1
|
Tennakoon M, Thotamune W, Payton JL, Karunarathne A. CaaX-motif-adjacent residues influence G protein gamma (Gγ) prenylation under suboptimal conditions. J Biol Chem 2023; 299:105269. [PMID: 37739036 PMCID: PMC10590752 DOI: 10.1016/j.jbc.2023.105269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/24/2023] Open
Abstract
Prenylation is an irreversible post-translational modification that supports membrane interactions of proteins involved in various cellular processes, including migration, proliferation, and survival. Dysregulation of prenylation contributes to multiple disorders, including cancers and vascular and neurodegenerative diseases. Prenyltransferases tether isoprenoid lipids to proteins via a thioether linkage during prenylation. Pharmacological inhibition of the lipid synthesis pathway by statins is a therapeutic approach to control hyperlipidemia. Building on our previous finding that statins inhibit membrane association of G protein γ (Gγ) in a subtype-dependent manner, we investigated the molecular reasoning for this differential inhibition. We examined the prenylation of carboxy-terminus (Ct) mutated Gγ in cells exposed to Fluvastatin and prenyl transferase inhibitors and monitored the subcellular localization of fluorescently tagged Gγ subunits and their mutants using live-cell confocal imaging. Reversible optogenetic unmasking-masking of Ct residues was used to probe their contribution to prenylation and membrane interactions of the prenylated proteins. Our findings suggest that specific Ct residues regulate membrane interactions of the Gγ polypeptide, statin sensitivity, and extent of prenylation. Our results also show a few hydrophobic and charged residues at the Ct are crucial determinants of a protein's prenylation ability, especially under suboptimal conditions. Given the cell and tissue-specific expression of different Gγ subtypes, our findings indicate a plausible mechanism allowing for statins to differentially perturb heterotrimeric G protein signaling in cells depending on their Gγ-subtype composition. Our results may also provide molecular reasoning for repurposing statins as Ras oncogene inhibitors and the failure of using prenyltransferase inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Mithila Tennakoon
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri, USA; Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA; Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, Missouri, USA
| | - Waruna Thotamune
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri, USA; Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA; Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, Missouri, USA
| | - John L Payton
- Department of Chemistry, Kenyon College, Gambier, Ohio, USA
| | - Ajith Karunarathne
- Department of Chemistry, Saint Louis University, Saint Louis, Missouri, USA; Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA; Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, Missouri, USA.
| |
Collapse
|
2
|
Ren Z, Zhang H, Yu H, Zhu X, Lin J. Roles of four targets in the pathogenesis of graves' orbitopathy. Heliyon 2023; 9:e19250. [PMID: 37810014 PMCID: PMC10558314 DOI: 10.1016/j.heliyon.2023.e19250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/29/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023] Open
Abstract
Graves' orbitopathy (GO) is an autoimmune disease that involves complex immune systems. The mainstays of clinical management for this disease are surgery, targeted drugs therapy, and no-targeted drugs drug therapy. targeted drugs can improve therapeutic efficacy and enhance the quality of life for GO patients. However, as a second-line treatment for GO, targeted drugs such as tocilizumab and rituximab have very limited therapeutic effects and may be accompanied by side effects. The introduction of Teprotumumab, which targets IGF-IR, has made significant progress in the clinical management of GO. The pathophysiology of GO still remains uncertain as it involves a variety of immune cells and fibroblast interactions as well as immune responses to relevant disease targets of action. Therfore, learning more about immune response feedback pathways and potential targets of action will assist in the treatment of GO. In this discussion, we explore the pathogenesis of GO and relevant work, and highlight four potential targets for GO: Interleukin-23 receptor (IL-23 R), Leptin receptor (LepR), Orbital fibroblast activating factors, and Plasminogen activator inhibitor-1 (PAI-1). A deeper understanding of the pathogenesis of GO and the role of potential target signaling pathways is crucial for effective treatment of this disease.
Collapse
Affiliation(s)
- Ziqiang Ren
- College of Life Sciences, Yantai University, Shandong, China
- Fengjin Biomedical Co., Ltd, Shandong, China
| | - Hailing Zhang
- College of Life Sciences, Yantai University, Shandong, China
| | - Haiwen Yu
- College of Life Sciences, Yantai University, Shandong, China
| | - Xiqiang Zhu
- Fengjin Biomedical Co., Ltd, Shandong, China
| | - Jian Lin
- College of Life Sciences, Yantai University, Shandong, China
| |
Collapse
|
3
|
Su X, Pang YT, Li W, Gumbart JC, Kelley J, Torres M. N-terminal intrinsic disorder is an ancestral feature of Gγ subunits that influences the balance between different Gβγ signaling axes in yeast. J Biol Chem 2023; 299:104947. [PMID: 37354971 PMCID: PMC10393545 DOI: 10.1016/j.jbc.2023.104947] [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: 03/23/2023] [Revised: 06/02/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
Activated G protein-coupled receptors promote the dissociation of heterotrimeric G proteins into Gα and Gβγ subunits that bind to effector proteins to drive intracellular signaling responses. In yeast, Gβγ subunits coordinate the simultaneous activation of multiple signaling axes in response to mating pheromones, including MAP kinase (MAPK)-dependent transcription, cell polarization, and cell cycle arrest responses. The Gγ subunit in this complex contains an N-terminal intrinsically disordered region that governs Gβγ-dependent signal transduction in yeast and mammals. Here, we demonstrate that N-terminal intrinsic disorder is likely an ancestral feature that has been conserved across different Gγ subtypes and organisms. To understand the functional contribution of structural disorder in this region, we introduced precise point mutations that produce a stepwise disorder-to-order transition in the N-terminal tail of the canonical yeast Gγ subunit, Ste18. Mutant tail structures were confirmed using circular dichroism and molecular dynamics and then substituted for the wildtype gene in yeast. We find that increasing the number of helix-stabilizing mutations, but not isometric mutation controls, has a negative and proteasome-independent effect on Ste18 protein levels as well as a differential effect on pheromone-induced levels of active MAPK/Fus3, but not MAPK/Kss1. When expressed at wildtype levels, we further show that mutants with an alpha-helical N terminus exhibit a counterintuitive shift in Gβγ signaling that reduces active MAPK/Fus3 levels whilst increasing cell polarization and cell cycle arrest. These data reveal a role for Gγ subunit intrinsically disordered regions in governing the balance between multiple Gβγ signaling axes.
Collapse
Affiliation(s)
- Xinya Su
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Yui Tik Pang
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Wei Li
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA; Southeast Center for Mathematics and Biology, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - J C Gumbart
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Joshua Kelley
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Matthew Torres
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA; Southeast Center for Mathematics and Biology, Georgia Institute of Technology, Atlanta, Georgia, USA.
| |
Collapse
|
4
|
Tennakoon M, Thotamune W, Payton JL, Karunarathne A. CaaX-motif adjacent residues control G protein prenylation under suboptimal conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.04.547731. [PMID: 37461501 PMCID: PMC10349941 DOI: 10.1101/2023.07.04.547731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Prenylation is a universal and irreversible post-translational modification that supports membrane interactions of proteins involved in various cellular processes, including migration, proliferation, and survival. Thus, dysregulation of prenylation contributes to multiple disorders, including cancers, vascular diseases, and neurodegenerative diseases. During prenylation, prenyltransferase enzymes tether metabolically produced isoprenoid lipids to proteins via a thioether linkage. Pharmacological inhibition of the lipid synthesis pathway by statins has long been a therapeutic approach to control hyperlipidemia. Building on our previous finding that statins inhibit membrane association of G protein γ (Gγ) in a subtype-dependent manner, we investigated the molecular reasoning for this differential. We examined the prenylation efficacy of carboxy terminus (Ct) mutated Gγ in cells exposed to Fluvastatin and prenyl transferase inhibitors and monitored the subcellular localization of fluorescently tagged Gγ subunits and their mutants using live-cell confocal imaging. Reversible optogenetic unmasking-masking of Ct residues was used to probe their contribution to the prenylation process and membrane interactions of the prenylated proteins. Our findings suggest that specific Ct residues regulate membrane interactions of the Gγ polypeptide statin sensitivity, and prenylation efficacy. Our results also show that a few hydrophobic and charged residues at the Ct are crucial determinants of a protein's prenylation ability, especially under suboptimal conditions. Given the cell and tissue-specific expression of different Gγ subtypes, our findings explain how and why statins differentially perturb heterotrimeric G protein signaling in specific cells and tissues. Our results may provide molecular reasoning for repurposing statins as Ras oncogene inhibitors and the failure of using prenyltransferase inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Mithila Tennakoon
- Department of Chemistry, Saint Louis University, Saint Louis, MO 63103, USA
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
- Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, MO 63103, USA
| | - Waruna Thotamune
- Department of Chemistry, Saint Louis University, Saint Louis, MO 63103, USA
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
- Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, MO 63103, USA
| | - John L. Payton
- Department of Chemistry, Kenyon College, Gambier, Ohio 43022, USA
| | - Ajith Karunarathne
- Department of Chemistry, Saint Louis University, Saint Louis, MO 63103, USA
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
- Institute for Drug and Biotherapeutic Innovation, Saint Louis University, Saint Louis, MO 63103, USA
| |
Collapse
|
5
|
Rajanala K, Wedegaertner PB. Gβγ signaling regulates microtubule-dependent control of Golgi integrity. Cell Signal 2023; 106:110630. [PMID: 36805843 PMCID: PMC10079639 DOI: 10.1016/j.cellsig.2023.110630] [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: 12/29/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Gβγ subunits regulate several non-canonical functions at distinct intracellular organelles. Previous studies have shown that Gβγ signaling at the Golgi is necessary to mediate vesicular protein transport function and to regulate mitotic Golgi fragmentation. Disruption of Golgi structure also occurs in response to microtubule depolymerizing agents, such as nocodazole. In this study, we use siRNA against Gβ1/2 or specific Gγ subunits to deplete their expression, and show that their knockdown causes a significant reduction in nocodazole-induced Golgi fragmentation. We establish that knockdown of Gβγ or inhibition of Gβγ with gallein resulted in decreased activation of protein kinase D (PKD) in response to nocodazole treatment. We demonstrate that restricting the amount of free Gβγ available for signaling by either inhibiting Gαi activation using pertussis toxin or by knockdown of the non-GPCR GEF, Girdin/GIV protein, results in a substantial decrease in nocodazole-induced Golgi fragmentation and PKD phosphorylation. Our results also indicate that depletion of Gβγ or inhibition with gallein or pertussis toxin significantly reduces the microtubule disruption-dependent Golgi fragmentation phenotype observed in cells transfected with mutant SOD1, a major causative protein in familial amyotrophic lateral sclerosis (ALS). These results provide compelling evidence that Gβγ signaling is critical for the regulation of Golgi integrity.
Collapse
Affiliation(s)
- Kalpana Rajanala
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Sidney Kimmel Medical College, Philadelphia, PA 19107, United States of America
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Sidney Kimmel Medical College, Philadelphia, PA 19107, United States of America.
| |
Collapse
|
6
|
Xu X, Wu G. Non-canonical Golgi-compartmentalized Gβγ signaling: mechanisms, functions, and therapeutic targets. Trends Pharmacol Sci 2023; 44:98-111. [PMID: 36494204 PMCID: PMC9901158 DOI: 10.1016/j.tips.2022.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
G protein Gβγ subunits are key mediators of G protein-coupled receptor (GPCR) signaling under physiological and pathological conditions; their inhibitors have been tested for the treatment of human disease. Conventional wisdom is that the Gβγ complex is activated and subsequently exerts its functions at the plasma membrane (PM). Recent studies have revealed non-canonical activation of Gβγ at intracellular organelles, where the Golgi apparatus is a major locale, via translocation or local activation. Golgi-localized Gβγ activates specific signaling cascades and regulates fundamental cell processes such as membrane trafficking, proliferation, and migration. More recent studies have shown that inhibiting Golgi-compartmentalized Gβγ signaling attenuates cardiomyocyte hypertrophy and prostate tumorigenesis, indicating new therapeutic targets. We review novel activation mechanisms and non-canonical functions of Gβγ at the Golgi, and discuss potential therapeutic interventions by targeting Golgi-biased Gβγ-directed signaling.
Collapse
Affiliation(s)
- Xin Xu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
7
|
Ubeysinghe S, Wijayaratna D, Kankanamge D, Karunarathne A. Molecular regulation of PLCβ signaling. Methods Enzymol 2023; 682:17-52. [PMID: 36948701 DOI: 10.1016/bs.mie.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Phospholipase C (PLC) enzymes convert the membrane phospholipid phosphatidylinositol-4,5-bisphosphate (PIP2) into inositol-1,4,5-triphosphate (IP3) and diacylglycerol (DAG). IP3 and DAG regulate numerous downstream pathways, eliciting diverse and profound cellular changes and physiological responses. In the six PLC subfamilies in higher eukaryotes, PLCβ is intensively studied due to its prominent role in regulating crucial cellular events underlying many processes including cardiovascular and neuronal signaling, and associated pathological conditions. In addition to GαqGTP, Gβγ generated upon G protein heterotrimer dissociation also regulates PLCβ activity. Here, we not only review how Gβγ directly activates PLCβ, and also extensively modulates Gαq-mediated PLCβ activity, but also provide a structure-function overview of PLC family members. Given that Gαq and PLCβ are oncogenes, and Gβγ shows unique cell-tissue-organ specific expression profiles, Gγ subtype-dependent signaling efficacies, and distinct subcellular activities, this review proposes that Gβγ is a major regulator of Gαq-dependent and independent PLCβ signaling.
Collapse
Affiliation(s)
| | | | - Dinesh Kankanamge
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Ajith Karunarathne
- Department of Chemistry, St. Louis University, St. Louis, MO, United States.
| |
Collapse
|
8
|
Kankanamge D, Tennakoon M, Karunarathne A, Gautam N. G protein gamma subunit, a hidden master regulator of GPCR signaling. J Biol Chem 2022; 298:102618. [PMID: 36272647 PMCID: PMC9678972 DOI: 10.1016/j.jbc.2022.102618] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/21/2022] Open
Abstract
Heterotrimeric G proteins (αβγ subunits) that are activated by G protein-coupled receptors (GPCRs) mediate the biological responses of eukaryotic cells to extracellular signals. The α subunits and the tightly bound βγ subunit complex of G proteins have been extensively studied and shown to control the activity of effector molecules. In contrast, the potential roles of the large family of γ subunits have been less studied. In this review, we focus on present knowledge about these proteins. Induced loss of individual γ subunit types in animal and plant models result in strikingly distinct phenotypes indicating that γ subtypes play important and specific roles. Consistent with these findings, downregulation or upregulation of particular γ subunit types result in various types of cancers. Clues about the mechanistic basis of γ subunit function have emerged from imaging the dynamic behavior of G protein subunits in living cells. This shows that in the basal state, G proteins are not constrained to the plasma membrane but shuttle between membranes and on receptor activation βγ complexes translocate reversibly to internal membranes. The translocation kinetics of βγ complexes varies widely and is determined by the membrane affinity of the associated γ subtype. On translocating, some βγ complexes act on effectors in internal membranes. The variation in translocation kinetics determines differential sensitivity and adaptation of cells to external signals. Membrane affinity of γ subunits is thus a parsimonious and elegant mechanism that controls information flow to internal cell membranes while modulating signaling responses.
Collapse
Affiliation(s)
- Dinesh Kankanamge
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Mithila Tennakoon
- Department of Chemistry, St Louis University, St Louis, Missouri, USA
| | | | - N Gautam
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA; Department of Genetics, Washington University School of Medicine, St Louis, Missouri, USA.
| |
Collapse
|
9
|
Xu X, Khater M, Wu G. The olfactory receptor OR51E2 activates ERK1/2 through the Golgi-localized Gβγ-PI3Kγ-ARF1 pathway in prostate cancer cells. Front Pharmacol 2022; 13:1009380. [PMID: 36313302 PMCID: PMC9606680 DOI: 10.3389/fphar.2022.1009380] [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: 08/01/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
The olfactory receptor OR51E2 is ectopically expressed in prostate tissues and regulates prostate cancer progression, but its function and regulation in oncogenic mitogen-activate protein kinase (MAPK) activation are poorly defined. Here we demonstrate that β-ionone, an OR51E2 agonist, dose-dependently activates extracellular signal-regulated kinases 1 and 2 (ERK1/2) in prostate cancer cells, with an EC50 value of approximate 20 μM and an efficiency comparable to other receptor agonists. We also find that CRISPR-Cas9-mediated knockout of Golgi-translocating Gγ9 subunit, phosphoinositide 3-kinase γ (PI3Kγ) and the small GTPase ADP-ribosylation factor 1 (ARF1), as well as pharmacological inhibition of Gβγ, PI3Kγ and Golgi-localized ARF1, each abolishes ERK1/2 activation by β-ionone. We further show that β-ionone significantly promotes ARF1 translocation to the Golgi and activates ARF1 that can be inhibited by Gγ9 and PI3Kγ depletion. Collectively, our data demonstrate that OR51E2 activates ERK1/2 through the Gβγ-PI3Kγ-ARF1 pathway that occurs spatially at the Golgi, and also provide important insights into MAPK hyper-activation in prostate cancer.
Collapse
|
10
|
Thor D. G protein-coupled receptors as regulators of pancreatic islet functionality. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119235. [PMID: 35151663 DOI: 10.1016/j.bbamcr.2022.119235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 01/03/2023]
Abstract
Glucose homeostasis is maintained by hormones secreted from different types of pancreatic islets and its dysregulation can result in diseases including diabetes mellitus. The secretion of hormones from pancreatic islets is highly complex and tightly controlled by G protein-coupled receptors (GPCRs). Moreover, GPCR signaling may play a role in enhancing islet cell replication and proliferation. Thus, targeting GPCRs offers a promising strategy for regulating the functionality of pancreatic islets. Here, available RNAseq datasets from human and mouse islets were used to identify the GPCR expression profile and the impact of GPCR signaling for normal islet functionality is discussed.
Collapse
Affiliation(s)
- Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany.
| |
Collapse
|
11
|
Liccardo F, Luini A, Di Martino R. Endomembrane-Based Signaling by GPCRs and G-Proteins. Cells 2022; 11:528. [PMID: 35159337 PMCID: PMC8834376 DOI: 10.3390/cells11030528] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 12/14/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) and G-proteins have a range of roles in many physiological and pathological processes and are among the most studied signaling proteins. A plethora of extracellular stimuli can activate the GPCR and can elicit distinct intracellular responses through the activation of specific transduction pathways. For many years, biologists thought that GPCR signaling occurred entirely on the plasma membrane. However, in recent decades, many lines of evidence have proved that the GPCRs and G-proteins may reside on endomembranes and can start or propagate signaling pathways through the organelles that form the secretory route. How these alternative intracellular signaling pathways of the GPCR and G-proteins influence the physiological and pathological function of the endomembranes is still under investigation. Here, we review the general role and classification of GPCRs and G-proteins with a focus on their signaling pathways in the membrane transport apparatus.
Collapse
Affiliation(s)
- Federica Liccardo
- Cardiovascular Research Institute, University of California San Francisco (UCSF), 555 Mission Bay Blvd., San Francisco, CA 94158, USA;
| | - Alberto Luini
- Istituto per L’endocrinologia e L’oncologia Sperimentale “Gaetano Salvatore” (IEOS)—Sede Secondaria, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Rosaria Di Martino
- Istituto per L’endocrinologia e L’oncologia Sperimentale “Gaetano Salvatore” (IEOS)—Sede Secondaria, Via Pietro Castellino 111, 80131 Napoli, Italy
| |
Collapse
|
12
|
Okada T, Nishida S, Zhang L, Ibrahim Mohamed NN, Wang T, Ijuin T, Kajimoto T, Nakamura SI. Constitutive activation of S1P receptors at the trans-Golgi network is required for surface transport carrier formation. iScience 2021; 24:103351. [PMID: 34805799 PMCID: PMC8590068 DOI: 10.1016/j.isci.2021.103351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/18/2021] [Accepted: 10/22/2021] [Indexed: 11/10/2022] Open
Abstract
The importance of the G-protein βγ subunits in the regulation of cargo transport from the trans-Golgi network (TGN) to the plasma membrane (PM) is well accepted; however, the molecular mechanism underlying the G-protein activation at the TGN remains unclear. We show here that sphingosine 1-phosphate (S1P) receptors at the PM were trafficked to the TGN in response to a surface transport cargo, temperature-sensitive vesicular stomatitis virus glycoprotein tagged with green fluorescent protein accumulation in the Golgi. The receptor internalization occurred in an S1P-independent manner but required phosphorylation by G-protein receptor kinase 2 and β-arrestin association before internalization. Continuously activated S1P receptors in a manner dependent on S1P at the TGN kept transmitting G-protein signals including the βγ subunits supply necessary for transport carrier formation at the TGN destined for the PM. S1P receptors traffic from the PM to Golgi in a surface cargo-dependent manner S1PR trafficking follows GRK2-dependent phosphorylation and β-arrestin binding S1PRs at the Golgi are continuously activated by S1P while sending G-protein signals S1PR/Gβγ signals at the Golgi are indispensable for surface transport carrier formation
Collapse
Affiliation(s)
- Taro Okada
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Susumu Nishida
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Lifang Zhang
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Nesma Nabil Ibrahim Mohamed
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.,Department of Agricultural Biochemistry, Faculty of Agriculture, Ain Shams University, Cairo, Egypt
| | - Tianyou Wang
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takeshi Ijuin
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Taketoshi Kajimoto
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Shun-Ichi Nakamura
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| |
Collapse
|
13
|
Tennakoon M, Senarath K, Kankanamge D, Chadee DN, Karunarathne A. A short C-terminal peptide in Gγ regulates Gβγ signaling efficacy. Mol Biol Cell 2021; 32:1446-1458. [PMID: 34106735 PMCID: PMC8351738 DOI: 10.1091/mbc.e20-11-0750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/03/2021] [Accepted: 06/04/2021] [Indexed: 01/03/2023] Open
Abstract
G protein beta-gamma (Gβγ) subunits anchor to the plasma membrane (PM) through the carboxy-terminal (CT) prenyl group in Gγ. This interaction is crucial for the PM localization and functioning of Gβγ, allowing GPCR-G protein signaling to proceed. The diverse Gγ family has 12 members, and we have recently shown that the signaling efficacies of major Gβγ effectors are Gγ-type dependent. This dependency is due to the distinct series of membrane-interacting abilities of Gγ. However, the molecular process allowing for Gβγ subunits to exhibit a discrete and diverse range of Gγ-type-dependent membrane affinities is unclear and cannot be explained using only the type of prenylation. The present work explores the unique designs of membrane-interacting CT residues in Gγ as a major source for this Gγ-type-dependent Gβγ signaling. Despite the type of prenylation, the results show signaling efficacy at the PM, and associated cell behaviors of Gβγ are governed by crucially located specific amino acids in the five to six residue preprenylation region of Gγ. The provided molecular picture of Gγ-membrane interactions may explain how cells gain Gγ-type-dependent G protein-GPCR signaling as well as how Gβγ elicits selective signaling at various subcellular compartments.
Collapse
Affiliation(s)
- Mithila Tennakoon
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606
| | - Kanishka Senarath
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606
| | - Dinesh Kankanamge
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606
| | - Deborah N. Chadee
- Department of Biological Sciences, The University of Toledo, Toledo, OH 43606
| | - Ajith Karunarathne
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606
| |
Collapse
|
14
|
Rajanala K, Klayman LM, Wedegaertner PB. Gβγ regulates mitotic Golgi fragmentation and G2/M cell cycle progression. Mol Biol Cell 2021; 32:br2. [PMID: 34260268 PMCID: PMC8684744 DOI: 10.1091/mbc.e21-04-0175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Heterotrimeric G proteins (αβγ) function at the cytoplasmic surface of a cell’s plasma membrane to transduce extracellular signals into cellular responses. However, numerous studies indicate that G proteins also play noncanonical roles at unique intracellular locations. Previous work has established that G protein βγ subunits (Gβγ) regulate a signaling pathway on the cytoplasmic surface of Golgi membranes that controls the exit of select protein cargo. Now, we demonstrate a novel role for Gβγ in regulating mitotic Golgi fragmentation, a key checkpoint of the cell cycle that occurs in the late G2 phase. We show that small interfering RNA–mediated depletion of Gβ1 and Gβ2 in synchronized cells causes a decrease in the number of cells with fragmented Golgi in late G2 and a delay of entry into mitosis and progression through G2/M. We also demonstrate that during G2/M Gβγ acts upstream of protein kinase D and regulates the phosphorylation of the Golgi structural protein GRASP55. Expression of Golgi-targeted GRK2ct, a Gβγ-sequestering protein used to inhibit Gβγ signaling, also causes a decrease in Golgi fragmentation and a delay in mitotic progression. These results highlight a novel role for Gβγ in regulation of Golgi structure.
Collapse
Affiliation(s)
- Kalpana Rajanala
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Sidney Kimmel Medical College, Philadelphia, PA 19107
| | - Lauren M Klayman
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Sidney Kimmel Medical College, Philadelphia, PA 19107
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Sidney Kimmel Medical College, Philadelphia, PA 19107
| |
Collapse
|
15
|
The PKD-Dependent Biogenesis of TGN-to-Plasma Membrane Transport Carriers. Cells 2021; 10:cells10071618. [PMID: 34203456 PMCID: PMC8303525 DOI: 10.3390/cells10071618] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/14/2021] [Accepted: 06/24/2021] [Indexed: 01/30/2023] Open
Abstract
Membrane trafficking is essential for processing and transport of proteins and lipids and to establish cell compartmentation and tissue organization. Cells respond to their needs and control the quantity and quality of protein secretion accordingly. In this review, we focus on a particular membrane trafficking route from the trans-Golgi network (TGN) to the cell surface: protein kinase D (PKD)-dependent pathway for constitutive secretion mediated by carriers of the TGN to the cell surface (CARTS). Recent findings highlight the importance of lipid signaling by organelle membrane contact sites (MCSs) in this pathway. Finally, we discuss our current understanding of multiple signaling pathways for membrane trafficking regulation mediated by PKD, G protein-coupled receptors (GPCRs), growth factors, metabolites, and mechanosensors.
Collapse
|
16
|
Tennakoon M, Senarath K, Kankanamge D, Ratnayake K, Wijayaratna D, Olupothage K, Ubeysinghe S, Martins-Cannavino K, Hébert TE, Karunarathne A. Subtype-dependent regulation of Gβγ signalling. Cell Signal 2021; 82:109947. [PMID: 33582184 PMCID: PMC8026654 DOI: 10.1016/j.cellsig.2021.109947] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 01/04/2023]
Abstract
G protein-coupled receptors (GPCRs) transmit information to the cell interior by transducing external signals to heterotrimeric G protein subunits, Gα and Gβγ subunits, localized on the inner leaflet of the plasma membrane. Though the initial focus was mainly on Gα-mediated events, Gβγ subunits were later identified as major contributors to GPCR-G protein signalling. A broad functional array of Gβγ signalling has recently been attributed to Gβ and Gγ subtype diversity, comprising 5 Gβ and 12 Gγ subtypes, respectively. In addition to displaying selectivity towards each other to form the Gβγ dimer, numerous studies have identified preferences of distinct Gβγ combinations for specific GPCRs, Gα subtypes and effector molecules. Importantly, Gβ and Gγ subtype-dependent regulation of downstream effectors, representing a diverse range of signalling pathways and physiological functions have been found. Here, we review the literature on the repercussions of Gβ and Gγ subtype diversity on direct and indirect regulation of GPCR/G protein signalling events and their physiological outcomes. Our discussion additionally provides perspective in understanding the intricacies underlying molecular regulation of subtype-specific roles of Gβγ signalling and associated diseases.
Collapse
Affiliation(s)
- Mithila Tennakoon
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | - Kanishka Senarath
- Genetics and Molecular Biology Unit, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Dinesh Kankanamge
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | - Kasun Ratnayake
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dhanushan Wijayaratna
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | - Koshala Olupothage
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | - Sithurandi Ubeysinghe
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA
| | | | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada.
| | - Ajith Karunarathne
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, OH 43606, USA.
| |
Collapse
|
17
|
Khater M, Bryant CN, Wu G. Gβγ translocation to the Golgi apparatus activates ARF1 to spatiotemporally regulate G protein-coupled receptor signaling to MAPK. J Biol Chem 2021; 296:100805. [PMID: 34022220 PMCID: PMC8215300 DOI: 10.1016/j.jbc.2021.100805] [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: 03/08/2021] [Revised: 05/11/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
After activation of G protein-coupled receptors, G protein βγ dimers may translocate from the plasma membrane to the Golgi apparatus (GA). We recently report that this translocation activates extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) via PI3Kγ; however, how Gβγ-PI3Kγ activates the ERK1/2 pathway is unclear. Here, we demonstrate that chemokine receptor CXCR4 activates ADP-ribosylation factor 1 (ARF1), a small GTPase important for vesicle-mediated membrane trafficking. This activation is blocked by CRISPR-Cas9-mediated knockout of the GA-translocating Gγ9 subunit. Inducible targeting of different Gβγ dimers to the GA can directly activate ARF1. CXCR4 activation and constitutive Gβγ recruitment to the GA also enhance ARF1 translocation to the GA. We further demonstrate that pharmacological inhibition and CRISPR-Cas9-mediated knockout of PI3Kγ markedly inhibit CXCR4-mediated and Gβγ translocation-mediated ARF1 activation. We also show that depletion of ARF1 by siRNA and CRISPR-Cas9 and inhibition of GA-localized ARF1 activation abolish ERK1/2 activation by CXCR4 and Gβγ translocation to the GA and suppress prostate cancer PC3 cell migration and invasion. Collectively, our data reveal a novel function for Gβγ translocation to the GA to activate ARF1 and identify GA-localized ARF1 as an effector acting downstream of Gβγ-PI3Kγ to spatiotemporally regulate G protein-coupled receptor signaling to mitogen-activated protein kinases.
Collapse
Affiliation(s)
- Mostafa Khater
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Christian N Bryant
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.
| |
Collapse
|
18
|
Kankanamge D, Ubeysinghe S, Tennakoon M, Pantula PD, Mitra K, Giri L, Karunarathne A. Dissociation of the G protein βγ from the Gq-PLCβ complex partially attenuates PIP2 hydrolysis. J Biol Chem 2021; 296:100702. [PMID: 33901492 PMCID: PMC8138763 DOI: 10.1016/j.jbc.2021.100702] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 04/09/2021] [Accepted: 04/21/2021] [Indexed: 01/14/2023] Open
Abstract
Phospholipase C β (PLCβ), which is activated by the Gq family of heterotrimeric G proteins, hydrolyzes the inner membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2), generating diacylglycerol and inositol 1,4,5-triphosphate (IP3). Because Gq and PLCβ regulate many crucial cellular processes and have been identified as major disease drivers, activation and termination of PLCβ signaling by the Gαq subunit have been extensively studied. Gq-coupled receptor activation induces intense and transient PIP2 hydrolysis, which subsequently recovers to a low-intensity steady-state equilibrium. However, the molecular underpinnings of this equilibrium remain unclear. Here, we explored the influence of signaling crosstalk between Gq and Gi/o pathways on PIP2 metabolism in living cells using single-cell and optogenetic approaches to spatially and temporally constrain signaling. Our data suggest that the Gβγ complex is a component of the highly efficient lipase GαqGTP-PLCβ-Gβγ. We found that over time, Gβγ dissociates from this lipase complex, leaving the less-efficient GαqGTP-PLCβ lipase complex and allowing the significant partial recovery of PIP2 levels. Our findings also indicate that the subtype of the Gγ subunit in Gβγ fine-tunes the lipase activity of Gq-PLCβ, in which cells expressing Gγ with higher plasma membrane interaction show lower PIP2 recovery. Given that Gγ shows cell- and tissue-specific subtype expression, our findings suggest the existence of tissue-specific distinct Gq-PLCβ signaling paradigms. Furthermore, these results also outline a molecular process that likely safeguards cells from excessive Gq signaling.
Collapse
Affiliation(s)
- Dinesh Kankanamge
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA
| | - Sithurandi Ubeysinghe
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA
| | - Mithila Tennakoon
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA
| | - Priyanka Devi Pantula
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, India
| | - Kishalay Mitra
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, India
| | - Lopamudra Giri
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, India
| | - Ajith Karunarathne
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio, USA.
| |
Collapse
|
19
|
Diversity of the Gβγ complexes defines spatial and temporal bias of GPCR signaling. Cell Syst 2021; 12:324-337.e5. [PMID: 33667409 DOI: 10.1016/j.cels.2021.02.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 12/09/2020] [Accepted: 02/04/2021] [Indexed: 01/04/2023]
Abstract
The signal transduction by G-protein-coupled receptors (GPCRs) is mediated by heterotrimeric G proteins composed from one of the 16 Gα subunits and the inseparable Gβγ complex assembled from a repertoire of 5 Gβ and 12 Gγ subunits. However, the functional role of compositional diversity in Gβγ complexes has been elusive. Using optical biosensors, we examined the function of all Gβγ combinations in living cells and uncovered two major roles of Gβγ diversity. First, we demonstrate that the identity of Gβγ subunits greatly influences the kinetics and efficacy of GPCR responses at the plasma membrane. Second, we show that different Gβγ combinations are selectively dispatched from the plasma membrane to various cellular organelles on a timescale from milliseconds to minutes. We describe the mechanisms regulating these processes and document their implications for GPCR signaling via various Gα subunits, thereby illustrating a role for the compositional diversity of G protein heterotrimers.
Collapse
|
20
|
Khater M, Wei Z, Xu X, Huang W, Lokeshwar BL, Lambert NA, Wu G. G protein βγ translocation to the Golgi apparatus activates MAPK via p110γ-p101 heterodimers. J Biol Chem 2021; 296:100325. [PMID: 33493514 PMCID: PMC7949113 DOI: 10.1016/j.jbc.2021.100325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 01/14/2023] Open
Abstract
The Golgi apparatus (GA) is a cellular organelle that plays a critical role in the processing of proteins for secretion. Activation of G protein-coupled receptors at the plasma membrane (PM) induces the translocation of G protein βγ dimers to the GA. However, the functional significance of this translocation is largely unknown. Here, we study PM-GA translocation of all 12 Gγ subunits in response to chemokine receptor CXCR4 activation and demonstrate that Gγ9 is a unique Golgi-translocating Gγ subunit. CRISPR-Cas9-mediated knockout of Gγ9 abolishes activation of extracellular signal-regulated kinase 1 and 2 (ERK1/2), two members of the mitogen-activated protein kinase family, by CXCR4. We show that chemically induced recruitment to the GA of Gβγ dimers containing different Gγ subunits activates ERK1/2, whereas recruitment to the PM is ineffective. We also demonstrate that pharmacological inhibition of phosphoinositide 3-kinase γ (PI3Kγ) and depletion of its subunits p110γ and p101 abrogate ERK1/2 activation by CXCR4 and Gβγ recruitment to the GA. Knockout of either Gγ9 or PI3Kγ significantly suppresses prostate cancer PC3 cell migration, invasion, and metastasis. Collectively, our data demonstrate a novel function for Gβγ translocation to the GA, via activating PI3Kγ heterodimers p110γ-p101, to spatiotemporally regulate mitogen-activated protein kinase activation by G protein-coupled receptors and ultimately control tumor progression.
Collapse
Affiliation(s)
- Mostafa Khater
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Zhe Wei
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Xin Xu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Wei Huang
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Bal L Lokeshwar
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Nevin A Lambert
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.
| |
Collapse
|
21
|
Ireland S, Ramnarayanan S, Fu M, Zhang X, Zhang J, Li J, Emebo D, Wang Y. Cytosolic Ca 2+ Modulates Golgi Structure Through PKCα-Mediated GRASP55 Phosphorylation. iScience 2020; 23:100952. [PMID: 32179476 PMCID: PMC7078314 DOI: 10.1016/j.isci.2020.100952] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/31/2020] [Accepted: 02/25/2020] [Indexed: 12/31/2022] Open
Abstract
It has been well documented that the ER responds to cellular stresses through the unfolded protein response (UPR), but it is unknown how the Golgi responds to similar stresses. In this study, we treated HeLa cells with ER stress inducers, thapsigargin (TG), tunicamycin (Tm), and dithiothreitol (DTT), and found that only TG treatment resulted in Golgi fragmentation. TG induced Golgi fragmentation at a low dose and short time when UPR was undetectable, indicating that Golgi fragmentation occurs independently of ER stress. Further experiments demonstrated that TG induces Golgi fragmentation through elevating intracellular Ca2+ and protein kinase Cα (PKCα) activity, which phosphorylates the Golgi stacking protein GRASP55. Significantly, activation of PKCα with other activating or inflammatory agents, including phorbol 12-myristate 13-acetate and histamine, modulates Golgi structure in a similar fashion. Hence, our study revealed a novel mechanism through which increased cytosolic Ca2+ modulates Golgi structure and function. Thapsigargin (TG) treatment leads to Golgi fragmentation independent of ER stress TG induces Golgi fragmentation through elevated cytosolic Ca2+ TG-induced cytosolic Ca2+ spikes activate PKCα that phosphorylates GRASP55 Histamine modulates the Golgi structure and function by a similar mechanism
Collapse
Affiliation(s)
- Stephen Ireland
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109-1085, USA
| | - Saiprasad Ramnarayanan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109-1085, USA
| | - Mingzhou Fu
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109-1085, USA
| | - Xiaoyan Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109-1085, USA
| | - Jianchao Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109-1085, USA
| | - Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109-1085, USA
| | - Dabel Emebo
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109-1085, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Biological Sciences Building, 1105 North University Avenue, Ann Arbor, MI 48109-1085, USA; Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109-1085, USA.
| |
Collapse
|
22
|
Abstract
The mammalian Golgi apparatus is a highly dynamic organelle, which is normally localized in the juxtanuclear space and plays an essential role in the regulation of cellular homeostasis. While posttranslational modification of cargo is mediated by the resident enzymes (glycosyltransferases, glycosidases, and kinases), the ribbon structure of Golgi and its cisternal stacking mostly rely on the cooperation of coiled-coil matrix golgins. Among them, giantin, GM130, and GRASPs are unique, because they form a tripartite complex and serve as Golgi docking sites for cargo delivered from the endoplasmic reticulum (ER). Golgi undergoes significant disorganization in many pathologies associated with a block of the ER-to-Golgi or intra-Golgi transport, including cancer, different neurological diseases, alcoholic liver damage, ischemic stress, viral infections, etc. In addition, Golgi fragments during apoptosis and mitosis. Here, we summarize and analyze clinically relevant observations indicating that Golgi fragmentation is associated with the selective loss of Golgi residency for some enzymes and, conversely, with the relocation of some cytoplasmic proteins to the Golgi. The central concept is that ER and Golgi stresses impair giantin docking site but have no impact on the GM130-GRASP65 complex, thus inducing mislocalization of giantin-sensitive enzymes only. This cardinally changes the processing of proteins by eliminating the pathways controlled by the missing enzymes and by activating the processes now driven by the GM130-GRASP65-dependent proteins. This type of Golgi disorganization is different from the one induced by the cytoskeleton alteration, which despite Golgi de-centralization, neither impairs function of golgins nor alters trafficking.
Collapse
Affiliation(s)
- A Petrosyan
- College of Medicine, Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA. .,The Nebraska Center for Integrated Biomolecular Communication, Lincoln, NE 68588, USA.,The Fred and Pamela Buffett Cancer Center, Omaha, NE 68106, USA
| |
Collapse
|
23
|
Smrcka AV, Fisher I. G-protein βγ subunits as multi-functional scaffolds and transducers in G-protein-coupled receptor signaling. Cell Mol Life Sci 2019; 76:4447-4459. [PMID: 31435698 PMCID: PMC6842434 DOI: 10.1007/s00018-019-03275-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/05/2019] [Accepted: 08/12/2019] [Indexed: 02/08/2023]
Abstract
G-protein βγ subunits are key participants in G-protein signaling. These subunits facilitate interactions between receptors and G proteins that are critical for the G protein activation cycle at the plasma membrane. In addition, they play roles in directly transducing signals to an ever expanding range of downstream targets, including integral membrane and cytosolic proteins. Emerging data indicate that Gβγ may play additional roles at intracellular compartments including endosomes, the Golgi apparatus, and the nucleus. Here, we discuss the molecular and structural basis for their ability to coordinate this wide range of cellular activities.
Collapse
Affiliation(s)
- Alan V Smrcka
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48104, USA.
| | - Isaac Fisher
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48104, USA
- Department of Pharmacology and Physiology, University of Rochester School of Medicine, Rochester, NY, 14629, USA
| |
Collapse
|
24
|
Vakilian M, Tahamtani Y, Ghaedi K. A review on insulin trafficking and exocytosis. Gene 2019; 706:52-61. [DOI: 10.1016/j.gene.2019.04.063] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/21/2022]
|
25
|
Optical approaches for single-cell and subcellular analysis of GPCR-G protein signaling. Anal Bioanal Chem 2019; 411:4481-4508. [PMID: 30927013 DOI: 10.1007/s00216-019-01774-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 01/05/2023]
Abstract
G protein-coupled receptors (GPCRs), G proteins, and their signaling associates are major signal transducers that control the majority of cellular signaling and regulate key biological functions including immune, neurological, cardiovascular, and metabolic processes. These pathways are targeted by over one-third of drugs on the market; however, the current understanding of their function is limited and primarily derived from cell-destructive approaches providing an ensemble of static, multi-cell information about the status and composition of molecules. Spatiotemporal behavior of molecules involved is crucial to understanding in vivo cell behaviors both in health and disease, and the advent of genetically encoded fluorescence proteins and small fluorophore-based biosensors has facilitated the mapping of dynamic signaling in cells with subcellular acuity. Since we and others have developed optogenetic methods to regulate GPCR-G protein signaling in single cells and subcellular regions using dedicated wavelengths, the desire to develop and adopt optogenetically amenable assays to measure signaling has motivated us to take a broader look at the available optical tools and approaches compatible with measuring single-cell and subcellular GPCR-G protein signaling. Here we review such key optical approaches enabling the examination of GPCR, G protein, secondary messenger, and downstream molecules such as kinase and lipid signaling in living cells. The methods reviewed employ both fluorescence and bioluminescence detection. We not only further elaborate the underlying principles of these sensors but also discuss the experimental criteria and limitations to be considered during their use in single-cell and subcellular signal mapping.
Collapse
|
26
|
Structural basis of Gip1 for cytosolic sequestration of G protein in wide-range chemotaxis. Nat Commun 2018; 9:4635. [PMID: 30401901 PMCID: PMC6219514 DOI: 10.1038/s41467-018-07035-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/10/2018] [Indexed: 12/15/2022] Open
Abstract
G protein interacting protein 1 (Gip1) binds and sequesters heterotrimeric G proteins in the cytosolic pool, thus regulating G protein-coupled receptor (GPCR) signalling for eukaryotic chemotaxis. Here, we report the underlying structural basis of Gip1 function. The crystal structure reveals that the region of Gip1 that binds to the G protein has a cylinder-like fold with a central hydrophobic cavity composed of six α-helices. Mutagenesis and biochemical analyses indicate that the hydrophobic cavity and the hydrogen bond network at the entrance of the cavity are essential for complex formation with the geranylgeranyl modification on the Gγ subunit. Mutations of the cavity impair G protein sequestration and translocation to the membrane from the cytosol upon receptor stimulation, leading to defects in chemotaxis at higher chemoattractant concentrations. These results demonstrate that the Gip1-dependent regulation of G protein shuttling ensures wide-range gradient sensing in eukaryotic chemotaxis. Gip1 sequesters heterotrimeric G proteins in the cytosolic pool which regulates G protein-coupled receptor signalling for eukaryotic chemotaxis. Here the authors provide the crystal structure of Gip1's G protein-binding region and show that mutations in this region lead to G protein sequestration and ultimately chemotaxis defects.
Collapse
|
27
|
Harvey BJ, Thomas W. Aldosterone-induced protein kinase signalling and the control of electrolyte balance. Steroids 2018; 133:67-74. [PMID: 29079406 DOI: 10.1016/j.steroids.2017.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/18/2017] [Accepted: 10/21/2017] [Indexed: 01/20/2023]
Abstract
Aldosterone acts through the mineralocorticoid receptor (MR) to modulate gene expression in target tissues. In the kidney, the principal action of aldosterone is to promote sodium conservation in the distal nephron and so indirectly enhance water conservation under conditions of hypotension. Over the last twenty years the rapid activation of protein kinase signalling cascades by aldosterone has been described in various tissues. This review describes the integration of rapid protein kinase D signalling responses with the non-genomic actions of aldosterone and transcriptional effects of MR activation.
Collapse
Affiliation(s)
- Brian J Harvey
- Molecular Medicine Laboratories, Royal College of Surgeons in Ireland, Education Centre, Beaumont Hospital, Dublin, Ireland
| | - Warren Thomas
- Molecular Medicine Laboratories, Royal College of Surgeons in Ireland, Education Centre, Beaumont Hospital, Dublin, Ireland; Perdana University - Royal College of Surgeons in Ireland School of Medicine, Serdang, Selangor, Malaysia.
| |
Collapse
|
28
|
Senarath K, Kankanamge D, Samaradivakara S, Ratnayake K, Tennakoon M, Karunarathne A. Regulation of G Protein βγ Signaling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 339:133-191. [PMID: 29776603 DOI: 10.1016/bs.ircmb.2018.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heterotrimeric guanine nucleotide-binding proteins (G proteins) deliver external signals to the cell interior, upon activation by the external signal stimulated G protein-coupled receptors (GPCRs).While the activated GPCRs control several pathways independently, activated G proteins control the vast majority of cellular and physiological functions, ranging from vision to cardiovascular homeostasis. Activated GPCRs dissociate GαGDPβγ heterotrimer into GαGTP and free Gβγ. Earlier, GαGTP was recognized as the primary signal transducer of the pathway and Gβγ as a passive signaling modality that facilitates the activity of Gα. However, Gβγ later found to regulate more number of pathways than GαGTP does. Once liberated from the heterotrimer, free Gβγ interacts and activates a diverse range of signaling regulators including kinases, lipases, GTPases, and ion channels, and it does not require any posttranslation modifications. Gβγ family consists of 48 members, which show cell- and tissue-specific expressions, and recent reports show that cells employ the subtype diversity in Gβγ to achieve desired signaling outcomes. In addition to activated GPCRs, which induce free Gβγ generation and the rate of GTP hydrolysis in Gα, which sequester Gβγ in the heterotrimer, terminating Gβγ signaling, additional regulatory mechanisms exist to regulate Gβγ activity. In this chapter, we discuss structure and function, subtype diversity and its significance in signaling regulation, effector activation, regulatory mechanisms as well as the disease relevance of Gβγ in eukaryotes.
Collapse
|
29
|
Senarath K, Payton JL, Kankanamge D, Siripurapu P, Tennakoon M, Karunarathne A. Gγ identity dictates efficacy of Gβγ signaling and macrophage migration. J Biol Chem 2018; 293:2974-2989. [PMID: 29317505 DOI: 10.1074/jbc.ra117.000872] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 01/04/2018] [Indexed: 11/06/2022] Open
Abstract
G protein βγ subunit (Gβγ) is a major signal transducer and controls processes ranging from cell migration to gene transcription. Despite having significant subtype heterogeneity and exhibiting diverse cell- and tissue-specific expression levels, Gβγ is often considered a unified signaling entity with a defined functionality. However, the molecular and mechanistic basis of Gβγ's signaling specificity is unknown. Here, we demonstrate that Gγ subunits, bearing the sole plasma membrane (PM)-anchoring motif, control the PM affinity of Gβγ and thereby differentially modulate Gβγ effector signaling in a Gγ-specific manner. Both Gβγ signaling activity and the migration rate of macrophages are strongly dependent on the PM affinity of Gγ. We also found that the type of C-terminal prenylation and five to six pre-CaaX motif residues at the PM-interacting region of Gγ control the PM affinity of Gβγ. We further show that the overall PM affinity of the Gβγ pool of a cell type is a strong predictor of its Gβγ signaling-activation efficacy. A kinetic model encompassing multiple Gγ types and parameterized for empirical Gβγ behaviors not only recapitulated experimentally observed signaling of Gβγ, but also suggested a Gγ-dependent, active-inactive conformational switch for the PM-bound Gβγ, regulating effector signaling. Overall, our results unveil crucial aspects of signaling and cell migration regulation by Gγ type-specific PM affinities of Gβγ.
Collapse
Affiliation(s)
- Kanishka Senarath
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio 43606
| | - John L Payton
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio 43606
| | - Dinesh Kankanamge
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio 43606
| | - Praneeth Siripurapu
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio 43606
| | - Mithila Tennakoon
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio 43606
| | - Ajith Karunarathne
- Department of Chemistry and Biochemistry, The University of Toledo, Toledo, Ohio 43606.
| |
Collapse
|
30
|
Bergeron V, Ghislain J, Vivot K, Tamarina N, Philipson LH, Fielitz J, Poitout V. Deletion of Protein Kinase D1 in Pancreatic β-Cells Impairs Insulin Secretion in High-Fat Diet-Fed Mice. Diabetes 2018; 67:71-77. [PMID: 29038309 PMCID: PMC5741145 DOI: 10.2337/db17-0982] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/09/2017] [Indexed: 12/29/2022]
Abstract
Ββ-Cell adaptation to insulin resistance is necessary to maintain glucose homeostasis in obesity. Failure of this mechanism is a hallmark of type 2 diabetes (T2D). Hence, factors controlling functional β-cell compensation are potentially important targets for the treatment of T2D. Protein kinase D1 (PKD1) integrates diverse signals in the β-cell and plays a critical role in the control of insulin secretion. However, the role of β-cell PKD1 in glucose homeostasis in vivo is essentially unknown. Using β-cell-specific, inducible PKD1 knockout mice (βPKD1KO), we examined the role of β-cell PKD1 under basal conditions and during high-fat feeding. βPKD1KO mice under a chow diet presented no significant difference in glucose tolerance or insulin secretion compared with mice expressing the Cre transgene alone; however, when compared with wild-type mice, both groups developed glucose intolerance. Under a high-fat diet, deletion of PKD1 in β-cells worsened hyperglycemia, hyperinsulinemia, and glucose intolerance. This was accompanied by impaired glucose-induced insulin secretion both in vivo in hyperglycemic clamps and ex vivo in isolated islets from high-fat diet-fed βPKD1KO mice without changes in islet mass. This study demonstrates an essential role for PKD1 in the β-cell adaptive secretory response to high-fat feeding in mice.
Collapse
Affiliation(s)
- Valérie Bergeron
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Julien Ghislain
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
| | - Kevin Vivot
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
| | | | | | - Jens Fielitz
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
31
|
Takauji Y, Kudo I, En A, Matsuo R, Hossain MN, Nakabayashi K, Miki K, Fujii M, Ayusawa D. GNG11 (G-protein subunit γ 11) suppresses cell growth with induction of reactive oxygen species and abnormal nuclear morphology in human SUSM-1 cells. Biochem Cell Biol 2017; 95:517-523. [PMID: 28380310 DOI: 10.1139/bcb-2016-0248] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Enforced expression of GNG11, G-protein subunit γ 11, induces cellular senescence in normal human diploid fibroblasts. We here examined the effect of the expression of GNG11 on the growth of immortalized human cell lines, and found that it suppressed the growth of SUSM-1 cells, but not of HeLa cells. We then compared these two cell lines to understand the molecular basis for the action of GNG11. We found that expression of GNG11 induced the generation of reactive oxygen species (ROS) and abnormal nuclear morphology in SUSM-1 cells but not in HeLa cells. Increased ROS generation by GNG11 would likely be caused by the down-regulation of the antioxidant enzymes in SUSM-1 cells. We also found that SUSM-1 cells, even under normal culture conditions, showed higher levels of ROS and higher incidence of abnormal nuclear morphology than HeLa cells, and that abnormal nuclear morphology was relevant to the increased ROS generation in SUSM-1 cells. Thus, SUSM-1 and HeLa cells showed differences in the regulation of ROS and nuclear morphology, which might account for their different responses to the expression of GNG11. Thus, SUSM-1 cells may provide a unique system to study the regulatory relationship between ROS generation, nuclear morphology, and G-protein signaling.
Collapse
Affiliation(s)
- Yuki Takauji
- a Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Ikuru Kudo
- a Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Atsuki En
- a Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Ryo Matsuo
- a Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Mohammad Nazir Hossain
- b Department of Biochemistry, Primeasia University, 9 Banani C/A Banani, Dhaka 1213, Bangladesh
| | - Kazuhiko Nakabayashi
- c Department of Maternal-Fetal Biology, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Kensuke Miki
- a Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan.,d Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, Kanagawa 231-0048, Japan
| | - Michihiko Fujii
- a Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Dai Ayusawa
- a Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan.,d Ichiban Life Corporation, 1-1-7 Horai-cho, Naka-ku, Yokohama, Kanagawa 231-0048, Japan
| |
Collapse
|
32
|
|
33
|
Klayman LM, Wedegaertner PB. Inducible Inhibition of Gβγ Reveals Localization-dependent Functions at the Plasma Membrane and Golgi. J Biol Chem 2016; 292:1773-1784. [PMID: 27994056 DOI: 10.1074/jbc.m116.750430] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/16/2016] [Indexed: 01/28/2023] Open
Abstract
Heterotrimeric G proteins signal at a variety of endomembrane locations, in addition to their canonical function at the cytoplasmic surface of the plasma membrane (PM), where they are activated by cell surface G protein-coupled receptors. Here we focus on βγ signaling at the Golgi, where βγ activates a signaling cascade, ultimately resulting in vesicle fission from the trans-Golgi network (TGN). To develop a novel molecular tool for inhibiting endogenous βγ in a spatial-temporal manner, we take advantage of a lipid association mutant of the widely used βγ inhibitor GRK2ct (GRK2ct-KERE) and the FRB/FKBP heterodimerization system. We show that GRK2ct-KERE cannot inhibit βγ function when expressed in cells, but recruitment to a specific membrane location recovers the ability of GRK2ct-KERE to inhibit βγ signaling. PM-recruited GRK2ct-KERE inhibits lysophosphatidic acid-induced phosphorylation of Akt, whereas Golgi-recruited GRK2ct-KERE inhibits cargo transport from the TGN to the PM. Moreover, we show that Golgi-recruited GRK2ct-KERE inhibits model basolaterally targeted but not apically targeted cargo delivery, for both PM-destined and secretory cargo, providing the first evidence of selectivity in terms of cargo transport regulated by βγ. Last, we show that Golgi fragmentation induced by ilimaquinone and nocodazole is blocked by βγ inhibition, demonstrating that βγ is a key regulator of multiple pathways that impact Golgi morphology. Thus, we have developed a new molecular tool, recruitable GRK2ct-KERE, to modulate βγ signaling at specific subcellular locations, and we demonstrate novel cargo selectivity for βγ regulation of TGN to PM transport and a novel role for βγ in mediating Golgi fragmentation.
Collapse
Affiliation(s)
- Lauren M Klayman
- From the Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Philip B Wedegaertner
- From the Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107.
| |
Collapse
|
34
|
Senarath K, Ratnayake K, Siripurapu P, Payton JL, Karunarathne A. Reversible G Protein βγ9 Distribution-Based Assay Reveals Molecular Underpinnings in Subcellular, Single-Cell, and Multicellular GPCR and G Protein Activity. Anal Chem 2016; 88:11450-11459. [PMID: 27778511 DOI: 10.1021/acs.analchem.6b02512] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Current assays to measure the activation of G protein coupled receptors (GPCRs) and G proteins are time-consuming, indirect, and expensive. Therefore, an efficient method which directly measures the ability of a ligand to govern GPCR-G protein interactions can help to understand the molecular underpinnings of the associated signaling. A live cell imaging-based approach is presented here to directly measure ligand-induced GPCR and G protein activity in real time. The number of active GPCRs governs G protein heterotrimer (αβγ) dissociation, thereby controlling the concentration of free βγ subunits. The described γ9 assay measures the GPCR activation-induced extent of the reversible βγ9 subunit exchange between the plasma membrane (PM) and internal membranes (IMs). Confocal microscopy-based γ9 assay quantitatively determines the concentration dependency of ligands on GPCR activation. Demonstrating the high-throughput screening (HTS) adaptability, the γ9 assay performed using an imaging plate reader measures the ligand-induced GPCR activation. This suggests that the γ9 assay can be employed to screen libraries of compounds for their ability to activate GPCRs. Together with subcellular optogenetics, the spatiotemporal sensitivity of the γ9 assay permits experimental determination of the limits of spatially restricted activation of GPCRs and G proteins in subcellular regions of single cells. This assay works effectively for GPCRs coupled to αi/o and αs heterotrimers, including light-sensitive GPCRs. In addition, computational modeling of experimental data from the assay is used to decipher intricate molecular details of the GPCR-G protein activation process. Overall, the γ9 assay provides a robust strategy for quantitative as well as qualitative determination of GPCR and G protein function on a single-cell, multicell, and subcellular level. This assay not only provides information about the inner workings of the signaling pathway, but it also strengthens GPCR deorphanization as well as drug discovery efforts.
Collapse
Affiliation(s)
- Kanishka Senarath
- Department of Chemistry and Biochemistry, The University of Toledo , Toledo, Ohio 43606, United States
| | - Kasun Ratnayake
- Department of Chemistry and Biochemistry, The University of Toledo , Toledo, Ohio 43606, United States
| | - Praneeth Siripurapu
- Department of Chemistry and Biochemistry, The University of Toledo , Toledo, Ohio 43606, United States
| | - John L Payton
- Department of Chemistry and Biochemistry, The University of Toledo , Toledo, Ohio 43606, United States
| | - Ajith Karunarathne
- Department of Chemistry and Biochemistry, The University of Toledo , Toledo, Ohio 43606, United States
| |
Collapse
|
35
|
Jensen DD, Zhao P, Jimenez-Vargas NN, Lieu T, Gerges M, Yeatman HR, Canals M, Vanner SJ, Poole DP, Bunnett NW. Protein Kinase D and Gβγ Subunits Mediate Agonist-evoked Translocation of Protease-activated Receptor-2 from the Golgi Apparatus to the Plasma Membrane. J Biol Chem 2016; 291:11285-99. [PMID: 27030010 PMCID: PMC4900274 DOI: 10.1074/jbc.m115.710681] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/17/2016] [Indexed: 12/12/2022] Open
Abstract
Agonist-evoked endocytosis of G protein-coupled receptors has been extensively studied. The mechanisms by which agonists stimulate mobilization and plasma membrane translocation of G protein-coupled receptors from intracellular stores are unexplored. Protease-activated receptor-2 (PAR2) traffics to lysosomes, and sustained protease signaling requires mobilization and plasma membrane trafficking of PAR2 from Golgi stores. We evaluated the contribution of protein kinase D (PKD) and Gβγ to this process. In HEK293 and KNRK cells, the PAR2 agonists trypsin and 2-furoyl-LIGRLO-NH2 activated PKD in the Golgi apparatus, where PKD regulates protein trafficking. PAR2 activation induced translocation of Gβγ, a PKD activator, to the Golgi apparatus, determined by bioluminescence resonance energy transfer between Gγ-Venus and giantin-Rluc8. Inhibitors of PKD (CRT0066101) and Gβγ (gallein) prevented PAR2-stimulated activation of PKD. CRT0066101, PKD1 siRNA, and gallein all inhibited recovery of PAR2-evoked Ca(2+) signaling. PAR2 with a photoconvertible Kaede tag was expressed in KNRK cells to examine receptor translocation from the Golgi apparatus to the plasma membrane. Irradiation of the Golgi region (405 nm) induced green-red photo-conversion of PAR2-Kaede. Trypsin depleted PAR2-Kaede from the Golgi apparatus and repleted PAR2-Kaede at the plasma membrane. CRT0066101 inhibited PAR2-Kaede translocation to the plasma membrane. CRT0066101 also inhibited sustained protease signaling to colonocytes and nociceptive neurons that naturally express PAR2 and mediate protease-evoked inflammation and nociception. Our results reveal a major role for PKD and Gβγ in agonist-evoked mobilization of intracellular PAR2 stores that is required for sustained signaling by extracellular proteases.
Collapse
Affiliation(s)
- Dane D Jensen
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Peishen Zhao
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Nestor N Jimenez-Vargas
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - TinaMarie Lieu
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Marina Gerges
- From the Monash Institute of Pharmaceutical Sciences and
| | | | - Meritxell Canals
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Stephen J Vanner
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Daniel P Poole
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia, the Departments of Anatomy and Neuroscience and
| | - Nigel W Bunnett
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia, Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia, and
| |
Collapse
|
36
|
Secretagogin affects insulin secretion in pancreatic β-cells by regulating actin dynamics and focal adhesion. Biochem J 2016; 473:1791-803. [PMID: 27095850 PMCID: PMC4901359 DOI: 10.1042/bcj20160137] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/18/2016] [Indexed: 01/03/2023]
Abstract
Secretagogin (SCGN), a Ca2+-binding protein having six EF-hands, is selectively expressed in pancreatic β-cells and neuroendocrine cells. Previous studies suggested that SCGN enhances insulin secretion by functioning as a Ca2+-sensor protein, but the underlying mechanism has not been elucidated. The present study explored the mechanism by which SCGN enhances glucose-induced insulin secretion in NIT-1 insulinoma cells. To determine whether SCGN influences the first or second phase of insulin secretion, we examined how SCGN affects the kinetics of insulin secretion in NIT-1 cells. We found that silencing SCGN suppressed the second phase of insulin secretion induced by glucose and H2O2, but not the first phase induced by KCl stimulation. Recruitment of insulin granules in the second phase of insulin secretion was significantly impaired by knocking down SCGN in NIT-1 cells. In addition, we found that SCGN interacts with the actin cytoskeleton in the plasma membrane and regulates actin remodelling in a glucose-dependent manner. Since actin dynamics are known to regulate focal adhesion, a critical step in the second phase of insulin secretion, we examined the effect of silencing SCGN on focal adhesion molecules, including FAK (focal adhesion kinase) and paxillin, and the cell survival molecules ERK1/2 (extracellular-signal-regulated kinase 1/2) and Akt. We found that glucose- and H2O2-induced activation of FAK, paxillin, ERK1/2 and Akt was significantly blocked by silencing SCGN. We conclude that SCGN controls glucose-stimulated insulin secretion and thus may be useful in the therapy of Type 2 diabetes.
Collapse
|
37
|
Hewavitharana T, Wedegaertner PB. PAQR3 regulates Golgi vesicle fission and transport via the Gβγ-PKD signaling pathway. Cell Signal 2015; 27:2444-51. [PMID: 26327583 PMCID: PMC4684484 DOI: 10.1016/j.cellsig.2015.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/21/2015] [Accepted: 08/26/2015] [Indexed: 01/06/2023]
Abstract
Heterotrimeric G proteins function at diverse subcellular locations, in addition to canonical signaling at the plasma membrane (PM). Gβγ signals at the Golgi, via protein kinase D (PKD), to regulate fission of PM-destined vesicles. However, the mechanism by which Gβγ is regulated at the Golgi in this process remains elusive. Recent studies have revealed that PAQR3 (Progestin and AdipoQ Receptor 3), also called RKTG (Raf Kinase Trapping to the Golgi), interacts with the Gβ subunit and localizes Gβ to the Golgi thereby inhibiting Gβγ signaling at the PM. Herein we show that, in contrast to this inhibition of canonical Gβγ signaling at the PM, PAQR3 promotes Gβγ signaling at the Golgi. Expression of PAQR3 causes fragmentation of the Golgi, while a Gβ binding-deficient mutant of PAQR3 does not cause Golgi fragmentation. Also, a C-terminal fragment of GRK2 (GRK2ct), which interacts with Gβγ and inhibits Gβγ signaling, and gallein, a small molecule inhibitor of Gβγ, are both able to inhibit PAQR3-mediated Golgi fragmentation. Furthermore, a dominant negative form of PKD (PKD-DN) and a pharmacological inhibitor of PKD, Gö6976, also inhibit PAQR3-mediated fragmentation of the Golgi. Importantly, expression of the Gβ binding-deficient mutant of PAQR3 inhibits the constitutive transport of the model cargo protein VSV-G from the Golgi to the PM, indicating the involvement of PAQR3 in Golgi-to PM vesicle transport and a dominant negative role for this mutant. Collectively, these results reveal a novel role for the newly characterized, Golgi-localized PAQR3 in regulating Gβγ at the non-canonical subcellular location of the Golgi and thus for controlling Golgi-to-PM protein transport via the Gβγ-PKD signaling pathway.
Collapse
Affiliation(s)
- Thamara Hewavitharana
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, 233 S. 10th St., 839 BLSB, Philadelphia, PA 19107, United States.
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, 233 S. 10th St., 839 BLSB, Philadelphia, PA 19107, United States.
| |
Collapse
|
38
|
Giri L, Patel AK, Karunarathne WKA, Kalyanaraman V, Venkatesh KV, Gautam N. A G-protein subunit translocation embedded network motif underlies GPCR regulation of calcium oscillations. Biophys J 2015; 107:242-54. [PMID: 24988358 DOI: 10.1016/j.bpj.2014.05.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 05/06/2014] [Accepted: 05/13/2014] [Indexed: 11/24/2022] Open
Abstract
G-protein βγ subunits translocate reversibly from the plasma membrane to internal membranes on receptor activation. Translocation rates differ depending on the γ subunit type. There is limited understanding of the role of the differential rates of Gβγ translocation in modulating signaling dynamics in a cell. Bifurcation analysis of the calcium oscillatory network structure predicts that the translocation rate of a signaling protein can regulate the damping of system oscillation. Here, we examined whether the Gβγ translocation rate regulates calcium oscillations induced by G-protein-coupled receptor activation. Oscillations in HeLa cells expressing γ subunit types with different translocation rates were imaged and quantitated. The results show that differential Gβγ translocation rates can underlie the diversity in damping characteristics of calcium oscillations among cells. Mathematical modeling shows that a translocation embedded motif regulates damping of G-protein-mediated calcium oscillations consistent with experimental data. The current study indicates that such a motif may act as a tuning mechanism to design oscillations with varying damping patterns by using intracellular translocation of a signaling component.
Collapse
Affiliation(s)
- Lopamudra Giri
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Anilkumar K Patel
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - W K Ajith Karunarathne
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Vani Kalyanaraman
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - K V Venkatesh
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India.
| | - N Gautam
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Department of Genetics, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
39
|
Lo IC, Gupta V, Midde KK, Taupin V, Lopez-Sanchez I, Kufareva I, Abagyan R, Randazzo PA, Farquhar MG, Ghosh P. Activation of Gαi at the Golgi by GIV/Girdin imposes finiteness in Arf1 signaling. Dev Cell 2015; 33:189-203. [PMID: 25865347 DOI: 10.1016/j.devcel.2015.02.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 11/25/2014] [Accepted: 02/09/2015] [Indexed: 01/22/2023]
Abstract
A long-held tenet of heterotrimeric G protein signal transduction is that it is triggered by G protein-coupled receptors (GPCRs) at the PM. Here, we demonstrate that Gi is activated in the Golgi by GIV/Girdin, a non-receptor guanine-nucleotide exchange factor (GEF). GIV-dependent activation of Gi at the Golgi maintains the finiteness of the cyclical activation of ADP-ribosylation factor 1 (Arf1), a fundamental step in vesicle traffic in all eukaryotes. Several interactions with other major components of Golgi trafficking-e.g., active Arf1, its regulator, ArfGAP2/3, and the adaptor protein β-COP-enable GIV to coordinately regulate Arf1 signaling. When the GIV-Gαi pathway is selectively inhibited, levels of GTP-bound Arf1 are elevated and protein transport along the secretory pathway is delayed. These findings define a paradigm in non-canonical G protein signaling at the Golgi, which places GIV-GEF at the crossroads between signals gated by the trimeric G proteins and the Arf family of monomeric GTPases.
Collapse
Affiliation(s)
- I-Chung Lo
- Departments of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vijay Gupta
- Departments of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Krishna K Midde
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vanessa Taupin
- Departments of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Marilyn G Farquhar
- Departments of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Pradipta Ghosh
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
40
|
Malik S, deRubio RG, Trembley M, Irannejad R, Wedegaertner PB, Smrcka AV. G protein βγ subunits regulate cardiomyocyte hypertrophy through a perinuclear Golgi phosphatidylinositol 4-phosphate hydrolysis pathway. Mol Biol Cell 2015; 26:1188-98. [PMID: 25609085 PMCID: PMC4357516 DOI: 10.1091/mbc.e14-10-1476] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Gβγ regulation of the perinuclear Golgi PI4P pathway and a separate pathway at the PM is required for ET-1–stimulated hypertrophy, and the efficacy of Gβγ inhibition in preventing heart failure may be due, in part, to its blocking both of these pathways. We recently identified a novel GPCR-dependent pathway for regulation of cardiac hypertrophy that depends on Golgi phosphatidylinositol 4-phosphate (PI4P) hydrolysis by a specific isoform of phospholipase C (PLC), PLCε, at the nuclear envelope. How stimuli are transmitted from cell surface GPCRs to activation of perinuclear PLCε is not clear. Here we tested the role of G protein βγ subunits. Gβγ inhibition blocked ET-1–stimulated Golgi PI4P depletion in neonatal and adult ventricular myocytes. Blocking Gβγ at the Golgi inhibited ET-1–dependent PI4P depletion and nuclear PKD activation. Translocation of Gβγ to the Golgi stimulated perinuclear Golgi PI4P depletion and nuclear PKD activation. Finally, blocking Gβγ at the Golgi or PM blocked ET-1–dependent cardiomyocyte hypertrophy. These data indicate that Gβγ regulation of the perinuclear Golgi PI4P pathway and a separate pathway at the PM is required for ET-1–stimulated hypertrophy, and the efficacy of Gβγ inhibition in preventing heart failure maybe due in part to its blocking both these pathways.
Collapse
Affiliation(s)
- S Malik
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| | - R G deRubio
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| | - M Trembley
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| | - R Irannejad
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94158
| | - P B Wedegaertner
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - A V Smrcka
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| |
Collapse
|
41
|
Anitei M, Chenna R, Czupalla C, Esner M, Christ S, Lenhard S, Korn K, Meyenhofer F, Bickle M, Zerial M, Hoflack B. A high-throughput siRNA screen identifies genes that regulate mannose 6-phosphate receptor trafficking. J Cell Sci 2014; 127:5079-92. [PMID: 25278553 DOI: 10.1242/jcs.159608] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The delivery of newly synthesized soluble lysosomal hydrolases to the endosomal system is essential for lysosome function and cell homeostasis. This process relies on the proper trafficking of the mannose 6-phosphate receptors (MPRs) between the trans-Golgi network (TGN), endosomes and the plasma membrane. Many transmembrane proteins regulating diverse biological processes ranging from virus production to the development of multicellular organisms also use these pathways. To explore how cell signaling modulates MPR trafficking, we used high-throughput RNA interference (RNAi) to target the human kinome and phosphatome. Using high-content image analysis, we identified 127 kinases and phosphatases belonging to different signaling networks that regulate MPR trafficking and/or the dynamic states of the subcellular compartments encountered by the MPRs. Our analysis maps the MPR trafficking pathways based on enzymes regulating phosphatidylinositol phosphate metabolism. Furthermore, it reveals how cell signaling controls the biogenesis of post-Golgi tubular carriers destined to enter the endosomal system through a SRC-dependent pathway regulating ARF1 and RAC1 signaling and myosin II activity.
Collapse
Affiliation(s)
- Mihaela Anitei
- Biotechnology Center, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Ramu Chenna
- Biotechnology Center, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Cornelia Czupalla
- Biotechnology Center, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Milan Esner
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Building A1, 62500 Brno, Czech Republic
| | - Sara Christ
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Steffi Lenhard
- Biotechnology Center, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Kerstin Korn
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Felix Meyenhofer
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Marc Bickle
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Marino Zerial
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Bernard Hoflack
- Biotechnology Center, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| |
Collapse
|
42
|
Abstract
Heterotrimeric G proteins transduce the ligand binding of transmembrane G protein coupled receptors into a variety of intracellular signaling pathways. Recently, heterotrimeric Gβγ subunit signaling at the Golgi complex has been shown to regulate the formation of vesicular transport carriers that deliver cargo from the Golgi to the plasma membrane. In addition to vesicles, membrane tubules have also been shown to mediate export from the Golgi complex, which requires the activity of cytoplasmic phospholipase A2 (PLA2) enzyme activity. Through the use of an in vitro reconstitution assay with isolated Golgi complexes, we provide evidence that Gβ1γ2 signaling also stimulates Golgi membrane tubule formation. In addition, we show that an inhibitor of Gβγ activation of PLA2 enzymes inhibits in vitro Golgi membrane tubule formation. Additionally, purified Gβγ protein stimulates membrane tubules in the presence of low (sub-threshold) cytosol concentrations. Importantly, this Gβγ stimulation of Golgi membrane tubule formation was inhibited by treatment with the PLA2 antagonist ONO-RS-082. These studies indicate that Gβ1γ2 signaling activates PLA2 enzymes required for Golgi membrane tubule formation, thus establishing a new layer of regulation for this process.
Collapse
Affiliation(s)
- Marie E Bechler
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - William J Brown
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| |
Collapse
|
43
|
Coria AS, Masseroni ML, Díaz Añel AM. Regulation of PKD1-mediated Golgi to cell surface trafficking by Gαq subunits. Biol Cell 2013; 106:30-43. [PMID: 24175919 DOI: 10.1111/boc.201300052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 10/28/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND INFORMATION Heterotrimeric GTP-binding proteins play a key role in cell trafficking regulation. Above all, specific Gβγ subunits have been shown to be a major component of a signal transduction pathway, which also involves phospholipases C (PLC), protein kinases C (PKC) and D (PKD), whose main function is to regulate transport between Golgi and plasma membrane. It was the involvement of PLC which led us to study the role of the other member of this G protein family, the α subunits, in the regulation of membrane fission at the Golgi apparatus. RESULTS Among constitutive active (QL) variants of different G protein α subunit sub-families, only GαqQL subunits were able to induce Golgi fragmentation, a phenotype that mainly reflects a membrane fission increase at this organelle. This phenotype was not observed with a GαqQL palmitoylation mutant, showing the need for a membrane-bounded subunit. Besides, GαqQL-dependent Golgi fission was blocked by specific PLC and PKC inhibitors, and in the presence of a PKD1-kinase dead variant. In addition, GαqQL was the only α subunit capable of inducing PKD1 phosphorylation. Finally, Vesicular Stomatitis Virus thermosensitive mutant glycoprotein (VSVG tsO45) transport assays have demonstrated that GαqQL acts directly on Golgi membranes to regulate trafficking between this organelle and plasma membrane. CONCLUSIONS All these results indicate Gαq subunits for the first time as a regulator of PKD-mediated intracellular trafficking between Golgi apparatus and plasma membrane, opening new perspectives in the understanding of internal trafficking regulation by external signals through G protein-coupled receptors.
Collapse
Affiliation(s)
- A Soledad Coria
- Laboratory of Neurobiology and Cell Biology, Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), CONICET and Universidad Nacional de Córdoba. Friuli 2434, Barrio Parque Vélez Sarsfield, Córdoba 5016, Provincia de Córdoba, Argentina
| | | | | |
Collapse
|
44
|
Regulation of Golgi signaling and trafficking by the KDEL receptor. Histochem Cell Biol 2013; 140:395-405. [DOI: 10.1007/s00418-013-1130-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2013] [Indexed: 12/31/2022]
|
45
|
Oner SS, Vural A, Lanier SM. Translocation of activator of G-protein signaling 3 to the Golgi apparatus in response to receptor activation and its effect on the trans-Golgi network. J Biol Chem 2013; 288:24091-103. [PMID: 23770668 DOI: 10.1074/jbc.m112.444505] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Group II activators of G-protein signaling play diverse functional roles through their interaction with Gαi, Gαt, and Gαo via a G-protein regulatory (GPR) motif that serves as a docking site for Gα-GDP. We recently reported the regulation of the AGS3-Gαi signaling module by a cell surface, seven-transmembrane receptor. Upon receptor activation, AGS3 reversibly dissociates from the cell cortex, suggesting that it may function as a signal transducer with downstream signaling implications, and this question is addressed in the current report. In HEK-293 and COS-7 cells expressing the α2A/D-AR and Gαi3, receptor activation resulted in the translocation of endogenous AGS3 and AGS3-GFP from the cell cortex to a juxtanuclear region, where it co-localized with markers of the Golgi apparatus (GA). The agonist-induced translocation of AGS3 was reversed by the α2-AR antagonist rauwolscine. The TPR domain of AGS3 was required for agonist-induced translocation of AGS3 from the cell cortex to the GA, and the translocation was blocked by pertussis toxin pretreatment or by the phospholipase Cβ inhibitor U73122. Agonist-induced translocation of AGS3 to the GA altered the functional organization and protein sorting at the trans-Golgi network. The regulated movement of AGS3 between the cell cortex and the GA offers unexpected mechanisms for modulating protein secretion and/or endosome recycling events at the trans-Golgi network.
Collapse
Affiliation(s)
- Sukru S Oner
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | |
Collapse
|
46
|
Lau WW, Chan AS, Poon LS, Zhu J, Wong YH. Gβγ-mediated activation of protein kinase D exhibits subunit specificity and requires Gβγ-responsive phospholipase Cβ isoforms. Cell Commun Signal 2013; 11:22. [PMID: 23561540 PMCID: PMC3637504 DOI: 10.1186/1478-811x-11-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 03/22/2013] [Indexed: 11/29/2022] Open
Abstract
Background Protein kinase D (PKD) constitutes a novel family of serine/threonine protein kinases implicated in fundamental biological activities including cell proliferation, survival, migration, and immune responses. Activation of PKD in these cellular activities has been linked to many extracellular signals acting through antigen receptor engagement, receptor tyrosine kinases, as well as G protein-coupled receptors. In the latter case, it is generally believed that the Gα subunits of the Gq family are highly effective in mediating PKD activation, whereas little is known with regard to the ability of Gβγ dimers and other Gα subunits to stimulate PKD. It has been suggested that the interaction between Gβγ and the PH domain of PKD, or the Gβγ-induced PLCβ/PKC activity is critical for the induction of PKD activation. However, the relative contribution of these two apparently independent events to Gβγ-mediated PKD activation has yet to be addressed. Results In this report, we demonstrate that among various members in the four G protein families, only the Gα subunits of the Gq family effectively activate all the three PKD isoforms (PKD1/2/3), while Gα subunits of other G protein families (Gs, Gi, and G12) are ineffective. Though the Gα subunits of Gi family are unable to stimulate PKD, receptors linked to Gi proteins are capable of triggering PKD activation in cell lines endogenously expressing (HeLa cells and Jurkat T-cells) or exogenously transfected with (HEK293 cells) Gβγ-sensitive PLCβ2/3 isoforms. This indicates that the Gi-mediated PKD activation is dependent on the released Gβγ dimers upon stimulation. Further investigation on individual Gβγ combinations (i.e. Gβ1 with Gγ1–13) revealed that, even if they can stimulate the PLCβ activity in a comparable manner, only those Gβ1γ dimers with γ2, γ3, γ4, γ5, γ7, and γ10 can serve as effective activators of PKD. We also demonstrated that Gi-mediated PKD activation is essential for the SDF-1α-induced chemotaxis on Jurkat T-cells. Conclusions Our current report illustrates that Gβγ dimers from the Gi proteins may activate PKD in a PLCβ2/3-dependent manner, and the specific identities of Gγ components within Gβγ dimers may determine this stimulatory action.
Collapse
Affiliation(s)
- Winnie Wi Lau
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.
| | | | | | | | | |
Collapse
|
47
|
Khan SM, Sleno R, Gora S, Zylbergold P, Laverdure JP, Labbé JC, Miller GJ, Hébert TE. The expanding roles of Gβγ subunits in G protein-coupled receptor signaling and drug action. Pharmacol Rev 2013; 65:545-77. [PMID: 23406670 DOI: 10.1124/pr.111.005603] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Gβγ subunits from heterotrimeric G proteins perform a vast array of functions in cells with respect to signaling, often independently as well as in concert with Gα subunits. However, the eponymous term "Gβγ" does not do justice to the fact that 5 Gβ and 12 Gγ isoforms have evolved in mammals to serve much broader roles beyond their canonical roles in cellular signaling. We explore the phylogenetic diversity of Gβγ subunits with a view toward understanding these expanded roles in different cellular organelles. We suggest that the particular content of distinct Gβγ subunits regulates cellular activity, and that the granularity of individual Gβ and Gγ action is only beginning to be understood. Given the therapeutic potential of targeting Gβγ action, this larger view serves as a prelude to more specific development of drugs aimed at individual isoforms.
Collapse
Affiliation(s)
- Shahriar M Khan
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1303, Montréal, Québec H3G 1Y6, Canada
| | | | | | | | | | | | | | | |
Collapse
|
48
|
G-protein signaling leverages subunit-dependent membrane affinity to differentially control βγ translocation to intracellular membranes. Proc Natl Acad Sci U S A 2012; 109:E3568-77. [PMID: 23213235 DOI: 10.1073/pnas.1205345109] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Activation of G-protein heterotrimers by receptors at the plasma membrane stimulates βγ-complex dissociation from the α-subunit and translocation to internal membranes. This intermembrane movement of lipid-modified proteins is a fundamental but poorly understood feature of cell signaling. The differential translocation of G-protein βγ-subunit types provides a valuable experimental model to examine the movement of signaling proteins between membranes in a living cell. We used live cell imaging, mathematical modeling, and in vitro measurements of lipidated fluorescent peptide dissociation from vesicles to determine the mechanistic basis of the intermembrane movement and identify the interactions responsible for differential translocation kinetics in this family of evolutionarily conserved proteins. We found that the reversible translocation is mediated by the limited affinity of the βγ-subunits for membranes. The differential kinetics of the βγ-subunit types are determined by variations among a set of basic and hydrophobic residues in the γ-subunit types. G-protein signaling thus leverages the wide variation in membrane dissociation rates among different γ-subunit types to differentially control βγ-translocation kinetics in response to receptor activation. The conservation of primary structures of γ-subunits across mammalian species suggests that there can be evolutionary selection for primary structures that confer specific membrane-binding affinities and consequent rates of intermembrane movement.
Collapse
|
49
|
Cancino J, Luini A. Signaling Circuits on the Golgi Complex. Traffic 2012; 14:121-34. [DOI: 10.1111/tra.12022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/12/2012] [Accepted: 10/12/2012] [Indexed: 01/21/2023]
|
50
|
Gehart H, Goginashvili A, Beck R, Morvan J, Erbs E, Formentini I, De Matteis M, Schwab Y, Wieland F, Ricci R. The BAR Domain Protein Arfaptin-1 Controls Secretory Granule Biogenesis at the trans-Golgi Network. Dev Cell 2012; 23:756-68. [DOI: 10.1016/j.devcel.2012.07.019] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 07/02/2012] [Accepted: 07/24/2012] [Indexed: 12/29/2022]
|