1
|
Al Aameri RFH, Alanisi EMA, Al Sallami D, Alberts I, Tischkau S, Rybak LP, Ramkumar V. Role of RGS17 in cisplatin-induced cochlear inflammation and ototoxicity via caspase-3 activation. Front Immunol 2025; 16:1470625. [PMID: 40061942 PMCID: PMC11885124 DOI: 10.3389/fimmu.2025.1470625] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/30/2025] [Indexed: 05/13/2025] Open
Abstract
Cisplatin is a chemotherapy drug used to treat different solid tumors, including ovarian, bladder, lung, and head and neck cancers. One of its significant side effects is ototoxicity, especially when high doses are required. Cisplatin-induced ototoxicity is associated with increased cochlear cell death resulting from DNA damage, caspase activation, oxidative stress, inflammation, and glutamate excitotoxicity. The regulator of G protein signaling 17 (RGS17), a member of the RGS-RZ subfamily, hastens the hydrolysis of GTP to GDP on the Gα subunit. In the current study, we demonstrate the role of RGS17 in cisplatin-induced cochlear inflammation and ototoxicity. C57BL/6J mice treated with two cycles of cisplatin (3.5 mg/kg) showed a significant elevation in ABR thresholds, along with loss of outer hair cells and inner hair cells synapse. Furthermore, immunohistochemical analysis revealed that cisplatin administration upregulates CXCL1, accompanied by an increase in the number of CD45 and CD68-positive immune cells. On the other hand, RGS17 knockout in hair cells protects against cisplatin-induced elevation of ABR thresholds, outer hair cell loss, cochlear inflammation, and inner hair cell synaptopathy. Moreover, RGS17 knockout downregulates CXCL1 immunolabeling and decreases the number of CD45 and CD68-positive immune cells induced by cisplatin. These results suggest that RGS17 is implicated in cisplatin ototoxicity, potentially by initiating the immune cascade, and indicate RGS17 as a relevant target for treating cisplatin ototoxicity.
Collapse
Affiliation(s)
- Raheem F. H. Al Aameri
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Biology, Wasit University, College of Applied Science, Wasit, Iraq
| | | | - Dheyaa Al Sallami
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Ian Alberts
- Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Shelley Tischkau
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Leonard P. Rybak
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Vickram Ramkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| |
Collapse
|
2
|
Wang J, Liang M, Shang Q, Qian H, An R, Liu H, Shao G, Li T, Liu X. Psilocin suppresses methamphetamine-induced hyperlocomotion and acquisition of conditioned place preference via D2R-mediated ERK signaling. CNS Neurosci Ther 2023; 29:831-841. [PMID: 36627756 PMCID: PMC9928547 DOI: 10.1111/cns.14054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/20/2022] [Accepted: 10/30/2022] [Indexed: 01/12/2023] Open
Abstract
AIM Psilocin is an active metabolite form of psilocybin and exerts psychoactive effects. Recent studies suggest that psilocin may have regulatory effects on abuse drugs, but the mechanisms remain unclear. In this study, we want to explore the effects of psilocin on methamphetamine (METH)-induced alterations of behavior in mice and its molecular mechanisms. METHODS Acute METH administration model and conditioned place preference (CPP) model were used to investigate the effects of psilocin on METH-induced alterations of behavior. Western blot was used to detect the expression of proteins. RESULTS In the acute 2 mg/kg METH administration model, 1 mg/kg psilocin counteracted METH-induced elevation of activity. In the 1 mg/kg METH-induced CPP model, 1 mg/kg psilocin inhibited CPP formation during the acquisition phase. However, psilocin did not impact METH extinction and relapse. Molecular results showed that the regulatory effect of psilocin on METH was underscored by altered expression of dopamine 2 receptor (D2R) and phosphorylated extra-cellular signal-regulated kinase (p-ERK) in the prefrontal cortex (PFC), nucleus accumbens (NAc), and ventral tegmental area (VTA). Trifluoperazine (TFP)-2HCl is a D2R inhibitor, and SCH772984 is a selective extra-cellular signal-regulated kinase (ERK) inhibitor that effectively inhibits ERK1/2 phosphorylation. The results indicated that 2 mg/kg TFP-2HCl and 10 mg/kg SCH772984 blocked METH-induced hyperactivity and acquisition of METH-induced CPP. CONCLUSION Psilocin has regulatory effects on METH-induced alterations of behavior in mice via D2R-mediated signal regulation of ERK phosphorylation.
Collapse
Affiliation(s)
- Jing Wang
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anChina,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation HarborXi'an Jiaotong UniversityXi'anChina
| | - Min Liang
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anChina,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation HarborXi'an Jiaotong UniversityXi'anChina
| | - Qing Shang
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anChina,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation HarborXi'an Jiaotong UniversityXi'anChina
| | - Hongyan Qian
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anChina,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation HarborXi'an Jiaotong UniversityXi'anChina
| | - Ran An
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anChina,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation HarborXi'an Jiaotong UniversityXi'anChina
| | - Hua Liu
- Key Laboratory of Forensic ToxicologyBeijingChina
| | - Gaojie Shao
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anChina,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation HarborXi'an Jiaotong UniversityXi'anChina
| | - Tao Li
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anChina,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation HarborXi'an Jiaotong UniversityXi'anChina
| | - Xinshe Liu
- College of Forensic MedicineXi'an Jiaotong University Health Science CenterXi'anChina,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation HarborXi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
3
|
Prenatal Amphetamine-Induced Dopaminergic Alteration in a Gender- and Estrogen-Dependent Manner. Neurochem Res 2022; 47:1317-1328. [PMID: 35147850 DOI: 10.1007/s11064-022-03531-1] [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: 10/15/2021] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 10/19/2022]
Abstract
Prenatal exposure to amphetamine induces changes in dopamine receptors in mesolimbic areas and alters locomotor response to amphetamine during adulthood. Sex differences have been reported in amphetamine-induced brain activity and stress sensitivity. We evaluated the effects of prenatal amphetamine exposure on locomotor activity, dopamine receptors and tyrosine hydroxylase mRNA expression in nucleus accumbens and caudate-putamen in response to amphetamine challenge in adult female and male rats. The role of estrogen in the response to restraint stress was analyzed in ovariectomized, prenatally amphetamine-exposed rats. Pregnant rats were treated with D-amphetamine during days 15-21 of gestation. Nucleus accumbens and caudate-putamen were processed for mRNA determination by real-time PCR. In nucleus accumbens, higher mRNA dopamine (D3) receptor expression was found in basal and D-amphetamine-challenge conditions in female than male, and prenatal amphetamine increased the difference. No sex differences were observed in caudate-putamen. Basal saline-treated females showed higher locomotor activity than males. Amphetamine challenge in prenatally amphetamine-exposed rats increased locomotor activity in males and reduced it in females. In nucleus accumbens, estrogen diminished mRNA D1, D2 and D3 receptor expression in basal, and D1 and D3 in ovariectomized stressed rats. Estrogen prevented the increase in tyrosine hydroxylase expression induced by stress in ovariectomized prenatally exposed rats. In conclusion, estrogen modulates mRNA levels of D1, D2 and D3 receptors and tyrosine hydroxylase expression in nucleus accumbens; prenatal amphetamine-exposure effects on D3 receptors and behavioral responses were gender dependent.
Collapse
|
4
|
Danielsson K, Lagström O, Ericson M, Söderpalm B, Adermark L. Subregion-specific effects on striatal neurotransmission and dopamine-signaling by acute and repeated amphetamine exposure. Neuropharmacology 2021; 194:108638. [PMID: 34116108 DOI: 10.1016/j.neuropharm.2021.108638] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/19/2021] [Accepted: 06/03/2021] [Indexed: 11/29/2022]
Abstract
Repeated administration of psychostimulants, such as amphetamine, is associated with a progressive increased sensitivity to some of the drug's effects, but tolerance towards others. We hypothesized that these adaptations in part could be linked to differential effects by amphetamine on dopaminergic signaling in striatal subregions. To test this theory, acute and long-lasting changes in dopaminergic neurotransmission were assessed in the nucleus accumbens (nAc) and the dorsomedial striatum (DMS) following amphetamine exposure in Wistar rats. By means of in vivo microdialysis, dopamine release induced by local administration of amphetamine was monitored in nAc and DMS of amphetamine naïve rats, and in rats subjected to five days of systemic amphetamine administration (2.0 mg/kg/day) followed by two weeks of withdrawal. In parallel, ex vivo electrophysiology was conducted to outline the effect of acute and repeated amphetamine exposure on striatal neurotransmission. The data shows that amphetamine increases dopamine in a concentration-dependent and subregion-specific manner. Furthermore, repeated administration of amphetamine followed by abstinence resulted in a selective decrease in baseline dopamine in the nAc, and a potentiation of the relative dopamine elevation after systemic amphetamine in the same area. Ex vivo electrophysiology demonstrated decreased excitatory neurotransmission in brain slices from amphetamine-treated animals, and a nAc selective shift in the responsiveness to the dopamine D2-receptor agonist quinpirole. These selective effects on dopamine signaling seen in striatal subregions after repeated drug exposure may partially explain why tolerance develops to the rewarding effects, but not towards the psychosis inducing properties of amphetamine.
Collapse
Affiliation(s)
- Klara Danielsson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Oona Lagström
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden; Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden; Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy University of Gothenburg, Sweden.
| |
Collapse
|
5
|
Differential Impact of Inhibitory G-Protein Signaling Pathways in Ventral Tegmental Area Dopamine Neurons on Behavioral Sensitivity to Cocaine and Morphine. eNeuro 2021; 8:ENEURO.0081-21.2021. [PMID: 33707203 PMCID: PMC8114902 DOI: 10.1523/eneuro.0081-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/21/2022] Open
Abstract
Drugs of abuse engage overlapping but distinct molecular and cellular mechanisms to enhance dopamine (DA) signaling in the mesocorticolimbic circuitry. DA neurons of the ventral tegmental area (VTA) are key substrates of drugs of abuse and have been implicated in addiction-related behaviors. Enhanced VTA DA neurotransmission evoked by drugs of abuse can engage inhibitory G-protein-dependent feedback pathways, mediated by GABAB receptors (GABABRs) and D2 DA receptors (D2Rs). Chemogenetic inhibition of VTA DA neurons potently suppressed baseline motor activity, as well as the motor-stimulatory effect of cocaine and morphine, confirming the critical influence of VTA DA neurons and inhibitory G-protein signaling in these neurons on this addiction-related behavior. To resolve the relative influence of GABABR-dependent and D2R-dependent signaling pathways in VTA DA neurons on behavioral sensitivity to drugs of abuse, we developed a neuron-specific viral CRISPR/Cas9 approach to ablate D2R and GABABR in VTA DA neurons. Ablation of GABABR or D2R did not impact baseline physiological properties or excitability of VTA DA neurons, but it did preclude the direct somatodendritic inhibitory influence of GABABR or D2R activation. D2R ablation potentiated the motor-stimulatory effect of cocaine in male and female mice, whereas GABABR ablation selectively potentiated cocaine-induced activity in male subjects only. Neither D2R nor GABABR ablation impacted morphine-induced motor activity. Collectively, our data show that cocaine and morphine differ in the extent to which they engage inhibitory G-protein-dependent feedback pathways in VTA DA neurons and highlight key sex differences that may impact susceptibility to various facets of addiction.
Collapse
|
6
|
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
|
7
|
Doyle MA, Stark AR, Fejes-Tóth G, Náray-Fejes-Tóth A, Mazei-Robison MS. Behavioral effects of SGK1 knockout in VTA and dopamine neurons. Sci Rep 2020; 10:14751. [PMID: 32901079 PMCID: PMC7478959 DOI: 10.1038/s41598-020-71681-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
Drugs of abuse cause significant neuroadaptations within the ventral tegmental area (VTA), with alterations in gene expression tied to changes in reward behavior. Serum- and glucocorticoid-inducible kinase 1 (SGK1) transcription, catalytic activity, and phosphorylation are upregulated in the VTA by chronic cocaine or morphine treatment, positioning SGK1 as a critical mediator of reward behavior. Using transgenic mouse models, we investigated the effect of SGK1 knockout in the VTA and in dopamine (DA) neurons to evaluate the necessity of protein expression for natural and drug reward behaviors. SGK1 knockdown in the VTA did not impact reward behaviors. Given VTA cellular heterogeneity, we also investigated a DA neuron-specific SGK1 knockout (KO). DA SGK1 KO significantly decreased body weight of adult mice as well as increased general locomotor activity; however, reward behaviors were similarly unaltered. Given that SGK1 mutants virally overexpressed in the VTA are capable of altering drug-associated behavior, our current results suggest that changes in SGK1 protein signaling may be distinct from expression. This work yields novel information on the impact of SGK1 deletion, critical for understanding the role of SGK1 signaling in the central nervous system and evaluating SGK1 as a potential therapeutic target for treatment of substance use disorders.
Collapse
Affiliation(s)
- Marie A Doyle
- Neuroscience Program, Michigan State University, 766 Service Rd, ISTB 5017, East Lansing, MI, 48824, USA
| | - Ali R Stark
- Neuroscience Program, Michigan State University, 766 Service Rd, ISTB 5017, East Lansing, MI, 48824, USA
| | - Geza Fejes-Tóth
- Department of Molecular and Systems Biology, Dartmouth University, Hanover, USA
| | | | - Michelle S Mazei-Robison
- Neuroscience Program, Michigan State University, 766 Service Rd, ISTB 5017, East Lansing, MI, 48824, USA.
- Department of Physiology, Michigan State University, East Lansing, USA.
| |
Collapse
|
8
|
Chen R, Ferris MJ, Wang S. Dopamine D2 autoreceptor interactome: Targeting the receptor complex as a strategy for treatment of substance use disorder. Pharmacol Ther 2020; 213:107583. [PMID: 32473160 PMCID: PMC7434700 DOI: 10.1016/j.pharmthera.2020.107583] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Dopamine D2 autoreceptors (D2ARs), located in somatodendritic and axon terminal compartments of dopamine (DA) neurons, function to provide a negative feedback regulatory control on DA neuron firing, DA synthesis, reuptake and release. Dysregulation of D2AR-mediated DA signaling is implicated in vulnerability to substance use disorder (SUD). Due to the extreme low abundance of D2ARs compared to postsynaptic D2 receptors (D2PRs) and the lack of experimental tools to differentiate the signaling of D2ARs from D2PRs, the regulation of D2ARs by drugs of abuse is poorly understood. The recent availability of conditional D2AR knockout mice and newly developed virus-mediated gene delivery approaches have provided means to specifically study the function of D2ARs at the molecular, cellular and behavioral levels. There is a growing revelation of novel mechanisms and new proteins that mediate D2AR activity, suggesting that D2ARs act cooperatively with an array of membrane and intracellular proteins to tightly control DA transmission. This review highlights D2AR-interacting partners including transporters, G-protein-coupled receptors, ion channels, intracellular signaling modulators, and protein kinases. The complexity of the D2AR interaction network illustrates the functional divergence of D2ARs. Pharmacological targeting of multiple D2AR-interacting partners may be more effective to restore disrupted DA homeostasis by drugs of abuse.
Collapse
Affiliation(s)
- Rong Chen
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America.
| | - Mark J Ferris
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America
| | - Shiyu Wang
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America
| |
Collapse
|
9
|
Almutairi F, Lee JK, Rada B. Regulator of G protein signaling 10: Structure, expression and functions in cellular physiology and diseases. Cell Signal 2020; 75:109765. [PMID: 32882407 PMCID: PMC7579743 DOI: 10.1016/j.cellsig.2020.109765] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 01/22/2023]
Abstract
Regulator of G protein signaling 10 (RGS10) belongs to the superfamily of RGS proteins, defined by the presence of a conserved RGS domain that canonically binds and deactivates heterotrimeric G-proteins. RGS proteins act as GTPase activating proteins (GAPs), which accelerate GTP hydrolysis on the G-protein α subunits and result in termination of signaling pathways downstream of G protein-coupled receptors. RGS10 is the smallest protein of the D/R12 subfamily and selectively interacts with Gαi proteins. It is widely expressed in many cells and tissues, with the highest expression found in the brain and immune cells. RGS10 expression is transcriptionally regulated via epigenetic mechanisms. Although RGS10 lacks multiple of the defined regulatory domains found in other RGS proteins, RGS10 contains post-translational modification sites regulating its expression, localization, and function. Additionally, RGS10 is a critical protein in the regulation of physiological processes in multiple cells, where dysregulation of its expression has been implicated in various diseases including Parkinson's disease, multiple sclerosis, osteopetrosis, chemoresistant ovarian cancer and cardiac hypertrophy. This review summarizes RGS10 features and its regulatory mechanisms, and discusses the known functions of RGS10 in cellular physiology and pathogenesis of several diseases.
Collapse
Affiliation(s)
- Faris Almutairi
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Jae-Kyung Lee
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| |
Collapse
|
10
|
Luessen DJ, Sun H, McGinnis MM, McCool BA, Chen R. Chronic intermittent ethanol exposure selectively alters the expression of Gα subunit isoforms and RGS subtypes in rat prefrontal cortex. Brain Res 2017; 1672:106-112. [PMID: 28736108 DOI: 10.1016/j.brainres.2017.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 10/19/2022]
Abstract
Chronic alcohol exposure induces pronounced changes in GPCR-mediated G-protein signaling. Recent microarray and RNA-seq analyses suggest associations between alcohol abuse and the expression of genes involved in G-protein signaling. The activity of G-proteins (e.g. Gαi/o and Gαq) is negatively modulated by regulator of G-protein signaling (RGS) proteins which are implicated in drugs of abuse including alcohol. The present study used 7days of chronic intermittent ethanol exposure followed by 24h withdrawal (CIE) to investigate changes in mRNA and protein levels of G-protein subunit isoforms and RGS protein subtypes in rat prefrontal cortex, a region associated with cognitive deficit attributed to excessive alcohol drinking. We found that this ethanol paradigm induced differential expression of Gα subunits and RGS subtypes. For example, there were increased mRNA and protein levels of Gαi1/3 subunits and no changes in the expression of Gαs and Gαq subunits in ethanol-treated animals. Moreover, CIE increased the mRNA but not the protein levels of Gαo. Additionally, a modest increase in Gαi2 mRNA level by CIE was accompanied by a pronounced increase in its protein level. Interestingly, we found that CIE increased mRNA and protein levels of RGS2, RGS4, RGS7 and RGS19 but had no effect on the expression of RGS5, RGS6, RGS8, RGS12 or RGS17. Changes in the expression of Gα subunits and RGS subtypes could contribute to the functional alterations of certain GPCRs following chronic ethanol exposure. The present study suggests that RGS proteins may be potential new targets for intervention of alcohol abuse via modification of Gα-mediated GPCR function.
Collapse
Affiliation(s)
- D J Luessen
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - H Sun
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - M M McGinnis
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - B A McCool
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - R Chen
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA.
| |
Collapse
|
11
|
Ledonne A, Mercuri NB. Current Concepts on the Physiopathological Relevance of Dopaminergic Receptors. Front Cell Neurosci 2017; 11:27. [PMID: 28228718 PMCID: PMC5296367 DOI: 10.3389/fncel.2017.00027] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/30/2017] [Indexed: 12/21/2022] Open
Abstract
Dopamine (DA) is a key neurotransmitter modulating essential functions of the central nervous system (CNS), like voluntary movement, reward, several cognitive functions and goal-oriented behaviors. The factual relevance of DAergic transmission can be well appreciated by considering that its dysfunction is recognized as a core alteration in several devastating neurological and psychiatric disorders, including Parkinson’s disease (PD) and associated movement disorders, as well as, schizophrenia, bipolar disorder, attention deficit hyperactivity disorder (ADHD) and addiction. Here we present an overview of the current knowledge on the involvement of DAergic receptors in the regulation of key physiological brain activities, and the consequences of their dysfunctions in brain disorders such as PD, schizophrenia and addiction.
Collapse
Affiliation(s)
- Ada Ledonne
- Department of Experimental Neuroscience, Santa Lucia Foundation Rome, Italy
| | - Nicola B Mercuri
- Department of Experimental Neuroscience, Santa Lucia FoundationRome, Italy; Department of Systems Medicine, University of Rome "Tor Vergata"Rome, Italy
| |
Collapse
|
12
|
Salvatore MF, Calipari ES, Jones SR. Regulation of Tyrosine Hydroxylase Expression and Phosphorylation in Dopamine Transporter-Deficient Mice. ACS Chem Neurosci 2016; 7:941-51. [PMID: 27124386 DOI: 10.1021/acschemneuro.6b00064] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Tyrosine hydroxylase (TH) and dopamine transporters (DATs) regulate dopamine (DA) neurotransmission at the biosynthesis and reuptake steps, respectively. Dysfunction or loss of these proteins occurs in impaired locomotor or addictive behavior, but little is known about the influence of DAT expression on TH function. Differences in TH phosphorylation, DA tissue content, l-DOPA biosynthesis, and DA turnover exist between the somatodendritic and terminal field compartments of nigrostriatal and mesoaccumbens pathways. We examined whether differential DAT expression affects these compartmental differences in DA regulation by comparing TH expression and phosphorylation at ser31 and ser40. In heterozygous DAT knockout (KO) (+/-) mice, DA tissue content and DA turnover were unchanged relative to wild-type mice, despite a 40% reduction in DAT protein expression. In DAT KO (-/-) mice, DA turnover increased in all DA compartments, but DA tissue content decreased (90-96%) only in terminal fields. TH protein expression and phosphorylation were differentially affected within DA pathway compartments by relative expression of DAT. TH protein decreased (∼74%), though to a significantly lesser extent than DA, in striatum and nucleus accumbens (NAc) in DAT -/- mice, with no decrease in substantia nigra or ventral tegmental area. Striatal ser31 TH phosphorylation and recovery of DA relative to TH protein expression in DAT +/- and DAT -/- mice decreased, whereas ser40 TH phosphorylation increased ∼2- to 3-fold in striatum and NAc of DAT -/- mice. These results suggest that DAT expression affects TH expression and phosphorylation largely in DA terminal field compartments, further corroborating evidence for dichotomous regulation of TH between somatodendritic and terminal field compartments of the nigrostriatal and mesoaccumbens pathways.
Collapse
Affiliation(s)
- Michael F. Salvatore
- Department
of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, United States
| | - Erin S. Calipari
- Department
of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Sara R. Jones
- Department
of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| |
Collapse
|