1
|
Leclerc NR, Dunne TM, Shrestha S, Johnson CP, Kelley JB. TOR signaling regulates GPCR levels on the plasma membrane and suppresses the Saccharomyces cerevisiae mating pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593412. [PMID: 38798445 PMCID: PMC11118302 DOI: 10.1101/2024.05.09.593412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Saccharomyces cerevisiae respond to mating pheromone through the GPCRs Ste2 and Ste3, which promote growth of a mating projection in response to ligand binding. This commitment to mating is nutritionally and energetically taxing, and so we hypothesized that the cell may suppress mating signaling during starvation. We set out to investigate negative regulators of the mating pathway in nutritionally depleted environments. Here, we report that nutrient deprivation led to loss of Ste2 from the plasma membrane. Recapitulating this effect with nitrogen starvation led us to hypothesize that it was due to TORC1 signaling. Rapamycin inhibition of TORC1 impacted membrane levels of all yeast GPCRs. Inhibition of TORC1 also dampened mating pathway output. Deletion analysis revealed that TORC1 repression leads to α-arrestin-directed CME through TORC2-Ypk1 signaling. We then set out to determine whether major downstream effectors of the TOR complexes also downregulate pathway output during mating. We found that autophagy contributes to pathway downregulation through analysis of strains lacking ATG8 . We also show that Ypk1 significantly reduced pathway output. Thus, both autophagy machinery and TORC2-Ypk1 signaling serve as attenuators of pheromone signaling during mating. Altogether, we demonstrate that the stress-responsive TOR complexes coordinate GPCR endocytosis and reduce the magnitude of pheromone signaling, in ligand-independent and ligand-dependent contexts. One Sentence Summary TOR signaling regulates the localization of all Saccharomyces cerevisiae GPCRs during starvation and suppress the mating pathway in the presence and absence of ligand.
Collapse
|
2
|
Nürnberg B, Beer-Hammer S, Reisinger E, Leiss V. Non-canonical G protein signaling. Pharmacol Ther 2024; 255:108589. [PMID: 38295906 DOI: 10.1016/j.pharmthera.2024.108589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/18/2023] [Accepted: 01/08/2024] [Indexed: 02/17/2024]
Abstract
The original paradigm of classical - also referred to as canonical - cellular signal transduction of heterotrimeric G proteins (G protein) is defined by a hierarchical, orthograde interaction of three players: the agonist-activated G protein-coupled receptor (GPCR), which activates the transducing G protein, that in turn regulates its intracellular effectors. This receptor-transducer-effector concept was extended by the identification of regulators and adapters such as the regulators of G protein signaling (RGS), receptor kinases like βARK, or GPCR-interacting arrestin adapters that are integrated into this canonical signaling process at different levels to enable fine-tuning. Finally, the identification of atypical signaling mechanisms of classical regulators, together with the discovery of novel modulators, added a new and fascinating dimension to the cellular G protein signal transduction. This heterogeneous group of accessory G protein modulators was coined "activators of G protein signaling" (AGS) proteins and plays distinct roles in canonical and non-canonical G protein signaling pathways. AGS proteins contribute to the control of essential cellular functions such as cell development and division, intracellular transport processes, secretion, autophagy or cell movements. As such, they are involved in numerous biological processes that are crucial for diseases, like diabetes mellitus, cancer, and stroke, which represent major health burdens. Although the identification of a large number of non-canonical G protein signaling pathways has broadened the spectrum of this cellular communication system, their underlying mechanisms, functions, and biological effects are poorly understood. In this review, we highlight and discuss atypical G protein-dependent signaling mechanisms with a focus on inhibitory G proteins (Gi) involved in canonical and non-canonical signal transduction, review recent developments and open questions, address the potential of new approaches for targeted pharmacological interventions.
Collapse
Affiliation(s)
- Bernd Nürnberg
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, and ICePhA Mouse Clinic, University of Tübingen, Wilhelmstraße 56, D-72074 Tübingen, Germany.
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, and ICePhA Mouse Clinic, University of Tübingen, Wilhelmstraße 56, D-72074 Tübingen, Germany
| | - Ellen Reisinger
- Gene Therapy for Hearing Impairment Group, Department of Otolaryngology - Head & Neck Surgery, University of Tübingen Medical Center, Elfriede-Aulhorn-Straße 5, D-72076 Tübingen, Germany
| | - Veronika Leiss
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, and ICePhA Mouse Clinic, University of Tübingen, Wilhelmstraße 56, D-72074 Tübingen, Germany
| |
Collapse
|
3
|
Li L, Xu Q, Tang C. RGS proteins and their roles in cancer: friend or foe? Cancer Cell Int 2023; 23:81. [PMID: 37118788 PMCID: PMC10148553 DOI: 10.1186/s12935-023-02932-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023] Open
Abstract
As negative modulators of G-protein-coupled receptors (GPCRs) signaling, regulators of G protein signaling (RGS) proteins facilitate various downstream cellular signalings through regulating kinds of heterotrimeric G proteins by stimulating the guanosine triphosphatase (GTPase) activity of G-protein α (Gα) subunits. The expression of RGS proteins is dynamically and precisely mediated by several different mechanisms including epigenetic regulation, transcriptional regulation -and post-translational regulation. Emerging evidence has shown that RGS proteins act as important mediators in controlling essential cellular processes including cell proliferation, survival -and death via regulating downstream cellular signaling activities, indicating that RGS proteins are fundamentally involved in sustaining normal physiological functions and dysregulation of RGS proteins (such as aberrant expression of RGS proteins) is closely associated with pathologies of many diseases such as cancer. In this review, we summarize the molecular mechanisms governing the expression of RGS proteins, and further discuss the relationship of RGS proteins and cancer.
Collapse
Affiliation(s)
- Lin Li
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China
| | - Qiang Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China
| | - Chao Tang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
4
|
Borges JI, Suster MS, Lymperopoulos A. Cardiac RGS Proteins in Human Heart Failure and Atrial Fibrillation: Focus on RGS4. Int J Mol Sci 2023; 24:6136. [PMID: 37047106 PMCID: PMC10147095 DOI: 10.3390/ijms24076136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023] Open
Abstract
The regulator of G protein signaling (RGS) proteins are crucial for the termination of G protein signals elicited by G protein-coupled receptors (GPCRs). This superfamily of cell membrane receptors, by far the largest and most versatile in mammals, including humans, play pivotal roles in the regulation of cardiac function and homeostasis. Perturbations in both the activation and termination of their G protein-mediated signaling underlie numerous heart pathologies, including heart failure (HF) and atrial fibrillation (AFib). Therefore, RGS proteins play important roles in the pathophysiology of these two devasting cardiac diseases, and several of them could be targeted therapeutically. Although close to 40 human RGS proteins have been identified, each RGS protein seems to interact only with a specific set of G protein subunits and GPCR types/subtypes in any given tissue or cell type. Numerous in vitro and in vivo studies in animal models, and also in diseased human heart tissue obtained from transplantations or tissue banks, have provided substantial evidence of the roles various cardiomyocyte RGS proteins play in cardiac normal homeostasis as well as pathophysiology. One RGS protein in particular, RGS4, has been reported in what are now decades-old studies to be selectively upregulated in human HF. It has also been implicated in protection against AFib via knockout mice studies. This review summarizes the current understanding of the functional roles of cardiac RGS proteins and their implications for the treatment of HF and AFib, with a specific focus on RGS4 for the aforementioned reasons but also because it can be targeted successfully with small organic molecule inhibitors.
Collapse
Affiliation(s)
| | | | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverrman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
5
|
Del Calvo G, Baggio Lopez T, Lymperopoulos A. The therapeutic potential of targeting cardiac RGS4. Ther Adv Cardiovasc Dis 2023; 17:17539447231199350. [PMID: 37724539 PMCID: PMC10510358 DOI: 10.1177/17539447231199350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/16/2023] [Indexed: 09/21/2023] Open
Abstract
G protein-coupled receptors (GPCRs) play pivotal roles in regulation of cardiac function and homeostasis. To function properly, every cell needs these receptors to be stimulated only when a specific extracellular stimulus is present, and to be silenced the moment that stimulus is removed. The regulator of G protein signaling (RGS) proteins are crucial for the latter to occur at the cell membrane, where the GPCR normally resides. Perturbations in both activation and termination of G protein signaling underlie numerous heart pathologies. Although more than 30 mammalian RGS proteins have been identified, each RGS protein seems to interact only with a specific set of G protein subunits and GPCR types/subtypes in any given tissue or cell type, and this applies to the myocardium as well. A large number of studies have provided substantial evidence for the roles various RGS proteins expressed in cardiomyocytes play in cardiac physiology and heart disease pathophysiology. This review summarizes the current understanding of the functional roles of cardiac RGS proteins and their implications for the treatment of specific heart diseases, such as heart failure and atrial fibrillation. We focus on cardiac RGS4 in particular, since this isoform appears to be selectively (among the RGS protein family) upregulated in human heart failure and is also the target of ongoing drug discovery efforts for the treatment of a variety of diseases.
Collapse
Affiliation(s)
- Giselle Del Calvo
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Teresa Baggio Lopez
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, HPD (Terry) Building/Room 1350, Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
6
|
Simke WC, Johnson CP, Hart AJ, Mayhue S, Craig PL, Sojka S, Kelley JB. Phosphorylation of RGS regulates MAP kinase localization and promotes completion of cytokinesis. Life Sci Alliance 2022; 5:5/10/e202101245. [PMID: 35985794 PMCID: PMC9394524 DOI: 10.26508/lsa.202101245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/03/2022] Open
Abstract
Phosphorylation of the RGS Sst2 alters its subcellular distribution, MAPK localization, and interaction with Kel1, which promotes coordination of polarized growth with completion of cytokinesis. Yeast use the G-protein–coupled receptor signaling pathway to detect and track the mating pheromone. The G-protein–coupled receptor pathway is inhibited by the regulator of G-protein signaling (RGS) Sst2 which induces Gα GTPase activity and inactivation of downstream signaling. G-protein signaling activates the MAPK Fus3, which phosphorylates the RGS; however, the role of this modification is unknown. We found that pheromone-induced RGS phosphorylation peaks early; the phospho-state of RGS controls its localization and influences MAPK spatial distribution. Surprisingly, phosphorylation of the RGS promotes completion of cytokinesis before pheromone-induced growth. Completion of cytokinesis in the presence of pheromone is promoted by the kelch-repeat protein, Kel1 and antagonized by the formin Bni1. We found that RGS complexes with Kel1 and prefers the unphosphorylatable RGS mutant. We also found overexpression of unphosphorylatable RGS exacerbates cytokinetic defects, whereas they are rescued by overexpression of Kel1. These data lead us to a model where Kel1 promotes completion of cytokinesis before pheromone-induced polarity but is inhibited by unphosphorylated RGS binding.
Collapse
Affiliation(s)
- William C Simke
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Cory P Johnson
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Andrew J Hart
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Sari Mayhue
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - P Lucas Craig
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Savannah Sojka
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Joshua B Kelley
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA .,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| |
Collapse
|
7
|
Lymperopoulos A, Suster MS, Borges JI. Cardiovascular GPCR regulation by regulator of G protein signaling proteins. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 193:145-166. [PMID: 36357075 DOI: 10.1016/bs.pmbts.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
G protein-coupled receptors (GPCRs) play pivotal roles in regulation of cardiovascular homeostasis across all vertebrate species, including humans. In terms of normal cellular function, termination of GPCR signaling via the heterotrimeric G proteins is equally (if not more) important to its stimulation. The Regulator of G protein Signaling (RGS) protein superfamily are indispensable for GPCR signaling cessation at the cell membrane, and thus, for cellular control of GPCR signaling and function. Perturbations in both activation and termination of G protein signaling underlie many examples of cardiovascular dysfunction and heart disease pathogenesis. Despite the plethora of over 30 members comprising the mammalian RGS protein superfamily, each member interacts with a specific set of second messenger pathways and GPCR types/subtypes in a tissue/cell type-specific manner. An increasing number of studies over the past two decades have provided compelling evidence for the involvement of various RGS proteins in physiological regulation of cardiovascular GPCRs and, consequently, also in the pathophysiology of several cardiovascular ailments. This chapter summarizes the current understanding of the functional roles of RGS proteins as they pertain to cardiovascular, i.e., heart, blood vessel, and platelet GPCR function, with a particular focus on their implications for chronic heart failure pathophysiology and therapy.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States.
| | - Malka S Suster
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States
| | - Jordana I Borges
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States
| |
Collapse
|
8
|
Fina ME, Wang J, Vedula P, Tang HY, Kashina A, Dong DW. Arginylation Regulates G-protein Signaling in the Retina. Front Cell Dev Biol 2022; 9:807345. [PMID: 35127722 PMCID: PMC8815403 DOI: 10.3389/fcell.2021.807345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/17/2021] [Indexed: 12/03/2022] Open
Abstract
Arginylation is a post-translational modification mediated by the arginyltransferase (Ate1). We recently showed that conditional deletion of Ate1 in the nervous system leads to increased light-evoked response sensitivities of ON-bipolar cells in the retina, indicating that arginylation regulates the G-protein signaling complexes of those neurons and/or photoreceptors. However, none of the key players in the signaling pathway were previously shown to be arginylated. Here we show that Gαt1, Gβ1, RGS6, and RGS7 are arginylated in the retina and RGS6 and RGS7 protein levels are elevated in Ate1 knockout, suggesting that arginylation plays a direct role in regulating their protein level and the G-protein-mediated responses in the retina.
Collapse
Affiliation(s)
- Marie E. Fina
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, United States
| | - Junling Wang
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, United States
| | - Pavan Vedula
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, United States
| | - Hsin-Yao Tang
- Proteomics and Metabolomics Facility, The Wistar Institute, Philadelphia, PA, United States
| | - Anna Kashina
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Anna Kashina, ; Dawei W. Dong,
| | - Dawei W. Dong
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Anna Kashina, ; Dawei W. Dong,
| |
Collapse
|
9
|
Garcia I, Orellana-Muñoz S, Ramos-Alonso L, Andersen AN, Zimmermann C, Eriksson J, Bøe SO, Kaferle P, Papamichos-Chronakis M, Chymkowitch P, Enserink JM. Kel1 is a phosphorylation-regulated noise suppressor of the pheromone signaling pathway. Cell Rep 2021; 37:110186. [PMID: 34965431 DOI: 10.1016/j.celrep.2021.110186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/01/2021] [Accepted: 12/07/2021] [Indexed: 11/26/2022] Open
Abstract
Mechanisms have evolved that allow cells to detect signals and generate an appropriate response. The accuracy of these responses relies on the ability of cells to discriminate between signal and noise. How cells filter noise in signaling pathways is not well understood. Here, we analyze noise suppression in the yeast pheromone signaling pathway and show that the poorly characterized protein Kel1 serves as a major noise suppressor and prevents cell death. At the molecular level, Kel1 prevents spontaneous activation of the pheromone response by inhibiting membrane recruitment of Ste5 and Far1. Only a hypophosphorylated form of Kel1 suppresses signaling, reduces noise, and prevents pheromone-associated cell death, and our data indicate that the MAPK Fus3 contributes to Kel1 phosphorylation. Taken together, Kel1 serves as a phospho-regulated suppressor of the pheromone pathway to reduce noise, inhibit spontaneous activation of the pathway, regulate mating efficiency, and prevent pheromone-associated cell death.
Collapse
Affiliation(s)
- Ignacio Garcia
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
| | - Sara Orellana-Muñoz
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
| | - Lucía Ramos-Alonso
- Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway; Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Aram N Andersen
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway; Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
| | - Christine Zimmermann
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Jens Eriksson
- Department of Medical Biochemistry and Microbiology, Uppsala University, 752 37 Uppsala, Sweden
| | - Stig Ove Bøe
- Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Petra Kaferle
- Institut Curie, PSL Research University, CNRS, UMR3664, Sorbonne Universities, Paris, France
| | - Manolis Papamichos-Chronakis
- Department of Molecular Physiology and Cell Signalling Institute of Systems, Molecular and Integrative Biology University of Liverpool, L69 7BE Liverpool, UK
| | - Pierre Chymkowitch
- Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway; Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Jorrit M Enserink
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, 0379 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway; Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway.
| |
Collapse
|
10
|
Fina ME, Wang J, Nikonov SS, Sterling S, Vardi N, Kashina A, Dong DW. Arginyltransferase (Ate1) regulates the RGS7 protein level and the sensitivity of light-evoked ON-bipolar responses. Sci Rep 2021; 11:9376. [PMID: 33931669 PMCID: PMC8087773 DOI: 10.1038/s41598-021-88628-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Regulator of G-protein signaling 7 (RGS7) is predominately present in the nervous system and is essential for neuronal signaling involving G-proteins. Prior studies in cultured cells showed that RGS7 is regulated via proteasomal degradation, however no protein is known to facilitate proteasomal degradation of RGS7 and it has not been shown whether this regulation affects G-protein signaling in neurons. Here we used a knockout mouse model with conditional deletion of arginyltransferase (Ate1) in the nervous system and found that in retinal ON bipolar cells, where RGS7 modulates a G-protein to signal light increments, deletion of Ate1 raised the level of RGS7. Electroretinographs revealed that lack of Ate1 leads to increased light-evoked response sensitivities of ON-bipolar cells, as well as their downstream neurons. In cultured mouse embryonic fibroblasts (MEF), RGS7 was rapidly degraded via proteasome pathway and this degradation was abolished in Ate1 knockout MEF. Our results indicate that Ate1 regulates RGS7 protein level by facilitating proteasomal degradation of RGS7 and thus affects G-protein signaling in neurons.
Collapse
Affiliation(s)
- Marie E Fina
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Junling Wang
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sergei S Nikonov
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Stephanie Sterling
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Noga Vardi
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anna Kashina
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Dawei W Dong
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
11
|
Calebiro D, Koszegi Z, Lanoiselée Y, Miljus T, O'Brien S. G protein-coupled receptor-G protein interactions: a single-molecule perspective. Physiol Rev 2020; 101:857-906. [PMID: 33331229 DOI: 10.1152/physrev.00021.2020] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors (GPCRs) regulate many cellular and physiological processes, responding to a diverse range of extracellular stimuli including hormones, neurotransmitters, odorants, and light. Decades of biochemical and pharmacological studies have provided fundamental insights into the mechanisms of GPCR signaling. Thanks to recent advances in structural biology, we now possess an atomistic understanding of receptor activation and G protein coupling. However, how GPCRs and G proteins interact in living cells to confer signaling efficiency and specificity remains insufficiently understood. The development of advanced optical methods, including single-molecule microscopy, has provided the means to study receptors and G proteins in living cells with unprecedented spatio-temporal resolution. The results of these studies reveal an unexpected level of complexity, whereby GPCRs undergo transient interactions among themselves as well as with G proteins and structural elements of the plasma membrane to form short-lived signaling nanodomains that likely confer both rapidity and specificity to GPCR signaling. These findings may provide new strategies to pharmaceutically modulate GPCR function, which might eventually pave the way to innovative drugs for common diseases such as diabetes or heart failure.
Collapse
Affiliation(s)
- Davide Calebiro
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| | - Zsombor Koszegi
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| | - Yann Lanoiselée
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| | - Tamara Miljus
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| | - Shannon O'Brien
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| |
Collapse
|
12
|
DiGiacomo V, Maziarz M, Luebbers A, Norris JM, Laksono P, Garcia-Marcos M. Probing the mutational landscape of regulators of G protein signaling proteins in cancer. Sci Signal 2020; 13:13/617/eaax8620. [PMID: 32019900 DOI: 10.1126/scisignal.aax8620] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The advent of deep-sequencing techniques has revealed that mutations in G protein-coupled receptor (GPCR) signaling pathways in cancer are more prominent than was previously appreciated. An emergent theme is that cancer-associated mutations tend to cause enhanced GPCR pathway activation to favor oncogenicity. Regulators of G protein signaling (RGS) proteins are critical modulators of GPCR signaling that dampen the activity of heterotrimeric G proteins through their GTPase-accelerating protein (GAP) activity, which is conferred by a conserved domain dubbed the "RGS-box." Here, we developed an experimental pipeline to systematically assess the mutational landscape of RGS GAPs in cancer. A pan-cancer bioinformatics analysis of the 20 RGS domains with GAP activity revealed hundreds of low-frequency mutations spread throughout the conserved RGS domain structure with a slight enrichment at positions that interface with G proteins. We empirically tested multiple mutations representing all RGS GAP subfamilies and sampling both G protein interface and noninterface positions with a scalable, yeast-based assay. Last, a subset of mutants was validated using G protein activity biosensors in mammalian cells. Our findings reveal that a sizable fraction of RGS protein mutations leads to a loss of function through various mechanisms, including disruption of the G protein-binding interface, loss of protein stability, or allosteric effects on G protein coupling. Moreover, our results also validate a scalable pipeline for the rapid characterization of cancer-associated mutations in RGS proteins.
Collapse
Affiliation(s)
- Vincent DiGiacomo
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Marcin Maziarz
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Alex Luebbers
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jillian M Norris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Pandu Laksono
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mikel Garcia-Marcos
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
13
|
O'Brien JB, Wilkinson JC, Roman DL. Regulator of G-protein signaling (RGS) proteins as drug targets: Progress and future potentials. J Biol Chem 2019; 294:18571-18585. [PMID: 31636120 PMCID: PMC6901330 DOI: 10.1074/jbc.rev119.007060] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors (GPCRs) play critical roles in regulating processes such as cellular homeostasis, responses to stimuli, and cell signaling. Accordingly, GPCRs have long served as extraordinarily successful drug targets. It is therefore not surprising that the discovery in the mid-1990s of a family of proteins that regulate processes downstream of GPCRs generated great excitement in the field. This finding enhanced the understanding of these critical signaling pathways and provided potentially new targets for pharmacological intervention. These regulators of G-protein signaling (RGS) proteins were viewed by many as nodes downstream of GPCRs that could be targeted with small molecules to tune signaling processes. In this review, we provide a brief overview of the discovery of RGS proteins and of the gradual and continuing discovery of their roles in disease states, focusing particularly on cancer and neurological disorders. We also discuss high-throughput screening efforts that have led to the discovery first of peptide-based and then of small-molecule inhibitors targeting a subset of the RGS proteins. We explore the unique mechanisms of RGS inhibition these chemical tools have revealed and highlight the most up-to-date studies using these tools in animal experiments. Finally, we discuss the future opportunities in the field, as there are clearly more avenues left to be explored and potentials to be realized.
Collapse
Affiliation(s)
- Joseph B O'Brien
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa 52242
| | - Joshua C Wilkinson
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa 52242
| | - David L Roman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa 52242; Iowa Neuroscience Institute, Iowa City, Iowa 52242; Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242.
| |
Collapse
|
14
|
Sieng M, Hayes MP, O'Brien JB, Andrew Fowler C, Houtman JC, Roman DL, Lyon AM. High-resolution structure of RGS17 suggests a role for Ca 2+ in promoting the GTPase-activating protein activity by RZ subfamily members. J Biol Chem 2019; 294:8148-8160. [PMID: 30940727 DOI: 10.1074/jbc.ra118.006059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 03/27/2019] [Indexed: 11/06/2022] Open
Abstract
Regulator of G protein signaling (RGS) proteins are negative regulators of G protein-coupled receptor (GPCR) signaling through their ability to act as GTPase-activating proteins (GAPs) for activated Gα subunits. Members of the RZ subfamily of RGS proteins bind to activated Gαo, Gαz, and Gαi1-3 proteins in the nervous system and thereby inhibit downstream pathways, including those involved in Ca2+-dependent signaling. In contrast to other RGS proteins, little is known about RZ subfamily structure and regulation. Herein, we present the 1.5-Å crystal structure of RGS17, the most complete and highest-resolution structure of an RZ subfamily member to date. RGS17 cocrystallized with Ca2+ bound to conserved positions on the predicted Gα-binding surface of the protein. Using NMR chemical shift perturbations, we confirmed that Ca2+ binds in solution to the same site. Furthermore, RGS17 had greater than 55-fold higher affinity for Ca2+ than for Mg2+ Finally, we found that Ca2+ promotes interactions between RGS17 and activated Gα and decreases the Km for GTP hydrolysis, potentially by altering the binding mechanism between these proteins. Taken together, these findings suggest that Ca2+ positively regulates RGS17, which may represent a general mechanism by which increased Ca2+ concentration promotes the GAP activity of the RZ subfamily, leading to RZ-mediated inhibition of Ca2+ signaling.
Collapse
Affiliation(s)
- Monita Sieng
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907
| | - Michael P Hayes
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242
| | - Joseph B O'Brien
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242
| | - C Andrew Fowler
- NMR Facility, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | - Jon C Houtman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa 52242
| | - David L Roman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242
| | - Angeline M Lyon
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907; Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907.
| |
Collapse
|
15
|
Squires KE, Montañez-Miranda C, Pandya RR, Torres MP, Hepler JR. Genetic Analysis of Rare Human Variants of Regulators of G Protein Signaling Proteins and Their Role in Human Physiology and Disease. Pharmacol Rev 2018; 70:446-474. [PMID: 29871944 PMCID: PMC5989036 DOI: 10.1124/pr.117.015354] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regulators of G protein signaling (RGS) proteins modulate the physiologic actions of many neurotransmitters, hormones, and other signaling molecules. Human RGS proteins comprise a family of 20 canonical proteins that bind directly to G protein-coupled receptors/G protein complexes to limit the lifetime of their signaling events, which regulate all aspects of cell and organ physiology. Genetic variations account for diverse human traits and individual predispositions to disease. RGS proteins contribute to many complex polygenic human traits and pathologies such as hypertension, atherosclerosis, schizophrenia, depression, addiction, cancers, and many others. Recent analysis indicates that most human diseases are due to extremely rare genetic variants. In this study, we summarize physiologic roles for RGS proteins and links to human diseases/traits and report rare variants found within each human RGS protein exome sequence derived from global population studies. Each RGS sequence is analyzed using recently described bioinformatics and proteomic tools for measures of missense tolerance ratio paired with combined annotation-dependent depletion scores, and protein post-translational modification (PTM) alignment cluster analysis. We highlight selected variants within the well-studied RGS domain that likely disrupt RGS protein functions and provide comprehensive variant and PTM data for each RGS protein for future study. We propose that rare variants in functionally sensitive regions of RGS proteins confer profound change-of-function phenotypes that may contribute, in newly appreciated ways, to complex human diseases and/or traits. This information provides investigators with a valuable database to explore variation in RGS protein function, and for targeting RGS proteins as future therapeutic targets.
Collapse
Affiliation(s)
- Katherine E Squires
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Carolina Montañez-Miranda
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Rushika R Pandya
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - Matthew P Torres
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| | - John R Hepler
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.)
| |
Collapse
|
16
|
Establishing a Split Luciferase Assay for Proteinkinase G (PKG) Interaction Studies. Int J Mol Sci 2018; 19:ijms19041180. [PMID: 29649180 PMCID: PMC5979328 DOI: 10.3390/ijms19041180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/04/2018] [Accepted: 04/04/2018] [Indexed: 02/06/2023] Open
Abstract
Nitric oxide (NO/cyclic guanosine monophosphate (cGMP)-regulated cellular mechanisms are involved in a variety of (patho-) physiological processes. One of the main effector molecules in this system, proteinkinase G (PKG), serves as a molecular switch by phosphorylating different target proteins and thereby turning them on or off. To date, only a few interaction partners of PKG have been described although the identification of protein–protein interactions (PPI) is indispensable for the understanding of cellular processes and diseases. Conventionally used methods to detect PPIs exhibit several disadvantages, e.g., co-immunoprecipitations, which depend on suitable high-affinity antibodies. Therefore, we established a cell-based protein-fragment complementation assay (PCA) for the identification of PKG target proteins. Here, a reporter protein (click beetle luciferase) is split into two fragments and fused to two different possible interaction partners. If interaction occurs, the reporter protein is functionally complemented and the catalyzed reaction can then be quantitatively measured. By using this technique, we confirmed the regulator of G-Protein signaling 2 (RGS2) as an interaction partner of PKGIα (a PKG-isoform) following stimulation with 8-Br-cGMP and 8-pCPT-cGMP. Hence, our results support the conclusion that the established approach could serve as a novel tool for the rapid, easy and cost-efficient detection of novel PKG target proteins.
Collapse
|
17
|
Cevheroğlu O, Becker JM, Son ÇD. GPCR-Gα protein precoupling: Interaction between Ste2p, a yeast GPCR, and Gpa1p, its Gα protein, is formed before ligand binding via the Ste2p C-terminal domain and the Gpa1p N-terminal domain. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:2435-2446. [PMID: 28958779 DOI: 10.1016/j.bbamem.2017.09.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 09/20/2017] [Accepted: 09/22/2017] [Indexed: 01/19/2023]
Abstract
G protein coupled receptors bind ligands that initiate intracellular signaling cascades via heterotrimeric G proteins. In this study, involvement of the N-terminal residues of yeast G-alpha (Gpa1p) with the C-terminal residues of a full-length or C-terminally truncated Ste2p were investigated using bioluminescence resonance energy transfer (BRET), a non-radiative energy transfer phenomenon where protein-protein interactions can be quantified between a donor bioluminescent molecule and a suitable acceptor fluorophore. Constitutive and position-dependent BRET signal was observed in the absence of agonist (α-factor). Upon the activation of the receptors with α-factor, no significant change in BRET signal was observed. The location of Ste2p-Gpa1p heterodimer was investigated using confocal fluorescence microscopy and bimolecular fluorescence complementation (BiFC) assay, a technique where two non-fluorescent fragments of a fluorescent protein reassemble in vivo to restore fluorescence property thereby directly reporting a protein-protein interaction. BiFC experiments resulted in a dimerization signal intracellularly during biosynthesis on the endoplasmic reticulum (ER) and on the plasma membrane (PM). The constitutive BRET and BiFC signals observed on ER between Ste2p and Gpa1p in their quiescent and activated states are indicative of pre-coupling between these two proteins. This study is the first to show that the extreme N-terminus of yeast G protein alpha subunit is in close proximity to its receptor. The data suggests a pre-coupled heterodimer prior to receptor activation. The images presented in this study are the first direct in vivo evidence showing the localization of receptor - G protein heterodimers during biosynthesis and before reaching the plasma membrane.
Collapse
Affiliation(s)
- Orkun Cevheroğlu
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996-0845, United States; Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupinar Blv. No: 1, Çankaya, Ankara, 06800, Turkey
| | - Jeffrey M Becker
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996-0845, United States
| | - Çağdaş D Son
- Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupinar Blv. No: 1, Çankaya, Ankara, 06800, Turkey.
| |
Collapse
|
18
|
Sjögren B. The evolution of regulators of G protein signalling proteins as drug targets - 20 years in the making: IUPHAR Review 21. Br J Pharmacol 2017; 174:427-437. [PMID: 28098342 DOI: 10.1111/bph.13716] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 12/11/2016] [Accepted: 01/08/2017] [Indexed: 12/11/2022] Open
Abstract
Regulators of G protein signalling (RGS) proteins are celebrating the 20th anniversary of their discovery. The unveiling of this new family of negative regulators of G protein signalling in the mid-1990s solved a persistent conundrum in the G protein signalling field, in which the rate of deactivation of signalling cascades in vivo could not be replicated in exogenous systems. Since then, there has been tremendous advancement in the knowledge of RGS protein structure, function, regulation and their role as novel drug targets. RGS proteins play an important modulatory role through their GTPase-activating protein (GAP) activity at active, GTP-bound Gα subunits of heterotrimeric G proteins. They also possess many non-canonical functions not related to G protein signalling. Here, an update on the status of RGS proteins as drug targets is provided, highlighting advances that have led to the inclusion of RGS proteins in the IUPHAR/BPS Guide to PHARMACOLOGY database of drug targets.
Collapse
Affiliation(s)
- B Sjögren
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
19
|
Rajagopal S, Shenoy SK. GPCR desensitization: Acute and prolonged phases. Cell Signal 2017; 41:9-16. [PMID: 28137506 DOI: 10.1016/j.cellsig.2017.01.024] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 01/25/2017] [Indexed: 01/04/2023]
Abstract
G protein-coupled receptors (GPCRs) transduce a wide array of extracellular signals and regulate virtually every aspect of physiology. While GPCR signaling is essential, overstimulation can be deleterious, resulting in cellular toxicity or uncontrolled cellular growth. Accordingly, nature has developed a number of mechanisms for limiting GPCR signaling, which are broadly referred to as desensitization, and refer to a decrease in response to repeated or continuous stimulation. Short-term desensitization occurs over minutes, and is primarily associated with β-arrestins preventing G protein interaction with a GPCR. Longer-term desensitization, referred to as downregulation, occurs over hours to days, and involves receptor internalization into vesicles, degradation in lysosomes and decreased receptor mRNA levels through unclear mechanisms. Phosphorylation of the receptor by GPCR kinases (GRKs) and the recruitment of β-arrestins is critical to both these short- and long-term desensitization mechanisms. In addition to phosphorylation, both the GPCR and β-arrestins are modified post-translationally in several ways, including by ubiquitination. For many GPCRs, receptor ubiquitination promotes degradation of agonist-activated receptors in the lysosomes. Other proteins also play important roles in desensitization, including phosphodiesterases, RGS family proteins and A-kinase-anchoring proteins. Together, this intricate network of kinases, ubiquitin ligases, and adaptor proteins orchestrate the acute and prolonged desensitization of GPCRs.
Collapse
Affiliation(s)
| | - Sudha K Shenoy
- Department of Medicine (Cardiology), Durham, NC, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
20
|
Bush A, Vasen G, Constantinou A, Dunayevich P, Patop IL, Blaustein M, Colman-Lerner A. Yeast GPCR signaling reflects the fraction of occupied receptors, not the number. Mol Syst Biol 2016; 12:898. [PMID: 28034910 PMCID: PMC5199120 DOI: 10.15252/msb.20166910] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
According to receptor theory, the effect of a ligand depends on the amount of agonist-receptor complex. Therefore, changes in receptor abundance should have quantitative effects. However, the response to pheromone in Saccharomyces cerevisiae is robust (unaltered) to increases or reductions in the abundance of the G-protein-coupled receptor (GPCR), Ste2, responding instead to the fraction of occupied receptor. We found experimentally that this robustness originates during G-protein activation. We developed a complete mathematical model of this step, which suggested the ability to compute fractional occupancy depends on the physical interaction between the inhibitory regulator of G-protein signaling (RGS), Sst2, and the receptor. Accordingly, replacing Sst2 by the heterologous hsRGS4, incapable of interacting with the receptor, abolished robustness. Conversely, forcing hsRGS4:Ste2 interaction restored robustness. Taken together with other results of our work, we conclude that this GPCR pathway computes fractional occupancy because ligand-bound GPCR-RGS complexes stimulate signaling while unoccupied complexes actively inhibit it. In eukaryotes, many RGSs bind to specific GPCRs, suggesting these complexes with opposing activities also detect fraction occupancy by a ratiometric measurement. Such complexes operate as push-pull devices, which we have recently described.
Collapse
Affiliation(s)
- Alan Bush
- Department of Physiology, Molecular and Cellular Biology, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Physiology, Molecular Biology and Neurosciences, National Research Council (CONICET), Buenos Aires, Argentina
| | - Gustavo Vasen
- Department of Physiology, Molecular and Cellular Biology, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Physiology, Molecular Biology and Neurosciences, National Research Council (CONICET), Buenos Aires, Argentina
| | - Andreas Constantinou
- Department of Physiology, Molecular and Cellular Biology, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Physiology, Molecular Biology and Neurosciences, National Research Council (CONICET), Buenos Aires, Argentina
| | - Paula Dunayevich
- Department of Physiology, Molecular and Cellular Biology, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Physiology, Molecular Biology and Neurosciences, National Research Council (CONICET), Buenos Aires, Argentina
| | - Inés Lucía Patop
- Department of Physiology, Molecular and Cellular Biology, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Physiology, Molecular Biology and Neurosciences, National Research Council (CONICET), Buenos Aires, Argentina
| | - Matías Blaustein
- Department of Physiology, Molecular and Cellular Biology, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Physiology, Molecular Biology and Neurosciences, National Research Council (CONICET), Buenos Aires, Argentina
| | - Alejandro Colman-Lerner
- Department of Physiology, Molecular and Cellular Biology, University of Buenos Aires, Buenos Aires, Argentina .,Institute of Physiology, Molecular Biology and Neurosciences, National Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
21
|
Tayou J, Wang Q, Jang GF, Pronin AN, Orlandi C, Martemyanov KA, Crabb JW, Slepak VZ. Regulator of G Protein Signaling 7 (RGS7) Can Exist in a Homo-oligomeric Form That Is Regulated by Gαo and R7-binding Protein. J Biol Chem 2016; 291:9133-47. [PMID: 26895961 DOI: 10.1074/jbc.m115.694075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Indexed: 11/06/2022] Open
Abstract
RGS (regulator of G protein signaling) proteins of the R7 subfamily (RGS6, -7, -9, and -11) are highly expressed in neurons where they regulate many physiological processes. R7 RGS proteins contain several distinct domains and form obligatory dimers with the atypical Gβ subunit, Gβ5 They also interact with other proteins such as R7-binding protein, R9-anchoring protein, and the orphan receptors GPR158 and GPR179. These interactions facilitate plasma membrane targeting and stability of R7 proteins and modulate their activity. Here, we investigated RGS7 complexes using in situ chemical cross-linking. We found that in mouse brain and transfected cells cross-linking causes formation of distinct RGS7 complexes. One of the products had the apparent molecular mass of ∼150 kDa on SDS-PAGE and did not contain Gβ5 Mass spectrometry analysis showed no other proteins to be present within the 150-kDa complex in the amount close to stoichiometric with RGS7. This finding suggested that RGS7 could form a homo-oligomer. Indeed, co-immunoprecipitation of differentially tagged RGS7 constructs, with or without chemical cross-linking, demonstrated RGS7 self-association. RGS7-RGS7 interaction required the DEP domain but not the RGS and DHEX domains or the Gβ5 subunit. Using transfected cells and knock-out mice, we demonstrated that R7-binding protein had a strong inhibitory effect on homo-oligomerization of RGS7. In contrast, our data indicated that GPR158 could bind to the RGS7 homo-oligomer without causing its dissociation. Co-expression of constitutively active Gαo prevented the RGS7-RGS7 interaction. These results reveal the existence of RGS protein homo-oligomers and show regulation of their assembly by R7 RGS-binding partners.
Collapse
Affiliation(s)
- Junior Tayou
- From the Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Qiang Wang
- From the Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Geeng-Fu Jang
- the Cole Eye Institute Cleveland Clinic, Cleveland, Ohio 44195, and
| | - Alexey N Pronin
- From the Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida 33136
| | - Cesare Orlandi
- the Department of Neuroscience, Scripps Research Institute, Jupiter, Florida 33458
| | - Kirill A Martemyanov
- the Department of Neuroscience, Scripps Research Institute, Jupiter, Florida 33458
| | - John W Crabb
- the Cole Eye Institute Cleveland Clinic, Cleveland, Ohio 44195, and
| | - Vladlen Z Slepak
- From the Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida 33136,
| |
Collapse
|
22
|
Knight A, Hemmings JL, Winfield I, Leuenberger M, Frattini E, Frenguelli BG, Dowell SJ, Lochner M, Ladds G. Discovery of Novel Adenosine Receptor Agonists That Exhibit Subtype Selectivity. J Med Chem 2016; 59:947-64. [DOI: 10.1021/acs.jmedchem.5b01402] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Anthony Knight
- Systems
Biology Doctoral Training Centre, University of Warwick, Coventry CV4 7AL, U.K
| | - Jennifer L. Hemmings
- Department
of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
| | - Ian Winfield
- Division
of Biomedical Cell Biology, Warwick Medical School, University of Warwick, Coventry CV4 7AL, U.K
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Michele Leuenberger
- Department
of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
| | - Eugenia Frattini
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | | | - Simon J. Dowell
- Department
of Platform Technology and Science, GlaxoSmithKline, Hertfordshire SG1 2NY, U.K
| | - Martin Lochner
- Department
of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
| | - Graham Ladds
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| |
Collapse
|
23
|
Taylor VG, Bommarito PA, Tesmer JJG. Structure of the Regulator of G Protein Signaling 8 (RGS8)-Gαq Complex: MOLECULAR BASIS FOR Gα SELECTIVITY. J Biol Chem 2016; 291:5138-45. [PMID: 26755720 DOI: 10.1074/jbc.m115.712075] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Indexed: 11/06/2022] Open
Abstract
Regulator of G protein signaling (RGS) proteins interact with activated Gα subunits via their RGS domains and accelerate the hydrolysis of GTP. Although the R4 subfamily of RGS proteins generally accepts both Gαi/o and Gαq/11 subunits as substrates, the R7 and R12 subfamilies select against Gαq/11. In contrast, only one RGS protein, RGS2, is known to be selective for Gαq/11. The molecular basis for this selectivity is not clear. Previously, the crystal structure of RGS2 in complex with Gαq revealed a non-canonical interaction that could be due to interfacial differences imposed by RGS2, the Gα subunit, or both. To resolve this ambiguity, the 2.6 Å crystal structure of RGS8, an R4 subfamily member, was determined in complex with Gαq. RGS8 adopts the same pose on Gαq as it does when bound to Gαi3, indicating that the non-canonical interaction of RGS2 with Gαq is due to unique features of RGS2. Based on the RGS8-Gαq structure, residues in RGS8 that contact a unique α-helical domain loop of Gαq were converted to those typically found in R12 subfamily members, and the reverse substitutions were introduced into RGS10, an R12 subfamily member. Although these substitutions perturbed their ability to stimulate GTP hydrolysis, they did not reverse selectivity. Instead, selectivity for Gαq seems more likely determined by whether strong contacts can be maintained between α6 of the RGS domain and Switch III of Gαq, regions of high sequence and conformational diversity in both protein families.
Collapse
Affiliation(s)
- Veronica G Taylor
- From the Life Sciences Institute and the Departments of Pharmacology and Biological Sciences, and the Program in Biophysics, University of Michigan, Ann Arbor, Michigan 48109
| | - Paige A Bommarito
- From the Life Sciences Institute and the Departments of Pharmacology and Biological Sciences, and
| | - John J G Tesmer
- From the Life Sciences Institute and the Departments of Pharmacology and Biological Sciences, and
| |
Collapse
|
24
|
Santhappan R, Crowder AT, Gouty S, Cox BM, Côté TE. Mu opioid receptor activation enhances regulator of G protein signaling 4 association with the mu opioid receptor/G protein complex in a GTP-dependent manner. J Neurochem 2015; 135:76-87. [PMID: 26119705 PMCID: PMC5034817 DOI: 10.1111/jnc.13222] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/19/2015] [Accepted: 06/25/2015] [Indexed: 11/26/2022]
Abstract
The interaction of Regulator of G protein Signaling 4 (RGS4) with the rat mu opioid receptor (MOR)/G protein complex was investigated. Solubilized MOR from rat brain membranes was immunoprecipitated in the presence of RGS4 with antibodies against the N-terminus of MOR (anti-MOR10-70 ). Activation of MOR with [D-Ala(2) , N-Me-Phe(4) , Gly(5) -ol] enkephalin (DAMGO) during immunoprecipitation caused a 150% increase in Goα and a 50% increase in RGS4 in the pellet. When 10 μM GTP was included with DAMGO, there was an additional 72% increase in RGS4 co-immunoprecipitating with MOR (p = 0.003). Guanosine 5'-O-(3-thiotriphosphate) (GTPγS) increased the amount of co-precipitating RGS4 by 93% (compared to DAMGO alone, p = 0.008), and the inclusion of GTPγS caused the ratio of MOR to RGS4 to be 1 : 1 (31 fmoles : 28 fmoles, respectively). GTPγS also increased the association of endogenous RGS4 with MOR. In His6 RGS4/Ni(2+) -NTA agarose pull down experiments, 0.3 μM GTPγS tripled the binding of Goα to His6 RGS4, whereas the addition of 100 μM GDP blocked this effect. Importantly, activation of solubilized MOR with DAMGO in the presence of 100 μM GDP and 0.3 μM GTPγS increased Goα binding to His6 RGS4/Ni(2+) -NTA agarose (p = 0.001). Regulators of G protein Signaling (RGS) shorten the time that G proteins are active. Activation of the mu opioid receptor (MOR) causes GTP to bind to and to activate Go (αoβγ). RGS4 then binds to the activated αo-GTP/MOR complex and accelerates the intrinsic GTPase of αo. After αo dissociates from MOR, RGS4 remains bound to the C-terminal region of MOR.
Collapse
Affiliation(s)
- Rema Santhappan
- Department of Pharmacology, The Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Alicia Tamara Crowder
- Department of Pharmacology, The Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Neuroscience Program, The Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Shawn Gouty
- Department of Pharmacology, The Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Brian M Cox
- Department of Pharmacology, The Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Neuroscience Program, The Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Thomas E Côté
- Department of Pharmacology, The Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Neuroscience Program, The Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
25
|
Chen CKJ. RGS Protein Regulation of Phototransduction. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 133:31-45. [PMID: 26123301 DOI: 10.1016/bs.pmbts.2015.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
First identified in yeast and worm and later in other species, the physiological importance of Regulators of G-protein Signaling (RGS) in mammals was first demonstrated at the turn of the century in mouse retinal photoreceptors, in which RGS9 is needed for timely recovery of rod phototransduction. The role of RGS in vision has also been established a synapse away in retinal depolarizing bipolar cells (DBCs), where RGS7 and RGS11 work redundantly and in a complex with Gβ5-S as GAPs for Goα in the metabotropic glutamate receptor 6 pathway situated at DBC dendritic tips. Much less is known on how RGS protein subserves vision in the rest of the visual system. The research into the roles of RGS proteins in vision holds great potential for many exciting new discoveries.
Collapse
Affiliation(s)
- Ching-Kang Jason Chen
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA; Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA; Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA.
| |
Collapse
|
26
|
Stewart A, Fisher RA. Introduction: G Protein-coupled Receptors and RGS Proteins. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 133:1-11. [PMID: 26123299 DOI: 10.1016/bs.pmbts.2015.03.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Here, we provide an overview of the role of regulator of G protein-signaling (RGS) proteins in signaling by G protein-coupled receptors (GPCRs), the latter of which represent the largest class of cell surface receptors in humans responsible for transducing diverse extracellular signals into the intracellular environment. Given that GPCRs regulate virtually every known physiological process, it is unsurprising that their dysregulation plays a causative role in many human diseases and they are targets of 40-50% of currently marketed pharmaceuticals. Activated GPCRs function as GTPase exchange factors for Gα subunits of heterotrimeric G proteins, promoting the formation of Gα-GTP and dissociated Gβγ subunits that regulate diverse effectors including enzymes, ion channels, and protein kinases. Termination of signaling is mediated by the intrinsic GTPase activity of Gα subunits leading to reformation of the inactive Gαβγ heterotrimer. RGS proteins determine the magnitude and duration of cellular responses initiated by many GPCRs by functioning as GTPase-accelerating proteins (GAPs) for specific Gα subunits. Twenty canonical mammalian RGS proteins, divided into four subfamilies, act as functional GAPs while almost 20 additional proteins contain nonfunctional RGS homology domains that often mediate interaction with GPCRs or Gα subunits. RGS protein biochemistry has been well elucidated in vitro, but the physiological functions of each RGS family member remain largely unexplored. This book summarizes recent advances employing modified model organisms that reveal RGS protein functions in vivo, providing evidence that RGS protein modulation of G protein signaling and GPCRs can be as important as initiation of signaling by GPCRs.
Collapse
Affiliation(s)
- Adele Stewart
- 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
|
27
|
Abstract
Proteins that interact coevolve their structures. When mutation disrupts the interaction, compensation by the partner occurs to restore interaction otherwise counterselection occurs. We show in this study how a destabilizing mutation in one protein is compensated by a stabilizing mutation in its protein partner and their coevolving path. The pathway in this case and likely a general principle of coevolution is that the compensatory change must tolerate both the original and derived structures with equivalence in function and activity. Evolution of the structure of signaling elements in a network is constrained by specific protein pair interactions, by requisite conformational changes, and by catalytic activity. The heterotrimeric G protein-coupled signaling is a paragon of this protein interaction/function complexity and our deep understanding of this pathway in diverse organisms lends itself to evolutionary study. Regulators of G protein Signaling (RGS) proteins accelerate the intrinsic GTP hydrolysis rate of the Gα subunit of the heterotrimeric G protein complex. An important RGS-contact site is a hydroxyl-bearing residue on the switch I region of Gα subunits in animals and most plants, such as Arabidopsis. The exception is the grasses (e.g., rice, maize, sugarcane, millets); these plants have Gα subunits that replaced the critical hydroxyl-bearing threonine with a destabilizing asparagine shown to disrupt interaction between Arabidopsis RGS protein (AtRGS1) and the grass Gα subunit. With one known exception (Setaria italica), grasses do not encode RGS genes. One parsimonious deduction is that the RGS gene was lost in the ancestor to the grasses and then recently acquired horizontally in the lineage S. italica from a nongrass monocot. Like all investigated grasses, S. italica has the Gα subunit with the destabilizing asparagine residue in the protein interface but, unlike other known grass genomes, still encodes an expressed RGS gene, SiRGS1. SiRGS1 accelerates GTP hydrolysis at similar concentration of both Gα subunits containing either the stabilizing (AtGPA1) or destabilizing (RGA1) interface residue. SiRGS1 does not use the hydroxyl-bearing residue on Gα to promote GAP activity and has a larger Gα-interface pocket fitting to the destabilizing Gα. These findings indicate that SiRGS1 adapted to a deleterious mutation on Gα using existing polymorphism in the RGS protein population.
Collapse
Affiliation(s)
- Daisuke Urano
- Department of Biology, University of North Carolina, Chapel Hill
| | - Taoran Dong
- Department of Genetics, University of Georgia
| | | | - Alan M Jones
- Department of Biology, University of North Carolina, Chapel Hill Department of Pharmacology, University of North Carolina, Chapel Hill
| |
Collapse
|
28
|
Tehseen M, Dumancic M, Briggs L, Wang J, Berna A, Anderson A, Trowell S. Functional coupling of a nematode chemoreceptor to the yeast pheromone response pathway. PLoS One 2014; 9:e111429. [PMID: 25415379 PMCID: PMC4240545 DOI: 10.1371/journal.pone.0111429] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 09/25/2014] [Indexed: 12/22/2022] Open
Abstract
Sequencing of the Caenorhabditis elegans genome revealed sequences encoding more than 1,000 G-protein coupled receptors, hundreds of which may respond to volatile organic ligands. To understand how the worm's simple olfactory system can sense its chemical environment there is a need to characterise a representative selection of these receptors but only very few receptors have been linked to a specific volatile ligand. We therefore set out to design a yeast expression system for assigning ligands to nematode chemoreceptors. We showed that while a model receptor ODR-10 binds to C. elegans Gα subunits ODR-3 and GPA-3 it cannot bind to yeast Gα. However, chimaeras between the nematode and yeast Gα subunits bound to both ODR-10 and the yeast Gβγ subunits. FIG2 was shown to be a superior MAP-dependent promoter for reporter expression. We replaced the endogenous Gα subunit (GPA1) of the Saccharomyces cerevisiae (ste2Δ sst2Δ far1Δ) triple mutant ("Cyb") with a Gpa1/ODR-3 chimaera and introduced ODR-10 as a model nematode GPCR. This strain showed concentration-dependent activation of the yeast MAP kinase pathway in the presence of diacetyl, the first time that the native form of a nematode chemoreceptor has been functionally expressed in yeast. This is an important step towards en masse de-orphaning of C. elegans chemoreceptors.
Collapse
Affiliation(s)
- Muhammad Tehseen
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, PO Box 1700, Canberra, ACT 2601, Australia
| | - Mira Dumancic
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, PO Box 1700, Canberra, ACT 2601, Australia
| | - Lyndall Briggs
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, PO Box 1700, Canberra, ACT 2601, Australia
| | - Jian Wang
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, PO Box 1700, Canberra, ACT 2601, Australia
| | - Amalia Berna
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, PO Box 1700, Canberra, ACT 2601, Australia
| | - Alisha Anderson
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, PO Box 1700, Canberra, ACT 2601, Australia
| | - Stephen Trowell
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, PO Box 1700, Canberra, ACT 2601, Australia
- * E-mail:
| |
Collapse
|
29
|
Kahn RA. GAPs: Terminator versus effector functions and the role(s) of ArfGAP1 in vesicle biogenesis. CELLULAR LOGISTICS 2014; 1:49-51. [PMID: 21686252 DOI: 10.4161/cl.1.2.15153] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Accepted: 02/14/2011] [Indexed: 11/19/2022]
Abstract
Whether your passion is to understand and reverse disease processes or "simply" a better understanding of how cells work, anyone wishing to understand cell regulation today must have a detailed and accurate understanding of regulatory GTPase mechanisms and their application to specific pathways. This is becoming increasingly difficult as the details of signaling by members of different families of GTPases and their regulators expand. But this is all the more reason to continually ask, which aspects of GTPase signaling are distinct to a GTPase or its subfamily and which are conserved throughout the superfamily? We each have slightly different views of the key aspects of GTPase signaling that are derived from the main GTPases studied in our own labs; e.g., translocation onto a membrane is an essential and integral aspect of Arf activation but not of other GTPases. However, one aspect of GTPase signaling that I had come to believe to be widespread and of general importance is not universally accepted. In fact, through my conversations at the recent FASEB summer research conference on "Arf Family GTPases" and reading of the literature in a graduate tutorial class, I realized that it is not known or accepted by the majority of researchers. The question is the role of GTPase activating proteins (GAPs) in signaling. Are they "pure" terminators of signaling or do they serve effector functions?
Collapse
Affiliation(s)
- Richard A Kahn
- Department of Biochemistry; Emory University School of Medicine; Atlanta, GA USA
| |
Collapse
|
30
|
Weston C, Poyner D, Patel V, Dowell S, Ladds G. Investigating G protein signalling bias at the glucagon-like peptide-1 receptor in yeast. Br J Pharmacol 2014; 171:3651-65. [PMID: 24712679 PMCID: PMC4128063 DOI: 10.1111/bph.12716] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 03/20/2014] [Accepted: 03/29/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE The glucagon-like peptide 1 (GLP-1) receptor performs an important role in glycaemic control, stimulating the release of insulin. It is an attractive target for treating type 2 diabetes. Recently, several reports of adverse side effects following prolonged use of GLP-1 receptor therapies have emerged: most likely due to an incomplete understanding of signalling complexities. EXPERIMENTAL APPROACH We describe the expression of the GLP-1 receptor in a panel of modified yeast strains that couple receptor activation to cell growth via single Gα/yeast chimeras. This assay enables the study of individual ligand-receptor G protein coupling preferences and the quantification of the effect of GLP-1 receptor ligands on G protein selectivity. KEY RESULTS The GLP-1 receptor functionally coupled to the chimeras representing the human Gαs, Gαi and Gαq subunits. Calculation of the dissociation constant for a receptor antagonist, exendin-3 revealed no significant difference between the two systems. We obtained previously unobserved differences in G protein signalling bias for clinically relevant therapeutic agents, liraglutide and exenatide; the latter displaying significant bias for the Gαi pathway. We extended the use of the system to investigate small-molecule allosteric compounds and the closely related glucagon receptor. CONCLUSIONS AND IMPLICATIONS These results provide a better understanding of the molecular events involved in GLP-1 receptor pleiotropic signalling and establish the yeast platform as a robust tool to screen for more selective, efficacious compounds acting at this important class of receptors in the future.
Collapse
Affiliation(s)
- C Weston
- Division of Biomedical Cell Biology, Warwick Medical School, University of Warwick, Coventry, UK
| | | | | | | | | |
Collapse
|
31
|
Jiang N, Xue R, Bu F, Tong X, Qiang J, Liu R. Decreased RGS6 expression is associated with poor prognosis in pancreatic cancer patients. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:4120-4127. [PMID: 25120791 PMCID: PMC4129026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 06/25/2014] [Indexed: 06/03/2023]
Abstract
Regulator of G-protein signaling 6 (RGS6), a member of a family of RGS proteins, has been reported to involve in multiple processes during tumor development. However, its role in pancreatic cancer has not been studied yet. In this study, we aimed to investigate the expression of RGS6 in pancreatic cancer and its role in predicting outcomes of patients with pancreatic cancer. We first measured the expression of RGS6 mRNA in 20 cases of tumor tissues and matched adjacent non-tumorous tissues by quantitative real-time PCR and examined RGS6 protein by immunohistochemistry in tissue microarrays containing 90 tumor and 90 paired adjacent non-tumor tissues. Decreased RGS6 mRNA detected in primary tumor, compared with their non-tumor counterparts. In addition, decreased RGS6 protein expression was associated with tumor differentiation (P = 0.027), pT classification (P = 0.034), smoking status (P = 0.041) and a poor survival (P = 0.007). Cox proportional hazards regression modeling analysis revealed that lymph node metastasis (P = 0.001; hazard ratio, 2.347, 95% CI, 1.387-3.972), tumor differentiation (P = 0.015; hazard ratio, 0.505, 95% CI, 0.291-0.876) and RGS6 expression (P = 0.048; hazard ratio, 0.567, 95% CI, 0.324-0.994) were three independent prognostic factors. Taken together, these date demonstrate that RGS6 decreases in tumor tissue and may serve as a novel biomarker for outcomes in pancreatic cancer patients and be a potential therapeutic target potential therapeutic target.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Surgical Oncology, Chinese People’s Liberation Army (PLA) General Hospital28 Fuxing Road, Beijing 100853, China
| | - Ruihua Xue
- Department of Surgical Oncology, Chinese People’s Liberation Army (PLA) General Hospital28 Fuxing Road, Beijing 100853, China
| | - Fangfang Bu
- Department of Cancer Center, Chinese People’s Liberation Army (PLA) General Hospital28 Fuxing Road, Beijing 100853, China
| | - Xin Tong
- Department of Cancer Center, Chinese People’s Liberation Army (PLA) General Hospital28 Fuxing Road, Beijing 100853, China
| | - Jiankun Qiang
- Department of Cancer Center, Chinese People’s Liberation Army (PLA) General Hospital28 Fuxing Road, Beijing 100853, China
| | - Rong Liu
- Department of Surgical Oncology, Chinese People’s Liberation Army (PLA) General Hospital28 Fuxing Road, Beijing 100853, China
| |
Collapse
|
32
|
Williams JW, Yau D, Sethakorn N, Kach J, Reed EB, Moore TV, Cannon J, Jin X, Xing H, Muslin AJ, Sperling AI, Dulin NO. RGS3 controls T lymphocyte migration in a model of Th2-mediated airway inflammation. Am J Physiol Lung Cell Mol Physiol 2013; 305:L693-701. [PMID: 24077945 DOI: 10.1152/ajplung.00214.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
T cell migration toward sites of antigen exposure is mediated by G protein signaling and is a key function in the development of immune responses. Regulators of G protein signaling (RGS) proteins modulate G protein signaling; however, their role in the regulation of adaptive immune responses has not been thoroughly explored. Herein we demonstrated abundant expression of the Gi/Gq-specific RGS3 in activated T cells, and that diminished RGS3 expression in a T cell thymoma increased cytokine-induced migration. To examine the role of endogenous RGS3 in vivo, mice deficient in the RGS domain (RGS3(ΔRGS)) were generated and tested in an experimental model of asthma. Compared with littermate controls, the inflammation in the RGS3(ΔRGS) mice was characterized by increased T cell numbers and the striking development of perivascular lymphoid structures. Surprisingly, while innate inflammatory cells were also increased in the lungs of RGS3(ΔRGS) mice, eosinophil numbers and Th2 cytokine production were equivalent to control mice. In contrast, T cell numbers in the draining lymph nodes (dLN) were reduced in the RGS3(ΔRGS), demonstrating a redistribution of T cells from the dLN to the lungs via increased RGS3(ΔRGS) T cell migration. Together these novel findings show a nonredundant role for endogenous RGS3 in controlling T cell migration in vitro and in an in vivo model of inflammation.
Collapse
Affiliation(s)
- Jesse W Williams
- Section of Pulmonary and Critical Care, Dept. of Medicine, The Univ. of Chicago, 5841 S. Maryland Ave., MC6076, Rm. M-648, Chicago, IL 60637.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Clement ST, Dixit G, Dohlman HG. Regulation of yeast G protein signaling by the kinases that activate the AMPK homolog Snf1. Sci Signal 2013; 6:ra78. [PMID: 24003255 DOI: 10.1126/scisignal.2004143] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Extracellular signals, such as nutrients and hormones, cue intracellular pathways to produce adaptive responses. Often, cells must coordinate their responses to multiple signals to produce an appropriate outcome. We showed that components of a glucose-sensing pathway acted on components of a heterotrimeric guanine nucleotide-binding protein (G protein)-mediated pheromone signaling pathway in the yeast Saccharomyces cerevisiae. We demonstrated that the G protein α subunit Gpa1 was phosphorylated in response to conditions of reduced glucose availability and that this phosphorylation event contributed to reduced pheromone-dependent stimulation of mitogen-activated protein kinases, gene transcription, cell morphogenesis, and mating efficiency. We found that Elm1, Sak1, and Tos3, the kinases that phosphorylate Snf1, the yeast homolog of adenosine monophosphate-activated protein kinase (AMPK), in response to limited glucose availability, also phosphorylated Gpa1 and contributed to the diminished mating response. Reg1, the regulatory subunit of the phosphatase PP1 that acts on Snf1, was likewise required to reverse the phosphorylation of Gpa1 and to maintain the mating response. Thus, the same kinases and phosphatase that regulate Snf1 also regulate Gpa1. More broadly, these results indicate that the pheromone signaling and glucose-sensing pathways communicate directly to coordinate cell behavior.
Collapse
Affiliation(s)
- Sarah T Clement
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
34
|
Bond M, Croft W, Tyson R, Bretschneider T, Davey J, Ladds G. Quantitative analysis of human ras localization and function in the fission yeast Schizosaccharomyces pombe. Yeast 2013; 30:145-56. [PMID: 23447405 DOI: 10.1002/yea.2949] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 02/06/2013] [Accepted: 02/12/2013] [Indexed: 12/30/2022] Open
Abstract
Ras signalling is central to fundamental and diverse cellular processes. In higher eukaryotes ras signalling is highly complex, involving multiple isoforms, regulatory proteins and effectors. As a consequence, the study of ras activity in mammalian systems presents a number of technical challenges. The model organism Schizosaccharomyces pombe has previously proved a key system for the study of human signalling components and provides an ideal model for the study of ras, as it contains just one ras protein (Ras1p), which is non-essential and controls a number of downstream processes. Here we present data demonstrating the quantitative analysis of three distinct Ras1-related signalling outputs, utilizing the three most abundant human ras isoforms, H-Ras, N-Ras and K-Ras4B, in Sz. pombe. Further, we have characterized the localization of these three human ras isoforms in Sz. pombe, utilizing quantitative image analysis techniques. These data indicate that all three human ras isoforms are functional in fission yeast, displaying differing localization patterns which correlate strongly with function in the regulation of pheromone response and cell shape. These data demonstrate that such yeast strains could provide powerful tools for the investigation of ras biology, and potentially in the development of cancer therapies.
Collapse
Affiliation(s)
- Michael Bond
- Division of Clinical Sciences, Warwick Medical School, Coventry, CV4 7AL, UK; Medical Research Council Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | | | | | | | | | | |
Collapse
|
35
|
Sethakorn N, Dulin NO. RGS expression in cancer: oncomining the cancer microarray data. J Recept Signal Transduct Res 2013; 33:166-71. [DOI: 10.3109/10799893.2013.773450] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
36
|
Fernandez-Piñar P, Alemán A, Sondek J, Dohlman HG, Molina M, Martín H. The Salmonella Typhimurium effector SteC inhibits Cdc42-mediated signaling through binding to the exchange factor Cdc24 in Saccharomyces cerevisiae. Mol Biol Cell 2012; 23:4430-43. [PMID: 23015760 PMCID: PMC3496616 DOI: 10.1091/mbc.e12-03-0243] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Expression of the Salmonella effector SteC in yeast leads to down-regulation of the mating and HOG pathways by Cdc42 inhibition. This is mediated by the SteC N-terminal domain through binding to the GEF Cdc24. SteC alters Cdc24 localization and also interacts with human GEF Vav1, suggesting that SteC could target Cdc42 function in host cells. Intracellular survival of Salmonella relies on the activity of proteins translocated into the host cell by type III secretion systems (T3SS). The protein kinase activity of the T3SS effector SteC is required for F-actin remodeling in host cells, although no SteC target has been identified so far. Here we show that expression of the N-terminal non-kinase domain of SteC down-regulates the mating and HOG pathways in Saccharomyces cerevisiae. Epistasis analyses using constitutively active components of these pathways indicate that SteC inhibits signaling at the level of the GTPase Cdc42. We demonstrate that SteC interacts through its N-terminal domain with the catalytic domain of Cdc24, the sole S. cerevisiae Cdc42 guanine nucleotide exchange factor (GEF). SteC also binds to the human Cdc24-like GEF protein Vav1. Moreover, expression of human Cdc42 suppresses growth inhibition caused by SteC. Of interest, the N-terminal SteC domain alters Cdc24 cellular localization, preventing its nuclear accumulation. These data reveal a novel functional domain within SteC, raising the possibility that this effector could also target GTPase function in mammalian cells. Our results also highlight the key role of the Cdc42 switch in yeast mating and HOG pathways and provide a new tool to study the functional consequences of Cdc24 localization.
Collapse
Affiliation(s)
- Pablo Fernandez-Piñar
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid, and Instituto Ramón y Cajal de Investigaciones Sanitarias, 28040 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
37
|
Ramanujam R, Yishi X, Liu H, Naqvi NI. Structure-function analysis of Rgs1 in Magnaporthe oryzae: role of DEP domains in subcellular targeting. PLoS One 2012; 7:e41084. [PMID: 22927898 PMCID: PMC3426613 DOI: 10.1371/journal.pone.0041084] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 06/18/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Rgs1, a prototypical Regulator of G protein Signaling, negatively modulates the cyclic AMP pathway thereby influencing various aspects of asexual development and pathogenesis in the rice-blast fungus Magnaporthe oryzae. Rgs1 possesses tandem DEP motifs (termed DEP-A and DEP-B; for Dishevelled, Egl-10, Pleckstrin) at the N-terminus, and a Gα-GTP interacting RGS catalytic core domain at the C-terminus. In this study, we focused on gaining further insights into the mechanisms of Rgs1 regulation and subcellular localization by characterizing the role(s) of the individual domains and the full-length protein during asexual development and pathogenesis in Magnaporthe. METHODOLOGY/PRINCIPAL FINDINGS Utilizing western blot analysis and specific antisera against the N- and C-terminal halves of Rgs1, we identify and report the in vivo endoproteolytic processing/cleavage of full-length Rgs1 that yields an N-terminal DEP and a RGS core domain. Independent expression of the resultant DEP-DEP half (N-Rgs1) or RGS core (C-Rgs1) fragments, failed to complement the rgs1Δ defects in colony morphology, aerial hyphal growth, surface hydrophobicity, conidiation, appressorium formation and infection. Interestingly, the full-length Rgs1-mCherry, as well as the tagged N-terminal DEP domains (individually or in conjunction) localized to distinct punctate vesicular structures in the cytosol, while the catalytic RGS core motif was predominantly vacuolar. CONCLUSIONS/SIGNIFICANCE Based on our data from sequence alignments, immuno-blot and microscopic analysis, we propose that the post-translational proteolytic processing of Rgs1 and the vacuolar sequestration of the catalytic RGS domain represents an important means of down regulating Rgs1 function and thus forming an additional and alternative means of regulating G protein signaling in Magnaporthe. We further hypothesize the prevalence of analogous mechanisms functioning in other filamentous fungi. Furthermore, we conclusively assign a specific vesicular/membrane targeting function for the N-terminal DEP domains of Rgs1 in the rice-blast fungus.
Collapse
Affiliation(s)
- Ravikrishna Ramanujam
- Fungal Patho-Biology Group, Temasek Life Sciences Laboratory, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Xu Yishi
- Fungal Patho-Biology Group, Temasek Life Sciences Laboratory, Singapore, Singapore
| | - Hao Liu
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Naweed I. Naqvi
- Fungal Patho-Biology Group, Temasek Life Sciences Laboratory, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
38
|
Kach J, Sethakorn N, Dulin NO. A finer tuning of G-protein signaling through regulated control of RGS proteins. Am J Physiol Heart Circ Physiol 2012; 303:H19-35. [PMID: 22542620 DOI: 10.1152/ajpheart.00764.2011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regulators of G-protein signaling (RGS) proteins are GTPase-activating proteins (GAP) for various Gα subunits of heterotrimeric G proteins. Through this mechanism, RGS proteins regulate the magnitude and duration of G-protein-coupled receptor signaling and are often referred to as fine tuners of G-protein signaling. Increasing evidence suggests that RGS proteins themselves are regulated through multiple mechanisms, which may provide an even finer tuning of G-protein signaling and crosstalk between G-protein-coupled receptors and other signaling pathways. This review summarizes the current data on the control of RGS function through regulated expression, intracellular localization, and covalent modification of RGS proteins, as related to cell function and the pathogenesis of diseases.
Collapse
Affiliation(s)
- Jacob Kach
- Department of Medicine, University of Chicago, Illinois, 60637, USA
| | | | | |
Collapse
|
39
|
Transcriptome and functional analysis of mating in the basidiomycete Schizophyllum commune. EUKARYOTIC CELL 2011; 11:571-89. [PMID: 22210832 DOI: 10.1128/ec.05214-11] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In this study, we undertook a functional characterization and transcriptome analysis that enabled a comprehensive study of the mating type loci of the mushroom Schizophyllum commune. Induced expression of both the bar2 receptor and the bap2(2) pheromone gene within 6 to 12 h after mates' contact was demonstrated by quantitative real-time PCR. Similar temporal expression patterns were confirmed for the allelic bbr1 receptor and bbp1 pheromone-encoding genes by Northern hybridization. Interestingly, the fusion of clamp connections to the subterminal cell was delayed in mating interactions in which one of the compatible partners expressed the bar2 receptor with a truncated C terminus. This developmental delay allowed the visualization of a green fluorescent protein (Gfp)-labeled truncated receptor at the cell periphery, consistent with a localization in the plasma membrane of unfused pseudoclamps. This finding does not support hypotheses envisioning a receptor localization to the nuclear membrane facilitating recognition between the two different nuclei present in each dikaryotic cell. Rather, Gfp fluorescence observed in such pseudoclamps indicated a role of receptor-pheromone interaction in clamp fusion. Transcriptome changes associated with mating interactions were analyzed in order to identify a role for pheromone-receptor interactions. We detected a total of 89 genes that were transcriptionally regulated in a mating type locus A-dependent manner, employing a cutoff of 5-fold changes in transcript abundance. Upregulation in cell cycle-related genes and downregulation of genes involved in metabolism were seen with this set of experiments. In contrast, mating type locus B-dependent transcriptome changes were observed in 208 genes, with a specific impact on genes related to cell wall and membrane metabolism, stress response, and the redox status of the cell.
Collapse
|
40
|
Kimple AJ, Bosch DE, Giguère PM, Siderovski DP. Regulators of G-protein signaling and their Gα substrates: promises and challenges in their use as drug discovery targets. Pharmacol Rev 2011; 63:728-49. [PMID: 21737532 PMCID: PMC3141876 DOI: 10.1124/pr.110.003038] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Because G-protein coupled receptors (GPCRs) continue to represent excellent targets for the discovery and development of small-molecule therapeutics, it is posited that additional protein components of the signal transduction pathways emanating from activated GPCRs themselves are attractive as drug discovery targets. This review considers the drug discovery potential of two such components: members of the "regulators of G-protein signaling" (RGS protein) superfamily, as well as their substrates, the heterotrimeric G-protein α subunits. Highlighted are recent advances, stemming from mouse knockout studies and the use of "RGS-insensitivity" and fast-hydrolysis mutations to Gα, in our understanding of how RGS proteins selectively act in (patho)physiologic conditions controlled by GPCR signaling and how they act on the nucleotide cycling of heterotrimeric G-proteins in shaping the kinetics and sensitivity of GPCR signaling. Progress is documented regarding recent activities along the path to devising screening assays and chemical probes for the RGS protein target, not only in pursuits of inhibitors of RGS domain-mediated acceleration of Gα GTP hydrolysis but also to embrace the potential of finding allosteric activators of this RGS protein action. The review concludes in considering the Gα subunit itself as a drug target, as brought to focus by recent reports of activating mutations to GNAQ and GNA11 in ocular (uveal) melanoma. We consider the likelihood of several strategies for antagonizing the function of these oncogene alleles and their gene products, including the use of RGS proteins with Gα(q) selectivity.
Collapse
Affiliation(s)
- Adam J Kimple
- Department of Pharmacology, UNC Neuroscience Center, UNC School of Medicine, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Suite 4010, Chapel Hill, NC 27599-7365, USA
| | | | | | | |
Collapse
|
41
|
Heterologous expression of GPCRs in fission yeast. Methods Mol Biol 2011. [PMID: 21607855 DOI: 10.1007/978-1-61779-126-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
In this chapter, we describe methods to heterologously express G protein-coupled receptors (GPCRs) in the fission yeast Schizosaccharomyces (Sz.) pombe. GPCRs regulate a diverse range of biological processes in all eukaryotic cells, including plants, insects, humans, and yeast. The high degree of conservation between GPCRs from different organisms has facilitated the development of a large number of model systems to enable study of this pharmaceutically important family of cell-surface receptors. Of the many model systems available for investigating GPCRs, yeast have proven to be one of the more attractive. Yeasts' amenability to both genetic and biochemical manipulation, a reduced number of endogenous GPCRs and their relative low culturing costs has facilitated their use in many high-throughput drug screens. Given the high number of detailed methods relating to the expression of GPCRs within budding yeast, we have focused our attention on the use of fission yeast as a model system. We describe the methods used and provide examples from our own experiences of expressing a number of human GPCRs in Sz. pombe cells.
Collapse
|
42
|
Sjögren B. Regulator of G protein signaling proteins as drug targets: current state and future possibilities. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2011; 62:315-47. [PMID: 21907914 DOI: 10.1016/b978-0-12-385952-5.00002-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Regulators of G protein signaling (RGS) proteins have emerged in the past two decades as novel drug targets in many areas of research. Their importance in regulating signaling via G protein-coupled receptors has become evident as numerous studies have been published on the structure and function of RGS proteins. A number of genetic models have also been developed, demonstrating the potential clinical importance of RGS proteins in various disease states, including central nervous system disorders, cardiovascular disease, diabetes, and several types of cancer. Apart from their classical mechanism of action as GTPase-activating proteins (GAPs), RGS proteins can also serve other noncanonical functions. This opens up a new approach to targeting RGS proteins in drug discovery as the view on the function of these proteins is constantly evolving. This chapter summarizes the latest development in RGS protein drug discovery with special emphasis on noncanonical functions and regulatory mechanisms of RGS protein expression. As more reports are being published on this group of proteins, it is becoming clear that modulation of GAP activity might not be the only way to therapeutically target RGS proteins.
Collapse
Affiliation(s)
- Benita Sjögren
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
43
|
Kaur K, Kehrl JM, Charbeneau RA, Neubig RR. RGS-insensitive Gα subunits: probes of Gα subtype-selective signaling and physiological functions of RGS proteins. Methods Mol Biol 2011; 756:75-98. [PMID: 21870221 DOI: 10.1007/978-1-61779-160-4_4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The Regulator of G protein Signaling (RGS) proteins were identified as a family in 1996 and humans have more than 30 such proteins. Their best known function is to suppress G Protein-Coupled Receptors (GPCR) signaling by increasing the rate of Gα turnoff through stimulation of GTPase activity (i.e., GTPase acceleration protein or GAP activity). The GAP activity of RGS proteins on the Gαi and Gαq family of G proteins can terminate signals initiated by both α and βγ subunits. RGS proteins also serve as scaffolds, assembling signal-regulating modules. Understanding the physiological roles of RGS proteins is of great importance, as GPCRs are major targets for drug development. The traditional method of using RGS knockout mice has provided some information about the role of RGS proteins but in many cases effects are modest, perhaps because of redundancy in RGS protein function. As an alternative approach, we have utilized a glycine-to-serine mutation in the switch 1 region of Gα subunits that prevents RGS binding. The mutation has no known effects on Gα binding to receptor, Gβγ, or effectors. Alterations in function resulting from the G>S mutation imply a role for both the specific mutated Gα subunit and its regulation by RGS protein activity. Mutant rodents expressing these G>S mutant Gα subunits have strong phenotypes and provide important information about specific physiological functions of Gαi2 and Gαo and their control by RGS. The conceptual framework behind this approach and a summary of recent results is presented in this chapter.
Collapse
Affiliation(s)
- Kuljeet Kaur
- Department of Pharmacology, The University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | | |
Collapse
|
44
|
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
|
45
|
Yang J, Huang J, Chatterjee TK, Twait E, Fisher RA. A novel mechanism involving coordinated regulation of nuclear levels and acetylation of NF-YA and Bcl6 activates RGS4 transcription. J Biol Chem 2010; 285:29760-9. [PMID: 20630860 PMCID: PMC2943308 DOI: 10.1074/jbc.m110.121459] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 07/13/2010] [Indexed: 11/06/2022] Open
Abstract
Neuronally enriched RGS4 plays a critical role attenuating G protein signaling in brain, although the mechanisms regulating RGS4 expression are unknown. Here we describe a novel mechanism for transcriptional activation of RGS4 in neuron-like PC6 cells, where RGS4 is markedly induced during confluence-induced growth arrest. Transcriptional activation of RGS4 in confluent PC6 cells was accompanied by impaired G(i/o)-dependent MAPK activation. In the human RGS4 gene promoter, we identified three phylogenetically conserved cis-elements: an inverted CCAAT box element (ICE), a cAMP response element, and a B-cell lymphoma 6 (Bcl6)-binding site. The ICE and the cAMP response element mediate activation, and the Bcl6 site mediates repression of RGS4 transcription. Activation of RGS4 transcription in confluent PC6 cells is accompanied by increases in NF-YA and C/EBPβ and decreases in Bcl6 levels in the nucleus. Increases in NF-YA and C/EBPβ lead to their increased binding to the RGS4 promoter in vivo, and dominant negative forms of these proteins repressed RGS4 promoter activity. Acetylation of NF-YA and Bcl6 were increased in postconfluent cells. Trichostatin A stimulation of RGS4 promoter activity, accompanied by increased binding of NF-YA and decreased binding of Bcl6 to the promoter, was abolished by mutation of the ICE and enhanced by mutation of the Bcl6 site. These findings demonstrate a dynamic and coordinated regulation of nuclear levels and acetylation status of trans-acting factors critical in determining the off/on state of the RGS4 promoter.
Collapse
Affiliation(s)
| | - Jie Huang
- From the Departments of Pharmacology and
| | - Tapan K. Chatterjee
- the Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio 45221
| | - Erik Twait
- Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | | |
Collapse
|
46
|
Freisinger CM, Fisher RA, Slusarski DC. Regulator of g protein signaling 3 modulates wnt5b calcium dynamics and somite patterning. PLoS Genet 2010; 6:e1001020. [PMID: 20628572 PMCID: PMC2900303 DOI: 10.1371/journal.pgen.1001020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 06/07/2010] [Indexed: 01/01/2023] Open
Abstract
Vertebrate development requires communication among cells of the embryo in order to define the body axis, and the Wnt-signaling network plays a key role in axis formation as well as in a vast array of other cellular processes. One arm of the Wnt-signaling network, the non-canonical Wnt pathway, mediates intracellular calcium release via activation of heterotrimeric G proteins. Regulator of G protein Signaling (RGS) proteins can accelerate inactivation of G proteins by acting as G protein GTPase-activating proteins (GAPs), however, the possible role of RGS proteins in non-canonical Wnt signaling and development is not known. Here, we identify rgs3 as having an overlapping expression pattern with wnt5b in zebrafish and reveal that individual knockdown of either rgs3 or wnt5b gene function produces similar somite patterning defects. Additionally, we describe endogenous calcium release dynamics in developing zebrafish somites and determine that both rgs3 and wnt5b function are required for appropriate frequency and amplitude of calcium release activity. Using rescue of gene knockdown and in vivo calcium imaging assays, we demonstrate that the activity of Rgs3 requires its ability to interact with Gα subunits and function as a G protein GAP. Thus, Rgs3 function is necessary for appropriate frequency and amplitude of calcium release during somitogenesis and is downstream of Wnt5 activity. These results provide the first evidence for an essential developmental role of RGS proteins in modulating the duration of non-canonical Wnt signaling. Vertebrate development requires communication among cells in order to define the body axis (front/back, head/tail, or left/right). Secreted factors such as Wnts play key roles in a vast array of cellular processes, including patterning of the body axis. One arm of the Wnt-signaling network, the non-canonical pathway, mediates intracellular calcium release via activation of heterotrimeric G proteins. Regulator of G protein Signaling (RGS) proteins can accelerate inactivation of G proteins by acting as G protein GAPs and are uniquely situated to control the amplitude of a Wnt signal. Here, we combine cellular, molecular, and genetic analyses with high resolution calcium imaging to identify a role for RGS modulation of Wnt-mediated calcium release dynamics and developmental patterning events. We find that loss of rgs3 gene function produced body patterning defects like those observed with loss of wnt5b gene function. Analysis of endogenous calcium release dynamics in developing zebrafish revealed that both rgs3 and wnt5b are required for appropriate frequency and amplitude of calcium release. Our results provide new evidence that a member of the RGS protein family is essential for modulating the non-canonical Wnt network to assure normal tissue patterning during development.
Collapse
Affiliation(s)
| | - Rory A. Fisher
- Department of Pharmacology, University of Iowa College of Medicine, Iowa City, Iowa, United States of America
| | - Diane C. Slusarski
- Department of Biology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
47
|
Regulators of G Protein Signaling Proteins as Targets for Drug Discovery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 91:81-119. [DOI: 10.1016/s1877-1173(10)91004-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
48
|
Tesmer JJG. Structure and function of regulator of G protein signaling homology domains. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 86:75-113. [PMID: 20374714 DOI: 10.1016/s1877-1173(09)86004-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
All regulator of G protein signaling (RGS) proteins contain a conserved domain of approximately 130 amino acids that binds to activated heterotrimeric G protein α subunits (Gα) and accelerates their rate of GTP hydrolysis. Homologous domains are found in at least six other protein families, including a family of Rho guanine nucleotide exchange factors (RhoGEFs) and the G protein-coupled receptor kinases (GRKs). Although some of the RhoGEF and GRK RGS-like domains can also bind to activated Gα subunits, they do so in distinct ways and with much lower levels of GTPase activation. In other protein families, the domains have as of yet no obvious relationship to heterotrimeric G protein signaling. These RGS homology (RH) domains are now recognized as mediators of extraordinarily diverse protein-protein interactions. Through these interactions, they play roles that range from enzyme to molecular scaffold to signal transducing module. In this review, the atomic structures of RH domains from RGS proteins, Axins, RhoGEFs, and GRKs are compared in light of what is currently known about their functional roles.
Collapse
Affiliation(s)
- John J G Tesmer
- Department of Pharmacology, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109‐2216, USA
| |
Collapse
|
49
|
Smith B, Hill C, Godfrey EL, Rand D, van den Berg H, Thornton S, Hodgkin M, Davey J, Ladds G. Dual positive and negative regulation of GPCR signaling by GTP hydrolysis. Cell Signal 2009; 21:1151-60. [PMID: 19285552 DOI: 10.1016/j.cellsig.2009.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Accepted: 03/04/2009] [Indexed: 10/21/2022]
Abstract
G protein-coupled receptors (GPCRs) regulate a variety of intracellular pathways through their ability to promote the binding of GTP to heterotrimeric G proteins. Regulator of G protein signaling (RGS) proteins increases the intrinsic GTPase activity of Galpha-subunits and are widely regarded as negative regulators of G protein signaling. Using yeast we demonstrate that GTP hydrolysis is not only required for desensitization, but is essential for achieving a high maximal (saturated level) response. Thus RGS-mediated GTP hydrolysis acts as both a negative (low stimulation) and positive (high stimulation) regulator of signaling. To account for this we generated a new kinetic model of the G protein cycle where Galpha(GTP) enters an inactive GTP-bound state following effector activation. Furthermore, in vivo and in silico experimentation demonstrates that maximum signaling output first increases and then decreases with RGS concentration. This unimodal, non-monotone dependence on RGS concentration is novel. Analysis of the kinetic model has revealed a dynamic network motif that shows precisely how inclusion of the inactive GTP-bound state for the Galpha produces this unimodal relationship.
Collapse
Affiliation(s)
- Benjamin Smith
- Molecular Organization and Assembly of Cells Centre, University of Warwick, Coventry, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
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]
|