1
|
Spicer MM, Weber MA, Luo Z, Yang J, Narayanan NS, Fisher RA. Regulator of G protein signaling 6 (RGS6) in dopamine neurons promotes EtOH seeking, behavioral reward, and susceptibility to relapse. Psychopharmacology (Berl) 2024; 241:2255-2269. [PMID: 38856764 PMCID: PMC11518640 DOI: 10.1007/s00213-024-06631-8] [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: 12/14/2023] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Mesolimbic dopamine (DA) transmission is believed to play a critical role in mediating reward responses to drugs of abuse, including alcohol (EtOH). The neurobiological mechanisms underlying EtOH-seeking behavior and dependence are not fully understood, and abstinence remains the only effective way to prevent alcohol use disorders (AUDs). Here, we developed novel RGS6fl/fl; DAT-iCreER mice to determine the role of RGS6 in DA neurons on EtOH consumption, reward, and relapse behaviors. We found that RGS6 is expressed in DA neurons in both human and mouse ventral tegmental area (VTA), and that RGS6 loss in mice upregulates DA transporter (DAT) expression in VTA DA neuron synaptic terminals. Remarkably, loss of RGS6 in DA neurons significantly reduced EtOH consumption, preference, and reward in a manner indistinguishable from that seen in RGS6-/- mice. Strikingly, RGS6 loss from DA neurons before or after EtOH behavioral reward is established significantly reduced (~ 50%) re-instatement of reward following extinguishment, demonstrating distinct roles of RGS6 in promoting reward and relapse susceptibility to EtOH. These studies identify DA neurons as the locus of RGS6 action in promoting EtOH consumption, preference, reward, and relapse. RGS6 is unique among R7 RGS proteins in promoting rather than suppressing behavioral responses to drugs of abuse and to modulate EtOH behavioral reward. This is a result of RGS6's pre-synaptic actions that we hypothesize promote VTA DA transmission by suppressing GPCR-Gαi/o-DAT signaling in VTA DA neurons. These studies identify RGS6 as a potential therapeutic target for behavioral reward and relapse to EtOH.
Collapse
Affiliation(s)
- Mackenzie M Spicer
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Matthew A Weber
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Zili Luo
- Department of Pediatrics, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Jianqi Yang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
| | - Nandakumar S Narayanan
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Rory A Fisher
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA.
| |
Collapse
|
2
|
Nguyen H, Glaaser IW, Slesinger PA. Direct modulation of G protein-gated inwardly rectifying potassium (GIRK) channels. Front Physiol 2024; 15:1386645. [PMID: 38903913 PMCID: PMC11187414 DOI: 10.3389/fphys.2024.1386645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/08/2024] [Indexed: 06/22/2024] Open
Abstract
Ion channels play a pivotal role in regulating cellular excitability and signal transduction processes. Among the various ion channels, G-protein-coupled inwardly rectifying potassium (GIRK) channels serve as key mediators of neurotransmission and cellular responses to extracellular signals. GIRK channels are members of the larger family of inwardly-rectifying potassium (Kir) channels. Typically, GIRK channels are activated via the direct binding of G-protein βγ subunits upon the activation of G-protein-coupled receptors (GPCRs). GIRK channel activation requires the presence of the lipid signaling molecule, phosphatidylinositol 4,5-bisphosphate (PIP2). GIRK channels are also modulated by endogenous proteins and other molecules, including RGS proteins, cholesterol, and SNX27 as well as exogenous compounds, such as alcohol. In the last decade or so, several groups have developed novel drugs and small molecules, such as ML297, GAT1508 and GiGA1, that activate GIRK channels in a G-protein independent manner. Here, we aim to provide a comprehensive overview focusing on the direct modulation of GIRK channels by G-proteins, PIP2, cholesterol, and novel modulatory compounds. These studies offer valuable insights into the underlying molecular mechanisms of channel function, and have potential implications for both basic research and therapeutic development.
Collapse
Affiliation(s)
| | | | - Paul A. Slesinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
3
|
Song J, Li M, Chen C, Zhou J, Wang L, Yan Y, She J, Tong L, Song Y. Regulator of G protein signaling protein 6 alleviates acute lung injury by inhibiting inflammation and promoting cell self-renewal in mice. Cell Mol Biol Lett 2023; 28:102. [PMID: 38066447 PMCID: PMC10709870 DOI: 10.1186/s11658-023-00488-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a disease with high mortality and morbidity. Regulator of G protein signaling protein 6 (RGS6), identified as a tumor suppressor gene, has received increasing attention owing to its close relationship with oxidative stress and inflammation. However, the association between ARDS and RGS6 has not been reported. METHODS Congruously regulated G protein-coupled receptor (GPCR)-related genes and differentially expressed genes (DEGs) in an acute lung injury (ALI) model were identified, and functional enrichment analysis was conducted. In an in vivo study, the effects of RGS6 knockout were studied in a mouse model of ALI induced by lipopolysaccharide (LPS). HE staining, ELISA, and immunohistochemistry were used to evaluate pathological changes and the degree of inflammation. In vitro, qRT‒PCR, immunofluorescence staining, and western blotting were used to determine the dynamic changes in RGS6 expression in cells. The RGS6 overexpression plasmid was constructed for transfection. qRT‒PCR was used to assess proinflammatory factors transcription. Western blotting and flow cytometry were used to evaluate apoptosis and reactive oxygen species (ROS) production. Organoid culture was used to assess the stemness and self-renewal capacity of alveolar epithelial type II cells (AEC2s). RESULTS A total of 110 congruously regulated genes (61 congruously upregulated and 49 congruously downregulated genes) were identified among GPCR-related genes and DEGs in the ALI model. RGS6 was downregulated in vivo and in vitro in the ALI model. RGS6 was expressed in the cytoplasm and accumulated in the nucleus after LPS stimulation. Compared with the control group, we found higher mortality, more pronounced body weight changes, more serious pulmonary edema and pathological damage, and more neutrophil infiltration in the RGS6 knockout group upon LPS stimulation in vivo. Moreover, AEC2s loss was significantly increased upon RGS6 knockout. Organoid culture assays showed slower alveolar organoid formation, fewer alveolar organoids, and impaired development of new structures after passaging upon RGS6 knockout. In addition, RGS6 overexpression decreased ROS production as well as proinflammatory factor transcription in macrophages and decreased apoptosis in epithelial cells. CONCLUSIONS RGS6 plays a protective role in ALI not only in early inflammatory responses but also in endogenous lung stem cell regeneration.
Collapse
Affiliation(s)
- Juan Song
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Pulmonary Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361000, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Miao Li
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Cuicui Chen
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Jian Zhou
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Linlin Wang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Yu Yan
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
| | - Jun She
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Respiratory Research Institute, Shanghai, 200032, China.
| | - Lin Tong
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Pulmonary Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361000, China.
- Shanghai Respiratory Research Institute, Shanghai, 200032, China.
| | - Yuanlin Song
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Pulmonary Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361000, China.
- Shanghai Respiratory Research Institute, Shanghai, 200032, China.
| |
Collapse
|
4
|
Spicer MM, Weber MA, Luo Z, Yang J, Narayanan NS, Fisher RA. Regulator of G protein signaling 6 (RGS6) in ventral tegmental area (VTA) dopamine neurons promotes EtOH seeking, behavioral reward and susceptibility to relapse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563844. [PMID: 37961154 PMCID: PMC10634791 DOI: 10.1101/2023.10.24.563844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Mesolimbic dopamine (DA) transmission is believed to play a critical role in mediating reward responses to drugs of abuse, including alcohol (EtOH). EtOH is the most abused substance worldwide with chronic consumption often leading to the development of dependence and abuse. Unfortunately, the neurobiological mechanisms underlying EtOH-seeking behavior and dependence are not fully understood, and abstinence remains the only effective way to prevent alcohol use disorders (AUDs). Here, we developed novel RGS6 fl/fl ; DAT-iCreER mice to determine the role of RGS6 in VTA DA neurons on EtOH consumption and reward behaviors. We found that RGS6 is expressed in DA neurons in both human and mouse VTA, and that RGS6 loss in mice upregulates DA transporter (DAT) expression in VTA DA neuron synaptic terminals. Remarkably, loss of RGS6 in VTA DA neurons significantly reduced EtOH consumption, preference, and reward in a manner indistinguishable from that seen in RGS6 -/- mice. Strikingly, RGS6 loss from VTA DA neurons before or after EtOH behavioral reward is established significantly reduced (∼50%) re-instatement of reward following extinguishment, demonstrating distinct roles of RGS6 in promoting reward and relapse susceptibility to EtOH. These studies illuminate a critical role of RGS6 in the mesolimbic circuit in promoting EtOH seeking, reward, and reinstatement. We propose that RGS6 functions to promote DA transmission through its function as a negative modulator of GPCR-Gα i/o -DAT signaling in VTA DA neurons. These studies identify RGS6 as a potential therapeutic target for behavioral reward and relapse to EtOH.
Collapse
|
5
|
DeBaker MC, Mitten EH, Rose TR, Marron Fernandez de Velasco E, Gao R, Lee AM, Wickman K. RGS6 negatively regulates inhibitory G protein signaling in dopamine neurons and positively regulates binge-like alcohol consumption in mice. Br J Pharmacol 2023; 180:2140-2155. [PMID: 36929333 PMCID: PMC10504421 DOI: 10.1111/bph.16071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/02/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND AND PURPOSE Drugs of abuse, including alcohol, increase dopamine in the mesocorticolimbic system via actions on dopamine neurons in the ventral tegmental area (VTA). Increased dopamine transmission can activate inhibitory G protein signalling pathways in VTA dopamine neurons, including those controlled by GABAB and D2 receptors. Members of the R7 subfamily of regulator of G protein signalling (RGS) proteins can regulate inhibitory G protein signalling, but their influence on VTA dopamine neurons is unclear. Here, we investigated the influence of RGS6, an R7 RGS family memberthat has been implicated in the regulation of alcohol consumption in mice, on inhibitory G protein signalling in VTA dopamine neurons. EXPERIMENTAL APPROACH We used molecular, electrophysiological and genetic approaches to probe the impact of RGS6 on inhibitory G protein signalling in VTA dopamine neurons and on binge-like alcohol consumption in mice. KEY RESULTS RGS6 is expressed in adult mouse VTA dopamine neurons and it modulates inhibitory G protein signalling in a receptor-dependent manner, tempering D2 receptor-induced somatodendritic currents and accelerating deactivation of synaptically evoked GABAB receptor-dependent responses. RGS6-/- mice exhibit diminished binge-like alcohol consumption, a phenotype replicated in female (but not male) mice lacking RGS6 selectively in VTA dopamine neurons. CONCLUSIONS AND IMPLICATIONS RGS6 negatively regulates GABAB - and D2 receptor-dependent inhibitory G protein signalling pathways in mouse VTA dopamine neurons and exerts a sex-dependent positive influence on binge-like alcohol consumption in adult mice. As such, RGS6 may represent a new diagnostic and/or therapeutic target for alcohol use disorder.
Collapse
Affiliation(s)
- Margot C. DeBaker
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN
| | - Eric H. Mitten
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN
| | - Timothy R. Rose
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | | | - Runbo Gao
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Anna M. Lee
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
6
|
Spicer MM, Yang J, Fu D, DeVore AN, Lauffer M, Atasoy NS, Atasoy D, Fisher RA. RGS6 mediates exercise-induced recovery of hippocampal neurogenesis, learning, and memory in an Alzheimer's mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537272. [PMID: 39185171 PMCID: PMC11343197 DOI: 10.1101/2023.04.17.537272] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Hippocampal neuronal loss causes cognitive dysfunction in Alzheimer's disease (AD). Adult hippocampal neurogenesis (AHN) is reduced in AD patients. Exercise stimulates AHN in rodents and improves memory and slows cognitive decline in AD patients. However, the molecular pathways for exercise-induced AHN and improved cognition in AD are poorly understood. Here, we show that voluntary running in APP SWE mice restores their hippocampal cognitive impairments to that of control mice. This cognitive rescue was abolished by RGS6 deletion in dentate gyrus (DG) neuronal progenitors (NPs), which also abolished running-mediated increases in AHN. AHN was reduced in sedentary APP SWE mice versus control mice, with basal AHN reduced by RGS6 deletion in DG NPs. RGS6 expression is significantly lower in the DG of AD patients. Thus, RGS6 mediates exercise-induced rescue of impaired cognition and AHN in AD mice, identifying RGS6 in DG NPs as a potential target to combat hippocampal neuron loss in AD. Teaser RGS6 expression in hippocampal NPCs promotes voluntary running-induced neurogenesis and restored cognition in APP SWE mice. Field Codes RGS6, Alzheimer's disease, adult hippocampal neurogenesis, neural precursor cells, dentate gyrus, exercise, learning/memory.
Collapse
|
7
|
Wang Z, Chen J, Wang S, Sun Z, Lei Z, Zhang HT, Huang J. RGS6 suppresses TGF-β-induced epithelial-mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with SMAD4. Cell Death Dis 2022; 13:656. [PMID: 35902557 PMCID: PMC9334288 DOI: 10.1038/s41419-022-05093-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 01/21/2023]
Abstract
Regulator of G-protein signaling 6 (RGS6) is a newly discovered tumor suppressor that has been shown to be protective in development of various cancers such as breast cancer and bladder cancer. But the mechanisms underlying these tumor-suppressing functions of RGS6 are not fully understood. Here, we discover a novel function of RGS6 in suppressing TGF-β-induced epithelial-mesenchymal transition (EMT) of non-small cell lung cancer (NSCLC) cells and in vivo NSCLC metastasis. Using both bioinformatics and experimental tools, we showed that RGS6 was downregulated in lung cancer tissues compared to noncancerous counterparts, and low expression of RGS6 was associated with poor survival of lung cancer patients. Overexpression of RGS6 suppressed TGF-β-induced EMT in vitro and TGF-β-promoted metastasis in vivo, by impairing gene expression of downstream effectors induced by the canonical TGF-β-SMAD signaling. The ability of RGS6 to suppress TGF-β-SMAD-mediated gene expression relied on its binding to SMAD4 to prevent complex formation between SMAD4 and SMAD2/3, but independent of its regulation of the G-protein signaling. Interaction between RGS6 and SMAD4 caused less nuclear entry of p-SMAD3 and SMAD4, resulting in inefficient SMAD3-mediated gene expression. Taken together, our findings reveal a novel and noncanonical role of RGS6 in regulation of TGF-β-induced EMT and metastasis of NSCLC and identify RGS6 as a prognostic marker and a potential novel target for NSCLC therapy.
Collapse
Affiliation(s)
- Zhao Wang
- grid.263761.70000 0001 0198 0694Soochow University Laboratory of Cancer Molecular Genetics, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China ,grid.263761.70000 0001 0198 0694Department of Genetics, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China
| | - Jun Chen
- grid.263761.70000 0001 0198 0694Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215006 China
| | - Shengjie Wang
- grid.263761.70000 0001 0198 0694Soochow University Laboratory of Cancer Molecular Genetics, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China ,grid.263761.70000 0001 0198 0694Department of Genetics, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China ,grid.89957.3a0000 0000 9255 8984Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang, 222000 China
| | - Zelong Sun
- grid.263761.70000 0001 0198 0694Soochow University Laboratory of Cancer Molecular Genetics, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China ,grid.263761.70000 0001 0198 0694Department of Genetics, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China
| | - Zhe Lei
- grid.263761.70000 0001 0198 0694Soochow University Laboratory of Cancer Molecular Genetics, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China ,grid.263761.70000 0001 0198 0694Department of Genetics, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China ,Suzhou Key Laboratory for Molecular Cancer Genetics, Suzhou, Jiangsu 215123 China
| | - Hong-Tao Zhang
- grid.263761.70000 0001 0198 0694Soochow University Laboratory of Cancer Molecular Genetics, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China ,grid.263761.70000 0001 0198 0694Department of Genetics, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China ,Suzhou Key Laboratory for Molecular Cancer Genetics, Suzhou, Jiangsu 215123 China
| | - Jie Huang
- grid.263761.70000 0001 0198 0694Soochow University Laboratory of Cancer Molecular Genetics, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China ,grid.263761.70000 0001 0198 0694Department of Genetics, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123 China ,Suzhou Key Laboratory for Molecular Cancer Genetics, Suzhou, Jiangsu 215123 China
| |
Collapse
|
8
|
Luo H, Marron Fernandez de Velasco E, Wickman K. Neuronal G protein-gated K + channels. Am J Physiol Cell Physiol 2022; 323:C439-C460. [PMID: 35704701 PMCID: PMC9362898 DOI: 10.1152/ajpcell.00102.2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels exert a critical inhibitory influence on neurons. Neuronal GIRK channels mediate the G protein-dependent, direct/postsynaptic inhibitory effect of many neurotransmitters including γ-aminobutyric acid (GABA), serotonin, dopamine, adenosine, somatostatin, and enkephalin. In addition to their complex regulation by G proteins, neuronal GIRK channel activity is sensitive to PIP2, phosphorylation, regulator of G protein signaling (RGS) proteins, intracellular Na+ and Ca2+, and cholesterol. The application of genetic and viral manipulations in rodent models, together with recent progress in the development of GIRK channel modulators, has increased our understanding of the physiological and behavioral impact of neuronal GIRK channels. Work in rodent models has also revealed that neuronal GIRK channel activity is modified, transiently or persistently, by various stimuli including exposure drugs of abuse, changes in neuronal activity patterns, and aversive experience. A growing body of preclinical and clinical evidence suggests that dysregulation of GIRK channel activity contributes to neurological diseases and disorders. The primary goals of this review are to highlight fundamental principles of neuronal GIRK channel biology, mechanisms of GIRK channel regulation and plasticity, the nascent landscape of GIRK channel pharmacology, and the potential relevance of GIRK channels to the pathophysiology and treatment of neurological diseases and disorders.
Collapse
Affiliation(s)
- Haichang Luo
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
9
|
Luquin E, Paternain B, Zugasti I, Santomá C, Mengual E. Stereological estimations and neurochemical characterization of neurons expressing GABAA and GABAB receptors in the rat pedunculopontine and laterodorsal tegmental nuclei. Brain Struct Funct 2022; 227:89-110. [PMID: 34510281 PMCID: PMC8741722 DOI: 10.1007/s00429-021-02375-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 08/31/2021] [Indexed: 11/29/2022]
Abstract
To better understand GABAergic transmission at two targets of basal ganglia downstream projections, the pedunculopontine (PPN) and laterodorsal (LDT) tegmental nuclei, the anatomical localization of GABAA and GABAB receptors was investigated in both nuclei. Specifically, the total number of neurons expressing the GABAA receptor γ2 subunit (GABAAR γ2) and the GABAB receptor R2 subunit (GABAB R2) in PPN and LDT was estimated using stereological methods, and the neurochemical phenotype of cells expressing each subunit was also determined. The mean number of non-cholinergic cells expressing GABAAR γ2 was 9850 ± 1856 in the PPN and 8285 ± 962 in the LDT, whereas those expressing GABAB R2 were 7310 ± 1970 and 9170 ± 1900 in the PPN and LDT, respectively. In addition, all cholinergic neurons in both nuclei co-expressed GABAAR γ2 and 95-98% of them co-expressed GABAB R2. Triple labeling using in situ hybridization revealed that 77% of GAD67 mRNA-positive cells in the PPT and 49% in the LDT expressed GABAAR γ2, while 90% (PPN) and 65% (LDT) of Vglut2 mRNA-positive cells also expressed GABAAR γ2. In contrast, a similar proportion (~2/3) of glutamatergic and GABAergic cells co-expressed GABAB R2 in both nuclei. The heterogeneous distribution of GABAAR and GABABR among non-cholinergic cells in PPN and LDT may give rise to physiological differences within each neurochemical subpopulation. In addition, the dissimilar proportion of GABAAR γ2-expressing glutamatergic and GABAergic neurons in the PPN and LDT may contribute to some of the functional differences found between the two nuclei.
Collapse
Affiliation(s)
- Esther Luquin
- Department of Pathology, Anatomy, and Physiology, School of Medicine, University of Navarra, Ed. Los Castaños, Irunlarrea 1, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Beatriz Paternain
- Department of Pathology, Anatomy, and Physiology, School of Medicine, University of Navarra, Ed. Los Castaños, Irunlarrea 1, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Inés Zugasti
- Department of Pathology, Anatomy, and Physiology, School of Medicine, University of Navarra, Ed. Los Castaños, Irunlarrea 1, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Carmen Santomá
- Department of Pathology, Anatomy, and Physiology, School of Medicine, University of Navarra, Ed. Los Castaños, Irunlarrea 1, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Elisa Mengual
- Department of Pathology, Anatomy, and Physiology, School of Medicine, University of Navarra, Ed. Los Castaños, Irunlarrea 1, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| |
Collapse
|
10
|
Mahata T, Sengar AS, Basak M, Das K, Pramanick A, Verma SK, Singh PK, Biswas S, Sarkar S, Saha S, Chatterjee S, Das M, Stewart A, Maity B. Hepatic Regulator of G Protein Signaling 6 (RGS6) drives non-alcoholic fatty liver disease by promoting oxidative stress and ATM-dependent cell death. Redox Biol 2021; 46:102105. [PMID: 34534913 PMCID: PMC8446788 DOI: 10.1016/j.redox.2021.102105] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 08/14/2021] [Indexed: 12/15/2022] Open
Abstract
The pathophysiological mechanism(s) driving non-alcoholic fatty liver disease, the most prevalent chronic liver disease globally, have yet to be fully elucidated. Here, we identify regulator of G protein signaling 6 (RGS6), up-regulated in the livers of NAFLD patients, as a critical mediator of hepatic steatosis, fibrosis, inflammation, and cell death. Human patients with high hepatic RGS6 expression exhibited a corresponding high inflammatory burden, pronounced insulin resistance, and poor liver function. In mice, liver-specific RGS6 knockdown largely ameliorated high fat diet (HFD)-driven oxidative stress, fibrotic remodeling, inflammation, lipid deposition and cell death. RGS6 depletion allowed for maintenance of mitochondrial integrity restoring redox balance, improving fatty acid oxidation, and preventing loss of insulin receptor sensitivity in hepatocytes. RGS6 is both induced by ROS and increases ROS generation acting as a key amplification node to exacerbate oxidative stress. In liver, RGS6 forms a direct complex with ATM kinase supported by key aspartate residues in the RGS domain and is both necessary and sufficient to drive hyperlipidemia-dependent ATM phosphorylation. pATM and markers of DNA damage (γH2AX) were also elevated in livers from NAFLD patients particularly in samples with high RGS6 protein content. Unsurprisingly, RGS6 knockdown prevented ATM phosphorylation in livers from HFD-fed mice. Further, RGS6 mutants lacking the capacity for ATM binding fail to facilitate palmitic acid-dependent hepatocyte apoptosis underscoring the importance of the RGS6-ATM complex in hyperlipidemia-dependent cell death. Inhibition of RGS6, then, may provide a viable means to prevent or reverse liver damage by mitigating oxidative liver damage.
Collapse
Affiliation(s)
- Tarun Mahata
- Centre of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Abhishek Singh Sengar
- Centre of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Madhuri Basak
- Centre of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Kiran Das
- Centre of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Arnab Pramanick
- Centre of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Sumit Kumar Verma
- Centre of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Praveen Kumar Singh
- Department of Surgery, Millers School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Sayan Biswas
- Department of Forensic Medicine, College of Medicine and Sagore Dutta Hospital, B.T. Road, Kamarhati, Kolkata, West Bengal, 700058, India
| | - Subhasish Sarkar
- Department of Surgery, College of Medicine and Sagore Dutta Hospital, B.T. Road, Kamarhati, Kolkata, West Bengal, 700058, India
| | - Sudipta Saha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India
| | - Suvro Chatterjee
- Department of Biotechnology, Anna University and Vascular Biology Laboratory, AU-KBC Research Centre, MIT Campus, Chennai, 600044, India
| | - Madhusudan Das
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, West Bengal, 700019, India.
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Biswanath Maity
- Centre of Biomedical Research, Sanjay Gandhi Post-Graduate Institute of Medical Sciences Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India.
| |
Collapse
|
11
|
Gao Y, Shen M, Gonzalez JC, Dong Q, Kannan S, Hoang JT, Eisinger BE, Pandey J, Javadi S, Chang Q, Wang D, Overstreet-Wadiche L, Zhao X. RGS6 Mediates Effects of Voluntary Running on Adult Hippocampal Neurogenesis. Cell Rep 2021; 32:107997. [PMID: 32755589 DOI: 10.1016/j.celrep.2020.107997] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 01/17/2023] Open
Abstract
Voluntary running enhances adult hippocampal neurogenesis, with consequences for hippocampal-dependent learning ability and mood regulation. However, the underlying mechanism remains unclear. Here, we show that voluntary running induces unique and dynamic gene expression changes specifically within the adult-born hippocampal neurons, with significant impact on genes involved in neuronal maturation and human diseases. We identify the regulator of G protein signaling 6 (RGS6) as a key factor that mediates running impact on adult-born neurons. RGS6 overexpression mimics the positive effects of voluntary running on morphological and physiological maturation of adult new neurons and reduced sensitivity of adult-born neurons to the inhibitory effect of GABAB (γ-Aminobutyric acid B) receptor activation. Knocking down RGS6 abolishes running-enhanced neuronal maturation and hippocampal neurogenesis-dependent learning and anxiolytic effect. Our study provides a data resource showing genome-wide intrinsic molecular changes in adult-born hippocampal neurons that contribute to voluntary running-induced neurogenesis.
Collapse
Affiliation(s)
- Yu Gao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Minjie Shen
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jose Carlos Gonzalez
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Qiping Dong
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sudharsan Kannan
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Johnson T Hoang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Brian E Eisinger
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jyotsna Pandey
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sahar Javadi
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
12
|
Mechanisms and Regulation of Neuronal GABA B Receptor-Dependent Signaling. Curr Top Behav Neurosci 2020; 52:39-79. [PMID: 32808092 DOI: 10.1007/7854_2020_129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
γ-Aminobutyric acid B receptors (GABABRs) are broadly expressed throughout the central nervous system where they play an important role in regulating neuronal excitability and synaptic transmission. GABABRs are G protein-coupled receptors that mediate slow and sustained inhibitory actions via modulation of several downstream effector enzymes and ion channels. GABABRs are obligate heterodimers that associate with diverse arrays of proteins to form modular complexes that carry out distinct physiological functions. GABABR-dependent signaling is fine-tuned and regulated through a multitude of mechanisms that are relevant to physiological and pathophysiological states. This review summarizes the current knowledge on GABABR signal transduction and discusses key factors that influence the strength and sensitivity of GABABR-dependent signaling in neurons.
Collapse
|
13
|
Ahlers-Dannen KE, Spicer MM, Fisher RA. RGS Proteins as Critical Regulators of Motor Function and Their Implications in Parkinson's Disease. Mol Pharmacol 2020; 98:730-738. [PMID: 32015009 DOI: 10.1124/mol.119.118836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/25/2020] [Indexed: 11/22/2022] Open
Abstract
Parkinson disease (PD) is a devastating, largely nonfamilial, age-related disorder caused by the progressive loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc). Release of DA from these neurons into the dorsal striatum is crucial for regulating movement and their loss causes PD. Unfortunately, the mechanisms underlying SNc neurodegeneration remain unclear, and currently there is no cure for PD, only symptomatic treatments. Recently, several regulator of G protein signaling (RGS) proteins have emerged as critical modulators of PD pathogenesis and/or motor dysfunction and dyskinesia: RGSs 4, 6, 9, and 10. Striatal RGS4 has been shown to exacerbate motor symptoms of DA loss by suppressing M4-autoreceptor-Gα i/o signaling in striatal cholinergic interneurons. RGS6 and RGS9 are key regulators of D2R-Gα i/o signaling in SNc DA neurons and striatal medium spiny neurons, respectively. RGS6, expressed in human and mouse SNc DA neurons, suppresses characteristic PD hallmarks in aged mice, including SNc DA neuron loss, motor deficits, and α-synuclein accumulation. After DA depletion, RGS9 (through its inhibition of medium spiny neuron D2R signaling) suppresses motor dysfunction induced by L-DOPA or D2R-selective agonists. RGS10 is highly expressed in microglia, the brain's resident immune cells. Within the SNc, RGS10 may promote DA neuron survival through the upregulation of prosurvival genes and inhibition of microglial inflammatory factor expression. Thus, RGSs 4, 6, 9, and 10 are critical modulators of cell signaling pathways that promote SNc DA neuron survival and/or proper motor control. Accordingly, these RGS proteins represent novel therapeutic targets for the treatment of PD pathology. SIGNIFICANCE STATEMENT: Parkinson disease (PD), the most common movement disorder, is a progressive neurodegenerative disease characterized by substantia nigra pars compacta (SNc) dopamine (DA) neuron loss and subsequent motor deficits. Current PD therapies only target disease motor symptomology and are fraught with side effects. Therefore, researchers have begun to explore alternative therapeutic options. Regulator of G protein signaling (RGS) proteins, whether primarily expressed in SNc DA neurons (RGS6), striatal neurons (RGSs 4 and 9), or microglia (RGS10), modulate key signaling pathways important for SNc DA neuron survival and/or proper motor control. As such, RGS proteins represent novel therapeutic targets in PD.
Collapse
Affiliation(s)
- Katelin E Ahlers-Dannen
- Department of Neuroscience and Pharmacology (K.E.A.-D., M.M.S., R.A.F.), Iowa Neuroscience Institute (R.A.F.), and Interdisciplinary Graduate Program in Molecular Medicine (M.M.S., R.A.F.), University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Mackenzie M Spicer
- Department of Neuroscience and Pharmacology (K.E.A.-D., M.M.S., R.A.F.), Iowa Neuroscience Institute (R.A.F.), and Interdisciplinary Graduate Program in Molecular Medicine (M.M.S., R.A.F.), University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Rory A Fisher
- Department of Neuroscience and Pharmacology (K.E.A.-D., M.M.S., R.A.F.), Iowa Neuroscience Institute (R.A.F.), and Interdisciplinary Graduate Program in Molecular Medicine (M.M.S., R.A.F.), University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
14
|
Malerba N, De Nittis P, Merla G. The Emerging Role of Gβ Subunits in Human Genetic Diseases. Cells 2019; 8:E1567. [PMID: 31817184 PMCID: PMC6952978 DOI: 10.3390/cells8121567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/23/2019] [Accepted: 11/29/2019] [Indexed: 12/20/2022] Open
Abstract
Environmental stimuli are perceived and transduced inside the cell through the activation of signaling pathways. One common type of cell signaling transduction network is initiated by G-proteins. G-proteins are activated by G-protein-coupled receptors (GPCRs) and transmit signals from hormones, neurotransmitters, and other signaling factors, thus controlling a number of biological processes that include synaptic transmission, visual photoreception, hormone and growth factors release, regulation of cell contraction and migration, as well as cell growth and differentiation. G-proteins mainly act as heterotrimeric complexes, composed of alpha, beta, and gamma subunits. In the last few years, whole exome sequencing and biochemical studies have shown causality of disease-causing variants in genes encoding G-proteins and human genetic diseases. This review focuses on the G-protein β subunits and their emerging role in the etiology of genetically inherited rare diseases in humans.
Collapse
Affiliation(s)
- Natascia Malerba
- Division of Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo (FG), Italy;
| | - Pasquelena De Nittis
- Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland;
| | - Giuseppe Merla
- Division of Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini, 71013 San Giovanni Rotondo (FG), Italy;
| |
Collapse
|
15
|
Luo Z, Ahlers-Dannen KE, Spicer MM, Yang J, Alberico S, Stevens HE, Narayanan NS, Fisher RA. Age-dependent nigral dopaminergic neurodegeneration and α-synuclein accumulation in RGS6-deficient mice. JCI Insight 2019; 5:126769. [PMID: 31120439 DOI: 10.1172/jci.insight.126769] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Parkinson's is primarily a non-familial, age-related disorder caused by α-synuclein accumulation and the progressive loss of dopamine neurons in the substantia nigra pars compacta (SNc). G protein-coupled receptor (GPCR)-cAMP signaling has been linked to a reduction in human Parkinson's incidence and α-synuclein expression. Neuronal cAMP levels are controlled by GPCRs coupled to Gs or Gi/o, which increase or decrease cAMP, respectively. Regulator of G protein signaling 6 (RGS6) powerfully inhibits Gi/o signaling. Therefore, we hypothesized that RGS6 suppresses D2 autoreceptor- Gi/o signaling in SNc dopamine neurons promoting neuronal survival and reducing α-synuclein expression. Here we provide novel evidence that RGS6 critically suppresses late-age-onset SNc dopamine neuron loss and α-synuclein accumulation. RGS6 is restrictively expressed in human SNc dopamine neurons and, despite their loss in Parkinson's, all surviving neurons express RGS6. RGS6-/- mice exhibit hyperactive D2 autoreceptors with reduced cAMP signaling in SNc dopamine neurons. Importantly, RGS6-/- mice recapitulate key sporadic Parkinson's hallmarks, including: SNc dopamine neuron loss, reduced nigrostriatal dopamine, motor deficits, and α-synuclein accumulation. To our knowledge, Rgs6 is the only gene whose loss phenocopies these features of human Parkinson's. Therefore, RGS6 is a key regulator of D2R-Gi/o signaling in SNc dopamine neurons, protecting against Parkinson's neurodegeneration and α-synuclein accumulation.
Collapse
Affiliation(s)
- Zili Luo
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Katelin E Ahlers-Dannen
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Mackenzie M Spicer
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Interdisciplinary Graduate Program of Molecular Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Jianqi Yang
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | | | - Hanna E Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Nandakumar S Narayanan
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Rory A Fisher
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
16
|
Inhibitory Signaling to Ion Channels in Hippocampal Neurons Is Differentially Regulated by Alternative Macromolecular Complexes of RGS7. J Neurosci 2018; 38:10002-10015. [PMID: 30315127 DOI: 10.1523/jneurosci.1378-18.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/01/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
The neuromodulatory effects of GABA on pyramidal neurons are mediated by GABAB receptors (GABABRs) that signal via a conserved G-protein-coupled pathway. Two prominent effectors regulated by GABABRs include G-protein inwardly rectifying K+ (GIRK) and P/Q/N type voltage-gated Ca2+ (CaV2) ion channels that control excitability and synaptic output of these neurons, respectively. Regulator of G-protein signaling 7 (RGS7) has been shown to control GABAB effects, yet the specificity of its impacts on effector channels and underlying molecular mechanisms is poorly understood. In this study, we show that hippocampal RGS7 forms two distinct complexes with alternative subunit configuration bound to either membrane protein R7BP (RGS7 binding protein) or orphan receptor GPR158. Quantitative biochemical experiments show that both complexes account for targeting nearly the entire pool of RGS7 to the plasma membrane. We analyzed the effect of genetic elimination in mice of both sexes and overexpression of various components of RGS7 complex by patch-clamp electrophysiology in cultured neurons and brain slices. We report that RGS7 prominently regulates GABABR signaling to CaV2, in addition to its known involvement in modulating GIRK. Strikingly, only complexes containing R7BP, but not GPR158, accelerated the kinetics of both GIRK and CaV2 modulation by GABABRs. In contrast, GPR158 overexpression exerted the opposite effect and inhibited RGS7-assisted temporal modulation of GIRK and CaV2 by GABA. Collectively, our data reveal mechanisms by which distinctly composed macromolecular complexes modulate the activity of key ion channels that mediate the inhibitory effects of GABA on hippocampal CA1 pyramidal neurons.SIGNIFICANCE STATEMENT This study identifies the contributions of distinct macromolecular complexes containing a major G-protein regulator to controlling key ion channel function in hippocampal neurons with implications for understanding molecular mechanisms underlying synaptic plasticity, learning, and memory.
Collapse
|
17
|
Neonatal brain injury causes cerebellar learning deficits and Purkinje cell dysfunction. Nat Commun 2018; 9:3235. [PMID: 30104642 PMCID: PMC6089917 DOI: 10.1038/s41467-018-05656-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 07/16/2018] [Indexed: 11/08/2022] Open
Abstract
Premature infants are more likely to develop locomotor disorders than term infants. In a chronic sub-lethal hypoxia (Hx) mouse model of neonatal brain injury, we recently demonstrated the presence of cellular and physiological changes in the cerebellar white matter. We also observed Hx-induced delay in Purkinje cell (PC) arborization. However, the behavioral consequences of these cellular alterations remain unexplored. Using the Erasmus Ladder to study cerebellar behavior, we report the presence of locomotor malperformance and long-term cerebellar learning deficits in Hx mice. Optogenetics experiments in Hx mice reveal a profound reduction in spontaneous and photoevoked PC firing frequency. Finally, treatment with a gamma-aminobutyric acid (GABA) reuptake inhibitor partially rescues locomotor performance and improves PC firing. Our results demonstrate a long-term miscoordination phenotype characterized by locomotor malperformance and cerebellar learning deficits in a mouse model of neonatal brain injury. Our findings also implicate the developing GABA network as a potential therapeutic target for prematurity-related locomotor deficits.
Collapse
|
18
|
TERUNUMA M. Diversity of structure and function of GABA B receptors: a complexity of GABA B-mediated signaling. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2018; 94:390-411. [PMID: 30541966 PMCID: PMC6374141 DOI: 10.2183/pjab.94.026] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/09/2018] [Indexed: 05/24/2023]
Abstract
γ-aminobutyric acid type B (GABAB) receptors are broadly expressed in the nervous system and play an important role in neuronal excitability. GABAB receptors are G protein-coupled receptors that mediate slow and prolonged inhibitory action, via activation of Gαi/o-type proteins. GABAB receptors mediate their inhibitory action through activating inwardly rectifying K+ channels, inactivating voltage-gated Ca2+ channels, and inhibiting adenylate cyclase. Functional GABAB receptors are obligate heterodimers formed by the co-assembly of R1 and R2 subunits. It is well established that GABAB receptors interact not only with G proteins and effectors but also with various proteins. This review summarizes the structure, subunit isoforms, and function of GABAB receptors, and discusses the complexity of GABAB receptors, including how receptors are localized in specific subcellular compartments, the mechanism regulating cell surface expression and mobility of the receptors, and the diversity of receptor signaling through receptor crosstalk and interacting proteins.
Collapse
Affiliation(s)
- Miho TERUNUMA
- Division of Oral Biochemistry, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
19
|
Chakravarti B, Yang J, Ahlers-Dannen KE, Luo Z, Flaherty HA, Meyerholz DK, Anderson ME, Fisher RA. Essentiality of Regulator of G Protein Signaling 6 and Oxidized Ca 2+/Calmodulin-Dependent Protein Kinase II in Notch Signaling and Cardiovascular Development. J Am Heart Assoc 2017; 6:JAHA.117.007038. [PMID: 29079565 PMCID: PMC5721783 DOI: 10.1161/jaha.117.007038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Congenital heart defects are the most common birth defects worldwide. Although defective Notch signaling is the major cause of mouse embryonic death from cardiovascular defects, how Notch signaling is regulated during embryonic vasculogenesis and heart development is poorly understood. Methods and Results Regulator of G protein signaling 6 (RGS6)−/−/Ca2+/calmodulin‐dependent protein kinase II (CaMKII)VV double mutant mice were developed by crossing RGS6−/− mice with mice expressing an oxidation‐resistant CaMKIIδ (CaMKIIVV), and the resulting embryonic defects/lethality were investigated using E7.5 to E15.5 embryos. While loss of either RGS6 or oxidized CaMKIIδ does not alter embryogenesis, their combined loss causes defective Notch signaling, severe cardiovascular defects, and embryonic lethality (≈E10.5–11.5). Embryos lacking RGS6 and expressing oxidation‐resistant CaMKIIδ exhibit reduced myocardial wall thickness, abnormal trabeculation, and arterial specification defects. Double mutants show vascular remodeling defects, including reduced neurovascularization, delayed neural tube maturation, and small dorsal aortae. These striking cardiovascular defects were accompanied by placental and yolk sac defects in angiogenesis, hematopoiesis, and vascular remodeling similar to what is seen with defective Notch1 signaling. Double mutant hearts, embryos, and yolk sacs exhibit profound downregulation of Notch1, Jagged 1, and Notch downstream target genes Hey1, Hey2, and Hey1L as well as impaired Notch1 signaling in embryos/hearts. Conclusions RGS6 and oxidized CaMKIIδ together function as novel critical upstream modulators of Notch signaling required for normal cardiovascular development and embryo survival. Their combined need indicates that they function in parallel pathways needed for Notch1 signaling in yolk sac, placenta and embryos. Thus, dysregulated embryonic RGS6 expression and oxidative activation of CaMKII may potentially contribute to congenital heart defects.
Collapse
Affiliation(s)
- Bandana Chakravarti
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Jianqi Yang
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA
| | | | - Zili Luo
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA
| | | | - David K Meyerholz
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Mark E Anderson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rory A Fisher
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA
| |
Collapse
|
20
|
Grammatopoulos DK. Regulation of G-protein coupled receptor signalling underpinning neurobiology of mood disorders and depression. Mol Cell Endocrinol 2017; 449:82-89. [PMID: 28229904 DOI: 10.1016/j.mce.2017.02.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/09/2017] [Indexed: 02/08/2023]
Abstract
G-protein coupled receptors (GPCRs) have long been at the center of investigations of the neurobiology of depression and mood disorders. Different facets of GPCR signalling pathways, including those controlling monoaminergic and neuropeptidergic hormonal systems are believed to be dysregulated in major depressive and bipolar disorders. Although these receptors are key molecular targets for a variety of therapeutic agents and continue to be the focus of intense pharmaceutical development, the molecular mechanisms activated by these GPCRs and underpin the pathological basis of mood disorders remain poorly understood. This review will discuss some of the emerging regulatory mechanisms of GPCR signaling in the central nervous system (CNS) involving protein-protein interactions, downstream effectors and cross-talk with other signaling molecules and their potential involvement in the neurobiology of psychiatric disease.
Collapse
Affiliation(s)
- Dimitris K Grammatopoulos
- Translational Medicine, Warwick Medical School & Clinical Biochemistry, Coventry and Warwickshire Pathology Service, United Kingdom.
| |
Collapse
|
21
|
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
|
22
|
Gerber KJ, Squires KE, Hepler JR. Roles for Regulator of G Protein Signaling Proteins in Synaptic Signaling and Plasticity. Mol Pharmacol 2015; 89:273-86. [PMID: 26655302 DOI: 10.1124/mol.115.102210] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/10/2015] [Indexed: 11/22/2022] Open
Abstract
The regulator of G protein signaling (RGS) family of proteins serves critical roles in G protein-coupled receptor (GPCR) and heterotrimeric G protein signal transduction. RGS proteins are best understood as negative regulators of GPCR/G protein signaling. They achieve this by acting as GTPase activating proteins (GAPs) for Gα subunits and accelerating the turnoff of G protein signaling. Many RGS proteins also bind additional signaling partners that either regulate their functions or enable them to regulate other important signaling events. At neuronal synapses, GPCRs, G proteins, and RGS proteins work in coordination to regulate key aspects of neurotransmitter release, synaptic transmission, and synaptic plasticity, which are necessary for central nervous system physiology and behavior. Accumulating evidence has revealed key roles for specific RGS proteins in multiple signaling pathways at neuronal synapses, regulating both pre- and postsynaptic signaling events and synaptic plasticity. Here, we review and highlight the current knowledge of specific RGS proteins (RGS2, RGS4, RGS7, RGS9-2, and RGS14) that have been clearly demonstrated to serve critical roles in modulating synaptic signaling and plasticity throughout the brain, and we consider their potential as future therapeutic targets.
Collapse
Affiliation(s)
- Kyle J Gerber
- Programs in Molecular and Systems Pharmacology (K.J.G., K.E.S., J.R.H.) and Neuroscience (J.R.H.), Department of Pharmacology (K.J.G., K.E.S., J.R.H.), Emory University School of Medicine, Atlanta, Georgia
| | - Katherine E Squires
- Programs in Molecular and Systems Pharmacology (K.J.G., K.E.S., J.R.H.) and Neuroscience (J.R.H.), Department of Pharmacology (K.J.G., K.E.S., J.R.H.), Emory University School of Medicine, Atlanta, Georgia
| | - John R Hepler
- Programs in Molecular and Systems Pharmacology (K.J.G., K.E.S., J.R.H.) and Neuroscience (J.R.H.), Department of Pharmacology (K.J.G., K.E.S., J.R.H.), Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
23
|
Doupnik CA. RGS Redundancy and Implications in GPCR-GIRK Signaling. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 123:87-116. [PMID: 26422983 DOI: 10.1016/bs.irn.2015.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Regulators of G protein signaling (RGS proteins) are key components of GPCR complexes, interacting directly with G protein α-subunits to enhance their intrinsic GTPase activity. The functional consequence is an accelerated termination of G protein effectors including certain ion channels. RGS proteins have a profound impact on the membrane-delimited gating behavior of G-protein-activated inwardly rectifying K(+) (GIRK) channels as demonstrated in reconstitution assays and recent RGS knockout mice studies. Akin to GPCRs and G protein αβγ subunits, multiple RGS isoforms are expressed within single GIRK-expressing neurons, suggesting functional redundancy and/or specificity in GPCR-GIRK channel signaling. The extent and impact of RGS redundancy in neuronal GPCR-GIRK channel signaling is currently not fully appreciated; however, recent studies from RGS knockout mice are providing important new clues on the impact of individual endogenous RGS proteins and the extent of RGS functional redundancy. Incorporating "tools" such as engineered RGS-resistant Gαi/o subunits provide an important assessment method for determining the impact of all endogenous RGS proteins on a given GPCR response and an accounting benchmark to assess the impact of individual RGS knockouts on overall RGS redundancy within a given neuron. Elucidating the degree of regulation attributable to specific RGS proteins in GIRK channel function will aid in the assessment of individual RGS proteins as viable therapeutic targets in epilepsy, ataxia's, memory disorders, and a growing list of neurological disorders.
Collapse
Affiliation(s)
- Craig A Doupnik
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida, USA.
| |
Collapse
|
24
|
Dansithong W, Paul S, Figueroa KP, Rinehart MD, Wiest S, Pflieger LT, Scoles DR, Pulst SM. Ataxin-2 regulates RGS8 translation in a new BAC-SCA2 transgenic mouse model. PLoS Genet 2015; 11:e1005182. [PMID: 25902068 PMCID: PMC4406435 DOI: 10.1371/journal.pgen.1005182] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 03/28/2015] [Indexed: 12/13/2022] Open
Abstract
Spinocerebellar ataxia type 2 (SCA2) is an autosomal dominant disorder with progressive degeneration of cerebellar Purkinje cells (PCs) and other neurons caused by expansion of a glutamine (Q) tract in the ATXN2 protein. We generated BAC transgenic lines in which the full-length human ATXN2 gene was transcribed using its endogenous regulatory machinery. Mice with the ATXN2 BAC transgene with an expanded CAG repeat (BAC-Q72) developed a progressive cellular and motor phenotype, whereas BAC mice expressing wild-type human ATXN2 (BAC-Q22) were indistinguishable from control mice. Expression analysis of laser-capture microdissected (LCM) fractions and regional expression confirmed that the BAC transgene was expressed in PCs and in other neuronal groups such as granule cells (GCs) and neurons in deep cerebellar nuclei as well as in spinal cord. Transcriptome analysis by deep RNA-sequencing revealed that BAC-Q72 mice had progressive changes in steady-state levels of specific mRNAs including Rgs8, one of the earliest down-regulated transcripts in the Pcp2-ATXN2[Q127] mouse line. Consistent with LCM analysis, transcriptome changes analyzed by deep RNA-sequencing were not restricted to PCs, but were also seen in transcripts enriched in GCs such as Neurod1. BAC-Q72, but not BAC-Q22 mice had reduced Rgs8 mRNA levels and even more severely reduced steady-state protein levels. Using RNA immunoprecipitation we showed that ATXN2 interacted selectively with RGS8 mRNA. This interaction was impaired when ATXN2 harbored an expanded polyglutamine. Mutant ATXN2 also reduced RGS8 expression in an in vitro coupled translation assay when compared with equal expression of wild-type ATXN2-Q22. Reduced abundance of Rgs8 in Pcp2-ATXN2[Q127] and BAC-Q72 mice supports our observations of a hyper-excitable mGluR1-ITPR1 signaling axis in SCA2, as RGS proteins are linked to attenuating mGluR1 signaling.
Collapse
Affiliation(s)
- Warunee Dansithong
- Department of Neurology, University of Utah, Salt Lake City, Utah, United States of America
| | - Sharan Paul
- Department of Neurology, University of Utah, Salt Lake City, Utah, United States of America
| | - Karla P. Figueroa
- Department of Neurology, University of Utah, Salt Lake City, Utah, United States of America
| | - Marc D. Rinehart
- Department of Neurology, University of Utah, Salt Lake City, Utah, United States of America
| | - Shaina Wiest
- Department of Neurology, University of Utah, Salt Lake City, Utah, United States of America
| | - Lance T. Pflieger
- Department of Neurology, University of Utah, Salt Lake City, Utah, United States of America
| | - Daniel R. Scoles
- Department of Neurology, University of Utah, Salt Lake City, Utah, United States of America
| | - Stefan M. Pulst
- Department of Neurology, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
25
|
Stewart A, Maity B, Fisher RA. Two for the Price of One: G Protein-Dependent and -Independent Functions of RGS6 In Vivo. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 133:123-51. [PMID: 26123305 DOI: 10.1016/bs.pmbts.2015.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Regulator of G protein signaling 6 (RGS6) is unique among the members of the RGS protein family as it remains the only protein with the demonstrated capacity to control G protein-dependent and -independent signaling cascades in vivo. RGS6 inhibits signaling mediated by γ-aminobutyric acid B receptors, serotonin 1A receptors, μ opioid receptors, and muscarinic acetylcholine 2 receptors. RGS6 deletion triggers distinct behavioral phenotypes resulting from potentiated signaling by these G protein-coupled receptors namely ataxia, a reduction in anxiety and depression, enhanced analgesia, and increased parasympathetic tone, respectively. In addition, RGS6 possesses potent proapoptotic and growth suppressive actions. In heart, RGS6-dependent reactive oxygen species (ROS) production promotes doxorubicin (Dox)-induced cardiomyopathy, while in cancer cells RGS6/ROS signaling is necessary for activation of the ataxia telangiectasia mutated/p53/apoptosis pathway required for the chemotherapeutic efficacy of Dox. Further, by facilitating Tip60 (trans-acting regulator protein of HIV type 1-interacting protein 60 kDa)-dependent DNA methyltransferase 1 degradation, RGS6 suppresses cellular transformation in response to oncogenic Ras. The culmination of these G protein-independent actions results in potent tumor suppressor actions of RGS6 in the murine mammary epithelium. This work summarizes evidence from human genetic studies and model animals implicating RGS6 in normal physiology, disease, and the pharmacological actions of multiple drugs. Though efforts by multiple laboratories have contributed to the ever-growing RGS6 oeuvre, the pleiotropic nature of this gene will likely lead to additional work detailing the importance of RGS6 in neuropsychiatric disorders, cardiovascular disease, and cancer.
Collapse
Affiliation(s)
- Adele Stewart
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Biswanath Maity
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Rory A Fisher
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
| |
Collapse
|
26
|
Regulator of G-protein signaling 6 (RGS6) in cardiology and oncology. Int J Cardiol 2015; 187:99-102. [PMID: 25828322 DOI: 10.1016/j.ijcard.2015.03.278] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/19/2015] [Indexed: 02/06/2023]
|
27
|
Kim G, Jung S, Son H, Kim S, Choi J, Lee DH, Roh GS, Kang SS, Cho GJ, Choi WS, Kim HJ. The GABAB receptor associates with regulators of G-protein signaling 4 protein in the mouse prefrontal cortex and hypothalamus. BMB Rep 2015; 47:324-9. [PMID: 24286319 PMCID: PMC4163873 DOI: 10.5483/bmbrep.2014.47.6.162] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Indexed: 01/29/2023] Open
Abstract
Regulators of G-protein signaling (RGS) proteins regulate certain G-protein-coupled receptor (GPCR)-mediated signaling pathways. The GABAB receptor (GABABR) is a GPCR that plays a role in the stress response. Previous studies indicate that acute immobilization stress (AIS) decreases RGS4 in the prefrontal cortex (PFC) and hypothalamus (HY) and suggest the possibility of a signal complex composed of RGS4 and GABABR. Therefore, in the present study, we tested whether RGS4 associates with GABABR in these brain regions. We found the co-localization of RGS4 and GABABR subtypes in the PFC and HY using double immunohistochemistry and confirmed a direct association between GABAB2R and RGS4 proteins using co-immunoprecipitation. Furthermore, we found that AIS decreased the amount of RGS4 bound to GABAB2R and the number of double-positive cells. These results indicate that GABABR forms a signal complex with RGS4 and suggests that RGS4 is a regulator of GABABR. [BMB Reports 2014; 47(6): 324-329]
Collapse
Affiliation(s)
- Gyeongwha Kim
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| | - Soonwoong Jung
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| | - Hyeonwi Son
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| | - Sujeong Kim
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| | - Jungil Choi
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| | - Dong Hoon Lee
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| | - Gu Seob Roh
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| | - Sang Soo Kang
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| | - Gyeong Jae Cho
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| | - Wan Sung Choi
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| | - Hyun Joon Kim
- Department of Anatomy and Neurobiology, Institute of Health Sciences, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University, Jinju 660-290, Korea
| |
Collapse
|
28
|
Regulator of G protein signaling 6 is a critical mediator of both reward-related behavioral and pathological responses to alcohol. Proc Natl Acad Sci U S A 2015; 112:E786-95. [PMID: 25646431 DOI: 10.1073/pnas.1418795112] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Alcohol is the most commonly abused drug worldwide, and chronic alcohol consumption is a major etiological factor in the development of multiple pathological sequelae, including alcoholic cardiomyopathy and hepatic cirrhosis. Here, we identify regulator of G protein signaling 6 (RGS6) as a critical regulator of both alcohol-seeking behaviors and the associated cardiac and hepatic morbidities through two mechanistically divergent signaling actions. RGS6(-/-) mice consume less alcohol when given free access and are less susceptible to alcohol-induced reward and withdrawal. Antagonism of GABA(B) receptors or dopamine D2 receptors partially reversed the reduction in alcohol consumption in RGS6(-/-) animals. Strikingly, dopamine transporter inhibition completely restored alcohol seeking in mice lacking RGS6. RGS6 deficiency was associated with alterations in the expression of genes controlling dopamine (DA) homeostasis and a reduction in DA levels in the striatum. Taken together, these data implicate RGS6 as an essential regulator of DA bioavailability. RGS6 deficiency also provided dramatic protection against cardiac hypertrophy and fibrosis, hepatic steatosis, and gastrointestinal barrier dysfunction and endotoxemia when mice were forced to consume alcohol. Although RGS proteins canonically function as G-protein regulators, RGS6-dependent, alcohol-mediated toxicity in the heart, liver, and gastrointestinal tract involves the ability of RGS6 to promote reactive oxygen species-dependent apoptosis, an action independent of its G-protein regulatory capacity. We propose that inhibition of RGS6 might represent a viable means to reduce alcohol cravings and withdrawal in human patients, while simultaneously protecting the heart and liver from further damage upon relapse.
Collapse
|
29
|
Raveh A, Turecek R, Bettler B. Mechanisms of fast desensitization of GABA(B) receptor-gated currents. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 73:145-65. [PMID: 25637440 DOI: 10.1016/bs.apha.2014.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
GABA(B) receptors (GABA(B)Rs) regulate the excitability of most neurons in the central nervous system by modulating the activity of enzymes and ion channels. In the sustained presence of the neurotransmitter γ-aminobutyric acid, GABA(B)Rs exhibit a time-dependent decrease in the receptor response-a phenomenon referred to as homologous desensitization. Desensitization prevents excessive receptor influences on neuronal activity. Much work focused on the mechanisms of GABA(B)R desensitization that operate at the receptor and control receptor expression at the plasma membrane. Over the past few years, it became apparent that GABA(B)Rs additionally evolved mechanisms for faster desensitization. These mechanisms operate at the G protein rather than at the receptor and inhibit G protein signaling within seconds of agonist exposure. The mechanisms for fast desensitization are ideally suited to regulate receptor-activated ion channel responses, which influence neuronal activity on a faster timescale than effector enzymes. Here, we provide an update on the mechanisms for fast desensitization of GABA(B)R responses and discuss physiological and pathophysiological implications.
Collapse
Affiliation(s)
- Adi Raveh
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Basel, Switzerland
| | - Rostislav Turecek
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Basel, Switzerland; Department of Auditory Neuroscience, Institute of Experimental Medicine, ASCR, Prague, Czech Republic
| | - Bernhard Bettler
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
30
|
de Velasco EMF, McCall N, Wickman K. GIRK Channel Plasticity and Implications for Drug Addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 123:201-38. [DOI: 10.1016/bs.irn.2015.05.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
31
|
GIRK Channels: A Potential Link Between Learning and Addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 123:239-77. [PMID: 26422987 DOI: 10.1016/bs.irn.2015.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The ability of drug-associated cues to reinitiate drug craving and seeking, even after long periods of abstinence, has led to the hypothesis that addiction represents a form of pathological learning, in which drugs of abuse hijack normal learning and memory processes to support long-term addictive behaviors. In this chapter, we review evidence suggesting that G protein-gated inwardly rectifying potassium (GIRK/Kir3) channels are one mechanism through which numerous drugs of abuse can modulate learning and memory processes. We will examine the role of GIRK channels in two forms of experience-dependent long-term changes in neuronal function: homeostatic plasticity and synaptic plasticity. We will also discuss how drug-induced changes in GIRK-mediated signaling can lead to changes that support the development and maintenance of addiction.
Collapse
|
32
|
Rgs6 is required for adult maintenance of dopaminergic neurons in the ventral substantia nigra. PLoS Genet 2014; 10:e1004863. [PMID: 25501001 PMCID: PMC4263397 DOI: 10.1371/journal.pgen.1004863] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/29/2014] [Indexed: 12/03/2022] Open
Abstract
Parkinson disease (PD) is characterized by the preferential, but poorly understood, vulnerability to degeneration of midbrain dopaminergic (mDA) neurons in the ventral substantia nigra compacta (vSNc). These sensitive mDA neurons express Pitx3, a transcription factor that is critical for their survival during development. We used this dependence to identify, by flow cytometry and expression profiling, the negative regulator of G-protein signaling Rgs6 for its restricted expression in these neurons. In contrast to Pitx3−/− mDA neurons that die during fetal (vSNc) or post-natal (VTA) period, the vSNc mDA neurons of Rgs6−/− mutant mice begin to exhibit unilateral signs of degeneration at around 6 months of age, and by one year cell loss is observed in a fraction of mice. Unilateral cell loss is accompanied by contralateral degenerating neurons that exhibit smaller cell size, altered morphology and reduced dendritic network. The degenerating neurons have low levels of tyrosine hydroxylase (TH) and decreased nuclear Pitx3; accordingly, expression of many Pitx3 target gene products is altered, including Vmat2, Bdnf, Aldh1a1 (Adh2) and Fgf10. These low TH neurons also express markers of increased dopamine signaling, namely increased DAT and phospho-Erk1/2 expression. The late onset degeneration may reflect the protective action of Rgs6 against excessive DA signaling throughout life. Rgs6-dependent protection is thus critical for adult survival and maintenance of the vSNc mDA neurons that are most affected in PD. The locomotor deficits associated with Parkinson disease result from the death of a specific subset of dopamine neurons in the ventral part of the midbrain. The reason for the greater sensitivity to degeneration of those, relative to other, neurons is not clear. Prior work showed that the Pitx3 transcription factor is specifically expressed in these neurons where it has a survival role during development. The present work identified a cell signaling component, Rgs6, that is also restricted to the sensitive neurons in the midbrain and that exerts a protective function, particularly late in life. While the loss of Rgs6 function may predispose or contribute to Parkinson disease, its stimulation may provide a novel therapeutic avenue to treat Parkinson disease.
Collapse
|
33
|
Lambert CS, Philpot RM, Engberg ME, Johns BE, Kim SH, Wecker L. Gait analysis and the cumulative gait index (CGI): Translational tools to assess impairments exhibited by rats with olivocerebellar ataxia. Behav Brain Res 2014; 274:334-43. [PMID: 25116252 DOI: 10.1016/j.bbr.2014.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/30/2014] [Accepted: 08/03/2014] [Indexed: 11/24/2022]
Abstract
Deviations from 'normal' locomotion exhibited by humans and laboratory animals may be determined using automated systems that capture both temporal and spatial gait parameters. Although many measures generated by these systems are unrelated and independent, some may be related and dependent, representing redundant assessments of function. To investigate this possibility, a treadmill-based system was used to capture gait parameters from normal and ataxic rats, and a multivariate analysis was conducted to determine deviations from normal. Rats were trained on the treadmill at two speeds, and gait parameters were generated prior to and following lesions of the olivocerebellar pathway. Control (non-lesioned) animals exhibited stable hindlimb gait parameters across assessments at each speed. Lesioned animals exhibited alterations in multiple hindlimb gait parameters, characterized by significant increases in stride frequency, braking duration, stance width, step angle, and paw angle and decreases in stride, stance, swing and propulsion durations, stride length and paw area. A principal component analysis of initial hindlimb measures indicated three uncorrelated factors mediating performance, termed Rhythmicity, Thrust and Contact. Deviation in the performance of each animal from the group mean was determined for each factor and values summed to yield the cumulative gait index (CGI), a single value reflecting variation within the group. The CGI for lesioned animals increased 2.3-fold relative to unlesioned animals. This study characterizes gait alterations in laboratory rats rendered ataxic by destruction of the climbing fiber pathway innervating Purkinje cells and demonstrates that a single index can be used to describe overall gait impairments.
Collapse
Affiliation(s)
- C S Lambert
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, USA; Laboratory of Neuropsychopharmacology, Department of Psychiatry and Behavioral Neurosciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - R M Philpot
- Laboratory of Neuropsychopharmacology, Department of Psychiatry and Behavioral Neurosciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - M E Engberg
- Laboratory of Neuropsychopharmacology, Department of Psychiatry and Behavioral Neurosciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - B E Johns
- Laboratory of Neuropsychopharmacology, Department of Psychiatry and Behavioral Neurosciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - S H Kim
- School of Physical Therapy and Rehabilitation Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - L Wecker
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, USA; Laboratory of Neuropsychopharmacology, Department of Psychiatry and Behavioral Neurosciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA; School of Physical Therapy and Rehabilitation Sciences, University of South Florida Morsani College of Medicine, Tampa, FL, USA; Department of Neurology, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
34
|
Tuggle K, Ali MW, Salazar H, Hooks SB. Regulator of G protein signaling transcript expression in human neural progenitor differentiation: R7 subfamily regulation by DNA methylation. Neurosignals 2014; 22:43-51. [PMID: 24903911 DOI: 10.1159/000362128] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/10/2014] [Indexed: 11/19/2022] Open
Abstract
G protein-coupled receptors (GPCRs) and their ligands are critical regulators of neural progenitor differentiation, and GPCR signaling pathways are regulated by regulator of G protein signaling (RGS) proteins. RGS protein expression is dynamically regulated, and we have recently described the epigenetic regulation of RGS transcript expression. Given the potential of RGS proteins to regulate GPCR signaling and the established role of epigenetic regulation in progenitor differentiation, we explored the impact of epigenetic regulation of RGS transcripts during in vitro differentiation of human neural progenitors. Here, we demonstrate robust upregulation of the RGS transcripts RGS4, RGS5, RGS6, RGS7, and RGS11 during neuronal differentiation, while DNA methyltransferase (DNMT) and histone deacetylase enzyme expression is suppressed during differentiation. Transcripts encoding R7 subfamily RGS proteins and the R7-binding partners R7BP and R9AP showed the greatest upregulation. Further, we showed that direct pharmacological inhibition of DNMT activity enhances expression of RGS2, RGS4, RGS5, RGS6, RGS7, RGS8, RGS9L, RGS10, and RGS14 as well as R7BP and R9AP transcripts in progenitors, consistent with regulation by DNMTs. Our results reveal marked upregulation of RGS expression during neuronal differentiation and suggest that decreased expression of DNMT enzymes during differentiation contributes to upregulation.
Collapse
Affiliation(s)
- Katie Tuggle
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Ga., USA
| | | | | | | |
Collapse
|
35
|
Ostrovskaya O, Xie K, Masuho I, Fajardo-Serrano A, Lujan R, Wickman K, Martemyanov KA. RGS7/Gβ5/R7BP complex regulates synaptic plasticity and memory by modulating hippocampal GABABR-GIRK signaling. eLife 2014; 3:e02053. [PMID: 24755289 PMCID: PMC3988575 DOI: 10.7554/elife.02053] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In the hippocampus, the inhibitory neurotransmitter GABA shapes the activity of the output pyramidal neurons and plays important role in cognition. Most of its inhibitory effects are mediated by signaling from GABAB receptor to the G protein-gated Inwardly-rectifying K+ (GIRK) channels. Here, we show that RGS7, in cooperation with its binding partner R7BP, regulates GABABR-GIRK signaling in hippocampal pyramidal neurons. Deletion of RGS7 in mice dramatically sensitizes GIRK responses to GABAB receptor stimulation and markedly slows channel deactivation kinetics. Enhanced activity of this signaling pathway leads to decreased neuronal excitability and selective disruption of inhibitory forms of synaptic plasticity. As a result, mice lacking RGS7 exhibit deficits in learning and memory. We further report that RGS7 is selectively modulated by its membrane anchoring subunit R7BP, which sets the dynamic range of GIRK responses. Together, these results demonstrate a novel role of RGS7 in hippocampal synaptic plasticity and memory formation. DOI:http://dx.doi.org/10.7554/eLife.02053.001 Neurons communicate with one another at junctions called synapses. The arrival of an electrical signal known as an action potential at the first cell causes molecules known as neurotransmitters to be released into the synapse. These molecules diffuse across the gap between the neurons and bind to receptors on the receiving cell. Some neurotransmitters, such as glutamate, activate cells when they bind to receptors, thus making it easier for the second neuron to ‘fire’ (i.e., to generate an action potential). By contrast, other neurotransmitters, such as GABA, usually make it harder for the second neuron to fire. Many of the effects of GABA involve a type of receptor called GABAB. When GABA binds to one of these receptors, a molecule called a G-protein is recruited to the receptor. This activates the G-protein, triggering a cascade of events inside the cell that lead ultimately to the opening of potassium ion channels, which as known as GIRKs, in the cell membrane. Positively charged potassium ions then leave the cell through these channels, and this makes it more difficult for the cell to fire. Now, Ostrovskaya et al. have revealed that a complex of three proteins regulates the interaction between GABAB receptors and GIRK channels. In neurons that lack either of these proteins, the receptors have less influence on GIRKs than in normal cells. Moreover, mice that lack one of the proteins (called RGS7) perform less well in various learning and memory tests: for example, they take longer than normal animals to learn the location of an escape platform in a water maze, or to retain a memory of a fearful event. By identifying the proteins that regulate the interaction between GABAB receptors and GIRKs, Ostrovskaya et al. have helped to unravel a key signaling cascade relevant to cognition. Given that GIRK channels have recently been implicated in Down’s syndrome, these insights may also increase understanding of cognitive impairments in neuropsychiatric disorders. DOI:http://dx.doi.org/10.7554/eLife.02053.002
Collapse
Affiliation(s)
- Olga Ostrovskaya
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | | | | | | | | | | | | |
Collapse
|
36
|
Stewart A, Maity B, Wunsch AM, Meng F, Wu Q, Wemmie JA, Fisher RA. Regulator of G-protein signaling 6 (RGS6) promotes anxiety and depression by attenuating serotonin-mediated activation of the 5-HT(1A) receptor-adenylyl cyclase axis. FASEB J 2014; 28:1735-44. [PMID: 24421401 DOI: 10.1096/fj.13-235648] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Targeting serotonin (5-HT) bioavailability with selective 5-HT reuptake inhibitors (SSRIs) remains the most widely used treatment for mood disorders. However, their limited efficacy, delayed onset of action, and side effects restrict their clinical utility. Endogenous regulator of G-protein signaling (RGS) proteins have been implicated as key inhibitors of 5-HT(1A)Rs, whose activation is believed to underlie the beneficial effects of SSRIs, but the identity of the specific RGS proteins involved remains unknown. We identify RGS6 as the critical negative regulator of 5-HT(1A)R-dependent antidepressant actions. RGS6 is enriched in hippocampal and cortical neurons, 5-HT(1A)R-expressing cells implicated in mood disorders. RGS6(-/-) mice exhibit spontaneous anxiolytic and antidepressant behavior rapidly and completely reversibly by 5-HT(1A)R blockade. Effects of the SSRI fluvoxamine and 5-HT(1A)R agonist 8-OH-DPAT were also potentiated in RGS6(+/-) mice. The phenotype of RGS6(-/-) mice was associated with decreased CREB phosphorylation in the hippocampus and cortex, implicating enhanced Gα(i)-dependent adenylyl cyclase inhibition as a possible causative factor in the behavior observed in RGS6(-/-) animals. Our results demonstrate that by inhibiting serotonergic innervation of the cortical-limbic neuronal circuit, RGS6 exerts powerful anxiogenic and prodepressant actions. These findings indicate that RGS6 inhibition may represent a viable means to treat mood disorders or enhance the efficacy of serotonergic agents.
Collapse
Affiliation(s)
- Adele Stewart
- 3Department of Pharmacology, University of Iowa, Carver College of Medicine, 51 Newton Rd., BSB 2-512, Iowa City, IA 52242, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Luján R, Marron Fernandez de Velasco E, Aguado C, Wickman K. New insights into the therapeutic potential of Girk channels. Trends Neurosci 2013; 37:20-9. [PMID: 24268819 DOI: 10.1016/j.tins.2013.10.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/24/2013] [Accepted: 10/25/2013] [Indexed: 01/01/2023]
Abstract
G protein-dependent signaling pathways control the activity of excitable cells of the nervous system and heart, and are the targets of neurotransmitters, clinically relevant drugs, and drugs of abuse. G protein-gated inwardly rectifying potassium (K(+)) (Girk/Kir3) channels are a key effector in inhibitory signaling pathways. Girk-dependent signaling contributes to nociception and analgesia, reward-related behavior, mood, cognition, and heart-rate regulation, and has been linked to epilepsy, Down syndrome, addiction, and arrhythmias. We discuss recent advances in our understanding of Girk channel structure, organization in signaling complexes, and plasticity, as well as progress on the development of subunit-selective Girk modulators. These findings offer new hope for the selective manipulation of Girk channels to treat a variety of debilitating afflictions.
Collapse
Affiliation(s)
- Rafael Luján
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Campus Biosanitario, C/Almansa 14, 02008 Albacete, Spain.
| | | | - Carolina Aguado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Campus Biosanitario, C/Almansa 14, 02008 Albacete, Spain
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, 321 Church Street South East, Minneapolis, MN 55455, USA.
| |
Collapse
|
38
|
Membrane channels as integrators of G-protein-mediated signaling. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:521-31. [PMID: 24028827 DOI: 10.1016/j.bbamem.2013.08.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 08/14/2013] [Accepted: 08/21/2013] [Indexed: 01/03/2023]
Abstract
A variety of extracellular stimuli regulate cellular responses via membrane receptors. A well-known group of seven-transmembrane domain-containing proteins referred to as G protein-coupled receptors, directly couple with the intracellular GTP-binding proteins (G proteins) across cell membranes and trigger various cellular responses by regulating the activity of several enzymes as well as ion channels. Many specific populations of ion channels are directly controlled by G proteins; however, indirect modulation of some channels by G protein-dependent phosphorylation events and lipid metabolism is also observed. G protein-mediated diverse modifications affect the ion channel activities and spatio-temporally regulate membrane potentials as well as of intracellular Ca(2+) concentrations in both excitatory and non-excitatory cells. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.
Collapse
|
39
|
Huang J, Stewart A, Maity B, Hagen J, Fagan RL, Yang J, Quelle DE, Brenner C, Fisher RA. RGS6 suppresses Ras-induced cellular transformation by facilitating Tip60-mediated Dnmt1 degradation and promoting apoptosis. Oncogene 2013; 33:3604-11. [PMID: 23995786 DOI: 10.1038/onc.2013.324] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 04/16/2013] [Accepted: 07/04/2013] [Indexed: 12/17/2022]
Abstract
The RAS protooncogene has a central role in regulation of cell proliferation, and point mutations leading to oncogenic activation of Ras occur in a large number of human cancers. Silencing of tumor-suppressor genes by DNA methyltransferase 1 (Dnmt1) is essential for oncogenic cellular transformation by Ras, and Dnmt1 is overexpressed in numerous human cancers. Here we provide new evidence that the pleiotropic regulator of G protein signaling (RGS) family member RGS6 suppresses Ras-induced cellular transformation by facilitating Tip60-mediated degradation of Dmnt1 and promoting apoptosis. Employing mouse embryonic fibroblasts from wild-type and RGS6(-/-) mice, we found that oncogenic Ras induced upregulation of RGS6, which in turn blocked Ras-induced cellular transformation. RGS6 functions to suppress cellular transformation in response to oncogenic Ras by downregulating Dnmt1 protein expression leading to inhibition of Dnmt1-mediated anti-apoptotic activity. Further experiments showed that RGS6 functions as a scaffolding protein for both Dnmt1 and Tip60 and is required for Tip60-mediated acetylation of Dnmt1 and subsequent Dnmt1 ubiquitylation and degradation. The RGS domain of RGS6, known only for its GTPase-activating protein activity toward Gα subunits, was sufficient to mediate Tip60 association with RGS6. This work demonstrates a novel signaling action for RGS6 in negative regulation of oncogene-induced transformation and provides new insights into our understanding of the mechanisms underlying Ras-induced oncogenic transformation and regulation of Dnmt1 expression. Importantly, these findings identify RGS6 as an essential cellular defender against oncogenic stress and a potential therapeutic target for developing new cancer treatments.
Collapse
Affiliation(s)
- J Huang
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - A Stewart
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - B Maity
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - J Hagen
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - R L Fagan
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - J Yang
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - D E Quelle
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - C Brenner
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - R A Fisher
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
40
|
Maity B, Stewart A, O'Malley Y, Askeland RW, Sugg SL, Fisher RA. Regulator of G protein signaling 6 is a novel suppressor of breast tumor initiation and progression. Carcinogenesis 2013; 34:1747-55. [PMID: 23598467 DOI: 10.1093/carcin/bgt128] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Breast cancer is a large global health burden and the most frequently diagnosed malignancy in women worldwide. Here, we utilize RGS6(-/-) mice to interrogate the role of regulator of G protein signaling 6 (RGS6), localized to the ductal epithelium in mouse and human breast, as a novel tumor suppressor in vivo. RGS6(-/-) mice exhibit accelerated 7,12-dimethylbenza[α]anthracene (DMBA)-induced tumor initiation and progression, as well as decreased overall survival. Analysis of carcinogenic aberrations in the mammary glands of DMBA-treated mice revealed a failure of the DNA damage response concurrent with augmented oncogenesis in RGS6(-/-) animals. Furthermore, RGS6 suppressed cell growth induced by either human epidermal growth factor receptor 2 or estrogen receptor activation in both MCF-7 breast cancer cells and mammary epithelial cells (MECs). MECs isolated from RGS6(-/-) mice also showed a deficit in DMBA-induced ATM/p53 activation, reactive oxygen species generation and apoptosis confirming that RGS6 is required for effective activation of the DNA damage response in these cells, a critical countermeasure against carcinogen-mediated genotoxic stress. The ability of RGS6 to simultaneously enhance DNA-damage-induced apoptotic signaling and suppress oncogenic cell growth likely underlie the accelerated tumorigenesis and cellular transformation observed in DMBA-treated RGS6(-/-) mice and isolated MECs, respectively. Unsurprisingly, spontaneous tumor formation was also seen in old female RGS6(-/-) but not in wild-type mice. Our finding that RGS6 is downregulated in all human breast cancer subtypes independent of their molecular classification indicates that obtaining a means to restore the growth suppressive and pro-apoptotic actions of RGS6 in breast might be a viable means to treat a large spectrum of breast tumors.
Collapse
Affiliation(s)
- Biswanath Maity
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | | | | | | | | |
Collapse
|
41
|
Seddik R, Jungblut SP, Silander OK, Rajalu M, Fritzius T, Besseyrias V, Jacquier V, Fakler B, Gassmann M, Bettler B. Opposite effects of KCTD subunit domains on GABA(B) receptor-mediated desensitization. J Biol Chem 2012; 287:39869-77. [PMID: 23035119 DOI: 10.1074/jbc.m112.412767] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
GABA(B) receptors assemble from principle and auxiliary subunits. The principle subunits GABA(B1) and GABA(B2) form functional heteromeric GABA(B(1,2)) receptors that associate with homotetramers of auxiliary KCTD8, -12, -12b, or -16 (named after their K(+) channel tetramerization domain) subunits. These auxiliary subunits constitute receptor subtypes with distinct functional properties. KCTD12 and -12b generate desensitizing receptor responses while KCTD8 and -16 generate largely non-desensitizing receptor responses. The structural elements of the KCTDs underlying these differences in desensitization are unknown. KCTDs are modular proteins comprising a T1 tetramerization domain, which binds to GABA(B2), and a H1 homology domain. KCTD8 and -16 contain an additional C-terminal H2 homology domain that is not sequence-related to the H1 domains. No functions are known for the H1 and H2 domains. Here we addressed which domains and sequence motifs in KCTD proteins regulate desensitization of the receptor response. We found that the H1 domains in KCTD12 and -12b mediate desensitization through a particular sequence motif, T/NFLEQ, which is not present in the H1 domains of KCTD8 and -16. In addition, the H2 domains in KCTD8 and -16 inhibit desensitization when expressed C-terminal to the H1 domains but not when expressed as a separate protein in trans. Intriguingly, the inhibitory effect of the H2 domain is sequence-independent, suggesting that the H2 domain sterically hinders desensitization by the H1 domain. Evolutionary analysis supports that KCTD12 and -12b evolved desensitizing properties by liberating their H1 domains from antagonistic H2 domains and acquisition of the T/NFLEQ motif.
Collapse
Affiliation(s)
- Riad Seddik
- Department of Biomedicine, University of Basel, 4056 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|