1
|
Wang L, Wang T, Lei Y, Su Y, Lin Y, Wu Z, Wu Q, Zhang S, Wang H. Unraveling the mechanisms of propofol-induced psychological dependence: a multi-omics approach linked to gut microbiota in hippocampal function. Front Med (Lausanne) 2025; 12:1539467. [PMID: 40248078 PMCID: PMC12005058 DOI: 10.3389/fmed.2025.1539467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Introduction Drug abuse is becoming a global public health crisis. According to the United Nations, the number of drug users worldwide has increased dramatically over the past decade, with a surge in the number of drug abusers. The problem was exacerbated by the expanding market for illicit drugs and the increasing availability of synthetic drugs such as fentanyl. Clinical drug abuse is a problem that requires particular attention, and the potential addictive properties of some drugs and their mechanisms of action are currently unknown, which limits the development and implementation of drug addiction intervention strategies. Methods Eight-week-old C57BL/6J mice were used as study subjects. A mental dependence model was established using the conditional position preference experiment (CPP), and the hippocampal tissues of the model mice were subjected to RNA-seq transcriptome sequencing, LC-MS non-targeted metabolome sequencing, and intestinal macro-genome sequencing in order to discover propofol mental dependence signature genes. Correlation analyses of transcriptomics and metabolomics were performed using the Spearman method, and gene-metabolite networks were mapped using Cytoscape software. Real-time fluorescence quantitative PCR and immunoprotein blotting (Western blotting) methods were used to validate the characterized genes. Results After the conditioned position preference experiment, the conditioned preference scores of the 75 mg/kg propofol and 2 g/kg alcohol groups were significantly higher than those of the control saline group. 152 differential genes and 214 differential metabolites were identified in the 75 mg/kg group. Cluster analysis revealed that changes in the neuroactive ligand receptor pathway were most pronounced. Gut microbiomics assays revealed significant changes in five differential enterobacterial phyla (Campylobacter phylum, Thick-walled phylum, Anaplasma phylum, Actinobacteria phylum, and Chlorella verticillata phylum) in the 75 mg/kg propofol group, which may be related to changes in the differential expression of dopamine. Discussion These findings suggest that 75 mg/kg propofol has a significant mind-dependent effect on the biology of drug addiction through neuroactive ligand-receptor interaction pathways in conjunction with the tricarboxylic acid cycle, and the metabolic pathways of alanine, aspartate, and glutamate that may influence intestinal microbial changes through bidirectional signaling.
Collapse
Affiliation(s)
- Li Wang
- Department of Basic Medical Sciences, Qinghai University Medical College, Xining, Qinghai, China
| | - Tangyi Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China
| | - Yadian Lei
- Department of Basic Medical Sciences, Qinghai University Medical College, Xining, Qinghai, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China
| | - Yudong Su
- Department of Basic Medical Sciences, Qinghai University Medical College, Xining, Qinghai, China
| | - Yuxin Lin
- Department of Basic Medical Sciences, Qinghai University Medical College, Xining, Qinghai, China
| | - Zhijing Wu
- Department of Basic Medical Sciences, Qinghai University Medical College, Xining, Qinghai, China
| | - Qiong Wu
- Department of Basic Medical Sciences, Qinghai University Medical College, Xining, Qinghai, China
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining, Qinghai, China
| | - Shoude Zhang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China
| | - Haiyan Wang
- Department of Basic Medical Sciences, Qinghai University Medical College, Xining, Qinghai, China
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai, China
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining, Qinghai, China
| |
Collapse
|
2
|
Serafini RA, Farzinpour Z, Patel V, Kelley AM, Estill M, Pryce KD, Sakloth F, Teague CD, Torres-Berrio A, Nestler EJ, Shen L, Akbarian S, Karkhanis AN, Blitzer RD, Zachariou V. Nucleus accumbens myocyte enhancer factor 2C mediates the maintenance of peripheral nerve injury-induced physiological and behavioral maladaptations. Pain 2024; 165:2733-2748. [PMID: 38985454 DOI: 10.1097/j.pain.0000000000003316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 04/18/2024] [Indexed: 07/11/2024]
Abstract
ABSTRACT Preclinical and clinical work has demonstrated altered plasticity and activity in the nucleus accumbens (NAc) under chronic pain states, highlighting critical therapeutic avenues for the management of chronic pain conditions. In this study, we demonstrate that myocyte enhancer factor 2C (MEF2C), a master regulator of neuronal activity and plasticity, is repressed in NAc neurons after prolonged spared nerve injury (SNI). Viral-mediated overexpression of Mef2c in NAc neurons partially ameliorated sensory hypersensitivity and emotional behaviors in mice with SNI, while also altering transcriptional pathways associated with synaptic signaling. Mef2c overexpression also reversed SNI-induced potentiation of phasic dopamine release and neuronal hyperexcitability in the NAc. Transcriptional changes induced by Mef2c overexpression were different than those observed after desipramine treatment, suggesting a mechanism of action different from antidepressants. Overall, we show that interventions in MEF2C-regulated mechanisms in the NAc are sufficient to disrupt the maintenance of chronic pain states, providing potential new treatment avenues for neuropathic pain.
Collapse
Affiliation(s)
- Randal A Serafini
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmacology, Physiology, & Biophysics, Chobanian and Avedisian School of Medicine at Boston University, Boston, MA, United States
| | - Zahra Farzinpour
- Department of Pharmacology, Physiology, & Biophysics, Chobanian and Avedisian School of Medicine at Boston University, Boston, MA, United States
| | - Vishwendra Patel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Abigail M Kelley
- Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| | - Molly Estill
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kerri D Pryce
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Farhana Sakloth
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Collin D Teague
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Angelica Torres-Berrio
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Schahram Akbarian
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Anushree N Karkhanis
- Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| | - Robert D Blitzer
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Venetia Zachariou
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmacology, Physiology, & Biophysics, Chobanian and Avedisian School of Medicine at Boston University, Boston, MA, United States
| |
Collapse
|
3
|
Avila-Zozaya M, Zachariou V. Genetic mouse models in opioid research: current status and future directions. J Neural Transm (Vienna) 2024; 131:491-494. [PMID: 38436758 DOI: 10.1007/s00702-024-02762-6] [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: 09/17/2023] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
Synthetic and semi-synthetic opioids are prescribed for the management of severe pain conditions, but their long-term use is often leading to physical dependence and addiction disorders. Understanding the complex neurobiology of the opioid system in preclinical models will be essential for the development of safe and efficacious analgesics. With rising numbers of synthetic opioid users and overdose cases, a better understanding of the neuroanatomical and cellular pathways associated with physical dependence and addiction is expected to guide treatment approaches for opioid use disorders. In this commentary, we highlight the importance of advanced genetic mouse models for studying the regional effects of opioid receptors, and we discuss the need of genetic mouse models for the investigation of the regional, circuit and cell compartment-specific role of intracellular mediators of opioid actions.
Collapse
Affiliation(s)
- Monserrat Avila-Zozaya
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Venetia Zachariou
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
4
|
Barrett JE, Shekarabi A, Inan S. Oxycodone: A Current Perspective on Its Pharmacology, Abuse, and Pharmacotherapeutic Developments. Pharmacol Rev 2023; 75:1062-1118. [PMID: 37321860 PMCID: PMC10595024 DOI: 10.1124/pharmrev.121.000506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/30/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023] Open
Abstract
Oxycodone, a semisynthetic derivative of naturally occurring thebaine, an opioid alkaloid, has been available for more than 100 years. Although thebaine cannot be used therapeutically due to the occurrence of convulsions at higher doses, it has been converted to a number of other widely used compounds that include naloxone, naltrexone, buprenorphine, and oxycodone. Despite the early identification of oxycodone, it was not until the 1990s that clinical studies began to explore its analgesic efficacy. These studies were followed by the pursuit of several preclinical studies to examine the analgesic effects and abuse liability of oxycodone in laboratory animals and the subjective effects in human volunteers. For a number of years oxycodone was at the forefront of the opioid crisis, playing a significant role in contributing to opioid misuse and abuse, with suggestions that it led to transitioning to other opioids. Several concerns were expressed as early as the 1940s that oxycodone had significant abuse potential similar to heroin and morphine. Both animal and human abuse liability studies have confirmed, and in some cases amplified, these early warnings. Despite sharing a similar structure with morphine and pharmacological actions also mediated by the μ-opioid receptor, there are several differences in the pharmacology and neurobiology of oxycodone. The data that have emerged from the many efforts to analyze the pharmacological and molecular mechanism of oxycodone have generated considerable insight into its many actions, reviewed here, which, in turn, have provided new information on opioid receptor pharmacology. SIGNIFICANCE STATEMENT: Oxycodone, a μ-opioid receptor agonist, was synthesized in 1916 and introduced into clinical use in Germany in 1917. It has been studied extensively as a therapeutic analgesic for acute and chronic neuropathic pain as an alternative to morphine. Oxycodone emerged as a drug with widespread abuse. This article brings together an integrated, detailed review of the pharmacology of oxycodone, preclinical and clinical studies of pain and abuse, and recent advances to identify potential opioid analgesics without abuse liability.
Collapse
Affiliation(s)
- James E Barrett
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University. Philadelphia, Pennsylvania
| | - Aryan Shekarabi
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University. Philadelphia, Pennsylvania
| | - Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University. Philadelphia, Pennsylvania
| |
Collapse
|
5
|
Serafini RA, Estill M, Pekarskaya EA, Sakloth F, Shen L, Javitch JA, Zachariou V. Tianeptine promotes lasting antiallodynic effects in a mouse model of neuropathic pain. Neuropsychopharmacology 2023; 48:1680-1689. [PMID: 37474762 PMCID: PMC10517169 DOI: 10.1038/s41386-023-01645-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/31/2023] [Accepted: 06/27/2023] [Indexed: 07/22/2023]
Abstract
Tricyclic antidepressants (TCAs), such as desipramine (DMI), are effective at managing neuropathic pain symptoms but often take several weeks to become effective and also lead to considerable side effects. Tianeptine (TIAN) is an atypical antidepressant that activates the mu-opioid receptor but does not produce analgesic tolerance or withdrawal in mice, nor euphoria in humans, at clinically-relevant doses. Here, we evaluate the efficacy of TIAN at persistently alleviating mechanical allodynia in the spared nerve injury (SNI) model of neuropathic pain, even well after drug clearance. After finding an accelerated onset of antiallodynic action compared to DMI, we used genetically modified mice to gain insight into RGS protein-associated pathways that modulate the efficacy of TIAN relative to DMI in models of neuropathic pain. Because we observed similar behavioral responses to both TIAN and DMI treatment in RGS4, RGSz1, and RGS9 knockout mice, we performed RNA sequencing on the NAc of TIAN- and DMI-treated mice after prolonged SNI to further clarify potential mechanisms underlying TIANs faster therapeutic actions. Our bioinformatic analysis revealed distinct transcriptomic signatures between the two drugs, with TIAN more directly reversing SNI-induced differentially expressed genes, and further predicted several upstream regulators that may be implicated in onset of action. This new understanding of the molecular pathways underlying TIAN action may enable the development of novel and more efficacious pharmacological approaches for the management of neuropathic pain.
Collapse
Affiliation(s)
- Randal A Serafini
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacology, Physiology and Biophysics, Chobanian & Avedisian School of Medicine at Boston University, Boston, MA, USA
| | - Molly Estill
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elizabeth A Pekarskaya
- Department of Neuroscience, Columbia University, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Farhana Sakloth
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | - Venetia Zachariou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pharmacology, Physiology and Biophysics, Chobanian & Avedisian School of Medicine at Boston University, Boston, MA, USA.
| |
Collapse
|
6
|
Wang Y, Hou X, Wei S, Yan J, Chen Z, Zhang M, Zhang Q, Lu Y, Zhang Q, Zheng T, Jia J, Dong B, Li Y, Zhang Y, Liang J, Li G. The roles of ubiquitin-proteasome system and regulator of G protein signaling 4 in behavioral sensitization induced by a single morphine exposure. Brain Behav 2023; 13:e2922. [PMID: 36793204 PMCID: PMC10013946 DOI: 10.1002/brb3.2922] [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: 11/22/2022] [Revised: 12/23/2022] [Accepted: 02/04/2023] [Indexed: 02/17/2023] Open
Abstract
AIMS Opioid addiction is a major public health issue, yet its underlying mechanism is still unknown. The aim of this study was to explore the roles of ubiquitin-proteasome system (UPS) and regulator of G protein signaling 4 (RGS4) in morphine-induced behavioral sensitization, a well-recognized animal model of opioid addiction. METHODS We explored the characteristics of RGS4 protein expression and polyubiquitination in the development of behavioral sensitization induced by a single morphine exposure in rats, and the effect of a selective proteasome inhibitor, lactacystin (LAC), on behavioral sensitization. RESULTS Polyubiquitination expression was increased in time-dependent and dose-related fashions during the development of behavioral sensitization, while RGS4 protein expression was not significantly changed during this phase. Stereotaxic administration of LAC into nucleus accumbens (NAc) core inhibited the establishment of behavioral sensitization. CONCLUSION UPS in NAc core is positively involved in behavioral sensitization induced by a single morphine exposure in rats. Polyubiquitination was observed during the development phase of behavioral sensitization, while RGS4 protein expression was not significantly changed, indicating that other members of RGS family might be substrate proteins in UPS-mediated behavioral sensitization.
Collapse
Affiliation(s)
- Yanting Wang
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xingzi Hou
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical SciencesPeking University Health Science CenterBeijingChina
| | - Shoupeng Wei
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMarylandUSA
| | - Jiaqing Yan
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhe Chen
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Mingyu Zhang
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qingying Zhang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical SciencesPeking University Health Science CenterBeijingChina
| | - Yingyuan Lu
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Natural Medicines, School of Pharmaceutical SciencesPeking University Health Science CenterBeijingChina
| | - Qingjie Zhang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Tiange Zheng
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Jingyi Jia
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Bin Dong
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ying Li
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yuanyuan Zhang
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jianhui Liang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Guohui Li
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
7
|
Guan Q, Velho RV, Sehouli J, Mechsner S. Endometriosis and Opioid Receptors: Are Opioids a Possible/Promising Treatment for Endometriosis? Int J Mol Sci 2023; 24:ijms24021633. [PMID: 36675147 PMCID: PMC9864914 DOI: 10.3390/ijms24021633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/17/2023] Open
Abstract
Endometriosis (EM), defined as the presence of endometrial-like tissue with surrounding smooth muscle cells outside the uterus, is a disregarded gynecological disease reported to affect 6-10% of women of reproductive age, with 30-50% of them suffering from chronic pelvic pain and infertility. Since the exact pathogenic mechanisms of EM are still unclear, no curative therapy is available. As pain is an important factor in EM, optimal analgesia should be sought, which to date has been treated primarily with non-steroidal anti-inflammatory drugs (NSAIDs), metamizole or, in extreme cases, opioids. Here, we review the pain therapy options, the mechanisms of pain development in EM, the endogenous opioid system and pain, as well as the opioid receptors and EM-associated pain. We also explore the drug abuse and addiction to opioids and the possible use of NOP receptors in terms of analgesia and improved tolerability as a target for EM-associated pain treatment. Emerging evidence has shown a promising functional profile of bifunctional NOP/MOP partial agonists as safe and nonaddictive analgesics. However, until now, the role of NOP receptors in EM has not been investigated. This review offers a thought which still needs further investigation but may provide potential options for relieving EM-associated pain.
Collapse
|
8
|
Co-Expression of Adaptor Protein FAM159B with Different Markers for Neuroendocrine Cells: An Immunocytochemical and Immunohistochemical Study. Int J Mol Sci 2022; 23:ijms232113503. [DOI: 10.3390/ijms232113503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/23/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022] Open
Abstract
Little is known about the adaptor protein FAM159B. Recently, FAM159B was shown to be particularly expressed in neuroendocrine cells and tissues, such as pancreatic islets and neuroendocrine cells of the bronchopulmonary and gastrointestinal tracts, as well as in different types of neuroendocrine tumours. To gain insights into possible interactions of FAM159B with other proteins and/or receptors, we analysed the co-expression of FAM159B and various neuroendocrine-specific markers in the cancer cell lines BON-1, PC-3, NCI-h82, OH-1, and A431 and also in human pancreatic tissues and pancreatic neuroendocrine tumours. The markers included prominent markers of neuroendocrine differentiation, such as chromogranin A (CgA), neuron-specific enolase (NSE), synaptophysin (SYP), insulinoma-associated protein 1 (INSM1), neural cell adhesion molecule 1 (NCAM1), serotonin (5-HT), somatostatin-14/28 (SST), and several receptors that are typically expressed by neuroendocrine cells, such as dopamine receptor 2 (D2R), somatostatin receptor (SSTR) 1, 2, 3, 4 and 5, and regulator of G-protein signalling 9 (RGS9). FAM159B was expressed evenly throughout the cytosol in all five cancer cell lines. Immunocytochemical and immunohistochemical analyses revealed co-expression of FAM159B with SYP, INSM1, RGS9, D2R, SSTR2, SSTR3, SSTR4, and SSTR5 and strong overlapping co-localisation with NSE. Double-labelling and co-immunoprecipitation Western blot analyses confirmed a direct association between FAM159B and NSE. These results suggest the involvement of FAM159B in several intracellular signalling pathways and a direct or indirect influence on diverse membrane proteins and receptors.
Collapse
|
9
|
Single nucleus transcriptomic analysis of rat nucleus accumbens reveals cell type-specific patterns of gene expression associated with volitional morphine intake. Transl Psychiatry 2022; 12:374. [PMID: 36075888 PMCID: PMC9458645 DOI: 10.1038/s41398-022-02135-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
Opioid exposure is known to cause transcriptomic changes in the nucleus accumbens (NAc). However, no studies to date have investigated cell type-specific transcriptomic changes associated with volitional opioid taking. Here, we use single nucleus RNA sequencing (snRNAseq) to comprehensively characterize cell type-specific alterations of the NAc transcriptome in rats self-administering morphine. One cohort of male Brown Norway rats was injected with acute morphine (10 mg/kg, i.p.) or saline. A second cohort of rats was allowed to self-administer intravenous morphine (1.0 mg/kg/infusion) for 10 consecutive days. Each morphine-experienced rat was paired with a yoked saline control rat. snRNAseq libraries were generated from NAc punches and used to identify cell type-specific gene expression changes associated with volitional morphine taking. We identified 1106 differentially expressed genes (DEGs) in the acute morphine group, compared to 2453 DEGs in the morphine self-administration group, across 27 distinct cell clusters. Importantly, we identified 1329 DEGs that were specific to morphine self-administration. DEGs were identified in novel clusters of astrocytes, oligodendrocytes, and D1R- and D2R-expressing medium spiny neurons in the NAc. Cell type-specific DEGs included Rgs9, Celf5, Oprm1, and Pde10a. Upregulation of Rgs9 and Celf5 in D2R-expressing neurons was validated by RNAscope. Approximately 85% of all oligodendrocyte DEGs, nearly all of which were associated with morphine taking, were identified in two subtypes. Bioinformatic analyses identified cell type-specific upstream regulatory mechanisms of the observed transcriptome alterations and downstream signaling pathways, including both novel and previously identified molecular pathways. These findings show that volitional morphine taking is associated with distinct cell type-specific transcriptomic changes in the rat NAc and highlight specific striatal cell populations and novel molecular substrates that could be targeted to reduce compulsive opioid taking.
Collapse
|
10
|
Sadat‐Shirazi M, Nouri Zadeh‐Tehrani S, Akbarabadi A, Mokri A, Taleb Zadeh Kasgari B, Zarrindast M. Exercise can restore behavioural and molecular changes of intergenerational morphine effects. Addict Biol 2022; 27:e13122. [PMID: 34931742 DOI: 10.1111/adb.13122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 11/28/2022]
Abstract
In our previous studies, the offspring of morphine-exposed parents (MEO) showed pharmacological tolerance to the morphine's reinforcing effect. According to the role of exercise in treatment of morphine addiction, the current study was designed to utilize exercise to improve the effect of parental morphine exposure on the morphine's reinforcing effect. Male and female rats received morphine for 10 days and were drug-free for another 10 days. Each morphine-exposed animal was allowed to mate either with a drug-naïve or a morphine-exposed rat. The offspring were divided into two groups: (1) offspring that were subjected to treadmill exercise and (2) offspring that were not subjected to exercise. The reinforcing effect of morphine was evaluated using conditioned place preference (CPP) and two-bottle choice (TBC) tests. Levels of dopamine receptors (D1DR and D2DR), μ-opioid receptor (MOR), and ΔFosB were evaluated in the nucleus accumbens. The MEO obtained lower preference scores in CPP and consumed morphine more than the control group in TBC. After 3 weeks of exercise, the reinforcing effect of morphine in the MEO was similar to the control. D1DR, D2DR, and MOR were increased in MEO compared with the controls before exercise. Levels of D1DR and MOR were decreased after exercise in the MEO; however, D1DR was increased in control. D2DR level did not change after exercise in MEO, but it increased in control group. Moreover, the level of ΔFosB was decreased among MEO while it was increased after exercise. In conclusion, exercise might modulate the reinforcing effect of morphine via alteration in levels of D1DR, MOR, and ΔFosB.
Collapse
Affiliation(s)
| | | | - Ardeshir Akbarabadi
- Iranian National Center for Addiction Studies Tehran University of Medical Sciences Tehran Iran
| | - Azarakhsh Mokri
- Iranian National Center for Addiction Studies Tehran University of Medical Sciences Tehran Iran
- Roozbeh Hospital Tehran University of Medical Sciences Tehran Iran
| | - Bahar Taleb Zadeh Kasgari
- Iranian National Center for Addiction Studies Tehran University of Medical Sciences Tehran Iran
- School of Biology, College of Science University of Tehran Tehran Iran
| | - Mohammad‐Reza Zarrindast
- Iranian National Center for Addiction Studies Tehran University of Medical Sciences Tehran Iran
- Department of Pharmacology, School of Medicine Tehran University of Medical Sciences Tehran Iran
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences Islamic Azad University Tehran Iran
- Endocrinology and Metabolism Research Institute Tehran University of Medical Science Tehran Iran
| |
Collapse
|
11
|
Optogenetic Stimulation of the Anterior Cingulate Cortex Modulates the Pain Processing in Neuropathic Pain: A Review. J Mol Neurosci 2021; 72:1-8. [PMID: 34505976 DOI: 10.1007/s12031-021-01898-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022]
Abstract
Neuropathic pain is characterized by hypersensitivity, hyperalgesia, and allodynia, which is caused by damage to the somatosensory nervous system. It substantially impairs the quality of life. The management of neuropathic pain is challenging and should comprise alternative therapies. Researchers working on neural modulation methods in the field of optogenetics have recently referred to novel techniques that involve the activation or inhibition of signaling proteins by specific wavelengths of light. The use of optogenetics in neuropathic pain facilitates the investigation of pain pathways involved in chronic pain and has the potential for therapeutic use. Neuropathic pain is often accompanied by negative stimuli involving a broad network of brain regions. In particular, the anterior cingulate cortex (ACC) is a part of the limbic system that has highly interconnected structures involved in processing components of pain. The ACC is a key region for acute pain perception as well as the development of neuropathic pain, characterized by long-term potentiation induced in pain pathways. The exact mechanism for neuropathic pain in the ACC is unclear. Current evidence supports the potential of optogenetics methods to modulate the neuronal activity in the ACC for neuropathic pain. We anticipate the neuronal modulation in the ACC will be used widely to manage neuropathic pain.
Collapse
|
12
|
Wang T, Zhu X, Yi H, Gu J, Liu S, Izenwasser S, Lemmon VP, Roy S, Hao S. Viral vector-mediated gene therapy for opioid use disorders. Exp Neurol 2021; 341:113710. [PMID: 33781732 DOI: 10.1016/j.expneurol.2021.113710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/26/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022]
Abstract
Chronic exposure to opioids typically results in adverse consequences. Opioid use disorder (OUD) is a disease of the CNS with behavioral, psychological, neurobiological, and medical manifestations. OUD induces a variety of changes of neurotransmitters/neuropeptides in the nervous system. Existing pharmacotherapy, such as opioid maintenance therapy (OMT) is the mainstay for the treatment of OUD, however, current opioid replacement therapy is far from effective for the majority of patients. Pharmacological therapy for OUD has been challenging for many reasons including debilitating side-effects. Therefore, developing an effective, non-pharmacological approach would be a critical advancement in improving and expanding treatment for OUD. Viral vector mediated gene therapy provides a potential new approach for treating opioid abused patients. Gene therapy can supply targeting gene products directly linked to the mechanisms of OUD to restore neurotransmitter and/or neuropeptides imbalance, and avoid the off-target effects of systemic administration of drugs. The most commonly used viral vectors in rodent studies of treatment of opioid-used disorder are based on recombinant adenovirus (AV), adeno-associated virus (AAV), lentiviral (LV) vectors, and herpes simplex virus (HSV) vectors. In this review, we will focus on the recent progress of viral vector mediated gene therapy in OUD, especially morphine tolerance and withdrawal.
Collapse
Affiliation(s)
- Tao Wang
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Xun Zhu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Hyun Yi
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Jun Gu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shue Liu
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sari Izenwasser
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Vance P Lemmon
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Shuanglin Hao
- Department of Anesthesiology, Perioperative Medicine & Pain Management, University of Miami Miller School of Medicine, Miami, FL, United States of America.
| |
Collapse
|
13
|
Therapeutic potential of targeting G protein-gated inwardly rectifying potassium (GIRK) channels in the central nervous system. Pharmacol Ther 2021; 223:107808. [PMID: 33476640 DOI: 10.1016/j.pharmthera.2021.107808] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022]
Abstract
G protein-gated inwardly rectifying potassium channels (Kir3/GirK) are important for maintaining resting membrane potential, cell excitability and inhibitory neurotransmission. Coupled to numerous G protein-coupled receptors (GPCRs), they mediate the effects of many neurotransmitters, neuromodulators and hormones contributing to the general homeostasis and particular synaptic plasticity processes, learning, memory and pain signaling. A growing number of behavioral and genetic studies suggest a critical role for the appropriate functioning of the central nervous system, as well as their involvement in many neurologic and psychiatric conditions, such as neurodegenerative diseases, mood disorders, attention deficit hyperactivity disorder, schizophrenia, epilepsy, alcoholism and drug addiction. Hence, GirK channels emerge as a very promising tool to be targeted in the current scenario where these conditions already are or will become a global public health problem. This review examines recent findings on the physiology, function, dysfunction, and pharmacology of GirK channels in the central nervous system and highlights the relevance of GirK channels as a worthful potential target to improve therapies for related diseases.
Collapse
|
14
|
Sakloth F, Polizu C, Bertherat F, Zachariou V. Regulators of G Protein Signaling in Analgesia and Addiction. Mol Pharmacol 2020; 98:739-750. [PMID: 32474445 PMCID: PMC7662521 DOI: 10.1124/mol.119.119206] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Regulator of G protein signaling (RGS) proteins are multifunctional proteins expressed in peripheral and neuronal cells, playing critical roles in development, physiologic processes, and pharmacological responses. RGS proteins primarily act as GTPase accelerators for activated Gα subunits of G-protein coupled receptors, but they may also modulate signal transduction by several other mechanisms. Over the last two decades, preclinical work identified members of the RGS family with unique and critical roles in intracellular responses to drugs of abuse. New information has emerged on the mechanisms by which RGS proteins modulate the efficacy of opioid analgesics in a brain region- and agonist-selective fashion. There has also been progress in the understanding of the protein complexes and signal transduction pathways regulated by RGS proteins in addiction and analgesia circuits. In this review, we summarize findings on the mechanisms by which RGS proteins modulate functional responses to opioids in models of analgesia and addiction. We also discuss reports on the regulation and function of RGS proteins in models of psychostimulant addiction. Using information from preclinical studies performed over the last 20 years, we highlight the diverse mechanisms by which RGS protein complexes control plasticity in response to opioid and psychostimulant drug exposure; we further discuss how the understanding of these pathways may lead to new opportunities for therapeutic interventions in G protein pathways. SIGNIFICANCE STATEMENT: Regulator of G protein signaling (RGS) proteins are signal transduction modulators, expressed widely in various tissues, including brain regions mediating addiction and analgesia. Evidence from preclinical work suggests that members of the RGS family act by unique mechanisms in specific brain regions to control drug-induced plasticity. This review highlights interesting findings on the regulation and function of RGS proteins in models of analgesia and addiction.
Collapse
Affiliation(s)
- Farhana Sakloth
- Nash Family Department of Neuroscience, and Friedman Brain Institute (F.S., C.P., F.B., V.Z.) and Department of Pharmacological Sciences (V.Z.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Claire Polizu
- Nash Family Department of Neuroscience, and Friedman Brain Institute (F.S., C.P., F.B., V.Z.) and Department of Pharmacological Sciences (V.Z.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Feodora Bertherat
- Nash Family Department of Neuroscience, and Friedman Brain Institute (F.S., C.P., F.B., V.Z.) and Department of Pharmacological Sciences (V.Z.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Venetia Zachariou
- Nash Family Department of Neuroscience, and Friedman Brain Institute (F.S., C.P., F.B., V.Z.) and Department of Pharmacological Sciences (V.Z.), Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
15
|
Abstract
Opioid use disorder (OUD) is a chronic relapsing disorder that, whilst initially driven by activation of brain reward neurocircuits, increasingly engages anti-reward neurocircuits that drive adverse emotional states and relapse. However, successful recovery is possible with appropriate treatment, although with a persisting propensity to relapse. The individual and public health burdens of OUD are immense; 26.8 million people were estimated to be living with OUD globally in 2016, with >100,000 opioid overdose deaths annually, including >47,000 in the USA in 2017. Well-conducted trials have demonstrated that long-term opioid agonist therapy with methadone and buprenorphine have great efficacy for OUD treatment and can save lives. New forms of the opioid receptor antagonist naltrexone are also being studied. Some frequently used approaches have less scientifically robust evidence but are nevertheless considered important, including community preventive strategies, harm reduction interventions to reduce adverse sequelae from ongoing use and mutual aid groups. Other commonly used approaches, such as detoxification alone, lack scientific evidence. Delivery of effective prevention and treatment responses is often complicated by coexisting comorbidities and inadequate support, as well as by conflicting public and political opinions. Science has a crucial role to play in informing public attitudes and developing fuller evidence to understand OUD and its associated harms, as well as in obtaining the evidence today that will improve the prevention and treatment interventions of tomorrow.
Collapse
|
16
|
Serafini RA, Pryce KD, Zachariou V. The Mesolimbic Dopamine System in Chronic Pain and Associated Affective Comorbidities. Biol Psychiatry 2020; 87:64-73. [PMID: 31806085 PMCID: PMC6954000 DOI: 10.1016/j.biopsych.2019.10.018] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/18/2022]
Abstract
Chronic pain is a complex neuropsychiatric disorder characterized by sensory, cognitive, and affective symptoms. Over the past 2 decades, researchers have made significant progress toward understanding the impact of mesolimbic dopamine circuitry in acute and chronic pain. These efforts have provided insights into the circuits and intracellular pathways in the brain reward center that are implicated in sensory and affective manifestations of chronic pain. Studies have also identified novel therapeutic targets as well as factors that affect treatment responsiveness. Dysregulation of dopamine function in the brain reward center may further promote comorbid mood disorders and vulnerability to addiction. This review discusses recent clinical and preclinical findings on the neuroanatomical and neurochemical adaptations triggered by prolonged pain states in the brain reward pathway. Furthermore, this discussion highlights evidence of mechanisms underlying comorbidities among pain, depression, and addiction.
Collapse
Affiliation(s)
- Randal A Serafini
- Nash Family Department of Neuroscience, Department of Pharmacological Sciences, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kerri D Pryce
- Nash Family Department of Neuroscience, Department of Pharmacological Sciences, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Venetia Zachariou
- Nash Family Department of Neuroscience, Department of Pharmacological Sciences, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
17
|
O'Brien JB, Wilkinson JC, Roman DL. Regulator of G-protein signaling (RGS) proteins as drug targets: Progress and future potentials. J Biol Chem 2019; 294:18571-18585. [PMID: 31636120 PMCID: PMC6901330 DOI: 10.1074/jbc.rev119.007060] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors (GPCRs) play critical roles in regulating processes such as cellular homeostasis, responses to stimuli, and cell signaling. Accordingly, GPCRs have long served as extraordinarily successful drug targets. It is therefore not surprising that the discovery in the mid-1990s of a family of proteins that regulate processes downstream of GPCRs generated great excitement in the field. This finding enhanced the understanding of these critical signaling pathways and provided potentially new targets for pharmacological intervention. These regulators of G-protein signaling (RGS) proteins were viewed by many as nodes downstream of GPCRs that could be targeted with small molecules to tune signaling processes. In this review, we provide a brief overview of the discovery of RGS proteins and of the gradual and continuing discovery of their roles in disease states, focusing particularly on cancer and neurological disorders. We also discuss high-throughput screening efforts that have led to the discovery first of peptide-based and then of small-molecule inhibitors targeting a subset of the RGS proteins. We explore the unique mechanisms of RGS inhibition these chemical tools have revealed and highlight the most up-to-date studies using these tools in animal experiments. Finally, we discuss the future opportunities in the field, as there are clearly more avenues left to be explored and potentials to be realized.
Collapse
Affiliation(s)
- Joseph B O'Brien
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa 52242
| | - Joshua C Wilkinson
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa 52242
| | - David L Roman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa 52242; Iowa Neuroscience Institute, Iowa City, Iowa 52242; Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242.
| |
Collapse
|
18
|
Sadat-Shirazi MS, Monfared Neirizi N, Matloob M, Safarzadeh M, Behrouzi M, Rajabpoor Dehdashti A, Ashabi G, Zarrindast MR. Possible involvement of nucleus accumbens D1-like dopamine receptors in the morphine-induced condition place preference in the offspring of morphine abstinent rats. Life Sci 2019; 233:116712. [DOI: 10.1016/j.lfs.2019.116712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/22/2019] [Accepted: 07/30/2019] [Indexed: 12/15/2022]
|
19
|
Regulators of G-Protein Signaling (RGS) Proteins Promote Receptor Coupling to G-Protein-Coupled Inwardly Rectifying Potassium (GIRK) Channels. J Neurosci 2018; 38:8737-8744. [PMID: 30150362 DOI: 10.1523/jneurosci.0516-18.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/31/2018] [Accepted: 08/20/2018] [Indexed: 01/01/2023] Open
Abstract
Regulators of G-protein signaling (RGS) proteins negatively modulate presynaptic μ-opioid receptor inhibition of GABA release in the ventrolateral periaqueductal gray (vlPAG). Paradoxically, we find that G-protein-coupled receptor (GPCR) activation of G-protein-gated inwardly rectifying K+ channels (GIRKs) in the vlPAG is reduced in an agonist- and receptor-dependent manner in transgenic knock-in mice of either sex expressing mutant RGS-insensitive Gαo proteins. μ-Opioid receptor agonist activation of GIRK currents was reduced for DAMGO and fentanyl but not for [Met5]-enkephalin acetate salt hydrate (ME) in the RGS-insensitive heterozygous (Het) mice compared with wild-type mice. The GABAB agonist baclofen-induced GIRK currents were also reduced in the Het mice. We confirmed the role of Gαo proteins in μ-opioid receptor and GABAB receptor signaling pathways in wild-type mice using myristoylated peptide inhibitors of Gαo1 and Gαi1-3 The results using these inhibitors indicate that receptor activation of GIRK channels is dependent on the preference of the agonist-stimulated receptor for Gαo versus that for Gαi. DAMGO and fentanyl-mediated GIRK currents were reduced in the presence of the Gαo1 inhibitor, but not the Gαi1-3 inhibitors. In contrast, the Gαo1 peptide inhibitor did not affect ME activation of GIRK currents, which is consistent with results in the Het mice, but the Gαi1-3 inhibitors significantly reduced ME-mediated GIRK currents. Finally, the reduction in GIRK activation in the Het mice plays a role in opioid- and baclofen-mediated spinal antinociception, but not supraspinal antinociception. Thus, our studies indicate that RGS proteins have multiple mechanisms of modulating GPCR signaling that produce negative and positive regulation of signaling depending on the effector.SIGNIFICANCE STATEMENT Regulators of G-protein signaling (RGS) proteins positively modulate GPCR coupling to GIRKs, and this coupling is critical for opioid- and baclofen-mediated spinal antinociception, whereas μ-opioid receptor-mediated supraspinal antinociception depends on presynaptic inhibition that is negatively regulated by RGS proteins. The identification of these opposite roles for RGS proteins has implications for signaling via other GPCRs.
Collapse
|
20
|
Selective Role of RGS9-2 in Regulating Retrograde Synaptic Signaling of Indirect Pathway Medium Spiny Neurons in Dorsal Striatum. J Neurosci 2018; 38:7120-7131. [PMID: 30006367 DOI: 10.1523/jneurosci.0493-18.2018] [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: 02/21/2018] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 11/21/2022] Open
Abstract
In the striatum, medium spiny neurons (MSNs) are heavily involved in controlling movement and reward. MSNs form two distinct populations expressing either dopamine receptor 1 (D1-MSN) or dopamine receptor 2 (D2-MSN), which differ in their projection targets and neurochemical composition. The activity of both types of MSNs is shaped by multiple neuromodulatory inputs processed by GPCRs that fundamentally impact their synaptic properties biasing behavioral outcomes. How these GPCR signaling cascades are regulated and what downstream targets they recruit in D1-MSN and D2-MSN populations are incompletely understood. In this study, we examined the cellular and molecular mechanisms underlying the action of RGS9-2, a key GPCR regulator in MSNs implicated in both movement control and actions of addictive drugs. Imaging cultured striatal neurons, we found that ablation of RGS9-2 significantly reduced calcium influx through NMDARs. Electrophysiological recordings in slices confirmed inhibition of NMDAR function in MSNs, resulting in enhanced AMPAR/NMDAR ratio. Accordingly, male mice lacking RGS9-2 displayed behavioral hypersensitivity to NMDAR blockade by MK-801 or ketamine. Recordings from genetically identified populations of striatal neurons revealed that these changes were selective to D2-MSNs. Surprisingly, we found that these postsynaptic effects resulted in remodeling of presynaptic inputs to D2-MSNs increasing the frequency of mEPSC and inhibiting paired-pulse ratio. Pharmacological dissection revealed that these adaptations were mediated by the NMDAR-dependent inhibition of retrograde endocannabinoid signaling from D2-MSNs to CB1 receptor on presynaptic terminals. Together, these data demonstrate a novel mechanism for pathway selective regulation of synaptic plasticity in MSNs controlled by GPCR signaling.SIGNIFICANCE STATEMENT This study identifies a role for a major G-protein regulator in controlling synaptic properties of striatal neurons in a pathway selective fashion.
Collapse
|
21
|
Kim J, Lee S, Kang S, Jeon TI, Kang MJ, Lee TH, Kim YS, Kim KS, Im HI, Moon C. Regulator of G-Protein Signaling 4 (RGS4) Controls Morphine Reward by Glutamate Receptor Activation in the Nucleus Accumbens of Mouse Brain. Mol Cells 2018; 41:454-464. [PMID: 29754475 PMCID: PMC5974622 DOI: 10.14348/molcells.2018.0023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/02/2018] [Accepted: 03/08/2018] [Indexed: 11/27/2022] Open
Abstract
Crosstalk between G-protein signaling and glutamatergic transmission within the brain reward circuits is critical for long-term emotional effects (depression and anxiety), cravings, and negative withdrawal symptoms associated with opioid addiction. A previous study showed that Regulator of G-protein signaling 4 (RGS4) may be implicated in opiate action in the nucleus accumbens (NAc). However, the mechanism of the NAc-specific RGS4 actions that induce the behavioral responses to opiates remains largely unknown. The present study used a short hairpin RNA (shRNA)-mediated knock-down of RGS4 in the NAc of the mouse brain to investigate the relationship between the activation of ionotropic glutamate receptors and RGS4 in the NAc during morphine reward. Additionally, the shRNA-mediated RGS4 knock-down was implemented in NAc/striatal primary-cultured neurons to investigate the role that striatal neurons have in the morphine-induced activation of ionotropic glutamate receptors. The results of this study show that the NAc-specific knockdown of RGS4 significantly increased the behaviors associated with morphine and did so by phosphorylation of the GluR1 (Ser831) and NR2A (Tyr1325) glutamate receptors in the NAc. Furthermore, the knock-down of RGS4 enhanced the phosphorylation of the GluR1 and NR2A glutamate receptors in the primary NAc/striatal neurons during spontaneous morphine withdrawal. These findings show a novel molecular mechanism of RGS4 in glutamatergic transmission that underlies the negative symptoms associated with morphine administration.
Collapse
Affiliation(s)
- Juhwan Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186,
Korea
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792,
Korea
- Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju 61186,
Korea
| | - Sueun Lee
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186,
Korea
| | - Sohi Kang
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186,
Korea
| | - Tae-Il Jeon
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju 61186,
Korea
| | - Man-Jong Kang
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju 61186,
Korea
| | - Tae-Hoon Lee
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju 61186,
Korea
- Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju 61186,
Korea
| | - Yong Sik Kim
- Department of Pharmacology, Seoul National University College of Medicine, Seoul 08826,
Korea
| | - Key-Sun Kim
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792,
Korea
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792,
Korea
| | - Heh-In Im
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792,
Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792,
Korea
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792,
Korea
| | - Changjong Moon
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186,
Korea
| |
Collapse
|
22
|
Suppression of RGSz1 function optimizes the actions of opioid analgesics by mechanisms that involve the Wnt/β-catenin pathway. Proc Natl Acad Sci U S A 2018; 115:E2085-E2094. [PMID: 29440403 DOI: 10.1073/pnas.1707887115] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Regulator of G protein signaling z1 (RGSz1), a member of the RGS family of proteins, is present in several networks expressing mu opioid receptors (MOPRs). By using genetic mouse models for global or brain region-targeted manipulations of RGSz1 expression, we demonstrated that the suppression of RGSz1 function increases the analgesic efficacy of MOPR agonists in male and female mice and delays the development of morphine tolerance while decreasing the sensitivity to rewarding and locomotor activating effects. Using biochemical assays and next-generation RNA sequencing, we identified a key role of RGSz1 in the periaqueductal gray (PAG) in morphine tolerance. Chronic morphine administration promotes RGSz1 activity in the PAG, which in turn modulates transcription mediated by the Wnt/β-catenin signaling pathway to promote analgesic tolerance to morphine. Conversely, the suppression of RGSz1 function stabilizes Axin2-Gαz complexes near the membrane and promotes β-catenin activation, thereby delaying the development of analgesic tolerance. These data show that the regulation of RGS complexes, particularly those involving RGSz1-Gαz, represents a promising target for optimizing the analgesic actions of opioids without increasing the risk of dependence or addiction.
Collapse
|
23
|
Gaspari S, Cogliani V, Manouras L, Anderson EM, Mitsi V, Avrampou K, Carr FB, Zachariou V. RGS9-2 Modulates Responses to Oxycodone in Pain-Free and Chronic Pain States. Neuropsychopharmacology 2017; 42:1548-1556. [PMID: 28074831 PMCID: PMC5436127 DOI: 10.1038/npp.2017.4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/28/2016] [Accepted: 12/23/2016] [Indexed: 12/11/2022]
Abstract
Regulator of G-protein signaling 9-2 (RGS9-2) is a striatal-enriched signal-transduction modulator known to have a critical role in the development of addiction-related behaviors following exposure to psychostimulants or opioids. RGS9-2 controls the function of several G-protein-coupled receptors, including dopamine receptor and mu opioid receptor (MOR). We previously showed that RGS9-2 complexes negatively control morphine analgesia, and promote the development of morphine tolerance. In contrast, RGS9-2 positively modulates the actions of other opioid analgesics, such as fentanyl and methadone. Here we investigate the role of RGS9-2 in regulating responses to oxycodone, an MOR agonist prescribed for the treatment of severe pain conditions that has addictive properties. Using mice lacking the Rgs9 gene (RGS9KO), we demonstrate that RGS9-2 positively regulates the rewarding effects of oxycodone in pain-free states, and in a model of neuropathic pain. Furthermore, although RGS9-2 does not affect the analgesic efficacy of oxycodone or the expression of physical withdrawal, it opposes the development of oxycodone tolerance, in both acute pain and chronic neuropathic pain models. Taken together, these data provide new information on the signal-transduction mechanisms that modulate the rewarding and analgesic actions of oxycodone.
Collapse
Affiliation(s)
- Sevasti Gaspari
- Icahn School of Medicine at Mount Sinai, Fishberg Department of Neuroscience and Friedman Brain Institute, New York, NY, USA
- University of Crete Faculty of Medicine, Department of Basic Sciences, Heraklion, Greece
| | - Valeria Cogliani
- Icahn School of Medicine at Mount Sinai, Fishberg Department of Neuroscience and Friedman Brain Institute, New York, NY, USA
| | - Lefteris Manouras
- University of Crete Faculty of Medicine, Department of Basic Sciences, Heraklion, Greece
| | - Ethan M Anderson
- Icahn School of Medicine at Mount Sinai, Fishberg Department of Neuroscience and Friedman Brain Institute, New York, NY, USA
| | - Vasiliki Mitsi
- Icahn School of Medicine at Mount Sinai, Fishberg Department of Neuroscience and Friedman Brain Institute, New York, NY, USA
| | - Kleopatra Avrampou
- Icahn School of Medicine at Mount Sinai, Fishberg Department of Neuroscience and Friedman Brain Institute, New York, NY, USA
| | - Fiona B Carr
- Icahn School of Medicine at Mount Sinai, Fishberg Department of Neuroscience and Friedman Brain Institute, New York, NY, USA
| | - Venetia Zachariou
- Icahn School of Medicine at Mount Sinai, Fishberg Department of Neuroscience and Friedman Brain Institute, New York, NY, USA
| |
Collapse
|
24
|
Cahill CM, Walwyn W, Taylor AMW, Pradhan AAA, Evans CJ. Allostatic Mechanisms of Opioid Tolerance Beyond Desensitization and Downregulation. Trends Pharmacol Sci 2016; 37:963-976. [PMID: 27670390 DOI: 10.1016/j.tips.2016.08.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 12/20/2022]
Abstract
Mechanisms of opioid tolerance have focused on adaptive modifications within cells containing opioid receptors, defined here as cellular allostasis, emphasizing regulation of the opioid receptor signalosome. We review additional regulatory and opponent processes involved in behavioral tolerance, and include mechanistic differences both between agonists (agonist bias), and between μ- and δ-opioid receptors. In a process we will refer to as pass-forward allostasis, cells modified directly by opioid drugs impute allostatic changes to downstream circuitry. Because of the broad distribution of opioid systems, every brain cell may be touched by pass-forward allostasis in the opioid-dependent/tolerant state. We will implicate neurons and microglia as interactive contributors to the cumulative allostatic processes creating analgesic and hedonic tolerance to opioid drugs.
Collapse
Affiliation(s)
- Catherine M Cahill
- Department of Anesthesiology and Perioperative Care, University of California, Irvine, 837 Health Sciences Road, Irvine, CA 92697, USA
| | - Wendy Walwyn
- Hatos Center for Neuropharmacology, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, 675 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Anna M W Taylor
- Hatos Center for Neuropharmacology, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, 675 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Amynah A A Pradhan
- Department of Psychiatry, University of Illinois at Chicago, 1601 West Taylor Street, Chicago, IL 60612, USA
| | - Christopher J Evans
- Hatos Center for Neuropharmacology, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, 675 Charles E. Young Drive South, Los Angeles, CA 90095, USA.
| |
Collapse
|
25
|
Sutton LP, Ostrovskaya O, Dao M, Xie K, Orlandi C, Smith R, Wee S, Martemyanov KA. Regulator of G-Protein Signaling 7 Regulates Reward Behavior by Controlling Opioid Signaling in the Striatum. Biol Psychiatry 2016; 80:235-45. [PMID: 26364547 PMCID: PMC4753143 DOI: 10.1016/j.biopsych.2015.07.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 07/02/2015] [Accepted: 07/27/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Morphine mediates its euphoric and analgesic effects by acting on the μ-opioid receptor (MOR). MOR belongs to the family of G-protein coupled receptors whose signaling efficiency is controlled by the regulator of G-protein signaling (RGS) proteins. Our understanding of the molecular diversity of RGS proteins that control MOR signaling, their circuit specific actions, and underlying cellular mechanisms is very limited. METHODS We used genetic approaches to ablate regulator of G-protein signaling 7 (RGS7) both globally and in specific neuronal populations. We used conditioned place preference and self-administration paradigms to examine reward-related behavior and a battery of tests to assess analgesia, tolerance, and physical dependence to morphine. Electrophysiology approaches were applied to investigate the impact of RGS7 on morphine-induced alterations in neuronal excitability and plasticity of glutamatergic synapses. At least three animals were used for each assessment. RESULTS Elimination of RGS7 enhanced reward, increased analgesia, delayed tolerance, and heightened withdrawal in response to morphine administration. RGS7 in striatal neurons was selectively responsible for determining the sensitivity of rewarding and reinforcing behaviors to morphine without affecting analgesia, tolerance, and withdrawal. In contrast, deletion of RGS7 in dopaminergic neurons did not influence morphine reward. RGS7 exerted its effects by controlling morphine-induced changes in excitability of medium spiny neurons in nucleus accumbens and gating the compositional plasticity of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid and N-methyl-D-aspartate receptors. CONCLUSIONS This study identifies RGS7 as a novel regulator of MOR signaling by dissecting its circuit specific actions and pinpointing its role in regulating morphine reward by controlling the activity of nucleus accumbens neurons.
Collapse
Affiliation(s)
- Laurie P. Sutton
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458 USA
| | - Olga Ostrovskaya
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458 USA
| | - Maria Dao
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL 33458 USA,Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458 USA
| | - Keqiang Xie
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458 USA
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458 USA
| | - Roy Smith
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458 USA
| | - Sunmee Wee
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL 33458 USA
| | - Kirill A. Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458 USA,Corresponding author: Dr. Kirill Martemyanov, Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, 3C2, Jupiter, FL 33458, Phone: (561) 228-2770,
| |
Collapse
|
26
|
Ahlers KE, Chakravarti B, Fisher RA. RGS6 as a Novel Therapeutic Target in CNS Diseases and Cancer. AAPS JOURNAL 2016; 18:560-72. [PMID: 27002730 DOI: 10.1208/s12248-016-9899-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/25/2016] [Indexed: 12/17/2022]
Abstract
Regulator of G protein signaling (RGS) proteins are gatekeepers regulating the cellular responses induced by G protein-coupled receptor (GPCR)-mediated activation of heterotrimeric G proteins. Specifically, RGS proteins determine the magnitude and duration of GPCR signaling by acting as a GTPase-activating protein for Gα subunits, an activity facilitated by their semiconserved RGS domain. The R7 subfamily of RGS proteins is distinguished by two unique domains, DEP/DHEX and GGL, which mediate membrane targeting and stability of these proteins. RGS6, a member of the R7 subfamily, has been shown to specifically modulate Gαi/o protein activity which is critically important in the central nervous system (CNS) for neuronal responses to a wide array of neurotransmitters. As such, RGS6 has been implicated in several CNS pathologies associated with altered neurotransmission, including the following: alcoholism, anxiety/depression, and Parkinson's disease. In addition, unlike other members of the R7 subfamily, RGS6 has been shown to regulate G protein-independent signaling mechanisms which appear to promote both apoptotic and growth-suppressive pathways that are important in its tumor suppressor function in breast and possibly other tissues. Further highlighting the importance of RGS6 as a target in cancer, RGS6 mediates the chemotherapeutic actions of doxorubicin and blocks reticular activating system (Ras)-induced cellular transformation by promoting degradation of DNA (cytosine-5)-methyltransferase 1 (DNMT1) to prevent its silencing of pro-apoptotic and tumor suppressor genes. Together, these findings demonstrate the critical role of RGS6 in regulating both G protein-dependent CNS pathology and G protein-independent cancer pathology implicating RGS6 as a novel therapeutic target.
Collapse
Affiliation(s)
- Katelin E Ahlers
- Department of Pharmacology, The Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 2-505 Bowen Science Building, Iowa City, Iowa, 52242, USA
| | - Bandana Chakravarti
- Department of Pharmacology, The Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 2-505 Bowen Science Building, Iowa City, Iowa, 52242, USA
| | - Rory A Fisher
- Department of Pharmacology, The Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 2-505 Bowen Science Building, Iowa City, Iowa, 52242, USA. .,Department of Internal Medicine, The Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
27
|
Abstract
This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants). This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
28
|
Maple AM, Smith KJ, Perna MK, Brown RW. Neonatal quinpirole treatment produces prepulse inhibition deficits in adult male and female rats. Pharmacol Biochem Behav 2015; 137:93-100. [DOI: 10.1016/j.pbb.2015.08.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/10/2015] [Accepted: 08/13/2015] [Indexed: 11/30/2022]
|
29
|
RGS9-2--controlled adaptations in the striatum determine the onset of action and efficacy of antidepressants in neuropathic pain states. Proc Natl Acad Sci U S A 2015; 112:E5088-97. [PMID: 26305935 DOI: 10.1073/pnas.1504283112] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The striatal protein Regulator of G-protein signaling 9-2 (RGS9-2) plays a key modulatory role in opioid, monoamine, and other G-protein-coupled receptor responses. Here, we use the murine spared-nerve injury model of neuropathic pain to investigate the mechanism by which RGS9-2 in the nucleus accumbens (NAc), a brain region involved in mood, reward, and motivation, modulates the actions of tricyclic antidepressants (TCAs). Prevention of RGS9-2 action in the NAc increases the efficacy of the TCA desipramine and dramatically accelerates its onset of action. By controlling the activation of effector molecules by G protein α and βγ subunits, RGS9-2 affects several protein interactions, phosphoprotein levels, and the function of the epigenetic modifier histone deacetylase 5, which are important for TCA responsiveness. Furthermore, information from RNA-sequencing analysis reveals that RGS9-2 in the NAc affects the expression of many genes known to be involved in nociception, analgesia, and antidepressant drug actions. Our findings provide novel information on NAc-specific cellular mechanisms that mediate the actions of TCAs in neuropathic pain states.
Collapse
|
30
|
Kravitz AV, Tomasi D, LeBlanc KH, Baler R, Volkow ND, Bonci A, Ferré S. Cortico-striatal circuits: Novel therapeutic targets for substance use disorders. Brain Res 2015; 1628:186-98. [PMID: 25863130 DOI: 10.1016/j.brainres.2015.03.048] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 03/04/2015] [Accepted: 03/28/2015] [Indexed: 01/08/2023]
Abstract
It is widely believed that substance use disorder (SUD) results from both pre-alterations (vulnerability) and/or post-alterations (drug effects) on cortico-striatal circuits. These circuits are essential for cognitive control, motivation, reward dependent learning, and emotional processing. As such, dysfunctions in cortico-striatal circuits are thought to relate to the core features of SUD, which include compulsive drug use, loss of the ability to control drug intake, and the emergence of negative emotional states (Koob and Volkow, 2010. Neuropsychopharmacology 35(1), 217-238). While the brain circuits underlying SUD have been studied in human patients largely through imaging studies, experiments in animals have allowed researchers to examine the specific cell-types within these circuits to reveal their role in behavior relevant to SUD. Here, we will review imaging studies on cortico-striatal systems that are altered in SUD, and describe animal experiments that relate SUD to specific neural projections and cell types within this circuitry. We will end with a discussion of novel clinical approaches such as deep brain stimulation (DBS), repeated transcranial magnetic stimulation (rTMS), and pharmacological targeting of G protein-coupled receptor (GPCR) heteromers that may provide promising avenues for modulating these circuits to combat SUD in humans.
Collapse
Affiliation(s)
- Alexxai V Kravitz
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA; National Institute of Drug Abuse, Baltimore, MD, USA
| | - Dardo Tomasi
- National Institute on Alcoholism and Alcohol Abuse, Bethesda, MD, USA
| | - Kimberly H LeBlanc
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Ruben Baler
- National Institute of Drug Abuse, Baltimore, MD, USA
| | - Nora D Volkow
- National Institute of Drug Abuse, Baltimore, MD, USA
| | - Antonello Bonci
- National Institute of Drug Abuse, Baltimore, MD, USA; Solomon H. Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Sergi Ferré
- National Institute of Drug Abuse, Baltimore, MD, USA
| |
Collapse
|
31
|
Woodard GE, Jardín I, Berna-Erro A, Salido GM, Rosado JA. Regulators of G-protein-signaling proteins: negative modulators of G-protein-coupled receptor signaling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 317:97-183. [PMID: 26008785 DOI: 10.1016/bs.ircmb.2015.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regulators of G-protein-signaling (RGS) proteins are a category of intracellular proteins that have an inhibitory effect on the intracellular signaling produced by G-protein-coupled receptors (GPCRs). RGS along with RGS-like proteins switch on through direct contact G-alpha subunits providing a variety of intracellular functions through intracellular signaling. RGS proteins have a common RGS domain that binds to G alpha. RGS proteins accelerate GTPase and thus enhance guanosine triphosphate hydrolysis through the alpha subunit of heterotrimeric G proteins. As a result, they inactivate the G protein and quickly turn off GPCR signaling thus terminating the resulting downstream signals. Activity and subcellular localization of RGS proteins can be changed through covalent molecular changes to the enzyme, differential gene splicing, and processing of the protein. Other roles of RGS proteins have shown them to not be solely committed to being inhibitors but behave more as modulators and integrators of signaling. RGS proteins modulate the duration and kinetics of slow calcium oscillations and rapid phototransduction and ion signaling events. In other cases, RGS proteins integrate G proteins with signaling pathways linked to such diverse cellular responses as cell growth and differentiation, cell motility, and intracellular trafficking. Human and animal studies have revealed that RGS proteins play a vital role in physiology and can be ideal targets for diseases such as those related to addiction where receptor signaling seems continuously switched on.
Collapse
Affiliation(s)
- Geoffrey E Woodard
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Isaac Jardín
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - A Berna-Erro
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - Gines M Salido
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - Juan A Rosado
- Department of Physiology, University of Extremadura, Caceres, Spain
| |
Collapse
|
32
|
Zhang Y, Qu H, Zhou Y, Wang Y, Zhang D, Yang X, Yang C, Xu M. The involvement of norepinephrine in pain modulation in the nucleus accumbens of morphine-dependent rats. Neurosci Lett 2015; 585:6-11. [DOI: 10.1016/j.neulet.2014.11.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/12/2014] [Accepted: 11/13/2014] [Indexed: 11/29/2022]
|
33
|
Terzi D, Gaspari S, Manouras L, Descalzi G, Mitsi V, Zachariou V. RGS9-2 modulates sensory and mood related symptoms of neuropathic pain. Neurobiol Learn Mem 2014; 115:43-8. [PMID: 25150149 DOI: 10.1016/j.nlm.2014.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 08/07/2014] [Accepted: 08/09/2014] [Indexed: 10/24/2022]
Abstract
The signal transduction modulator Rgs9-2 (Regulator of G protein signaling 9-2) plays a key role in dopaminergic and opioidergic transmission in the striatum. Rgs9-2 is a potent modulator of opiate reward and analgesia, but its role in chronic pain remains unknown. Here, we use the spared nerve injury model (SNI), to evaluate the influence of Rgs9-2 in sensory symptoms, as well as in anxiety and depression-like behaviors observed under neuropathic pain conditions. Our data demonstrate that knockout of the Rgs9 gene reduces the intensity of thermal hyperalgesia and mechanical allodynia the first few days after nerve injury. This small, but significant effect is only observed at early time points after nerve injury, whereas after the first week of SNI, Rgs9 knockout (Rgs9KO) and Rgs9 wildtype (Rgs9WT) mice show similar levels of mechanical allodynia and thermal hyperalgesia. Furthermore, Rgs9-2 deletion exacerbates anxiety and depression like behaviors several weeks after the emergence of the neuropathic pain symptoms. Our findings also reveal a temporal and regional regulation of Rgs9-2 protein expression by neuropathic pain, as Rgs9-2 levels are reduced in the spinal cord a few days after nerve injury, whereas decreased Rgs9-2 levels in the Nucleus Accumbens (NAc) are only observed several weeks after nerve injury. Thus, adaptations in Rgs9-2 activity in the spinal cord and in the NAc may contribute to sensory and affective components of neuropathic pain.
Collapse
Affiliation(s)
- Dimitra Terzi
- University of Crete Faculty of Medicine, Department of Basic Sciences, Heraklion, Crete 71003, Greece
| | - Sevasti Gaspari
- University of Crete Faculty of Medicine, Department of Basic Sciences, Heraklion, Crete 71003, Greece
| | - Lefteris Manouras
- University of Crete Faculty of Medicine, Department of Basic Sciences, Heraklion, Crete 71003, Greece
| | - Giannina Descalzi
- Icahn School of Medicine at Mount Sinai, Fishberg Department of Neuroscience and Friedman Brain Institute, Department of Pharmacology and Systems Therapeutics, United States
| | - Vassiliki Mitsi
- University of Crete Faculty of Medicine, Department of Basic Sciences, Heraklion, Crete 71003, Greece
| | - Venetia Zachariou
- University of Crete Faculty of Medicine, Department of Basic Sciences, Heraklion, Crete 71003, Greece; Icahn School of Medicine at Mount Sinai, Fishberg Department of Neuroscience and Friedman Brain Institute, Department of Pharmacology and Systems Therapeutics, United States.
| |
Collapse
|
34
|
Carr FB, Zachariou V. Nociception and pain: lessons from optogenetics. Front Behav Neurosci 2014; 8:69. [PMID: 24723861 PMCID: PMC3971183 DOI: 10.3389/fnbeh.2014.00069] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/18/2014] [Indexed: 12/29/2022] Open
Abstract
The process of pain perception begins in the periphery by activation of nociceptors. From here nociceptive signals are conveyed via the dorsal horn of the spinal cord to multiple brain regions, where pain is perceived. Despite great progress in pain research in recent years, many questions remain regarding nociceptive circuitry and behavior, in both acute nociception and chronic pain states. Techniques that allow for selective activation of neuronal subpopulations in vivo can provide a better understanding of these complex pathways. Here we review the studies to date that have employed novel optogenetic tools to improve our understanding of the pain pathway at the peripheral, spinal and supraspinal levels.
Collapse
Affiliation(s)
- Fiona B Carr
- Fishberg Department of Neuroscience, Department of Pharmacology and Systems Therapeutics and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Venetia Zachariou
- Fishberg Department of Neuroscience, Department of Pharmacology and Systems Therapeutics and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| |
Collapse
|