1
|
Eshrati S, Nikbakhtzadeh M, Arezoomandan R, Fattahi A. Efficacy of minocycline in substance use disorder: A systematic review of preclinical and clinical studies. Pharmacol Biochem Behav 2025; 250:173982. [PMID: 39993507 DOI: 10.1016/j.pbb.2025.173982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/30/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Addiction is a serious condition that leads to negative changes in the central nervous system. Although there have been significant advancements in medication treatments for substance use disorders (SUDs), it is clear that there is a need to implement these developments in clinical settings to explore new therapeutic approaches for helping individuals with SUDs. Minocycline, a semi-synthetic second-generation tetracycline, possesses neuroprotective and anti-inflammatory properties. Recent studies have shown promising results when using this drug for the treatment of substance misuse. This study aimed to review the pre-clinical and clinical studies assessing the therapeutic efficacy of minocycline on drug-related outcomes, including reward, tolerance, withdrawal, impairments, and toxicity. We conducted a systematic review to assess the effectiveness of minocycline in ameliorating drug-induced outcomes per the PRISMA guidelines. Electronic medical databases Web of Science, PubMed/Medline, Scopus, and Google Scholar were searched from databases from their inception date until December 2023. 56 of the 623 articles met the eligible criteria for analysis. Of the 56 articles reviewed, 51 were conducted on animals, while 5 involved human subjects. Our study indicates that the majority of animal studies have primarily focused on morphine and alcohol, with no research found to date on the effects of cannabis. This review highlights minocycline's potential in addiction treatment through its effects on anti-inflammatory mechanisms, neuroprotection, regulation of synaptic plasticity. Results of this study suggest that although minocycline shows promise in experiments, its effectiveness in humans may be limited by dosage, individual variability, and addiction's complexity. Further clinical studies are required to clarify the optimal dose, duration of administration, and delivery route and focus on identifying specific conditions where it may be most effective.
Collapse
Affiliation(s)
- Sahar Eshrati
- Addiction Department, School of Behavioral Sciences and Mental Health (Tehran Institute of Psychiatry), Iran University of Medical Sciences, Tehran, Iran
| | - Marjan Nikbakhtzadeh
- Department of Physiology, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Reza Arezoomandan
- Addiction Department, School of Behavioral Sciences and Mental Health (Tehran Institute of Psychiatry), Iran University of Medical Sciences, Tehran, Iran; School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Azin Fattahi
- Addiction Department, School of Behavioral Sciences and Mental Health (Tehran Institute of Psychiatry), Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Mabry SJ, Cao X, Zhu Y, Rowe C, Patel S, González-Arancibia C, Romanazzi T, Saleeby DP, Elam A, Lee HT, Turkmen S, Lauzon SN, Hernandez CE, Sun H, Wu H, Carter AM, Galli A. Fusobacterium nucleatum determines the expression of amphetamine-induced behavioral responses through an epigenetic phenomenon. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633210. [PMID: 39868090 PMCID: PMC11761806 DOI: 10.1101/2025.01.15.633210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Amphetamines (AMPHs) are psychostimulants commonly used for the treatment of neuropsychiatric disorders. They are also misused (AMPH use disorder; AUD), with devastating outcomes. Recent studies have implicated dysbiosis in the pathogenesis of AUD. However, the mechanistic roles of microbes in AUD are unknown. Fusobacterium nucleatum (Fn) is a bacterium that increases in abundance in both rats and humans upon AMPH exposure. Fn releases short-chain fatty acids (SCFAs), bacterial byproducts thought to play a fundamental role in the gut-brain axis as well as the pathogenesis of AUD. We demonstrate that in gnotobiotic Drosophila melanogaster, colonization with Fn or dietary supplementation of the SCFA butyrate, a potent inhibitor of histone deacetylases (HDACs), enhances the psychomotor and rewarding properties of AMPH as well as its ability to promote male sexual motivation. Furthermore, solely HDAC1 RNAi targeted inhibition recapitulates these enhancements, pointing to a specific process underlying this Fn phenomenon. Of note is that the expression of these AMPH behaviors is determined by the increase in extracellular dopamine (DA) levels that result from AMPH-induced reversal of DA transporter (DAT) function, termed non-vesicular DA release (NVDR). The magnitude of AMPH-induced NVDR is dictated, at least in part, by DAT expression levels. Consistent with our behavioral data, we show that Fn, butyrate, and HDAC1 inhibition enhance NVDR by elevating DAT expression. Thus, the participation of Fn in AUD stems from its ability to release butyrate and inhibit HDAC1. These data offer a microbial target and probiotic interventions for AUD treatment.
Collapse
Affiliation(s)
- Samuel J Mabry
- University of Alabama Birmingham, Department of Surgery, Birmingham, Alabama
| | - Xixi Cao
- Oregon Health & Science University, School of Dentistry, Portland, Oregon
| | - Yanqi Zhu
- University of Alabama Birmingham, Department of Surgery, Birmingham, Alabama
| | - Caleb Rowe
- University of Alabama Birmingham, Department of Surgery, Birmingham, Alabama
| | - Shalin Patel
- University of Alabama Birmingham, Department of Surgery, Birmingham, Alabama
| | | | - Tiziana Romanazzi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - David P Saleeby
- University of Alabama Birmingham, Department of Surgery, Birmingham, Alabama
| | - Anna Elam
- University of Alabama Birmingham, Department of Psychiatry, Birmingham, Alabama
| | - Hui-Ting Lee
- University of Alabama Birmingham, Department of Chemistry, Birmingham, Alabama
| | - Serhat Turkmen
- Howard Hughes Medical Institute, University of Alabama Birmingham, Department of Cell, Developmental, and Integrative Biology, Birmingham, Alabama
| | - Shelby N Lauzon
- Howard Hughes Medical Institute, University of Alabama Birmingham, Department of Cell, Developmental, and Integrative Biology, Birmingham, Alabama
| | - Cesar E Hernandez
- University of Alabama Birmingham, Department of Surgery, Birmingham, Alabama
| | - HaoSheng Sun
- Howard Hughes Medical Institute, University of Alabama Birmingham, Department of Cell, Developmental, and Integrative Biology, Birmingham, Alabama
| | - Hui Wu
- Oregon Health & Science University, School of Dentistry, Portland, Oregon
| | - Angela M Carter
- University of Alabama Birmingham, Department of Surgery, Birmingham, Alabama
- University of Alabama Birmingham, Center for Inter-systemic Networks and Enteric Medical Advances (CINEMA), Birmingham, Alabama
| | - Aurelio Galli
- University of Alabama Birmingham, Department of Surgery, Birmingham, Alabama
- University of Alabama Birmingham, Center for Inter-systemic Networks and Enteric Medical Advances (CINEMA), Birmingham, Alabama
| |
Collapse
|
3
|
Xie L, Zhuang Z, Guo B, Huang Y, Shi X, Huang Z, Xu Z, Chen Y, Cao Y, Zheng Y, Wu R, Ma S. Ketamine induced gut microbiota dysbiosis and barrier and hippocampal dysfunction in rats. iScience 2024; 27:111089. [PMID: 39493883 PMCID: PMC11530865 DOI: 10.1016/j.isci.2024.111089] [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: 05/12/2024] [Revised: 08/02/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
The microbiota-gut-brain axis (MGBA) plays a pivotal role in drug addiction. However, the pathophysiological mechanism of MGBA in ketamine addiction remains elusive. The present study investigated the ketamine-induced gut microbiota disorders, intestinal barrier dysfunction, and the alterations in brain function, using a conditioned place preference (CPP) model of ketamine addiction in rats. Compared with the control group, ketamine induced decreased amplitude of low-frequency fluctuation (ALFF) values in the hippocampus, and pyknotic nuclei and concentrated cytoplasm in hippocampal neurons, as well as alterations in gut microbiota composition, shortened ileum villi, and thinner colonic mucosa. We also found that the abundance of gut microbiota exhibited correlations with CPP score, hippocampal ALFF value, length of ileum villi, and thickness of colonic mucosa. Our findings provide evidence for abnormal alterations in the MGBA of ketamine-addicted rats, which improves our understating of the mechanism of ketamine addiction and the potential for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Lei Xie
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Zelin Zhuang
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Baowen Guo
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yuehua Huang
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Xiaoyan Shi
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Zikai Huang
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Ziquan Xu
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yanbin Chen
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yuyin Cao
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yanmin Zheng
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Renhua Wu
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, China
| | - Shuhua Ma
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
4
|
Mai Y, Cheng Z, Wang Z, Hu T, Zhang Y, Yuan X, Xu X, Fan Y, Ge F, Shi P, Wang J, Yang X, Guan X. Pathological polarizations from microglia to astrocyte contributes to spatial memory deficit in methamphetamine abstinence mice. Cereb Cortex 2024; 34:bhae281. [PMID: 38981852 DOI: 10.1093/cercor/bhae281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/17/2024] [Accepted: 06/22/2024] [Indexed: 07/11/2024] Open
Abstract
Previously, we found that dCA1 A1-like polarization of astrocytes contributes a lot to the spatial memory deficit in methamphetamine abstinence mice. However, the underlying mechanism remains unclear, resulting in a lack of promising therapeutic targets. Here, we found that methamphetamine abstinence mice exhibited an increased M1-like microglia and A1-like astrocytes, together with elevated levels of interleukin 1α and tumor necrosis factor α in dCA1. In vitro, the M1-like BV2 microglia cell medium, containing high levels of Interleukin 1α and tumor necrosis factor α, elevated A1-like polarization of astrocytes, which weakened their capacity for glutamate clearance. Locally suppressing dCA1 M1-like microglia activation with minocycline administration attenuated A1-like polarization of astrocytes, ameliorated dCA1 neurotoxicity, and, most importantly, rescued spatial memory in methamphetamine abstinence mice. The effective time window of minocycline treatment on spatial memory is the methamphetamine exposure period, rather than the long-term methamphetamine abstinence.
Collapse
Affiliation(s)
- Yuning Mai
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Zhen Cheng
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Ze Wang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Tao Hu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Yuanyuan Zhang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Xiya Yuan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Xing Xu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Yu Fan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Feifei Ge
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Pengbo Shi
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Jun Wang
- Department of Toxicology, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China
| | - Xin Yang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Xiaowei Guan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| |
Collapse
|
5
|
da Silva MCM, de Souza Ferreira LP, Giustina AD. Could immunotherapy be a hope for addiction treatment? Clinics (Sao Paulo) 2024; 79:100347. [PMID: 38583393 PMCID: PMC11002847 DOI: 10.1016/j.clinsp.2024.100347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/26/2024] [Accepted: 03/10/2024] [Indexed: 04/09/2024] Open
Affiliation(s)
| | - Luiz Philipe de Souza Ferreira
- Department of Morphology and Genetics, Structural and Functional Biology Graduate Program, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Amanda Della Giustina
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| |
Collapse
|
6
|
Grodin EN. Neuroimmune modulators as novel pharmacotherapies for substance use disorders. Brain Behav Immun Health 2024; 36:100744. [PMID: 38435721 PMCID: PMC10906159 DOI: 10.1016/j.bbih.2024.100744] [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: 09/20/2023] [Revised: 12/20/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
One promising avenue of research is the use of neuroimmune modulators to treat substance use disorders (SUDs). Neuroimmune modulators target the interactions between the nervous system and immune system, which have been found to play a crucial role in the development and maintenance of SUDs. Multiple classes of substances produce alterations to neuroimmune signaling and peripheral immune function, including alcohol, opioids, and psychostimulants Preclinical studies have shown that neuroimmune modulators can reduce drug-seeking behavior and prevent relapse in animal models of SUDs. Additionally, early-phase clinical trials have demonstrated the safety and feasibility of using neuroimmune modulators as a treatment for SUDs in humans. These therapeutics can be used as stand-alone treatments or as adjunctive. This review summarizes the current state of the field and provides future directions with a specific focus on personalized medicine.
Collapse
Affiliation(s)
- Erica N. Grodin
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
- Cousins Center for Psychoneuroimmunology, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
7
|
Kusui Y, Izuo N, Tokuhara R, Asano T, Nitta A. Neuronal activation of nucleus accumbens by local methamphetamine administration induces cognitive impairment through microglial inflammation in mice. J Pharmacol Sci 2024; 154:127-138. [PMID: 38395513 DOI: 10.1016/j.jphs.2023.12.003] [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: 09/04/2023] [Revised: 11/27/2023] [Accepted: 12/07/2023] [Indexed: 02/25/2024] Open
Abstract
More than half of methamphetamine (METH) users present with cognitive impairment, making it difficult for them to reintegrate into society. However, the mechanisms of METH-induced cognitive impairment remain unclear. METH causes neuronal hyperactivation in the nucleus accumbens (NAc) by aberrantly releasing dopamine, which triggers dependence. In this study, to clarify the involvement of hyperactivation of NAc in METH-induced cognitive impairment, mice were locally microinjected with METH into NAc (mice with METH (NAc)) and investigated their cognitive phenotype. Mice with METH (NAc) exhibited cognitive dysfunction in behavioral analyses and decreased long-term potentiation in the hippocampus, with NAc activation confirmed by expression of FosB, a neuronal activity marker. In the hippocampus of mice with METH (NAc), activated microglia, but not astroglia, and upregulated microglia-related genes, Il1b and C1qa were observed. Finally, administration of minocycline, a tetracycline antibiotic with suppressive effect on microglial activation, to mice with METH (NAc) ameliorated cognitive impairment and synaptic dysfunction by suppressing the increased expression of Il1b and C1qa in the hippocampus. In conclusion, activation of NAc by injection of METH into NAc elicited cognitive impairment by facilitating immune activation in mice. This study suggests that immunological intervention could be a therapeutic strategy for addiction-related cognitive disturbances.
Collapse
Affiliation(s)
- Yuka Kusui
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Naotaka Izuo
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
| | - Reika Tokuhara
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Takashi Asano
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Atsumi Nitta
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
| |
Collapse
|
8
|
Lai M, Fu D, Li X, Zhuang D, Wang M, Xu Z, Liu H, Shen H, Xu P, Zhou W. N-Isopropylbenzylamine-induced conditioned place preference, sensitization behaviour and self-administration in rodents. Addict Biol 2024; 29:e13370. [PMID: 38353028 PMCID: PMC10898833 DOI: 10.1111/adb.13370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 02/16/2024]
Abstract
N-Isopropylbenzylamine (N-ipb), a chain isomer of methamphetamine (METH) with similar physical properties, has been used as a substitute for METH in seized drug samples. However, the abuse potential of N-ipb remains unclear. Therefore, this study aimed to evaluate the abuse potential of N-ipb in comparison to METH, by using conditioned place preference (CPP), locomotor sensitization and intravenous self-administration tests. The results showed that N-ipb at a dose of 3 mg·kg-1 significantly induced CPP in mice, which was comparable to the effect of METH at 1 mg·kg-1 . Either acute or repeated N-ipb injections (1 or 3 mg·kg-1 ) failed to raise the locomotor activity. However, acute treatment with 10 mg·kg-1 N-ipb elevated the locomotor activity compared with saline, while chronic injection of 10 mg·kg-1 N-ipb induced a delayed and attenuated sensitization compared with 1 mg·kg-1 METH. Rats could acquire N-ipb self-administration at a dose of 1 mg·kg-1 ·infusion-1 , and a typical inverted U-shaped dose-response curve was obtained for N-ipb. The mean dose of N-ipb that maintained the maximum response was greater than that of METH, indicating that N-ipb is less potent for reinforcement than METH. In the economic behavioural analysis, comparison of essential values derived from the demand elasticity revealed that N-ipb is less efficacy as a reinforcer than METH. The present data demonstrate that N-ipb functions as a reinforcer and has a potential for abuse. However, the potency of psychomotor stimulation and the reinforcing effectiveness of N-ipb are lower than those of METH.
Collapse
Affiliation(s)
- Miaojun Lai
- Department of PsychiatryAffiliated Kangning Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Addiction Research of Zhejiang ProvinceNingboChina
| | - Dan Fu
- Department of PsychiatryAffiliated Kangning Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Addiction Research of Zhejiang ProvinceNingboChina
| | - Xiangyu Li
- Office of China National Narcotics Control CommissionChina Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics ControlBeijingChina
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic CenterMinistry of Public SecurityBeijingChina
| | - Dingding Zhuang
- Department of PsychiatryAffiliated Kangning Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Addiction Research of Zhejiang ProvinceNingboChina
| | - Majie Wang
- Department of PsychiatryAffiliated Kangning Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Addiction Research of Zhejiang ProvinceNingboChina
| | - Zeming Xu
- Department of PsychiatryAffiliated Kangning Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Addiction Research of Zhejiang ProvinceNingboChina
| | - Huifen Liu
- Department of PsychiatryAffiliated Kangning Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Addiction Research of Zhejiang ProvinceNingboChina
| | - Haowei Shen
- Faculty of Physiology & Pharmacology, School of MedicineNingbo UniversityNingboChina
| | - Peng Xu
- Office of China National Narcotics Control CommissionChina Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics ControlBeijingChina
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic CenterMinistry of Public SecurityBeijingChina
| | - Wenhua Zhou
- Department of PsychiatryAffiliated Kangning Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Addiction Research of Zhejiang ProvinceNingboChina
| |
Collapse
|
9
|
Brown KT, Levis SC, O'Neill CE, Levy C, Rice KC, Watkins LR, Bachtell RK. Toll-like receptor 4 antagonists reduce cocaine-primed reinstatement of drug seeking. Psychopharmacology (Berl) 2023; 240:1587-1600. [PMID: 37286899 PMCID: PMC10732226 DOI: 10.1007/s00213-023-06392-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/23/2023] [Indexed: 06/09/2023]
Abstract
RATIONALE Cocaine can increase inflammatory neuroimmune markers, including chemokines and cytokines characteristic of innate inflammatory responding. Prior work indicates that the Toll-like receptor 4 (TLR4) initiates this response, and administration of TLR4 antagonists provides mixed evidence that TLR4 contributes to cocaine reward and reinforcement. OBJECTIVE These studies utilize (+)-naltrexone, the TLR4 antagonist, and mu-opioid inactive enantiomer to examine the role of TLR4 on cocaine self-administration and cocaine seeking in rats. METHODS (+)-Naltrexone was continuously administered via an osmotic mini-pump during the acquisition or maintenance of cocaine self-administration. The motivation to acquire cocaine was assessed using a progressive ratio schedule following either continuous and acute (+)-naltrexone administration. The effects of (+)-naltrexone on cocaine seeking were assessed using both a cue craving model and a drug-primed reinstatement model. The highly selective TLR4 antagonist, lipopolysaccharide from Rhodobacter sphaeroides (LPS-Rs), was administered into the nucleus accumbens to determine the effectiveness of TLR4 blockade on cocaine-primed reinstatement. RESULTS (+)-Naltrexone administration did not alter the acquisition or maintenance of cocaine self-administration. Similarly, (+)-naltrexone was ineffective at altering the progressive ratio responding. Continuous administration of (+)-naltrexone during forced abstinence did not impact cued cocaine seeking. Acute systemic administration of (+)-naltrexone dose-dependently decreased cocaine-primed reinstatement of previously extinguished cocaine seeking, and administration of LPS-Rs into the nucleus accumbens shell also reduced cocaine-primed reinstatement of cocaine seeking. DISCUSSION These results complement previous studies suggesting that the TLR4 plays a role in cocaine-primed reinstatement of cocaine seeking, but may have a more limited role in cocaine reinforcement.
Collapse
Affiliation(s)
- Kyle T Brown
- Department of Psychology and Neuroscience and Center for Neuroscience, Boulder, CO, USA
| | - Sophia C Levis
- Department of Psychology and Neuroscience and Center for Neuroscience, Boulder, CO, USA
| | - Casey E O'Neill
- Department of Psychology and Neuroscience and Center for Neuroscience, Boulder, CO, USA
| | - Catherine Levy
- Department of Psychology and Neuroscience and Center for Neuroscience, Boulder, CO, USA
| | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Linda R Watkins
- Department of Psychology and Neuroscience and Center for Neuroscience, Boulder, CO, USA
| | - Ryan K Bachtell
- Department of Psychology and Neuroscience and Center for Neuroscience, Boulder, CO, USA.
- Institute for Behavioral Genetics University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
10
|
Cheng Y, Dempsey RE, Roodsari SK, Shuboni-Mulligan DD, George O, Sanford LD, Guo ML. Cocaine Regulates NLRP3 Inflammasome Activity and CRF Signaling in a Region- and Sex-Dependent Manner in Rat Brain. Biomedicines 2023; 11:1800. [PMID: 37509440 PMCID: PMC10376186 DOI: 10.3390/biomedicines11071800] [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: 05/03/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Cocaine, one of the most abused drugs worldwide, is capable of activating microglia in vitro and in vivo. Several neuroimmune pathways have been suggested to play roles in cocaine-mediated microglial activation. Previous work showed that cocaine activates microglia in a region-specific manner in the brains of self-administered mice. To further characterize the effects of cocaine on microglia and neuroimmune signaling in vivo, we utilized the brains from both sexes of outbred rats with cocaine self-administration to explore the activation status of microglia, NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activity, corticotropin-releasing factor (CRF) signaling, and NF-κB levels in the striatum and hippocampus (HP). Age-matched rats of the same sex (drug naïve) served as controls. Our results showed that cocaine increased neuroinflammation in the striatum and HP of both sexes with a relatively higher increases in male brains. In the striatum, cocaine upregulated NLRP3 inflammasome activity and CRF levels in males but not in females. In contrast, cocaine increased NLRP3 inflammasome activity in the HP of females but not in males, and no effects on CRF signaling were observed in this region of either sex. Interestingly, cocaine increased NF-κB levels in the striatum and HP with no sex difference. Taken together, our results provide evidence that cocaine can exert region- and sex-specific differences in neuroimmune signaling in the brain. Targeting neuroimmune signaling has been suggested as possible treatment for cocaine use disorders (CUDs). Our current results indicate that sex should be taken into consideration when determining the efficacy of these new therapeutic approaches.
Collapse
Affiliation(s)
- Yan Cheng
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Rachael Elizabeth Dempsey
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Soheil Kazemi Roodsari
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Dorela D Shuboni-Mulligan
- Sleep Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Olivier George
- Department of Psychiatry, School of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Larry D Sanford
- Sleep Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Ming-Lei Guo
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| |
Collapse
|
11
|
Karimi-Haghighi S, Chavoshinezhad S, Mozafari R, Noorbakhsh F, Borhani-Haghighi A, Haghparast A. Neuroinflammatory Response in Reward-Associated Psychostimulants and Opioids: A Review. Cell Mol Neurobiol 2023; 43:649-682. [PMID: 35461410 PMCID: PMC11415174 DOI: 10.1007/s10571-022-01223-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/26/2022] [Indexed: 11/03/2022]
Abstract
Substance abuse is one of the significant problems in social and public health worldwide. Vast numbers of evidence illustrate that motivational and reinforcing impacts of addictive drugs are primarily attributed to their ability to change dopamine signaling in the reward circuit. However, the roles of classic neurotransmitters, especially dopamine and neuromodulators, monoamines, and neuropeptides, in reinforcing characteristics of abused drugs have been extensively investigated. It has recently been revealed that central immune signaling includes cascades of chemokines and proinflammatory cytokines released by neurons and glia via downstream intracellular signaling pathways that play a crucial role in mediating rewarding behavioral effects of drugs. More interestingly, inflammatory responses in the central nervous system modulate the mesolimbic dopamine signaling and glutamate-dependent currents induced by addictive drugs. This review summarized researches in the alterations of inflammatory responses accompanied by rewarding and reinforcing properties of addictive drugs, including cocaine, methamphetamine, and opioids that were evaluated by conditioned place preference and self-administration procedures as highly common behavioral tests to investigate the motivational and reinforcing impacts of addictive drugs. The neuroinflammatory responses affect the rewarding properties of psychostimulants and opioids.
Collapse
Affiliation(s)
- Saeideh Karimi-Haghighi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19615-1178, Tehran, Iran
| | - Sara Chavoshinezhad
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Roghayeh Mozafari
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19615-1178, Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | | | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19615-1178, Tehran, Iran.
| |
Collapse
|
12
|
Smiley CE, Wood SK. Stress- and drug-induced neuroimmune signaling as a therapeutic target for comorbid anxiety and substance use disorders. Pharmacol Ther 2022; 239:108212. [PMID: 35580690 DOI: 10.1016/j.pharmthera.2022.108212] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
Stress and substance use disorders remain two of the most highly prevalent psychiatric conditions and are often comorbid. While individually these conditions have a debilitating impact on the patient and a high cost to society, the symptomology and treatment outcomes are further exacerbated when they occur together. As such, there are few effective treatment options for these patients, and recent investigation has sought to determine the neural processes underlying the co-occurrence of these disorders to identify novel treatment targets. One such mechanism that has been linked to stress- and addiction-related conditions is neuroimmune signaling. Increases in inflammatory factors across the brain have been heavily implicated in the etiology of these disorders, and this review seeks to determine the nature of this relationship. According to the "dual-hit" hypothesis, also referred to as neuroimmune priming, prior exposure to either stress or drugs of abuse can sensitize the neuroimmune system to be hyperresponsive when exposed to these insults in the future. This review completes an examination of the literature surrounding stress-induced increases in inflammation across clinical and preclinical studies along with a summarization of the evidence regarding drug-induced alterations in inflammatory factors. These changes in neuroimmune profiles are also discussed within the context of their impact on the neural circuitry responsible for stress responsiveness and addictive behaviors. Further, this review explores the connection between neuroimmune signaling and susceptibility to these conditions and highlights the anti-inflammatory pharmacotherapies that may be used for the treatment of stress and substance use disorders.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| |
Collapse
|
13
|
Wang Q, Guo X, Yue Q, Zhu S, Guo L, Li G, Zhou Q, Xiang Y, Chen G, Yin W, Sun J. Exploring the role and mechanism of gut microbiota in methamphetamine addiction using antibiotic treatment followed by fecal microbiota transplantation. Anat Rec (Hoboken) 2022; 306:1149-1164. [PMID: 36054423 DOI: 10.1002/ar.25055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/11/2022] [Accepted: 07/12/2022] [Indexed: 11/09/2022]
Abstract
Recently, the role of the gut microbiota in the context of drug addiction has attracted the attention of researchers; however, the specific effects and underlying mechanisms require further exploration. To accomplish this, C57BL/6 mice were firstly treated with methamphetamine (MA). Conditioned place preference (CPP) behavior changes, gut permeability and function, microglial activation, and inflammatory cytokine expression were systematically analyzed in antibiotics-treated mice with microbiota depletion and in fecal microbiota transplantation mice with microbiota reconstitution. MA treatment altered microbiota composition and caused gut dysbiosis. Depletion of gut microbiota with antibiotics inhibited MA-induced CPP formation, and fecal microbiota transplantation reversed this inhibition. Mechanistic analyses indicated that antibiotic treatment decreased gut permeability and neuroinflammation, while fecal microbiota transplantation offset the impact of antibiotic treatment. Additionally, MA-induced microglial activation was suppressed by antibiotics but restored by microbiota transplantation, and this correlated well with the CPP score. Compared to antibiotic treatment, microbiota transplantation significantly increased 5-HT4 receptor expression in both the nucleus accumbens and the hippocampus. Furthermore, when fecal microbiota from healthy mice was transplanted into MA-treated mice, the CPP scores decreased. Our results provide a novel avenue for understanding MA addiction and suggest a potential future intervention strategy.
Collapse
Affiliation(s)
- Qiuting Wang
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Xiuwen Guo
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Qingwei Yue
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Shaowei Zhu
- Department of Neurology Qilu Hospital of Shandong University Jinan China
| | - Liying Guo
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Guibao Li
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Qidi Zhou
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Yunzhi Xiang
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Ganggang Chen
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Wei Yin
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Jinhao Sun
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| |
Collapse
|
14
|
Ortinski PI, Reissner KJ, Turner J, Anderson TA, Scimemi A. Control of complex behavior by astrocytes and microglia. Neurosci Biobehav Rev 2022; 137:104651. [PMID: 35367512 PMCID: PMC9119927 DOI: 10.1016/j.neubiorev.2022.104651] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/28/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
Evidence that glial cells influence behavior has been gaining a steady foothold in scientific literature. Out of the five main subtypes of glial cells in the brain, astrocytes and microglia have received an outsized share of attention with regard to shaping a wide spectrum of behavioral phenomena and there is growing appreciation that the signals intrinsic to these cells as well as their interactions with surrounding neurons reflect behavioral history in a brain region-specific manner. Considerable regional diversity of glial cell phenotypes is beginning to be recognized and may contribute to behavioral outcomes arising from circuit-specific computations within and across discrete brain nuclei. Here, we summarize current knowledge on the impact of astrocyte and microglia activity on behavioral outcomes, with a specific focus on brain areas relevant to higher cognitive control, reward-seeking, and circadian regulation.
Collapse
Affiliation(s)
- P I Ortinski
- Department of Neuroscience, University of Kentucky, USA
| | - K J Reissner
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, USA
| | - J Turner
- Department of Pharmaceutical Sciences, University of Kentucky, USA
| | - T A Anderson
- Department of Neuroscience, University of Kentucky, USA
| | - A Scimemi
- Department of Biology, State University of New York at Albany, USA
| |
Collapse
|
15
|
Roodsari SK, Cheng Y, Reed KM, Wellman LL, Sanford LD, Kim WK, Guo ML. Sleep Disturbance Alters Cocaine-Induced Locomotor Activity: Involvement of Striatal Neuroimmune and Dopamine Signaling. Biomedicines 2022; 10:biomedicines10051161. [PMID: 35625897 PMCID: PMC9138453 DOI: 10.3390/biomedicines10051161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 12/10/2022] Open
Abstract
Sleep disorders have high comorbidity with drug addiction and function as major risk factors for developing drug addiction. Recent studies have indicated that both sleep disturbance (SD) and abused drugs could activate microglia, and that increased neuroinflammation plays a critical role in the pathogenesis of both diseases. Whether microglia are involved in the contribution of chronic SDs to drug addiction has never been explored. In this study, we employed a mouse model of sleep fragmentation (SF) with cocaine treatment and examined their locomotor activities, as well as neuroinflammation levels and dopamine signaling in the striatum, to assess their interaction. We also included mice with, or without, SF that underwent cocaine withdrawal and challenge. Our results showed that SF significantly blunted cocaine-induced locomotor stimulation while having marginal effects on locomotor activity of mice with saline injections. Meanwhile, SF modulated the effects of cocaine on neuroimmune signaling in the striatum and in ex vivo isolated microglia. We did not observe differences in dopamine signaling in the striatum among treatment groups. In mice exposed to cocaine and later withdrawal, SF reduced locomotor sensitivity and also modulated neuroimmune and dopamine signaling in the striatum. Taken together, our results suggested that SF was capable of blunting cocaine-induced psychoactive effects through modulating neuroimmune and dopamine signaling. We hypothesize that SF could affect neuroimmune and dopamine signaling in the brain reward circuitry, which might mediate the linkage between sleep disorders and drug addiction.
Collapse
Affiliation(s)
- Soheil Kazemi Roodsari
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (S.K.R.); (Y.C.); (K.M.R.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
| | - Yan Cheng
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (S.K.R.); (Y.C.); (K.M.R.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
| | - Kirstin M. Reed
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (S.K.R.); (Y.C.); (K.M.R.)
| | - Laurie L. Wellman
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Larry D. Sanford
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Woong-Ki Kim
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Ming-Lei Guo
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (S.K.R.); (Y.C.); (K.M.R.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
- Correspondence: ; Tel.: +1-757-446-5891
| |
Collapse
|
16
|
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
|
17
|
NLRP3 Inflammasome Is Involved in Cocaine-Mediated Potentiation on Behavioral Changes in CX3CR1-Deficient Mice. J Pers Med 2021; 11:jpm11100963. [PMID: 34683104 PMCID: PMC8540128 DOI: 10.3390/jpm11100963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/25/2021] [Accepted: 09/22/2021] [Indexed: 11/30/2022] Open
Abstract
Microglia, the primary immunocompetent cells of the brain, are suggested to play a role in the development of drug addiction. Previous studies have identified the microglia-derived pro-inflammatory factor IL1β can promote the progression of cocaine addiction. Additionally, the activation status of microglia and “two-hit hypothesis” have been proposed in the field of drug addiction to explain how early life stress (ELS) could significantly increase the incidence of drug addiction in later life. However, the mechanisms underlying microglia prime and full activation and their roles in drug addiction remain greatly unexplored. Here, we employed CX3CR1-GFP mice (CX3CR1 functional deficiency, CX3CR1−/−) to explore whether primed microglia could potentiate cocaine-mediated behavioral changes and the possible underlying mechanisms. CX3CR1−/− mice revealed higher hyperlocomotion activity and conditional place preference than wild-type (WT) mice did under cocaine administration. In parallel, CX3CR1−/− mice showed higher activity of NLR family pyrin domain-containing 3 (NLRP3) inflammasome than WT mice. Interestingly, CX3CR1 deficiency itself could prime NLRP3 signaling by increasing the expression of NLPR3 and affect lysosome biogenesis under basal conditions. Taken together, our findings demonstrated that the functional status of microglia could have an impact on cocaine-mediated reward effects, and NLRP3 inflammasome activity was associated with this phenomenon. This study was consistent with the two-hit hypothesis and provided solid evidence to support the involvement of microglia in drug addiction. Targeting the NLRP3 inflammasome may represent a novel therapeutic approach for ameliorating or blocking the development of drug addiction.
Collapse
|
18
|
Mogali S, Askalsky P, Madera G, Jones JD, Comer SD. Minocycline attenuates oxycodone-induced positive subjective responses in non-dependent, recreational opioid users. Pharmacol Biochem Behav 2021; 209:173241. [PMID: 34298029 DOI: 10.1016/j.pbb.2021.173241] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 01/28/2023]
Abstract
BACKGROUND Recent data suggest that glial cells may be involved in the analgesic effects and abuse liability of opioids. Preclinical studies have demonstrated that mu-opioid-receptor-selective agonists, such as oxycodone, activate glia and increase the release of cytokines, causing a suppression of opioid-induced analgesic effects. Preclinical studies also show that certain medications, such as the broad-spectrum tetracycline antibiotic minocycline, inhibit opioid-induced glial activation and thereby enhance the analgesic effects of opioids. Importantly, minocycline reduces the rewarding effects of opioids at the same doses that it enhances opioid-induced analgesia. AIMS The purpose of the present study was to assess the effects of acute administration of minocycline on the subjective, physiological, and analgesic effects of oxycodone in human research volunteers. DESIGN This study was a within-subject, randomized, double-blind outpatient study. Participants completed five separate sessions in which they received 0, 100, or 200 mg minocycline (MINO) simultaneously with either 0 or 40 mg oxycodone (OXY). The subjective, physiological, and analgesic effects of OXY were measured before and repeatedly after drug administration. SETTINGS AND PARTICIPANTS Participants were between 21 and 45 years of age, non-treatment seeking, non-dependent recreational opioid users (N = 12). This study was conducted between 2013 and 2014 at the New York State Psychiatric Institute in New York, NY. FINDINGS MINO 100 and 200 mg were safe and well-tolerated in combination with OXY 40 mg. MINO 200 mg administered with OXY 40 mg attenuated OXY-induced positive subjective effects such as "Good Effect" and "Liking" compared to OXY alone. MINO did not alter the physiological or analgesic effects of OXY. CONCLUSIONS MINO may attenuate the abuse liability of mu-opioid-receptor-selective agonists.
Collapse
Affiliation(s)
- S Mogali
- Division on Substance Use Disorders, New York State Psychiatric Institute, Department of Psychiatry, Vagelos College of Physicians and Surgeons of Columbia University, 1051 Riverside Dr., Unit 66, New York, NY 10032, United States of America.
| | - P Askalsky
- NYU Langone School of Medicine, Department of Psychiatry, New York, NY 10016, United States of America
| | - G Madera
- Weill Cornell Medical College, 515 East 71st Street, New York, NY 10021, United States of America
| | - J D Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute, Department of Psychiatry, Vagelos College of Physicians and Surgeons of Columbia University, 1051 Riverside Dr., Unit 66, New York, NY 10032, United States of America
| | - S D Comer
- Division on Substance Use Disorders, New York State Psychiatric Institute, Department of Psychiatry, Vagelos College of Physicians and Surgeons of Columbia University, 1051 Riverside Dr., Unit 66, New York, NY 10032, United States of America
| |
Collapse
|
19
|
Namba MD, Leyrer-Jackson JM, Nagy EK, Olive MF, Neisewander JL. Neuroimmune Mechanisms as Novel Treatment Targets for Substance Use Disorders and Associated Comorbidities. Front Neurosci 2021; 15:650785. [PMID: 33935636 PMCID: PMC8082184 DOI: 10.3389/fnins.2021.650785] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Recent studies examining the neurobiology of substance abuse have revealed a significant role of neuroimmune signaling as a mechanism through which drugs of abuse induce aberrant changes in synaptic plasticity and contribute to substance abuse-related behaviors. Immune signaling within the brain and the periphery critically regulates homeostasis of the nervous system. Perturbations in immune signaling can induce neuroinflammation or immunosuppression, which dysregulate nervous system function including neural processes associated with substance use disorders (SUDs). In this review, we discuss the literature that demonstrates a role of neuroimmune signaling in regulating learning, memory, and synaptic plasticity, emphasizing specific cytokine signaling within the central nervous system. We then highlight recent preclinical studies, within the last 5 years when possible, that have identified immune mechanisms within the brain and the periphery associated with addiction-related behaviors. Findings thus far underscore the need for future investigations into the clinical potential of immunopharmacology as a novel approach toward treating SUDs. Considering the high prevalence rate of comorbidities among those with SUDs, we also discuss neuroimmune mechanisms of common comorbidities associated with SUDs and highlight potentially novel treatment targets for these comorbid conditions. We argue that immunopharmacology represents a novel frontier in the development of new pharmacotherapies that promote long-term abstinence from drug use and minimize the detrimental impact of SUD comorbidities on patient health and treatment outcomes.
Collapse
Affiliation(s)
- Mark D. Namba
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | - Erin K. Nagy
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | | |
Collapse
|
20
|
Angoa-Pérez M, Kuhn DM. Evidence for Modulation of Substance Use Disorders by the Gut Microbiome: Hidden in Plain Sight. Pharmacol Rev 2021; 73:571-596. [PMID: 33597276 PMCID: PMC7896134 DOI: 10.1124/pharmrev.120.000144] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The gut microbiome modulates neurochemical function and behavior and has been implicated in numerous central nervous system (CNS) diseases, including developmental, neurodegenerative, and psychiatric disorders. Substance use disorders (SUDs) remain a serious threat to the public well-being, yet gut microbiome involvement in drug abuse has received very little attention. Studies of the mechanisms underlying SUDs have naturally focused on CNS reward circuits. However, a significant body of research has accumulated over the past decade that has unwittingly provided strong support for gut microbiome participation in drug reward. β-Lactam antibiotics have been employed to increase glutamate transporter expression to reverse relapse-induced release of glutamate. Sodium butyrate has been used as a histone deacetylase inhibitor to prevent drug-induced epigenetic alterations. High-fat diets have been used to alter drug reward because of the extensive overlap of the circuitry mediating them. This review article casts these approaches in a different light and makes a compelling case for gut microbiome modulation of SUDs. Few factors alter the structure and composition of the gut microbiome more than antibiotics and a high-fat diet, and butyrate is an endogenous product of bacterial fermentation. Drugs such as cocaine, alcohol, opiates, and psychostimulants also modify the gut microbiome. Therefore, their effects must be viewed on a complex background of cotreatment-induced dysbiosis. Consideration of the gut microbiome in SUDs should have the beneficial effects of expanding the understanding of SUDs and aiding in the design of new therapies based on opposing the effects of abused drugs on the host's commensal bacterial community. SIGNIFICANCE STATEMENT: Proposed mechanisms underlying substance use disorders fail to acknowledge the impact of drugs of abuse on the gut microbiome. β-Lactam antibiotics, sodium butyrate, and high-fat diets are used to modify drug seeking and reward, overlooking the notable capacity of these treatments to alter the gut microbiome. This review aims to stimulate research on substance abuse-gut microbiome interactions by illustrating how drugs of abuse share with antibiotics, sodium butyrate, and fat-laden diets the ability to modify the host microbial community.
Collapse
Affiliation(s)
- Mariana Angoa-Pérez
- Research and Development Service, John D. Dingell VA Medical Center, and Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Donald M Kuhn
- Research and Development Service, John D. Dingell VA Medical Center, and Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
21
|
Rathitharan G, Truong J, Tong J, McCluskey T, Meyer JH, Mizrahi R, Warsh J, Rusjan P, Kennedy JL, Houle S, Kish SJ, Boileau I. Microglia imaging in methamphetamine use disorder: a positron emission tomography study with the 18 kDa translocator protein radioligand [F-18]FEPPA. Addict Biol 2021; 26:e12876. [PMID: 32017280 DOI: 10.1111/adb.12876] [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: 10/29/2019] [Revised: 01/08/2020] [Accepted: 01/12/2020] [Indexed: 01/19/2023]
Abstract
Activation of brain microglial cells, microgliosis, has been linked to methamphetamine (MA)-seeking behavior, suggesting that microglia could be a new therapeutic target for MA use disorder. Animal data show marked brain microglial activation following acute high-dose MA, but microglial status in human MA users is uncertain, with one positron emission tomography (PET) investigation reporting massively and globally increased translocator protein 18 kDa (TSPO; [C-11](R)-PK11195) binding, a biomarker for microgliosis, in MA users. Our aim was to measure binding of a second-generation TSPO radioligand, [F-18]FEPPA, in brain of human chronic MA users. Regional total volume of distribution (VT ) of [F-18]FEPPA was estimated with a two-tissue compartment model with arterial plasma input function for 10 regions of interest in 11 actively using MA users and 26 controls. A RM-ANOVA corrected for TSPO rs6971 polymorphism was employed to test significance. There was no main effect of group on [F-18]FEPPA VT (P = .81). No significant correlations between [F-18]FEPPA VT and MA use duration, weekly dosage, blood MA concentrations, regional brain volumes, and self-reported craving were observed. Our preliminary findings, consistent with our earlier postmortem data, do not suggest substantial brain microgliosis in MA use disorder but do not rule out microglia as a therapeutic target in MA addiction. Absence of increased [F-18]FEPPA TSPO binding might be related to insufficient MA dose or blunting of microglial response following repeated MA exposure, as suggested by some animal data.
Collapse
Affiliation(s)
- Gausiha Rathitharan
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Jennifer Truong
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Junchao Tong
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
| | - Tina McCluskey
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
| | - Jeffrey H. Meyer
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Romina Mizrahi
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Jerry Warsh
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Pablo Rusjan
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - James L. Kennedy
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Sylvain Houle
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
| | - Stephen J. Kish
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Isabelle Boileau
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| |
Collapse
|
22
|
Romero-Miguel D, Lamanna-Rama N, Casquero-Veiga M, Gómez-Rangel V, Desco M, Soto-Montenegro ML. Minocycline in neurodegenerative and psychiatric diseases: An update. Eur J Neurol 2020; 28:1056-1081. [PMID: 33180965 DOI: 10.1111/ene.14642] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Minocycline is a broad-spectrum antibiotic, effective as a chronic treatment for recurrent bacterial infections. Beyond its antibiotic action, minocycline also has important anti-inflammatory, antioxidant and antiapoptotic properties. Its efficacy has therefore been evaluated in many neurodegenerative and psychiatric diseases that have an inflammatory basis. Our aim was to review preclinical and clinical studies performed in neurological and psychiatric diseases whose treatment involved the use of minocycline and thereby to discern the possible beneficial effect of minocycline in these disorders. METHODS Completed and ongoing preclinical studies and clinical trials of minocycline for both neurodegenerative diseases and psychiatric disorders, published from January 1995 to January 2020, were identified through searching relevant databases (https://www.ncbi.nlm.nih.gov/pubmed/, https://clinicaltrials.gov/). A total of 74 preclinical studies and 44 clinical trials and open-label studies were selected. RESULTS The results of the nearly 20 years of research identified are diverse. While minocycline mostly proved to be effective in animal models, clinical results showed divergent outcomes, with positive results in some studies counterbalanced by a number of cases with no significant improvements. Specific data for each disease are further individually described in this review. CONCLUSIONS Despite minocycline demonstrating antioxidant and anti-inflammatory effects, discrepancies between preclinical and clinical data indicate that we should be cautious in analyzing the outcomes. Improving and standardizing protocols and refining animal models could help us to determine if minocycline really is a useful drug in the treatment of these pathologies.
Collapse
Affiliation(s)
| | | | - Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,CIBER de Salud Mental (CIBERSAM), Madrid
| | | | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,CIBER de Salud Mental (CIBERSAM), Madrid.,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - María Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,CIBER de Salud Mental (CIBERSAM), Madrid
| |
Collapse
|
23
|
Wan X, Fujita Y, Chang L, Wei Y, Ma L, Wuyun G, Pu Y, Hammock BD, Hashimoto K. Lack of rewarding effects of a soluble epoxide hydrolase inhibitor TPPU in mice: Comparison with morphine. Neuropsychopharmacol Rep 2020; 40:412-416. [PMID: 32896112 PMCID: PMC7722641 DOI: 10.1002/npr2.12136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/05/2020] [Accepted: 08/16/2020] [Indexed: 12/19/2022] Open
Abstract
AIM Although opioids have been used as treatment of neuropathic pain, opioids have abuse potential in humans. Since soluble epoxide hydrolase (sEH) in the metabolism of polyunsaturated fatty acids plays a key role in the pain, sEH inhibitors would be promising new therapeutic drugs for neuropathic pain. In this study, we examined the effect of the sEH inhibitor TPPU on rewarding effects in mice using the conditioned place preference (CPP) paradigm. METHODS The rewarding effects of morphine (10 mg/kg) and TPPU (3, 10, or 30 mg/kg) in mice were examined using CPP paradigm. Furthermore, the effect of TPPU (30 mg/kg) on morphine-induced rewarding effects was examined. RESULTS TPPU (3, 10, or 30 mg/kg) did not increase CPP scores in the mice whereas morphine significantly increased CPP scores in the mice. Furthermore, pretreatment with TPPU did not block the rewarding effects of morphine in the mice, suggesting that sEH does not play a role in the rewarding effect of morphine. CONCLUSION This study suggests that TPPU did not have rewarding effects in rodents. This would make sEH inhibitors potential therapeutic drugs without abuse potential for neuropathic pain.
Collapse
Affiliation(s)
- Xiayun Wan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Yuko Fujita
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Lijia Chang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Yan Wei
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Li Ma
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Gerile Wuyun
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Yaoyu Pu
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Bruce D Hammock
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| |
Collapse
|
24
|
Burkovetskaya ME, Liu Q, Vadukoot AK, Gautam N, Alnouti Y, Kumar S, Miczek K, Buch S, Hopkins CR, Guo M. KVA-D-88, a Novel Preferable Phosphodiesterase 4B Inhibitor, Decreases Cocaine-Mediated Reward Properties in Vivo. ACS Chem Neurosci 2020; 11:2231-2242. [PMID: 32609488 DOI: 10.1021/acschemneuro.0c00170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cocaine addiction remains a major public concern throughout the world especially in developed countries. In the last three decades, significant achievements have led to a greater understanding of the signaling pathways involved in the development of cocaine addiction; however, there are no FDA-approved treatments available to reverse or block this brain disease due to either the unsatisfactory therapeutic efficacy or severe side effects. Previous studies have demonstrated that chronic exposure to cocaine elevates levels of cyclic AMP (cAMP) as a neuroadaptative response in reward-related brain regions. Phosphodiesterase 4 (PDE4) inhibitors, which elevate cAMP levels, have been shown to block cocaine-mediated behavioral changes related to psychoactive and reinforcing properties. Unfortunately, previously studied PDE4 inhibitors induce severe side-effects, which limit their clinical usage. In this study, we identified a novel PDE4B inhibitor, KVA-D-88, with an improved selectivity profile compared to previous compounds (e.g., rolipram). Pharmacokinetic studies have shown that this compound is brain penetrant and preferably acts on PDE4B compared to PDE4D in vitro, alluding to less unwanted side effects with KVA-D-88 in vivo. Interestingly, pretreatment with KVA-D-88 significantly inhibited cocaine-induced hyperlocomotor activity. In cocaine self-administering mice with differential schedules, KVA-D-88 strikingly decreased the number of active nose-pokes and cocaine infusions and reduced the break point. Taken together, our findings demonstrate that this novel PDE4 inhibitor, KVA-D-88, could inhibit cocaine-mediated rewarding effects implying its potential clinical usage for cocaine addiction.
Collapse
Affiliation(s)
- Maria E. Burkovetskaya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Qiaoling Liu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Anish K. Vadukoot
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Nagsen Gautam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Yazen Alnouti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Sushil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Klaus Miczek
- Department of Psychology, Tufts University, 530 Boston Ave, Medford, Massachusetts 02155, United States
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Corey R. Hopkins
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Minglei Guo
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia 23507, United States
| |
Collapse
|
25
|
Mahmoudiasl GR, Abbaszadeh HA, Rezaei-Tavirani M, Abdollahifar MA, Sadeghi Y, Khoramgah MS, Niknazar S, Darabi S. Postmortem Study of Molecular and Histological Changes in the CA1 Hippocampal Region of Chronic Methamphetamine User. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 18:2067-2082. [PMID: 32184870 PMCID: PMC7059073 DOI: 10.22037/ijpr.2019.15483.13123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Methamphetamine (Meth) is recognized as one of the most important new distributed abused drug that causes severe damage to the different parts of the brain, especially hippocampus. Previous studies have demonstrated that Meth can induce apoptosis and cell death in the brain. In this study, we evaluated the long-term effects of Meth abuse in the CA1 region of postmortem hippocampus. Postmortem molecular and histological analysis was performed for five non-addicted subjects and five Meth addicted ones. Iba-1 (microglia) and glial fibrillary acidic protein, GFAP (astrocytes) expression were assayed by western blotting and immunohistochemistry (IHC) methods. Histopathological assessment was done with stereological counts of hippocampal cells stained with hematoxylin and eosin (H and E). Tunel staining was used to detect DNA damage in human brains. In addition, protein-protein interaction analysis network was investigated. Western blotting and immunohistochemistry assay showed overexpression of GFAP and Iba-1 protein in the CA1 hippocampal region of Meth users’ brain. Stereological analysis in the CA1 region revealed increased neuron degeneration. Furthermore, significant apoptosis and cell death were confirmed by Tunel assay in the hippocampus. The prominent role of TLR4, IL1B, CASP1, and NLRP3 in the molecular mechanism of Meth was highlighted via PPI network analysis. Chronic Meth use can induce GFAP and Iba-1 upregulation and neuronal apoptosis in the CA1 region of the postmortem hippocampus.
Collapse
Affiliation(s)
- Gholam-Reza Mahmoudiasl
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yousef Sadeghi
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Sadat Khoramgah
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Niknazar
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Darabi
- Cellular and Molecular Research Center, Faculty of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
26
|
Catale C, Bussone S, Lo Iacono L, Carola V. Microglial alterations induced by psychoactive drugs: A possible mechanism in substance use disorder? Semin Cell Dev Biol 2019; 94:164-175. [PMID: 31004753 DOI: 10.1016/j.semcdb.2019.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/19/2019] [Accepted: 03/29/2019] [Indexed: 12/11/2022]
Abstract
Recently, the xenobiotic hypothesis has implicated the immune system in targeting substances of abuse as foreign molecules and stimulating inflammatory responses. Microglial cells are the resident immune cells of the central nervous system and function in homeostatic surveillance. Microglial changes that are induced by exposure to substances of abuse appear to mediate in part the establishment of addiction and the persistence of drug-mediated biological and behavioral changes. In this context, interest in the study of drug-microglia interactions has increased recently. This review summarizes the most recent preclinical rodent and clinical studies on the interaction between microglia and various classes of drugs of abuse, such as ethanol, psychostimulants, and opioids. The principal biological mechanisms of the communication between substances of abuse and microglia will be described to consider putative mechanisms of the establishment of drug addiction and future potential targets for treating substance use disorder.
Collapse
Affiliation(s)
- Clarissa Catale
- Department of Psychology, University of Rome "La Sapienza", Via dei Marsi, 78, 00185 Rome, Italy
| | - Silvia Bussone
- Department of Dynamic and Clinical Psychology, University of Rome "La Sapienza", Via degli Apuli 1, 00185 Rome, Italy
| | - Luisa Lo Iacono
- Department of Psychology, University of Rome "La Sapienza", Via dei Marsi, 78, 00185 Rome, Italy; IRCCS Santa Lucia Foundation, Via Fosso di Fiorano 64, 00143 Rome, Italy
| | - Valeria Carola
- Department of Dynamic and Clinical Psychology, University of Rome "La Sapienza", Via degli Apuli 1, 00185 Rome, Italy; IRCCS Santa Lucia Foundation, Via Fosso di Fiorano 64, 00143 Rome, Italy.
| |
Collapse
|
27
|
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: 103] [Impact Index Per Article: 17.2] [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
|
28
|
Dave S, Chen L, Yu C, Seaton M, Khodr CE, Al-Harthi L, Hu XT. Methamphetamine decreases K + channel function in human fetal astrocytes by activating the trace amine-associated receptor type-1. J Neurochem 2018; 148:29-45. [PMID: 30295919 DOI: 10.1111/jnc.14606] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 09/28/2018] [Accepted: 10/03/2018] [Indexed: 12/22/2022]
Abstract
Methamphetamine (Meth) is a potent and commonly abused psychostimulant. Meth alters neuron and astrocyte activity; yet the underlying mechanism(s) is not fully understood. Here we assessed the impact of acute Meth on human fetal astrocytes (HFAs) using whole-cell patch-clamping. We found that HFAs displayed a large voltage-gated K+ efflux (IKv ) through Kv /Kv -like channels during membrane depolarization, and a smaller K+ influx (Ikir ) via inward-rectifying Kir /Kir -like channels during membrane hyperpolarization. Meth at a 'recreational' (20 μM) or toxic/fatal (100 μM) concentration depolarized resting membrane potential (RMP) and suppressed IKv/Kv-like . These changes were associated with a decreased time constant (Ƭ), and mimicked by blocking the two-pore domain K+ (K2P )/K2P -like and Kv /Kv -like channels, respectively. Meth also diminished IKir/Kir-like , but only at toxic/fatal levels. Given that Meth is a potent agonist for the trace amine-associated receptor type-1 (TAAR1), and TAAR1-coupled cAMP/cAMP-activated protein kinase (PKA) cascade, we further evaluated whether the Meth impact on K+ efflux was mediated by this pathway. We found that antagonizing TAAR1 with N-(3-Ethoxyphenyl)-4-(1-pyrrolidinyl)-3-(trifluoromethyl)benzamide (EPPTB) reversed Meth-induced suppression of IKv/Kv-like ; and inhibiting PKA activity by H89 abolished Meth effects on suppressing IKv/Kv-like . Antagonizing TAAR1 might also attenuate Meth-induced RMP depolarization. Voltage-gated Ca2+ currents were not detected in HFAs. These novel findings demonstrate that Meth suppresses IKv/Kv-like by facilitating the TAAR1/Gs /cAMP/PKA cascade and altering the kinetics of Kv /Kv -like channel gating, but reduces K2P /K2P -like channel activity through other pathway(s), in HFAs. Given that Meth-induced decrease in astrocytic K+ efflux through K2P /K2P -like and Kv /Kv -like channels reduces extracellular K+ levels, such reduction could consequently contribute to a decreased excitability of surrounding neurons. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Sonya Dave
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Lihua Chen
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Chunjiang Yu
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Melanie Seaton
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Christina E Khodr
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Lena Al-Harthi
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Xiu-Ti Hu
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
29
|
Arezoomandan R, Riahi E, Haghparast A. Minocycline increases firing rates of accumbal neurons and modifies the effects of morphine on neuronal activity. Addict Biol 2018; 23:1055-1066. [PMID: 28961365 DOI: 10.1111/adb.12557] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 08/15/2017] [Accepted: 08/18/2017] [Indexed: 01/02/2023]
Abstract
Accumulating evidence indicated that minocycline, a glial cell modulator, is able to modify a variety of morphine effects. Here, we investigated minocycline effects on electrical activity of nucleus accumbens (NAc) neurons using single unit recording in urethane-anesthetized rats. In addition, we investigated whether minocycline can modify the effects of morphine on NAc neural activity during reinstatement of morphine-seeking behavior. Minocycline increased the NAc firing activity in intact animals. Electrophysiological recording in morphine-treated animals was performed, following the acquisition of morphine-induced conditioned place preference (5 mg/kg, s.c., 3 days) and a drug-free extinction period. In acutely minocycline- treated animals, the neurons were recorded for 40 minutes following a single injection of either minocycline (50 μg/5 μl, i.c.v.) or saline. Then a priming dose of morphine (1 mg/kg, s.c.) was injected while the recording was continued for an additional 40 minutes. Minocycline significantly increased the firing rates of neurons and significantly modified morphine inhibitory effects on NAc neurons. In subchronically minocycline-treated groups, the rats were given daily injections of minocycline (50 μg/5 μl, i.c.v) during the extinction period. Then, on the reinstatement day, NAc neurons were recorded for 10 minutes, the priming dose of morphine was administered and the recording was continued for 45 minutes. Our results showed the failure of minocycline to significantly modify the inhibitory effects of morphine. In conclusion, our findings indicated that minocycline modifies morphine-induced decreases in the firing rates of NAc neurons in the reinstatement phase.
Collapse
Affiliation(s)
- Reza Arezoomandan
- Addiction Department, School of Behavioral Sciences and Mental Health (Tehran Institute of Psychiatry); Iran University of Medical Sciences; Tehran Iran
| | - Esmail Riahi
- Physiology Department, School of Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
30
|
Gajbhiye SV, Tripathi RK, Salve B, Petare A, Potey AV. Evaluation of effect of minocycline on rewarding potential and alcohol relapse in place preference model in mice. Neurosci Lett 2017; 649:28-33. [DOI: 10.1016/j.neulet.2017.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/28/2017] [Accepted: 04/03/2017] [Indexed: 01/28/2023]
|
31
|
Zhang JC, Yao W, Hashimoto K. Brain-derived Neurotrophic Factor (BDNF)-TrkB Signaling in Inflammation-related Depression and Potential Therapeutic Targets. Curr Neuropharmacol 2017; 14:721-31. [PMID: 26786147 PMCID: PMC5050398 DOI: 10.2174/1570159x14666160119094646] [Citation(s) in RCA: 377] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 10/08/2015] [Accepted: 11/10/2015] [Indexed: 12/20/2022] Open
Abstract
Depression is the most prevalent and among the most debilitating of psychiatric disorders. The precise neurobiology of this illness is unknown. Several lines of evidence suggest that peripheral and central inflammation plays a role in depressive symptoms, and that anti-inflammatory drugs can improve depressive symptoms in patients with inflammation-related depression. Signaling via brain-derived neurotrophic factor (BDNF) and its receptor, tropomycin receptor kinase B (TrkB) plays a key role in the pathophysiology of depression and in the therapeutic mechanisms of antidepressants. A recent paper showed that lipopolysaccharide (LPS)-induced inflammation gave rise to depression-like phenotype by altering BDNF-TrkB signaling in the prefrontal cortex, hippocampus, and nucleus accumbens, areas thought to be involved in the antidepressant effects of TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF) and TrkB antagonist, ANA-12. Here we provide an overview of the tryptophan-kynurenine pathway and BDNF-TrkB signaling in the pathophysiology of inflammation-induced depression, and propose mechanistic actions for potential therapeutic agents. Additionally, the authors discuss the putative role of TrkB agonists and antagonists as novel therapeutic drugs for inflammation-related depression.
Collapse
Affiliation(s)
| | | | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chiba 260-8670, Japan
| |
Collapse
|
32
|
Arezoomandan R, Moradi M, Attarzadeh-Yazdi G, Tomaz C, Haghparast A. Administration of activated glial condition medium in the nucleus accumbens extended extinction and intensified reinstatement of methamphetamine-induced conditioned place preference. Brain Res Bull 2016; 125:106-16. [PMID: 27346277 DOI: 10.1016/j.brainresbull.2016.06.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/17/2016] [Accepted: 06/21/2016] [Indexed: 02/08/2023]
Abstract
Methamphetamine (METH) is a psychostimulant drug with significant abuse potential and neurotoxic effects. A high percentage of users relapse to use after detoxification and no effective medication has been developed for treatment of METH addiction. Developing evidences indicated the role of glial cells in drugs abused related phenomena. However, little is known about the role of these cells in the maintenance and reinstatement of METH-seeking behaviors. Therefore, the current study was conducted to clarify the role of glial cells in the maintenance and reinstatement of METH-induced conditioned place preference (CPP) in rats. Astrocyte condition medium (ACM) and neuroglia conditioned medium (NCM) are liquid mediums prepared from primary astrocyte and neuroglia cells. These mediums seem to contain many factors that release by glia cells. CPP was induced by systemic administration of METH (1mg/kg for 5days, s.c.). Following the establishment of CPP, the rats were given daily bilateral injections (0.5μl/side) of either vehicle, ACM or NCM into the nucleus accumbens (NAc) and then were tested for the maintenance and reinstatement. Intra-NAc administration of ACM treated with METH, could extend the extinction period and also, intensified the magnitude of METH reinstatement. Furthermore, intra-accumbal administration of NCM treated with METH notably delayed the extinction period by four days and significantly increased the magnitude of CPP score in the reinstatement phase compared to the post-test phase. Collectively, these findings suggested that activation of glial cells may be involved in the maintenance and reinstatement of METH-seeking behaviors. It provides new evidence that glia cells might be considered as a potential target for the treatment of METH addiction.
Collapse
Affiliation(s)
- Reza Arezoomandan
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Addiction Department, Center of Excellence in Psychiatry and Clinical Psychology, School of Behavioral Sciences and Mental Health (Institute of Tehran Psychiatry), Iran University of Medical Sciences, Tehran, Iran
| | - Marzieh Moradi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghassem Attarzadeh-Yazdi
- Molecular Medicine Research Centre, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Iran National Science Foundation, Tehran, Iran
| | - Carlos Tomaz
- Department of Physiological Sciences, University of Brasilia, Brasilia, Brazil
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Iran National Science Foundation, Tehran, Iran.
| |
Collapse
|
33
|
Krasnova IN, Justinova Z, Cadet JL. Methamphetamine addiction: involvement of CREB and neuroinflammatory signaling pathways. Psychopharmacology (Berl) 2016; 233:1945-62. [PMID: 26873080 PMCID: PMC5627363 DOI: 10.1007/s00213-016-4235-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/04/2016] [Indexed: 02/06/2023]
Abstract
RATIONALE AND OBJECTIVES Addiction to psychostimulant methamphetamine (METH) remains a major public health problem in the world. Animal models that use METH self-administration incorporate many features of human drug-taking behavior and are very helpful in elucidating mechanisms underlying METH addiction. These models are also helping to decipher the neurobiological substrates of associated neuropsychiatric complications. This review summarizes our work on the influence of METH self-administration on dopamine systems, transcription and immune responses in the brain. METHODS We used the rat model of METH self-administration with extended access (15 h/day for eight consecutive days) to investigate the effects of voluntary METH intake on the markers of dopamine system integrity and changes in gene expression observed in the brain at 2 h-1 month after cessation of drug exposure. RESULTS Extended access to METH self-administration caused changes in the rat brain that are consistent with clinical findings reported in neuroimaging and postmortem studies of human METH addicts. In addition, gene expression studies using striatal tissues from METH self-administering rats revealed increased expression of genes involved in cAMP response element binding protein (CREB) signaling pathway and in the activation of neuroinflammatory response in the brain. CONCLUSION These data show an association of METH exposure with activation of neuroplastic and neuroinflammatory cascades in the brain. The neuroplastic changes may be involved in promoting METH addiction. Neuroinflammatory processes in the striatum may underlie cognitive deficits, depression, and parkinsonism reported in METH addicts. Therapeutic approaches that include suppression of neuroinflammation may be beneficial to addicted patients.
Collapse
Affiliation(s)
- Irina N. Krasnova
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA,Corresponding authors: Irina N. Krasnova, Ph.D., Molecular Neuropsychiatry Research Branch, NIDA/NIH/DHHS, 251 Bayview Blvd, Baltimore, MD 21224, Tel. 443-74-2658, Fax 443-740-2856, , Jean Lud Cadet, M.D., Molecular Neuropsychiatry Research Branch, NIDA/NIH/DHHS, 251 Bayview Blvd., Baltimore, MD 21224, Tel. 443-740-2656, Fax 443-740-2856,
| | - Zuzana Justinova
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS Baltimore, MD 21224, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA, NIH, DHHS, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| |
Collapse
|
34
|
Mediouni S, Marcondes MCG, Miller C, McLaughlin JP, Valente ST. The cross-talk of HIV-1 Tat and methamphetamine in HIV-associated neurocognitive disorders. Front Microbiol 2015; 6:1164. [PMID: 26557111 PMCID: PMC4615951 DOI: 10.3389/fmicb.2015.01164] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/07/2015] [Indexed: 12/15/2022] Open
Abstract
Antiretroviral therapy has dramatically improved the lives of human immunodeficiency virus 1 (HIV-1) infected individuals. Nonetheless, HIV-associated neurocognitive disorders (HAND), which range from undetectable neurocognitive impairments to severe dementia, still affect approximately 50% of the infected population, hampering their quality of life. The persistence of HAND is promoted by several factors, including longer life expectancies, the residual levels of virus in the central nervous system (CNS) and the continued presence of HIV-1 regulatory proteins such as the transactivator of transcription (Tat) in the brain. Tat is a secreted viral protein that crosses the blood–brain barrier into the CNS, where it has the ability to directly act on neurons and non-neuronal cells alike. These actions result in the release of soluble factors involved in inflammation, oxidative stress and excitotoxicity, ultimately resulting in neuronal damage. The percentage of methamphetamine (MA) abusers is high among the HIV-1-positive population compared to the general population. On the other hand, MA abuse is correlated with increased viral replication, enhanced Tat-mediated neurotoxicity and neurocognitive impairments. Although several strategies have been investigated to reduce HAND and MA use, no clinically approved treatment is currently available. Here, we review the latest findings of the effects of Tat and MA in HAND and discuss a few promising potential therapeutic developments.
Collapse
Affiliation(s)
- Sonia Mediouni
- Department of Infectious Diseases, The Scripps Research Institute , Jupiter, FL, USA
| | | | - Courtney Miller
- Department of Metabolism and Aging, The Scripps Research Institute , Jupiter, FL, USA ; Department of Neuroscience, The Scripps Research Institute , Jupiter, FL, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, University of Florida , Gainesville, FL, USA
| | - Susana T Valente
- Department of Infectious Diseases, The Scripps Research Institute , Jupiter, FL, USA
| |
Collapse
|
35
|
R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry 2015; 5:e632. [PMID: 26327690 PMCID: PMC5068814 DOI: 10.1038/tp.2015.136] [Citation(s) in RCA: 445] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 07/30/2015] [Accepted: 07/30/2015] [Indexed: 01/18/2023] Open
Abstract
Although the efficacy of racemate ketamine, a rapid onset and sustained antidepressant, for patients with treatment-resistant depression was a serendipitous finding, clinical use of ketamine is limited, due to psychotomimetic side effects and abuse liability. Behavioral and side-effect evaluation tests were applied to compare the two stereoisomers of ketamine. To elucidate their potential therapeutic mechanisms, we examined the effects of these stereoisomers on brain-derived neurotrophic factor (BDNF)-TrkB signaling, and synaptogenesis in selected brain regions. In the social defeat stress and learned helplessness models of depression, R-ketamine showed a greater potency and longer-lasting antidepressant effect than S-ketamine (esketamine). Furthermore, R-ketamine induced a more potent beneficial effect on decreased dendritic spine density, BDNF-TrkB signaling and synaptogenesis in the prefrontal cortex (PFC), CA3 and dentate gyrus (DG) of the hippocampus from depressed mice compared with S-ketamine. However, neither stereoisomer affected these alterations in the nucleus accumbens of depressed mice. In behavioral tests for side effects, S-ketamine, but not R-ketamine, precipitated behavioral abnormalities, such as hyperlocomotion, prepulse inhibition deficits and rewarding effects. In addition, a single dose of S-ketamine, but not R-ketamine, caused a loss of parvalbumin (PV)-positive cells in the prelimbic region of the medial PFC and DG. These findings suggest that, unlike S-ketamine, R-ketamine can elicit a sustained antidepressant effect, mediated by increased BDNF-TrkB signaling and synaptogenesis in the PFC, DG and CA3. R-ketamine appears to be a potent, long-lasting and safe antidepressant, relative to S-ketamine, as R-ketamine appears to be free of psychotomimetic side effects and abuse liability.
Collapse
|
36
|
Arezoomandan R, Haghparast A. Administration of the glial cell modulator, minocycline, in the nucleus accumbens attenuated the maintenance and reinstatement of morphine-seeking behavior. Can J Physiol Pharmacol 2015; 94:257-64. [PMID: 26745749 DOI: 10.1139/cjpp-2015-0209] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Relapse to drug use is one of the most difficult clinical problems in treating addiction. Glial activation has been linked with the drug abuse, and the glia modulators such as minocycline can modulate the drug abuse effects. The aim of the present study was to determine whether minocycline could attenuate the maintenance and reinstatement of morphine. Conditioned place preference (CPP) was induced by subcutaneous injection of morphine (5 mg/kg) for 3 days. Following the acquisition of the CPP, the rats were given daily bilateral intra-NAc injections of either minocycline (1, 5, and 10 μg/0.5 μL) or saline (0.5 μL). The animals were tested for conditioning score 60 min after each injection. To induce the reinstatement, a priming dose of morphine (1 mg/kg) was injected 1 day after the final extinction day. The morphine-induced CPP lasted for 7 days after cessation of morphine treatment. Our data revealed that a priming dose of morphine could reinstate the extinguished morphine-induced CPP. Daily intra-accumbal injection of minocycline during the extinction period blocked the maintenance of morphine CPP and also attenuated the priming-induced reinstatement. Our findings indicated that minocycline could facilitate the extinction and attenuate the reinstatement of morphine. These results provided new evidence that minocycline might be considered as a promising therapeutic agent for the treatment of several symptoms associated with morphine abuse.
Collapse
Affiliation(s)
- Reza Arezoomandan
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Mannangatti P, NarasimhaNaidu K, Damaj MI, Ramamoorthy S, Jayanthi LD. A Role for p38 Mitogen-activated Protein Kinase-mediated Threonine 30-dependent Norepinephrine Transporter Regulation in Cocaine Sensitization and Conditioned Place Preference. J Biol Chem 2015; 290:10814-27. [PMID: 25724654 DOI: 10.1074/jbc.m114.612192] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Indexed: 01/08/2023] Open
Abstract
The noradrenergic and p38 mitogen-activated protein kinase (p38 MAPK) systems are implicated in cocaine-elicited behaviors. Previously, we demonstrated a role for p38 MAPK-mediated norepinephrine transporter (NET) Thr(30) phosphorylation in cocaine-induced NET up-regulation (Mannangatti, P., Arapulisamy, O., Shippenberg, T. S., Ramamoorthy, S., and Jayanthi, L. D. (2011) J. Biol. Chem. 286, 20239-20250). The present study explored the functional interaction between p38 MAPK-mediated NET regulation and cocaine-induced behaviors. In vitro cocaine treatment of mouse prefrontal cortex synaptosomes resulted in enhanced NET function, surface expression, and phosphorylation. Pretreatment with PD169316, a p38 MAPK inhibitor, completely blocked cocaine-mediated NET up-regulation and phosphorylation. In mice, in vivo administration of p38 MAPK inhibitor SB203580 completely blocked cocaine-induced NET up-regulation and p38 MAPK activation in the prefrontal cortex and nucleus accumbens. When tested for cocaine-induced locomotor sensitization and conditioned place preference (CPP), mice receiving SB203580 on cocaine challenge day or on postconditioning test day exhibited significantly reduced cocaine sensitization and CPP. A transactivator of transcription (TAT) peptide strategy was utilized to test the involvement of the NET-Thr(30) motif. In vitro treatment of synaptosomes with TAT-NET-Thr(30) (wild-type peptide) completely blocked cocaine-mediated NET up-regulation and phosphorylation. In vivo administration of TAT-NET-Thr(30) peptide but not TAT-NET-T30A (mutant peptide) completely blocked cocaine-mediated NET up-regulation and phosphorylation. In the cocaine CPP paradigm, mice receiving TAT-NET-Thr(30) but not TAT-NET-T30A on postconditioning test day exhibited significantly reduced cocaine CPP. Following extinction, TAT-NET-Thr(30) when given prior to cocaine challenge significantly reduced reinstatement of cocaine CPP. These results demonstrate that the direct inhibition of p38 MAPK or the manipulation of NET-Thr(30) motif/phosphorylation via a TAT peptide strategy prevents cocaine-induced NET up-regulation, locomotor sensitization, and CPP, suggesting a role for Thr(30)-linked NET regulation in cocaine-elicited behaviors.
Collapse
Affiliation(s)
- Padmanabhan Mannangatti
- From the Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298
| | | | - Mohamad Imad Damaj
- From the Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Sammanda Ramamoorthy
- From the Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia 23298
| | | |
Collapse
|
38
|
Minocycline, an antibiotic with inhibitory effect on microglial activation, attenuates the maintenance and reinstatement of methamphetamine-seeking behavior in rat. Prog Neuropsychopharmacol Biol Psychiatry 2014; 53:142-8. [PMID: 24768984 DOI: 10.1016/j.pnpbp.2014.04.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 03/31/2014] [Accepted: 04/11/2014] [Indexed: 01/01/2023]
Abstract
Methamphetamine (METH) is a major criminal justice and public health problem. Repeated use of METH causes dependence in humans and there are currently no particular pharmacological treatments for METH addiction. Glial cell activation is linked with METH abuse and METH administration causes activation of these cells in many areas of the brain. Many studies have demonstrated that glial cell modulators can modulate drug abuse effects. In this study, we examined the effect of the putative microglial inhibitor, minocycline on maintenance and prime-induced reinstatement of METH seeking behavior using the conditioned place preference (CPP) paradigm. CPP induced with METH (1 mg/kg, i.p. for 3 days) lasted for 11 days after cessation of METH treatment and priming dose of METH (0.5 mg/kg, i.p.) reinstated the extinguished METH-induced CPP. Daily treatment of minocycline (40 mg/kg, i.p.) followed by establishment of CPP blocked the maintenance of METH-induced CPP and also could attenuate priming-induced reinstatement. Furthermore, daily bilateral intra-accumbal injection of minocycline (10 and 20 μg/0.5 μl saline), during extinction period blocked the maintenance of METH CPP but just the highest dose of that could attenuate priming-induced reinstatement. We showed that minocycline administration during extinction period could facilitate extinction and maybe abolish the ability of drug-related cues evoke reinstatement, suggesting that minocycline might be considered as a promising therapeutic agent in preventing relapse in METH dependent individuals.
Collapse
|
39
|
Wakida N, Kiguchi N, Saika F, Nishiue H, Kobayashi Y, Kishioka S. CC-chemokine ligand 2 facilitates conditioned place preference to methamphetamine through the activation of dopamine systems. J Pharmacol Sci 2014; 125:68-73. [PMID: 24748435 DOI: 10.1254/jphs.14032fp] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Methamphetamine addiction is characterized by drug craving caused by stimulation of the reward system. Because neuroinflammation underlies several neurological disorders, we investigated whether CC-chemokine ligand 2 (CCL2) participates in the methamphetamine dependence using mice. Upregulation of CCL2 but not CC-chemokine receptor 2 (CCR2), a dominant receptor for CCL2, mRNA in both the prefrontal cortex (PFC) and nucleus accumbens (NAC) was observed after methamphetamine (3 mg/kg, s.c.) administration. Using immunohistochemistry, high CCL2 protein levels localized to neurons in the PFC and NAC. In the conditioned place preference (CPP) test, methamphetamine (0.3 - 3 mg/kg, s.c.) induced a CPP, reflecting psychic dependence on methamphetamine, in a dose-dependent manner. The CPP to methamphetamine was attenuated by RS504393 (1 mg/kg, s.c.), a CCR2 antagonist. Moreover, methamphetamine increased phosphorylated tyrosine hydroxylase (pTH) levels in the ventral tegmental area (VTA). Increased levels of pTH in the VTA by methamphetamine was also suppressed by RS504393. Furthermore, intracerebroventricular injection of recombinant CCL2 increased pTH levels in the VTA. Taken together, we demonstrate that activation of dopamine neurons, which enhances reward-system activity, via the CCL2-CCR2 axis plays a crucial role in psychic dependence on methamphetamine. Novel treatments targeting this machinery may be effective for drug addiction.
Collapse
Affiliation(s)
- Naoki Wakida
- Department of Pharmacology, Wakayama Medical University, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Ray LA, Roche DJO, Heinzerling K, Shoptaw S. Opportunities for the development of neuroimmune therapies in addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 118:381-401. [PMID: 25175870 DOI: 10.1016/b978-0-12-801284-0.00012-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Studies have implicated neuroinflammatory processes in the pathophysiology of various psychiatric conditions, including addictive disorders. Neuroimmune signaling represents an important and relatively poorly understood biological process in drug addiction. The objective of this review is to update the field on recent developments in neuroimmune therapies for addiction. First, we review studies of neuroinflammation in relation to alcohol and methamphetamine dependence followed by a section on neuroinflammation and accompanying neurocognitive dysfunction in HIV infection and concomitant substance abuse. Second, we provide a review of pharmacotherapies with neuroimmune properties and their potential development for the treatment of addictions. Pharmacotherapies covered in this review include ibudilast, minocycline, doxycycline, topiramate, indomethacin, rolipram, anakinra (IL-1Ra), peroxisome proliferator-activated receptor agonists, naltrexone, and naloxone. Lastly, summary and future directions are provided with recommendations for how to efficiently translate preclinical findings into clinical studies that can ultimately lead to novel and more effective pharmacotherapies for addiction.
Collapse
Affiliation(s)
- Lara A Ray
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA.
| | - Daniel J O Roche
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
| | - Keith Heinzerling
- Department of Family Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Steve Shoptaw
- Department of Family Medicine, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
41
|
Beardsley PM, Hauser KF. Glial modulators as potential treatments of psychostimulant abuse. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:1-69. [PMID: 24484974 DOI: 10.1016/b978-0-12-420118-7.00001-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glia (including astrocytes, microglia, and oligodendrocytes), which constitute the majority of cells in the brain, have many of the same receptors as neurons, secrete neurotransmitters and neurotrophic and neuroinflammatory factors, control clearance of neurotransmitters from synaptic clefts, and are intimately involved in synaptic plasticity. Despite their prevalence and spectrum of functions, appreciation of their potential general importance has been elusive since their identification in the mid-1800s, and only relatively recently have they been gaining their due respect. This development of appreciation has been nurtured by the growing awareness that drugs of abuse, including the psychostimulants, affect glial activity, and glial activity, in turn, has been found to modulate the effects of the psychostimulants. This developing awareness has begun to illuminate novel pharmacotherapeutic targets for treating psychostimulant abuse, for which targeting more conventional neuronal targets has not yet resulted in a single, approved medication. In this chapter, we discuss the molecular pharmacology, physiology, and functional relationships that the glia have especially in the light in which they present themselves as targets for pharmacotherapeutics intended to treat psychostimulant abuse disorders. We then review a cross section of preclinical studies that have manipulated glial processes whose behavioral effects have been supportive of considering the glia as drug targets for psychostimulant-abuse medications. We then close with comments regarding the current clinical evaluation of relevant compounds for treating psychostimulant abuse, as well as the likelihood of future prospects.
Collapse
Affiliation(s)
| | - Kurt F Hauser
- Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
42
|
Ren Q, Zhang JC, Fujita Y, Ma M, Wu J, Hashimoto K. Effects of TrkB agonist 7,8-dihydroxyflavone on sensory gating deficits in mice after administration of methamphetamine. Pharmacol Biochem Behav 2013; 106:124-7. [PMID: 23567202 DOI: 10.1016/j.pbb.2013.03.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 03/21/2013] [Accepted: 03/29/2013] [Indexed: 12/31/2022]
Abstract
Several lines of evidence suggest that the brain-derived neurotrophic factor (BDNF)-tropomyosin-related kinase B (TrkB) signaling pathway plays a role in behavioral abnormalities observed after administration of psychostimulants, such as methamphetamine (METH). This study was undertaken to examine whether the potent TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF) could improve prepulse inhibition (PPI) deficits in mice seen after a single dose of METH. Treatment with 7,8-DHF (3.0, 10 or 30 mg/kg) improved PPI deficits in mice associated with exposure to METH (3.0 mg/kg), in a dose dependent manner. Furthermore, co-administration of ANA-12 (0.5 mg/kg), a TrkB antagonist, significantly blocked the effects of 7,8-DHF (30 mg/kg) on METH-induced PPI deficits. In contrast, administration of 5,7-dihydroxyflavone (5,7-DHF: 30 mg/kg), an inactive TrkB ligand, did not affect METH-induced PPI deficits in mice. An in vivo microdialysis study in conscious mice showed that 7,8-DHF (30 mg/kg) significantly attenuated increased dopamine release in the striatum, after METH administration (3 mg/kg). This study suggests that 7,8-DHF can improve PPI deficits in these mice, through the inhibition of METH-induced dopamine release. Therefore, it is likely that TrkB agonists, such as 7,8-DHF, may constitute a novel class of therapeutic drugs for neuropsychiatric diseases such as METH-use disorder and schizophrenia.
Collapse
Affiliation(s)
- Qian Ren
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | | | | | | | | | | |
Collapse
|
43
|
Snider SE, Hendrick ES, Beardsley PM. Glial cell modulators attenuate methamphetamine self-administration in the rat. Eur J Pharmacol 2013; 701:124-30. [PMID: 23375937 DOI: 10.1016/j.ejphar.2013.01.016] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 12/17/2012] [Accepted: 01/16/2013] [Indexed: 12/11/2022]
Abstract
Neuroinflammation induced by activated microglia and astrocytes can be elicited by drugs of abuse. Methamphetamine administration activates glial cells and increases proinflammatory cytokine production, and there is recent evidence of a linkage between glial cell activation and drug abuse-related behavior. We have previously reported that ibudilast (AV411; 3-isobutyryl-2-isopropylpyrazolo-[1,5-a]pyridine), which inhibits phosphodiesterase (PDE) and pro-inflammatory activity, blocks reinstatement of methamphetamine-maintained responding in rats, and that ibudilast and AV1013, an amino analog of ibudilast, which has similar glial-attenuating properties but limited PDE activity, attenuate methamphetamine-induced locomotor activity and sensitization in mice. The present study's objective was to determine whether co-administered ibudilast, AV1013, or minocycline, which is a tetracycline derivative that also suppresses methamphetamine-induced glial activation, would attenuate active methamphetamine i.v. self-administration in Long-Evans hooded rats. Rats were initially trained to press a lever for 0.1mg/kg/inf methamphetamine according to a FR1 schedule during 2-h daily sessions. Once stable responding was obtained, twice daily ibudilast (1, 7.5, 10mg/kg), AV1013 (1, 10, 30mg/kg), or once daily minocycline (10, 30, 60mg/kg), or their corresponding vehicles, were given i.p. for three consecutive days during methamphetamine (0.001, 0.03, 0.1mg/kg/inf) self-administration. Ibudilast, AV1013, and minocycline all significantly (p<0.05) reduced responding maintained by 0.03mg/kg/inf methamphetamine that had maintained the highest level of infusions under vehicle conditions. These results suggest that targeting glial cells may provide a novel approach to pharmacotherapy for treating methamphetamineabuse.
Collapse
Affiliation(s)
- Sarah E Snider
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | | | | |
Collapse
|
44
|
What is behind the non-antibiotic properties of minocycline? Pharmacol Res 2012; 67:18-30. [PMID: 23085382 DOI: 10.1016/j.phrs.2012.10.006] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 09/13/2012] [Accepted: 10/09/2012] [Indexed: 11/24/2022]
Abstract
Minocycline is a second-generation, semi-synthetic tetracycline that has been in use in therapy for over 30 years for its antibiotic properties against both Gram-positive and Gram-negative bacteria. It displays antibiotic activity due to its ability to bind to the 30S ribosomal subunit of bacteria and thus inhibit protein synthesis. More recently, it has been described to exert a variety of biological actions beyond its antimicrobial activity, including anti-inflammatory and anti-apoptotic activities, inhibition of proteolysis, as well as suppression of angiogenesis and tumor metastasis, which have been confirmed in different experimental models of non-infectious diseases. There are also many studies that have focused on the mechanisms involved in these non-antibiotic properties of minocycline, including anti-oxidant activity, inhibition of several enzyme activities, inhibition of apoptosis and regulation of immune cell activation and proliferation. This review summarizes the current findings in this topic, mainly focusing on the mechanisms underlying the immunomodulatory and anti-inflammatory activities of minocycline.
Collapse
|
45
|
Hutchinson MR, Northcutt AL, Hiranita T, Wang X, Lewis SS, Thomas J, van Steeg K, Kopajtic TA, Loram LC, Sfregola C, Galer E, Miles NE, Bland ST, Amat J, Rozeske RR, Maslanik T, Chapman TR, Strand KA, Fleshner M, Bachtell RK, Somogyi AA, Yin H, Katz JL, Rice KC, Maier SF, Watkins LR. Opioid activation of toll-like receptor 4 contributes to drug reinforcement. J Neurosci 2012; 32:11187-200. [PMID: 22895704 PMCID: PMC3454463 DOI: 10.1523/jneurosci.0684-12.2012] [Citation(s) in RCA: 231] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Revised: 06/14/2012] [Accepted: 06/25/2012] [Indexed: 01/08/2023] Open
Abstract
Opioid action was thought to exert reinforcing effects solely via the initial agonism of opioid receptors. Here, we present evidence for an additional novel contributor to opioid reward: the innate immune pattern-recognition receptor, toll-like receptor 4 (TLR4), and its MyD88-dependent signaling. Blockade of TLR4/MD2 by administration of the nonopioid, unnatural isomer of naloxone, (+)-naloxone (rats), or two independent genetic knock-outs of MyD88-TLR4-dependent signaling (mice), suppressed opioid-induced conditioned place preference. (+)-Naloxone also reduced opioid (remifentanil) self-administration (rats), another commonly used behavioral measure of drug reward. Moreover, pharmacological blockade of morphine-TLR4/MD2 activity potently reduced morphine-induced elevations of extracellular dopamine in rat nucleus accumbens, a region critical for opioid reinforcement. Importantly, opioid-TLR4 actions are not a unidirectional influence on opioid pharmacodynamics, since TLR4(-/-) mice had reduced oxycodone-induced p38 and JNK phosphorylation, while displaying potentiated analgesia. Similar to our recent reports of morphine-TLR4/MD2 binding, here we provide a combination of in silico and biophysical data to support (+)-naloxone and remifentanil binding to TLR4/MD2. Collectively, these data indicate that the actions of opioids at classical opioid receptors, together with their newly identified TLR4/MD2 actions, affect the mesolimbic dopamine system that amplifies opioid-induced elevations in extracellular dopamine levels, therefore possibly explaining altered opioid reward behaviors. Thus, the discovery of TLR4/MD2 recognition of opioids as foreign xenobiotic substances adds to the existing hypothesized neuronal reinforcement mechanisms, identifies a new drug target in TLR4/MD2 for the treatment of addictions, and provides further evidence supporting a role for central proinflammatory immune signaling in drug reward.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/blood
- Analysis of Variance
- Animals
- Conditioning, Operant/drug effects
- Conditioning, Operant/physiology
- Dopamine/metabolism
- Dose-Response Relationship, Drug
- Drug Administration Routes
- Hyperalgesia/drug therapy
- Hyperalgesia/physiopathology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Microdialysis
- Mitogen-Activated Protein Kinase 1/metabolism
- Models, Molecular
- Myeloid Differentiation Factor 88/deficiency
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Nucleus Accumbens/drug effects
- Nucleus Accumbens/metabolism
- Pain Threshold/drug effects
- Pain Threshold/physiology
- Phosphorylation/drug effects
- Protein Binding/drug effects
- Protein Binding/genetics
- Rats
- Rats, Sprague-Dawley
- Reaction Time/drug effects
- Reinforcement, Psychology
- Self Administration
- Signal Transduction/drug effects
- Time Factors
- Toll-Like Receptor 4/agonists
- Toll-Like Receptor 4/deficiency
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
- M R Hutchinson
- Department of Psychology and Neuroscience, University of Colorado-Boulder, Boulder, Colorado 80309, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Horio M, Kohno M, Fujita Y, Ishima T, Inoue R, Mori H, Hashimoto K. Role of serine racemase in behavioral sensitization in mice after repeated administration of methamphetamine. PLoS One 2012; 7:e35494. [PMID: 22530033 PMCID: PMC3329469 DOI: 10.1371/journal.pone.0035494] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 03/20/2012] [Indexed: 02/07/2023] Open
Abstract
Background The N-methyl-D-aspartate (NMDA) receptors play a role in behavioral abnormalities observed after administration of the psychostimulant, methamphetamine (METH). Serine racemase (SRR) is an enzyme which synthesizes D-serine, an endogenous co-agonist of NMDA receptors. Using Srr knock-out (KO) mice, we investigated the role of SRR on METH-induced behavioral abnormalities in mice. Methodology/Principal Findings Evaluations of behavior in acute hyperlocomotion, behavioral sensitization, and conditioned place preference (CPP) were performed. The role of SRR on the release of dopamine (DA) in the nucleus accumbens after administration of METH was examined using in vivo microdialysis technique. Additionally, phosphorylation levels of ERK1/2 proteins in the striatum, frontal cortex and hippocampus were examined using Western blot analysis. Acute hyperlocomotion after a single administration of METH (3 mg/kg) was comparable between wild-type (WT) and Srr-KO mice. However, repeated administration of METH (3 mg/kg/day, once daily for 5 days) resulted in behavioral sensitization in WT, but not Srr-KO mice. Pretreatment with D-serine (900 mg/kg, 30 min prior to each METH treatment) did not affect the development of behavioral sensitization after repeated METH administration. In the CPP paradigm, METH-induced rewarding effects were demonstrable in both WT and Srr-KO mice. In vivo microdialysis study showed that METH (1 mg/kg)-induced DA release in the nucleus accumbens of Srr-KO mice previously treated with METH was significantly lower than that of the WT mice previously treated with METH. Interestingly, a single administration of METH (3 mg/kg) significantly increased the phosphorylation status of ERK1/2 in the striatum of WT, but not Srr-KO mice. Conclusions/Significance These findings suggest first, that SRR plays a role in the development of behavioral sensitization in mice after repeated administration of METH, and second that phosphorylation of ERK1/2 by METH may contribute to the development of this sensitization as seen in WT but not Srr-KO mice.
Collapse
Affiliation(s)
- Mao Horio
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Mami Kohno
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Yuko Fujita
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Tamaki Ishima
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Ran Inoue
- Department of Molecular Neuroscience, Toyama University Graduate School of Medicine, Toyama, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Toyama University Graduate School of Medicine, Toyama, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
- * E-mail:
| |
Collapse
|