1
|
Goldberg LR, Baskin BM, Adla Y, Beierle JA, Kelliher JC, Yao EJ, Kirkpatrick SL, Reed ER, Jenkins DF, Cox J, Luong AM, Luttik KP, Scotellaro JA, Drescher TA, Crotts SB, Yazdani N, Ferris MT, Johnson WE, Mulligan MK, Bryant CD. Atp1a2 and Kcnj9 are candidate genes underlying sensitivity to oxycodone-induced locomotor activation and withdrawal-induced anxiety-like behaviors in C57BL/6 substrains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589731. [PMID: 38798314 PMCID: PMC11123399 DOI: 10.1101/2024.04.16.589731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Opioid use disorder is heritable, yet its genetic etiology is largely unknown. C57BL/6J and C57BL/6NJ mouse substrains exhibit phenotypic diversity in the context of limited genetic diversity which together can facilitate genetic discovery. Here, we found C57BL/6NJ mice were less sensitive to oxycodone (OXY)-induced locomotor activation versus C57BL/6J mice in a conditioned place preference paradigm. Narrow-sense heritability was estimated at 0.22-0.31, implicating suitability for genetic analysis. Quantitative trait locus (QTL) mapping in an F2 cross identified a chromosome 1 QTL explaining 7-12% of the variance in OXY locomotion and anxiety-like withdrawal in the elevated plus maze. A second QTL for EPM withdrawal behavior on chromosome 5 near Gabra2 (alpha-2 subunit of GABA-A receptor) explained 9% of the variance. To narrow the chromosome 1 locus, we generated recombinant lines spanning 163-181 Mb, captured the QTL for OXY locomotor traits and withdrawal, and fine-mapped a 2.45-Mb region (170.16-172.61 Mb). Transcriptome analysis identified five, localized striatal cis-eQTL transcripts and two were confirmed at the protein level (KCNJ9, ATP1A2). Kcnj9 codes for a potassium channel (GIRK3) that is a major effector of mu opioid receptor signaling. Atp1a2 codes for a subunit of a Na+/K+ ATPase enzyme that regulates neuronal excitability and shows functional adaptations following chronic opioid administration. To summarize, we identified two candidate genes underlying the physiological and behavioral properties of opioids, with direct preclinical relevance to investigators employing these widely used substrains and clinical relevance to human genetic studies of opioid use disorder.
Collapse
Affiliation(s)
- Lisa R. Goldberg
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- Graduate Program in Biomolecular Pharmacology, Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA USA
| | - Britahny M. Baskin
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- T32 Training Program on Development of Medications for Substance Use Disorder, Center for Drug Discovery, Northeastern University
| | - Yahia Adla
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Jacob A. Beierle
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- Graduate Program in Biomolecular Pharmacology, Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA USA
- Transformative Training Program in Addiction Science, Boston University
| | - Julia C. Kelliher
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Emily J. Yao
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Stacey L. Kirkpatrick
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Eric R. Reed
- Graduate Program in Bioinformatics, Boston University, Boston, MA USA
| | - David F. Jenkins
- Graduate Program in Bioinformatics, Boston University, Boston, MA USA
| | - Jiayi Cox
- Genetics and Graduate Program in Genetics and Genomics, Program in Biomedical Sciences, Boston University Chobanian & Avedisian School of Medicine
| | - Alexander M. Luong
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Kimberly P. Luttik
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Julia A. Scotellaro
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- Undergraduate Research Opportunity Program (UROP), Boston University
| | - Timothy A. Drescher
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Sydney B. Crotts
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
| | - Neema Yazdani
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- Graduate Program in Biomolecular Pharmacology, Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA USA
- Transformative Training Program in Addiction Science, Boston University
| | - Martin T. Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, NC USA
| | - W. Evan Johnson
- Division of Infectious Disease, Department of Medicine, Center for Data Science, Rutgers University, New Jersey, USA
| | - Megan K. Mulligan
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN USA
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Department of Pharmaceutical Sciences and Center for Drug Discovery, Northeastern University, Boston, MA USA
- T32 Training Program on Development of Medications for Substance Use Disorder, Center for Drug Discovery, Northeastern University
| |
Collapse
|
2
|
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
|
3
|
Sakloth F, Sanchez-Reyes OB, Ruiz A, Nicolais A, Serafini RA, Pryce KD, Bertherat F, Torres-Berrío A, Gomes I, Devi LA, Wacker D, Zachariou V. A Regional and Projection-Specific Role of RGSz1 in the Ventrolateral Periaqueductal Grey in the Modulation of Morphine Reward. Mol Pharmacol 2023; 103:1-8. [PMID: 36310031 PMCID: PMC11033942 DOI: 10.1124/molpharm.122.000528] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 09/12/2022] [Accepted: 10/11/2022] [Indexed: 02/03/2023] Open
Abstract
Opioid analgesics exert their therapeutic and adverse effects by activating μ opioid receptors (MOPR); however, functional responses to MOPR activation are modulated by distinct signal transduction complexes within the brain. The ventrolateral periaqueductal gray (vlPAG) plays a critical role in modulation of nociception and analgesia, but the exact intracellular pathways associated with opioid responses in this region are not fully understood. We previously showed that knockout of the signal transduction modulator Regulator of G protein Signaling z1 (RGSz1) enhanced analgesic responses to opioids, whereas it decreased the rewarding efficacy of morphine. Here, we applied viral mediated gene transfer methodology and delivered adeno-associated virus (AAV) expressing Cre recombinase to the vlPAG of RGSz1fl\fl mice to demonstrate that downregulation of RGSz1 in this region decreases sensitivity to morphine in the place preference paradigm, under pain-free as well as neuropathic pain states. We also used retrograde viral vectors along with flippase-dependent Cre vectors to conditionally downregulate RGSz1 in vlPAG projections to the ventral tegmental area (VTA) and show that downregulation of RGSz1 prevents the development of place conditioning to low morphine doses. Consistent with the role for RGSz1 as a negative modulator of MOPR activity, RGSz1KO enhances opioid-induced cAMP inhibition in periaqueductal gray (PAG) membranes. Furthermore, using a new generation of bioluminescence resonance energy transfer (BRET) sensors, we demonstrate that RGSz1 modulates Gαz but not other Gαi family subunits and selectively impedes MOPR-mediated Gαz signaling events invoked by morphine and other opioids. Our work highlights a regional and circuit-specific role of the G protein-signaling modulator RGSz1 in morphine reward, providing insights on midbrain intracellular pathways that control addiction-related behaviors. SIGNIFICANCE STATEMENT: This study used advanced genetic mouse models to highlight the role of the signal transduction modulator named RGSz1 in responses to clinically used opioid analgesics. We show that RGSz1 controls the rewarding efficacy of opioids by actions in ventrolateral periaqueductal gray projections to the ventral tegmental area, a key component of the midbrain dopamine pathway. These studies highlight novel mechanisms by which pain-modulating structures control the rewarding efficacy of opioids.
Collapse
Affiliation(s)
- Farhana Sakloth
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Omar B Sanchez-Reyes
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Anne Ruiz
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Andrew Nicolais
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Randal A Serafini
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Kerri D Pryce
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Feodora Bertherat
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Angélica Torres-Berrío
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Ivone Gomes
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Lakshmi A Devi
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Daniel Wacker
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Venetia Zachariou
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| |
Collapse
|
4
|
Darira SV, Sutton LP. The interaction, mechanism and function of GPR158-RGS7 cross-talk. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 193:167-176. [PMID: 36357076 DOI: 10.1016/bs.pmbts.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
GPR158 is an orphan G protein-coupled receptor (GPCR) that is broadly expressed in the brain and displays unique structural characteristics and signaling mechanisms. GPR158 is a binding partner for the regulator of G protein signaling 7 (RGS7) and augments its expression, subcellular localization, and catalytic activity. Recent cryo-electron microscopy (cryo-EM) studies have revealed the structure of GPR158 alone and in complex with RGS7. The GPR158-RGS7 complex is shown to be regulated by chronic stress exposure and is a modulator of stress-induced depression. This review highlights the signaling mechanism and function of GPR158-RGS7 and provides a context for the unique formation of GPCR-RGS complexes.
Collapse
Affiliation(s)
- Shradha V Darira
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, United States
| | - Laurie P Sutton
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD, United States.
| |
Collapse
|
5
|
Sosnowski DW, Jaffe AE, Tao R, Deep-Soboslay A, Shu C, Sabunciyan S, Kleinman JE, Hyde TM, Maher BS. Differential expression of NPAS4 in the dorsolateral prefrontal cortex following opioid overdose. DRUG AND ALCOHOL DEPENDENCE REPORTS 2022; 3:100040. [PMID: 36845993 PMCID: PMC9948892 DOI: 10.1016/j.dadr.2022.100040] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 06/18/2023]
Abstract
Background Although preclinical models reveal the neurobiological pathways altered through opioid abuse, comprehensive assessments of gene expression in human brain samples are needed. Moreover, less is known about gene expression in response to fatal overdose. The primary goal of the present study was to compare gene expression in the dorsolateral prefrontal cortex (DLPFC) between brain samples of individuals who died of acute opioid intoxication and group-matched controls. Methods Postmortem tissue samples of the DLPFC from 153 deceased individuals (Mage = 35.4; 62% male; 77% European ancestry). Study groups included 72 brain samples from individuals who died of acute opioid intoxication, 53 psychiatric controls, and 28 normal controls. Whole transcriptome RNA-sequencing was used to generate exon counts, and differential expression was tested using limma-voom. Analyses were adjusted for relevant sociodemographic characteristics, technical covariates, and cryptic relatedness using quality surrogate variables. Weighted correlation network analysis and gene set enrichment analyses also were conducted. Results Two genes were differentially expressed in opioid samples compared to control samples. The top gene, NPAS4, was downregulated in opioid samples (log2FC = -2.47, adj. p = .049) and has been implicated in opioid, cocaine, and methamphetamine use. Weighted correlation network analysis revealed 15 gene modules associated with opioid overdose, though no intramodular hub genes were related to opioid overdose, nor were pathways related to opioid overdose enriched for differential expression. Conclusions Results provide preliminary evidence that NPAS4 is implicated in opioid overdose, and more research is needed to understand its role in opioid abuse and associated outcomes.
Collapse
Affiliation(s)
- David W. Sosnowski
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway, Hampton House, Baltimore, MD 21205, United States
| | - Andrew E. Jaffe
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway, Hampton House, Baltimore, MD 21205, United States
- Lieber Institute for Brain Development, United States
| | - Ran Tao
- Lieber Institute for Brain Development, United States
| | | | - Chang Shu
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway, Hampton House, Baltimore, MD 21205, United States
- Department of Pediatrics, Columbia University Irving Medical Center, United States
| | | | - Joel E. Kleinman
- Lieber Institute for Brain Development, United States
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, United States
| | | | - Brion S. Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway, Hampton House, Baltimore, MD 21205, United States
| |
Collapse
|
6
|
Stoveken HM, Fernandez-Vega V, Muntean BS, Patil DN, Shumate J, Bannister TD, Scampavia L, Spicer TP, Martemyanov KA. Identification of Potential Modulators of the RGS7/Gβ5/R7BP Complex. SLAS DISCOVERY 2021; 26:1177-1188. [PMID: 34112017 DOI: 10.1177/24725552211020679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Regulators of G protein signaling (RGS) proteins serve as critical regulatory nodes to limit the lifetime and extent of signaling via G protein-coupled receptors (GPCRs). Previously, approaches to pharmacologically inhibit RGS activity have mostly focused on the inhibition of GTPase activity by interrupting the interaction of RGS proteins with the G proteins they regulate. However, several RGS proteins are also regulated by association with binding partners. A notable example is the mammalian RGS7 protein, which has prominent roles in metabolic control, vision, reward, and actions of opioid analgesics. In vivo, RGS7 exists in complex with the binding partners type 5 G protein β subunit (Gβ5) and R7 binding protein (R7BP), which control its stability and activity, respectively. Targeting the whole RGS7/Gβ5/R7BP protein complex affords the opportunity to allosterically tune opioid receptor signaling following opioid engagement while potentially bypassing undesirable side effects. Hence, we implemented a novel strategy to pharmacologically target the interaction between RGS7/Gβ5 and R7BP. To do so, we searched for protein complex inhibitors using a time-resolved fluorescence resonance energy transfer (FRET)-based high-throughput screening (HTS) assay that measures compound-mediated alterations in the FRET signal between RGS7/Gβ5 and R7BP. We performed two HTS campaigns, each screening ~100,000 compounds from the Scripps Drug Discovery Library (SDDL). Each screen yielded more than 100 inhibitors, which will be described herein.
Collapse
Affiliation(s)
- Hannah M Stoveken
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | | | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Dipak N Patil
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Justin Shumate
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Thomas D Bannister
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Louis Scampavia
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Timothy P Spicer
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | | |
Collapse
|
7
|
Striatal RGS7 Regulates Depression-Related Behaviors and Stress-Induced Reinstatement of Cocaine Conditioned Place Preference. eNeuro 2021; 8:ENEURO.0365-20.2020. [PMID: 33402347 PMCID: PMC7986533 DOI: 10.1523/eneuro.0365-20.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
The striatum plays a key role in both reward-related and affective behaviors and disruptions to this circuit contributes to depression and drug addiction. However, our understanding of the molecular factors that facilitate and modify these processes are incomplete. Striatal function is modulated by G-protein-coupled receptors (GPCRs) that process vast neuromodulatory inputs. GPCR signaling is negatively regulated by regulator of G-protein signaling (Rgs) proteins. In this study, we examine the role of striatal Rgs proteins in depressive-like and reward-related behaviors in male mice. Using a genetic mouse model with specific elimination of Rgs7 in striatal neurons we found that these mice exhibit an anxiolytic-like and antidepressant-like phenotype. In contrast, knock-out of Rgs9, an abundant Rgs protein in the same neuronal population did not affect the behavioral outcome in the depressive-like tests. Mice lacking striatal Rgs7 did not show significant differences in cocaine-induced psychomotor activation, sensitization or conditional place preference (CPP). Interestingly, loss of Rgs7 in the striatum made mice resilient to stress-induced but not drug-induced reinstatement of cocaine CPP. Analysis of striatal proteome revealed that loss of Rgs7 selectively affected expression of several networks, most prominently including proteins involved in translation and vesicular exocytosis. Together, these findings begin to demonstrate the specific contribution of Rgs7 acting in the striatum toward depression as it relates to stress-induced reinstatement of drug use.
Collapse
|
8
|
Heinsbroek JA, De Vries TJ, Peters J. Glutamatergic Systems and Memory Mechanisms Underlying Opioid Addiction. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a039602. [PMID: 32341068 DOI: 10.1101/cshperspect.a039602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Glutamate is the main excitatory neurotransmitter in the brain and is of critical importance for the synaptic and circuit mechanisms that underlie opioid addiction. Opioid memories formed over the course of repeated drug use and withdrawal can become powerful stimuli that trigger craving and relapse, and glutamatergic neurotransmission is essential for the formation and maintenance of these memories. In this review, we discuss the mechanisms by which glutamate, dopamine, and opioid signaling interact to mediate the primary rewarding effects of opioids, and cover the glutamatergic systems and circuits that mediate the expression, extinction, and reinstatement of opioid seeking over the course of opioid addiction.
Collapse
Affiliation(s)
- Jasper A Heinsbroek
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Taco J De Vries
- Amsterdam Neuroscience, Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, VU University, 1081HV Amsterdam, The Netherlands.,Amsterdam Neuroscience, Department of Anatomy and Neurosciences, VU University Medical Center, 1081HZ Amsterdam, The Netherlands
| | - Jamie Peters
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
9
|
Gαo is a major determinant of cAMP signaling in the pathophysiology of movement disorders. Cell Rep 2021; 34:108718. [PMID: 33535037 PMCID: PMC7903328 DOI: 10.1016/j.celrep.2021.108718] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/07/2020] [Accepted: 01/11/2021] [Indexed: 01/20/2023] Open
Abstract
The G protein alpha subunit o (Gαo) is one of the most abundant proteins in the nervous system, and pathogenic mutations in its gene (GNAO1) cause movement disorder. However, the function of Gαo is ill defined mechanistically. Here, we show that Gαo dictates neuromodulatory responsiveness of striatal neurons and is required for movement control. Using in vivo optical sensors and enzymatic assays, we determine that Gαo provides a separate transduction channel that modulates coupling of both inhibitory and stimulatory dopamine receptors to the cyclic AMP (cAMP)-generating enzyme adenylyl cyclase. Through a combination of cell-based assays and rodent models, we demonstrate that GNAO1-associated mutations alter Gαo function in a neuron-type-specific fashion via a combination of a dominant-negative and loss-of-function mechanisms. Overall, our findings suggest that Gαo and its pathological variants function in specific circuits to regulate neuromodulatory signals essential for executing motor programs. Muntean et al. describe biochemical, cellular, and physiological mechanisms by which the heterotrimeric G protein subunit Gαo controls neuromodulatory signaling in the striatum and elucidate mechanisms by which Gαo mutations compromise movements in GNAO1 disorder.
Collapse
|
10
|
Regulators of G protein signalling as pharmacological targets for the treatment of neuropathic pain. Pharmacol Res 2020; 160:105148. [PMID: 32858121 DOI: 10.1016/j.phrs.2020.105148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 07/24/2020] [Accepted: 08/10/2020] [Indexed: 12/30/2022]
Abstract
Neuropathic pain, a specific type of chronic pain resulting from persistent nervous tissue lesions, is a debilitating condition that affects about 7% of the population. This condition remains particularly difficult to treat because of the poor understanding of its underlying mechanisms. Drugs currently used to alleviate this chronic pain syndrome are of limited benefit due to their lack of efficacy and the elevated risk of side effects, especially after a prolonged period of treatment. Although drugs targeting G protein-coupled receptors (GPCR) also have several limitations, such as progressive loss of efficacy due to receptor desensitization or unavoidable side effects due to wide receptor distribution, the identification of several molecular partners that contribute to the fine-tuning of receptor activity has raised new opportunities for the development of alternative therapeutic approaches. Regulators of G protein signalling (RGS) act intracellularly by influencing the coupling process and activity of G proteins, and are amongst the best-characterized physiological modulators of GPCR. Changes in RGS expression have been documented in a range of models of neuropathic pain, or after prolonged treatment with diverse analgesics, and could participate in altered pain processing as well as impaired physiological or pharmacological control of nociceptive signals. The present review summarizes the experimental data that implicates RGS in the development of pain with focus on the pathological mechanisms of neuropathic pain, including the impact of neuropathic lesions on RGS expression and, reciprocally, the influence of modifying RGS on GPCRs involved in the modulation of nociception as well as on the outcome of pain. In this context, we address the question of the relevance of RGS as promising targets in the treatment of neuropathic pain.
Collapse
|
11
|
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 DOI: 10.1124/mol.119.119206] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [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
|
12
|
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 DOI: 10.1074/jbc.rev119.007060] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
13
|
Sutton LP, Muntean BS, Ostrovskaya O, Zucca S, Dao M, Orlandi C, Song C, Xie K, Martemyanov KA. NF1-cAMP signaling dissociates cell type-specific contributions of striatal medium spiny neurons to reward valuation and motor control. PLoS Biol 2019; 17:e3000477. [PMID: 31600280 PMCID: PMC6805008 DOI: 10.1371/journal.pbio.3000477] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/22/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
The striatum plays a fundamental role in motor learning and reward-related behaviors that are synergistically shaped by populations of D1 dopamine receptor (D1R)- and D2 dopamine receptor (D2R)-expressing medium spiny neurons (MSNs). How various neurotransmitter inputs converging on common intracellular pathways are parsed out to regulate distinct behavioral outcomes in a neuron-specific manner is poorly understood. Here, we reveal that distinct contributions of D1R-MSNs and D2R-MSNs towards reward and motor behaviors are delineated by the multifaceted signaling protein neurofibromin 1 (NF1). Using genetic mouse models, we show that NF1 in D1R-MSN modulates opioid reward, whereas loss of NF1 in D2R-MSNs delays motor learning by impeding the formation and consolidation of repetitive motor sequences. We found that motor learning deficits upon NF1 loss were associated with the disruption in dopamine signaling to cAMP in D2R-MSN. Restoration of cAMP levels pharmacologically or chemogenetically rescued the motor learning deficits seen upon NF1 loss in D2R-MSN. Our findings illustrate that multiplex signaling capabilities of MSNs are deployed at the level of intracellular pathways to achieve cell-specific control over behavioral outcomes. A mouse genetic study reveals that the multifaceted signaling protein neurofibromin (known for its role in the human genetic disease neurofibromatosis type 1) plays a key role in differential routing of motor and reward signals in populations of striatal medium spiny neurons.
Collapse
Affiliation(s)
- Laurie P. Sutton
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Brian S. Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Olga Ostrovskaya
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Stefano Zucca
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Maria Dao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Chenghui Song
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Keqiang Xie
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Kirill A. Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
- * E-mail:
| |
Collapse
|
14
|
Muntean BS, Patil DN, Madoux F, Fossetta J, Scampavia L, Spicer TP, Martemyanov KA. A High-Throughput Time-Resolved Fluorescence Energy Transfer Assay to Screen for Modulators of RGS7/Gβ5/R7BP Complex. Assay Drug Dev Technol 2019; 16:150-161. [PMID: 29658790 DOI: 10.1089/adt.2017.839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are excellent drug targets exploited by majority of the Food and Drug Administration-approved medications, but when modulated, are often accompanied by significant adverse effects. Targeting of other elements in GPCR pathways for improved safety and efficacy is thus an unmet need. The strength of GPCR signaling is tightly regulated by regulators of G protein signaling (RGS) proteins, making them attractive drug targets. We focused on a prominent RGS complex in the brain consisting of RGS7 and its binding partners Gβ5 and R7BP. These complexes play critical roles in regulating multiple GPCRs and essential physiological processes, yet no small molecule modulators are currently available to modify its function. In this study, we report a novel high-throughput approach to screen for small molecule modulators of the intramolecular transitions in the RGS7/Gβ5/R7BP complex known to be involved in its allosteric regulation. We developed a time-resolved fluorescence energy transfer-based in vitro assay that utilizes full-length recombinant proteins and shows consistency, excellent assay statistics, and high level of sensitivity. We demonstrated the potential of this approach by screening two compound libraries (LOPAC 1280 and MicroSource Spectrum). This study confirms the feasibility of the chosen strategy for identifying small molecule modulators of RGS7/Gβ5/R7BP complex for impacting signaling downstream of the GPCRs.
Collapse
Affiliation(s)
- Brian S Muntean
- 1 Department of Neuroscience, The Scripps Research Institute , Jupiter, Florida
| | - Dipak N Patil
- 1 Department of Neuroscience, The Scripps Research Institute , Jupiter, Florida
| | - Franck Madoux
- 2 Department of Molecular Medicine, The Scripps Research Institute , Jupiter, Florida
| | | | - Louis Scampavia
- 2 Department of Molecular Medicine, The Scripps Research Institute , Jupiter, Florida
| | - Timothy P Spicer
- 2 Department of Molecular Medicine, The Scripps Research Institute , Jupiter, Florida
| | | |
Collapse
|
15
|
Alugubelly N, Mohammad AN, Edelmann MJ, Nanduri B, Sayed M, Park JW, Carr RL. Proteomic and transcriptional profiling of rat amygdala following social play. Behav Brain Res 2019; 376:112210. [PMID: 31493430 DOI: 10.1016/j.bbr.2019.112210] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 09/03/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Abstract
Social play is the most characteristic form of social interaction which is necessary for adolescents to develop proper cognitive, emotional, and social competency. The information available on neural substrates and the mechanism involved in social play is limited. This study characterized social play by proteomic and transcriptional profiling studies. Social play was performed on male Sprague Dawley rats on postnatal day 38 and protein and gene expression in the amygdala was determined following behavioral testing. The proteomic analysis led to the identification of 170 differentially expressed proteins (p ≤ 0.05) with 67 upregulated and 103 downregulated proteins. The transcriptomic analysis led to the identification of 188 genes (FDR ≤ 0.05) with 55 upregulated and 133 downregulated genes. DAVID analysis of gene/protein expression data revealed that social play altered GABAergic signaling, glutamatergic signaling, and G-protein coupled receptor (GPCR) signaling. These data suggest that the synaptic levels of GABA and glutamate increased during play. Ingenuity Pathway Analysis (IPA) confirmed these alterations. IPA also revealed that differentially expressed genes/proteins in our data had significant over representation of neurotransmitter signaling systems, including the opioid, serotonin, and dopamine systems, suggesting that play alters the systems involved in the regulation of reward. In addition, corticotropin-releasing hormone signaling was altered indicating that an increased level of stress occurs during play. Overall, our data suggest that increased inhibitory GPCR signaling in these neurotransmitter pathways occurs following social play as a physiological response to regulate the induced level of reward and stress and to maintain the excitatory-inhibitory balance in the neurotransmitter systems.
Collapse
Affiliation(s)
- Navatha Alugubelly
- Center for Environmental Health Sciences, MS, USA; Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS, USA
| | - Afzaal N Mohammad
- Center for Environmental Health Sciences, MS, USA; Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS, USA
| | - Mariola J Edelmann
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Bindu Nanduri
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS, USA
| | - Mohammed Sayed
- Department of Computer Engineering and Computer Science, KY, USA
| | - Juw Won Park
- Department of Computer Engineering and Computer Science, KY, USA; KBRIN Bioinformatics Core, University of Louisville, KY, USA.
| | - Russell L Carr
- Center for Environmental Health Sciences, MS, USA; Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS, USA.
| |
Collapse
|
16
|
Israeli R, Asli A, Avital-Shacham M, Kosloff M. RGS6 and RGS7 Discriminate between the Highly Similar Gα i and Gα o Proteins Using a Two-Tiered Specificity Strategy. J Mol Biol 2019; 431:3302-3311. [PMID: 31153905 DOI: 10.1016/j.jmb.2019.05.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/12/2019] [Accepted: 05/23/2019] [Indexed: 11/15/2022]
Abstract
RGS6 and RGS7 are regulators of G protein signaling (RGS) proteins that inactivate heterotrimeric (αβγ) G proteins and mediate diverse biological functions, such as cardiac and neuronal signaling. Uniquely, both RGS6 and RGS7 can discriminate between Gαo and Gαi1-two similar Gα subunits that belong to the same Gi sub-family. Here, we show that the isolated RGS domains of RGS6 and RGS7 are sufficient to achieve this specificity. We identified three specific RGS6/7 "disruptor residues" that can attenuate RGS interactions toward Gα subunits and demonstrated that their insertion into a representative high-activity RGS causes a significant, yet non-specific, reduction in activity. We further identified a unique "modulatory" residue that bypasses this negative effect, specifically toward Gαo. Hence, the exquisite specificity of RGS6 and RGS7 toward closely related Gα subunits is achieved via a two-tier specificity system, whereby a Gα-specific modulatory motif overrides the inhibitory effect of non-specific disruptor residues. Our findings expand the understanding of the molecular toolkit used by the RGS family to achieve specific interactions with selected Gα subunits-emphasizing the functional importance of the RGS domain in determining the activity and selectivity of RGS R7 sub-family members toward particular Gα subunits.
Collapse
Affiliation(s)
- Ran Israeli
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Ali Asli
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Meirav Avital-Shacham
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Mickey Kosloff
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
17
|
Orlandi C, Sutton LP, Muntean BS, Song C, Martemyanov KA. Homeostatic cAMP regulation by the RGS7 complex controls depression-related behaviors. Neuropsychopharmacology 2019; 44:642-653. [PMID: 30546127 PMCID: PMC6333837 DOI: 10.1038/s41386-018-0238-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 01/27/2023]
Abstract
Affective disorders arise from abnormal responses of the brain to prolonged exposure to challenging environmental stimuli. Recent work identified the orphan receptor GPR158 as a molecular link between chronic stress and depression. Here we reveal a non-canonical mechanism by which GPR158 exerts its effects on stress-induced depression by the complex formation with Regulator of G protein Signaling 7 (RGS7). Chronic stress promotes membrane recruitment of RGS7 via GPR158 in the medial prefrontal cortex (mPFC). The resultant complex suppresses homeostatic regulation of cAMP by inhibitory GPCRs in the region. Accordingly, RGS7 loss in mice induces an antidepressant-like phenotype and resiliency to stress, whereas its restoration within the mPFC is sufficient to rescue this phenotype in a GPR158-dependent way. These findings mechanistically link the unusual orphan receptor-RGS complex to a major stress mediator, the cAMP system and suggest new avenues for pharmacological interventions in affective disorders.
Collapse
Affiliation(s)
- Cesare Orlandi
- 0000000122199231grid.214007.0Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Laurie P. Sutton
- 0000000122199231grid.214007.0Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Brian S. Muntean
- 0000000122199231grid.214007.0Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Chenghui Song
- 0000000122199231grid.214007.0Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458 USA
| | - Kirill A. Martemyanov
- 0000000122199231grid.214007.0Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458 USA
| |
Collapse
|
18
|
Patil DN, Rangarajan ES, Novick SJ, Pascal BD, Kojetin DJ, Griffin PR, Izard T, Martemyanov KA. Structural organization of a major neuronal G protein regulator, the RGS7-Gβ5-R7BP complex. eLife 2018; 7:e42150. [PMID: 30540250 PMCID: PMC6310461 DOI: 10.7554/elife.42150] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/12/2018] [Indexed: 01/03/2023] Open
Abstract
Signaling by the G-protein-coupled receptors (GPCRs) plays fundamental role in a vast number of essential physiological functions. Precise control of GPCR signaling requires action of regulators of G protein signaling (RGS) proteins that deactivate heterotrimeric G proteins. RGS proteins are elaborately regulated and comprise multiple domains and subunits, yet structural organization of these assemblies is poorly understood. Here, we report a crystal structure and dynamics analyses of the multisubunit complex of RGS7, a major regulator of neuronal signaling with key roles in controlling a number of drug target GPCRs and links to neuropsychiatric disease, metabolism, and cancer. The crystal structure in combination with molecular dynamics and mass spectrometry analyses reveals unique organizational features of the complex and long-range conformational changes imposed by its constituent subunits during allosteric modulation. Notably, several intermolecular interfaces in the complex work in synergy to provide coordinated modulation of this key GPCR regulator.
Collapse
Affiliation(s)
- Dipak N Patil
- Department of NeuroscienceThe Scripps Research InstituteJupiterUnited States
| | - Erumbi S Rangarajan
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteJupiterUnited States
| | - Scott J Novick
- Department of Molecular MedicineThe Scripps Research InstituteJupiterUnited States
| | - Bruce D Pascal
- Department of Molecular MedicineThe Scripps Research InstituteJupiterUnited States
| | - Douglas J Kojetin
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteJupiterUnited States
| | - Patrick R Griffin
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteJupiterUnited States
- Department of Molecular MedicineThe Scripps Research InstituteJupiterUnited States
| | - Tina Izard
- Department of Integrative Structural and Computational BiologyThe Scripps Research InstituteJupiterUnited States
| | | |
Collapse
|
19
|
Squires KE, Montañez-Miranda C, Pandya RR, Torres MP, Hepler JR. Genetic Analysis of Rare Human Variants of Regulators of G Protein Signaling Proteins and Their Role in Human Physiology and Disease. Pharmacol Rev 2018; 70:446-474. [PMID: 29871944 DOI: 10.1124/pr.117.015354] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regulators of G protein signaling (RGS) proteins modulate the physiologic actions of many neurotransmitters, hormones, and other signaling molecules. Human RGS proteins comprise a family of 20 canonical proteins that bind directly to G protein-coupled receptors/G protein complexes to limit the lifetime of their signaling events, which regulate all aspects of cell and organ physiology. Genetic variations account for diverse human traits and individual predispositions to disease. RGS proteins contribute to many complex polygenic human traits and pathologies such as hypertension, atherosclerosis, schizophrenia, depression, addiction, cancers, and many others. Recent analysis indicates that most human diseases are due to extremely rare genetic variants. In this study, we summarize physiologic roles for RGS proteins and links to human diseases/traits and report rare variants found within each human RGS protein exome sequence derived from global population studies. Each RGS sequence is analyzed using recently described bioinformatics and proteomic tools for measures of missense tolerance ratio paired with combined annotation-dependent depletion scores, and protein post-translational modification (PTM) alignment cluster analysis. We highlight selected variants within the well-studied RGS domain that likely disrupt RGS protein functions and provide comprehensive variant and PTM data for each RGS protein for future study. We propose that rare variants in functionally sensitive regions of RGS proteins confer profound change-of-function phenotypes that may contribute, in newly appreciated ways, to complex human diseases and/or traits. This information provides investigators with a valuable database to explore variation in RGS protein function, and for targeting RGS proteins as future therapeutic targets.
Collapse
Affiliation(s)
- Katherine E Squires
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Carolina Montañez-Miranda
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Rushika R Pandya
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Matthew P Torres
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - John R Hepler
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| |
Collapse
|
20
|
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: 3.7] [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
|
21
|
Abstract
This paper is the thirty-ninth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2016 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, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and CUNY Neuroscience Collaborative, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
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: 4.0] [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
|
Doyen PJ, Vergouts M, Pochet A, Desmet N, van Neerven S, Brook G, Hermans E. Inflammation-associated regulation of RGS in astrocytes and putative implication in neuropathic pain. J Neuroinflammation 2017; 14:209. [PMID: 29078779 PMCID: PMC5658970 DOI: 10.1186/s12974-017-0971-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/26/2017] [Indexed: 02/07/2023] Open
Abstract
Background Regulators of G-protein signaling (RGS) are major physiological modulators of G-protein-coupled receptors (GPCR) signaling. Several GPCRs expressed in both neurons and astrocytes participate in the central control of pain processing, and the reduced efficacy of analgesics in neuropathic pain conditions may rely on alterations in RGS function. The expression and the regulation of RGS in astrocytes is poorly documented, and we herein hypothesized that neuroinflammation which is commonly observed in neuropathic pain could influence RGS expression in astrocytes. Methods In a validated model of neuropathic pain, the spared nerve injury (SNI), the regulation of RGS2, RGS3, RGS4, and RGS7 messenger RNA (mRNA) was examined up to 3 weeks after the lesion. Changes in the expression of the same RGS were also studied in cultured astrocytes exposed to defined activation protocols or to inflammatory cytokines. Results We evidenced a differential regulation of these RGS in the lumbar spinal cord of animals undergoing SNI. In particular, RGS3 appeared upregulated at early stages after the lesion whereas expression of RGS2 and RGS4 was decreased at later stages. Decrease in RGS7 expression was already observed after 3 days and outlasted until 21 days after the lesion. In cultured astrocytes, we observed that changes in the culture conditions distinctly influenced the constitutive expression of these RGS. Also, brief exposures (4 to 8 h) to either interleukin-1β, interleukin-6, or tumor necrosis factor α caused rapid changes in the mRNA levels of the RGS, which however did not strictly recapitulate the regulations observed in the spinal cord of lesioned animals. Longer exposure (48 h) to inflammatory cytokines barely influenced RGS expression, confirming the rapid but transient regulation of these cell signaling modulators. Conclusion Changes in the environment of astrocytes mimicking the inflammation observed in the model of neuropathic pain can affect RGS expression. Considering the role of astrocytes in the onset and progression of neuropathic pain, we propose that the inflammation-mediated modulation of RGS in astrocytes constitutes an adaptive mechanism in a context of neuroinflammation and may participate in the regulation of nociception.
Collapse
Affiliation(s)
- Pierre J Doyen
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Maxime Vergouts
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Amandine Pochet
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Nathalie Desmet
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Sabien van Neerven
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Gary Brook
- Institute for Neuropathology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Emmanuel Hermans
- Neuropharmacology, Institute of Neuroscience, Université Catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium.
| |
Collapse
|
24
|
Park TY, Nishida KS, Wilson CM, Jaiswal S, Scott J, Hoy AR, Selwyn RG, Dardzinski BJ, Choi KH. Effects of isoflurane anesthesia and intravenous morphine self-administration on regional glucose metabolism ([ 18 F]FDG-PET) of male Sprague-Dawley rats. Eur J Neurosci 2017; 45:922-931. [PMID: 28196306 DOI: 10.1111/ejn.13542] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 02/07/2017] [Accepted: 02/08/2017] [Indexed: 01/22/2023]
Abstract
Although certain drugs of abuse are known to disrupt brain glucose metabolism (BGluM), the effects of opiates on BGluM are not well characterized. Moreover, preclinical positron emission tomography (PET) studies anesthetize animals during the scan, which limits clinical applications. We investigated the effects of (i) isoflurane anesthesia and (ii) intravenous morphine self-administration (MSA) on BGluM in rats. Jugular vein cannulated adult male Sprague-Dawley rats self-administered either saline (SSA) or morphine (0.5 mg/kg/infusion, 4 h/day for 12 days). All animals were scanned twice with [18 F]-fluoro-deoxy-glucose (FDG)-PET/CT at a baseline and at 2-day withdrawal from self-administration. After the IV injection of FDG, one batch of animals (n = 14) was anesthetized with isoflurane and the other batch (n = 16) was kept awake during the FDG uptake (45 min). After FDG uptake, all animals were anesthetized in order to perform a PET/CT scan (30 min). Isoflurane anesthesia, as compared to the awake condition, reduced BGluM in the olfactory, cortex, thalamus, and basal ganglia, while increasing BGluM in the midbrain, hypothalamus, hippocampus, and cerebellum. Morphine self-administered animals exhibited withdrawal signs (piloerection and increased defecation), drug seeking, and locomotor stimulation to morphine (0.5 mg/kg) during the 2 day withdrawal. The BGluM in the striatum was increased in the MSA group as compared to the SSA group; this effect was observed only in the isoflurane anesthesia, not the awake condition. These findings suggest that the choice of the FDG uptake condition may be important in preclinical PET studies and increased BGluM in the striatum may be associated with opiate seeking in withdrawal.
Collapse
Affiliation(s)
- Thomas Y Park
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Center for the Study of Traumatic Stress, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kevin S Nishida
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Center for the Study of Traumatic Stress, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Colin M Wilson
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Radiology, University of New Mexico, Albuquerque, NM, USA
| | - Shalini Jaiswal
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jessica Scott
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Andrew R Hoy
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Reed G Selwyn
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Radiology, University of New Mexico, Albuquerque, NM, USA.,Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Bernard J Dardzinski
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kwang H Choi
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.,Center for the Study of Traumatic Stress, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
25
|
Aguado C, Orlandi C, Fajardo-Serrano A, Gil-Minguez M, Martemyanov KA, Luján R. Cellular and Subcellular Localization of the RGS7/Gβ5/R7BP Complex in the Cerebellar Cortex. Front Neuroanat 2016; 10:114. [PMID: 27965545 PMCID: PMC5127842 DOI: 10.3389/fnana.2016.00114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/10/2016] [Indexed: 11/13/2022] Open
Abstract
A member of regulator of G-protein signaling family, RGS7, is an essential modulator of signaling through GABAB receptors. RGS7 functions as a macromolecular complex with type 5 G protein β (Gβ5) and R7 binding protein (R7BP) to control the localization and function of the resultant heterotrimeric complexes. Here, we used co-immunoprecipitation, in situ hybridization, histoblot and immunohistochemical techniques at the light and electron microscopic level to advance understanding of RGS7-Gβ5-R7BP complexes in the central nervous system, focusing on distinct neuronal populations in the cerebellar cortex. Histoblot analysis showed that RGS7, Gβ5 and R7BP proteins were widely expressed in the brain, with mostly an overlapping pattern and showing a high expression level in the molecular layer of the cerebellar cortex. Co-immunoprecipitation experiments established that the RGS7/Gβ5 forms complexes with R7BP in the cerebellum. At the cellular level, RGS7 and R7BP mRNAs were expressed at the highest level in Purkinje cells (PCs) and Golgi cells, and at low levels in granule cells. Immunohistochemistry confirmed that labeling for RGS7, Gβ5 and R7BP were present in the three neuronal populations and concentrated in dendrites and spines. At the electron microscopic level, immunolabeling for RGS7, Gβ5 and R7BP proteins was found both at postsynaptic and presynaptic sites and showed similar distribution patterns. Immunoreactivity for the three proteins was mostly localized along the extrasynaptic plasma membrane of dendritic shafts and spines of PCs and to a lesser extent, in axon terminals (AT) establishing excitatory synapses. Quantitative analysis of immunogold particles for RGS7, Gβ5 and R7BP revealed that they are non-uniformly distributed along the surface of PCs, and show enrichment around excitatory synapses on dendritic spines. We further report that deletion of R7BP in mice reduced the targeting of both RGS7 and Gβ5 to the plasma membrane. Altogether, these data support the existence of macromolecular complexes composed of RGS7-Gβ5-R7BP in PCs. The location at post- and pre-synaptic sites in PCs spines-parallel fiber synapses suggests their involvement in the modulation of glutamatergic neurotransmission in the cerebellar cortex.
Collapse
Affiliation(s)
- Carolina Aguado
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Spain
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute Jupiter, FL, USA
| | - Ana Fajardo-Serrano
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Spain
| | - Mercedes Gil-Minguez
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Spain
| | | | - Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha Albacete, Spain
| |
Collapse
|
26
|
Muntean BS, Martemyanov KA. Association with the Plasma Membrane Is Sufficient for Potentiating Catalytic Activity of Regulators of G Protein Signaling (RGS) Proteins of the R7 Subfamily. J Biol Chem 2016; 291:7195-204. [PMID: 26811338 PMCID: PMC4807299 DOI: 10.1074/jbc.m115.713446] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 01/21/2016] [Indexed: 12/23/2022] Open
Abstract
Regulators of G protein Signaling (RGS) promote deactivation of heterotrimeric G proteins thus controlling the magnitude and kinetics of responses mediated by G protein-coupled receptors (GPCR). In the nervous system, RGS7 and RGS9-2 play essential role in vision, reward processing, and movement control. Both RGS7 and RGS9-2 belong to the R7 subfamily of RGS proteins that form macromolecular complexes with R7-binding protein (R7BP). R7BP targets RGS proteins to the plasma membrane and augments their GTPase-accelerating protein (GAP) activity, ultimately accelerating deactivation of G protein signaling. However, it remains unclear if R7BP serves exclusively as a membrane anchoring subunit or further modulates RGS proteins to increase their GAP activity. To directly answer this question, we utilized a rapidly reversible chemically induced protein dimerization system that enabled us to control RGS localization independent from R7BP in living cells. To monitor kinetics of Gα deactivation, we coupled this strategy with measuring changes in the GAP activity by bioluminescence resonance energy transfer-based assay in a cellular system containing μ-opioid receptor. This approach was used to correlate changes in RGS localization and activity in the presence or absence of R7BP. Strikingly, we observed that RGS activity is augmented by membrane recruitment, in an orientation independent manner with no additional contributions provided by R7BP. These findings argue that the association of R7 RGS proteins with the membrane environment provides a major direct contribution to modulation of their GAP activity.
Collapse
Affiliation(s)
- Brian S Muntean
- From the Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458
| | - Kirill A Martemyanov
- From the Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458
| |
Collapse
|
27
|
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: 25] [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
|