1
|
McNeill SM, Zhao P. The roles of RGS proteins in cardiometabolic disease. Br J Pharmacol 2024; 181:2319-2337. [PMID: 36964984 DOI: 10.1111/bph.16076] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/12/2023] [Accepted: 03/20/2023] [Indexed: 03/27/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are the most prominent receptors on the surface of the cell and play a central role in the regulation of cardiac and metabolic functions. GPCRs transmit extracellular stimuli to the interior of the cells by activating one or more heterotrimeric G proteins. The duration and intensity of G protein-mediated signalling are tightly controlled by a large array of intracellular mediators, including the regulator of G protein signalling (RGS) proteins. RGS proteins selectively promote the GTPase activity of a subset of Gα subunits, thus serving as negative regulators in a pathway-dependent manner. In the current review, we summarise the involvement of RGS proteins in cardiometabolic function with a focus on their tissue distribution, mechanisms of action and dysregulation under various disease conditions. We also discuss the potential therapeutic applications for targeting RGS proteins in treating cardiometabolic conditions and current progress in developing RGS modulators. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Samantha M McNeill
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Peishen Zhao
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins (CCeMMP), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
2
|
Ahlers-Dannen KE, Yang J, Spicer MM, Fu D, DeVore A, Fisher RA. A splice acceptor variant in RGS6 associated with intellectual disability, microcephaly, and cataracts disproportionately promotes expression of a subset of RGS6 isoforms. J Hum Genet 2024; 69:145-152. [PMID: 38332109 PMCID: PMC11485174 DOI: 10.1038/s10038-024-01220-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024]
Abstract
Intellectual disability (ID) is associated with an increased risk of developing psychiatric disorders, suggesting a common underlying genetic factor. Importantly, altered signaling and/or expression of regulator of G protein signaling 6 (RGS6) is associated with ID and numerous psychiatric disorders. RGS6 is highly conserved and undergoes complex alternative mRNA splicing producing ~36 protein isoforms with high sequence similarity historically necessitating a global approach in functional studies. However, our recent analysis in mice revealed RGS6 is most highly expressed in CNS with RGS6L(+GGL) isoforms predominating. A previously reported genetic variant in intron 17 of RGS6 (c.1369-1G>C), associated with ID, may provide further clues into RGS6L(+GGL) isoform functional delineation. This variant was predicted to alter a highly conserved canonical 3' acceptor site creating an alternative branch point within exon 18 (included in a subset of RGS6L(+GGL) transcripts) and a frameshift forming an early stop codon. We previously identified this alternative splice site and demonstrated its use generates RGS6Lζ(+GGL) isoforms. Here, we show that the c.1369-1G>C variant disrupts the canonical, preferred (>90%) intron 17 splice site and leads to the exclusive use of the alternate exon 18 splice site, inducing disproportionate expression of a subset of isoforms, particularly RGS6Lζ(+GGL). Furthermore, RGS6 global knockout mice do not exhibit ID. Thus, ID caused by the c.1369-1G>C variant likely results from altered RGS6 isoform expression, rather than RGS6 isoform loss. In summary, these studies highlight the importance of proper RGS6 splicing and identify a previously unrecognized role of G protein signaling in ID.
Collapse
Affiliation(s)
- K E Ahlers-Dannen
- Department of Neuroscience and Pharmacology, The Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - J Yang
- Department of Neuroscience and Pharmacology, The Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - M M Spicer
- Department of Neuroscience and Pharmacology, The Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - D Fu
- Department of Neuroscience and Pharmacology, The Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - A DeVore
- Department of Neuroscience and Pharmacology, The Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - R A Fisher
- Department of Neuroscience and Pharmacology, The Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
3
|
STMN2 overexpression promotes cell proliferation and EMT in pancreatic cancer mediated by WNT/β-catenin signaling. Cancer Gene Ther 2023; 30:472-480. [PMID: 36460804 DOI: 10.1038/s41417-022-00568-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/03/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
STMN2, as a key regulator in microtubule disassembly and dynamics, has recently been shown to participate in cancer development. However, the corresponding role in pancreatic ductal adenocarcinoma (PC), to our knowledge, has not been reported yet. In the current study, we systematically investigate the potential role of STMN2 in the progression of PC in vitro and vivo. Overexpression of STMN2 was prevalently observed in 81 human cases of PC tissues compared with that in the paired adjacent pancreas (54.3% vs 18.5%, P < 0.01), which was positively associated with multiple advanced clinical stages of PC patients (tumor size, T stage, lymph-node metastasis and the poor prognosis). Meanwhile, a close correlation between high STMN2 and cytoplasmic/nuclear β-catenin expression (P = 0.007) was observed in PC tissues and cell lines. STMN2 overexpression induced EMT and cell proliferation in vitro via stimulating EMT-like cellular morphology, cell motility and proliferation, and the change of EMT (Snail1, E-cadherin and Vimentin) and Cyclin D1 signaling. However, XAV939 inhibited STMN2 overexpression-enhanced EMT and proliferation. Conversely, KY19382 reversed STMN2 silencing- inhibited EMT and cell proliferation in vitro. Furthermore, activated STMN2 and β-catenin were co-localized in cytoplasm/nuclear in vitro. β-catenin/TCF-mediated the transcription of STMN2 by the potential binding sites (TTCAAAG). Finally, STMN2 promoted subcutaneous tumor growth following the activation of EMT and Cyclin D1 signaling. STMN2 overexpression promotes the aggressive clinical stage of PC patients and promotes EMT and cell proliferation in vitro and vivo. β-catenin/TCF-mediated the transcription of STMN2.
Collapse
|
4
|
The Potential Role of R4 Regulators of G Protein Signaling (RGS) Proteins in Type 2 Diabetes Mellitus. Cells 2022; 11:cells11233897. [PMID: 36497154 PMCID: PMC9739376 DOI: 10.3390/cells11233897] [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: 10/25/2022] [Revised: 11/26/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex and heterogeneous disease that primarily results from impaired insulin secretion or insulin resistance (IR). G protein-coupled receptors (GPCRs) are proposed as therapeutic targets for T2DM. GPCRs transduce signals via the Gα protein, playing an integral role in insulin secretion and IR. The regulators of G protein signaling (RGS) family proteins can bind to Gα proteins and function as GTPase-activating proteins (GAP) to accelerate GTP hydrolysis, thereby terminating Gα protein signaling. Thus, RGS proteins determine the size and duration of cellular responses to GPCR stimulation. RGSs are becoming popular targeting sites for modulating the signaling of GPCRs and related diseases. The R4 subfamily is the largest RGS family. This review will summarize the research progress on the mechanisms of R4 RGS subfamily proteins in insulin secretion and insulin resistance and analyze their potential value in the treatment of T2DM.
Collapse
|
5
|
RGS6 Drives Spinal Cord Injury by Inhibiting AMPK Pathway in Mice. DISEASE MARKERS 2022; 2022:4535652. [PMID: 35510037 PMCID: PMC9061016 DOI: 10.1155/2022/4535652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 11/18/2022]
Abstract
Objective. Oxidative stress and inflammation play critical roles in the pathogenesis of spinal cord injury (SCI). Regulator of G protein signaling 6 (RGS6) is involved in controlling ROS generation and inflammatory response under different contexts. This study is aimed at investigating its role and underlying mechanism in SCI. Methods. Contusive SCI mouse models were generated, and lentiviral vectors were injected to silence or overexpress RGS6 in the spinal cord. To inhibit AMP-activated protein kinase (AMPK) activity, SCI mice were intraperitoneally injected with compound C (20 mg/kg) every two days. Oxidative and inflammatory markers were detected. Results. Spinal RGS6 expression was elevated upon SCI stimulation. RGS6 knockdown suppressed, while RGS6 overexpression aggravated oxidative stress, inflammation, and SCI in mice. Mechanistically, RGS6 elevation during SCI deactivated AMPK pathway, thereby exacerbating oxidative stress and inflammation in SCI mice. Conclusion. RGS6 is required for the initiation and progression of SCI, and knocking down RGS6 may provide promising therapeutic strategies for SCI patients.
Collapse
|
6
|
Li Z, Wang D, Liao H, Zhang S, Guo W, Chen L, Lu L, Huang T, Cai YD. Exploring the Genomic Patterns in Human and Mouse Cerebellums Via Single-Cell Sequencing and Machine Learning Method. Front Genet 2022; 13:857851. [PMID: 35309141 PMCID: PMC8930846 DOI: 10.3389/fgene.2022.857851] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/09/2022] [Indexed: 12/29/2022] Open
Abstract
In mammals, the cerebellum plays an important role in movement control. Cellular research reveals that the cerebellum involves a variety of sub-cell types, including Golgi, granule, interneuron, and unipolar brush cells. The functional characteristics of cerebellar cells exhibit considerable differences among diverse mammalian species, reflecting a potential development and evolution of nervous system. In this study, we aimed to recognize the transcriptional differences between human and mouse cerebellum in four cerebellar sub-cell types by using single-cell sequencing data and machine learning methods. A total of 321,387 single-cell sequencing data were used. The 321,387 cells included 4 cell types, i.e., Golgi (5,048, 1.57%), granule (250,307, 77.88%), interneuron (60,526, 18.83%), and unipolar brush (5,506, 1.72%) cells. Our results showed that by using gene expression profiles as features, the optimal classification model could achieve very high even perfect performance for Golgi, granule, interneuron, and unipolar brush cells, respectively, suggesting a remarkable difference between the genomic profiles of human and mouse. Furthermore, a group of related genes and rules contributing to the classification was identified, which might provide helpful information for deepening the understanding of cerebellar cell heterogeneity and evolution.
Collapse
Affiliation(s)
- ZhanDong Li
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Deling Wang
- Department of Radiology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - HuiPing Liao
- Eye Institute of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - ShiQi Zhang
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Lin Lu
- Department of Radiology, Columbia University Medical Center, New York, NY, United States
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
7
|
Characteristics of amniotic mesenchymal stromal cells derived from term and preterm labor. Taiwan J Obstet Gynecol 2022; 61:51-56. [PMID: 35181046 DOI: 10.1016/j.tjog.2021.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2020] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Amniotic mesenchymal stromal cells (AMSCs) can be obtained from the mesoderm of human amniotic membrane. AMSCs derived from term baby show increased expression of genes associated with apoptosis and senescence. The objective of this study was to examine gene expression profiles of AMSCs derived from preterm (preterm AMSCs) and term labors (term AMSCs) and analyze common and different mechanisms. MATERIALS AND METHODS We isolated and cultured AMSCs from 43 placentas: 27 from term labor and 16 from preterm labor. Microarray analysis and gene network analysis were performed to compare gene expression profile (GEP) of preterm (n = 6) with term AMSCs (n = 10). Senescence-associated gene (CDKN2A and CDKN2B) expression was also measured by reverse transcription quantitative PCR. RESULTS GEP demonstrated that preterm AMSCs showed upregulation of nicotinamide adenine dinucleotide biosynthetic process and downregulation of extracellular matrix, cholesterol import and transport, lipid storage, and maintenance of location. CDKN2A and CDKN2B genes showed similar expression levels between term and preterm AMSCs. CDKN2A gene expression was correlated with CDKN2B expression and population doubling time. Compared to term AMSCs, preterm AMSCs showed significantly different expression of genes associated with inflammatory response which could be one of the major players in labor events. CONCLUSION Increased CDKN2A expression in AMSCs is associated with placental membrane aging which participates in both preterm and term labor. To the best of our knowledge, this is the first report to demonstrate the association of AMSCs with labor.
Collapse
|
8
|
Sakloth F, Polizu C, Bertherat F, Zachariou V. Regulators of G Protein Signaling in Analgesia and Addiction. Mol Pharmacol 2020; 98:739-750. [PMID: 32474445 PMCID: PMC7662521 DOI: 10.1124/mol.119.119206] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Regulator of G protein signaling (RGS) proteins are multifunctional proteins expressed in peripheral and neuronal cells, playing critical roles in development, physiologic processes, and pharmacological responses. RGS proteins primarily act as GTPase accelerators for activated Gα subunits of G-protein coupled receptors, but they may also modulate signal transduction by several other mechanisms. Over the last two decades, preclinical work identified members of the RGS family with unique and critical roles in intracellular responses to drugs of abuse. New information has emerged on the mechanisms by which RGS proteins modulate the efficacy of opioid analgesics in a brain region- and agonist-selective fashion. There has also been progress in the understanding of the protein complexes and signal transduction pathways regulated by RGS proteins in addiction and analgesia circuits. In this review, we summarize findings on the mechanisms by which RGS proteins modulate functional responses to opioids in models of analgesia and addiction. We also discuss reports on the regulation and function of RGS proteins in models of psychostimulant addiction. Using information from preclinical studies performed over the last 20 years, we highlight the diverse mechanisms by which RGS protein complexes control plasticity in response to opioid and psychostimulant drug exposure; we further discuss how the understanding of these pathways may lead to new opportunities for therapeutic interventions in G protein pathways. SIGNIFICANCE STATEMENT: Regulator of G protein signaling (RGS) proteins are signal transduction modulators, expressed widely in various tissues, including brain regions mediating addiction and analgesia. Evidence from preclinical work suggests that members of the RGS family act by unique mechanisms in specific brain regions to control drug-induced plasticity. This review highlights interesting findings on the regulation and function of RGS proteins in models of analgesia and addiction.
Collapse
Affiliation(s)
- Farhana Sakloth
- Nash Family Department of Neuroscience, and Friedman Brain Institute (F.S., C.P., F.B., V.Z.) and Department of Pharmacological Sciences (V.Z.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Claire Polizu
- Nash Family Department of Neuroscience, and Friedman Brain Institute (F.S., C.P., F.B., V.Z.) and Department of Pharmacological Sciences (V.Z.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Feodora Bertherat
- Nash Family Department of Neuroscience, and Friedman Brain Institute (F.S., C.P., F.B., V.Z.) and Department of Pharmacological Sciences (V.Z.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Venetia Zachariou
- Nash Family Department of Neuroscience, and Friedman Brain Institute (F.S., C.P., F.B., V.Z.) and Department of Pharmacological Sciences (V.Z.), Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
9
|
Morin A, Goncalves J, Moog S, Castro-Vega LJ, Job S, Buffet A, Fontenille MJ, Woszczyk J, Gimenez-Roqueplo AP, Letouzé E, Favier J. TET-Mediated Hypermethylation Primes SDH-Deficient Cells for HIF2α-Driven Mesenchymal Transition. Cell Rep 2020; 30:4551-4566.e7. [DOI: 10.1016/j.celrep.2020.03.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 01/24/2020] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
|
10
|
Development of a bimolecular luminescence complementation assay for RGS: G protein interactions in cells. Anal Biochem 2017; 522:10-17. [PMID: 28115169 DOI: 10.1016/j.ab.2017.01.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 01/10/2017] [Accepted: 01/18/2017] [Indexed: 02/03/2023]
Abstract
Cell based assessment tools and screening platforms are the preferred paradigm for small molecule identification and validation due to selectively identifying molecules with cellular activity and validation of compound activity against target proteins in their native environment. With respect to Regulator of G Protein Signaling (RGS) proteins, current cell based methodologies are either low throughput or monitor downstream signaling consequences. The increasing number of reports indicating RGS function in various disease pathogeneses highlights the need for a robust RGS inhibitor discovery and characterization paradigm. Promega's NanoBit Protein Complementation Assay utilizes NanoLuc, an engineered luciferase with enhanced luminescence characteristics which allow for both robust and kinetic assessment of protein interaction formation and disruption. Here we characterized 15 separate RGS: G protein interactions using this system. The binding profile of RGS: Gα interactions correlates to prior published biochemical binding profiles of these proteins. Additionally, we demonstrated this system is suitable for high throughput screening efforts via calculation of Z-factors for three of the interactions and demonstrated that a known small molecule inhibitor of RGS4 disrupts the RGS4: Gαi1 protein-protein interaction. In conclusion, the NanoBit Protein Complementation Assay holds promise as a robust platform for discovery and characterization of RGS inhibitors.
Collapse
|
11
|
Carter MG, Smagghe BJ, Stewart AK, Rapley JA, Lynch E, Bernier KJ, Keating KW, Hatziioannou VM, Hartman EJ, Bamdad CC. A Primitive Growth Factor, NME7AB , Is Sufficient to Induce Stable Naïve State Human Pluripotency; Reprogramming in This Novel Growth Factor Confers Superior Differentiation. Stem Cells 2016; 34:847-59. [PMID: 26749426 DOI: 10.1002/stem.2261] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 11/10/2015] [Accepted: 11/26/2015] [Indexed: 12/28/2022]
Abstract
Scientists have generated human stem cells that in some respects mimic mouse naïve cells, but their dependence on the addition of several extrinsic agents, and their propensity to develop abnormal karyotype calls into question their resemblance to a naturally occurring "naïve" state in humans. Here, we report that a recombinant, truncated human NME7, referred to as NME7AB here, induces a stable naïve-like state in human embryonic stem cells and induced pluripotent stem cells without the use of inhibitors, transgenes, leukemia inhibitory factor (LIF), fibroblast growth factor 2 (FGF2), feeder cells, or their conditioned media. Evidence of a naïve state includes reactivation of the second X chromosome in female source cells, increased expression of naïve markers and decreased expression of primed state markers, ability to be clonally expanded and increased differentiation potential. RNA-seq analysis shows vast differences between the parent FGF2 grown, primed state cells, and NME7AB converted cells, but similarities to altered gene expression patterns reported by others generating naïve-like stem cells via the use of biochemical inhibitors. Experiments presented here, in combination with our previous work, suggest a mechanistic model of how human stem cells regulate self-replication: an early naïve state driven by NME7, which cannot itself limit self-replication and a later naïve state regulated by NME1, which limits self-replication when its multimerization state shifts from the active dimer to the inactive hexamer.
Collapse
Affiliation(s)
- M G Carter
- Minerva Biotechnologies, Waltham, Massachusetts, USA
| | - B J Smagghe
- Minerva Biotechnologies, Waltham, Massachusetts, USA
| | - A K Stewart
- Minerva Biotechnologies, Waltham, Massachusetts, USA
| | - J A Rapley
- Minerva Biotechnologies, Waltham, Massachusetts, USA
| | - E Lynch
- Minerva Biotechnologies, Waltham, Massachusetts, USA
| | - K J Bernier
- Minerva Biotechnologies, Waltham, Massachusetts, USA
| | - K W Keating
- Minerva Biotechnologies, Waltham, Massachusetts, USA
| | | | - E J Hartman
- Minerva Biotechnologies, Waltham, Massachusetts, USA
| | | |
Collapse
|
12
|
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: 1.0] [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
|
13
|
Woodard GE, Jardín I, Berna-Erro A, Salido GM, Rosado JA. Regulators of G-protein-signaling proteins: negative modulators of G-protein-coupled receptor signaling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 317:97-183. [PMID: 26008785 DOI: 10.1016/bs.ircmb.2015.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regulators of G-protein-signaling (RGS) proteins are a category of intracellular proteins that have an inhibitory effect on the intracellular signaling produced by G-protein-coupled receptors (GPCRs). RGS along with RGS-like proteins switch on through direct contact G-alpha subunits providing a variety of intracellular functions through intracellular signaling. RGS proteins have a common RGS domain that binds to G alpha. RGS proteins accelerate GTPase and thus enhance guanosine triphosphate hydrolysis through the alpha subunit of heterotrimeric G proteins. As a result, they inactivate the G protein and quickly turn off GPCR signaling thus terminating the resulting downstream signals. Activity and subcellular localization of RGS proteins can be changed through covalent molecular changes to the enzyme, differential gene splicing, and processing of the protein. Other roles of RGS proteins have shown them to not be solely committed to being inhibitors but behave more as modulators and integrators of signaling. RGS proteins modulate the duration and kinetics of slow calcium oscillations and rapid phototransduction and ion signaling events. In other cases, RGS proteins integrate G proteins with signaling pathways linked to such diverse cellular responses as cell growth and differentiation, cell motility, and intracellular trafficking. Human and animal studies have revealed that RGS proteins play a vital role in physiology and can be ideal targets for diseases such as those related to addiction where receptor signaling seems continuously switched on.
Collapse
Affiliation(s)
- Geoffrey E Woodard
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Isaac Jardín
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - A Berna-Erro
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - Gines M Salido
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - Juan A Rosado
- Department of Physiology, University of Extremadura, Caceres, Spain
| |
Collapse
|
14
|
Chauvin S, Sobel A. Neuronal stathmins: A family of phosphoproteins cooperating for neuronal development, plasticity and regeneration. Prog Neurobiol 2015; 126:1-18. [DOI: 10.1016/j.pneurobio.2014.09.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/23/2014] [Accepted: 09/29/2014] [Indexed: 02/06/2023]
|
15
|
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: 1.1] [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
|
16
|
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: 3.3] [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
|
17
|
Regulator of G protein signaling 19 suppresses Ras-induced neoplastic transformation and tumorigenesis. Cancer Lett 2013; 339:33-41. [PMID: 23911936 DOI: 10.1016/j.canlet.2013.07.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/12/2013] [Accepted: 07/24/2013] [Indexed: 11/20/2022]
Abstract
Regulator of G protein signaling 19 (RGS19) has recently been shown to inhibit Ras activation by upregulating the tumor metastasis suppressor Nm23. Here, we have examined the effect of RGS19 on Ras-induced oncogenesis. Coexpression of RGS19, but not RGS20, in NIH3T3 cells effectively suppressed neoplastic transformation and tumorigenesis induced by the oncogenic Ras(GV) mutant. In non-small cell lung carcinoma H1299 cells that harbor Ras mutations, shRNA-mediated knockdown of RGS19 facilitated tumorigenesis with the early appearance of large tumors in nude mice assays. Collectively, these results suggest that expression of RGS19 can suppress the oncogenic actions of Ras.
Collapse
|
18
|
Cheng YC, Scotting PJ, Hsu LS, Lin SJ, Shih HY, Hsieh FY, Wu HL, Tsao CL, Shen CJ. Zebrafish rgs4 is essential for motility and axonogenesis mediated by Akt signaling. Cell Mol Life Sci 2013; 70:935-50. [PMID: 23052218 PMCID: PMC11113239 DOI: 10.1007/s00018-012-1178-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Revised: 09/19/2012] [Accepted: 09/24/2012] [Indexed: 12/21/2022]
Abstract
The schizophrenia susceptibility gene, Rgs4, is one of the most intensively studied regulators of G-protein signaling members, well known to be fundamental in regulating neurotransmission. However, little is known about its role in the developing nervous system. We have isolated zebrafish rgs4 and shown that it is transcribed in the developing nervous system. Rgs4 knockdown did not affect neuron number and patterning but resulted in locomotion defects and aberrant development of axons. This was confirmed using a selective Rgs4 inhibitor, CCG-4986. Rgs4 knockdown also attenuated the level of phosphorylated-Akt1, and injection of constitutively-activated AKT1 rescued the motility defects and axonal phenotypes in the spinal cord but not in the hindbrain and trigeminal neurons. Our in vivo analysis reveals a novel role for Rgs4 in regulating axonogenesis during embryogenesis, which is mediated by another schizophrenia-associated gene, Akt1, in a region-specific manner.
Collapse
Affiliation(s)
- Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, 33383, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Antypa M, Faux C, Eichele G, Parnavelas JG, Andrews WD. Differential gene expression in migratory streams of cortical interneurons. Eur J Neurosci 2012; 34:1584-94. [PMID: 22103416 PMCID: PMC3401901 DOI: 10.1111/j.1460-9568.2011.07896.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cortical interneurons originate in the ganglionic eminences of the subpallium and migrate into the cortex in well-defined tangential streams. At the start of corticogenesis, two streams of migrating neurons are evident: a superficial one at the level of the preplate (PPL), and a deeper one at the level of the intermediate zone (IZ). Currently, little is known about the signalling mechanisms that regulate interneuron migration, and almost nothing is known about the molecules that may be involved in their choice of migratory stream. Here, we performed a microarray analysis, comparing the changes in gene expression between cells migrating in the PPL and those migrating in the IZ at embryonic day 13.5. This analysis identified genes, many of them novel, that were upregulated in one of the two streams. Moreover, polymerase chain reaction, in situ hybridization experiments and immunohistochemistry showed the expression of these genes in interneurons migrating within the PPL or IZ, suggesting that they play a role in their migration and choice of stream.
Collapse
Affiliation(s)
- Mary Antypa
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | |
Collapse
|
20
|
Palmitoylation of stathmin family proteins domain A controls Golgi versus mitochondrial subcellular targeting. Biol Cell 2012; 100:577-89. [DOI: 10.1042/bc20070119] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
21
|
Ip AKC, Tso PH, Lee MMK, Wong YH. Elevated expression of RGS19 impairs the responsiveness of stress-activated protein kinases to serum. Mol Cell Biochem 2011; 362:159-68. [PMID: 22045062 DOI: 10.1007/s11010-011-1138-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 10/22/2011] [Indexed: 12/17/2022]
Abstract
Regulators of G protein signaling (RGS proteins) serve as GTPase activating proteins for the signal transducing Gα subunits. RGS19, also known as Gα-interacting protein (GAIP), has been shown to subserve other functions such as the regulation of macroautophagy and growth factor signaling. We have recently demonstrated that the expression of RGS19 in human embryonic kidney (HEK) 293 cells resulted in the disruption of serum-induced mitogenic response along the classical Ras/Raf/MEK/ERK pathway. Here, we further examined the effect of RGS19 expression on the stress-activated protein kinases (SAPKs). Both c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) became non-responsive to serum in 293/RGS19 cells, yet the two SAPKs responded to UV irradiation or osmotic stress induced by sorbitol. Kinases upstream of JNK and p38 MAPK, including MKK3/6, MKK4, and MLK3, also failed to respond to serum stimulation in 293/RGS19 cells. Serum-induced activation of the small GTPases Rac1 and Cdc42 was similarly suppressed in these cells. Our results indicate that elevated expression of RGS19 can severely disrupt the regulation of MAPKs by small GTPases.
Collapse
Affiliation(s)
- Angel K C Ip
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | | | | | | |
Collapse
|
22
|
RGS19 stimulates cell proliferation by deregulating cell cycle control and enhancing Akt signaling. Cancer Lett 2011; 309:199-208. [DOI: 10.1016/j.canlet.2011.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 05/09/2011] [Accepted: 06/01/2011] [Indexed: 11/13/2022]
|
23
|
Huang J, Yang J, Maity B, Mayuzumi D, Fisher RA. Regulator of G protein signaling 6 mediates doxorubicin-induced ATM and p53 activation by a reactive oxygen species-dependent mechanism. Cancer Res 2011; 71:6310-9. [PMID: 21859827 DOI: 10.1158/0008-5472.can-10-3397] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Doxorubicin (DXR), among the most widely used cancer chemotherapy agents, promotes cancer cell death via activation of ataxia telangiectasia mutated (ATM) and the resultant upregulation of tumor suppressor p53. The exact mechanism by which DXR activates ATM is not fully understood. Here, we discovered a novel role for regulator of G protein signaling 6 (RGS6) in mediating activation of ATM and p53 by DXR. RGS6 was robustly induced by DXR, and genetic loss of RGS6 dramatically impaired DXR-induced activation of ATM and p53, as well as its in vivo apoptotic actions in heart. The ability of RGS6 to promote p53 activation in response to DXR was independent of RGS6 interaction with G proteins but required ATM. RGS6 mediated activation of ATM and p53 by DXR via a reactive oxygen species (ROS)-dependent and DNA damage-independent mechanism. This mechanism represents the primary means by which DXR promotes activation of the ATM-p53 apoptosis pathway that underlies its cytotoxic activity. Our findings contradict the canonical theories that DXR activates ATM primarily by promoting DNA damage either directly or indirectly (via ROS) and that RGS6 function is mediated by its interactions with G proteins. These findings reveal a new mechanism for the chemotherapeutic actions of DXR and identify RGS6 as a novel target for cancer chemotherapy.
Collapse
Affiliation(s)
- Jie Huang
- Department of Pharmacology, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|
24
|
Sibbel SP, Talbert ME, Bowden DW, Haffner SM, Taylor KD, Chen YDI, Wagenknecht LE, Langefeld CD, Norris JM. RGS6 variants are associated with dietary fat intake in Hispanics: the IRAS Family Study. Obesity (Silver Spring) 2011; 19:1433-8. [PMID: 21233807 PMCID: PMC3683650 DOI: 10.1038/oby.2010.333] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recently, a genome-wide association scan was completed in the IRAS (Insulin Resistance Atherosclerosis Study) Family Study (IRASFS) Hispanic-American cohort. Multiple single-nucleotide polymorphisms (SNPs) in the G-protein signaling 6 (RGS6) gene were found to be associated with adiposity phenotypes. RGS6 has shown downstream antagonistic interplay with opioid receptors, targets of fatty/sugary food agonists. The possibility that RGS6 promotes tolerance and tachyphylaxis among the opioid receptor is a plausible pathway for overconsuming fat/sugar-laden food. Therefore, we hypothesized that RGS6 variants are associated with intake of fatty/sugary foods. In 932 Hispanics from San Antonio and San Luis Valley, CO, the following dietary intake variables were assessed using the Block Brief 2000 food frequency questionnaire: total calories, total fat, % calories from fat, % calories from saturated fat, protein, % calories from protein, carbohydrates, % calories from carbohydrates, and daily frequency of servings of fats/oils/sweets. We tested for association between 23 SNPs in RGS6 and dietary intake using a variance components measured genotype approach. All models were adjusted for gender, recruitment site, admixture, BMI, and age. Using an additive genetic model, rs1402064 was associated with higher intake of fats/oils/sweets, total calories, total fat and saturated fat (P = 0.0007, 0.026, 0.023, and 0.024). SNPs rs847330 and rs847354 were associated with higher intake of fats/oils/sweets (P = 0.002 and 0.018), total fat (P = 0.040 and 0.048) and saturated fat (P = 0.044 and 0.041). Finally, rs769148 was associated with higher intake of fats/oils/sweets (P = 0.002). RGS6 is a new candidate gene for adiposity traits that may be associated with a behavioral tendency toward fat-laden food intake.
Collapse
Affiliation(s)
- Scott P. Sibbel
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado, USA
| | - Matthew E. Talbert
- Department of Biochemistry, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Donald W. Bowden
- Department of Biochemistry, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Steve M. Haffner
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Kent D. Taylor
- Medical Genetics Institute, Burns and Allen Cedars-Sinai Research Institute, Los Angeles, California, USA
| | - Yii-Der I. Chen
- Medical Genetics Institute, Burns and Allen Cedars-Sinai Research Institute, Los Angeles, California, USA
| | - Lynne E. Wagenknecht
- Department of Epidemiology, Division of Public Health Sciences, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Carl D. Langefeld
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest University, Winston-Salem, North Carolina, USA.
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
25
|
Sharma M, Celver J, Kovoor A. Regulator of G protein signaling 9-2 (RGS9-2) mRNA is up regulated during neuronal differentiation of mouse embryonic stem cells. Neurosci Lett 2011; 502:123-8. [PMID: 21616123 DOI: 10.1016/j.neulet.2011.05.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 05/05/2011] [Accepted: 05/09/2011] [Indexed: 01/12/2023]
Abstract
In this study we demonstrate up-regulation of mRNA for Regulator of G protein Signaling (RGS) 6, 7, 9 and 11, R7 family RGS binding protein (R7BP) and RGS9 anchor protein (R9AP) during neuronal differentiation of mouse embryonic stem cells (mESCs). This expression pattern was most robust for RGS9 whose transcript level was low in undifferentiated mESCs but increased over 125 fold when differentiating mESCs began to exhibit a neuronal precursor cell (NPC) phenotype. In addition, we demonstrate that RGS9 mRNA is expressed in neuronal stem cells isolated from embryonic mouse cortex. The expression of RGS9 in two distinct populations of NPCs suggests that RGS9 and its accessory proteins may play an important role in neuron development.
Collapse
Affiliation(s)
- Meenakshi Sharma
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | | | | |
Collapse
|
26
|
Maity B, Yang J, Huang J, Askeland RW, Bera S, Fisher RA. Regulator of G protein signaling 6 (RGS6) induces apoptosis via a mitochondrial-dependent pathway not involving its GTPase-activating protein activity. J Biol Chem 2010; 286:1409-19. [PMID: 21041304 DOI: 10.1074/jbc.m110.186700] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Regulator of G protein signaling 6 (RGS6) is a member of a family of proteins called RGS proteins, which function as GTPase-activating proteins (GAPs) for Gα subunits. Given the role of RGS6 as a G protein GAP, the link between G protein activation and cancer, and a reduction of cancer risk in humans expressing a RGS6 SNP leading to its increased translation, we hypothesized that RGS6 might function to inhibit growth of cancer cells. Here, we show a marked down-regulation of RGS6 in human mammary ductal epithelial cells that correlates with the progression of their transformation. RGS6 exhibited impressive antiproliferative actions in breast cancer cells, including inhibition of cell growth and colony formation and induction of cell cycle arrest and apoptosis by mechanisms independent of p53. RGS6 activated the intrinsic pathway of apoptosis involving regulation of Bax/Bcl-2, mitochondrial outer membrane permeabilization (MOMP), cytochrome c release, activation of caspases-3 and -9, and poly(ADP-ribose) polymerase cleavage. RGS6 promoted loss of mitochondrial membrane potential (ΔΨ(m)) and increases in reactive oxygen species (ROS). RGS6-induced caspase activation and loss of ΔΨ(m) was mediated by ROS, suggesting an amplification loop in which ROS provided a feed forward signal to induce MOMP, caspase activation, and cell death. Loss of RGS6 in mouse embryonic fibroblasts dramatically impaired doxorubicin-induced growth suppression and apoptosis. Surprisingly, RGS6-induced apoptosis in both breast cancer cells and mouse embryonic fibroblasts does not require its GAP activity toward G proteins. This work demonstrates a novel signaling action of RGS6 in cell death pathways and identifies it as a possible therapeutic target for treatment of breast cancer.
Collapse
Affiliation(s)
- Biswanath Maity
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | |
Collapse
|
27
|
RGS19 enhances cell proliferation through its C-terminal PDZ motif. Cell Signal 2010; 22:1700-7. [PMID: 20599498 DOI: 10.1016/j.cellsig.2010.06.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 06/24/2010] [Indexed: 01/18/2023]
Abstract
Regulator of G protein signaling 19 (RGS19), also known as Galpha-interacting protein (GAIP), is a GTPase activating protein (GAP) for Galpha(i) subunits. Apart from its GAP function, RGS19 has been implicated in growth factor signaling through binding to GAIP-interacting protein C-terminus (GIPC) via its C-terminal PDZ-binding motif. To gain additional insight on its function, we have stably expressed RGS19 in a number of mammalian cell lines and examined its effect on cell proliferation. Interestingly, overexpression of RGS19 stimulated the growth of HEK293, PC12, Caco2, and NIH3T3 cells. This growth promoting effect was not shared by other RGS proteins including RGS4, RGS10 and RGS20. Despite its ability to stimulate cell proliferation, RGS19 failed to induce neoplastic transformation in NIH3T3 cells as determined by focus formation and soft-agar assays, and it did not induce tumor growth in athymic nude mice. Deletion mutants of RGS19 lacking the PDZ-binding motif failed to complex with GIPC and did not exhibit any growth promoting effect. Overexpression of GIPC alone in HEK293 cells stimulated cell proliferation whereas its knockdown in H1299 non-small cell lung carcinomas suppressed cell proliferation. This study demonstrates that RGS19, in addition to acting as a GAP, is able to stimulate cell proliferation in a GIPC-dependent manner.
Collapse
|
28
|
McCoy KL, Hepler JR. Regulators of G protein signaling proteins as central components of G protein-coupled receptor signaling complexes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 86:49-74. [PMID: 20374713 DOI: 10.1016/s1877-1173(09)86003-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The regulators of G protein signaling (RGS) proteins bind directly to G protein alpha (Gα) subunits to regulate the signaling functions of Gα and their linked G protein-coupled receptors (GPCRs). Recent studies indicate that RGS proteins also interact with GPCRs, not just G proteins, to form preferred functional pairs. Interactions between GPCRs and RGS proteins may be direct or indirect (via a linker protein) and are dictated by the receptors, rather than the linked G proteins. Emerging models suggest that GPCRs serve as platforms for assembling an overlapping and distinct constellation of signaling proteins that perform receptor-specific signaling tasks. Compelling evidence now indicates that RGS proteins are central components of these GPCR signaling complexes. This review will outline recent discoveries of GPCR/RGS pairs as well as new data in support of the idea that GPCRs serve as platforms for the formation of multiprotein signaling complexes.
Collapse
Affiliation(s)
- Kelly L McCoy
- Department of Pharmacology, G205 Rollins Research Center, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
29
|
Slepak VZ. Structure, function, and localization of Gβ5-RGS complexes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 86:157-203. [PMID: 20374716 DOI: 10.1016/s1877-1173(09)86006-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Members of the R7 subfamily of regulator of G protein signaling (RGS) proteins (RGS6, 7, 9, and 11) exist as heterodimers with the G protein beta subunit Gβ5. These protein complexes are only found in neurons and are defined by the presence of three domains: DEP/DHEX, Gβ5/GGL, and RGS. This article summarizes published work in the following areas: (1) the functional significance of structural organization of Gβ5-R7 complexes, (2) regional distribution of Gβ5-R7 in the nervous system and regulation of R7 family expression, (3) subcellular localization of Gβ5-R7 complexes, and (4) novel binding partners of Gβ5-R7 proteins. The review points out some contradictions between observations made by different research groups and highlights the importance of using alternative experimental approaches to obtain conclusive information about Gβ5-R7 function in vivo.
Collapse
Affiliation(s)
- Vladlen Z Slepak
- Department of Molecular and Cellular Pharmacology and the Neuroscience Program, University of Miami School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
30
|
The R7 RGS protein family: multi-subunit regulators of neuronal G protein signaling. Cell Biochem Biophys 2009; 54:33-46. [PMID: 19521673 DOI: 10.1007/s12013-009-9052-9] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 05/27/2009] [Indexed: 01/09/2023]
Abstract
G protein-coupled receptor signaling pathways mediate the transmission of signals from the extracellular environment to the generation of cellular responses, a process that is critically important for neurons and neurotransmitter action. The ability to promptly respond to rapidly changing stimulation requires timely inactivation of G proteins, a process controlled by a family of specialized proteins known as regulators of G protein signaling (RGS). The R7 group of RGS proteins (R7 RGS) has received special attention due to their pivotal roles in the regulation of a range of crucial neuronal processes such as vision, motor control, reward behavior, and nociception in mammals. Four proteins in this group, RGS6, RGS7, RGS9, and RGS11, share a common molecular organization of three modules: (i) the catalytic RGS domain, (ii) a GGL domain that recruits G beta(5), an outlying member of the G protein beta subunit family, and (iii) a DEP/DHEX domain that mediates interactions with the membrane anchor proteins R7BP and R9AP. As heterotrimeric complexes, R7 RGS proteins not only associate with and regulate a number of G protein signaling pathway components, but have also been found to form complexes with proteins that are not traditionally associated with G protein signaling. This review summarizes our current understanding of the biology of the R7 RGS complexes including their structure/functional organization, protein-protein interactions, and physiological roles.
Collapse
|
31
|
Huang J, Fisher RA. Chapter 5 Nuclear Trafficking of Regulator of G Protein Signaling Proteins and Their Roles in the Nucleus. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 86:115-56. [DOI: 10.1016/s1877-1173(09)86005-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
32
|
Tso PH, Morris CJ, Yung LY, Ip NY, Wong YH. Multiple Gi Proteins Participate in Nerve Growth Factor-Induced Activation of c-Jun N-terminal Kinases in PC12 Cells. Neurochem Res 2008; 34:1101-12. [DOI: 10.1007/s11064-008-9880-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2008] [Indexed: 01/21/2023]
|
33
|
Yung LY, Tso PH, Wu EH, Yu JC, Ip NY, Wong YH. Nerve growth factor-induced stimulation of p38 mitogen-activated protein kinase in PC12 cells is partially mediated via Gi/o proteins. Cell Signal 2008; 20:1538-44. [DOI: 10.1016/j.cellsig.2008.04.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 04/10/2008] [Accepted: 04/11/2008] [Indexed: 12/21/2022]
|
34
|
Mahapatra NR, Taupenot L, Courel M, Mahata SK, O'Connor DT. The trans-Golgi proteins SCLIP and SCG10 interact with chromogranin A to regulate neuroendocrine secretion. Biochemistry 2008; 47:7167-78. [PMID: 18549247 DOI: 10.1021/bi7019996] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Secretion of proteins and peptides from eukaryotic cells takes place by both constitutive and regulated pathways. Regulated secretion may involve interplay of proteins that are currently unknown. Recent studies suggest an important role of chromogranin A (CHGA) in the regulated secretory pathway in neuroendocrine cells, but the mechanism by which CHGA enters the regulated pathway, or even triggers the formation of the pathway, remains unclear. In this study, we used a transcriptome/proteome-wide approach, to discover binding partners for CHGA, by employing a phage display cDNA library method. Several proteins within or adjacent to the secretory pathway were initially detected as binding partners of recombinant human CHGA. We then focused on the trans-Golgi protein SCLIP (STMN3) and its stathmin paralog SCG10 (STMN2) for functional study. Co-immunoprecipitation experiments confirmed the interaction of each of these two proteins with CHGA in vitro. SCLIP and SCG10 were colocalized to the Golgi apparatus of chromaffin cells in vivo and shared localization with CHGA as it transited the Golgi. Downregulation of either SCLIP or SCG10 by synthetic siRNAs virtually abolished chromaffin cell secretion of a transfected CHGA-EAP chimera (expressing CHGA fused to an enzymatic reporter, and trafficked to the regulated pathway). SCLIP siRNA also decreased the level of secretion of endogenous CHGA and SCG2, as well as transfected human growth hormone, while SCG10 siRNA decreased the level of regulated secretion of endogenous CHGB. Moreover, a dominant negative mutant of SCG10 (Cys 22,Cys 24-->Ala 22,Ala 24) significantly blocked secretion of the transfected CHGA-EAP chimera. A decrease in the buoyant density of chromaffin granules was observed after downregulation of SCG10 by siRNA, suggesting participation of these stathmins in granule formation or maturation. We conclude that SCLIP and SCG10 interact with CHGA, share partial colocalization in the Golgi apparatus, and may be necessary for typical transmitter storage and release from chromaffin cells.
Collapse
Affiliation(s)
- Nitish R Mahapatra
- Department of Medicine, Center for Human Genetics and Genomics, University of California at San Diego, La Jolla, California 92093-0838, USA.
| | | | | | | | | |
Collapse
|
35
|
Yau DM, Sethakorn N, Taurin S, Kregel S, Sandbo N, Camoretti-Mercado B, Sperling AI, Dulin NO. Regulation of Smad-mediated gene transcription by RGS3. Mol Pharmacol 2008; 73:1356-61. [PMID: 18287247 PMCID: PMC3329871 DOI: 10.1124/mol.108.044990] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Regulator of G protein signaling (RGS) proteins are united into a family by the presence of the homologous RGS domain that binds the alpha subunits of heterotrimeric G proteins and accelerates their GTPase activity. A member of this family, RGS3 regulates the signaling mediated by G(q) and G(i) proteins by binding the corresponding Galpha subunits. Here we show that RGS3 interacts with the novel partners Smad2, Smad3, and Smad4-the transcription factors that are activated through a transforming growth factor-beta (TGF-beta) receptor signaling. This interaction is mediated by the region of RGS3 outside of the RGS domain and by Smad's Mad homology 2 domain. Overexpression of RGS3 results in inhibition of Smad-mediated gene transcription. RGS3 does not affect TGF-beta-induced Smad phosphorylation, but it prevents heteromerization of Smad3 with Smad4, which is required for transcriptional activity of Smads. This translates to functional inhibition of TGF-beta-induced myofibroblast differentiation by RGS3. In conclusion, this study identifies a novel, noncanonical role of RGS3 in regulation of TGF-beta signaling through its interaction with Smads and interfering with Smad heteromerization.
Collapse
Affiliation(s)
- Douglas M Yau
- Section of Pulmonary and Critical Care Medicine, the University of Chicago Department of Medicine, 5841 S. Maryland Ave, MC 6076, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Baldassa S, Gnesutta N, Fascio U, Sturani E, Zippel R. SCLIP, a Microtubule-destabilizing Factor, Interacts with RasGRF1 and Inhibits Its Ability to Promote Rac Activation and Neurite Outgrowth. J Biol Chem 2007; 282:2333-45. [PMID: 17135267 DOI: 10.1074/jbc.m604495200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RasGRF1 is a neuron-specific guanine nucleotide exchange factor for the small GTPases Ras and Rac. It is implicated in the regulation of memory formation and in the development of tolerance to drug abuse, although the mechanisms have been elucidated only in part. Here we report the isolation, by the yeast two-hybrid screen, of the microtubule-destabilizing factor SCLIP (SCG10-like protein) as a novel RasGRF1-interacting protein. This interaction requires the region spanning the Dbl-homology domain of RasGRF1, endowed with catalytic activity on Rac. In search for a possible function we found by biochemical means that SCLIP influences the signaling properties of RasGRF1, greatly reducing its ability to activate the Rac/p38 MAPK pathway, while the Ras/Erk one remains unaffected. Moreover, a potential role is suggested by transfection studies in neuronal PC12 cells in which RasGRF1 induces neurite outgrowth, and coexpression of SCLIP counteracts this effect, causing a dramatic decrease in the percentage of cells bearing neurites, which also appear significantly shortened. This study unveils a physical and functional interaction between RasGRF1 and SCLIP. We suggest that this novel interplay may have possible implications in mechanisms that regulate neuronal morphology and structural plasticity.
Collapse
Affiliation(s)
- Simona Baldassa
- Department of Biomolecular Sciences and Biotechnology, University of Milan, Italy.
| | | | | | | | | |
Collapse
|
37
|
Bogoch Y, Biala YN, Linial M, Weinstock M. Anxiety induced by prenatal stress is associated with suppression of hippocampal genes involved in synaptic function. J Neurochem 2006; 101:1018-30. [PMID: 17254015 DOI: 10.1111/j.1471-4159.2006.04402.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Exposure of pregnant women or animals to stress during a critical period of foetal brain development increases the likelihood of anxiety, depression and learning deficits that are associated with structural alterations in the offspring hippocampus. In this study, we report the effect of gestational stress in rats on anxiogenic behaviour and hippocampal gene expression of their 23-day-old female offspring. As the rat brain continues to develop after birth, we also used the procedure of handling (H) during the first 10 days of life to reverse the anxiogenic behaviour of prenatally stressed (PS) rats. By means of micro-array analysis on hippocampal extracts, we found that the expression of about 6.1% of 9505 valid genes was significantly altered by prenatal stress (p<0.05). Of these, 48% were over-expressed and 52% under-expressed. The latter included approximately 300 genes that participate in axonal growth, regulation of ion channels and transporters, trafficking of synaptic vesicles and neurotransmitter release. About 30% of the genes that were down-regulated in PS rats were restored to control levels by H. These include genes that play a role in pre-synaptic organization and function. Our results provide a possible relationship between hippocampal gene expression and changes in behaviour resulting from prenatal stress.
Collapse
Affiliation(s)
- Y Bogoch
- Department of Biological Chemistry, Life Science Institute, The Hebrew University, Jerusalem, Israel
| | | | | | | |
Collapse
|
38
|
Poulain FE, Sobel A. The "SCG10-LIke Protein" SCLIP is a novel regulator of axonal branching in hippocampal neurons, unlike SCG10. Mol Cell Neurosci 2006; 34:137-46. [PMID: 17145186 DOI: 10.1016/j.mcn.2006.10.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Revised: 10/11/2006] [Accepted: 10/17/2006] [Indexed: 02/03/2023] Open
Abstract
Stathmin family phosphoproteins participate in the control of microtubule dynamics and have been proposed to be involved in the control of neuronal differentiation. We examined the biological properties and functions of SCLIP, the stathmin family protein most similar to SCG10, a widely studied and recognized neuronal differentiation marker. SCLIP, like SCG10, is present from the earliest stages of hippocampal neuron differentiation in culture at vesicle-like structures following dynamic microtubules. Its inhibition by RNA interference resulted in increased axonal branching, revealing a novel biological role for SCLIP, distinct from SCG10 whose down-regulation in the same conditions promoted growth cone expansion. The enhanced axonal branching resulted from the formation of collateral lamellar protrusions, with cytoskeleton reorganization typical of normal branching. In addition to revealing a novel function for SCLIP in axonal morphogenesis, our results demonstrate for the first time that stathmin family proteins fulfill different and complementary roles during neuronal differentiation.
Collapse
Affiliation(s)
- Fabienne E Poulain
- INSERM, U706, and Université Pierre et Marie Curie-Paris 6, Institut du Fer à Moulin, 17 rue du Fer-à-Moulin, Paris F-75005, France
| | | |
Collapse
|
39
|
Abstract
Regulators of G-protein signalling (RGS) proteins are a large and diverse family initially identified as GTPase activating proteins (GAPs) of heterotrimeric G-protein Galpha-subunits. At least some can also influence Galpha activity through either effector antagonism or by acting as guanine nucleotide dissociation inhibitors (GDIs). As our understanding of RGS protein structure and function has developed, so has the realisation that they play roles beyond G-protein regulation. Such diversity of function is enabled by the variety of RGS protein structure and their ability to interact with other cellular molecules including phospholipids, receptors, effectors and scaffolds. The activity, sub-cellular distribution and expression levels of RGS proteins are dynamically regulated, providing a layer of complexity that has yet to be fully elucidated.
Collapse
Affiliation(s)
- Gary B Willars
- Department of Cell Physiology and Pharmacology, University of Leicester, University Road, Leicester LE1 9HN, UK.
| |
Collapse
|
40
|
Heo K, Ha SH, Chae YC, Lee S, Oh YS, Kim YH, Kim SH, Kim JH, Mizoguchi A, Itoh TJ, Kwon HM, Ryu SH, Suh PG. RGS2 promotes formation of neurites by stimulating microtubule polymerization. Cell Signal 2006; 18:2182-92. [PMID: 16820281 DOI: 10.1016/j.cellsig.2006.05.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Accepted: 05/11/2006] [Indexed: 11/22/2022]
Abstract
Regulator of G-protein signaling (RGS) proteins interact with alpha subunits of heterotrimeric G-proteins via the RGS domain and attenuate their activity by accelerating GTPase activity. RGS2, a member of the RGS family, regulates synaptic development via hereto unknown mechanism. In this study, we found that RGS2 directly interacted with tubulin via a short region at the N-terminus: amino acids 41-60. RGS2 enhanced microtubule polymerization in vitro, and the tubulin binding region was necessary and sufficient for this activity. In Vero cells, polymerization of microtubule was stimulated when peptides containing the tubulin binding region were microinjected. Immunocytochemical analysis showed that endogenous RGS2 was localized at the termini of neurites in differentiated PC12 cells. Over-expression of RGS2 enhanced the nerve growth factor-induced neurite outgrowth in PC12 cells, while specific knock-down of endogenous RGS2 suppressed the neurite outgrowth. These findings demonstrate that RGS2 contributes to the neuronal cell differentiation via regulation of microtubule dynamics.
Collapse
Affiliation(s)
- Kyun Heo
- Division of Molecular and Life Science, Pohang University of Science and Technology, Pohang, Kyungbuk 790-784, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Cai Y, Wu P, Ozen M, Yu Y, Wang J, Ittmann M, Liu M. Gene expression profiling and analysis of signaling pathways involved in priming and differentiation of human neural stem cells. Neuroscience 2006; 138:133-48. [PMID: 16414199 DOI: 10.1016/j.neuroscience.2005.11.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Revised: 11/08/2005] [Accepted: 11/08/2005] [Indexed: 01/02/2023]
Abstract
Human neural stem cells have the ability to differentiate into all three major cell types in the CNS including neurons, astrocytes and oligodendrocytes. The multipotency of human neural stem cells shed a light on the possibility of using stem cells as a therapeutic tool for various neurological disorders including neurodegenerative diseases and neurotrauma that involve a loss of functional neurons. We have discovered previously a priming procedure to direct primarily cultured human neural stem cells to differentiate into almost pure neurons when grafted into adult CNS. However, the molecular mechanism underlying this phenomenon is still unknown. To unravel transcriptional changes of human neural stem cells upon priming, cDNA microarray was used to study temporal changes in human neural stem cell gene expression profile during priming and differentiation. As a result, transcriptional levels of 520 annotated genes were detected changed in at least at two time points during the priming process. In addition, transcription levels of more than 3000 hypothetical protein encoding genes and EST genes were modulated during the priming and differentiation processes of human neural stem cells. We further analyzed the named genes and grouped them into 14 functional categories. Of particular interest, key cell signal transduction pathways, including the G-protein-mediated signaling pathways (heterotrimeric and small monomeric GTPase pathways), the Wnt signaling pathway and the TGF-beta pathway, are modulated by the neural stem cell priming, suggesting important roles of these key signaling pathways in priming and differentiation of human neural stem cells.
Collapse
Affiliation(s)
- Y Cai
- Alkek Institute of Biosciences and Technology, Department of Medical Biochemistry and Genetics, Texas A&M University System Health Science Center, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Nakazawa T, Morii H, Tamai M, Mori N. Selective upregulation of RB3/stathmin4 by ciliary neurotrophic factor following optic nerve axotomy. Brain Res 2005; 1061:97-106. [PMID: 16256088 DOI: 10.1016/j.brainres.2005.08.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Revised: 08/22/2005] [Accepted: 08/28/2005] [Indexed: 11/16/2022]
Abstract
In this study, we examined the cellular responses of stathmin-related proteins in the rat retina following optic nerve (ON) axotomy. To examine the distribution of stathmin-related gene products, we performed semi-quantitative reverse transcription polymerase chain reaction (RT-PCR), in situ hybridization (ISH) and immunohistochemical analyses. Retrograde labeling using a fluorescein tracer, fluorogold (FG), was used for the identification of retinal ganglion cells (RGCs). RT-PCR and ISH analyses indicated that the expression of RB3 was specifically increased in the ganglion cell layer (GCL) comparing to other members of stathmin-related gene family examined 3 days following the ON axotomy. When brain-derived neurotrophic factor was administrated intravitreously, the induction of RB3 mRNA sustained up to 7 days after axotomy, although the peak induction level was unchanged. In contrast, ciliary neurotrophic factor (CNTF) administration increased the peak level of RB3 mRNA induction significantly at 3 days after axotomy. Immunohistochemistry in combination with the retrograde labeling of axotomized cells by FG revealed that RB3 was increased following axotomy in FG-labeled RGCs. These data suggest that RB3 is the unique response protein in the stathmin-related proteins following ON axotomy and the induced RB3 may play a critical role in the CNTF-induced response on the axotomized RGCs, e.g. axonal regeneration and/or neuroprotection.
Collapse
Affiliation(s)
- Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | | | | | | |
Collapse
|
43
|
Oh JE, Karlmark KR, Shin JH, Pollak A, Freilinger A, Hengstschläger M, Lubec G. Differentiation of neuroblastoma cell line N1E-115 involves several signaling cascades. Neurochem Res 2005; 30:333-48. [PMID: 16018577 DOI: 10.1007/s11064-005-2607-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
No systematic searches for differential expression of signaling proteins (SP) in undifferentiated vs. differentiated cell lineages were published and herein we used protein profiling for this purpose. The NIE-115 cell line was cultivated and an aliquot was differentiated with dimethylsulfoxide (DMSO), that is known to lead to a neuronal phenotype. Cell lysates were prepared, run on two-dimensional gel electrophoresis followed by MALDI-TOF-TOF identification of proteins and maps of identified SPs were generated. Seven SPs were comparable, 27 SPs: GTP-binding/Ras-related proteins, kinases, growth factors, calcium binding proteins, phosphatase-related proteins were observed in differentiated NIE-115 cells and eight SPs of the groups mentioned above were observed in undifferentiated cells only. Switching-on/off of several individual SPs from different signaling cascades during the differentiation process is a key to understand mechanisms involved. The findings reported herein are challenging in vitro and in vivo studies to confirm a functional role for deranged SPs.
Collapse
Affiliation(s)
- Ji-eun Oh
- Department of Pediatrics, Medical University of Vienna, Währinger Gürtel 18, A 1090, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
44
|
Charbaut E, Chauvin S, Enslen H, Zamaroczy S, Sobel A. Two separate motifs cooperate to target stathmin-related proteins to the Golgi complex. J Cell Sci 2005; 118:2313-23. [PMID: 15870110 DOI: 10.1242/jcs.02349] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The appropriate targeting of membrane-associated proteins involves a diversity of motifs including post-translational modifications and specific protein sequences. Phosphoproteins of the stathmin family are important regulators of microtubule dynamics, in particular in the developing and mature nervous system. Whereas stathmin is cytosolic, SCG10, SCLIP and the splice variants RB3/RB3′/RB3″ are associated with Golgi and vesicular membranes, through their palmitoylated N-terminal A domains. In order to identify essential motifs involved in this specific targeting, we examined the subcellular distribution of various subdomains derived from domain A of SCG10 fused with GFP. We show that the Golgi localization of SCG10 results from the cooperation of two motifs: a membrane-anchoring palmitoylation motif and a newly identified Golgi-specifying sequence. The latter displayed no targeting activity by itself, but retained a Golgi-specifying activity when associated with another membrane-anchoring palmitoylation motif derived from the protein GAP-43. We further identified critical residues for the specific Golgi targeting of domain A. Altogether, our results give new insight into the regulation of the subcellular localization of stathmin family proteins, an important feature of their physiological functions in differentiating and mature neural cells. More generally we provide new information on essential mechanisms of functional protein subcellular targeting.
Collapse
Affiliation(s)
- Elodie Charbaut
- INSERM, U706, Neurosignalisation Moléculaire et Cellulaire, Institut du Fer à Moulin, 17 rue du Fer à Moulin, and Université Pierre et Marie Curie (UPMC), 4 place Jussieu, Paris, 75005 France
| | | | | | | | | |
Collapse
|
45
|
Reis EM, Ojopi EPB, Alberto FL, Rahal P, Tsukumo F, Mancini UM, Guimarães GS, Thompson GMA, Camacho C, Miracca E, Carvalho AL, Machado AA, Paquola ACM, Cerutti JM, da Silva AM, Pereira GG, Valentini SR, Nagai MA, Kowalski LP, Verjovski-Almeida S, Tajara EH, Dias-Neto E, Bengtson MH, Canevari RA, Carazzolle MF, Colin C, Costa FF, Costa MCR, Estécio MRH, Esteves LICV, Federico MHH, Guimarães PEM, Hackel C, Kimura ET, Leoni SG, Maciel RMB, Maistro S, Mangone FRR, Massirer KB, Matsuo SE, Nobrega FG, Nóbrega MP, Nunes DN, Nunes F, Pandolfi JR, Pardini MIMC, Pasini FS, Peres T, Rainho CA, dos Reis PP, Rodrigus-Lisoni FCC, Rogatto SR, dos Santos A, dos Santos PCC, Sogayar MC, Zanelli CF. Large-scale Transcriptome Analyses Reveal New Genetic Marker Candidates of Head, Neck, and Thyroid Cancer. Cancer Res 2005; 65:1693-9. [PMID: 15753364 DOI: 10.1158/0008-5472.can-04-3506] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A detailed genome mapping analysis of 213,636 expressed sequence tags (EST) derived from nontumor and tumor tissues of the oral cavity, larynx, pharynx, and thyroid was done. Transcripts matching known human genes were identified; potential new splice variants were flagged and subjected to manual curation, pointing to 788 putatively new alternative splicing isoforms, the majority (75%) being insertion events. A subset of 34 new splicing isoforms (5% of 788 events) was selected and 23 (68%) were confirmed by reverse transcription-PCR and DNA sequencing. Putative new genes were revealed, including six transcripts mapped to well-studied chromosomes such as 22, as well as transcripts that mapped to 253 intergenic regions. In addition, 2,251 noncoding intronic RNAs, eventually involved in transcriptional regulation, were found. A set of 250 candidate markers for loss of heterozygosis or gene amplification was selected by identifying transcripts that mapped to genomic regions previously known to be frequently amplified or deleted in head, neck, and thyroid tumors. Three of these markers were evaluated by quantitative reverse transcription-PCR in an independent set of individual samples. Along with detailed clinical data about tumor origin, the information reported here is now publicly available on a dedicated Web site as a resource for further biological investigation. This first in silico reconstruction of the head, neck, and thyroid transcriptomes points to a wealth of new candidate markers that can be used for future studies on the molecular basis of these tumors. Similar analysis is warranted for a number of other tumors for which large EST data sets are available.
Collapse
Affiliation(s)
- Eduardo M Reis
- Departamento de Bioquímica, Faculdade de Medicina, Universidade de São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Martin-McCaffrey L, Willard FS, Pajak A, Dagnino L, Siderovski DP, D'Souza SJA. Analysis of interactions between regulator of G-protein signaling-14 and microtubules. Methods Enzymol 2005; 390:240-58. [PMID: 15488182 DOI: 10.1016/s0076-6879(04)90016-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Microtubules are dynamic polymers essential for mitosis and cell division, intracellular transport, and maintaining cell organization and structure. Microtubule dynamics are tightly controlled in a context-specific manner by a myriad of microtubule-associated proteins. We have identified regulator of G-protein signaling-14 (RGS14) as a microtubule-associated protein. RGS14 is a component of the mitotic apparatus that binds directly to and stabilizes microtubules in vitro and is essential for the first cell division in the mouse embryo. This article describes methods used for examining the impact of the RGS14/microtubule interaction in vivo and in vitro.
Collapse
|
47
|
Berman DM, Wang Y, Liu Z, Dong Q, Burke LA, Liotta LA, Fisher R, Wu X. A functional polymorphism in RGS6 modulates the risk of bladder cancer. Cancer Res 2004; 64:6820-6. [PMID: 15375002 DOI: 10.1158/0008-5472.can-04-1916] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RGS proteins negatively regulate heterotrimeric G protein signaling. Recent reports have shown that RGS proteins modulate neuronal, cardiovascular, and lymphocytic activity, yet their role in carcinogenesis has not been explored. In an epidemiologic study of 477 bladder cancer patients and 446 matched controls, three noncoding single-nucleotide polymorphisms (SNPs) in RGS2 and RGS6 were each associated with a statistically significant reduction in bladder cancer risk. The risk of bladder cancer was reduced by 74% in those individuals with the variant genotype at all three SNPs (odds ratio, 0.26; 95% confidence interval, 0.09-0.71). When the SNPs were analyzed separately, the RGS6-rs2074647 (C-->T) polymorphism conferred the greatest overall reduction in risk of bladder cancer (odds ratio, 0.66; 95% confidence interval, 0.46-0.95). These reductions in risk were more pronounced in ever smokers, suggesting a gene-environment interaction. In transfection assays, the RGS6-rs2074647 (C-->T) polymorphism increased the activity of a luciferase-RGS fusion protein by 2.9-fold, suggesting that this SNP is functionally significant. Finally, we demonstrate that RGS2 transcripts and several splice variants of RGS6 are expressed in bladder cancer cells. These data provide the first evidence that RGS proteins may be important modulators of cancer risk and validate RGS6 as a target for further study.
Collapse
Affiliation(s)
- David M Berman
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Witt-Enderby PA, Jarzynka MJ, Krawitt BJ, Melan MA. Knock-down of RGS4 and beta tubulin in CHO cells expressing the human MT1 melatonin receptor prevents melatonin-induced receptor desensitization. Life Sci 2004; 75:2703-15. [PMID: 15369705 DOI: 10.1016/j.lfs.2004.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Accepted: 08/04/2004] [Indexed: 11/18/2022]
Abstract
Previously, it has been shown that chronic melatonin exposure in MT1-CHO cells results in receptor desensitization while at the same time producing drastic morphological changes. The addition of a depolymerizing agent during the melatonin pretreatment period prevents MT1 receptor desensitization and the changes in cellular morphology. The lack of morphological change in the presence of a depolymerizing agent is easily explained by the inability of the microtubules to polymerize, however, the prevention of receptor desensitization is a little more complex and may involve G-protein activation. The goal of this study was to determine whether melatonin-induced MT1 receptor desensitization is regulated by proteins known to regulate G-protein activation states, beta-tubulin and RGS4,using anti sense knockdown approaches. The expression of RGS4 mRNA in CHO cells was confirmed using RT PCR and successful knockdown of each was confirmed by western blot analysis or quantitative PCR. Pretreatment of MT1-CHO cells, transfected with the nonsense probes and exposed to melatonin, resulted in a desensitization of the receptor, an increase in forskolin-induced cAMP accumulation, an increase in 2-[125I]-iodomelatonin binding and no change in the affinity of melatonin for the MT1 receptor. However, knockdown of either beta-tubulin or RGS4 in MT1-CHO cells followed by pretreatment with melatonin attenuated the desensitization of melatonin receptors, decreased total 2-[125I]-iodomelatonin binding, and did not affect neither the forskolin response nor the affinity of melatonin for the MT1 receptor. Perhaps RGS4 and beta-tubulin modulate Galpha-GDP and Galpha-GTP states thus modulating MT1 melatonin receptor function.
Collapse
Affiliation(s)
- P A Witt-Enderby
- Division of Pharmaceutical Sciences, Department of Pharmacology-Toxicology, Duquesne University School of Pharmacy, 421 Mellon Hall, Pittsburgh, PA 15282, USA.
| | | | | | | |
Collapse
|
49
|
Grenningloh G, Soehrman S, Bondallaz P, Ruchti E, Cadas H. Role of the microtubule destabilizing proteins SCG10 and stathmin in neuronal growth. ACTA ACUST UNITED AC 2004; 58:60-9. [PMID: 14598370 DOI: 10.1002/neu.10279] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The related proteins SCG10 and stathmin are highly expressed in the developing nervous system. Recently it was discovered that they are potent microtubule destabilizing factors. While stathmin is expressed in a variety of cell types and shows a cytosolic distribution, SCG10 is neuron-specific and membrane-associated. It contains an N-terminal targeting sequence that mediates its transport to the growing tips of axons and dendrites. SCG10 accumulates in the central domain of the growth cone, a region that also contains highly dynamic microtubules. These dynamic microtubules are known to be important for growth cone advance and responses to guidance cues. Because overexpression of SCG10 strongly enhances neurite outgrowth, SCG10 appears to be an important factor for the dynamic assembly and disassembly of growth cone microtubules during axonal elongation. Phosphorylation negatively regulates the microtubule destabilizing activity of SCG10 and stathmin, suggesting that these proteins may link extracellular signals to the rearrangement of the neuronal cytoskeleton. A role for these proteins in axonal elongation is also supported by their growth-associated expression pattern in nervous system development as well as during neuronal regeneration.
Collapse
Affiliation(s)
- Gabriele Grenningloh
- Institut de Biologie Cellulaire et de Morphologie, Université de Lausanne, 1005 Lausanne, Suisse
| | | | | | | | | |
Collapse
|
50
|
Nixon AB, Casey PJ. Analysis of the Regulation of Microtubule Dynamics by Interaction of RGSZ1 (RGS20) with the Neuronal Stathmin, SCG10. Methods Enzymol 2004; 390:53-64. [PMID: 15488170 DOI: 10.1016/s0076-6879(04)90004-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Regulators of G protein signaling (RGS proteins) are a diverse family of proteins that act to negatively regulate signaling by heterotrimeric G proteins; however, recent data have implied additional functions for RGS proteins. Previously, we employed the yeast two-hybrid system and identified the microtubule-destabilizing protein, superior cervical ganglia neural-specific 10 protein (SCG10), as a potential effector protein of RGSZ1. This article describes the expression and biochemical purification of both RGSZ1 and SCG10 and details the development of various in vitro assays to evaluate microtubule polymerization?depolymerization. Both turbidimetric and microscopy-based assays can be employed to study the impact that RGS proteins have on SCG10 function. The application of these in vitro assays may help identify a novel role for RGS proteins in regulating the cytoskeletal network.
Collapse
Affiliation(s)
- Andrew B Nixon
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|