1
|
McRae AN, Ticho AL, Liu Y, Ricardo-Silgado ML, Mangena NN, Jassir FF, Gonzalez-Izundegui D, Calderon G, Rohakhtar FR, Simon V, Li Y, Leggett C, Hurtado D, LaRusso N, Acosta AJ. Regulator of G-protein signaling expression in human intestinal enteroendocrine cells and potential role in satiety hormone secretion in health and obesity. EBioMedicine 2024; 107:105283. [PMID: 39142076 PMCID: PMC11367526 DOI: 10.1016/j.ebiom.2024.105283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Gut L-type enteroendocrine cells (EECs) are intestinal chemosensory cells that secrete satiety hormones GLP-1 and PYY in response to activation of G-protein coupled receptors (GPCRs) by luminal components of nutrient digestion and microbial fermentation. Regulator of G-protein Signaling (RGS) proteins are negative regulators of GPCR signaling. The expression profile of RGS in EECs, and their potential role in satiety hormone secretion and obesity is unknown. METHODS Transcriptomic profiling of RGS was completed in native colonic EECs was completed using single-cell RNA sequencing (scRNA-Seq) in lean and obesity, and human jejunal EECs with data obtained from a publicly available RNAseq dataset (GSE114853). RGS validation studies were completed using whole mucosal intestinal tissue obtained during endoscopy in 61 patients (n = 42 OB, n = 19 Lean); a subset of patients' postprandial plasma was assayed for GLP-1 and PYY. Ex vivo human intestinal cultures and in vitro NCI-H716 cells overexpressing RGS9 were exposed to GLP-1 secretagogues in conjunction with a nonselective RGS-inhibitor and assayed for GLP-1 secretion. FINDINGS Transcriptomic profiling of colonic and jejunal enteroendocrine cells revealed a unique RGS expression profile in EECs, and further within GLP-1+ L-type EECs. In obesity the RGS expression profile was altered in colonic EECs. Human gut RGS9 expression correlated positively with BMI and negatively with postprandial GLP-1 and PYY. RGS inhibition in human intestinal cultures increased GLP-1 release from EECs ex vivo. NCI-H716 cells overexpressing RGS9 displayed defective nutrient-stimulated GLP-1 secretion. INTERPRETATION This study introduces the expression profile of RGS in human EECs, alterations in obesity, and suggests a role for RGS proteins as modulators of GLP-1 and PYY secretion from intestinal EECs. FUNDING AA is supported by the NIH(C-Sig P30DK84567, K23 DK114460), a Pilot Award from the Mayo Clinic Center for Biomedical Discovery, and a Translational Product Development Fund from The Mayo Clinic Center for Clinical and Translational Science Office of Translational Practice in partnership with the University of Minnesota Clinical and Translational Science Institute.
Collapse
Affiliation(s)
- Alison N McRae
- Precision Medicine for Obesity Program and Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Alexander L Ticho
- Precision Medicine for Obesity Program and Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Yuanhang Liu
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Maria Laura Ricardo-Silgado
- Precision Medicine for Obesity Program and Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Nothando N Mangena
- Precision Medicine for Obesity Program and Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Fauzi Feris Jassir
- Precision Medicine for Obesity Program and Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Daniel Gonzalez-Izundegui
- Precision Medicine for Obesity Program and Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Gerardo Calderon
- Precision Medicine for Obesity Program and Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | - Vernadette Simon
- Center for Individualized Medicine (CIM), Mayo Clinic, Rochester, MN, USA
| | - Ying Li
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Cadman Leggett
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Daniela Hurtado
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition Mayo Clinic, Jacksonville, FL, USA
| | - Nicholas LaRusso
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Andres J Acosta
- Precision Medicine for Obesity Program and Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Philibert CE, Garcia-Marcos M. Smooth operator(s): dialing up and down neurotransmitter responses by G-protein regulators. Trends Cell Biol 2024:S0962-8924(24)00140-5. [PMID: 39054106 DOI: 10.1016/j.tcb.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
G-protein-coupled receptors (GPCRs) are essential mediators of neuromodulation and prominent pharmacological targets. While activation of heterotrimeric G-proteins (Gαβɣ) by GPCRs is essential in this process, much less is known about the postreceptor mechanisms that influence G-protein activity. Neurons express G-protein regulators that shape the amplitude and kinetics of GPCR-mediated synaptic responses. Although many of these operate by directly altering how G-proteins handle guanine-nucleotides enzymatically, recent discoveries have revealed alternative mechanisms by which GPCR-stimulated G-protein responses are modulated at the synapse. In this review, we cover the molecular basis for, and consequences of, the action of two G-protein regulators that do not affect the enzymatic activity of G-proteins directly: Gα inhibitory interacting protein (GINIP), which binds active Gα subunits, and potassium channel tetramerization domain-containing 12 (KCTD12), which binds active Gβγ subunits.
Collapse
Affiliation(s)
- Clementine E Philibert
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Mikel Garcia-Marcos
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; Department of Biology, College of Arts and Sciences, Boston University, Boston, MA 02115, USA.
| |
Collapse
|
3
|
Shen H, Yuan J, Tong D, Chen B, Yu E, Chen G, Peng C, Chang W, E J, Cao F. Regulator of G protein signaling 16 restrains apoptosis in colorectal cancer through disrupting TRAF6-TAB2-TAK1-JNK/p38 MAPK signaling. Cell Death Dis 2024; 15:438. [PMID: 38906869 PMCID: PMC11192724 DOI: 10.1038/s41419-024-06803-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024]
Abstract
Colorectal cancer (CRC) remains a major global cause of cancer-related mortality, lacking effective biomarkers and therapeutic targets. Revealing the critical pathogenic factors of CRC and the underlying mechanisms would offer potential therapeutic strategies for clinical application. G protein signaling (RGS) protein family modulators play essential role within regulating downstream signaling of GPCR receptors, with function in cancers unclear. Our study focused on the expression patterns of RGS proteins in CRC, identifying Regulator of G protein signaling 16 (RGS16) as a prospective diagnostic and therapeutic target. Analyzing 899 CRC tissues revealed elevated RGS16 levels, correlating with clinicopathological features and CRC prognosis by immunohistochemistry (IHC) combined with microarray. We confirmed the elevated RGS16 protein level in CRC, and found that patients with RGS16-high tumors exhibited decreased disease-specific survival (DSS) and disease-free survival (DFS) compared to those with low RGS16 expression. Functional assays demonstrated that RGS16 promoted the CRC progression, knockdown of RGS16 led to significantly increased apoptosis rates of CRC in vitro and in vivo. Notably, we also confirmed these phenotypes of RGS16 in organoids originated from resected primary human CRC tissues. Mechanistically, RGS16 restrained JNK/P38-mediated apoptosis in CRC cells through disrupting the recruitment of TAB2/TAK1 to TRAF6. This study provides insights into addressing the challenges posed by CRC, offering avenues for clinical translation.
Collapse
Affiliation(s)
- Hao Shen
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Environmental and Occupational Health, Naval Medical University, Shanghai, China
| | - Jie Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Health Management, Beidaihe Rest and Recuperation Center of PLA Joint Logistics Support Force, Qinhuangdao, China
| | - Dafeng Tong
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Bingchen Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Enda Yu
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Guanglei Chen
- Department of Health Management, Beidaihe Rest and Recuperation Center of PLA Joint Logistics Support Force, Qinhuangdao, China
| | - Cheng Peng
- Department of Health Management, Beidaihe Rest and Recuperation Center of PLA Joint Logistics Support Force, Qinhuangdao, China
| | - Wenjun Chang
- Department of Environmental and Occupational Health, Naval Medical University, Shanghai, China.
| | - Jifu E
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Fuao Cao
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China.
| |
Collapse
|
4
|
Leclerc NR, Dunne TM, Shrestha S, Johnson CP, Kelley JB. TOR signaling regulates GPCR levels on the plasma membrane and suppresses the Saccharomyces cerevisiae mating pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593412. [PMID: 38798445 PMCID: PMC11118302 DOI: 10.1101/2024.05.09.593412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Saccharomyces cerevisiae respond to mating pheromone through the GPCRs Ste2 and Ste3, which promote growth of a mating projection in response to ligand binding. This commitment to mating is nutritionally and energetically taxing, and so we hypothesized that the cell may suppress mating signaling during starvation. We set out to investigate negative regulators of the mating pathway in nutritionally depleted environments. Here, we report that nutrient deprivation led to loss of Ste2 from the plasma membrane. Recapitulating this effect with nitrogen starvation led us to hypothesize that it was due to TORC1 signaling. Rapamycin inhibition of TORC1 impacted membrane levels of all yeast GPCRs. Inhibition of TORC1 also dampened mating pathway output. Deletion analysis revealed that TORC1 repression leads to α-arrestin-directed CME through TORC2-Ypk1 signaling. We then set out to determine whether major downstream effectors of the TOR complexes also downregulate pathway output during mating. We found that autophagy contributes to pathway downregulation through analysis of strains lacking ATG8 . We also show that Ypk1 significantly reduced pathway output. Thus, both autophagy machinery and TORC2-Ypk1 signaling serve as attenuators of pheromone signaling during mating. Altogether, we demonstrate that the stress-responsive TOR complexes coordinate GPCR endocytosis and reduce the magnitude of pheromone signaling, in ligand-independent and ligand-dependent contexts. One Sentence Summary TOR signaling regulates the localization of all Saccharomyces cerevisiae GPCRs during starvation and suppress the mating pathway in the presence and absence of ligand.
Collapse
|
5
|
Torres-Rodriguez MD, Lee SG, Roy Choudhury S, Paul R, Selvam B, Shukla D, Jez JM, Pandey S. Structure-function analysis of plant G-protein regulatory mechanisms identifies key Gα-RGS protein interactions. J Biol Chem 2024; 300:107252. [PMID: 38569936 PMCID: PMC11061236 DOI: 10.1016/j.jbc.2024.107252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
Heterotrimeric GTP-binding protein alpha subunit (Gα) and its cognate regulator of G-protein signaling (RGS) protein transduce signals in eukaryotes spanning protists, amoeba, animals, fungi, and plants. The core catalytic mechanisms of the GTPase activity of Gα and the interaction interface with RGS for the acceleration of GTP hydrolysis seem to be conserved across these groups; however, the RGS gene is under low selective pressure in plants, resulting in its frequent loss. Our current understanding of the structural basis of Gα:RGS regulation in plants has been shaped by Arabidopsis Gα, (AtGPA1), which has a cognate RGS protein. To gain a comprehensive understanding of this regulation beyond Arabidopsis, we obtained the x-ray crystal structures of Oryza sativa Gα, which has no RGS, and Selaginella moellendorffi (a lycophyte) Gα that has low sequence similarity with AtGPA1 but has an RGS. We show that the three-dimensional structure, protein-protein interaction with RGS, and the dynamic features of these Gα are similar to AtGPA1 and metazoan Gα. Molecular dynamic simulation of the Gα-RGS interaction identifies the contacts established by specific residues of the switch regions of GTP-bound Gα, crucial for this interaction, but finds no significant difference due to specific amino acid substitutions. Together, our data provide valuable insights into the regulatory mechanisms of plant G-proteins but do not support the hypothesis of adaptive co-evolution of Gα:RGS proteins in plants.
Collapse
Affiliation(s)
| | - Soon Goo Lee
- Department of Molecular & Cellular Biology, Kennesaw State University, Kennesaw, Georgia, USA
| | - Swarup Roy Choudhury
- Donald Danforth Plant Science Center, St Louis, Missouri, USA; Department of Biology, Indian Institute of Science Education and Research, Tirupati, India
| | - Rabindranath Paul
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Balaji Selvam
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Diwakar Shukla
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Joseph M Jez
- Department of Biology, Washington University in St Louis, St Louis, Missouri, USA
| | - Sona Pandey
- Donald Danforth Plant Science Center, St Louis, Missouri, USA.
| |
Collapse
|
6
|
Ding X, Li X, Jiang Y, Li Y, Li H, Shang L, Feng G, Zhang H, Xu Z, Yang L, Li B, Zhao RC. RGS20 promotes non-small cell lung carcinoma proliferation via autophagy activation and inhibition of the PKA-Hippo signaling pathway. Cancer Cell Int 2024; 24:93. [PMID: 38431606 PMCID: PMC10909273 DOI: 10.1186/s12935-024-03282-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/24/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Novel therapeutic targets are urgently needed for treating drug-resistant non-small cell lung cancer (NSCLC) and overcoming drug resistance to molecular-targeted therapies. Regulator of G protein signaling 20 (RGS20) is identified as an upregulated factor in many cancers, yet its specific role and the mechanism through which RGS20 functions in NSCLC remain unclear. Our study aimed to identify the role of RGS20 in NSCLC prognosis and delineate associated cellular and molecular pathways. METHODS Immunohistochemistry and lung cancer tissue microarray were used to verify the expression of RGS20 between NSCLC patients. CCK8 and cell cloning were conducted to determine the proliferation ability of H1299 and Anip973 cells in vitro. Furthermore, Transcriptome sequencing was performed to show enrichment genes and pathways. Immunofluorescence was used to detect the translocation changes of YAP to nucleus. Western blotting demonstrated different expressions of autophagy and the Hippo-PKA signal pathway. In vitro and in vivo experiments verified whether overexpression of RGS20 affect the proliferation and autophagy of NSCLC through regulating the Hippo pathway. RESULTS The higher RGS20 expression was found to be significantly correlated with a poorer five-year survival rate. Further, RGS20 accelerated cell proliferation by increasing autophagy. Transcriptomic sequencing suggested the involvement of the Hippo signaling pathway in the action of RGS20 in NSCLC. RGS20 activation reduced YAP phosphorylation and facilitated its nuclear translocation. Remarkably, inhibiting Hippo signaling with GA-017 promoted cell proliferation and activated autophagy in RGS20 knock-down cells. However, forskolin, a GPCR activator, increased YAP phosphorylation and reversed the promoting effect of RGS20 in RGS20-overexpressing cells. Lastly, in vivo experiments further confirmed role of RGS20 in aggravating tumorigenicity, as its overexpression increased NSCLC cell proliferation. CONCLUSION Our findings indicate that RGS20 drives NSCLC cell proliferation by triggering autophagy via the inhibition of PKA-Hippo signaling. These insights support the role of RGS20 as a promising novel molecular marker and a target for future targeted therapies in lung cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Ding
- School of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University, Qingdao, China
| | - Xiaoxia Li
- School of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University, Qingdao, China
| | - Yanxia Jiang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yujun Li
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Li
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lipeng Shang
- School of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University, Qingdao, China
| | - Guilin Feng
- School of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University, Qingdao, China
| | - Huhu Zhang
- School of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University, Qingdao, China
| | - Ziyuan Xu
- School of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University, Qingdao, China
| | - Lina Yang
- School of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University, Qingdao, China.
| | - Bing Li
- School of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University, Qingdao, China.
| | - Robert Chunhua Zhao
- School of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University, Qingdao, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
- School of Life Sciences, Shanghai University, Shanghai, China.
| |
Collapse
|
7
|
Nürnberg B, Beer-Hammer S, Reisinger E, Leiss V. Non-canonical G protein signaling. Pharmacol Ther 2024; 255:108589. [PMID: 38295906 DOI: 10.1016/j.pharmthera.2024.108589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/18/2023] [Accepted: 01/08/2024] [Indexed: 02/17/2024]
Abstract
The original paradigm of classical - also referred to as canonical - cellular signal transduction of heterotrimeric G proteins (G protein) is defined by a hierarchical, orthograde interaction of three players: the agonist-activated G protein-coupled receptor (GPCR), which activates the transducing G protein, that in turn regulates its intracellular effectors. This receptor-transducer-effector concept was extended by the identification of regulators and adapters such as the regulators of G protein signaling (RGS), receptor kinases like βARK, or GPCR-interacting arrestin adapters that are integrated into this canonical signaling process at different levels to enable fine-tuning. Finally, the identification of atypical signaling mechanisms of classical regulators, together with the discovery of novel modulators, added a new and fascinating dimension to the cellular G protein signal transduction. This heterogeneous group of accessory G protein modulators was coined "activators of G protein signaling" (AGS) proteins and plays distinct roles in canonical and non-canonical G protein signaling pathways. AGS proteins contribute to the control of essential cellular functions such as cell development and division, intracellular transport processes, secretion, autophagy or cell movements. As such, they are involved in numerous biological processes that are crucial for diseases, like diabetes mellitus, cancer, and stroke, which represent major health burdens. Although the identification of a large number of non-canonical G protein signaling pathways has broadened the spectrum of this cellular communication system, their underlying mechanisms, functions, and biological effects are poorly understood. In this review, we highlight and discuss atypical G protein-dependent signaling mechanisms with a focus on inhibitory G proteins (Gi) involved in canonical and non-canonical signal transduction, review recent developments and open questions, address the potential of new approaches for targeted pharmacological interventions.
Collapse
Affiliation(s)
- Bernd Nürnberg
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, and ICePhA Mouse Clinic, University of Tübingen, Wilhelmstraße 56, D-72074 Tübingen, Germany.
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, and ICePhA Mouse Clinic, University of Tübingen, Wilhelmstraße 56, D-72074 Tübingen, Germany
| | - Ellen Reisinger
- Gene Therapy for Hearing Impairment Group, Department of Otolaryngology - Head & Neck Surgery, University of Tübingen Medical Center, Elfriede-Aulhorn-Straße 5, D-72076 Tübingen, Germany
| | - Veronika Leiss
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, and ICePhA Mouse Clinic, University of Tübingen, Wilhelmstraße 56, D-72074 Tübingen, Germany
| |
Collapse
|
8
|
Garcia-Marcos M. Heterotrimeric G protein signaling without GPCRs: The Gα-binding-and-activating (GBA) motif. J Biol Chem 2024; 300:105756. [PMID: 38364891 PMCID: PMC10943482 DOI: 10.1016/j.jbc.2024.105756] [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: 10/28/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
Heterotrimeric G proteins (Gαβγ) are molecular switches that relay signals from 7-transmembrane receptors located at the cell surface to the cytoplasm. The function of these receptors is so intimately linked to heterotrimeric G proteins that they are named G protein-coupled receptors (GPCRs), showcasing the interdependent nature of this archetypical receptor-transducer axis of transmembrane signaling in eukaryotes. It is generally assumed that activation of heterotrimeric G protein signaling occurs exclusively by the action of GPCRs, but this idea has been challenged by the discovery of alternative mechanisms by which G proteins can propagate signals in the cell. This review will focus on a general principle of G protein signaling that operates without the direct involvement of GPCRs. The mechanism of G protein signaling reviewed here is mediated by a class of G protein regulators defined by containing an evolutionarily conserved sequence named the Gα-binding-and-activating (GBA) motif. Using the best characterized proteins with a GBA motif as examples, Gα-interacting vesicle-associated protein (GIV)/Girdin and dishevelled-associating protein with a high frequency of leucine residues (DAPLE), this review will cover (i) the mechanisms by which extracellular cues not relayed by GPCRs promote the coupling of GBA motif-containing regulators with G proteins, (ii) the structural and molecular basis for how GBA motifs interact with Gα subunits to facilitate signaling, (iii) the relevance of this mechanism in different cellular and pathological processes, including cancer and birth defects, and (iv) strategies to manipulate GBA-G protein coupling for experimental therapeutics purposes, including the development of rationally engineered proteins and chemical probes.
Collapse
Affiliation(s)
- Mikel Garcia-Marcos
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA; Department of Biology, College of Arts & Sciences, Boston University, Boston, Massachusetts, USA.
| |
Collapse
|
9
|
Wenbo L, Liangyu X, Zhiyong L, Gongchang Y, Yuanzhen C, Bin S. Status and trends of RGS16 based on data visualization analysis: A review. Medicine (Baltimore) 2024; 103:e36981. [PMID: 38363937 PMCID: PMC10869050 DOI: 10.1097/md.0000000000036981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/22/2023] [Indexed: 02/18/2024] Open
Abstract
G-protein signaling regulator 16 (RGS16) has been confirmed that RGS16 is associated with cancer, neurodegenerative diseases, and cardiovascular diseases. Moreover, many studies have shown that RGS16 can be used as a biomarker for cancer diagnosis and prognosis. We used CiteSpace and VOS viewer software to perform a bibliometric analysis of 290 publications in the core collection of Web of Science. All the articles come from 399 institutions, including 618 authors, 179 journals, 40 countries, 115 keywords, 1 language, two types of papers, and reviews. The United States has the largest number of publications. The Research Center of Allergy and Infectious Diseases (NIAID) publishes the most papers, Emory University is the most recent of all institutions with the most recent results in the RGS16 study. Cell biology is the most studied discipline, and the most studied topic is migration. Drury published RGS16-related articles with the most citations (n = 15), and Berman published articles with the most citations (n = 106). The biological applications of RGS16 are currently a hot area of RGS16 research, including inflammation, cancer, ulcerative colitis, metabolic acidosis, platelet activation, and thrombosis. The current scientometrics study provides an overview of RGS16 research from 1995 to 2022. This study provides an overview of current and potential future research hotspots in the field of RGS16 and can be used as a resource for interested researchers.
Collapse
Affiliation(s)
- Liu Wenbo
- Bone Biomechanics Engineering Laboratory of Shandong Province, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xie Liangyu
- Bone Biomechanics Engineering Laboratory of Shandong Province, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Lu Zhiyong
- Bone Biomechanics Engineering Laboratory of Shandong Province, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yu Gongchang
- Bone Biomechanics Engineering Laboratory of Shandong Province, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Chen Yuanzhen
- Bone Biomechanics Engineering Laboratory of Shandong Province, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shi Bin
- Bone Biomechanics Engineering Laboratory of Shandong Province, Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Shandong Traditional Chinese Medicine University, Jinan, Shandong Province, China
| |
Collapse
|
10
|
Qi D, Liu C, Zhang Z, Liu X, Kang P. Construction of a Lung Adenocarcinoma Prognostic Model Utilizing Serine and Glycine Metabolism-Related Genes. J Proteome Res 2024; 23:797-808. [PMID: 38212294 DOI: 10.1021/acs.jproteome.3c00700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
The objective of this study was to construct a prognostic model by utilizing serine/glycine metabolism-related genes (SGMGs), thus establishing a risk score for lung adenocarcinoma (LUAD). Based on the TCGA-LUAD and SGMG data set, two subtypes with different SGMG expression levels were identified by clustering analysis. Thirteen differential expression genes were used to construct RiskScore by Cox regression. GSE72094 data set was used for validation. The survival characteristics, immune features, and potential benefits of chemotherapy drugs were analyzed for two risk groups. RiskScore was constructed based on the genes ABCC12, RIC3, CYP4B1, SFTPB, CACNA2D2, IGF2BP1, NTSR1, DKK1, CREG2, PITX3, RGS20, FETUB, and IGFBP1. Patients in the low-risk (LR) group exhibited a superior overall survival. In addition, aDCs, iDSs, mast cells, neutrophils, HLA, and type II IFN were more abundant in the LR group with higher IPS scores and lower TIDE scores. In contrast, NK cells, APC coinhibition, and MHC-I were more common in the high-risk (HR) group, which may be more sensitive to chemotherapy drugs such as cisplatin, oxaliplatin, and nilotinib. RiskScore was a promising biomarker that can be used to distinguish LUAD prognosis, immune features, and sensitivity to chemotherapy drugs.
Collapse
Affiliation(s)
- Dongdong Qi
- Department of Thoracic Surgery, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Chengjun Liu
- Department of Thoracic Surgery, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Zuwang Zhang
- Department of Thoracic Surgery, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Xun Liu
- Department of Thoracic Surgery, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Poming Kang
- Department of Thoracic Surgery, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| |
Collapse
|
11
|
Luebbers A, Gonzalez-Hernandez AJ, Zhou M, Eyles SJ, Levitz J, Garcia-Marcos M. Dissecting the molecular basis for the modulation of neurotransmitter GPCR signaling by GINIP. Structure 2024; 32:47-59.e7. [PMID: 37989308 PMCID: PMC10872408 DOI: 10.1016/j.str.2023.10.010] [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: 05/04/2023] [Revised: 09/23/2023] [Accepted: 10/25/2023] [Indexed: 11/23/2023]
Abstract
It is well established that G-protein-coupled receptors (GPCRs) stimulated by neurotransmitters are critical for neuromodulation. Much less is known about how heterotrimeric G-protein (Gαβγ) regulation after receptor-mediated activation contributes to neuromodulation. Recent evidence indicates that the neuronal protein GINIP shapes GPCR inhibitory neuromodulation via a unique mechanism of G-protein regulation that controls pain and seizure susceptibility. However, the molecular basis of this mechanism remains ill-defined because the structural determinants of GINIP responsible for binding and regulating G proteins are not known. Here, we combined hydrogen-deuterium exchange mass spectrometry, computational structure predictions, biochemistry, and cell-based biophysical assays to demonstrate an effector-like binding mode of GINIP to Gαi. Specific amino acids of GINIP's PHD domain first loop are essential for G-protein binding and subsequent regulation of Gαi-GTP and Gβγ signaling upon neurotransmitter GPCR stimulation. In summary, these findings shed light onto the molecular basis for a post-receptor mechanism of G-protein regulation that fine-tunes inhibitory neuromodulation.
Collapse
Affiliation(s)
- Alex Luebbers
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | | | - Myles Zhou
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Stephen J Eyles
- Mass Spectrometry Core Facility, Institute for Applied Life Sciences (IALS), University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10064, USA; Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mikel Garcia-Marcos
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; Department of Biology, College of Arts & Sciences, Boston University, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Ham D, Ahn D, Chung C, Chung KY. Isolation and conformational analysis of the Gα α-helical domain. Biochem Biophys Res Commun 2023; 685:149153. [PMID: 37913692 DOI: 10.1016/j.bbrc.2023.149153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
Heterotrimeric G proteins (G proteins), composed of Gα, Gβ, and Gγ subunits, are the major downstream signaling molecules of the G protein-coupled receptors. Upon activation, Gα undergoes conformational changes both in the Ras-like domain (RD) and the α-helical domain (AHD), leading to the dissociation of Gα from Gβγ and subsequent regulation of downstream effector proteins. Gα RD mediate the most of classical functions of Gα. However, the role of Gα AHD is relatively not well elucidated despite its much higher sequence differences between Gα subtypes than those between Gα RD. Here, we isolated AHD from Gαs, Gαi1, and Gαq to provide tools for examining Gα AHD. We investigated the conformational dynamics of the isolated Gα AHD compared to those of the GDP-bound Gα. The results showed higher local conformational dynamics of Gα AHD not only at the domain interfaces but also in regions further away from the domain interfaces. This finding is consistent with the conformation of Gα AHD in the receptor-bound nucleotide-free state. Therefore, the isolated Gα AHD could provide a platform for studying the functions of Gα AHD, such as identification of the Gα AHD-binding proteins.
Collapse
Affiliation(s)
- Donghee Ham
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Donghoon Ahn
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Chiwoon Chung
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
13
|
Wang B, Jiang B, Du L, Chen W, Zhang Q, Chen W, Ding M, Cao W, Gao J, Deng Y, Fu Y, Li Y, Xiao Y, Diao W, Guo H. Tumor-intrinsic RGS1 potentiates checkpoint blockade response via ATF3-IFNGR1 axis. Oncoimmunology 2023; 12:2279800. [PMID: 38264343 PMCID: PMC10804258 DOI: 10.1080/2162402x.2023.2279800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/01/2023] [Indexed: 01/25/2024] Open
Abstract
Background Non-responsiveness is a major barrier in current cancer immune checkpoint blockade therapies, and the mechanism has not been elucidated yet. Therefore, it is necessary to discover the mechanism and biomarkers of tumor immunotherapeutic resistance. Methods Bioinformatics analysis was performed based on CD8+ T cell infiltration in multiple tumor databases to screen out genes related to anti-tumor immunity. Associations between Regulator of G-protein signaling 1 (RGS1) and IFNγ-STAT1 signaling, and MHCI antigen presentation pathway were examined by RT-qPCR, western blotting, and flow cytometry. The modulatory mechanisms of RGS1 were investigated via CHIP-qPCR and dual-luciferase assay. The clinical and therapeutic implications of RGS1 were comprehensively investigated using tumor cell lines, mouse models, and clinical samples receiving immunotherapy. Results RGS1 was identified as the highest gene positively correlated with immunogenicity among RGS family. Inhibition of RGS1 in neoplastic cells dampened anti-tumor immune response and elicited resistance to immunotherapy in both renal and lung murine subcutaneous tumors. Mechanistically, RGS1 enhanced the binding of activating transcription factor 3 (ATF3) to the promoter of interferon gamma receptor 1 (IFNGR1), activated STAT1 and the subsequent expression of IFNγ-inducible genes, especially CXCL9 and MHC class I (MHCI), thereby influenced CD8+ T cell infiltration and antigen presentation and processing. Clinically, lower expression level of RGS1 was associated with resistance of PD1 inhibition therapy and shortened progression-free survival among 21 NSCLC patients receiving immunotherapy. Conclusions Together, these findings uncover a novel mechanism that elicits immunotherapy resistance and highlight the function of tumor-intrinsic RGS1, which brings new insights for future strategies to sensitize anti-PD1 immunotherapy.
Collapse
Affiliation(s)
- Baojun Wang
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Bo Jiang
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Lin Du
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Southeast University Medical School, Nanjing, Jiangsu, China
| | - Wenyuan Chen
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qing Zhang
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wei Chen
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Meng Ding
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenmin Cao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jie Gao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yongming Deng
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yan Li
- Department of Respiratory and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yonglong Xiao
- Department of Respiratory and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Wenli Diao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
14
|
Park JC, Luebbers A, Dao M, Semeano A, Nguyen AM, Papakonstantinou MP, Broselid S, Yano H, Martemyanov KA, Garcia-Marcos M. Fine-tuning GPCR-mediated neuromodulation by biasing signaling through different G protein subunits. Mol Cell 2023; 83:2540-2558.e12. [PMID: 37390816 PMCID: PMC10527995 DOI: 10.1016/j.molcel.2023.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/28/2023] [Accepted: 06/02/2023] [Indexed: 07/02/2023]
Abstract
G-protein-coupled receptors (GPCRs) mediate neuromodulation through the activation of heterotrimeric G proteins (Gαβγ). Classical models depict that G protein activation leads to a one-to-one formation of Gα-GTP and Gβγ species. Each of these species propagates signaling by independently acting on effectors, but the mechanisms by which response fidelity is ensured by coordinating Gα and Gβγ responses remain unknown. Here, we reveal a paradigm of G protein regulation whereby the neuronal protein GINIP (Gα inhibitory interacting protein) biases inhibitory GPCR responses to favor Gβγ over Gα signaling. Tight binding of GINIP to Gαi-GTP precludes its association with effectors (adenylyl cyclase) and, simultaneously, with regulator-of-G-protein-signaling (RGS) proteins that accelerate deactivation. As a consequence, Gαi-GTP signaling is dampened, whereas Gβγ signaling is enhanced. We show that this mechanism is essential to prevent the imbalances of neurotransmission that underlie increased seizure susceptibility in mice. Our findings reveal an additional layer of regulation within a quintessential mechanism of signal transduction that sets the tone of neurotransmission.
Collapse
Affiliation(s)
- Jong-Chan Park
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Alex Luebbers
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Maria Dao
- U.F. Scripps Biomedical Research, University of Florida, Jupiter, FL 33458, USA
| | - Ana Semeano
- Department of Pharmaceutical Sciences, Center for Drug Discovery, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA 02115, USA
| | - Anh Minh Nguyen
- Department of Pharmaceutical Sciences, Center for Drug Discovery, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA 02115, USA
| | - Maria P Papakonstantinou
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Stefan Broselid
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Hideaki Yano
- Department of Pharmaceutical Sciences, Center for Drug Discovery, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA 02115, USA
| | | | - Mikel Garcia-Marcos
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; Department of Biology, College of Arts & Sciences, Boston University, Boston, MA 02115, USA.
| |
Collapse
|
15
|
Schalamun M, Molin EM, Schmoll M. RGS4 impacts carbohydrate and siderophore metabolism in Trichoderma reesei. BMC Genomics 2023; 24:372. [PMID: 37400774 PMCID: PMC10316542 DOI: 10.1186/s12864-023-09467-2] [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: 12/15/2022] [Accepted: 06/20/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Adaptation to complex, rapidly changing environments is crucial for evolutionary success of fungi. The heterotrimeric G-protein pathway belongs to the most important signaling cascades applied for this task. In Trichoderma reesei, enzyme production, growth and secondary metabolism are among the physiological traits influenced by the G-protein pathway in a light dependent manner. RESULTS Here, we investigated the function of the SNX/H-type regulator of G-protein signaling (RGS) protein RGS4 of T. reesei. We show that RGS4 is involved in regulation of cellulase production, growth, asexual development and oxidative stress response in darkness as well as in osmotic stress response in the presence of sodium chloride, particularly in light. Transcriptome analysis revealed regulation of several ribosomal genes, six genes mutated in RutC30 as well as several genes encoding transcription factors and transporters. Importantly, RGS4 positively regulates the siderophore cluster responsible for fusarinine C biosynthesis in light. The respective deletion mutant shows altered growth on nutrient sources related to siderophore production such as ornithine or proline in a BIOLOG phenotype microarray assay. Additionally, growth on storage carbohydrates as well as several intermediates of the D-galactose and D-arabinose catabolic pathway is decreased, predominantly in light. CONCLUSIONS We conclude that RGS4 mainly operates in light and targets plant cell wall degradation, siderophore production and storage compound metabolism in T. reesei.
Collapse
Affiliation(s)
- Miriam Schalamun
- AIT Austrian Institute of Technology GmbH, Bioresources Unit, Center for Health & Bioresources, Konrad Lorenz Strasse 24, Tulln, 3430 Austria
| | - Eva Maria Molin
- AIT Austrian Institute of Technology GmbH, Bioresources Unit, Center for Health & Bioresources, Konrad Lorenz Strasse 24, Tulln, 3430 Austria
| | - Monika Schmoll
- AIT Austrian Institute of Technology GmbH, Bioresources Unit, Center for Health & Bioresources, Konrad Lorenz Strasse 24, Tulln, 3430 Austria
- Division of Terrestrial Ecosystem Research, Centre of Microbiology and Ecosystem Science, University of Vienna, Djerassiplatz 1, Vienna, 1030 Austria
| |
Collapse
|
16
|
Li L, Xu Q, Tang C. RGS proteins and their roles in cancer: friend or foe? Cancer Cell Int 2023; 23:81. [PMID: 37118788 PMCID: PMC10148553 DOI: 10.1186/s12935-023-02932-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023] Open
Abstract
As negative modulators of G-protein-coupled receptors (GPCRs) signaling, regulators of G protein signaling (RGS) proteins facilitate various downstream cellular signalings through regulating kinds of heterotrimeric G proteins by stimulating the guanosine triphosphatase (GTPase) activity of G-protein α (Gα) subunits. The expression of RGS proteins is dynamically and precisely mediated by several different mechanisms including epigenetic regulation, transcriptional regulation -and post-translational regulation. Emerging evidence has shown that RGS proteins act as important mediators in controlling essential cellular processes including cell proliferation, survival -and death via regulating downstream cellular signaling activities, indicating that RGS proteins are fundamentally involved in sustaining normal physiological functions and dysregulation of RGS proteins (such as aberrant expression of RGS proteins) is closely associated with pathologies of many diseases such as cancer. In this review, we summarize the molecular mechanisms governing the expression of RGS proteins, and further discuss the relationship of RGS proteins and cancer.
Collapse
Affiliation(s)
- Lin Li
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China
| | - Qiang Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China
| | - Chao Tang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
17
|
Luebbers A, Zhou M, Eyles SJ, Garcia-Marcos M. Dissecting the molecular basis for the modulation of neurotransmitter GPCR signaling by GINIP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.20.537566. [PMID: 37131787 PMCID: PMC10153262 DOI: 10.1101/2023.04.20.537566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
It is well-established that activation of heterotrimeric G-proteins (Gαβγ) by G-protein-coupled receptors (GPCRs) stimulated by neurotransmitters is a key mechanism underlying neuromodulation. Much less is known about how G-protein regulation after receptor-mediated activation contributes to neuromodulation. Recent evidence indicates that the neuronal protein GINIP shapes GPCR inhibitory neuromodulation via a unique mechanism of G-protein regulation that controls neurological processes like pain and seizure susceptibility. However, the molecular basis of this mechanism remains ill-defined because the structural determinants of GINIP responsible for binding Gαi subunits and regulating G-protein signaling are not known. Here, we combined hydrogen-deuterium exchange mass-spectrometry, protein folding predictions, bioluminescence resonance energy transfer assays, and biochemical experiments to identify the first loop of the PHD domain of GINIP as an obligatory requirement for Gαi binding. Surprisingly, our results support a model in which GINIP undergoes a long-range conformational change to accommodate Gαi binding to this loop. Using cell-based assays, we demonstrate that specific amino acids in the first loop of the PHD domain are essential for the regulation of Gαi-GTP and free Gβγ signaling upon neurotransmitter GPCR stimulation. In summary, these findings shed light onto the molecular basis for a post-receptor mechanism of G-protein regulation that fine-tunes inhibitory neuromodulation.
Collapse
Affiliation(s)
- Alex Luebbers
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Myles Zhou
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Stephen J Eyles
- Mass Spectrometry Core Facility, Institute for Applied Life Sciences (IALS), University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Mikel Garcia-Marcos
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
- Department of Biology, College of Arts & Sciences, Boston University, Boston, MA 02115, USA
| |
Collapse
|
18
|
Koval A, Larasati YA, Savitsky M, Solis GP, Good JM, Quinodoz M, Rivolta C, Superti-Furga A, Katanaev VL. In-depth molecular profiling of an intronic GNAO1 mutant as the basis for personalized high-throughput drug screening. MED 2023; 4:311-325.e7. [PMID: 37001522 DOI: 10.1016/j.medj.2023.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND The GNAO1 gene, encoding the major neuronal G protein Gαo, is mutated in a subset of pediatric encephalopathies. Most such mutations consist of missense variants. METHODS In this study, we present a precision medicine workflow combining next-generation sequencing (NGS) diagnostics, molecular etiology analysis, and personalized drug discovery. FINDINGS We describe a patient carrying a de novo intronic mutation (NM_020988.3:c.724-8G>A), leading to epilepsy-negative encephalopathy with motor dysfunction from the second decade. Our data show that this mutation creates a novel splice acceptor site that in turn causes an in-frame insertion of two amino acid residues, Pro-Gln, within the regulatory switch III region of Gαo. This insertion misconfigures the switch III loop and creates novel interactions with the catalytic switch II region, resulting in increased GTP uptake, defective GTP hydrolysis, and aberrant interactions with effector proteins. In contrast, intracellular localization, Gβγ interactions, and G protein-coupled receptor (GPCR) coupling of the Gαo[insPQ] mutant protein remain unchanged. CONCLUSIONS This in-depth analysis characterizes the heterozygous c.724-8G>A mutation as partially dominant negative, providing clues to the molecular etiology of this specific pathology. Further, this analysis allows us to establish and validate a high-throughput screening platform aiming at identifying molecules that could correct the aberrant biochemical functions of the mutant Gαo. FUNDING This work was supported by the Joint Seed Money Funding scheme between the University of Geneva and the Hebrew University of Jerusalem.
Collapse
Affiliation(s)
- Alexey Koval
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Yonika A Larasati
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Mikhail Savitsky
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Gonzalo P Solis
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Jean-Marc Good
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Mathieu Quinodoz
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland; Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Carlo Rivolta
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland; Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Andrea Superti-Furga
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - Vladimir L Katanaev
- Translational Research Center in Oncohaematology, Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690090 Vladivostok, Russia.
| |
Collapse
|
19
|
Montañez-Miranda C, Perszyk RE, Harbin NH, Okalova J, Ramineni S, Traynelis SF, Hepler JR. Functional Assessment of Cancer-Linked Mutations in Sensitive Regions of Regulators of G Protein Signaling Predicted by Three-Dimensional Missense Tolerance Ratio Analysis. Mol Pharmacol 2023; 103:21-37. [PMID: 36384958 PMCID: PMC10955721 DOI: 10.1124/molpharm.122.000614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/04/2022] [Accepted: 10/18/2022] [Indexed: 11/18/2022] Open
Abstract
Regulators of G protein signaling (RGS) proteins modulate G protein-coupled receptor (GPCR) signaling by acting as negative regulators of G proteins. Genetic variants in RGS proteins are associated with many diseases, including cancers, although the impact of these mutations on protein function is uncertain. Here we analyze the RGS domains of 15 RGS protein family members using a novel bioinformatic tool that measures the missense tolerance ratio (MTR) using a three-dimensional (3D) structure (3DMTR). Subsequent permutation analysis can define the protein regions that are most significantly intolerant (P < 0.05) in each dataset. We further focused on RGS14, RGS10, and RGS4. RGS14 exhibited seven significantly tolerant and seven significantly intolerant residues, RGS10 had six intolerant residues, and RGS4 had eight tolerant and six intolerant residues. Intolerant and tolerant-control residues that overlap with pathogenic cancer mutations reported in the COSMIC cancer database were selected to define the functional phenotype. Using complimentary cellular and biochemical approaches, proteins were tested for effects on GPCR-Gα activation, Gα binding properties, and downstream cAMP levels. Identified intolerant residues with reported cancer-linked mutations RGS14-R173C/H and RGS4-K125Q/E126K, and tolerant RGS14-S127P and RGS10-S64T resulted in a loss-of-function phenotype in GPCR-G protein signaling activity. In downstream cAMP measurement, tolerant RGS14-D137Y and RGS10-S64T and intolerant RGS10-K89M resulted in change of function phenotypes. These findings show that 3DMTR identified intolerant residues that overlap with cancer-linked mutations cause phenotypic changes that negatively impact GPCR-G protein signaling and suggests that 3DMTR is a potentially useful bioinformatics tool for predicting functionally important protein residues. SIGNIFICANCE STATEMENT: Human genetic variant/mutation information has expanded rapidly in recent years, including cancer-linked mutations in regulator of G protein signaling (RGS) proteins. However, experimental testing of the impact of this vast catalogue of mutations on protein function is not feasible. We used the novel bioinformatics tool three-dimensional missense tolerance ratio (3DMTR) to define regions of genetic intolerance in RGS proteins and prioritize which cancer-linked mutants to test. We found that 3DMTR more accurately classifies loss-of-function mutations in RGS proteins than other databases thereby offering a valuable new research tool.
Collapse
Affiliation(s)
- Carolina Montañez-Miranda
- Department of Pharmacology and Chemical Biology (C.M.-M., R.E.P., N.H.H., S.R., S.F.T., J.R.H.) and Aflac Cancer and Blood Disorders Center, Department of Pediatrics (J.O.), Emory University School of Medicine, Atlanta, Georgia
| | - Riley E Perszyk
- Department of Pharmacology and Chemical Biology (C.M.-M., R.E.P., N.H.H., S.R., S.F.T., J.R.H.) and Aflac Cancer and Blood Disorders Center, Department of Pediatrics (J.O.), Emory University School of Medicine, Atlanta, Georgia
| | - Nicholas H Harbin
- Department of Pharmacology and Chemical Biology (C.M.-M., R.E.P., N.H.H., S.R., S.F.T., J.R.H.) and Aflac Cancer and Blood Disorders Center, Department of Pediatrics (J.O.), Emory University School of Medicine, Atlanta, Georgia
| | - Jennifer Okalova
- Department of Pharmacology and Chemical Biology (C.M.-M., R.E.P., N.H.H., S.R., S.F.T., J.R.H.) and Aflac Cancer and Blood Disorders Center, Department of Pediatrics (J.O.), Emory University School of Medicine, Atlanta, Georgia
| | - Suneela Ramineni
- Department of Pharmacology and Chemical Biology (C.M.-M., R.E.P., N.H.H., S.R., S.F.T., J.R.H.) and Aflac Cancer and Blood Disorders Center, Department of Pediatrics (J.O.), Emory University School of Medicine, Atlanta, Georgia
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology (C.M.-M., R.E.P., N.H.H., S.R., S.F.T., J.R.H.) and Aflac Cancer and Blood Disorders Center, Department of Pediatrics (J.O.), Emory University School of Medicine, Atlanta, Georgia
| | - John R Hepler
- Department of Pharmacology and Chemical Biology (C.M.-M., R.E.P., N.H.H., S.R., S.F.T., J.R.H.) and Aflac Cancer and Blood Disorders Center, Department of Pediatrics (J.O.), Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
20
|
Williams AV, Peña CJ, Ramos-Maciel S, Laman-Maharg A, Ordoñez-Sanchez E, Britton M, Durbin-Johnson B, Settles M, Hao R, Yokoyama S, Xu C, Luo PX, Dwyer T, Bhela S, Black AM, Labonté B, Serafini RA, Ruiz A, Neve RL, Zachariou V, Nestler EJ, Trainor BC. Comparative Transcriptional Analyses in the Nucleus Accumbens Identifies RGS2 as a Key Mediator of Depression-Related Behavior. Biol Psychiatry 2022; 92:942-951. [PMID: 36075764 PMCID: PMC9794384 DOI: 10.1016/j.biopsych.2022.06.030] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Major depressive disorder is one of the most commonly diagnosed mental illnesses worldwide, with a higher prevalence in women than in men. Although currently available pharmacological therapeutics help many individuals, they are not effective for most. Animal models have been important for the discovery of molecular alterations in stress and depression, but difficulties in adapting animal models of depression for females has impeded progress in developing novel therapeutic treatments that may be more efficacious for women. METHODS Using the California mouse social defeat model, we took a multidisciplinary approach to identify stress-sensitive molecular targets that have translational relevance for women. We determined the impact of stress on transcriptional profiles in male and female California mouse nucleus accumbens (NAc) and compared these results with data from postmortem samples of the NAc from men and women diagnosed with major depressive disorder. RESULTS Our cross-species computational analyses identified Rgs2 (regulator of G protein signaling 2) as a transcript downregulated by social defeat stress in female California mice and in women with major depressive disorder. RGS2 plays a key role in signal regulation of neuropeptide and neurotransmitter receptors. Viral vector-mediated overexpression of Rgs2 in the NAc restored social approach and sucrose preference in stressed female California mice. CONCLUSIONS These studies show that Rgs2 acting in the NAc has functional properties that translate to changes in anxiety- and depression-related behavior. Future studies should investigate whether targeting Rgs2 represents a novel target for treatment-resistant depression in women.
Collapse
Affiliation(s)
- Alexia V Williams
- Department of Psychology, University of California, Davis, Davis, California
| | - Catherine J Peña
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Princeton Neuroscience Institute, Princeton, New Jersey
| | | | | | - Evelyn Ordoñez-Sanchez
- Department of Psychology, University of California, Davis, Davis, California; Department of Psychology, Temple University, Philadelphia, Pennsylvania
| | - Monica Britton
- Bioinformatics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, California
| | | | - Matt Settles
- Bioinformatics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, California
| | - Rebecca Hao
- Department of Psychology, University of California, Davis, Davis, California
| | - Sae Yokoyama
- Department of Psychology, University of California, Davis, Davis, California
| | - Christine Xu
- Department of Psychology, University of California, Davis, Davis, California
| | - Pei X Luo
- Department of Psychology, University of California, Davis, Davis, California
| | - Tjien Dwyer
- Department of Psychology, University of California, Davis, Davis, California
| | - Shanu Bhela
- Department of Psychology, University of California, Davis, Davis, California
| | - Alexis M Black
- Department of Psychology, University of California, Davis, Davis, California
| | - Benoit Labonté
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry and Neuroscience, Laval University, Québec, Quebec, Canada
| | - Randal Alex Serafini
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anne Ruiz
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, Boston, Massachusetts
| | - Venetia Zachariou
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brian C Trainor
- Department of Psychology, University of California, Davis, Davis, California.
| |
Collapse
|
21
|
The Potential Role of R4 Regulators of G Protein Signaling (RGS) Proteins in Type 2 Diabetes Mellitus. Cells 2022; 11:cells11233897. [PMID: 36497154 PMCID: PMC9739376 DOI: 10.3390/cells11233897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/26/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex and heterogeneous disease that primarily results from impaired insulin secretion or insulin resistance (IR). G protein-coupled receptors (GPCRs) are proposed as therapeutic targets for T2DM. GPCRs transduce signals via the Gα protein, playing an integral role in insulin secretion and IR. The regulators of G protein signaling (RGS) family proteins can bind to Gα proteins and function as GTPase-activating proteins (GAP) to accelerate GTP hydrolysis, thereby terminating Gα protein signaling. Thus, RGS proteins determine the size and duration of cellular responses to GPCR stimulation. RGSs are becoming popular targeting sites for modulating the signaling of GPCRs and related diseases. The R4 subfamily is the largest RGS family. This review will summarize the research progress on the mechanisms of R4 RGS subfamily proteins in insulin secretion and insulin resistance and analyze their potential value in the treatment of T2DM.
Collapse
|
22
|
Cabrera IE, Oza Y, Carrillo AJ, Collier LA, Wright SJ, Li L, Borkovich KA. Regulator of G Protein Signaling Proteins Control Growth, Development and Cellulase Production in Neurospora crassa. J Fungi (Basel) 2022; 8:jof8101076. [PMID: 36294641 PMCID: PMC9604755 DOI: 10.3390/jof8101076] [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: 08/30/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
Heterotrimeric (αβγ) G protein signaling pathways are critical environmental sensing systems found in eukaryotic cells. Exchange of GDP for GTP on the Gα subunit leads to its activation. In contrast, GTP hydrolysis on the Gα is accelerated by Regulator of G protein Signaling (RGS) proteins, resulting in a return to the GDP-bound, inactive state. Here, we analyzed growth, development and extracellular cellulase production in strains with knockout mutations in the seven identified RGS genes (rgs-1 to rgs-7) in the filamentous fungus, Neurospora crassa. We compared phenotypes to those of strains with either knockout mutations or expressing predicted constitutively activated, GTPase-deficient alleles for each of the three Gα subunit genes (gna-1Q204L, gna-2Q205L or gna-3Q208L). Our data revealed that six RGS mutants have taller aerial hyphae than wild type and all seven mutants exhibit reduced asexual sporulation, phenotypes shared with strains expressing the gna-1Q204L or gna-3Q208L allele. In contrast, Δrgs-1 and Δrgs-3 were the only RGS mutants with a slower growth rate phenotype, a defect in common with gna-1Q204L strains. With respect to female sexual development, Δrgs-1 possessed defects most similar to gna-3Q208L strains, while those of Δrgs-2 mutants resembled strains expressing the gna-1Q204L allele. Finally, we observed that four of the seven RGS mutants had significantly different extracellular cellulase levels relative to wild type. Of interest, the Δrgs-2 mutant had no detectable activity, similar to the gna-3Q208L strain. In contrast, the Δrgs-1 and Δrgs-4 mutants and gna-1Q204L and gna-2Q205L strains exhibited significantly higher cellulase activity than wild type. With the exception of sexual development, our results demonstrate the greatest number of genetic interactions between rgs-1 and gna-1 and rgs-2 and gna-3 in N. crassa.
Collapse
|
23
|
Dahlen SA, Bernadyn TF, Dixon AJ, Sun B, Xia J, Owens EA, Osei-Owusu P. Dual loss of regulator of G protein signaling 2 and 5 exacerbates ventricular myocyte arrhythmias and disrupts the fine-tuning of G i/o signaling. J Mol Cell Cardiol 2022; 170:34-46. [PMID: 35661621 DOI: 10.1016/j.yjmcc.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/22/2022] [Accepted: 05/14/2022] [Indexed: 10/18/2022]
Abstract
AIMS Cardiac contractility, essential to maintaining proper cardiac output and circulation, is regulated by G protein-coupled receptor (GPCR) signaling. Previously, the absence of regulator of G protein signaling (RGS) 2 and 5, separately, was shown to cause G protein dysregulation, contributing to modest blood pressure elevation and exaggerated cardiac hypertrophic response to pressure-overload. Whether RGS2 and 5 redundantly control G protein signaling to maintain cardiovascular homeostasis is unknown. Here we examined how the dual absence of RGS2 and 5 (Rgs2/5 dbKO) affects blood pressure and cardiac structure and function. METHODS AND RESULTS We found that Rgs2/5 dbKO mice showed left ventricular dilatation at baseline by echocardiography. Cardiac contractile response to dobutamine stress test was sex-dependently reduced in male Rgs2/5 dbKO relative to WT mice. When subjected to surgery-induced stress, male Rgs2/5 dbKO mice had 75% mortality within 72-96 h after surgery, accompanied by elevated baseline blood pressure and decreased cardiac contractile function. At the cellular level, cardiomyocytes (CM) from Rgs2/5 dbKO mice showed augmented Ca2+ transients and increased incidence of arrhythmia without augmented contractile response to electrical field stimulation (EFS) and activation of β-adrenergic receptors (βAR) with isoproterenol. Dual loss of Rgs2 and 5 suppressed forskolin-induced cAMP production, which was restored by Gi/o inactivation with pertussis toxin that also reduced arrhythmogenesis during EFS or βAR stimulation. Cardiomyocyte NCX and PMCA mRNA expression was unaffected in Rgs2/5 dbKO male mice. However, there was an exaggerated elevation of EFS-induced cytoplasmic Ca2+ in the presence of SERCA blockade with thapsigargin. CONCLUSIONS We conclude that RGS2 and 5 promote normal ventricular rhythm by coordinating their regulatory activity towards Gi/o signaling and facilitating cardiomyocyte calcium handling.
Collapse
Affiliation(s)
- Shelby A Dahlen
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States of America
| | - Tyler F Bernadyn
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America
| | - Alethia J Dixon
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States of America
| | - Bo Sun
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States of America
| | - Jingsheng Xia
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States of America
| | - Elizabeth A Owens
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America
| | - Patrick Osei-Owusu
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States of America; Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America.
| |
Collapse
|
24
|
RGS20 Promotes Tumor Progression through Modulating PI3K/AKT Signaling Activation in Penile Cancer. JOURNAL OF ONCOLOGY 2022; 2022:1293622. [PMID: 35498542 PMCID: PMC9042636 DOI: 10.1155/2022/1293622] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 11/20/2022]
Abstract
Regulator of G protein signaling 20 (RGS20) plays an important role in regulating neuronal G protein-coupled receptor signaling; however, its expression and oncogenic function in penile cancer (PC) remains unclear. Here, we observed high RGS20 expression in PC tissues compared to normal/adjacent penile tissues, which was closely associated with tumor stage, nodal status, and pelvic metastasis in our PC cohort. The cellular functional analysis of RGS20 revealed that manipulation of the RGS20 expression markedly affected cell viability, BrdU incorporation, soft agar clonogenesis, caspase-3 activity, and cell migration/invasion in PC cell models. Moreover, RGS20 could interact with PI3K p85α subunit and regulate PI3K/AKT signaling activation in PC cell lines. Knockdown of the PI3K p85α or p110α subunit attenuated cell viability, BrdU incorporation, soft agar clonogenesis, and cell migration/invasion in PC cell lines. In contrast, the overexpression of constitutively activated PI3K p110α mutant restored cell proliferation and cell migration/invasion caused by RGS20 depletion in PC cells. Consistent with the in vitro findings, RGS20 depletion attenuated PI3K/AKT signaling activation and suppressed tumor growth in a murine xenograft model. Importantly, the high RGS20 expression was associated with PI3K/AKT signaling activation and unfavorable progression-free/overall survival, highlighting the clinical relevance of RGS20/PI3K/AKT signaling in PC. In conclusion, the aberrant RGS20 expression may serve as a diagnostic and prognostic marker for PC. RGS20 may promote PC progression through modulating PI3K/AKT signaling activation, which may assist with the development of RGS20-targeting therapeutics in the future.
Collapse
|
25
|
Miete C, Solis GP, Koval A, Brückner M, Katanaev VL, Behrens J, Bernkopf DB. Gαi2-induced conductin/axin2 condensates inhibit Wnt/β-catenin signaling and suppress cancer growth. Nat Commun 2022; 13:674. [PMID: 35115535 PMCID: PMC8814139 DOI: 10.1038/s41467-022-28286-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 01/14/2022] [Indexed: 12/25/2022] Open
Abstract
Conductin/axin2 is a scaffold protein negatively regulating the pro-proliferative Wnt/β-catenin signaling pathway. Accumulation of scaffold proteins in condensates frequently increases their activity, but whether condensation contributes to Wnt pathway inhibition by conductin remains unclear. Here, we show that the Gαi2 subunit of trimeric G-proteins induces conductin condensation by targeting a polymerization-inhibiting aggregon in its RGS domain, thereby promoting conductin-mediated β-catenin degradation. Consistently, transient Gαi2 expression inhibited, whereas knockdown activated Wnt signaling via conductin. Colorectal cancers appear to evade Gαi2-induced Wnt pathway suppression by decreased Gαi2 expression and inactivating mutations, associated with shorter patient survival. Notably, the Gαi2-activating drug guanabenz inhibited Wnt signaling via conductin, consequently reducing colorectal cancer growth in vitro and in mouse models. In summary, we demonstrate Wnt pathway inhibition via Gαi2-triggered conductin condensation, suggesting a tumor suppressor function for Gαi2 in colorectal cancer, and pointing to the FDA-approved drug guanabenz for targeted cancer therapy.
Collapse
Affiliation(s)
- Cezanne Miete
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Gonzalo P Solis
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, 1211, Geneva 4, Geneva, Switzerland
| | - Alexey Koval
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, 1211, Geneva 4, Geneva, Switzerland
| | - Martina Brückner
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Vladimir L Katanaev
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, 1211, Geneva 4, Geneva, Switzerland
- School of Biomedicine, Far Eastern Federal University, 690922, Vladivostok, Russia
| | - Jürgen Behrens
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Dominic B Bernkopf
- Experimental Medicine II, Nikolaus-Fiebiger-Center, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany.
| |
Collapse
|
26
|
Zhang JK, Ding MJ, Liu H, Shi JH, Wang ZH, Wen PH, Zhang Y, Yan B, Guo DF, Zhang XD, Tao RL, Yan ZP, Zhang Y, Liu Z, Guo WZ, Zhang SJ. Regulator of G-protein signaling 14 protects the liver from ischemia-reperfusion injury by suppressing TGF-β-activated kinase 1 activation. Hepatology 2022; 75:338-352. [PMID: 34455616 PMCID: PMC9300117 DOI: 10.1002/hep.32133] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/25/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Hepatic ischemia-reperfusion injury (IRI) is a common complication of hepatectomy and liver transplantation. However, the mechanisms underlying hepatic IRI have not been fully elucidated. Regulator of G-protein signaling 14 (RGS14) is a multifunctional scaffolding protein that integrates the G-protein and mitogen-activated protein kinase (MAPK) signaling pathways. However, the role of RGS14 in hepatic IRI remains unclear. APPROACH AND RESULTS We found that RGS14 expression increased in mice subjected to hepatic ischemia-reperfusion (IR) surgery and during hypoxia reoxygenation in hepatocytes. We constructed global RGS14 knockout (RGS14-KO) and hepatocyte-specific RGS14 transgenic (RGS14-TG) mice to establish 70% hepatic IRI models. Histological hematoxylin and eosin staining, levels of alanine aminotransferase and aspartate aminotransferase, expression of inflammatory factors, and apoptosis were used to assess liver damage and function in these models. We found that RGS14 deficiency significantly aggravated IR-induced liver injury and activated hepatic inflammatory responses and apoptosis in vivo and in vitro. Conversely, RGS14 overexpression exerted the opposite effect of the RGS14-deficient models. Phosphorylation of TGF-β-activated kinase 1 (TAK1) and its downstream effectors c-Jun N-terminal kinase (JNK) and p38 increased in the liver tissues of RGS14-KO mice but was repressed in those of RGS14-TG mice. Furthermore, inhibition of TAK1 phosphorylation rescued the effect of RGS14 deficiency on JNK and p38 activation, thus blocking the inflammatory responses and apoptosis. CONCLUSIONS RGS14 plays a protective role in hepatic IR by inhibiting activation of the TAK1-JNK/p38 signaling pathway. This may be a potential therapeutic strategy for reducing incidences of hepatic IRI in the future.
Collapse
Affiliation(s)
- Jia-Kai Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Ming-Jie Ding
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Hui Liu
- Tongren Hospital of Wuhan University & Wuhan Third HospitalWuhanChina
| | - Ji-Hua Shi
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Zhi-Hui Wang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Pei-Hao Wen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Yi Zhang
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Bing Yan
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Dan-Feng Guo
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Xiao-Dan Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Ruo-Lin Tao
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Zhi-Ping Yan
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Yan Zhang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhen Liu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Wen-Zhi Guo
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| | - Shui-Jun Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina.,Henan Research & Development International Joint Laboratory for Organ Transplantation ImmunomodulationZhengzhouChina
| |
Collapse
|
27
|
Fina ME, Wang J, Vedula P, Tang HY, Kashina A, Dong DW. Arginylation Regulates G-protein Signaling in the Retina. Front Cell Dev Biol 2022; 9:807345. [PMID: 35127722 PMCID: PMC8815403 DOI: 10.3389/fcell.2021.807345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/17/2021] [Indexed: 12/03/2022] Open
Abstract
Arginylation is a post-translational modification mediated by the arginyltransferase (Ate1). We recently showed that conditional deletion of Ate1 in the nervous system leads to increased light-evoked response sensitivities of ON-bipolar cells in the retina, indicating that arginylation regulates the G-protein signaling complexes of those neurons and/or photoreceptors. However, none of the key players in the signaling pathway were previously shown to be arginylated. Here we show that Gαt1, Gβ1, RGS6, and RGS7 are arginylated in the retina and RGS6 and RGS7 protein levels are elevated in Ate1 knockout, suggesting that arginylation plays a direct role in regulating their protein level and the G-protein-mediated responses in the retina.
Collapse
Affiliation(s)
- Marie E. Fina
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, United States
| | - Junling Wang
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, United States
| | - Pavan Vedula
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, United States
| | - Hsin-Yao Tang
- Proteomics and Metabolomics Facility, The Wistar Institute, Philadelphia, PA, United States
| | - Anna Kashina
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Anna Kashina, ; Dawei W. Dong,
| | - Dawei W. Dong
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Anna Kashina, ; Dawei W. Dong,
| |
Collapse
|
28
|
Bai Y, Hu M, Chen Z, Wei J, Du H. Single-Cell Transcriptome Analysis Reveals RGS1 as a New Marker and Promoting Factor for T-Cell Exhaustion in Multiple Cancers. Front Immunol 2021; 12:767070. [PMID: 34956194 PMCID: PMC8692249 DOI: 10.3389/fimmu.2021.767070] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/15/2021] [Indexed: 01/04/2023] Open
Abstract
T-cell exhaustion is one of the main reasons of tumor immune escape. Using single-cell transcriptome data of CD8+ T cells in multiple cancers, we identified different cell types, in which Pre_exhaust and exhausted T cells participated in negative regulation of immune system process. By analyzing the coexpression network patterns and differentially expressed genes of Pre_exhaust, exhausted, and effector T cells, we identified 35 genes related to T-cell exhaustion, whose high GSVA scores were associated with significantly poor prognosis in various cancers. In the differentially expressed genes, RGS1 showed the greatest fold change in Pre_exhaust and exhausted cells of three cancers compared with effector T cells, and high expression of RGS1 was also associated with poor prognosis in various cancers. Additionally, RGS1 protein was upregulated significantly in tumor tissues in the immunohistochemistry verification. Furthermore, RGS1 displayed positive correlation with the 35 genes, especially highly correlated with PDCD1, CTLA4, HAVCR2, and TNFRSF9 in CD8+ T cells and cancer tissues, indicating the important roles of RGS1 in CD8+ T-cell exhaustion. Considering the GTP-hydrolysis activity of RGS1 and significantly high mRNA and protein expression in cancer tissues, we speculated that RGS1 potentially mediate the T-cell retention to lead to the persistent antigen stimulation, resulting in T-cell exhaustion. In conclusion, our findings suggest that RGS1 is a new marker and promoting factor for CD8+ T-cell exhaustion and provide theoretical basis for research and immunotherapy of exhausted cells.
Collapse
Affiliation(s)
- Yunmeng Bai
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital, Shenzhen, China
| | - Meiling Hu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Zixi Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Jinfen Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
29
|
Almutairi F, Sarr D, Tucker SL, Fantone K, Lee JK, Rada B. RGS10 Reduces Lethal Influenza Infection and Associated Lung Inflammation in Mice. Front Immunol 2021; 12:772288. [PMID: 34912341 PMCID: PMC8667315 DOI: 10.3389/fimmu.2021.772288] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/10/2021] [Indexed: 01/05/2023] Open
Abstract
Seasonal influenza epidemics represent a significant global health threat. The exacerbated immune response triggered by respiratory influenza virus infection causes severe pulmonary damage and contributes to substantial morbidity and mortality. Regulator of G-protein signaling 10 (RGS10) belongs to the RGS protein family that act as GTPase activating proteins for heterotrimeric G proteins to terminate signaling pathways downstream of G protein-coupled receptors. While RGS10 is highly expressed in immune cells, in particular monocytes and macrophages, where it has strong anti-inflammatory effects, its physiological role in the respiratory immune system has not been explored yet. Here, we show that Rgs10 negatively modulates lung immune and inflammatory responses associated with severe influenza H1N1 virus respiratory infection in a mouse model. In response to influenza A virus challenge, mice lacking RGS10 experience enhanced weight loss and lung viral titers, higher mortality and significantly faster disease onset. Deficiency of Rgs10 upregulates the levels of several proinflammatory cytokines and chemokines and increases myeloid leukocyte accumulation in the infected lung, markedly neutrophils, monocytes, and inflammatory monocytes, which is associated with more pronounced lung damage. Consistent with this, influenza-infected Rgs10-deficent lungs contain more neutrophil extracellular traps and exhibit higher neutrophil elastase activities than wild-type lungs. Overall, these findings propose a novel, in vivo role for RGS10 in the respiratory immune system controlling myeloid leukocyte infiltration, viral clearance and associated clinical symptoms following lethal influenza challenge. RGS10 also holds promise as a new, potential therapeutic target for respiratory infections.
Collapse
Affiliation(s)
- Faris Almutairi
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, United States
| | - Demba Sarr
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Samantha L. Tucker
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Kayla Fantone
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Jae-Kyung Lee
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
30
|
Sahly AN, Shevell M, Sadleir LG, Myers KA. SUDEP risk and autonomic dysfunction in genetic epilepsies. Auton Neurosci 2021; 237:102907. [PMID: 34773737 DOI: 10.1016/j.autneu.2021.102907] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 10/11/2021] [Accepted: 11/06/2021] [Indexed: 01/02/2023]
Abstract
The underlying pathophysiology of sudden unexpected death in epilepsy (SUDEP) remains unclear. This phenomenon is likely multifactorial, and there is considerable evidence that genetic factors play a role. There are certain genetic causes of epilepsy in which the risk of SUDEP appears to be increased relative to epilepsy overall. For individuals with pathogenic variants in genes including SCN1A, SCN1B, SCN8A, SCN2A, GNB5, KCNA1 and DEPDC5, there are varying degrees of evidence to suggest an increased risk for sudden death. Why the risk for sudden death is higher is not completely clear; however, in many cases pathogenic variants in these genes are also associated with autonomic dysfunction, which is hypothesized as a contributing factor to SUDEP. We review the evidence for increased SUDEP risk for patients with epilepsy due to pathogenic variants in these genes, and also discuss what is known about autonomic dysfunction in these contexts.
Collapse
Affiliation(s)
- Ahmed N Sahly
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Department of Neurosciences, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | - Michael Shevell
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Research Institute of the McGill University Medical Centre, Montreal, Quebec, Canada
| | - Lynette G Sadleir
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Kenneth A Myers
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Research Institute of the McGill University Medical Centre, Montreal, Quebec, Canada.
| |
Collapse
|
31
|
Li C, Vides A, Kim D, Xue J, Zhao Y, Lito P. The G protein signaling regulator RGS3 enhances the GTPase activity of KRAS. Science 2021; 374:197-201. [PMID: 34618566 PMCID: PMC9295010 DOI: 10.1126/science.abf1730] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recently reported to be effective in patients with lung cancer, KRASG12C inhibitors bind to the inactive, or guanosine diphosphate (GDP)–bound, state of the oncoprotein and require guanosine triphosphate (GTP) hydrolysis for inhibition. However, KRAS mutations prevent the catalytic arginine of GTPase-activating proteins (GAPs) from enhancing an otherwise slow hydrolysis rate. If KRAS mutants are indeed insensitive to GAPs, it is unclear how KRASG12C hydrolyzes sufficient GTP to allow inactive state–selective inhibition. Here, we show that RGS3, a GAP previously known for regulating G protein–coupled receptors, can also enhance the GTPase activity of mutant and wild-type KRAS proteins. Our study reveals an unexpected mechanism that inactivates KRAS and explains the vulnerability to emerging clinically effective therapeutics.
Collapse
Affiliation(s)
- Chuanchuan Li
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY 10065
| | - Alberto Vides
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY 10065
| | - Dongsung Kim
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY 10065
| | - Jenny Xue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY 10065
- Tri-Institutional MD-PhD Program, Weill Cornell Medical College and Rockefeller University and Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Yulei Zhao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY 10065
| | - Piro Lito
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY 10065
- Tri-Institutional MD-PhD Program, Weill Cornell Medical College and Rockefeller University and Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
32
|
Degrandmaison J, Grisé O, Parent JL, Gendron L. Differential barcoding of opioid receptors trafficking. J Neurosci Res 2021; 100:99-128. [PMID: 34559903 DOI: 10.1002/jnr.24949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 07/25/2021] [Accepted: 08/05/2021] [Indexed: 11/09/2022]
Abstract
Over the past several years, studies have highlighted the δ-opioid receptor (DOPr) as a promising therapeutic target for chronic pain management. While exhibiting milder undesired effects than most currently prescribed opioids, its specific agonists elicit effective analgesic responses in numerous animal models of chronic pain, including inflammatory, neuropathic, diabetic, and cancer-related pain. However, as compared with the extensively studied μ-opioid receptor, the molecular mechanisms governing its trafficking remain elusive. Recent advances have denoted several significant particularities in the regulation of DOPr intracellular routing, setting it apart from the other members of the opioid receptor family. Although they share high homology, each opioid receptor subtype displays specific amino acid patterns potentially involved in the regulation of its trafficking. These precise motifs or "barcodes" are selectively recognized by regulatory proteins and therefore dictate several aspects of the itinerary of a receptor, including its anterograde transport, internalization, recycling, and degradation. With a specific focus on the regulation of DOPr trafficking, this review will discuss previously reported, as well as potential novel trafficking barcodes within the opioid and nociceptin/orphanin FQ opioid peptide receptors, and their impact in determining distinct interactomes and physiological responses.
Collapse
Affiliation(s)
- Jade Degrandmaison
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Quebec Network of Junior Pain Investigators, QC, Canada
| | - Olivier Grisé
- Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Luc Parent
- Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Louis Gendron
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Quebec Pain Research Network, QC, Canada
| |
Collapse
|
33
|
Asli A, Higazy-Mreih S, Avital-Shacham M, Kosloff M. Residue-level determinants of RGS R4 subfamily GAP activity and specificity towards the G i subfamily. Cell Mol Life Sci 2021; 78:6305-6318. [PMID: 34292354 PMCID: PMC11072900 DOI: 10.1007/s00018-021-03898-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/18/2021] [Accepted: 07/09/2021] [Indexed: 01/01/2023]
Abstract
The structural basis for the GTPase-accelerating activity of regulators of G protein signaling (RGS) proteins, as well as the mechanistic basis for their specificity in interacting with the heterotrimeric (αβγ) G proteins they inactivate, is not sufficiently understood at the family level. Here, we used biochemical assays to compare RGS domains across the RGS family and map those individual residues that favorably contribute to GTPase-accelerating activity, and those residues responsible for attenuating RGS domain interactions with Gα subunits. We show that conserved interactions of RGS residues with both the Gα switch I and II regions are crucial for RGS activity, while the reciprocal effects of "modulatory" and "disruptor" residues selectively modulate RGS activity. Our results quantify how specific interactions between RGS domains and Gα subunits are set by a balance between favorable RGS residue interactions with particular Gα switch regions, and unfavorable interactions with the Gα helical domain.
Collapse
Affiliation(s)
- Ali Asli
- The Department of Human Biology, Faculty of Natural Science, University of Haifa, 199 Aba Khoushy Ave., Mt. Carmel, 3498838, Haifa, Israel
| | - Sabreen Higazy-Mreih
- The Department of Human Biology, Faculty of Natural Science, University of Haifa, 199 Aba Khoushy Ave., Mt. Carmel, 3498838, Haifa, Israel
| | - Meirav Avital-Shacham
- The Department of Human Biology, Faculty of Natural Science, University of Haifa, 199 Aba Khoushy Ave., Mt. Carmel, 3498838, Haifa, Israel
| | - Mickey Kosloff
- The Department of Human Biology, Faculty of Natural Science, University of Haifa, 199 Aba Khoushy Ave., Mt. Carmel, 3498838, Haifa, Israel.
| |
Collapse
|
34
|
Almutairi F, Tucker SL, Sarr D, Rada B. PI3K/ NF-κB-dependent TNF-α and HDAC activities facilitate LPS-induced RGS10 suppression in pulmonary macrophages. Cell Signal 2021; 86:110099. [PMID: 34339853 PMCID: PMC8406451 DOI: 10.1016/j.cellsig.2021.110099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022]
Abstract
Regulator of G-protein signaling 10 (RGS10) is a member of the superfamily of RGS proteins that canonically act as GTPase activating proteins (GAPs). RGS proteins accelerate GTP hydrolysis on the G-protein α subunits and result in termination of signaling pathways downstream of G protein-coupled receptors. Beyond its GAP function, RGS10 has emerged as an anti-inflammatory protein by inhibiting LPS-mediated NF-κB activation and expression of inflammatory cytokines, in particular TNF-α. Although RGS10 is abundantly expressed in resting macrophages, previous studies have shown that RGS10 expression is suppressed in macrophages following Toll-like receptor 4 (TLR4) activation by LPS. However, the molecular mechanism by which LPS induces Rgs10 silencing has not been clearly defined. The goal of the current study was to determine whether LPS silences Rgs10 expression through an NF-κB-mediated proinflammatory mechanism in pulmonary macrophages, a unique type of innate immune cells. We demonstrate that Rgs10 transcript and RGS10 protein levels are suppressed upon LPS treatment in the murine MH-S alveolar macrophage cell line. We show that pharmacological inhibition of PI3K/ NF-κB/p300 (NF-κB co-activator)/TNF-α signaling cascade and the activities of HDAC (1-3) enzymes block LPS-induced silencing of Rgs10 in MH-S cells as well as microglial BV2 cells and BMDMs. Further, loss of RGS10 generated by using CRISPR/Cas9 amplifies NF-κB phosphorylation and inflammatory gene expression following LPS treatment in MH-S cells. Together, our findings strongly provide critical insight into the molecular mechanism underlying RGS10 suppression by LPS in pulmonary macrophages.
Collapse
Affiliation(s)
- Faris Almutairi
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Samantha L Tucker
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Demba Sarr
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| |
Collapse
|
35
|
Stoveken HM, Fernandez-Vega V, Muntean BS, Patil DN, Shumate J, Bannister TD, Scampavia L, Spicer TP, Martemyanov KA. Identification of Potential Modulators of the RGS7/Gβ5/R7BP Complex. SLAS DISCOVERY 2021; 26:1177-1188. [PMID: 34112017 DOI: 10.1177/24725552211020679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Regulators of G protein signaling (RGS) proteins serve as critical regulatory nodes to limit the lifetime and extent of signaling via G protein-coupled receptors (GPCRs). Previously, approaches to pharmacologically inhibit RGS activity have mostly focused on the inhibition of GTPase activity by interrupting the interaction of RGS proteins with the G proteins they regulate. However, several RGS proteins are also regulated by association with binding partners. A notable example is the mammalian RGS7 protein, which has prominent roles in metabolic control, vision, reward, and actions of opioid analgesics. In vivo, RGS7 exists in complex with the binding partners type 5 G protein β subunit (Gβ5) and R7 binding protein (R7BP), which control its stability and activity, respectively. Targeting the whole RGS7/Gβ5/R7BP protein complex affords the opportunity to allosterically tune opioid receptor signaling following opioid engagement while potentially bypassing undesirable side effects. Hence, we implemented a novel strategy to pharmacologically target the interaction between RGS7/Gβ5 and R7BP. To do so, we searched for protein complex inhibitors using a time-resolved fluorescence resonance energy transfer (FRET)-based high-throughput screening (HTS) assay that measures compound-mediated alterations in the FRET signal between RGS7/Gβ5 and R7BP. We performed two HTS campaigns, each screening ~100,000 compounds from the Scripps Drug Discovery Library (SDDL). Each screen yielded more than 100 inhibitors, which will be described herein.
Collapse
Affiliation(s)
- Hannah M Stoveken
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | | | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Dipak N Patil
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Justin Shumate
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Thomas D Bannister
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Louis Scampavia
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Timothy P Spicer
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | | |
Collapse
|
36
|
Fina ME, Wang J, Nikonov SS, Sterling S, Vardi N, Kashina A, Dong DW. Arginyltransferase (Ate1) regulates the RGS7 protein level and the sensitivity of light-evoked ON-bipolar responses. Sci Rep 2021; 11:9376. [PMID: 33931669 PMCID: PMC8087773 DOI: 10.1038/s41598-021-88628-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Regulator of G-protein signaling 7 (RGS7) is predominately present in the nervous system and is essential for neuronal signaling involving G-proteins. Prior studies in cultured cells showed that RGS7 is regulated via proteasomal degradation, however no protein is known to facilitate proteasomal degradation of RGS7 and it has not been shown whether this regulation affects G-protein signaling in neurons. Here we used a knockout mouse model with conditional deletion of arginyltransferase (Ate1) in the nervous system and found that in retinal ON bipolar cells, where RGS7 modulates a G-protein to signal light increments, deletion of Ate1 raised the level of RGS7. Electroretinographs revealed that lack of Ate1 leads to increased light-evoked response sensitivities of ON-bipolar cells, as well as their downstream neurons. In cultured mouse embryonic fibroblasts (MEF), RGS7 was rapidly degraded via proteasome pathway and this degradation was abolished in Ate1 knockout MEF. Our results indicate that Ate1 regulates RGS7 protein level by facilitating proteasomal degradation of RGS7 and thus affects G-protein signaling in neurons.
Collapse
Affiliation(s)
- Marie E Fina
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Junling Wang
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sergei S Nikonov
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Stephanie Sterling
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Noga Vardi
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anna Kashina
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Dawei W Dong
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
37
|
Ren J, Wei W, Tan L, Yang Q, Lu Q, Ding H, Yue Y, Tian Y, Hao L, Wang M, Li J. Inhibition of regulator of G protein signaling 10, aggravates rheumatoid arthritis progression by promoting NF-κB signaling pathway. Mol Immunol 2021; 134:236-246. [PMID: 33836352 DOI: 10.1016/j.molimm.2021.03.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/10/2021] [Accepted: 03/18/2021] [Indexed: 02/05/2023]
Abstract
Rheumatoid arthritis (RA) is the most common inflammatory arthropathy, with evidence pointing to an immune-mediated etiology that propagates chronic inflammation. Although targeted immune therapeutics and aggressive treatment strategies have substantially improved, a complete understanding of the associated pathological mechanisms of the disease remains elusive. This study aimed at investigating whether regulator of G protein signaling 10 (RGS10) could affect rheumatoid arthritis (RA) pathology by regulating the immune response. A DBA/J1 mouse model of RA was established and evaluated for disease severity. RGS10 expression was inhibited by adeno-associated virus in vivo. Moreover, small interfering RNA was used to downregulate RGS10 expression in raw 264.7 cells in vitro. Results showed that RGS10 inhibition augmented RA severity, and attenuated the increase in expression of inflammatory factors. Furthermore, activated NF-κB signaling pathways were detected following RGS10 inhibition. These results revealed that RGS10 inhibition directly aggravated the RA pathological process by activating the NF-κB signaling pathway. Therefore, RGS10 is a promising novel therapeutic target for RA treatment with a potential clinical impact.
Collapse
Affiliation(s)
- Jie Ren
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Wei Wei
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Liangyu Tan
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Qin Yang
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Qiuyu Lu
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Handong Ding
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Yuan Yue
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Ye Tian
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Liang Hao
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Min Wang
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China.
| | - Jinle Li
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China.
| |
Collapse
|
38
|
Wendimu MY, Alqinyah M, Vella S, Dean P, Almutairi F, Davila-Rivera R, Rayatpisheh S, Wohlschlegel J, Moreno S, Hooks SB. RGS10 physically and functionally interacts with STIM2 and requires store-operated calcium entry to regulate pro-inflammatory gene expression in microglia. Cell Signal 2021; 83:109974. [PMID: 33705894 DOI: 10.1016/j.cellsig.2021.109974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/19/2021] [Accepted: 03/04/2021] [Indexed: 01/14/2023]
Abstract
Chronic activation of microglia is a driving factor in the progression of neuroinflammatory diseases, and mechanisms that regulate microglial inflammatory signaling are potential targets for novel therapeutics. Regulator of G protein Signaling 10 is the most abundant RGS protein in microglia, where it suppresses inflammatory gene expression and reduces microglia-mediated neurotoxicity. In particular, microglial RGS10 downregulates the expression of pro-inflammatory mediators including cyclooxygenase 2 (COX-2) following stimulation with lipopolysaccharide (LPS). However, the mechanism by which RGS10 affects inflammatory signaling is unknown and is independent of its canonical G protein targeted mechanism. Here, we sought to identify non-canonical RGS10 interacting partners that mediate its anti-inflammatory mechanism. Through RGS10 co-immunoprecipitation coupled with mass spectrometry, we identified STIM2, an endoplasmic reticulum (ER) localized calcium sensor and a component of the store-operated calcium entry (SOCE) machinery, as a novel RGS10 interacting protein in microglia. Direct immunoprecipitation experiments confirmed RGS10-STIM2 interaction in multiple microglia and macrophage cell lines, as well as in primary cells, with no interaction observed with the homologue STIM1. We further determined that STIM2, Orai channels, and the calcium-dependent phosphatase calcineurin are essential for LPS-induced COX-2 production in microglia, and this pathway is required for the inhibitory effect of RGS10 on COX-2. Additionally, our data demonstrated that RGS10 suppresses SOCE triggered by ER calcium depletion and that ER calcium depletion, which induces SOCE, amplifies pro-inflammatory genes. In addition to COX-2, we also show that RGS10 suppresses the expression of pro-inflammatory cytokines in microglia in response to thrombin and LPS stimulation, and all of these effects require SOCE. Collectively, the physical and functional links between RGS10 and STIM2 suggest a complex regulatory network connecting RGS10, SOCE, and pro-inflammatory gene expression in microglia, with broad implications in the pathogenesis and treatment of chronic neuroinflammation.
Collapse
Affiliation(s)
- Menbere Y Wendimu
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, United States of America
| | - Mohammed Alqinyah
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, United States of America
| | - Stephen Vella
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, United States of America
| | - Phillip Dean
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, United States of America
| | - Faris Almutairi
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, United States of America
| | - Roseanne Davila-Rivera
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, United States of America
| | - Shima Rayatpisheh
- Department of Biological Chemistry, University of California, Los Angeles 90095, United States of America
| | - James Wohlschlegel
- Department of Biological Chemistry, University of California, Los Angeles 90095, United States of America
| | - Silvia Moreno
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, United States of America
| | - Shelley B Hooks
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, United States of America.
| |
Collapse
|
39
|
Fuentes N, McCullough M, Panettieri RA, Druey KM. RGS proteins, GRKs, and beta-arrestins modulate G protein-mediated signaling pathways in asthma. Pharmacol Ther 2021; 223:107818. [PMID: 33600853 DOI: 10.1016/j.pharmthera.2021.107818] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 12/17/2022]
Abstract
Asthma is a highly prevalent disorder characterized by chronic lung inflammation and reversible airways obstruction. Pathophysiological features of asthma include episodic and reversible airway narrowing due to increased bronchial smooth muscle shortening in response to external and host-derived mediators, excessive mucus secretion into the airway lumen, and airway remodeling. The aberrant airway smooth muscle (ASM) phenotype observed in asthma manifests as increased sensitivity to contractile mediators (EC50) and an increase in the magnitude of contraction (Emax); collectively these attributes have been termed "airways hyper-responsiveness" (AHR). This defining feature of asthma can be promoted by environmental factors including airborne allergens, viruses, and air pollution and other irritants. AHR reduces airway caliber and obstructs airflow, evoking clinical symptoms such as cough, wheezing and shortness of breath. G-protein-coupled receptors (GPCRs) have a central function in asthma through their impact on ASM and airway inflammation. Many but not all treatments for asthma target GPCRs mediating ASM contraction or relaxation. Here we discuss the roles of specific GPCRs, G proteins, and their associated signaling pathways, in asthma, with an emphasis on endogenous mechanisms of GPCR regulation of ASM tone and lung inflammation including regulators of G-protein signaling (RGS) proteins, G-protein coupled receptor kinases (GRKs), and β-arrestin.
Collapse
Affiliation(s)
- Nathalie Fuentes
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States of America
| | - Morgan McCullough
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States of America
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers University School of Medicine, New Brunswick, NJ, United States of America
| | - Kirk M Druey
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, NIAID/NIH, Bethesda, MD, United States of America.
| |
Collapse
|
40
|
Hariharan A, Weir N, Robertson C, He L, Betsholtz C, Longden TA. The Ion Channel and GPCR Toolkit of Brain Capillary Pericytes. Front Cell Neurosci 2020; 14:601324. [PMID: 33390906 PMCID: PMC7775489 DOI: 10.3389/fncel.2020.601324] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Brain pericytes reside on the abluminal surface of capillaries, and their processes cover ~90% of the length of the capillary bed. These cells were first described almost 150 years ago (Eberth, 1871; Rouget, 1873) and have been the subject of intense experimental scrutiny in recent years, but their physiological roles remain uncertain and little is known of the complement of signaling elements that they employ to carry out their functions. In this review, we synthesize functional data with single-cell RNAseq screens to explore the ion channel and G protein-coupled receptor (GPCR) toolkit of mesh and thin-strand pericytes of the brain, with the aim of providing a framework for deeper explorations of the molecular mechanisms that govern pericyte physiology. We argue that their complement of channels and receptors ideally positions capillary pericytes to play a central role in adapting blood flow to meet the challenge of satisfying neuronal energy requirements from deep within the capillary bed, by enabling dynamic regulation of their membrane potential to influence the electrical output of the cell. In particular, we outline how genetic and functional evidence suggest an important role for Gs-coupled GPCRs and ATP-sensitive potassium (KATP) channels in this context. We put forth a predictive model for long-range hyperpolarizing electrical signaling from pericytes to upstream arterioles, and detail the TRP and Ca2+ channels and Gq, Gi/o, and G12/13 signaling processes that counterbalance this. We underscore critical questions that need to be addressed to further advance our understanding of the signaling topology of capillary pericytes, and how this contributes to their physiological roles and their dysfunction in disease.
Collapse
Affiliation(s)
- Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Colin Robertson
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Liqun He
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Christer Betsholtz
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Medicine Huddinge (MedH), Karolinska Institutet & Integrated Cardio Metabolic Centre, Huddinge, Sweden
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
41
|
Wang HH, de Bari O, Arnatt CK, Liu M, Portincasa P, Wang DQH. Activation of Estrogen Receptor G Protein-Coupled Receptor 30 Enhances Cholesterol Cholelithogenesis in Female Mice. Hepatology 2020; 72:2077-2089. [PMID: 32112420 PMCID: PMC8157628 DOI: 10.1002/hep.31212] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/21/2020] [Accepted: 02/13/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND AIMS Estrogen is an important risk factor for cholesterol gallstone disease because women are twice as likely as men to form gallstones. The classical estrogen receptor α (ERα), but not ERβ, in the liver plays a critical role in the formation of estrogen-induced gallstones in female mice. The molecular mechanisms underlying the lithogenic effect of estrogen on gallstone formation have become more complicated with the identification of G protein-coupled receptor 30 (GPR30), an estrogen receptor. APPROACH AND RESULTS We investigated the biliary and gallstone phenotypes in ovariectomized female GPR30-/- , ERα-/- , and wild-type mice injected intramuscularly with the potent GPR30-selective agonist G-1 at 0 or 1 μg/day and fed a lithogenic diet for 8 weeks. The activation of GPR30 by G-1 enhanced cholelithogenesis by suppressing expression of cholesterol 7α-hydroxylase, the rate-limiting enzyme for the classical pathway of bile salt synthesis. These metabolic abnormalities led to an increase in biliary cholesterol concentrations in company with hepatic hyposecretion of biliary bile salts, thereby inducing cholesterol-supersaturated gallbladder bile and accelerating cholesterol crystallization. G-1 also impairs gallbladder emptying, leading to sluggish gallbladder motility and promoting the development of biliary sludge in the early stage of gallstone formation. The prevalence rates of gallstones were 80% in wild-type and ERα-/- mice treated with G-1 compared to 10% in wild-type mice receiving no G-1. However, no gallstones were formed in GPR30-/- mice treated with G-1. CONCLUSIONS GPR30 produces additional lithogenic actions, working independently of ERα, to increase susceptible to gallstone formation in female mice; both GPR30 and ERα are potential therapeutic targets for cholesterol gallstone disease, particularly in women and patients exposed to high levels of estrogen.
Collapse
Affiliation(s)
- Helen H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY;,Department of Internal Medicine, Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, MO
| | - Ornella de Bari
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, MO
| | | | - Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, Clinica Medica “A. Murri”, University of Bari Medical School, Bari, Italy
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY;,Department of Internal Medicine, Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, MO
| |
Collapse
|
42
|
DeHelian D, Gupta S, Wu J, Thorsheim C, Estevez B, Cooper M, Litts K, Lee-Sundlov MM, Hoffmeister KM, Poncz M, Ma P, Brass LF. RGS10 and RGS18 differentially limit platelet activation, promote platelet production, and prolong platelet survival. Blood 2020; 136:1773-1782. [PMID: 32542378 PMCID: PMC7544544 DOI: 10.1182/blood.2019003251] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 05/22/2020] [Indexed: 12/23/2022] Open
Abstract
G protein-coupled receptors are critical mediators of platelet activation whose signaling can be modulated by members of the regulator of G protein signaling (RGS) family. The 2 most abundant RGS proteins in human and mouse platelets are RGS10 and RGS18. While each has been studied individually, critical questions remain about the overall impact of this mode of regulation in platelets. Here, we report that mice missing both proteins show reduced platelet survival and a 40% decrease in platelet count that can be partially reversed with aspirin and a P2Y12 antagonist. Their platelets have increased basal (TREM)-like transcript-1 expression, a leftward shift in the dose/response for a thrombin receptor-activating peptide, an increased maximum response to adenosine 5'-diphosphate and TxA2, and a greatly exaggerated response to penetrating injuries in vivo. Neither of the individual knockouts displays this constellation of findings. RGS10-/- platelets have an enhanced response to agonists in vitro, but platelet count and survival are normal. RGS18-/- mice have a 15% reduction in platelet count that is not affected by antiplatelet agents, nearly normal responses to platelet agonists, and normal platelet survival. Megakaryocyte number and ploidy are normal in all 3 mouse lines, but platelet recovery from severe acute thrombocytopenia is slower in RGS18-/- and RGS10-/-18-/- mice. Collectively, these results show that RGS10 and RGS18 have complementary roles in platelets. Removing both at the same time discloses the extent to which this regulatory mechanism normally controls platelet reactivity in vivo, modulates the hemostatic response to injury, promotes platelet production, and prolongs platelet survival.
Collapse
Affiliation(s)
- Daniel DeHelian
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Shuchi Gupta
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Jie Wu
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Chelsea Thorsheim
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Brian Estevez
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia PA
| | - Matthew Cooper
- Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Kelly Litts
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Melissa M Lee-Sundlov
- Department of Biochemistry and
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Milwaukee, WI; and
| | - Karin M Hoffmeister
- Department of Biochemistry and
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Versiti Blood Research Institute, Milwaukee, WI; and
| | - Mortimer Poncz
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia PA
| | - Peisong Ma
- Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Lawrence F Brass
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
43
|
Masuho I, Balaji S, Muntean BS, Skamangas NK, Chavali S, Tesmer JJG, Babu MM, Martemyanov KA. A Global Map of G Protein Signaling Regulation by RGS Proteins. Cell 2020; 183:503-521.e19. [PMID: 33007266 PMCID: PMC7572916 DOI: 10.1016/j.cell.2020.08.052] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 07/03/2020] [Accepted: 08/29/2020] [Indexed: 12/24/2022]
Abstract
The control over the extent and timing of G protein signaling is provided by the regulator of G protein signaling (RGS) proteins that deactivate G protein α subunits (Gα). Mammalian genomes encode 20 canonical RGS and 16 Gα genes with key roles in physiology and disease. To understand the principles governing the selectivity of Gα regulation by RGS, we examine the catalytic activity of all canonical human RGS proteins and their selectivity for a complete set of Gα substrates using real-time kinetic measurements in living cells. The data reveal rules governing RGS-Gα recognition, the structural basis of its selectivity, and provide principles for engineering RGS proteins with defined selectivity. The study also explores the evolution of RGS-Gα selectivity through ancestral reconstruction and demonstrates how naturally occurring non-synonymous variants in RGS alter signaling. These results provide a blueprint for decoding signaling selectivity and advance our understanding of molecular recognition principles. Systematic analysis reveals G protein selectivity of all canonical RGS proteins RGS proteins rely on selectivity bar codes for selective G protein recognition Transplantation of bar codes across RGS proteins switches their G protein preferences Natural variants, mutations, and evolution shape RGS selectivity
Collapse
Affiliation(s)
- Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Santhanam Balaji
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Departments of Structural Biology and Center for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Nickolas K Skamangas
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Sreenivas Chavali
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Karakambadi Road, Tirupati 517 507, India
| | - John J G Tesmer
- Departments of Biological Sciences and Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907-2054, USA
| | - M Madan Babu
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Departments of Structural Biology and Center for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA.
| |
Collapse
|
44
|
Chen R, Ferris MJ, Wang S. Dopamine D2 autoreceptor interactome: Targeting the receptor complex as a strategy for treatment of substance use disorder. Pharmacol Ther 2020; 213:107583. [PMID: 32473160 PMCID: PMC7434700 DOI: 10.1016/j.pharmthera.2020.107583] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Dopamine D2 autoreceptors (D2ARs), located in somatodendritic and axon terminal compartments of dopamine (DA) neurons, function to provide a negative feedback regulatory control on DA neuron firing, DA synthesis, reuptake and release. Dysregulation of D2AR-mediated DA signaling is implicated in vulnerability to substance use disorder (SUD). Due to the extreme low abundance of D2ARs compared to postsynaptic D2 receptors (D2PRs) and the lack of experimental tools to differentiate the signaling of D2ARs from D2PRs, the regulation of D2ARs by drugs of abuse is poorly understood. The recent availability of conditional D2AR knockout mice and newly developed virus-mediated gene delivery approaches have provided means to specifically study the function of D2ARs at the molecular, cellular and behavioral levels. There is a growing revelation of novel mechanisms and new proteins that mediate D2AR activity, suggesting that D2ARs act cooperatively with an array of membrane and intracellular proteins to tightly control DA transmission. This review highlights D2AR-interacting partners including transporters, G-protein-coupled receptors, ion channels, intracellular signaling modulators, and protein kinases. The complexity of the D2AR interaction network illustrates the functional divergence of D2ARs. Pharmacological targeting of multiple D2AR-interacting partners may be more effective to restore disrupted DA homeostasis by drugs of abuse.
Collapse
Affiliation(s)
- Rong Chen
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America.
| | - Mark J Ferris
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America
| | - Shiyu Wang
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America
| |
Collapse
|
45
|
Yuan G, Yang S, Liu M, Yang S. RGS12 is required for the maintenance of mitochondrial function during skeletal development. Cell Discov 2020; 6:59. [PMID: 32922858 PMCID: PMC7459111 DOI: 10.1038/s41421-020-00190-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial morphology and function are crucial for tissue homeostasis, such as for skeletal development, but the cellular and molecular mechanisms remain unclear. Here, we provide evidence that regulator of G-protein signaling 12 (RGS12) is present in the mitochondria of primary chondrocytes and cartilage tissues. Deletion of RGS12 in type II collagen-positive cells led to a significant decrease in mitochondrial number, membrane potential, and oxidative phosphorylation function. Mechanistically, RGS12 promoted the function of ATP5A as an enhancer of tyrosine phosphorylation. Mice with RGS12 deficiency in the chondrocyte lineage showed serious body retardation, decreased bone mass, and chondrocyte apoptosis due to the defective activity of ATP synthase. To our knowledge, this is the first report that RGS12 is required for maintaining the function of mitochondria, which may allow it to orchestrate responses to cellular homeostasis.
Collapse
Affiliation(s)
- Gongsheng Yuan
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA USA
| | - Shuting Yang
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA USA
| | - Min Liu
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA USA
| | - Shuying Yang
- Department of Basic and Translational Sciences, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA USA.,The Penn Center for Musculoskeletal Disorders, University of Pennsylvania, School of Medicine, Philadelphia, PA USA.,Center for Innovation & Precision Dentistry, University of Pennsylvania, School of Dental Medicine, School of Engineering and Applied Sciences, Philadelphia, PA USA
| |
Collapse
|
46
|
Almutairi F, Lee JK, Rada B. Regulator of G protein signaling 10: Structure, expression and functions in cellular physiology and diseases. Cell Signal 2020; 75:109765. [PMID: 32882407 PMCID: PMC7579743 DOI: 10.1016/j.cellsig.2020.109765] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 01/22/2023]
Abstract
Regulator of G protein signaling 10 (RGS10) belongs to the superfamily of RGS proteins, defined by the presence of a conserved RGS domain that canonically binds and deactivates heterotrimeric G-proteins. RGS proteins act as GTPase activating proteins (GAPs), which accelerate GTP hydrolysis on the G-protein α subunits and result in termination of signaling pathways downstream of G protein-coupled receptors. RGS10 is the smallest protein of the D/R12 subfamily and selectively interacts with Gαi proteins. It is widely expressed in many cells and tissues, with the highest expression found in the brain and immune cells. RGS10 expression is transcriptionally regulated via epigenetic mechanisms. Although RGS10 lacks multiple of the defined regulatory domains found in other RGS proteins, RGS10 contains post-translational modification sites regulating its expression, localization, and function. Additionally, RGS10 is a critical protein in the regulation of physiological processes in multiple cells, where dysregulation of its expression has been implicated in various diseases including Parkinson's disease, multiple sclerosis, osteopetrosis, chemoresistant ovarian cancer and cardiac hypertrophy. This review summarizes RGS10 features and its regulatory mechanisms, and discusses the known functions of RGS10 in cellular physiology and pathogenesis of several diseases.
Collapse
Affiliation(s)
- Faris Almutairi
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Jae-Kyung Lee
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| |
Collapse
|
47
|
Regulators of G protein signalling as pharmacological targets for the treatment of neuropathic pain. Pharmacol Res 2020; 160:105148. [PMID: 32858121 DOI: 10.1016/j.phrs.2020.105148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 07/24/2020] [Accepted: 08/10/2020] [Indexed: 12/30/2022]
Abstract
Neuropathic pain, a specific type of chronic pain resulting from persistent nervous tissue lesions, is a debilitating condition that affects about 7% of the population. This condition remains particularly difficult to treat because of the poor understanding of its underlying mechanisms. Drugs currently used to alleviate this chronic pain syndrome are of limited benefit due to their lack of efficacy and the elevated risk of side effects, especially after a prolonged period of treatment. Although drugs targeting G protein-coupled receptors (GPCR) also have several limitations, such as progressive loss of efficacy due to receptor desensitization or unavoidable side effects due to wide receptor distribution, the identification of several molecular partners that contribute to the fine-tuning of receptor activity has raised new opportunities for the development of alternative therapeutic approaches. Regulators of G protein signalling (RGS) act intracellularly by influencing the coupling process and activity of G proteins, and are amongst the best-characterized physiological modulators of GPCR. Changes in RGS expression have been documented in a range of models of neuropathic pain, or after prolonged treatment with diverse analgesics, and could participate in altered pain processing as well as impaired physiological or pharmacological control of nociceptive signals. The present review summarizes the experimental data that implicates RGS in the development of pain with focus on the pathological mechanisms of neuropathic pain, including the impact of neuropathic lesions on RGS expression and, reciprocally, the influence of modifying RGS on GPCRs involved in the modulation of nociception as well as on the outcome of pain. In this context, we address the question of the relevance of RGS as promising targets in the treatment of neuropathic pain.
Collapse
|
48
|
Koch JN, Dahlen SA, Owens EA, Osei-Owusu P. Regulator of G Protein Signaling 2 Facilitates Uterine Artery Adaptation During Pregnancy in Mice. J Am Heart Assoc 2020; 8:e010917. [PMID: 31030617 PMCID: PMC6512123 DOI: 10.1161/jaha.118.010917] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Decreased uterine blood flow is known to contribute to pregnancy complications such as gestational hypertension and preeclampsia. Previously, we showed that the loss of regulator of G protein signaling 2 ( RGS 2), a GTP ase activating protein for Gq/11 and Gi/o class G proteins, decreases uterine blood flow in the nonpregnant state in mice. Here, we examined the effects of the absence of RGS 2 and 5 on uterine blood flow and uterine vascular structure and function at early, mid, and late gestation, as well as peripartum period in mice. Methods and Results Abdominal Doppler ultrasonography was performed on adult female wild-type, Rgs2-/-, and Rgs5-/- mice at pre-pregnancy, gestational days 10, 15, and 18, and postpartum day 3. Uterine artery structure and function were also assessed by vessel myograph studies. At mid-pregnancy, uterine blood flow decreased in both Rgs2-/- and Rgs5-/- mice, whereas resistive index increased only in Rgs2-/- mice. In uterine arteries from wild-type mice, mRNA expression of RGS 2 and 4 increased, whereas RGS 5 expression remained elevated at mid-pregnancy. These changes in gene expression were unique to uterine arteries because they were absent in mesenteric arteries and the aorta of wild-type mice. In Rgs2-/- mice, uterine artery medial cross-sectional area and G protein-coupled receptor-mediated vasoconstriction increased in mid-pregnancy, implicating a role for RGS 2 in structural and functional remodeling of uterine arteries during pregnancy. In contrast, RGS 5 absence increased vasoconstriction only in the peripartum period. Conclusions These data together indicate that RGS 2 plays a critical role in the structural and functional remodeling of uterine arteries to impact uterine blood flow during pregnancy. Targeting the signaling pathway regulated by RGS 2 may therefore be a therapeutic strategy for ameliorating utero-placental perfusion disorders during pregnancy.
Collapse
Affiliation(s)
- Jennifer N Koch
- 1 Department of Pharmacology and Physiology Drexel University College of Medicine Philadelphia PA
| | - Shelby A Dahlen
- 1 Department of Pharmacology and Physiology Drexel University College of Medicine Philadelphia PA
| | - Elizabeth A Owens
- 1 Department of Pharmacology and Physiology Drexel University College of Medicine Philadelphia PA
| | - Patrick Osei-Owusu
- 1 Department of Pharmacology and Physiology Drexel University College of Medicine Philadelphia PA
| |
Collapse
|
49
|
Patella F, Cutler DF. RGS4 controls secretion of von Willebrand factor to the subendothelial matrix. J Cell Sci 2020; 133:jcs247312. [PMID: 32576664 DOI: 10.1242/jcs.247312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/11/2020] [Indexed: 11/20/2022] Open
Abstract
The haemostatic protein von Willebrand factor (VWF) exists in plasma and subendothelial pools. The plasma pools are secreted from endothelial storage granules, Weibel-Palade bodies (WPBs), by basal secretion with a contribution from agonist-stimulated secretion, and the subendothelial pool is secreted into the subendothelial matrix by a constitutive pathway not involving WPBs. We set out to determine whether the constitutive release of subendothelial VWF is actually regulated and, if so, what functional consequences this might have. Constitutive VWF secretion can be increased by a range of factors, including changes in VWF expression, levels of TNF and other environmental cues. An RNA-seq analysis revealed that expression of regulator of G protein signalling 4 (RGS4) was reduced in endothelial cells (HUVECs) grown under these conditions. siRNA RGS4 treatment of HUVECs increased constitutive basolateral secretion of VWF, probably by affecting the anterograde secretory pathway. In a simple model of endothelial damage, we show that RGS4-silenced cells increased platelet recruitment onto the subendothelial matrix under flow. These results show that changes in RGS4 expression alter levels of subendothelial VWF, affecting platelet recruitment. This introduces a novel control over VWF function.
Collapse
Affiliation(s)
- Francesca Patella
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Daniel F Cutler
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| |
Collapse
|
50
|
Hernandez KR, Karim ZA, Qasim H, Druey KM, Alshbool FZ, Khasawneh FT. Regulator of G-Protein Signaling 16 Is a Negative Modulator of Platelet Function and Thrombosis. J Am Heart Assoc 2020; 8:e011273. [PMID: 30791801 PMCID: PMC6474914 DOI: 10.1161/jaha.118.011273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background Members of the regulator of G‐protein signaling (RGS) family inhibit G‐protein coupled receptor signaling by modulating G‐protein activity. In platelets, there are 3 different RGS isoforms that are expressed at the protein level, including RGS16. Recently, we have shown that CXCL12 regulates platelet function via RGS16. However, the role of RGS16 in platelet function and thrombus formation is poorly defined. Methods and Results We used a genetic knockout mouse model approach to examine the role(s) of RGS16 in platelet activation by using a host of in vitro and in vivo assays. We observed that agonist‐induced platelet aggregation, secretion, and integrin activation were much more pronounced in platelets from the RGS16 knockout (Rgs16−/−) mice relative to their wild type (Rgs16+/+) littermates. Furthermore, the Rgs16−/− mice had a markedly shortened bleeding time and were more susceptible to vascular injury–associated thrombus formation than the controls. Conclusions These findings support a critical role for RGS16 in regulating hemostatic and thrombotic functions of platelets in mice. Hence, RGS16 represents a potential therapeutic target for modulating platelet function.
Collapse
Affiliation(s)
- Keziah R Hernandez
- 1 Pharmaceutical Sciences, School of Pharmacy The University of Texas at El Paso TX
| | - Zubair A Karim
- 1 Pharmaceutical Sciences, School of Pharmacy The University of Texas at El Paso TX
| | - Hanan Qasim
- 1 Pharmaceutical Sciences, School of Pharmacy The University of Texas at El Paso TX
| | - Kirk M Druey
- 2 Molecular Signal Transduction Section Laboratory of Allergic Diseases NIAID/NIH Bethesda MD
| | - Fatima Z Alshbool
- 1 Pharmaceutical Sciences, School of Pharmacy The University of Texas at El Paso TX
| | - Fadi T Khasawneh
- 1 Pharmaceutical Sciences, School of Pharmacy The University of Texas at El Paso TX
| |
Collapse
|