1
|
Juliano VAL, Albernaz-Mariano KA, Covre LHH, Jucá PM, Pereira RM, Shigeo-de-Almeida A, Sampaio LL, Duque EDA, Munhoz CD. Neurobiological intersections of stress and substance use disorders. Front Neurosci 2025; 19:1548372. [PMID: 40376607 PMCID: PMC12078238 DOI: 10.3389/fnins.2025.1548372] [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: 12/19/2024] [Accepted: 04/07/2025] [Indexed: 05/18/2025] Open
Abstract
Substance use has been intertwined with human history for millennia. Throughout the ages, people have consumed various substances for medicinal, spiritual, and recreational reasons, although occasional use differs significantly from substance use disorders (SUDs). Exposure to lifetime stressors constitutes a significant risk factor for both psychiatric disorders and SUD development and relapse. Indeed, hypothalamic-pituitary-adrenal (HPA) axis modulation, alterations in neuroanatomical and neurotransmitter systems, as well as neuroinflammation are common features of stress-related mood disorders and SUDs. In this mini-review, we will explore how stress exposure influences the SUDs' neurobiological basis on different scales-from large neural circuitries to specific molecular mechanisms-and discuss novel targets for potential treatments.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Carolina Demarchi Munhoz
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
2
|
Liu Y, Wang XQ, Zhang P, Haghparast A, He WB, Zhang JJ. Research progress of DNA methylation on the regulation of substance use disorders and the mechanisms. Front Cell Neurosci 2025; 19:1566001. [PMID: 40230379 PMCID: PMC11994631 DOI: 10.3389/fncel.2025.1566001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Drug abuse can damage the central nervous system and lead to substance use disorder (SUD). SUD is influenced by both genetic and environmental factors. Genes determine an individual's susceptibility to drug, while the dysregulation of epigenome drives the abnormal transcription processes, promoting the development of SUD. One of the most widely studied epigenetic mechanisms is DNA methylation, which can be inherited stably. In ontogeny, DNA methylation pattern is dynamic. DNA dysmethylation is prevalent in drug-related psychiatric disorders, resulting in local hypermethylation and transcriptional silencing of related genes. In this review, we summarize the role and regulatory mechanisms of DNA methylation in cocaine, opioids, and methamphetamine in terms of drug exposure, addiction memory, withdrawal relapse, intergenerational inheritance, and focus on cell-specific aspects of the studies with a view to suggesting possible therapeutic regimens for targeting methylation in both human and animal research.
Collapse
Affiliation(s)
- Ya Liu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Xiao-Qian Wang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Peng Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Abbas Haghparast
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Wen-Bin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Jian-Jun Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
3
|
Sakai Y, Hattori J, Morikawa Y, Matsumura T, Jimbo S, Suenami K, Takayama T, Nagai A, Michiue T, Ikari A, Matsunaga T. α-Pyrrolidinooctanophenone facilitates activation of human microglial cells via ROS/STAT3-dependent pathway. Forensic Toxicol 2025; 43:142-154. [PMID: 39652148 PMCID: PMC11782452 DOI: 10.1007/s11419-024-00708-x] [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: 09/16/2024] [Accepted: 11/24/2024] [Indexed: 01/31/2025]
Abstract
PURPOSE Pyrrolidinophenone derivatives (PPs) are amphetamine-like designer drugs containing a pyrrolidine ring, and their adverse effects resemble those of methamphetamine (METH). Microglial activation has been recently suggested as a key event in eliciting the adverse effects against dysfunction of the central nervous system. The aim of this study is to clarify the mechanisms of microglial activation induced by PPs. METHODS We employed the human microglial cell line HMC3 to assess microglial activation induced by PPs and evaluated the capacities for proliferation and interleukin-6 (IL-6) production that are characteristic features of the activation events. RESULTS The WST-1 assay indicated that viability of HMC3 cells was increased by treatment with sublethal concentrations (5-20 µM) of α-pyrrolidinooctanophenone (α-POP), a highly lipophilic PP, whereas it was decreased by treatment with concentrations above 40 µM. Treatment with sublethal α-POP concentrations up-regulated the expression and secretion of IL-6. Additionally, α-POP-induced increase in cell viability was restored by pretreating with N-acetyl-L-cysteine, a reactive oxygen species (ROS) scavenger, and stattic, an inhibitor of signal transducer and activator of transcription 3 (STAT3), respectively, suggesting that activation of the ROS/STAT3 pathway is involved in the α-POP-induced activation of HMC3 cells. The increases in cell viability were also observed in HMC3 cells treated with other α-POP derivatives and METH. CONCLUSIONS These results suggest that enhanced productions of ROS and IL-6 are also involved in microglial activation by drug treatment and that HMC3 cell-based system is available to evaluate accurately the microglial activation induced by abused drugs.
Collapse
Affiliation(s)
- Yuji Sakai
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan.
| | - Junta Hattori
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu, 502-8585, Japan
| | - Yoshifumi Morikawa
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Toshihiro Matsumura
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu, 502-8585, Japan
| | - Shunsuke Jimbo
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Koichi Suenami
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Tomohiro Takayama
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Atsushi Nagai
- Department of Legal Medicine, Graduate School of Medicine, Gifu University, Gifu, 501-1194, Japan
| | - Tomomi Michiue
- Department of Legal Medicine, Graduate School of Medicine, Gifu University, Gifu, 501-1194, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Toshiyuki Matsunaga
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu, 502-8585, Japan
| |
Collapse
|
4
|
Tang X, Xiang X, Yu Y, Huang S, Pan C, Gan S, Yao Y. Surgery-induced neuroinflammatory transcriptional programs in medial prefrontal cortex of mice during early phase of perioperative neurocognitive disorders. PeerJ 2024; 12:e18664. [PMID: 39650559 PMCID: PMC11623096 DOI: 10.7717/peerj.18664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/18/2024] [Indexed: 12/11/2024] Open
Abstract
Patients receiving anesthesia and surgery may experience cognitive dysfunction, memory deficits, and mental disturbances, which are referred to as perioperative neurocognitive disorders (PND). The function of the medial prefrontal cortex (mPFC) is disrupted during early phase of PND. To gain insight into the mechanisms of PND, we collected mouse mPFC tissues 6 h post-surgery and performed RNA sequencing analysis. In total, 178 differentially expressed genes (DEGs) were identified, including 105 upregulated and 73 downregulated genes. Bioinformatic analysis highlighted the significant enrichment of these DEGs in several immune-related biological processes and signaling pathways, suggesting that pronounced neuroinflammatory transcriptional programming in the mPFC was evoked during early phase of PND. Interleukin-6 level increased in both serum and mPFC, while the mRNA levels of Il-6, Tnf-α, and Il-1β remained unchanged. Taken together, our findings suggest that a distinct and acute neuroinflammatory response in the mPFC is evoked after peripheral surgery, which might play a key role in the development of PND.
Collapse
Affiliation(s)
- Xiaodong Tang
- Department of Anesthesiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xuwu Xiang
- Department of Anesthesiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yang Yu
- Department of Anesthesiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuyuan Huang
- Department of Anesthesiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Caifei Pan
- Department of Anesthesiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuyuan Gan
- Department of Anesthesiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yongxing Yao
- Department of Anesthesiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Balan I, Boero G, Chéry SL, McFarland MH, Lopez AG, Morrow AL. Neuroactive Steroids, Toll-like Receptors, and Neuroimmune Regulation: Insights into Their Impact on Neuropsychiatric Disorders. Life (Basel) 2024; 14:582. [PMID: 38792602 PMCID: PMC11122352 DOI: 10.3390/life14050582] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/18/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Pregnane neuroactive steroids, notably allopregnanolone and pregnenolone, exhibit efficacy in mitigating inflammatory signals triggered by toll-like receptor (TLR) activation, thus attenuating the production of inflammatory factors. Clinical studies highlight their therapeutic potential, particularly in conditions like postpartum depression (PPD), where the FDA-approved compound brexanolone, an intravenous formulation of allopregnanolone, effectively suppresses TLR-mediated inflammatory pathways, predicting symptom improvement. Additionally, pregnane neurosteroids exhibit trophic and anti-inflammatory properties, stimulating the production of vital trophic proteins and anti-inflammatory factors. Androstane neuroactive steroids, including estrogens and androgens, along with dehydroepiandrosterone (DHEA), display diverse effects on TLR expression and activation. Notably, androstenediol (ADIOL), an androstane neurosteroid, emerges as a potent anti-inflammatory agent, promising for therapeutic interventions. The dysregulation of immune responses via TLR signaling alongside reduced levels of endogenous neurosteroids significantly contributes to symptom severity across various neuropsychiatric disorders. Neuroactive steroids, such as allopregnanolone, demonstrate efficacy in alleviating symptoms of various neuropsychiatric disorders and modulating neuroimmune responses, offering potential intervention avenues. This review emphasizes the significant therapeutic potential of neuroactive steroids in modulating TLR signaling pathways, particularly in addressing inflammatory processes associated with neuropsychiatric disorders. It advances our understanding of the complex interplay between neuroactive steroids and immune responses, paving the way for personalized treatment strategies tailored to individual needs and providing insights for future research aimed at unraveling the intricacies of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Irina Balan
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Giorgia Boero
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA;
| | - Samantha Lucenell Chéry
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Minna H. McFarland
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alejandro G. Lopez
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Nagy EK, Overby PF, Leyrer-Jackson JM, Carfagno VF, Acuña AM, Olive MF. Methamphetamine and the Synthetic Cathinone 3,4-Methylenedioxypyrovalerone (MDPV) Produce Persistent Effects on Prefrontal and Striatal Microglial Morphology and Neuroimmune Signaling Following Repeated Binge-like Intake in Male and Female Rats. Brain Sci 2024; 14:435. [PMID: 38790414 PMCID: PMC11118022 DOI: 10.3390/brainsci14050435] [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: 03/21/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Psychostimulants alter cellular morphology and activate neuroimmune signaling in a number of brain regions, yet few prior studies have investigated their persistence beyond acute abstinence or following high levels of voluntary drug intake. In this study, we examined the effects of the repeated binge-like self-administration (96 h/week for 3 weeks) of methamphetamine (METH) and 21 days of abstinence in female and male rats on changes in cell density, morphology, and cytokine levels in two addiction-related brain regions-the prefrontal cortex (PFC) and dorsal striatum (DStr). We also examined the effects of similar patterns of intake of the cocaine-like synthetic cathinone derivative 3,4-methylenedioxypyrovalerone (MDPV) or saline as a control. Robust levels of METH and MDPV intake (~500-1000 infusions per 96 h period) were observed in both sexes. We observed no changes in astrocyte or neuron density in either region, but decreases in dendritic spine densities were observed in PFC pyramidal and DStr medium spiny neurons. The microglial cell density was decreased in the PFC of METH self-administering animals, accompanied by evidence of microglial apoptosis. Changes in microglial morphology (e.g., decreased territorial volume and ramification and increased cell soma volume) were also observed, indicative of an inflammatory-like state. Multiplex analyses of PFC and DStr cytokine content revealed elevated levels of various interleukins and chemokines only in METH self-administering animals, with region- and sex-dependent effects. Our findings suggest that voluntary binge-like METH or MDPV intake induces similar cellular perturbations in the brain, but they are divergent neuroimmune responses that persist beyond the initial abstinence phase.
Collapse
Affiliation(s)
- Erin K. Nagy
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, AZ 85287, USA
| | - Paula F. Overby
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, AZ 85287, USA
| | - Jonna M. Leyrer-Jackson
- Department of Medical Education, School of Medicine, Creighton University, Phoenix, AZ 85012, USA
| | - Vincent F. Carfagno
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Amanda M. Acuña
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, AZ 85287, USA
- Interdisciplinary Graduate Program in Neuroscience, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - M. Foster Olive
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, AZ 85287, USA
- Interdisciplinary Graduate Program in Neuroscience, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
7
|
Nagy EK, Leyrer-Jackson JM, Hood LE, Acuña AM, Olive MF. Effects of repeated binge intake of the pyrovalerone cathinone derivative 3,4-methylenedioxypyrovalerone on prefrontal cytokine levels in rats - a preliminary study. Front Behav Neurosci 2023; 17:1275968. [PMID: 38025384 PMCID: PMC10668493 DOI: 10.3389/fnbeh.2023.1275968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Drugs of abuse activate neuroimmune signaling in addiction-related regions of the brain, including the prefrontal cortex (PFC) which mediates executive control, attention, and behavioral inhibition. Traditional psychostimulants including methamphetamine and cocaine are known to induce PFC inflammation, yet the effects of synthetic cathinone derivatives are largely unexplored. In this study, we examined the ability of repeated binge-like intake of the pyrovalerone cathinone derivative 3,4-methylenedioxypyrovalerone (MDPV) to alter cytokine profiles in the PFC. Male and female rats were allowed to intravenously self-administer MDPV (0.05 mg/kg/infusion) or saline as a control under conditions of prolonged binge-like access, consisting of three 96 h periods of drug access interspersed with 72 h of forced abstinence. Three weeks following cessation of drug availability, PFC cytokine levels were assessed using antibody arrays. Employing the unsupervised clustering and regression analysis tool CytoMod, a single module of co-signaling cytokines associated with MDPV intake regardless of sex was identified. With regards to specific cytokines, MDPV intake was positively associated with PFC levels of VCAM-1/CD106 and negatively associated with levels of Flt-3 ligand. These findings indicate that prolonged MDPV intake causes changes in PFC cytokine levels that persist into abstinence; however, the functional ramifications of these changes remain to be fully elucidated.
Collapse
Affiliation(s)
- Erin K. Nagy
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, AZ, United States
| | - Jonna M. Leyrer-Jackson
- Department of Medical Education, School of Medicine, Creighton University, Phoenix, AZ, United States
| | - Lauren E. Hood
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, AZ, United States
| | - Amanda M. Acuña
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, AZ, United States
- Interdisciplinary Graduate Program in Neuroscience, School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - M. Foster Olive
- Department of Psychology, Behavioral Neuroscience and Comparative Psychology Area, Arizona State University, Tempe, AZ, United States
- Interdisciplinary Graduate Program in Neuroscience, School of Life Sciences, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
8
|
Vilca SJ, Margetts AV, Pollock TA, Tuesta LM. Transcriptional and epigenetic regulation of microglia in substance use disorders. Mol Cell Neurosci 2023; 125:103838. [PMID: 36893849 PMCID: PMC10247513 DOI: 10.1016/j.mcn.2023.103838] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/17/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
Microglia are widely known for their role in immune surveillance and for their ability to refine neurocircuitry during development, but a growing body of evidence suggests that microglia may also play a complementary role to neurons in regulating the behavioral aspects of substance use disorders. While many of these efforts have focused on changes in microglial gene expression associated with drug-taking, epigenetic regulation of these changes has yet to be fully understood. This review provides recent evidence supporting the role of microglia in various aspects of substance use disorder, with particular focus on changes to the microglial transcriptome and the potential epigenetic mechanisms driving these changes. Further, this review discusses the latest technical advances in low-input chromatin profiling and highlights the current challenges for studying these novel molecular mechanisms in microglia.
Collapse
Affiliation(s)
- Samara J Vilca
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| | - Alexander V Margetts
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| | - Tate A Pollock
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America
| | - Luis M Tuesta
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States of America.
| |
Collapse
|
9
|
Machado da Silva MC, Iglesias LP, Candelario-Jalil E, Khoshbouei H, Moreira FA, de Oliveira ACP. Role of Microglia in Psychostimulant Addiction. Curr Neuropharmacol 2023; 21:235-259. [PMID: 36503452 PMCID: PMC10190137 DOI: 10.2174/1570159x21666221208142151] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022] Open
Abstract
The use of psychostimulant drugs can modify brain function by inducing changes in the reward system, mainly due to alterations in dopaminergic and glutamatergic transmissions in the mesocorticolimbic pathway. However, the etiopathogenesis of addiction is a much more complex process. Previous data have suggested that microglia and other immune cells are involved in events associated with neuroplasticity and memory, which are phenomena that also occur in addiction. Nevertheless, how dependent is the development of addiction on the activity of these cells? Although the mechanisms are not known, some pathways may be involved. Recent data have shown psychoactive substances may act directly on immune cells, alter their functions and induce various inflammatory mediators that modulate synaptic activity. These could, in turn, be involved in the pathological alterations that occur in substance use disorder. Here, we extensively review the studies demonstrating how cocaine and amphetamines modulate microglial number, morphology, and function. We also describe the effect of these substances in the production of inflammatory mediators and a possible involvement of some molecular signaling pathways, such as the toll-like receptor 4. Although the literature in this field is scarce, this review compiles the knowledge on the neuroimmune axis that is involved in the pathogenesis of addiction, and suggests some pharmacological targets for the development of pharmacotherapy.
Collapse
Affiliation(s)
- Maria Carolina Machado da Silva
- Department of Pharmacology, Neuropharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil;
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lia Parada Iglesias
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fabrício Araujo Moreira
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
10
|
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: 13] [Impact Index Per Article: 4.3] [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
|
11
|
Rungnirundorn T, Krusong K, Kalayasiri R, Maes M. Leukocyte telomere length is not shortened in methamphetamine dependence or methamphetamine-induced psychosis but is increased following traumatic events. World J Biol Psychiatry 2022; 23:613-621. [PMID: 34895035 PMCID: PMC9991870 DOI: 10.1080/15622975.2021.2016957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 01/25/2023]
Abstract
OBJECTIVE This study aims to examine the effects of methamphetamine (MA) use and dependence and MA withdrawal symptoms on the telomere length and whether shortening of the latter is associated with MA-induced psychosis (MIP). METHODS This study included 185 MA-abuse, 118 MA-dependent, and 67 MIP patients, diagnosed using DSM-IV criteria. The Semi-structured Assessment for Drug Dependence and Alcoholism (SSADDA) questionnaire was employed to collect MA-related data. MIP was confirmed using the Methamphetamine Experience Questionnaire (MEQ). The leukocyte telomere length was measured using real-time polymerase chain reaction measuring the Telomere/Single gene ratio (T/S ratio). Data were analysed using multivariate statistical analyses. RESULTS There were no significant associations between the T/S ratio and severity of MA-use, MIP, and MA withdrawal symptoms. MIP was significantly predicted by alcohol dependence, antisocial personality disorder, and MA-use severity. There were significantly positive associations between the T/S ratio and previous traumatic and life-threatening events. The T/S ratio was not affected by alcohol and nicotine dependence. Alcohol and nicotine dependence, antisocial personality disorder, and severity of MA use increased risk of MA withdrawal symptoms. CONCLUSION MIP and MA-use severity are not associated with leukocyte telomere length, but previous traumatic and life-threatening events are associated with increased telomere length.
Collapse
Affiliation(s)
| | - Kuakarun Krusong
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Rasmon Kalayasiri
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
- IMPACT Strategic Research Centre, Deakin University, PO Box 281, Geelong, VIC, 3220, Australia
| |
Collapse
|
12
|
Methamphetamine Induces Systemic Inflammation and Anxiety: The Role of the Gut–Immune–Brain Axis. Int J Mol Sci 2022; 23:ijms231911224. [PMID: 36232524 PMCID: PMC9569811 DOI: 10.3390/ijms231911224] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 12/21/2022] Open
Abstract
Methamphetamine (METH) is a highly addictive drug abused by millions of users worldwide, thus becoming a global health concern with limited management options. The inefficiency of existing treatment methods has driven research into understanding the mechanisms underlying METH-induced disorders and finding effective treatments. This study aims to understand the complex interactions of the gastrointestinal–immune–nervous systems following an acute METH dose administration as one of the potential underlying molecular mechanisms concentrating on the impact of METH abuse on gut permeability. Findings showed a decreased expression of tight junction proteins ZO-1 and EpCAm in intestinal tissue and the presence of FABP-1 in sera of METH treated mice suggests intestinal wall disruption. The increased presence of CD45+ immune cells in the intestinal wall further confirms gut wall inflammation/disruption. In the brain, the expression of inflammatory markers Ccl2, Cxcl1, IL-1β, TMEM119, and the presence of albumin were higher in METH mice compared to shams, suggesting METH-induced blood–brain barrier disruption. In the spleen, cellular and gene changes are also noted. In addition, mice treated with an acute dose of METH showed anxious behavior in dark and light, open field, and elevated maze tests compared to sham controls. The findings on METH-induced inflammation and anxiety may provide opportunities to develop effective treatments for METH addiction in the future.
Collapse
|
13
|
Fan R, Shen Y, Li X, Luo H, Zhang P, Liu Y, Si Z, Zhou W, Liu Y. The effect of the NLRP1 inflammasome on methamphetamine-induced cognitive impairment in rats. Drug Alcohol Depend 2022; 237:109537. [PMID: 35752024 DOI: 10.1016/j.drugalcdep.2022.109537] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/11/2022] [Accepted: 06/08/2022] [Indexed: 11/03/2022]
Abstract
Methamphetamine (METH) use disorder has been shown to be in high comorbidity with cognitive deficits. METH-induced cognitive deficits are accompanied by neurotoxicity which could result from neuroinflammation. The potential role of NLRP1 inflammasome (NLRP1) and the downstream signalling pathway in METH-induced cognitive impairment was explored in the current study. Cognitive functions and the changes of NLRP1/Caspase-1/GSDMD signalling pathway were firstly determined in rats receiving daily injections of METH. Subsequently, the effects of aspirin-triggered-lipoxin A4 (ATL), a potent anti-inflammatory mediator, and NLRP1 siRNA was investigated were investigated in both METH-treated rats and HT22 cells. METH induces significant cognitive deficits in rats, using the NOR test. METH-induced cognitive impairment was in line with increased activities of NLRP1, cleaved-Caspase-11, IL-1β and TNF-α and the presence of GSDMD-mediated pyroptosis in the hippocampus of rats. NLRP1 inhibition by ATL significantly attenuated METH-induced cognitive impairment, in conjunction with the decreased activities of NLRP1 and cleaved-Caspase-1, IL-1β and TNF-α. ATL and NLRP1 siRNA also prevented the presence of apoptosis in the hippocampus of METH-treated rats and the cell death in METH-treated HT22 cells. These results reveal a novel role of NLRP1 and the downstream signaling pathways in the complex actions of METH-induced cognitive deficits.
Collapse
Affiliation(s)
- Runyue Fan
- School of Public Health, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang 315211, PR China
| | - Yao Shen
- School of Public Health, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang 315211, PR China
| | - Xiaofang Li
- School of Teaching and Education, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang 315211, PR China
| | - Hu Luo
- School of Teaching and Education, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang 315211, PR China
| | - Peng Zhang
- School of Public Health, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang 315211, PR China
| | - Yingying Liu
- School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang 315211, PR China
| | - Zizhen Si
- Department of Pharmacy, the Affiliated Hospital of Ningbo University Medical School, Ningbo , Zhejiang 315211, PR China; Department of Physiology and Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang 315211, PR China
| | - Wenhua Zhou
- Kangning Hospital, 1 South Zhuangyu Road, Ningbo, Zhejiang 315201, PR China
| | - Yu Liu
- Department of Physiology and Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang 315211, PR China.
| |
Collapse
|
14
|
Garmabi B, Mohaddes R, Rezvani F, Mohseni F, Khastar H, Khaksari M. Erythropoietin improve spatial memory impairment following methamphetamine neurotoxicity by inhibition of apoptosis, oxidative stress and neuroinflammation in CA1 area of hippocampus. J Chem Neuroanat 2022; 124:102137. [PMID: 35842017 DOI: 10.1016/j.jchemneu.2022.102137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Methamphetamine (METH) is one of the most widely used addictive drugs, and addiction to it is on the rise all over the world. METH abuse has long-term damaging effects that reduce memory and impair cognitive functions. According to studies, the observed effects are strongly related to the nerve cell damage caused by METH, which leads to neurotoxicity. Some of these intra-neuronal events include dopamine oxidation, excitotoxicity, and oxidative stress. Erythropoietin (EPO) is a hormone produced primarily by the kidneys and, in small quantities, by the liver. Studies have shown that EPO exhibits considerable neuroprotective effects. This study aimed to investigate the protective effects of EPO on METH neurotoxicity. METHODS Initially, 48 male Wistar rats, weighing 250-300 g, were randomly assigned to four groups: control (n = 12), METH (n = 12), and METH+EPO (2500, 5000 IU/kg/IP- n = 12). METH was injected intraperitoneally at a dose of 40 mg per kg of body weight (four injections of 10 mg every two hours) to induce neurotoxicity. EPO was injected at doses of 2500 and 5000 IU/kg seven days after the last METH administration (ip). Morris water maze test was performed following EPO injection (1 day after the last dose) to assess spatial memory. The brains were removed after the behavioral test, biochemical evaluations and immunohistochemistry (caspase-3 and GFAP) was performed. RESULTS The results showed that EPO treatment significantly improved spatial memory impairment (P < 0.01), compared to the METH group, EPO was a significant reduction in malondialdehyde and TNF-α (P < 0.01), as well as an increase in superoxide dismutase (P < 0.05) and glutathione-PX (P < 0.01). Furthermore, EPO treatment significantly reduced the number of GFAP positive cells (P < 0.01) and caspase 3 (P < 0.001) in the hippocampus (CA1 region). CONCLUSIONS The study findings suggested that EPO may have great neuroprotective effects on METH neurotoxicity due to its anti-inflammatory, antioxidant, and antiapoptotic properties.
Collapse
Affiliation(s)
- Behzad Garmabi
- Neurosciences Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Reza Mohaddes
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Fatemeh Rezvani
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Fahimeh Mohseni
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Hossein Khastar
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mehdi Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| |
Collapse
|
15
|
Long-term exercise at different intensities can reduce the inflammatory response in the brains of methamphetamine-treated mice. Biochem Biophys Res Commun 2022; 613:201-206. [DOI: 10.1016/j.bbrc.2022.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
|
16
|
Zeng Y, Chen Y, Zhang S, Ren H, Xia J, Liu M, Shan B, Ren Y. Natural Products in Modulating Methamphetamine-Induced Neuronal Apoptosis. Front Pharmacol 2022; 12:805991. [PMID: 35058785 PMCID: PMC8764133 DOI: 10.3389/fphar.2021.805991] [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: 10/31/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Methamphetamine (METH), an amphetamine-type psychostimulant, is highly abused worldwide. Chronic abuse of METH causes neurodegenerative changes in central dopaminergic neurons with numerous neuropsychiatric consequences. Neuronal apoptosis plays a critical role in METH-induced neurotoxicity and may provide promising pharmacological targets for preventing and treating METH addiction. In recent years, accumulating evidence has revealed that natural products may possess significant potentials to inhibit METH-evoked neuronal apoptosis. In this review, we summarized and analyzed the improvement effect of natural products on METH-induced neuronal apoptosis and their potential molecular mechanisms on modulating dopamine release, oxidative stress, mitochondrial-dependent apoptotic pathway, endoplasmic reticulum stress-mediated apoptotic pathway, and neuroinflammation. Hopefully, this review may highlight the potential value of natural products in modulating METH-caused neuronal apoptosis and provide useful information for future research and developments of novel and efficacious pharmacotherapies in this field.
Collapse
Affiliation(s)
- Yiwei Zeng
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunhui Chen
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Su Zhang
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Ren
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jialin Xia
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengnan Liu
- Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, China
| | - Baozhi Shan
- School of Humanities, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yulan Ren
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
17
|
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: 31] [Impact Index Per Article: 10.3] [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
|
18
|
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
|
19
|
Canedo T, Portugal CC, Socodato R, Almeida TO, Terceiro AF, Bravo J, Silva AI, Magalhães JD, Guerra-Gomes S, Oliveira JF, Sousa N, Magalhães A, Relvas JB, Summavielle T. Astrocyte-derived TNF and glutamate critically modulate microglia activation by methamphetamine. Neuropsychopharmacology 2021; 46:2358-2370. [PMID: 34400780 PMCID: PMC8581027 DOI: 10.1038/s41386-021-01139-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/12/2021] [Accepted: 07/24/2021] [Indexed: 02/06/2023]
Abstract
Methamphetamine (Meth) is a powerful illicit psychostimulant, widely used for recreational purposes. Besides disrupting the monoaminergic system and promoting oxidative brain damage, Meth also causes neuroinflammation, contributing to synaptic dysfunction and behavioral deficits. Aberrant activation of microglia, the largest myeloid cell population in the brain, is a common feature in neurological disorders triggered by neuroinflammation. In this study, we investigated the mechanisms underlying the aberrant activation of microglia elicited by Meth in the adult mouse brain. We found that binge Meth exposure caused microgliosis and disrupted risk assessment behavior (a feature that usually occurs in individuals who abuse Meth), both of which required astrocyte-to-microglia crosstalk. Mechanistically, Meth triggered a detrimental increase of glutamate exocytosis from astrocytes (in a process dependent on TNF production and calcium mobilization), promoting microglial expansion and reactivity. Ablating TNF production, or suppressing astrocytic calcium mobilization, prevented Meth-elicited microglia reactivity and re-established risk assessment behavior as tested by elevated plus maze (EPM). Overall, our data indicate that glial crosstalk is critical to relay alterations caused by acute Meth exposure.
Collapse
Affiliation(s)
- Teresa Canedo
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Camila Cabral Portugal
- Glial Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
| | - Renato Socodato
- grid.5808.50000 0001 1503 7226Glial Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Tiago Oliveira Almeida
- grid.5808.50000 0001 1503 7226Glial Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana Filipa Terceiro
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Joana Bravo
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana Isabel Silva
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - João Duarte Magalhães
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Sónia Guerra-Gomes
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - João Filipe Oliveira
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal ,grid.410922.c0000 0001 0180 6901IPCA-EST-2Ai, Polytechnic Institute of Cávado and Ave, Applied Artificial Intelligence, Barcelos, Portugal
| | - Nuno Sousa
- grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Magalhães
- grid.5808.50000 0001 1503 7226Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal ,grid.5808.50000 0001 1503 7226Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - João Bettencourt Relvas
- grid.5808.50000 0001 1503 7226Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal ,grid.5808.50000 0001 1503 7226Glial Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Teresa Summavielle
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde and IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal. .,ESS.PP, Escola Superior de Saúde do Politécnico do Porto, Porto, Portugal.
| |
Collapse
|
20
|
Kobeissy FH, Shakkour Z, Hayek SE, Mohamed W, Gold MS, Wang KKW. Elevation of Pro-inflammatory and Anti-inflammatory Cytokines in Rat Serum after Acute Methamphetamine Treatment and Traumatic Brain Injury. J Mol Neurosci 2021; 72:158-168. [PMID: 34542809 DOI: 10.1007/s12031-021-01886-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/05/2021] [Indexed: 11/24/2022]
Abstract
The use of methamphetamine (METH) is a growing worldwide epidemic that bears grave societal implications. METH is known to exert its neurotoxic effects on the dopaminergic and serotonergic systems of the brain. In addition to this classical studied mechanism of damage, findings from our laboratory and others have shown that acute METH treatment and mechanical injury, i.e. traumatic brain injury (TBI), share common cell injury mechanism(s). Since neuro-inflammation is a signature event in TBI, we hypothesize that certain cytokine levels might also be altered in rat brain exposed to an acute METH insult. In this study, using a cytokine antibody array chip, we evaluated the serum levels of 19 cytokines in rats 24 h after exposure to a 40 mg/kg acute regimen of METH. Data were compared to rats subjected to experimental TBI using the controlled cortical impact (CCI) injury model and saline controls. Sandwich ELISA method was used to further validate some of the findings obtained from the antibody cytokine array. We confirmed that three major inflammatory-linked cytokines (IL-1β, IL-6, and IL-10) were elevated in the METH and TBI groups compared to the saline group. Such finding suggests the involvement of an inflammatory process in these brain insults, indicating that METH use is, in fact, a stressor to the immune system where systemic involvement of an altered cytokine profile may play a major role in mediating chemical brain injury after METH use.
Collapse
Affiliation(s)
- Firas H Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Program for Neurotrauma, Neuroproteomics, and Biomarkers Research, Gainesville, FL, USA.,Department of Emergency Medicine, University of Florida, Gainesville, FL, USA
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samer El Hayek
- Department of Psychiatry, American University of Beirut, Beirut, Lebanon
| | - Wael Mohamed
- Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Al Minufya, Egypt.,Basic medical science department, Kulliyyah of Medicine, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Mark S Gold
- Washington University School of Medicine, Department of Psychiatry, and National Council, Washington University in St. Louis, Institute for Public Health, St. Louis, MO, USA
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics, and Biomarkers Research, Gainesville, FL, USA. .,Department of Emergency Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
21
|
Rahimian R, Wakid M, O'Leary LA, Mechawar N. The emerging tale of microglia in psychiatric disorders. Neurosci Biobehav Rev 2021; 131:1-29. [PMID: 34536460 DOI: 10.1016/j.neubiorev.2021.09.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/18/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
As the professional phagocytes of the brain, microglia orchestrate the immunological response and play an increasingly important role in maintaining homeostatic brain functions. Microglia are activated by pathological events or slight alterations in brain homeostasis. This activation is dependent on the context and type of stressor or pathology. Through secretion of cytokines, chemokines and growth factors, microglia can strongly influence the response to a stressor and can, therefore, determine the pathological outcome. Psychopathologies have repeatedly been associated with long-lasting priming and sensitization of cerebral microglia. This review focuses on the diversity of microglial phenotype and function in health and psychiatric disease. We first discuss the diverse homeostatic functions performed by microglia and then elaborate on context-specific spatial and temporal microglial heterogeneity. Subsequently, we summarize microglia involvement in psychopathologies, namely major depressive disorder, schizophrenia and bipolar disorder, with a particular focus on post-mortem studies. Finally, we postulate microglia as a promising novel therapeutic target in psychiatry through antidepressant and antipsychotic treatment.
Collapse
Affiliation(s)
- Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada
| | - Marina Wakid
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Liam Anuj O'Leary
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
22
|
Gruol DL, Hernandez RV, Roberts A. Alcohol Enhances Responses to High Frequency Stimulation in Hippocampus from Transgenic Mice with Increased Astrocyte Expression of IL-6. Cell Mol Neurobiol 2021; 41:1299-1310. [PMID: 32562098 PMCID: PMC7749046 DOI: 10.1007/s10571-020-00902-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/09/2020] [Indexed: 12/29/2022]
Abstract
Recent studies show that alcohol exposure can induce glial production of neuroimmune factors in the CNS. Of these, IL-6 has gained attention because it is involved in a number of important physiological and pathophysiological processes that could be affected by alcohol-induced CNS production of IL-6, particularly under conditions of excessive alcohol use. For example, IL-6 has been shown to play a role in hippocampal behaviors and synaptic plasticity (long-term potentiation; LTP) associated with memory and learning. Surprisingly, in our in vitro studies of LTP at the Schaffer collateral to CA1 pyramidal neuron synapse in hippocampus from transgenic mice that express elevated levels of astrocyte produced IL-6 (TG), LTP was not altered by the increased levels of IL-6. However, exposure to acute alcohol revealed neuroadaptive changes that served to protect LTP against the alcohol-induced reduction of LTP observed in hippocampus from non-transgenic control mice (WT). Here we examined the induction phase of LTP to assess if presynaptic neuroadaptive changes occurred in the hippocampus of TG mice that contributed to the resistance of LTP to alcohol. Results are consistent with a role for IL-6-induced neuroadaptive effects on presynaptic mechanisms involved in transmitter release in the resistance of LTP to alcohol in hippocampus from the TG mice. These actions are important with respect to a role for IL-6 in physiological and pathophysiological processes in the CNS and in CNS actions of alcohol, especially when excessive alcohol used is comorbid with conditions associated with elevated levels of IL-6 in the CNS.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, SR301, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - Ruben V Hernandez
- Neuroscience Department, SR301, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Amanda Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
23
|
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
|
24
|
Limanaqi F, Busceti CL, Celli R, Biagioni F, Fornai F. Autophagy as a gateway for the effects of methamphetamine: From neurotransmitter release and synaptic plasticity to psychiatric and neurodegenerative disorders. Prog Neurobiol 2021; 204:102112. [PMID: 34171442 DOI: 10.1016/j.pneurobio.2021.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
As a major eukaryotic cell clearing machinery, autophagy grants cell proteostasis, which is key for neurotransmitter release, synaptic plasticity, and neuronal survival. In line with this, besides neuropathological events, autophagy dysfunctions are bound to synaptic alterations that occur in mental disorders, and early on, in neurodegenerative diseases. This is also the case of methamphetamine (METH) abuse, which leads to psychiatric disturbances and neurotoxicity. While consistently altering the autophagy machinery, METH produces behavioral and neurotoxic effects through molecular and biochemical events that can be recapitulated by autophagy blockade. These consist of altered physiological dopamine (DA) release, abnormal stimulation of DA and glutamate receptors, as well as oxidative, excitotoxic, and neuroinflammatory events. Recent molecular insights suggest that METH early impairs the autophagy machinery, though its functional significance remains to be investigated. Here we discuss evidence suggesting that alterations of DA transmission and autophagy are intermingled within a chain of events underlying behavioral alterations and neurodegenerative phenomena produced by METH. Understanding how METH alters the autophagy machinery is expected to provide novel insights into the neurobiology of METH addiction sharing some features with psychiatric disorders and parkinsonism.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy
| | | | - Roberta Celli
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy; IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy.
| |
Collapse
|
25
|
Hadizadeh-Bazaz M, Vaezi G, Khaksari M, Hojati V. Curcumin attenuates spatial memory impairment by anti-oxidative, anti-apoptosis, and anti-inflammatory mechanism against methamphetamine neurotoxicity in male Wistar rats: Histological and biochemical changes. Neurotoxicology 2021; 84:208-217. [PMID: 33819551 DOI: 10.1016/j.neuro.2021.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Methamphetamine is used extensively around the world as a psychostimulant. The complications related to methamphetamine include methamphetamine-induced neurotoxicity, mainly involving intraneuronal processes, such as oxidative stress and excitotoxicity. Curcumin is effective against neuronal injury due to its antioxidant, anti-inflammatory effects. In this study, we examined the protective effects of curcumin against methamphetamine neurotoxicity. METHODS Sixty male Wistar rats were divided into the following groups: control (n = 12), DMSO (n = 12), methamphetamine (n = 12), and methamphetamine + curcumin (100 and 200 mg/kg, respectively, intraperitoneal [IP]; n = 12). Neurotoxicity was induced by 40 mg/kg of methamphetamine administrated through 4 injections (4 × 10 mg/kg, q2h, IP). Curcumin (100 and 200 mg/kg) was administered at 7 days after the last methamphetamine injection. By using a Morris water maze task, the hippocampus-dependent memory and spatial learning were evaluated 1 day after the last curcumin injection. Then, the animal brains were isolated for biochemical measurements, as well as glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor protein-1(Iba-1) and caspase-3 immunohistochemical staining. RESULTS The current study demonstrated that administration of curcumin significantly attenuates spatial memory impairment (P < 0.01) following methamphetamine neurotoxicity. Curcumin caused a significant increase in the levels of superoxide dismutase and glutathione peroxidase (P < 0.05). However, it decreased tumor necrosis factor (TNF-α) (P < 0.05) and malondialdehyde (P < 0.01) levels as compared to the methamphetamine group. Also, curcumin significantly reduced Iba-1 (P < 0. 01), GFAP and caspase-3 positive cells in the hippocampus (P < 0.001). CONCLUSION Curcumin exerted neuroprotective effects on methamphetamine neurotoxicity because of its antioxidant and anti-inflammatory effect.
Collapse
Affiliation(s)
| | - Golamhassan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Mehdi Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Vida Hojati
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
26
|
Ghazvini H, Tirgar F, Khodamoradi M, Akbarnejad Z, Rafaiee R, Seyedhosseini Tamijani SM, Asadi-Shekaari M, Esmaeilpour K, Sheibani V. Ovarian hormones prevent methamphetamine-induced anxiety-related behaviors and neuronal damage in ovariectomized rats. Neurosci Lett 2021; 746:135652. [PMID: 33482310 DOI: 10.1016/j.neulet.2021.135652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 01/09/2023]
Abstract
Methamphetamine (METH) may cause long‒lasting neurotoxic effects and cognitive impairment. On the other hand, the ovarian hormones estrogen and progesterone have neuroprotective effects. In the current study, we aimed to examine the effects of estrogen and progesterone on anxiety‒like behavior and neuronal damage in METH‒exposed ovariectomized (OVX) rats. Three weeks after ovariectomy, the animals received estrogen (1 mg/kg, i.p.), or progesterone (8 mg/kg, i.p.), or estrogen plus progesterone (with the same doses), or vehicle during 7 consecutive days (days 22-28). On day 28, OVX rats were exposed to a single‒day METH regimen (6 mg/kg, four s.c. Injections, with 2 h interval) 30 min after the hormone treatment. The next day (on day 29), the animals were assessed for anxiety‒related behaviors using the open field and elevated plus‒maze tasks. The animals were then sacrificed and brain water content, cell apoptosis and expression of IL-1β were evaluated. The findings showed that treatment with estrogen or progesterone alone in METH‒exposed rats significantly improved hyperthermia, anxiety‒like behavior, neuronal damage, and inflammation in the CA1 area. Also, treatment with estrogen plus progesterone improved hyperthermia and brain edema. Taken together, the findings suggest that treatment with ovarian hormones can partially prevent hyperthermia and anxiety‒related behaviors induced by METH in OVX rats, which could be accompanied by their neuroprotective effects in the hippocampus.
Collapse
Affiliation(s)
- Hamed Ghazvini
- Department of Neuroscience, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Tirgar
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Khodamoradi
- Substance Abuse Prevention Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeinab Akbarnejad
- ENT and Head and Neck Research Center and Department, Hazrat Rasoul Akram Hospital, The Five Senses Health Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Raheleh Rafaiee
- Department of Neuroscience, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Majid Asadi-Shekaari
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Khadijeh Esmaeilpour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
27
|
Lucerne KE, Kiraly DD. The role of gut-immune-brain signaling in substance use disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:311-370. [PMID: 33648673 DOI: 10.1016/bs.irn.2020.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Substance use disorders (SUDs) are debilitating neuropsychiatric conditions that exact enormous costs in terms of loss of life and individual suffering. While much progress has been made defining the neurocircuitry and intracellular signaling cascades that contribute to SUDs, these studies have yielded limited effective treatment options. This has prompted greater exploration of non-traditional targets in addiction. Emerging data suggest inputs from peripheral systems, such as the immune system and the gut microbiome, impact multiple neuropsychiatric diseases, including SUDs. Until recently the gut microbiome, peripheral immune system, and the CNS have been studied independently; however, current work shows the gut microbiome and immune system critically interact to modulate brain function. Additionally, the gut microbiome and immune system intimately regulate one another via extensive bidirectional communication. Accumulating evidence suggests an important role for gut-immune-brain communication in the pathogenesis of substance use disorders. Thus, a better understanding of gut-immune-brain signaling could yield important insight to addiction pathology and potential treatment options.
Collapse
Affiliation(s)
- Kelsey E Lucerne
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
28
|
Inflammation but not programmed cell death is activated in methamphetamine-dependent patients: Relevance to the brain function. Int J Psychophysiol 2020; 157:42-50. [PMID: 32976886 DOI: 10.1016/j.ijpsycho.2020.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/31/2020] [Accepted: 09/11/2020] [Indexed: 11/23/2022]
Abstract
Animal studies have shown that methamphetamine (MA) induces neurodegeneration through programmed cell death, however, the effects of MA on human brain and the extent of induced neural degeneration is not well understood. Given that the dose and duration of MA administration differ in animals and humans, we evaluated MA effects on active users considering brain damage mechanisms. Nineteen active MA-dependent patients and 18 healthy controls performed the color-word Stroop task, during fMRI and their blood samples were collected. Human enzyme-linked immunosorbent assays (ELISA) and quantitative PCR were applied to measure circulating proteins and miRNAs involved in various programmed cell death pathways (apoptosis, necroptosis, and autophagy), brain damage and neuroinflammation. Results showed the performance deficit in color-word Stroop task in MA abusers as well as higher activations of the right inferior and middle temporal gyri detected by fMRI. Structural MRI revealed increased white matter volume in MA-dependent patients in the superior and medial frontal gyri, and left/right middle temporal gyrus. Molecular analyses detected no significant differences in the plasma levels of the studied proteins and miRNAs of MA-dependent patients and controls except the higher levels of MBP, S100B, and TNFα in MA abusers. Results showed that MA induced physiological and structural changes accompanied by inflammation and release of damage-associated molecules in MA-dependent patients.
Collapse
|
29
|
Wei Y, Shah R. Substance Use Disorder in the COVID-19 Pandemic: A Systematic Review of Vulnerabilities and Complications. Pharmaceuticals (Basel) 2020; 13:E155. [PMID: 32708495 PMCID: PMC7407364 DOI: 10.3390/ph13070155] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 01/18/2023] Open
Abstract
As the world endures the coronavirus disease 2019 (COVID-19) pandemic, the conditions of 35 million vulnerable individuals struggling with substance use disorders (SUDs) worldwide have not received sufficient attention for their special health and medical needs. Many of these individuals are complicated by underlying health conditions, such as cardiovascular and lung diseases and undermined immune systems. During the pandemic, access to the healthcare systems and support groups is greatly diminished. Current research on COVID-19 has not addressed the unique challenges facing individuals with SUDs, including the heightened vulnerability and susceptibility to the disease. In this systematic review, we will discuss the pathogenesis and pathology of COVID-19, and highlight potential risk factors and complications to these individuals. We will also provide insights and considerations for COVID-19 treatment and prevention in patients with SUDs.
Collapse
Affiliation(s)
- Yufeng Wei
- Department of Chemistry, New Jersey City University, Jersey City, NJ 07305, USA;
| | | |
Collapse
|
30
|
Abstract
The pervasive and devastating nature of substance use disorders underlies the need for the continued development of novel pharmacotherapies. We now know that glia play a much greater role in neuronal processes than once believed. The various types of glial cells (e.g., astrocytes, microglial, oligodendrocytes) participate in numerous functions that are crucial to healthy central nervous system function. Drugs of abuse have been shown to interact with glia in ways that directly contribute to the pharmacodynamic effects responsible for their abuse potential. Through their effect upon glia, drugs of abuse also alter brain function resulting in behavioral changes associated with substance use disorders. Therefore, drug-induced changes in glia and inflammation within the central nervous system (neuroinflammation) have been investigated to treat various aspects of drug abuse and dependence. This article presents a brief overview of the effects of each of the major classes of addictive drugs on glia. Next, the paper reviews the pre-clinical and clinical studies assessing the effects that glial modulators have on abuse-related behavioral effects, such as pleasure, withdrawal, and motivation. There is a strong body of pre-clinical literature demonstrating the general effectiveness of several glia-modulating drugs in models of reward and relapse. Clinical studies have also yielded promising results, though not as robust. There is still much to disentangle regarding the integration between addictive drugs and glial cells. Improved understanding of the relationship between glia and the pathophysiology of drug abuse should allow for more precise exploration in the development and testing of glial-directed treatments for substance use disorders.
Collapse
Affiliation(s)
- Jermaine D. Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
31
|
Protective Effects of Melatonin on Methamphetamine-Induced Blood-Brain Barrier Dysfunction in Rat Model. Neurotox Res 2020; 37:640-660. [PMID: 31900895 DOI: 10.1007/s12640-019-00156-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/26/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022]
Abstract
The specialized brain endothelial cells interconnected by unique junctions and adhesion molecules are distinctive features of the blood-brain barrier (BBB), maintaining the homeostasis of the cerebral microenvironment. This study was designed to investigate the protective effects of melatonin on methamphetamine (METH)-induced alterations of BBB integrity. Wistar rats were randomly distributed into groups and underwent melatonin pretreatment and escalating-high doses of METH treatment. Immunohistochemistry was performed to demonstrate the BBB leakage. Protein and RNA samples were isolated from hippocampal and prefrontal cortical tissues and measured expression levels of molecular markers associated with BBB structural components and inflammatory processes. METH provoked the loss of zonula occludens (ZO)-1, occludin, and claudin-5 tight junction proteins. Furthermore, METH caused an excessive increase in matrix metalloproteinase-9 (MMP-9) enzyme, intercellular adhesion molecule 1 (ICAM-1), and vascular cell adhesion molecule 1 (VCAM-1) and the increase in NAD(P)H oxidase 2 (NOX2). Melatonin exerted the protective effects by recovering tight junction loss; attenuating excessive MMP-9, NOX2, and cell adhesion molecule expression; and reducing serum albumin in the brain. Our results also showed the protective effects of melatonin against METH neurotoxic profiles, characterized by reactive gliosis: microglia (integrin-αM) and astrocyte (GFAP); an excessive upregulation of primary pro-inflammatory cytokines: interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α); activation of neuroinflammatory signaling: nuclear factor-kappa B (NF-κB); and suppression of anti-oxidative signaling: nuclear factor erythroid 2-related factor (Nrf2), that may exacerbate BBB structural impairment. Our results provide insights into the beneficial effects of melatonin against METH-induced BBB disruption and mechanisms that play detrimental roles in BBB impairment by in vivo design.
Collapse
|
32
|
Nolan RA, Reeb KL, Rong Y, Matt SM, Johnson HS, Runner K, Gaskill PJ. Dopamine activates NF-κB and primes the NLRP3 inflammasome in primary human macrophages. Brain Behav Immun Health 2019; 2. [PMID: 33665636 PMCID: PMC7929492 DOI: 10.1016/j.bbih.2019.100030] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Induction of innate immune genes in the brain is thought to be a major factor in the development of addiction to substances of abuse. As the major component of the innate immune system in the brain, aberrant activation of myeloid cells such as macrophages and microglia due to substance use may mediate neuroinflammation and contribute to the development of addiction. All addictive drugs modulate the dopaminergic system and our previous studies have identified dopamine as a pro-inflammatory modulator of macrophage function. However, the mechanism that mediates this effect is currently unknown. Inflammatory activation of macrophages and induction of cytokine production is often mediated by the transcription factor NF-κB, and prior studies have shown that dopamine can modulate NF-κB activity in T-cells and other non-immune cell lines. Here we demonstrated that dopamine can activate NF-κB in primary human macrophages, resulting in the induction of its downstream targets including the NLRP3 inflammasome and the inflammatory cytokine IL-1β. These data also indicate that dopamine primes but does not activate the NLRP3 inflammasome in human macrophages. Activation of NF-κB was required for dopamine-mediated increases in IL-1β, as an inhibitor of NF-κB was able to abrogate the effects of dopamine on production of these cytokines. Connecting an increase in extracellular dopamine to NF-κB activation and inflammation suggests specific intracellular targets that could be used to ameliorate the inflammatory impact of dopamine in neuroinflammatory conditions associated with myeloid cell activation such as addiction. Dopamine exposure primes, but does not activate the NLRP3 inflammasome. Inflammasome priming can be mediated, at least partially, by a dopamine-induced increase in the activation and nuclear translocation of NF-κB in primary human macrophages. Dopamine additively increases the impact of cytomegalovirus on NF-κB activation in macrophages. Dopamine priming increases IL-1β release in response to inflammasome activation.
Collapse
Affiliation(s)
- R A Nolan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - K L Reeb
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - Y Rong
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - S M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - H S Johnson
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - K Runner
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102
| |
Collapse
|
33
|
Kays JS, Yamamoto BK. Evaluation of Microglia/Macrophage Cells from Rat Striatum and Prefrontal Cortex Reveals Differential Expression of Inflammatory-Related mRNA after Methamphetamine. Brain Sci 2019; 9:brainsci9120340. [PMID: 31775383 PMCID: PMC6955783 DOI: 10.3390/brainsci9120340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/19/2022] Open
Abstract
RNA sequencing (RNAseq) can be a powerful tool in the identification of transcriptional changes after drug treatment. RNAseq was utilized to determine expression changes in Fluorescence-activated cell sorted (FACS) CD11b/c+ cells from the striatum (STR) and prefrontal cortex (PFC) of male Sprague-Dawley rats after a methamphetamine (METH) binge dosing regimen. Resident microglia and infiltrating macrophages were collected 2 h or 3 days after drug administration. Gene expression changes indicated there was an increase toward an overall pro-inflammatory state, or M1 polarization, along with what appears to be a subset of cells that differentiated toward the anti-inflammatory M2 polarization. In general, there were significantly more mRNA expression changes in the STR than the PFC and more at 2 h post-binge METH than at 3 days post-binge METH. Additionally, Ingenuity® Pathway Analysis along with details of RNA expression changes revealed cyclo-oxygenase 2 (COX2)-driven prostaglandin (PG) E2 synthesis, glutamine uptake, and the Nuclear factor erythroid2-related factor 2 (NRF2) canonical pathway in microglia were associated with the binge administration regimen of METH.
Collapse
|
34
|
Wang X, Northcutt AL, Cochran TA, Zhang X, Fabisiak TJ, Haas ME, Amat J, Li H, Rice KC, Maier SF, Bachtell RK, Hutchinson MR, Watkins LR. Methamphetamine Activates Toll-Like Receptor 4 to Induce Central Immune Signaling within the Ventral Tegmental Area and Contributes to Extracellular Dopamine Increase in the Nucleus Accumbens Shell. ACS Chem Neurosci 2019; 10:3622-3634. [PMID: 31282647 DOI: 10.1021/acschemneuro.9b00225] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Methamphetamine (METH) is a globally abused, highly addictive stimulant. While investigations of the rewarding and motivational effects of METH have focused on neuronal actions, increasing evidence suggests that METH can also target microglia, the innate immune cells of the central nervous system, causing release of proinflammatory mediators and therefore amplifying the reward changes in the neuronal activity induced by METH. However, how METH induces neuroinflammatory responses within the central nervous system (CNS) is unknown. Herein, we provide direct evidence that METH creates neuroinflammation, at least in part, via the activation of the innate immune Toll-like receptor 4 (TLR4). Biophysical studies revealed that METH bound to MD-2, the key coreceptor of TLR4. Molecular dynamics simulations showed METH binding stabilized the active heterotetramer (TLR4/MD-2)2 conformation. Classic TLR4 antagonists LPS-RS and TAK-242 attenuated METH induced NF-κB activation of microglia, whereas added MD-2 protein boosted METH-induced NF-κB activation. Systemically administered METH (1 mg/kg) was found to specifically up-regulate expression of both CD11b (microglial activation marker) and the proinflammatory cytokine interleukin 6 (IL-6) mRNAs in the ventral tegmental area (VTA), but not in either the nucleus accumbens shell (NAc) or prefrontal cortex (PFC). Systemic administration of a nonopioid, blood-brain barrier permeable TLR4 antagonist (+)-naloxone inhibited METH-induced activation of microglia and IL-6 mRNA overexpression in VTA. METH was found to increase conditioned place preference (CPP) as well as extracellular dopamine concentrations in the NAc, with both effects suppressed by the nonopioid TLR4 antagonist (+)-naloxone. Furthermore, intra-VTA injection of LPS-RS or IL-6 neutralizing antibody suppressed METH-induced elevation of extracellular NAc dopamine. Taken together, this series of studies demonstrate that METH-induced neuroinflammation is, at least in part, mediated by TLR4-IL6 signaling within the VTA, which has the downstream effect of elevating dopamine in the NAc shell. These results provide a novel understanding of the neurobiological mechanisms underlying acute METH reward that includes a critical role for central immune signaling and offers a new target for medication development for treating drug abuse.
Collapse
Affiliation(s)
- Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- Department of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Alexis L. Northcutt
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Thomas A. Cochran
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Xiaozheng Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Timothy J. Fabisiak
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Mackenzie E. Haas
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Jose Amat
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Kenner C. Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland 20892, United States
| | - Steven F. Maier
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | - Ryan K. Bachtell
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| | | | - Linda R. Watkins
- Department of Psychology and Neuroscience and the Center for Neuroscience, University of Colorado at Boulder, Boulder, Colorado 80309, United States
| |
Collapse
|
35
|
Combination of acute intravenous methamphetamine injection and LPS challenge facilitate leukocyte infiltration into the central nervous system of C57BL/6 mice. Int Immunopharmacol 2019; 75:105751. [PMID: 31319359 DOI: 10.1016/j.intimp.2019.105751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022]
Abstract
Methamphetamine (METH) is a stimulant of the central nervous system (CNS) that causes behavioral changes in users. METH is slowly cleared from brain tissue and its chronic use is neurotoxic. METH also alters the cellular and chemical components of inflammation. However, little is known about the effect of a single intravenous dose of METH followed by bacterial lipopolysaccharide (LPS) injection on cellular infiltration and cytokine release in brain tissue. Using a murine model of acute METH administration and flow cytometry, we found that combination of METH and LPS stimulate the infiltration of macrophages (F4/80+cells) and neutrophils (Ly-6G+cells) into the CNS. Histological sections of the brainstem of METH-treated and LPS-challenged C57BL/6 mice demonstrated considerable leukocyte infiltration relative to untreated, LPS, and METH groups. Moreover, rodents treated with LPS alone or combined with METH showed elevated levels of pro-inflammatory cytokines mRNA in brain tissue. Our observations are important because recognizing neuroinflammatory changes after acute METH administration might help us to understand METH-induced neurotoxicity in users.
Collapse
|
36
|
Sepehrian A, Shakeri N, Abednatanzi H, Soheili S. The Effect of Methylphenidate and Aerobic Exercise on Renal Function of Male Rats. INTERNATIONAL JOURNAL OF BASIC SCIENCE IN MEDICINE 2019. [DOI: 10.15171/ijbsm.2019.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introduction: Inadequate awareness and knowledge exists regarding the effects of stimulant drugs on renal health among athletes. The present study aimed to determine the effects of methylphenidate, as a stimulant drug, and aerobic exercise on renal function in rats. Materials and Methods: Eighty male rats were randomly divided into 8 groups (n=10 per group) including control (Co), aerobic exercise sham (AE Sh), drug sham (D Sh), aerobic exercise (AE), the effective dose of drug (ED, 10 mg/kg), 3 times of effective dose (TED, 30 mg/kg), aerobic exercise-effective dose (AE-ED), and aerobic exercise-three times of effective dose (AE-TED). The drug was orally administrated to the animals, and then they were placed on a rat treadmill after 30 minutes. The physical activity (25 m/min) was performed 30 minutes a day, 3 days a week for two months. Twenty-four hours after the last session of AE, blood samples were taken from the rats and serum creatinine (Cr) and blood urea nitrogen (BUN) were determined. Results: The results showed that serum Cr and BUN levels were not significantly different in the exercise group compared to the control groups (i.e., Co, AE Sham, and D Sham). However, serum BUN and Cr significantly increased in the AE-ED and AE-TED groups compared to the AE group (PCr=0.001 and PBUN=0.001). Conclusion: In general, significant increases in the serum BUN and Cr levels in rats received methylphenidate indicated decreased renal function in these animals.
Collapse
Affiliation(s)
- Ali Sepehrian
- Ph.D. Student of Department of Physical Education and Sport Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nader Shakeri
- Department of Physical Education and Sport Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hosein Abednatanzi
- Department of Physical Education and Sport Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shahram Soheili
- Department of Physical Education and Sport Sciences, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
37
|
Kohno M, Link J, Dennis LE, McCready H, Huckans M, Hoffman WF, Loftis JM. Neuroinflammation in addiction: A review of neuroimaging studies and potential immunotherapies. Pharmacol Biochem Behav 2019; 179:34-42. [PMID: 30695700 DOI: 10.1016/j.pbb.2019.01.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 01/22/2019] [Accepted: 01/25/2019] [Indexed: 12/29/2022]
Abstract
Addiction is a worldwide public health problem and this article reviews scientific advances in identifying the role of neuroinflammation in the genesis, maintenance, and treatment of substance use disorders. With an emphasis on neuroimaging techniques, this review examines human studies of addiction using positron emission tomography to identify binding of translocator protein (TSPO), which is upregulated in reactive glial cells and activated microglia during pathological states. High TSPO levels have been shown in methamphetamine use but exhibits variable patterns in cocaine use. Alcohol and nicotine use, however, are associated with lower TSPO levels. We discuss how mechanistic differences at the neurotransmitter and circuit level in the neural effects of these agents and subsequent immune response may explain these observations. Finally, we review the potential of anti-inflammatory drugs, including ibudilast, minocycline, and pioglitazone, to ameliorate the behavioral and cognitive consequences of addiction.
Collapse
Affiliation(s)
- Milky Kohno
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Jeanne Link
- Center for Radiochemistry Research, Knight Cardiovascular Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA
| | - Laura E Dennis
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Holly McCready
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Marilyn Huckans
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Mental Health and Clinical Neurosciences Division, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - William F Hoffman
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Mental Health and Clinical Neurosciences Division, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Jennifer M Loftis
- Research & Development Service, Veterans Affairs Portland Health Care System, 3710 SW US Veterans Hospital Road, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University and Veterans Affairs Portland Health Care System, Portland, OR, USA.
| |
Collapse
|
38
|
Mitchell CM, El Jordi O, Yamamoto BK. Inflammatory mechanisms of abused drugs. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
39
|
Papageorgiou M, Raza A, Fraser S, Nurgali K, Apostolopoulos V. Methamphetamine and its immune-modulating effects. Maturitas 2018; 121:13-21. [PMID: 30704560 DOI: 10.1016/j.maturitas.2018.12.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022]
Abstract
The recreational use of methamphetamine (METH, or ice) is a global burden. It pervades and plagues contemporary society; it has been estimated that there are up to 35 million users worldwide. METH is a highly addictive psychotropic compound which acts on the central nervous system, and chronic use can induce psychotic behavior. METH has the capacity to modulate immune cells, giving the drug long-term effects which may manifest as neuropsychiatric disorders, and that increase susceptibility to communicable diseases, such as HIV. In addition, changes to the cytokine balance have been associated with compromise of the blood-brain barrier, resulting to alterations to brain plasticity, creating lasting neurotoxicity. Immune-related signaling pathways are key to further evaluating how METH impacts host immunity through these neurological and peripheral modifications. Combining this knowledge with current data on inflammatory responses will improve understanding of how the adaptive and innate immunity responds to METH, how this can activate premature-ageing processes and how METH exacerbates disturbances that lead to non-communicable age-related diseases, including cardiovascular disease, stroke, depression and dementia.
Collapse
Affiliation(s)
- Marco Papageorgiou
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Ali Raza
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Sarah Fraser
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Regenerative Medicine and StemCells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia.
| | | |
Collapse
|
40
|
Ghanbari F, Khaksari M, Vaezi G, Hojati V, Shiravi A. Hydrogen Sulfide Protects Hippocampal Neurons Against Methamphetamine Neurotoxicity Via Inhibition of Apoptosis and Neuroinflammation. J Mol Neurosci 2018; 67:133-141. [DOI: 10.1007/s12031-018-1218-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/11/2018] [Indexed: 12/28/2022]
|
41
|
Linker KE, Cross SJ, Leslie FM. Glial mechanisms underlying substance use disorders. Eur J Neurosci 2018; 50:2574-2589. [PMID: 30240518 DOI: 10.1111/ejn.14163] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 12/28/2022]
Abstract
Addiction is a devastating disorder that produces persistent maladaptive changes to the central nervous system, including glial cells. Although there is an extensive body of literature examining the neuronal mechanisms of substance use disorders, effective therapies remain elusive. Glia, particularly microglia and astrocytes, have an emerging and meaningful role in a variety of processes beyond inflammation and immune surveillance, and may represent a promising therapeutic target. Indeed, glia actively modulate neurotransmission, synaptic connectivity and neural circuit function, and are critically poised to contribute to addictive-like brain states and behaviors. In this review, we argue that glia influence the cellular, molecular, and synaptic changes that occur in neurons following drug exposure, and that this cellular relationship is critically modified following drug exposure. We discuss direct actions of abused drugs on glial function through immune receptors, such as Toll-like receptor 4, as well as other mechanisms. We highlight how drugs of abuse affect glia-neural communication, and the profound effects that glial-derived factors have on neuronal excitability, structure, and function. Recent research demonstrates that glia have brain region-specific functions, and glia in different brain regions have distinct contributions to drug-associated behaviors. We will also evaluate the evidence demonstrating that glial activation is essential for drug reward and drug-induced dopamine release, and highlight clinical evidence showing that glial mechanisms contribute to drug abuse liability. In this review, we synthesize the extensive evidence that glia have a unique, pivotal, and underappreciated role in the development and maintenance of addiction.
Collapse
Affiliation(s)
- K E Linker
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - S J Cross
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - F M Leslie
- Department of Pharmacology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
42
|
Shafahi M, Vaezi G, Shajiee H, Sharafi S, Khaksari M. Crocin Inhibits Apoptosis and Astrogliosis of Hippocampus Neurons Against Methamphetamine Neurotoxicity via Antioxidant and Anti-inflammatory Mechanisms. Neurochem Res 2018; 43:2252-2259. [PMID: 30259275 DOI: 10.1007/s11064-018-2644-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/11/2018] [Accepted: 09/19/2018] [Indexed: 01/19/2023]
Abstract
Methamphetamine (METH) is a stimulant drug, which can cause neurotoxicity and increase the risk of neurodegenerative disorders. The mechanisms of acute METH intoxication comprise intra-neuronal events including oxidative stress, dopamine oxidation, and excitotoxicity. According to recent studies, crocin protects neurons by functioning as an anti-oxidant, anti-inflammatory, and anti-apoptotic compound. Accordingly, this study aimed to determine if crocin can protect against METH-induced neurotoxicity. Seventy-two male Wistar rats that weighed 260-300 g were randomly allocated to six groups of control (n = 12), crocin 90 mg/kg group (n = 12), METH (n = 12), METH + crocin 30 mg/kg (n = 12), METH + crocin 60 mg/kg (n = 12), and METH + crocin 90 mg/kg (n = 12). METH neurotoxicity was induced by 40 mg/kg of METH in four injections (e.g., 4 × 10 mg/kg q. 2 h, IP). Crocin was intraperitoneally (IP) injected at 30 min, 24 h, and 48 h after the final injection of METH. Seven days after METH injection, the rats' brains were removed for biochemical assessment using the ELISA technique, and immunohistochemistry staining was used for caspase-3 and glial fibrillary acidic protein (GFAP) detection. Crocin treatment could significantly increase superoxide dismutase (P < 0.05) and glutathione (P < 0.01) levels and reduce malondialdehyde and TNF-α in comparison with the METH group (P < 0.05). Moreover, crocin could significantly decline the level of caspase-3 and GFAP-positive cells in the CA1 region (P < 0.01). According to the results, crocin exerts neuroprotective effects on METH neurotoxicity via the inhibition of apoptosis and neuroinflammation.
Collapse
Affiliation(s)
- Monire Shafahi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Golamhassan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Hooman Shajiee
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Shahram Sharafi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Mehdi Khaksari
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| |
Collapse
|
43
|
Yang X, Wang Y, Li Q, Zhong Y, Chen L, Du Y, He J, Liao L, Xiong K, Yi CX, Yan J. The Main Molecular Mechanisms Underlying Methamphetamine- Induced Neurotoxicity and Implications for Pharmacological Treatment. Front Mol Neurosci 2018; 11:186. [PMID: 29915529 PMCID: PMC5994595 DOI: 10.3389/fnmol.2018.00186] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/14/2018] [Indexed: 01/07/2023] Open
Abstract
Methamphetamine (METH) is a popular new-type psychostimulant drug with complicated neurotoxicity. In spite of mounting evidence on METH-induced damage of neural cell, the accurate mechanism of toxic effect of the drug on central nervous system (CNS) has not yet been completely deciphered. Besides, effective treatment strategies toward METH neurotoxicity remain scarce and more efficacious drugs are to be developed. In this review, we summarize cellular and molecular bases that might contribute to METH-elicited neurotoxicity, which mainly include oxidative stress, excitotoxicity, and neuroinflammation. We also discuss some drugs that protect neural cells suffering from METH-induced neurotoxic consequences. We hope more in-depth investigations of exact details that how METH produces toxicity in CNS could be carried out in future and the development of new drugs as natural compounds and immunotherapies, including clinic trials, are expected.
Collapse
Affiliation(s)
- Xue Yang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yong Wang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Qiyan Li
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yaxian Zhong
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Liangpei Chen
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yajun Du
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Jing He
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Lvshuang Liao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Chun-xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
44
|
The relationship between interleukin-6 and functional connectivity in methamphetamine users. Neurosci Lett 2018; 677:49-54. [PMID: 29689344 DOI: 10.1016/j.neulet.2018.04.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 03/23/2018] [Accepted: 04/19/2018] [Indexed: 01/09/2023]
Abstract
Methamphetamine (MA) causes an increase in pro-inflammatory cytokines in animal models and in humans. Resulting activation of microglia and neuro-inflammation could, via effects on reward networks, mediate behavioral characteristics of addiction. We examined the relationship between interleukin-6 (IL-6) and corticolimbic and striatolimbic resting-state functional connectivity (RSFC). Thirty adults diagnosed with MA dependence and 20 control subjects underwent a resting-state functional magnetic resonance imaging (fMRI) scan and gave a blood sample for determination of plasma IL-6 levels. Seed-based RSFC analyses were performed to examine the interactive effect of group and IL-6 on ventral striatal and prefrontal connectivity. Within the MA group, IL-6 levels were positively related to striatolimbic RSFC but negatively related to corticostriatal RSFC. Our findings with IL-6 support the idea that inflammation may at least partly mediate the link among MA use disorder, RSFC, and behavior, possibly via effects on mesolimbic and mesocortical dopaminergic systems.
Collapse
|
45
|
Tay TL, Béchade C, D'Andrea I, St-Pierre MK, Henry MS, Roumier A, Tremblay ME. Microglia Gone Rogue: Impacts on Psychiatric Disorders across the Lifespan. Front Mol Neurosci 2018; 10:421. [PMID: 29354029 PMCID: PMC5758507 DOI: 10.3389/fnmol.2017.00421] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022] Open
Abstract
Microglia are the predominant immune response cells and professional phagocytes of the central nervous system (CNS) that have been shown to be important for brain development and homeostasis. These cells present a broad spectrum of phenotypes across stages of the lifespan and especially in CNS diseases. Their prevalence in all neurological pathologies makes it pertinent to reexamine their distinct roles during steady-state and disease conditions. A major question in the field is determining whether the clustering and phenotypical transformation of microglial cells are leading causes of pathogenesis, or potentially neuroprotective responses to the onset of disease. The recent explosive growth in our understanding of the origin and homeostasis of microglia, uncovering their roles in shaping of the neural circuitry and synaptic plasticity, allows us to discuss their emerging functions in the contexts of cognitive control and psychiatric disorders. The distinct mesodermal origin and genetic signature of microglia in contrast to other neuroglial cells also make them an interesting target for the development of therapeutics. Here, we review the physiological roles of microglia, their contribution to the effects of environmental risk factors (e.g., maternal infection, early-life stress, dietary imbalance), and their impact on psychiatric disorders initiated during development (e.g., Nasu-Hakola disease (NHD), hereditary diffuse leukoencephaly with spheroids, Rett syndrome, autism spectrum disorders (ASDs), and obsessive-compulsive disorder (OCD)) or adulthood (e.g., alcohol and drug abuse, major depressive disorder (MDD), bipolar disorder (BD), schizophrenia, eating disorders and sleep disorders). Furthermore, we discuss the changes in microglial functions in the context of cognitive aging, and review their implication in neurodegenerative diseases of the aged adult (e.g., Alzheimer’s and Parkinson’s). Taking into account the recent identification of microglia-specific markers, and the availability of compounds that target these cells selectively in vivo, we consider the prospect of disease intervention via the microglial route.
Collapse
Affiliation(s)
- Tuan Leng Tay
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Catherine Béchade
- INSERM UMR-S 839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Ivana D'Andrea
- INSERM UMR-S 839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris, France.,Institut du Fer à Moulin, Paris, France
| | | | - Mathilde S Henry
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Anne Roumier
- INSERM UMR-S 839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Marie-Eve Tremblay
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada.,Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
| |
Collapse
|
46
|
Persons AL, Bradaric BD, Dodiya HB, Ohene-Nyako M, Forsyth CB, Keshavarzian A, Shaikh M, Napier TC. Colon dysregulation in methamphetamine self-administering HIV-1 transgenic rats. PLoS One 2018; 13:e0190078. [PMID: 29293553 PMCID: PMC5749763 DOI: 10.1371/journal.pone.0190078] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023] Open
Abstract
The integrity and function of the gut is impaired in HIV-infected individuals, and gut pathogenesis may play a role in several HIV-associated disorders. Methamphetamine is a popular illicit drug abused by HIV-infected individuals. However, the effect of methamphetamine on the gut and its potential to exacerbate HIV-associated gut pathology is not known. To shed light on this scenario, we evaluated colon barrier pathology in a rat model of the human comorbid condition. Intestinal barrier integrity and permeability were assessed in drug-naïve Fischer 344 HIV-1 transgenic (Tg) and non-Tg rats, and in Tg and non-Tg rats instrumented with jugular cannulae trained to self-administer methamphetamine or serving as saline-yoked controls. Intestinal permeability was determined by measuring the urine content of orally gavaged sugars. Intestinal barrier integrity was evaluated by immunoblotting or immunofluorescence of colon claudin-1 and zonula occludens-1 (ZO-1), two major tight junction proteins that regulate gut epithelial paracellular permeability. Both non-Tg and Tg rats self-administered moderate amounts of methamphetamine. These amounts were sufficient to increase colon permeability, reduce protein level of claudin-1, and reduce claudin-1 and ZO-1 immunofluorescence in Tg rats relative to non-Tg rats. Methamphetamine decreased tight junction immunofluorescence in non-Tg rats, with a similar, but non-significant trend observed in Tg rats. However, the effect of methamphetamine on tight junction proteins was subthreshold to gut leakiness. These findings reveal that both HIV-1 proteins and methamphetamine alter colon barrier integrity, and indicate that the gut may be a pathogenic site for these insults.
Collapse
Affiliation(s)
- Amanda L. Persons
- Department of Psychiatry, Rush University Medical Center, Chicago, IL, United States of America
- Department of Physician Assistant Studies, Rush University Medical Center, Chicago, IL, United States of America
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- * E-mail:
| | - Brinda D. Bradaric
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Health Sciences, Rush University Medical Center, Chicago, IL, United States of America
| | - Hemraj B. Dodiya
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, United States of America
| | - Michael Ohene-Nyako
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, United States of America
| | - Christopher B. Forsyth
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States of America
| | - Ali Keshavarzian
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States of America
| | - Maliha Shaikh
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States of America
| | - T. Celeste Napier
- Department of Psychiatry, Rush University Medical Center, Chicago, IL, United States of America
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, United States of America
| |
Collapse
|
47
|
Intranasal insulin treatment alleviates methamphetamine induced anxiety-like behavior and neuroinflammation. Neurosci Lett 2017; 660:122-129. [DOI: 10.1016/j.neulet.2017.09.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 01/03/2023]
|
48
|
Bachtell RK, Jones JD, Heinzerling KG, Beardsley PM, Comer SD. Glial and neuroinflammatory targets for treating substance use disorders. Drug Alcohol Depend 2017; 180:156-170. [PMID: 28892721 PMCID: PMC5790191 DOI: 10.1016/j.drugalcdep.2017.08.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/28/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND The plenary session at the 2016 Behavior, Biology and Chemistry: Translational Research in Addiction Conference focused on glia as potential players in the development, persistence and treatment of substance use disorders. Glia partake in various functions that are important for healthy brain activity. Drugs of abuse alter glial cell activity producing several perturbations in brain function that are thought to contribute to behavioral changes associated with substance use disorders. Consequently, drug-induced changes in glia-driven processes in the brain represent potential targets for pharmacotherapeutics treating substance use disorders. METHODS Four speakers presented preclinical and clinical research illustrating the effects that glial modulators have on abuse-related behavioral effects of psychostimulants and opioids. This review highlights some of these findings and expands its focus to include other research focused on drug-induced glia abnormalities and glia-focused treatment approaches in substance use disorders. RESULTS Preclinical findings show that drugs of abuse induce neuroinflammatory signals and disrupt glutamate homeostasis through their interaction with microglia and astrocytes. Preclinical and clinical studies testing the effects of glial modulators show general effectiveness in reducing behaviors associated with substance use disorders. CONCLUSIONS The contribution of drug-induced glial activity continues to emerge as an intriguing target for substance use disorder treatments. Clinical investigations of glial modulators have yielded promising results on substance use measures and indicate that they are generally safe and well-tolerated. However, results have not been entirely positive and more questions remain for continued exploration in the development and testing of glial-directed treatments for substance use disorders.
Collapse
Affiliation(s)
- Ryan K. Bachtell
- Department of Psychology and Neuroscience, and Center for Neuroscience, UCB 345, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Jermaine D. Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute and College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Keith G. Heinzerling
- Department of Family Medicine and Center for Behavioral and Addiction Medicine, UCLA, Los Angeles, CA, USA
| | - Patrick M. Beardsley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, VA 23298, USA
| | - Sandra D. Comer
- Division on Substance Use Disorders, New York State Psychiatric Institute and College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
49
|
Zhang A, Wu L, Chen Z, Huang G, Lu X. Methamphetamine Causes Photoreceptor Cell Damage Through Promoting Polarization of Macrophages and Inducing Inflammatory Response. Int J Toxicol 2017; 36:403-409. [PMID: 28731367 DOI: 10.1177/1091581817718473] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Classically activated macrophages (M1) are proinflammatory effectors and closely related to the progression of neurotoxicity. As a powerful psychostimulant and addictive drug, methamphetamine (Meth) abuse could result in long-lasting abnormalities in retina. This study investigated the effect of Meth at nontoxic concentration on macrophage activation state and its resultant toxicity to photoreceptor cells. Results showed that cytotoxicity was caused by Meth on 661 W cells after coculturing with RAW264.7 macrophage. RAW264.7 cells tended to switch to the M1 phenotype, releasing more proinflammatory cytokines after treatment with Meth. Meth could also upregulate the M1-related gene and protein expression. Our study demonstrated that Meth promoted macrophage polarization from M0 to M1 and induced inflammatory response, providing the scientific rationale for the photoreceptor cell damage caused by the Meth abuse.
Collapse
Affiliation(s)
- Aihui Zhang
- 1 Department of Ophthalmology, Liaocheng Brain Hospital affiliated to Liaocheng People's Hospital, Liaocheng, People's Republic of China
| | - Laiwei Wu
- 2 Southern Medical University, Guangzhou, People's Republic of China
- 3 Department of Ophthalmology, Huizhou Municipal Central Hospital, Huizhou, People's Republic of China
| | - Zilin Chen
- 3 Department of Ophthalmology, Huizhou Municipal Central Hospital, Huizhou, People's Republic of China
| | - Guoqiang Huang
- 4 Department of Ophthalmology, Meizhou People's Hospital, Meizhou, People's Republic of China
| | - Xiaohe Lu
- 5 Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
50
|
Ginkgolide B Suppresses Methamphetamine-Induced Microglial Activation Through TLR4-NF-κB Signaling Pathway in BV2 Cells. Neurochem Res 2017; 42:2881-2891. [DOI: 10.1007/s11064-017-2309-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/14/2017] [Accepted: 05/22/2017] [Indexed: 01/20/2023]
|