS-RS treated stressed-mice. As a conclusion, the antagonism of TLR4 could provide therapeutic value in the treatment of stress-induced addiction.
Collapse
Affiliation(s)
- Huei Gau Chuang
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - NurNaznee Hirni Abd Aziz
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Jia Hui Wong
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Muzaimi Mustapha
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Jafri Malin Abdullah
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Zamzuri Idris
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Zuraidah Abdullah
- Biomedicine Program, School of Health Science, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Aziza Alrafiah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Sangu Muthuraju
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston 77240, TX, USA.
| |
Collapse
|
44
|
Drug addiction co-morbidity with alcohol: Neurobiological insights. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 157:409-472. [PMID: 33648675 DOI: 10.1016/bs.irn.2020.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Addiction is a chronic disorder that consists of a three-stage cycle of binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation. These stages involve, respectively, neuroadaptations in brain circuits involved in incentive salience and habit formation, stress surfeit and reward deficit, and executive function. Much research on addiction focuses on the neurobiology underlying single drug use. However, alcohol use disorder (AUD) can be co-morbid with substance use disorder (SUD), called dual dependence. The limited epidemiological data on dual dependence indicates that there is a large population of individuals suffering from addiction who are dependent on more than one drug and/or alcohol, yet dual dependence remains understudied in addiction research. Here, we review neurobiological data on neurotransmitter and neuropeptide systems that are known to contribute to addiction pathology and how the involvement of these systems is consistent or divergent across drug classes. In particular, we highlight the dopamine, opioid, corticotropin-releasing factor, norepinephrine, hypocretin/orexin, glucocorticoid, neuroimmune signaling, endocannabinoid, glutamate, and GABA systems. We also discuss the limited research on these systems in dual dependence. Collectively, these studies demonstrate that the use of multiple drugs can produce neuroadaptations that are distinct from single drug use. Further investigation into the neurobiology of dual dependence is necessary to develop effective treatments for addiction to multiple drugs.
Collapse
|
45
|
García-Cabrerizo R, Carbia C, O Riordan KJ, Schellekens H, Cryan JF. Microbiota-gut-brain axis as a regulator of reward processes. J Neurochem 2021; 157:1495-1524. [PMID: 33368280 DOI: 10.1111/jnc.15284] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/08/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
Our gut harbours trillions of microorganisms essential for the maintenance of homeostasis and host physiology in health and disease. In the last decade, there has been a growing interest in understanding the bidirectional pathway of communication between our microbiota and the central nervous system. With regard to reward processes there is accumulating evidence from both animal and human studies that this axis may be a key factor in gating reward valence. Focusing on the mesocorticolimbic pathway, we will discuss how the intestinal microbiota is involved in regulating brain reward functions, both in natural (i.e. eating, social or sexual behaviours) and non-natural reinforcers (drug addiction behaviours including those relevant to alcohol, psychostimulants, opioids and cannabinoids). We will integrate preclinical and clinical evidence suggesting that the microbiota-gut-brain axis could be implicated in the development of disorders associated with alterations in the reward system and how it may be targeted as a promising therapeutic strategy. Cover Image for this issue: https://doi.org/10.1111/jnc.15065.
Collapse
Affiliation(s)
| | - Carina Carbia
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Harriet Schellekens
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
46
|
Rush CR, Stoops WW, Lile JA, Alcorn JL, Bolin BL, Reynolds AR, Hays LR, Rayapati AO. Topiramate-phentermine combinations reduce cocaine self-administration in humans. Drug Alcohol Depend 2021; 218:108413. [PMID: 33290875 DOI: 10.1016/j.drugalcdep.2020.108413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
RATIONALE Cocaine use disorder is an unrelenting public health concern. Despite nearly four decades of research, an FDA approved medication is not yet available. OBJECTIVES The objective of this human laboratory study was to demonstrate the initial efficacy, safety and tolerability of topiramate-phentermine combinations for cocaine use disorder. METHODS Thirty-one (31) participants with cocaine use disorder completed this mixed-model inpatient laboratory study. Participants were maintained on topiramate (0 [N = 11], 50 [N = 9] or 100 [N = 11] mg/day). Each topiramate group was concurrently maintained on phentermine (0, 15, 30 mg). Drug self-administration, subjective responses and cardiovascular effects following acute doses of intranasal cocaine (0, 40, 80 mg) were determined during separate experimental sessions after at least seven (7) days of maintenance on each condition. RESULTS The three groups of participants were well matched demographically and generally did not differ significantly in their responses to a range of doses of intranasal cocaine (0, 10, 20, 40, 80 mg) during a medical safety session. Maintenance on topiramate and phentermine alone significantly decreased cocaine self-administration although these effects were modest in magnitude. Combining topiramate and phentermine robustly decreased cocaine self-administration. Topiramate and phentermine were well tolerated alone and combined, as well as in conjunction with cocaine. CONCLUSIONS The results of the present study support advancing topiramate-phentermine combinations as a putative pharmacotherapeutic for cocaine use disorder.
Collapse
Affiliation(s)
- Craig R Rush
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY, 40536-0086, USA.
| | - William W Stoops
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY, 40536-0086, USA; Center on Drug and Alcohol Research, University of Kentucky College of Medicine, 845 Angliana Ave, Lexington, KY, 40508, USA
| | - Joshua A Lile
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY, 40536-0086, USA; Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY, 40509-1810, USA; Department of Psychology, University of Kentucky College of Arts and Sciences, 171 Funkhouser Drive, Lexington, KY, 40506-0044, USA
| | - Joseph L Alcorn
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY, 40536-0086, USA
| | - B Levi Bolin
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY, 40536-0086, USA
| | - Anna R Reynolds
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY, 40536-0086, USA
| | - Lon R Hays
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY, 40509-1810, USA
| | - Abner O Rayapati
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY, 40509-1810, USA
| |
Collapse
|
47
|
Fitting S, McRae M, Hauser KF. Opioid and neuroHIV Comorbidity - Current and Future Perspectives. J Neuroimmune Pharmacol 2020; 15:584-627. [PMID: 32876803 PMCID: PMC7463108 DOI: 10.1007/s11481-020-09941-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022]
Abstract
With the current national opioid crisis, it is critical to examine the mechanisms underlying pathophysiologic interactions between human immunodeficiency virus (HIV) and opioids in the central nervous system (CNS). Recent advances in experimental models, methodology, and our understanding of disease processes at the molecular and cellular levels reveal opioid-HIV interactions with increasing clarity. However, despite the substantial new insight, the unique impact of opioids on the severity, progression, and prognosis of neuroHIV and HIV-associated neurocognitive disorders (HAND) are not fully understood. In this review, we explore, in detail, what is currently known about mechanisms underlying opioid interactions with HIV, with emphasis on individual HIV-1-expressed gene products at the molecular, cellular and systems levels. Furthermore, we review preclinical and clinical studies with a focus on key considerations when addressing questions of whether opioid-HIV interactive pathogenesis results in unique structural or functional deficits not seen with either disease alone. These considerations include, understanding the combined consequences of HIV-1 genetic variants, host variants, and μ-opioid receptor (MOR) and HIV chemokine co-receptor interactions on the comorbidity. Lastly, we present topics that need to be considered in the future to better understand the unique contributions of opioids to the pathophysiology of neuroHIV. Graphical Abstract Blood-brain barrier and the neurovascular unit. With HIV and opiate co-exposure (represented below the dotted line), there is breakdown of tight junction proteins and increased leakage of paracellular compounds into the brain. Despite this, opiate exposure selectively increases the expression of some efflux transporters, thereby restricting brain penetration of specific drugs.
Collapse
Affiliation(s)
- Sylvia Fitting
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-3270, USA
| | - MaryPeace McRae
- Department of Pharmacotherapy and Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 1217 East Marshall Street, Richmond, VA, 23298-0613, USA.
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298-0709, USA.
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, 203 East Cary Street, Richmond, VA, 23298-0059, USA.
| |
Collapse
|
48
|
Wei ZX, Chen L, Zhang JJ, Cheng Y. Aberrations in peripheral inflammatory cytokine levels in substance use disorders: a meta-analysis of 74 studies. Addiction 2020; 115:2257-2267. [PMID: 32533781 DOI: 10.1111/add.15160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/02/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023]
Abstract
AIMS To characterize the peripheral inflammatory cytokine profile in people with substance use disorders (SUDs). DESIGN Systematic review and meta-analysis. SETTING Clinical studies that evaluated peripheral blood inflammatory cytokine levels in patients with SUDs and healthy controls PARTICIPANTS: SUD patients and healthy controls. MEASUREMENTS PubMed and Web of Science were systematically searched for relevant studies. Two investigators independently selected studies and extracted data. A total of 77 articles were included in the meta-analysis, containing 5649 patients with SUDs and 4643 healthy controls. Data were pooled using a random-effects model by the Comprehensive Meta-Analysis version 2 software. FINDINGS Concentrations of interleukin (IL)-6) in 32 studies, tumor necrosis factor (TNF)-α in 28 studies, IL-10 in 20 studies, IL-8 in 17 studies, C-reactive protein in 14 studies, IL-4 in 10 studies, IL-12 in seven studies, monocyte chemoattractant protein (MCP)-1 in 6 studies, TNF-receptor 2 (TNF-R2) in four studies and granulocyte-macrophage colony-stimulating factor (GM-CSF) in three studies were significantly higher in patients with SUDs compared with healthy controls, while concentrations of leptin in 14 studies were significantly lower in patients with SUDs compared with healthy controls. The findings were inconclusive for the associations between interferon-γ, IL-1β, IL-2, IL-1 receptor antagonist (IL-1RA), transforming growth factor (TGF)-β1, G-CSF, C-C motif chemokine 11, TGF-α and SUDs. CONCLUSIONS People with substance use disorders (SUDs) appear to have higher peripheral concentrations of IL-4, IL-6, IL-8, IL-10, IL-12, TNF-α, C-reactive protein, MCP-1, TNF-R2 and GM-CSF and lower peripheral concentrations of leptin than people without SUDs. This strengthens the view that SUD is accompanied by an inflammatory response.
Collapse
Affiliation(s)
- Ze-Xu Wei
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Lei Chen
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Jian-Jun Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yong Cheng
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| |
Collapse
|
49
|
Wu R, Li JX. Toll-Like Receptor 4 Signaling and Drug Addiction. Front Pharmacol 2020; 11:603445. [PMID: 33424612 PMCID: PMC7793839 DOI: 10.3389/fphar.2020.603445] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022] Open
Abstract
The emphasis of neuronal alterations and adaptations have long been the main focus of the studies of the mechanistic underpinnings of drug addiction. Recent studies have begun to appreciate the role of innate immune system, especially toll-like receptor 4 (TLR4) signaling in drug reward-associated behaviors and physiology. Drugs like opioids, alcohol and psychostimulants activate TLR4 signaling and subsequently induce proinflammatory responses, which in turn contributes to the development of drug addiction. Inhibition of TLR4 or its downstream effectors attenuated the reinforcing effects of opioids, alcohol and psychostimulants, and this effect is also involved in the withdrawal and relapse-like behaviors of different drug classes. However, conflicting results also argue that TLR4-related immune response may play a minimal part in drug addiction. This review discussed the preclinical evidence that whether TLR4 signaling is involved in multiple drug classes action and the possible mechanisms underlying this effect. Moreover, clinical studies which examined the potential efficacy of immune-base pharmacotherapies in treating drug addiction are also discussed.
Collapse
Affiliation(s)
- Ruyan Wu
- School of Medicine, Yangzhou University, Yangzhou, China.,Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
50
|
Figueroa-Hall LK, Paulus MP, Savitz J. Toll-Like Receptor Signaling in Depression. Psychoneuroendocrinology 2020; 121:104843. [PMID: 32911436 PMCID: PMC7883590 DOI: 10.1016/j.psyneuen.2020.104843] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/09/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022]
Abstract
Depression is one of the most prevalent, disabling, and costly mental illnesses currently affecting over 300 million people worldwide. A subset of depressed patients display inflammation as indicated by increased levels of proinflammatory mediators in the blood and cerebrospinal fluid. Longitudinal and experimental studies suggest that this inflammatory profile may causally contribute to the initiation, maintenance, or recurrence of depressive episodes in the context of major depressive disorder (MDD). While the mechanistic pathways that mediate these depressogenic effects have not yet been fully elucidated, toll-like receptor (TLR) signaling is one potential common inflammatory pathway. In this review, we focus on the role that inflammation plays in depression, TLR signaling and its plasticity as a candidate pathway, its regulation by micro ribonucleic acids (miRNAs), and their potential as diagnostic biomarkers for identification of inflammatory subtypes of depression. Pre-clinical and clinical studies have demonstrated that TLR expression and TLR signaling regulators are associated with MDD. Further, TLR expression and signaling is in-turn, regulated in part by miRNAs and some TLR-responsive miRNAs indirectly modulate pathways that are implicated in MDD pathophysiology. These data suggest an intersection between TLR signaling regulation and MDD-linked pathways. While these studies suggest that miRNAs play a role in the pathophysiology of MDD via their regulatory effects on TLR pathways, the utility of miRNAs as biomarkers and potential treatment targets remains to be determined. Developing new and innovative techniques or adapting established immunological approaches to mental health, should be at the forefront in moving the field forward, especially in terms of categorization of inflammatory subtypes in MDD.
Collapse
Affiliation(s)
| | - Martin P Paulus
- Laureate Institute for Brain Research, 6655 S. Yale Ave, Tulsa, OK, 74136, United States; Oxley College of Health Sciences, 1215 S. Boulder Ave W., The University of Tulsa, Tulsa, OK, 74199, United States.
| | - Jonathan Savitz
- Laureate Institute for Brain Research, 6655 S. Yale Ave, Tulsa, OK, 74136, United States; Oxley College of Health Sciences, 1215 S. Boulder Ave W., The University of Tulsa, Tulsa, OK, 74199, United States.
| |
Collapse
|