1
|
Mandal S, Flood BE, Lunzer M, Kumar D, Bailey JV. Fluoride and gallein regulate polyphosphate accumulation in dental caries-associated Lacticaseibacillus. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001519. [PMID: 39607745 PMCID: PMC11604172 DOI: 10.1099/mic.0.001519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Inorganic polyphosphates (polyPs) are energy-storing biopolymers synthesized by all three domains of life. PolyP accumulation has been well studied with respect to its role in stress response, but its role in dental disease has received less attention. Dental decay can be promoted by changes in pH as well as the chemical activity of ions such as phosphate in oral fluids at the enamel interface. Previous work has shown that the drawdown of phosphate from biofilm fluids can alter the saturation state of oral fluids to thermodynamically favour mineral dissolution. The members of the Lactobacillaceae are known to accumulate polyP and play a role in early-stage and late-stage dental caries. In this study, we examined the effects of potential metabolic inhibitors on polyP accumulation in Lacticaseibacillus rhamnosus. We observed that two inhibitors of the enzyme responsible for polyP synthesis, gallein and fluoride, inhibited polyP accumulation in a balanced medium. However, fluoride and gallein treatments amended with either glucose or lactate were found to enhance polyP accumulation. These results illustrate the potential complexity of polyP metabolism in the oral environment.
Collapse
Affiliation(s)
- Subhrangshu Mandal
- Department of Earth & Environmental Sciences, University of Minnesota – Twin Cities, Minneapolis, MN 55455, USA
- Department of Botany, Visva Bharati University, Bolpur, West Bengal, India
| | - Beverly E. Flood
- Department of Earth & Environmental Sciences, University of Minnesota – Twin Cities, Minneapolis, MN 55455, USA
| | - Mark Lunzer
- Department of Diagnostic and Biological Sciences, University of Minnesota – Twin Cities, Minneapolis, MN 55455, USA
| | - Dhiraj Kumar
- Department of Diagnostic and Biological Sciences, University of Minnesota – Twin Cities, Minneapolis, MN 55455, USA
| | - Jake V. Bailey
- Department of Earth & Environmental Sciences, University of Minnesota – Twin Cities, Minneapolis, MN 55455, USA
| |
Collapse
|
2
|
Wei W, Smrcka AV. Internalized β2-Adrenergic Receptors Oppose PLC-Dependent Hypertrophic Signaling. Circ Res 2024; 135:e24-e38. [PMID: 38813686 PMCID: PMC11223973 DOI: 10.1161/circresaha.123.323201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Chronically elevated neurohumoral drive, and particularly elevated adrenergic tone leading to β-adrenergic receptor (β-AR) overstimulation in cardiac myocytes, is a key mechanism involved in the progression of heart failure. β1-AR (β1-adrenergic receptor) and β2-ARs (β2-adrenergic receptor) are the 2 major subtypes of β-ARs present in the human heart; however, they elicit different or even opposite effects on cardiac function and hypertrophy. For example, chronic activation of β1-ARs drives detrimental cardiac remodeling while β2-AR signaling is protective. The underlying molecular mechanisms for cardiac protection through β2-ARs remain unclear. METHODS β2-AR signaling mechanisms were studied in isolated neonatal rat ventricular myocytes and adult mouse ventricular myocytes using live cell imaging and Western blotting methods. Isolated myocytes and mice were used to examine the roles of β2-AR signaling mechanisms in the regulation of cardiac hypertrophy. RESULTS Here, we show that β2-AR activation protects against hypertrophy through inhibition of phospholipaseCε signaling at the Golgi apparatus. The mechanism for β2-AR-mediated phospholipase C inhibition requires internalization of β2-AR, activation of Gi and Gβγ subunit signaling at endosome and ERK (extracellular regulated kinase) activation. This pathway inhibits both angiotensin II and Golgi-β1-AR-mediated stimulation of phosphoinositide hydrolysis at the Golgi apparatus ultimately resulting in decreased PKD (protein kinase D) and histone deacetylase 5 phosphorylation and protection against cardiac hypertrophy. CONCLUSIONS This reveals a mechanism for β2-AR antagonism of the phospholipase Cε pathway that may contribute to the known protective effects of β2-AR signaling on the development of heart failure.
Collapse
Affiliation(s)
- Wenhui Wei
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Alan V. Smrcka
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| |
Collapse
|
3
|
Sanchez GA, Smrcka AV, Jutkiewicz EM. Biasing G βγ Downstream Signaling with Gallein Inhibits Development of Morphine Tolerance and Potentiates Morphine-Induced Nociception in a Tolerant State. Mol Pharmacol 2024; 106:47-55. [PMID: 38769020 PMCID: PMC11187686 DOI: 10.1124/molpharm.124.000875] [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: 01/19/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024] Open
Abstract
Opioid analgesics are widely used as a treatment option for pain management and relief. However, the misuse of opioid analgesics has contributed to the current opioid epidemic in the United States. Prescribed opioids such as morphine, codeine, oxycodone, and fentanyl are mu-opioid receptor (MOR) agonists primarily used in the clinic to treat pain or during medical procedures, but development of tolerance limits their utility for treatment of chronic pain. Here we explored the effects of biasing Gβγ signaling on tolerance development after chronic morphine treatment in vivo. We hypothesized that biasing Gβγ signaling with gallein could prevent activation of regulatory signaling pathways that result in tolerance to antinociceptive effects of MOR agonists. Gallein has been shown to bind to Gβγ and inhibit interactions of Gβγ with phospholipase-Cβ3 (PLCβ3) or G-protein-coupled receptor kinase 2 (GRK2) but not G-protein inwardly rectifying potassium (GIRK) channels. In mice, morphine-induced antinociception was evaluated in the 55°C warm water tail withdrawal assay. We used two paradigms for gallein treatment: administration during and after three times-daily morphine administration. Our results show that gallein cotreatment during repeated administration of morphine decreased opioid tolerance development and that gallein treatment in an opioid-tolerant state enhanced the potency of morphine. Mechanistically, our data suggest that PLCβ3 is necessary for potentiating effects of gallein in an opioid-tolerant state but not in preventing the development of tolerance. These studies demonstrate that small molecules that target Gβγ signaling could reduce the need for large doses of opioid analgesics to treat pain by producing an opioid-sparing effect. SIGNIFICANCE STATEMENT: Biasing Gβγ signaling prevents tolerance to repeated morphine administration in vivo and potentiates the antinociceptive effects of morphine in an opioid-tolerant state. Mechanistically, phospholipase-Cβ is necessary for potentiating effects of gallein in an opioid-tolerant state but not in preventing the development of tolerance. This study identifies a novel treatment strategy to decrease the development of tolerance to the analgesic effects of mu-opioid receptor agonists, which are necessary to improve pain treatment and decrease the incidence of opioid use disorder.
Collapse
Affiliation(s)
- Gissell A Sanchez
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
4
|
Manning JJ, Finlay DB, Glass M. GPCR kinase subtype requirements for arrestin-2 and -3 translocation to the cannabinoid CB 1 receptor and the consequences on G protein signalling. Biochem Pharmacol 2024; 224:116190. [PMID: 38604257 DOI: 10.1016/j.bcp.2024.116190] [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: 11/30/2023] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
Arrestins are key negative regulators of G Protein-Coupled Receptors (GPCRs) through mediation of G protein desensitisation and receptor internalisation. Arrestins can also contribute to signal transduction by scaffolding downstream signalling effectors for activation. GPCR kinase (GRK) enzymes phosphorylate the intracellular C-terminal domain, or intracellular loop regions of GPCRs to promote arrestin interaction. There are seven different GRK subtypes, which may uniquely phosphorylate the C-terminal tail in a type of 'phosphorylation barcode,' potentially differentially contributing to arrestin translocation and arrestin-dependent signalling. Such contributions may be exploited to develop arrestin-biased ligands. Here, we examine the effect of different GRK subtypes on the ability to promote translocation of arrestin-2 and arrestin-3 to the cannabinoid CB1 receptor (CB1) with a range of ligands. We find that most GRK subtypes (including visual GRK1) can enhance arrestin-2 and -3 translocation to CB1, and that GRK-dependent changes in arrestin-2 and arrestin-3 translocation were broadly shared for most agonists tested. GRK2/3 generally enhanced arrestin translocation more than the other GRK subtypes, with some small differences between ligands. We also explore the interplay between G protein activity and GRK2/3-dependent arrestin translocation, highlighting that high-efficacy G protein agonists will cause GRK2/3 dependent arrestin translocation. This study supports the hypothesis that arrestin-biased ligands for CB1 must engage GRK5/6 rather than GRK2/3, and G protein-biased ligands must have inherently low efficacy.
Collapse
Affiliation(s)
- Jamie J Manning
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - David B Finlay
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Michelle Glass
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
5
|
Lin J, Scullion L, Garland CJ, Dora K. Gβγ subunit signalling underlies neuropeptide Y-stimulated vasoconstriction in rat mesenteric and coronary arteries. Br J Pharmacol 2023; 180:3045-3058. [PMID: 37460913 PMCID: PMC10953346 DOI: 10.1111/bph.16192] [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: 02/13/2023] [Revised: 06/27/2023] [Accepted: 07/09/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Raised serum concentrations of the sympathetic co-transmitter neuropeptide Y (NPY) are linked to cardiovascular diseases. However, the signalling mechanism for vascular smooth muscle (VSM) constriction to NPY is poorly understood. Therefore, the present study investigated the mechanisms of NPY-induced vasoconstriction in rat small mesenteric (RMA) and coronary (RCA) arteries. EXPERIMENTAL APPROACH Third-order mesenteric or intra-septal arteries from male Wistar rats were assessed in wire myographs for isometric tension, VSM membrane potential and VSM intracellular Ca2+ events. KEY RESULTS NPY stimulated concentration-dependent vasoconstriction in both RMA and RCA, which was augmented by blocking NO synthase or endothelial denudation in RMA. NPY-mediated vasoconstriction was blocked by the selective Y1 receptor antagonist BIBO 3304 and Y1 receptor protein expression was detected in both the VSM and endothelial cells in RMA and RCA. The selective Gβγ subunit inhibitor gallein and the PLC inhibitor U-73122 attenuated NPY-induced vasoconstriction. Signalling via the Gβγ-PLC pathway stimulated VSM Ca2+ waves and whole-field synchronised Ca2+ flashes in RMA and increased the frequency of Ca2+ flashes in myogenically active RCA. Furthermore, in RMA, the Gβγ pathway linked NPY to VSM depolarization and generation of action potential-like spikes associated with intense vasoconstriction. This depolarization activated L-type voltage-gated Ca2+ channels, as nifedipine abolished NPY-mediated vasoconstriction. CONCLUSIONS AND IMPLICATIONS These data suggest that the Gβγ subunit, which dissociates upon Y1 receptor activation, initiates VSM membrane depolarization and Ca2+ mobilisation to cause vasoconstriction. This model may help explain the development of microvascular vasospasm during raised sympathetic nerve activity.
Collapse
Affiliation(s)
- JinHeng Lin
- Department of PharmacologyUniversity of OxfordOxfordUK
| | | | | | - Kim Dora
- Department of PharmacologyUniversity of OxfordOxfordUK
| |
Collapse
|
6
|
Zhu H, Liu X, Wang X, Li Y, Ma F, Tan B, Zhou P, Fu F, Su R. Gβγ subunit inhibitor decreases DOM-induced head twitch response via the PLCβ/IP3/Ca 2+/ERK and cAMP signaling pathways. Eur J Pharmacol 2023; 957:176038. [PMID: 37657742 DOI: 10.1016/j.ejphar.2023.176038] [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: 03/12/2023] [Revised: 08/17/2023] [Accepted: 08/30/2023] [Indexed: 09/03/2023]
Abstract
AIMS (-)-2,5-dimethoxy-4-methylamphetamine (DOM) induces the head-twitch response (HTR) primarily by activating the serotonin 5-hydroxytryptamine 2A receptor (5-HT2A receptor) in mice. However, the mechanisms underlying 5-HT2A receptor activation and the HTR remain elusive. Gβγ subunits are a potential treatment target in numerous diseases. The present study investigated the mechanism whereby Gβγ subunits influence DOM-induced HTR. MAIN METHODS The effects of the Gβγ inhibitor 3',4',5',6'-tetrahydroxyspiro[2-benzofuran-3,9'-xanthene]-1-one (gallein) and antagonistic peptide βARKct (β-adrenergic receptor kinase C-terminal fragment) on DOM-induced HTR were studied via an HTR test. The activation of the phospholipase C β (PLCβ)/inositol triphosphate (IP3)/calcium (Ca2+) signaling pathway and extracellular signal-regulated kinase (ERK) following Gβγ subunit inhibition was detected by western blotting, Homogeneous Time-Resolved Fluorescence (HTRF) inositol phosphate (IP1) assay and Fluorometric Imaging Plate Reader (FLIPR) calcium 6 assay. The Gβγ subunit-mediated regulation of cyclic adenosine monophosphate (cAMP) was assessed via a GloSensor™ cAMP assay. KEY FINDINGS The Gβγ subunit inhibitors gallein and βARKct reduced DOM-induced HTR in C57BL/6J mice. Like the 5-HT2A receptor-selective antagonist (R)-[2,3-di(methoxy)phenyl]-[1-[2-(4-fluorophenyl)ethyl]piperidin-4-yl]methanol (M100907), gallein inhibited PLCβ phosphorylation (pPLCβ), IP1 production, Ca2+ transients, ERK1/2 phosphorylation (pERK1/2) and cAMP accumulation induced by DOM in human embryonic kidney (HEK) 293T cells stably or transiently transfected with the human 5-HT2A receptor. Moreover, PLCβ protein inhibitor 1-[6-[[(8R,9S,13S,14S,17S)-3-methoxy-13-methyl-6,7,8,9,11,12,14,15,16,17-decahydrocyclopenta[a]phenanthren-17-yl]amino]hexyl]pyrrole-2,5-dione (U73122) (10 nmol/mouse), intracellular Ca2+ blocker 6-[6-[6-[5-acetamido-4,6-dihydroxy-2-(sulfooxymethyl)oxan-3-yl]oxy-2-carboxy-4-hydroxy-5-sulfooxyoxan-3-yl]oxy-2-(hydroxymethyl)-5-(sulfoamino)-4-sulfooxyoxan-3-yl]oxy-3,4-dihydroxy-5-sulfooxyoxane-2-carboxylic acid (heparin) (5 nmol/mouse), L-type Ca2+ channel blocker 3-O-(2-methoxyethyl) 5-O-propan-2-yl 2,6-dimethyl-4-(3-nitrophenyl)-1,4-dihydropyridine-3,5-dicarboxylate (nimodipine) (4 mg/kg), mitogen extracellular regulating kinase 1/2 (MEK1/2) inhibitor (Z)-3-amino-3-(4-aminophenyl)sulfanyl-2-[2-(trifluoromethyl)phenyl]prop-2-enenitrile (SL327) (30 mg/kg), and Gαs protein selective antagonist 4,4',4″,4‴-(Carbonylbis-(imino-5,1,3-benzenetriylbis(carbonylimino)))tetrakisbenzene-1,3-disulfonic acid (NF449) (10 nmol/mouse) reduced DOM-induced HTR in C57BL/6J mice. SIGNIFICANCE The Gβγ subunits potentially mediate the HTR after 5-HT2A receptor activation via the PLCβ/IP3/Ca2+/ERK1/2 and cAMP signaling pathways. Inhibitors targeting the Gβγ subunits potentially inhibit the hallucinogenic effects of 5-HT2A receptor agonists.
Collapse
Affiliation(s)
- Huili Zhu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China; School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Xiaoqian Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Xiaoxuan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Yulei Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Fang Ma
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Bo Tan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Peilan Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China.
| | - Fenghua Fu
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China.
| |
Collapse
|
7
|
Wei W, Smrcka AV. Internalized β2-Adrenergic Receptors Inhibit Subcellular Phospholipase C-Dependent Cardiac Hypertrophic Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544153. [PMID: 37333278 PMCID: PMC10274790 DOI: 10.1101/2023.06.07.544153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Chronically elevated neurohumoral drive, and particularly elevated adrenergic tone leading to β-adrenergic receptor (β-AR) overstimulation in cardiac myocytes, is a key mechanism involved in the progression of heart failure. β1-AR and β2-ARs are the two major subtypes of β-ARs present in the human heart, however, they elicit different or even opposite effects on cardiac function and hypertrophy. For example, chronic activation of β1ARs drives detrimental cardiac remodeling while β2AR signaling is protective. The underlying molecular mechanisms for cardiac protection through β2ARs remain unclear. Here we show that β2-AR protects against hypertrophy through inhibition of PLCε signaling at the Golgi apparatus. The mechanism for β2AR-mediated PLC inhibition requires internalization of β2AR, activation of Gi and Gβγ subunit signaling at endosomes and ERK activation. This pathway inhibits both angiotensin II and Golgi-β1-AR-mediated stimulation of phosphoinositide hydrolysis at the Golgi apparatus ultimately resulting in decreased PKD and HDAC5 phosphorylation and protection against cardiac hypertrophy. This reveals a mechanism for β2-AR antagonism of the PLCε pathway that may contribute to the known protective effects of β2-AR signaling on the development of heart failure.
Collapse
Affiliation(s)
- Wenhui Wei
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Alan V. Smrcka
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| |
Collapse
|
8
|
Zhao J, DiGiacomo V, Ferreras-Gutierrez M, Dastjerdi S, Ibáñez de Opakua A, Park JC, Luebbers A, Chen Q, Beeler A, Blanco FJ, Garcia-Marcos M. Small-molecule targeting of GPCR-independent noncanonical G-protein signaling in cancer. Proc Natl Acad Sci U S A 2023; 120:e2213140120. [PMID: 37098067 PMCID: PMC10160980 DOI: 10.1073/pnas.2213140120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 03/06/2023] [Indexed: 04/26/2023] Open
Abstract
Activation of heterotrimeric G-proteins (Gαβγ) by G-protein-coupled receptors (GPCRs) is a quintessential mechanism of cell signaling widely targeted by clinically approved drugs. However, it has become evident that heterotrimeric G-proteins can also be activated via GPCR-independent mechanisms that remain untapped as pharmacological targets. GIV/Girdin has emerged as a prototypical non-GPCR activator of G proteins that promotes cancer metastasis. Here, we introduce IGGi-11, a first-in-class small-molecule inhibitor of noncanonical activation of heterotrimeric G-protein signaling. IGGi-11 binding to G-protein α-subunits (Gαi) specifically disrupted their engagement with GIV/Girdin, thereby blocking noncanonical G-protein signaling in tumor cells and inhibiting proinvasive traits of metastatic cancer cells. In contrast, IGGi-11 did not interfere with canonical G-protein signaling mechanisms triggered by GPCRs. By revealing that small molecules can selectively disable noncanonical mechanisms of G-protein activation dysregulated in disease, these findings warrant the exploration of therapeutic modalities in G-protein signaling that go beyond targeting GPCRs.
Collapse
Affiliation(s)
- Jingyi Zhao
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
| | - Vincent DiGiacomo
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
| | | | - Shiva Dastjerdi
- Department of Chemistry, Boston University, College of Arts & Sciences, Boston, MA02115
| | | | - Jong-Chan Park
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
| | - Alex Luebbers
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
| | - Qingyan Chen
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
| | - Aaron Beeler
- Department of Chemistry, Boston University, College of Arts & Sciences, Boston, MA02115
| | - Francisco J. Blanco
- Centro de Investigaciones Biológicas-Centro Superior de Investigaciones Cientificas, Madrid, Spain
| | - Mikel Garcia-Marcos
- Department of Biochemistry & Cell Biology, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA02118
- Department of Biology, College of Arts & Sciences, Boston University, Boston, MA02115
| |
Collapse
|
9
|
Zhao J, DiGiacomo V, Ferreras-Gutierrez M, Dastjerdi S, de Opakua AI, Park JC, Luebbers A, Chen Q, Beeler A, Blanco FJ, Garcia-Marcos M. Small-molecule targeting of GPCR-independent non-canonical G protein signaling inhibits cancer progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.18.529092. [PMID: 36824907 PMCID: PMC9949157 DOI: 10.1101/2023.02.18.529092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Activation of heterotrimeric G-proteins (Gαβγ) by G-protein-coupled receptors (GPCRs) is a quintessential mechanism of cell signaling widely targeted by clinically-approved drugs. However, it has become evident that heterotrimeric G-proteins can also be activated via GPCR-independent mechanisms that remain untapped as pharmacological targets. GIV/Girdin has emerged as a prototypical non-GPCR activator of G proteins that promotes cancer metastasis. Here, we introduce IGGi-11, a first-in-class smallmolecule inhibitor of non-canonical activation of heterotrimeric G-protein signaling. IGGi-11 binding to G-protein α-subunits (Gαi) specifically disrupted their engagement with GIV/Girdin, thereby blocking non-canonical G-protein signaling in tumor cells, and inhibiting pro-invasive traits of metastatic cancer cells in vitro and in mice. In contrast, IGGi-11 did not interfere with canonical G-protein signaling mechanisms triggered by GPCRs. By revealing that small molecules can selectively disable non-canonical mechanisms of G-protein activation dysregulated in disease, these findings warrant the exploration of therapeutic modalities in G-protein signaling that go beyond targeting GPCRs.
Collapse
|
10
|
Abd El-Hafeez AA, Sun N, Chakraborty A, Ear J, Roy S, Chamarthi P, Rajapakse N, Das S, Luker KE, Hazra TK, Luker GD, Ghosh P. Regulation of DNA damage response by trimeric G-proteins. iScience 2023; 26:105973. [PMID: 36756378 PMCID: PMC9900518 DOI: 10.1016/j.isci.2023.105973] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 08/14/2022] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Upon sensing DNA double-strand breaks (DSBs), eukaryotic cells either die or repair DSBs via one of the two competing pathways, i.e., non-homologous end-joining (NHEJ) or homologous recombination (HR). We show that cell fate after DSBs hinges on GIV/Girdin, a guanine nucleotide-exchange modulator of heterotrimeric Giα•βγ protein. GIV suppresses HR by binding and sequestering BRCA1, a key coordinator of multiple steps within the HR pathway, away from DSBs; it does so using a C-terminal motif that binds BRCA1's BRCT-modules via both phospho-dependent and -independent mechanisms. Using another non-overlapping C-terminal motif GIV binds and activates Gi and enhances the "free" Gβγ→PI-3-kinase→Akt pathway, which promotes survival and is known to suppress HR, favor NHEJ. Absence of GIV, or loss of either of its C-terminal motifs enhanced cell death upon genotoxic stress. Because GIV selectively binds other BRCT-containing proteins suggests that G-proteins may fine-tune sensing, repair, and survival after diverse types of DNA damage.
Collapse
Affiliation(s)
- Amer Ali Abd El-Hafeez
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Pharmacology and Experimental Oncology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Nina Sun
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Anirban Chakraborty
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jason Ear
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Biological Sciences Department, California State Polytechnic University, Pomona, CA 91768, USA
| | - Suchismita Roy
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Pranavi Chamarthi
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Navin Rajapakse
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Soumita Das
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | - Kathryn E. Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| | - Tapas K. Hazra
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gary D. Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI 48109-2099, USA
- Department of Microbiology and Immunology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Moores Comprehensive Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Veterans Affairs Medical Center, La Jolla, CA, USA
| |
Collapse
|
11
|
Xu X, Wu G. Non-canonical Golgi-compartmentalized Gβγ signaling: mechanisms, functions, and therapeutic targets. Trends Pharmacol Sci 2023; 44:98-111. [PMID: 36494204 PMCID: PMC9901158 DOI: 10.1016/j.tips.2022.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
G protein Gβγ subunits are key mediators of G protein-coupled receptor (GPCR) signaling under physiological and pathological conditions; their inhibitors have been tested for the treatment of human disease. Conventional wisdom is that the Gβγ complex is activated and subsequently exerts its functions at the plasma membrane (PM). Recent studies have revealed non-canonical activation of Gβγ at intracellular organelles, where the Golgi apparatus is a major locale, via translocation or local activation. Golgi-localized Gβγ activates specific signaling cascades and regulates fundamental cell processes such as membrane trafficking, proliferation, and migration. More recent studies have shown that inhibiting Golgi-compartmentalized Gβγ signaling attenuates cardiomyocyte hypertrophy and prostate tumorigenesis, indicating new therapeutic targets. We review novel activation mechanisms and non-canonical functions of Gβγ at the Golgi, and discuss potential therapeutic interventions by targeting Golgi-biased Gβγ-directed signaling.
Collapse
Affiliation(s)
- Xin Xu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
12
|
Zhang Y, Zhang J, Wang J, Chen H, Ouyang L, Wang Y. Targeting GRK2 and GRK5 for treating chronic degenerative diseases: Advances and future perspectives. Eur J Med Chem 2022; 243:114668. [DOI: 10.1016/j.ejmech.2022.114668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022]
|
13
|
Sanchez GA, Jutkiewicz EM, Ingram S, Smrcka AV. Coincident Regulation of PLC β Signaling by Gq-Coupled and μ-Opioid Receptors Opposes Opioid-Mediated Antinociception. Mol Pharmacol 2022; 102:269-279. [PMID: 36116788 PMCID: PMC11033930 DOI: 10.1124/molpharm.122.000541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
Pain management is an important problem worldwide. The current frontline approach for pain management is the use of opioid analgesics. The primary analgesic target of opioids is the μ-opioid receptor (MOR). Deletion of phospholipase Cβ3 (PLCβ3) or selective inhibition of Gβγ regulation of PLCβ3 enhances the potency of the antinociceptive effects of morphine suggesting a novel strategy for achieving opioid-sparing effects. Here we investigated a potential mechanism for regulation of PLC signaling downstream of MOR in human embryonic kidney 293 cells and found that MOR alone could not stimulate PLC but rather required a coincident signal from a Gq-coupled receptor. Knockout of PLCβ3 or pharmacological inhibition of its upstream regulators, Gβγ or Gq, ex vivo in periaqueductal gray slices increased the potency of the selective MOR agonist [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin acetate salt in inhibiting presynaptic GABA release. Finally, inhibition of Gq- G protein-coupled receptor coupling in mice enhanced the antinociceptive effects of morphine. These data support a model where Gq and Gβγ-dependent signaling cooperatively regulate PLC activation to decrease MOR-dependent antinociceptive potency. Ultimately, this could lead to identification of new non-MOR targets that would allow for lower-dose utilization of opioid analgesics. SIGNIFICANCE STATEMENT: Previous work demonstrated that deletion of phospholipase Cβ3 (PLCβ3) in mice potentiates μ-opioid receptor (MOR)-dependent antinociception. How PLCβ3 is regulated downstream of MOR had not been clearly defined. We show that PLC-dependent diacylglycerol generation is cooperatively regulated by MOR-Gβγ and Gq-coupled receptor signaling through PLCβ3 and that blockade of either Gq-signaling or Gβγ signaling enhances the potency of opioids in ex vivo brain slices and in vivo. These results reveal potential novel strategies for improving opioid analgesic potency and safety.
Collapse
Affiliation(s)
- Gissell A Sanchez
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (G.A.S., E.M.J., A.V.S.) and Department of Neurologic Surgery, Oregon Health Sciences University, Portland, Oregon (S.I.)
| | - Emily M Jutkiewicz
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (G.A.S., E.M.J., A.V.S.) and Department of Neurologic Surgery, Oregon Health Sciences University, Portland, Oregon (S.I.)
| | - Susan Ingram
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (G.A.S., E.M.J., A.V.S.) and Department of Neurologic Surgery, Oregon Health Sciences University, Portland, Oregon (S.I.)
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (G.A.S., E.M.J., A.V.S.) and Department of Neurologic Surgery, Oregon Health Sciences University, Portland, Oregon (S.I.)
| |
Collapse
|
14
|
Ferrero KM, Koch WJ. GRK2 in cardiovascular disease and its potential as a therapeutic target. J Mol Cell Cardiol 2022; 172:14-23. [PMID: 35878706 DOI: 10.1016/j.yjmcc.2022.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/29/2022] [Accepted: 07/19/2022] [Indexed: 01/25/2023]
Abstract
Cardiovascular diseases (CVDs) represent the leading cause of death globally. Despite major advances in the field of pharmacological CVD treatments, particularly in the field of heart failure (HF) research, case numbers and overall mortality remain high and have trended upwards over the last few years. Thus, identifying novel molecular targets for developing HF therapeutics remains a key research focus. G protein-coupled receptors (GPCRs) are critical myocardial signal transducers which regulate cardiac contractility, growth, adaptation and metabolism. Additionally, GPCR dysregulation underlies multiple models of cardiac pathology, and most pharmacological therapeutics currently used in HF target these receptors. Currently-approved treatments have improved patient outcomes, but therapies to stop or reverse HF are lacking. A recent focus on GPCR intracellular-regulating proteins such as GPCR kinases (GRKs) has uncovered GRK2 as a promising target for combating HF. Current literature strongly establishes increased levels and activity of GRK2 in multiple models of CVD. Additionally, the GRK2 interactome includes numerous proteins which interact with differential domains of GRK2 to modulate both beneficial and deleterious signaling pathways in the heart, indicating that these domains can be targeted with a high level of specificity unique to various cardiac pathologies. These data support the premise that GRK2 should be at the forefront of a novel investigative drug search. This perspective reviews cardiac GPCRs, describes the structure and functions of GRK2 in cardiac function and maladaptive pathology, and summarizes the ongoing and future research for targeting this critical kinase across cellular, animal and human models of cardiac dysfunction and HF.
Collapse
Affiliation(s)
- Kimberly M Ferrero
- Lewis Katz School of Medicine at Temple University, Department of Cardiovascular Sciences, Philadelphia, PA, USA; Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Philadelphia, PA, USA
| | - Walter J Koch
- Lewis Katz School of Medicine at Temple University, Department of Cardiovascular Sciences, Philadelphia, PA, USA; Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Dai SA, Hu Q, Gao R, Blythe EE, Touhara KK, Peacock H, Zhang Z, von Zastrow M, Suga H, Shokat KM. State-selective modulation of heterotrimeric Gαs signaling with macrocyclic peptides. Cell 2022; 185:3950-3965.e25. [PMID: 36170854 PMCID: PMC9747239 DOI: 10.1016/j.cell.2022.09.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/08/2022] [Accepted: 09/07/2022] [Indexed: 01/26/2023]
Abstract
The G protein-coupled receptor cascade leading to production of the second messenger cAMP is replete with pharmacologically targetable proteins, with the exception of the Gα subunit, Gαs. GTPases remain largely undruggable given the difficulty of displacing high-affinity guanine nucleotides and the lack of other drug binding sites. We explored a chemical library of 1012 cyclic peptides to expand the chemical search for inhibitors of this enzyme class. We identified two macrocyclic peptides, GN13 and GD20, that antagonize the active and inactive states of Gαs, respectively. Both macrocyclic peptides fine-tune Gαs activity with high nucleotide-binding-state selectivity and G protein class-specificity. Co-crystal structures reveal that GN13 and GD20 distinguish the conformational differences within the switch II/α3 pocket. Cell-permeable analogs of GN13 and GD20 modulate Gαs/Gβγ signaling in cells through binding to crystallographically defined pockets. The discovery of cyclic peptide inhibitors targeting Gαs provides a path for further development of state-dependent GTPase inhibitors.
Collapse
Affiliation(s)
- Shizhong A Dai
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Qi Hu
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Rong Gao
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Emily E Blythe
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kouki K Touhara
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hayden Peacock
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ziyang Zhang
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Mark von Zastrow
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Kevan M Shokat
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
16
|
Cxcl10 chemokine induces migration of ING4-deficient breast cancer cells via a novel crosstalk mechanism between the Cxcr3 and Egfr receptors. Mol Cell Biol 2021; 42:e0038221. [PMID: 34871062 DOI: 10.1128/mcb.00382-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The chemokine Cxcl10 has been associated with poor prognosis in breast cancer, but the mechanism is not well understood. Our previous study have shown that CXCL10 was repressed by the ING4 tumor suppressor, suggesting a potential inverse functional relationship. We thus investigated a role for Cxcl10 in the context of ING4 deficiencies in breast cancer. We first analyzed public gene expression datasets and found that patients with CXCL10-high/ING4-low expressing tumors had significantly reduced disease-free survival in breast cancer. In vitro, Cxcl10 induced migration of ING4-deleted breast cancer cells, but not of ING4-intact cells. Using inhibitors, we found that Cxcl10-induced migration of ING4-deleted cells required Cxcr3, Egfr, and the Gβγ subunits downstream of Cxcr3, but not Gαi. Immunofluorescent imaging showed that Cxcl10 induced early transient colocalization between Cxcr3 and Egfr in both ING4-intact and ING4-deleted cells, which recurred only in ING4-deleted cells. A peptide agent that binds to the internal juxtamembrane domain of Egfr inhibited Cxcr3/Egfr colocalization and cell migration. Taken together, these results presented a novel mechanism of Cxcl10 that elicits migration of ING4-deleted cells, in part by inducing a physical or proximal association between Cxcr3 and Egfr and signaling downstream via Gβγ. These results further indicated that ING4 plays a critical role in the regulation of Cxcl10 signaling that enables breast cancer progression.
Collapse
|
17
|
Seibel-Ehlert U, Plank N, Inoue A, Bernhardt G, Strasser A. Label-Free Investigations on the G Protein Dependent Signaling Pathways of Histamine Receptors. Int J Mol Sci 2021; 22:9739. [PMID: 34575903 PMCID: PMC8467282 DOI: 10.3390/ijms22189739] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 01/14/2023] Open
Abstract
G protein activation represents an early key event in the complex GPCR signal transduction process and is usually studied by label-dependent methods targeting specific molecular events. However, the constrained environment of such "invasive" techniques could interfere with biological processes. Although histamine receptors (HRs) represent (evolving) drug targets, their signal transduction is not fully understood. To address this issue, we established a non-invasive dynamic mass redistribution (DMR) assay for the human H1-4Rs expressed in HEK cells, showing excellent signal-to-background ratios above 100 for histamine (HIS) and higher than 24 for inverse agonists with pEC50 values consistent with literature. Taking advantage of the integrative nature of the DMR assay, the involvement of endogenous Gαq/11, Gαs, Gα12/13 and Gβγ proteins was explored, pursuing a two-pronged approach, namely that of classical pharmacology (G protein modulators) and that of molecular biology (Gα knock-out HEK cells). We showed that signal transduction of hH1-4Rs occurred mainly, but not exclusively, via their canonical Gα proteins. For example, in addition to Gαi/o, the Gαq/11 protein was proven to contribute to the DMR response of hH3,4Rs. Moreover, the Gα12/13 was identified to be involved in the hH2R mediated signaling pathway. These results are considered as a basis for future investigations on the (patho)physiological role and the pharmacological potential of H1-4Rs.
Collapse
Affiliation(s)
- Ulla Seibel-Ehlert
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040 Regensburg, Germany; (N.P.); (G.B.)
| | - Nicole Plank
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040 Regensburg, Germany; (N.P.); (G.B.)
| | - Asuka Inoue
- Department of Pharmacological Sciences, Tohoku University, Sendai 980-8578, Japan;
| | - Guenther Bernhardt
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040 Regensburg, Germany; (N.P.); (G.B.)
| | - Andrea Strasser
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, 93040 Regensburg, Germany; (N.P.); (G.B.)
| |
Collapse
|
18
|
Lyu C, Ye Y, Lensing MM, Wagner KU, Weigel RJ, Chen S. Targeting Gi/o protein-coupled receptor signaling blocks HER2-induced breast cancer development and enhances HER2-targeted therapy. JCI Insight 2021; 6:e150532. [PMID: 34343132 PMCID: PMC8492335 DOI: 10.1172/jci.insight.150532] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/30/2021] [Indexed: 12/02/2022] Open
Abstract
GPCRs are highly desirable drug targets for human disease. Although GPCR dysfunction drives development and progression of many tumors, including breast cancer (BC), targeting individual GPCRs has limited efficacy as a cancer therapy because numerous GPCRs are activated. Here, we sought a new way of blocking GPCR activation in HER2+ BC by targeting a subgroup of GPCRs that couple to Gi/o proteins (Gi/o-GPCRs). In mammary epithelial cells of transgenic mouse models, and BC cell lines, HER2 hyperactivation altered GPCR expression, particularly, Gi/o-GPCR expression. Gi/o-GPCR stimulation transactivated EGFR and HER2 and activated the PI3K/AKT and Src pathways. If we uncoupled Gi/o-GPCRs from their cognate Gi/o proteins by pertussis toxin (PTx), then BC cell proliferation and migration was inhibited in vitro and HER2-driven tumor formation and metastasis were suppressed in vivo. Moreover, targeting Gi/o-GPCR signaling via PTx, PI3K, or Src inhibitors enhanced HER2-targeted therapy. These results indicate that, in BC cells, HER2 hyperactivation drives aberrant Gi/o-GPCR signaling and Gi/o-GPCR signals converge on the PI3K/AKT and Src signaling pathways to promote cancer progression and resistance to HER2-targeted therapy. Our findings point to a way to pharmacologically deactivate GPCR signaling to block tumor growth and enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Cancan Lyu
- Department of Neuroscience and Pharmacology, The University of Iowa Carver College of Medicine, Iowa City, United States of America
| | - Yuanchao Ye
- Department of Neuroscience and Pharmacology, The University of Iowa Carver College of Medicine, Iowa City, United States of America
| | - Maddison M Lensing
- Department of Neuroscience and Pharmacology, The University of Iowa Carver College of Medicine, Iowa City, United States of America
| | - Kay-Uwe Wagner
- Department of Oncology, Wayne State University School of Medicine, Detroit, United States of America
| | - Ronald J Weigel
- Department of Surgery, The University of Iowa Carver College of Medicine, Iowa City, United States of America
| | - Songhai Chen
- Department of Neuroscience and Pharmacology, The University of Iowa Carver College of Medicine, Iowa City, United States of America
| |
Collapse
|
19
|
Abd El-hafeez AA, Sun N, Chakraborty A, Ear J, Roy S, Chamarthi P, Rajapakse N, Das S, Luker KE, Hazra TK, Luker GD, Ghosh P. Regulation of DNA damage response by trimeric G-protein Signaling.. [DOI: 10.1101/2021.07.21.452842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
AbstractUpon sensing DNA double-strand breaks (DSBs), eukaryotic cells either die or repair DSBs via one of two competing pathways, i.e., non-homologous end-joining (NHEJ) or homologous recombination (HR). We show that cell fate after DNA damage hinges on the guanine nucleotide-exchange modulator of heterotrimeric G-protein, Giα•βγ, GIV/Girdin. GIV suppresses HR by binding and sequestering BRCA1, a key coordinator of multiple steps within the HR pathway, away from DSBs; it does so using a C-terminal motif that binds BRCA1’s BRCT-modules via both phospho-dependent and -independent mechanisms. GIV promotes NHEJ, and binds and activates Gi and enhances the ‘free’ Gβγ→PI-3-kinase→Akt pathway, thus revealing the enigmatic origin of prosurvival Akt signals during dsDNA repair. Absence of GIV, or the loss of either of its two functions impaired DNA repair, and induced cell death when challenged with numerous cytotoxic agents. That GIV selectively binds few other BRCT-containing proteins suggests convergent signaling such that heterotrimeric G-proteins may finetune sensing, repair, and outcome after DNA damage.GRAPHIC ABSTRACTHIGHLIGHTSNon-receptor G protein modulator, GIV/Girdin binds BRCA1Binding occurs in both canonical and non-canonical modesGIV sequesters BRCA1 away from dsDNA breaks, suppresses HRActivation of Gi by GIV enhances Akt signals, favors NHEJIN BRIEFIn this work, the authors show that heterotrimeric G protein signaling that is triggered by non-receptor GEF, GIV/Girdin, in response to double-stranded DNA breaks is critical for decisive signaling events which favor non-homologous end-joining (NHEJ) and inhibit homologous recombination (HR).
Collapse
|
20
|
Valencia-Guzmán CJ, Castro-Ruiz JE, García-Gasca T, Rojas-Molina A, Romo-Mancillas A, Luna-Vázquez FJ, Rojas-Molina JI, Ibarra-Alvarado C. Endothelial TRP channels and cannabinoid receptors are involved in affinin-induced vasodilation. Fitoterapia 2021; 153:104985. [PMID: 34237389 DOI: 10.1016/j.fitote.2021.104985] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/24/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023]
Abstract
Affinin is mainly recognized by its antinociceptive effect. Recently, our research group demonstrated that this compound produces vasodilation via activation of the gasotransmitters signaling pathways. However, the molecular targets of affinin were not identified. Considering the structural similarity of this alkamide with anandamide, we hypothesized that affinin-induced vasodilation could involve participation of TRP channels and cannabinoid receptors. In this work, by using the isolated rat aorta assay, we assessed involvement of TRP channels, the cannabinoid system, and the HNO-CGRP-TRPA1 pathway on the mechanism of action of affinin. Additionally, we measured NO and H2S levels elicited by affinin on rat aorta homogenates and carried out computer simulations of molecular interactions between affinin and the TRPA1 and TRPV1 channels and the CB1 receptor. Our results indicated that affinin induces an increase in aortic NO and H2S levels. We found evidence that the vasodilator effect induced by affinin involves activation of TRPA1 and TRPV1 channels and the CB1 and eCB receptors. In silico analyses showed that affinin is able to bind with high affinity to these molecular targets. Moreover, we also proved that affinin-induced vasodilation is partly mediated via activation of the HNO-TRPA1-CGRP pathway. Based on these results we propose a novel mechanism of action to explain the vasodilatory effect of affinin, which could be developed as an alternative drug to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Christian J Valencia-Guzmán
- Posgrado en Ciencias Químico Biológicas, Facultad de Química, Universidad Autónoma de Querétaro, Cerro de las Campanas S/N, Querétaro 76010, Mexico; Laboratorio de Investigación Química y Farmacológica de Productos Naturales, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico.
| | - Jesús E Castro-Ruiz
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Juriquilla, Querétaro 76230, Mexico.
| | - Teresa García-Gasca
- Laboratorio de Biología Celular y Molecular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Campus Juriquilla, Querétaro 76230, Mexico.
| | - Alejandra Rojas-Molina
- Laboratorio de Investigación Química y Farmacológica de Productos Naturales, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico.
| | - Antonio Romo-Mancillas
- Laboratorio de Diseño Asistido por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico.
| | - Francisco J Luna-Vázquez
- Laboratorio de Investigación Química y Farmacológica de Productos Naturales, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico
| | - Juana I Rojas-Molina
- Laboratorio de Investigación Química y Farmacológica de Productos Naturales, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico
| | - César Ibarra-Alvarado
- Laboratorio de Investigación Química y Farmacológica de Productos Naturales, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico.
| |
Collapse
|
21
|
GRKs and Epac1 Interaction in Cardiac Remodeling and Heart Failure. Cells 2021; 10:cells10010154. [PMID: 33466800 PMCID: PMC7830799 DOI: 10.3390/cells10010154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/25/2022] Open
Abstract
β-adrenergic receptors (β-ARs) play a major role in the physiological regulation of cardiac function through signaling routes tightly controlled by G protein-coupled receptor kinases (GRKs). Although the acute stimulation of β-ARs and the subsequent production of cyclic AMP (cAMP) have beneficial effects on cardiac function, chronic stimulation of β-ARs as observed under sympathetic overdrive promotes the development of pathological cardiac remodeling and heart failure (HF), a leading cause of mortality worldwide. This is accompanied by an alteration in cAMP compartmentalization and the activation of the exchange protein directly activated by cAMP 1 (Epac1) signaling. Among downstream signals of β-ARs, compelling evidence indicates that GRK2, GRK5, and Epac1 represent attractive therapeutic targets for cardiac disease. Here, we summarize the pathophysiological roles of GRK2, GRK5, and Epac1 in the heart. We focus on their signalosome and describe how under pathological settings, these proteins can cross-talk and are part of scaffolded nodal signaling systems that contribute to a decreased cardiac function and HF development.
Collapse
|
22
|
Tian JY, Chi CL, Bian G, Guo FJ, Wang XQ, Yu B. A novel GPCR target in correlation with androgen deprivation therapy for prostate cancer drug discovery. Basic Clin Pharmacol Toxicol 2020; 128:195-203. [PMID: 32991779 DOI: 10.1111/bcpt.13499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 12/24/2022]
Abstract
Most prostate carcinomas require androgen stimulation to grow, and for nearly 70 years, androgen ablation therapy has been one of the central therapeutic strategies against advanced prostate cancer. Although most tumours initially respond to this therapy, some will be acquired resistant and progress to metastatic castration-resistant (mCRPC) disease which clinically tends to progress more rapidly than earlier disease manifestations. The underlying molecular biology of mCRPC is highly complex, and numerous mechanisms have been proposed that promote and retain androgen independence. In various clinical and preclinical data explored, the nature of intracellular signalling pathways mediating mitogenic acquired resistant effects of GPCRs in prostate cancer is poorly defined. G-protein-coupled receptor kinase 2 (GRK2) contributes to the modulation of basic cellular functions-such as cell proliferation, survival or motility-and is involved in metabolic homeostasis, inflammation or angiogenic processes. Moreover, altered GRK2 levels are starting to be reported in different tumoural contexts and shown to promote breast tumourigenesis or to trigger the tumoural angiogenic switch. Thus, we are exploring recent findings that present unexpected opportunities to interfere with major tumourigenic signals by manipulating GPCR-mediated pathways.
Collapse
Affiliation(s)
- Jing-Yan Tian
- Department of Urology, Second Division of The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Chang-Liang Chi
- Department of Urology, Second Division of The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Ge Bian
- Department of Urology, Second Division of The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Feng-Jun Guo
- Department of Gynaecology and Obstetrics, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Xiao-Qing Wang
- Department of Urology, Second Division of The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Bing Yu
- Department of Urology, Second Division of The First Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
23
|
Chung YK, Wong YH. Re‐examining the ‘Dissociation Model’ of G protein activation from the perspective of Gβγ signaling. FEBS J 2020; 288:2490-2501. [DOI: 10.1111/febs.15605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 12/27/2022]
Affiliation(s)
- Yin Kwan Chung
- Division of Life Science and Biotechnology Research Institute Hong Kong University of Science and Technology Hong Kong China
| | - Yung Hou Wong
- Division of Life Science and Biotechnology Research Institute Hong Kong University of Science and Technology Hong Kong China
- State Key Laboratory of Molecular Neuroscience the Molecular Neuroscience Center Hong Kong University of Science and Technology Kowloon China
| |
Collapse
|
24
|
Katan M, Cockcroft S. Phospholipase C families: Common themes and versatility in physiology and pathology. Prog Lipid Res 2020; 80:101065. [PMID: 32966869 DOI: 10.1016/j.plipres.2020.101065] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/14/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022]
Abstract
Phosphoinositide-specific phospholipase Cs (PLCs) are expressed in all mammalian cells and play critical roles in signal transduction. To obtain a comprehensive understanding of these enzymes in physiology and pathology, a detailed structural, biochemical, cell biological and genetic information is required. In this review, we cover all these aspects to summarize current knowledge of the entire superfamily. The families of PLCs have expanded from 13 enzymes to 16 with the identification of the atypical PLCs in the human genome. Recent structural insights highlight the common themes that cover not only the substrate catalysis but also the mechanisms of activation. This involves the release of autoinhibitory interactions that, in the absence of stimulation, maintain classical PLC enzymes in their inactive forms. Studies of individual PLCs provide a rich repertoire of PLC function in different physiologies. Furthermore, the genetic studies discovered numerous mutated and rare variants of PLC enzymes and their link to human disease development, greatly expanding our understanding of their roles in diverse pathologies. Notably, substantial evidence now supports involvement of different PLC isoforms in the development of specific cancer types, immune disorders and neurodegeneration. These advances will stimulate the generation of new drugs that target PLC enzymes, and will therefore open new possibilities for treatment of a number of diseases where current therapies remain ineffective.
Collapse
Affiliation(s)
- Matilda Katan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Shamshad Cockcroft
- Department of Neuroscience, Physiology and Pharmacology, Division of Biosciences, University College London, 21 University Street, London WC1E 6JJ, UK.
| |
Collapse
|
25
|
G proteins: binary switches in health and disease. Cent Eur J Immunol 2020; 45:364-367. [PMID: 33437192 PMCID: PMC7789995 DOI: 10.5114/ceji.2020.101271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 04/29/2018] [Indexed: 12/18/2022] Open
Abstract
Cell signaling plays critical role in health and disease. The normal functioning of body depends on the homeostasis of immunity players. One of the very important cell signaling participants is G protein-coupled receptor (GPCR). GPCRs transduce extracellular signals into target cell by binding to and activating different G proteins (G αβγ, families Gi, Gs, Gq/11, G12/13) leading to range of different functions. Abnormal GPCRs signaling leads to various abnormalities, including but not limited to, cancer, pain, cardiac problems, and asthma. Mutations, which lead to activation or inactivation of GPCR pathways, permanently alter the pathways controlled by these receptors. A large number of human cancer incidence is a consequence of genetic abnormalities in signaling pathways, which influence cell division. Some bacteria and pathogens may interfere with the GPCR signaling pathways for their survival and immune evasion. Inhibition of GPCR signaling by small inhibitors is a novel way to treat various pathological conditions. There are several types of GPCRs in human genome, which due to their central role in health and disease, are the target of many commercially available drugs. Importantly, GPCRs have huge impact on drug discovery and approximately 30% of current drug targets are GPCRs. There is a need of further studies to explore more the role of G protein and the GPCRs in human health and how certain mutations can lead to disease state. Such studies may be important to adjust the signaling pathways for health improvement.
Collapse
|
26
|
Abstract
Heterotrimeric G proteins are the core upstream elements that transduce and amplify the cellular signals from G protein-coupled receptors (GPCRs) to intracellular effectors. GPCRs are the largest family of membrane proteins encoded in the human genome and are the targets of about one-third of prescription medicines. However, to date, no single therapeutic agent exerts its effects via perturbing heterotrimeric G protein function, despite a plethora of evidence linking G protein malfunction to human disease. Several recent studies have brought to light that the Gq family-specific inhibitor FR900359 (FR) is unexpectedly efficacious in silencing the signaling of Gq oncoproteins, mutant Gq variants that mostly exist in the active state. These data not only raise the hope that researchers working in drug discovery may be able to potentially strike Gq oncoproteins from the list of undruggable targets, but also raise questions as to how FR achieves its therapeutic effect. Here, we place emphasis on these recent studies and explain why they expand our pharmacological armamentarium for targeting Gq protein oncogenes as well as broaden our mechanistic understanding of Gq protein oncogene function. We also highlight how this novel insight impacts the significance and utility of using G(q) proteins as targets in drug discovery efforts.
Collapse
Affiliation(s)
- Evi Kostenis
- Section of Molecular, Cellular and Pharmacobiology, Institute of Pharmaceutical Biology, Nussallee 6, 53115 Bonn, Germany.
| | - Eva Marie Pfeil
- Section of Molecular, Cellular and Pharmacobiology, Institute of Pharmaceutical Biology, Nussallee 6, 53115 Bonn, Germany
| | - Suvi Annala
- Section of Molecular, Cellular and Pharmacobiology, Institute of Pharmaceutical Biology, Nussallee 6, 53115 Bonn, Germany
| |
Collapse
|
27
|
Cao Z, Yan L, Shen Z, Chen Y, Shi Y, He X, Zhou N. A novel splice variant of Gαq-coupled Bombyx CAPA-PVK receptor 1 functions as a specific Gαi/o-linked receptor for CAPA-PK. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118718. [PMID: 32289337 DOI: 10.1016/j.bbamcr.2020.118718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 01/04/2023]
Abstract
Alternative splicing enables G protein-coupled receptor (GPCR) genes to greatly increase the number of structurally and functionally distinct receptor isoforms. However, the functional role and relevance of the individual GPCR splice variants in regulating physiological processes are still to be assessed. A naturally occurring alternative splice variant of Bombyx CAPA-PVK receptor, BomCAPA-PVK-R1-Δ341, has been shown to act as a dominant-negative protein to regulate cell surface expression and function of the canonical CAPA-PVK receptor. Herein, using functional assays, we identify the splice variant Δ341 as a specific receptor for neuropeptide CAPA-PK, and upon activation, Δ341 signals to ERK1/2 pathway. Further characterization demonstrates that Δ341 couples to Gαi/o, distinct from the Gαq-coupled canonical CAPA-PVK receptor, triggering ERK1/2 phosphorylation through Gβγ-PI3K-PKCζ signaling cascade. Moreover, our ELISA data show that the ligand-dependent internalization of the splice variant Δ341 is significantly impaired due to lack of GRKs-mediated phosphorylation sites. Our findings highlight the potential of this knowledge for molecular, pharmacological and physiological studies on GPCR splice variants in the future.
Collapse
Affiliation(s)
- Zheng Cao
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lili Yan
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhangfei Shen
- Department of Economic Zoology, College of Animal Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yu Chen
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ying Shi
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaobai He
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
28
|
Lieu M, Traynham CJ, de Lucia C, Pfleger J, Piedepalumbo M, Roy R, Petovic J, Landesberg G, Forrester SJ, Hoffman M, Grisanti LA, Yuan A, Gao E, Drosatos K, Eguchi S, Scalia R, Tilley DG, Koch WJ. Loss of dynamic regulation of G protein-coupled receptor kinase 2 by nitric oxide leads to cardiovascular dysfunction with aging. Am J Physiol Heart Circ Physiol 2020; 318:H1162-H1175. [PMID: 32216616 PMCID: PMC7346533 DOI: 10.1152/ajpheart.00094.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nitric oxide (NO) and S-nitrosothiol (SNO) are considered cardio- and vasoprotective substances. We now understand that one mechanism in which NO/SNOs provide cardiovascular protection is through their direct inhibition of cardiac G protein-coupled receptor (GPCR) kinase 2 (GRK2) activity via S-nitrosylation of GRK2 at cysteine 340 (C340). This maintains GPCR homeostasis, including β-adrenergic receptors, through curbing receptor GRK2-mediated desensitization. Previously, we have developed a knockin mouse (GRK2-C340S) where endogenous GRK2 is resistant to dynamic S-nitrosylation, which led to increased GRK2 desensitizing activity. This unchecked regulation of cardiac GRK2 activity resulted in significantly more myocardial damage after ischemic injury that was resistant to NO-mediated cardioprotection. Although young adult GRK2-C340S mice show no overt phenotype, we now report that as these mice age, they develop significant cardiovascular dysfunction due to the loss of SNO-mediated GRK2 regulation. This pathological phenotype is apparent as early as 12 mo of age and includes reduced cardiac function, increased cardiac perivascular fibrosis, and maladaptive cardiac hypertrophy, which are common maladies found in patients with cardiovascular disease (CVD). There are also vascular reactivity and aortic abnormalities present in these mice. Therefore, our data demonstrate that a chronic and global increase in GRK2 activity is sufficient to cause cardiovascular remodeling and dysfunction, likely due to GRK2’s desensitizing effects in several tissues. Because GRK2 levels have been reported to be elevated in elderly CVD patients, GRK2-C340 mice can give insight into the aged-molecular landscape leading to CVD. NEW & NOTEWORTHY Research on G protein-coupled receptor kinase 2 (GRK2) in the setting of cardiovascular aging is largely unknown despite its strong established functions in cardiovascular physiology and pathophysiology. This study uses a mouse model of chronic GRK2 overactivity to further investigate the consequences of long-term GRK2 on cardiac function and structure. We report for the first time that chronic GRK2 overactivity was able to cause cardiac dysfunction and remodeling independent of surgical intervention, highlighting the importance of GRK activity in aged-related heart disease.
Collapse
Affiliation(s)
- Melissa Lieu
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Christopher J Traynham
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Claudio de Lucia
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Jessica Pfleger
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Michela Piedepalumbo
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania.,Department of Medical, Surgical, Neurological, Metabolic, and Aging Sciences, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Rajika Roy
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Jennifer Petovic
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Gavin Landesberg
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Matthew Hoffman
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Laurel A Grisanti
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania.,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Ancai Yuan
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Erhe Gao
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Konstantinos Drosatos
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Douglas G Tilley
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Busserolles J, Lolignier S, Kerckhove N, Bertin C, Authier N, Eschalier A. Replacement of current opioid drugs focusing on MOR-related strategies. Pharmacol Ther 2020; 210:107519. [PMID: 32165137 DOI: 10.1016/j.pharmthera.2020.107519] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
The scarcity and limited risk/benefit ratio of painkillers available on the market, in addition to the opioid crisis, warrant reflection on new innovation strategies. The pharmacopoeia of analgesics is based on products that are often old and derived from clinical empiricism, with limited efficacy or spectrum of action, or resulting in an unsatisfactory tolerability profile. Although they are reference analgesics for nociceptive pain, opioids are subject to the same criticism. The use of opium as an analgesic is historical. Morphine was synthesized at the beginning of the 19th century. The efficacy of opioids is limited in certain painful contexts and these drugs can induce potentially serious and fatal adverse effects. The current North American opioid crisis, with an ever-rising number of deaths by opioid overdose, is a tragic illustration of this. It is therefore legitimate to develop research into molecules likely to maintain or increase opioid efficacy while improving their tolerability. Several avenues are being explored including targeting of the mu opioid receptor (MOR) splice variants, developing biased agonists or targeting of other receptors such as heteromers with MOR. Ion channels acting as MOR effectors, are also targeted in order to offer compounds without MOR-dependent adverse effects. Another route is to develop opioid analgesics with peripheral action or limited central nervous system (CNS) access. Finally, endogenous opioids used as drugs or compounds that modify the metabolism of endogenous opioids (Dual ENKephalinase Inhibitors) are being developed. The aim of the present review is to present these various targets/strategies with reference to current indications for opioids, concerns about their widespread use, particularly in chronic non-cancer pains, and ways of limiting the risk of opioid abuse and misuse.
Collapse
Affiliation(s)
- Jérôme Busserolles
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Stéphane Lolignier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Nicolas Kerckhove
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Célian Bertin
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Nicolas Authier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
30
|
Lee N, Jae Y, Kim M, Cho T, Lee C, Hong YR, Hyeon DY, Ahn S, Kwon H, Kim K, Jung JH, Chae S, Shin JO, Bok J, Byun Y, Hwang D, Koo J. A pathogen-derived metabolite induces microglial activation via odorant receptors. FEBS J 2020; 287:3841-3870. [PMID: 32003140 DOI: 10.1111/febs.15234] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/10/2019] [Accepted: 01/27/2020] [Indexed: 12/21/2022]
Abstract
Microglia (MG), the principal neuroimmune sentinels in the brain, continuously sense changes in their environment and respond to invading pathogens, toxins, and cellular debris, thereby affecting neuroinflammation. Microbial pathogens produce small metabolites that influence neuroinflammation, but the molecular mechanisms that determine whether pathogen-derived small metabolites affect microglial activation of neuroinflammation remain to be elucidated. We hypothesized that odorant receptors (ORs), the largest subfamily of G protein-coupled receptors, are involved in microglial activation by pathogen-derived small metabolites. We found that MG express high levels of two mouse ORs, Olfr110 and Olfr111, which recognize a pathogenic metabolite, 2-pentylfuran, secreted by Streptococcus pneumoniae. These interactions activate MG to engage in chemotaxis, cytokine production, phagocytosis, and reactive oxygen species generation. These effects were mediated through the Gαs -cyclic adenosine monophosphate-protein kinase A-extracellular signal-regulated kinase and Gβγ -phospholipase C-Ca2+ pathways. Taken together, our results reveal a novel interplay between the pathogen-derived metabolite and ORs, which has major implications for our understanding of microglial activation by pathogen recognition. DATABASE: Model data are available in the PMDB database under the accession number PM0082389.
Collapse
Affiliation(s)
- NaHye Lee
- Department of New Biology, DGIST, Daegu, Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu, Korea
| | - YoonGyu Jae
- Department of New Biology, DGIST, Daegu, Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu, Korea
| | - Minhyung Kim
- Center for Plant Aging Research, DGIST, Daegu, Korea
| | - TaeHo Cho
- Department of New Biology, DGIST, Daegu, Korea
| | - ChaeEun Lee
- Department of New Biology, DGIST, Daegu, Korea
| | - Yu Ri Hong
- Department of New Biology, DGIST, Daegu, Korea
| | | | - Sanghyun Ahn
- Center for Plant Aging Research, DGIST, Daegu, Korea
| | - Hongmok Kwon
- College of Pharmacy, Korea University, Sejong, Korea
| | - Kyul Kim
- College of Pharmacy, Korea University, Sejong, Korea
| | - Jae Hoon Jung
- Center for Plant Aging Research, DGIST, Daegu, Korea
| | - Sehyun Chae
- Center for Plant Aging Research, DGIST, Daegu, Korea
| | - Jeong-Oh Shin
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Youngjoo Byun
- College of Pharmacy, Korea University, Sejong, Korea
| | - Daehee Hwang
- Center for Plant Aging Research, DGIST, Daegu, Korea.,Department of Biological Sciences, Seoul National University, Korea
| | | |
Collapse
|
31
|
Rogers TJ. Bidirectional Regulation of Opioid and Chemokine Function. Front Immunol 2020; 11:94. [PMID: 32076421 PMCID: PMC7006827 DOI: 10.3389/fimmu.2020.00094] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
The opioid family of GPCRs consists of the classical opioid receptors, designated μ-, κ-, and δ-opioid receptors, and the orphanin-FQ receptor, and these proteins are expressed on both neuronal and hematopoietic cells. A number of laboratories have reported that an important degree of cross-talk can occur between the opioid receptors and the chemokine and chemokine receptor families. As a part of this, the opioid receptors are known to regulate the expression of certain chemokines and chemokine receptors, including those that possess strong pro-inflammatory activity. At the level of receptor function, it is clear that certain members of the chemokine family can mediate cross-desensitization of the opioid receptors. Conversely, the opioid receptors are all able to induce heterologous desensitization of some of the chemokine receptors. Consequently, activation of one or more of the opioid receptors can selectively cross-desensitize chemokine receptors and regulate chemokine function. These cross-talk processes have significant implications for the inflammatory response, since the regulation of both the recruitment of inflammatory cells, as well as the sensation of pain, can be controlled in this way.
Collapse
Affiliation(s)
- Thomas J Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
32
|
Li J, Ge Y, Huang JX, Strømgaard K, Zhang X, Xiong XF. Heterotrimeric G Proteins as Therapeutic Targets in Drug Discovery. J Med Chem 2019; 63:5013-5030. [PMID: 31841625 DOI: 10.1021/acs.jmedchem.9b01452] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heterotrimeric G proteins are molecular switches in GPCR signaling pathways and regulate a plethora of physiological and pathological processes. GPCRs are efficient drug targets, and more than 30% of the drugs in use target them. However, selectively targeting an individual GPCR may be undesirable in various multifactorial diseases in which multiple receptors are involved. In addition, abnormal activation or expression of G proteins is frequently associated with diseases. Furthermore, G proteins harboring mutations often result in malignant diseases. Thus, targeting G proteins instead of GPCRs might provide alternative approaches for combating these diseases. In this review, we discuss the biochemistry of heterotrimeric G proteins, describe the G protein-associated diseases, and summarize the currently known modulators that can regulate the activities of G proteins. The outlook for targeting G proteins to treat diverse diseases is also included in this manuscript.
Collapse
Affiliation(s)
- Jian Li
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Yang Ge
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Jun-Xiang Huang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Xiaolei Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| | - Xiao-Feng Xiong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, Guangdong, P. R. China
| |
Collapse
|
33
|
Kunselman JM, Zajac AS, Weinberg ZY, Puthenveedu MA. Homologous Regulation of Mu Opioid Receptor Recycling by G βγ , Protein Kinase C, and Receptor Phosphorylation. Mol Pharmacol 2019; 96:702-710. [PMID: 31575621 PMCID: PMC6820217 DOI: 10.1124/mol.119.117267] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/14/2019] [Indexed: 12/20/2022] Open
Abstract
Membrane trafficking and receptor signaling are two fundamental cellular processes that interact constantly. Although how trafficking regulates signaling is well studied, how signaling pathways regulate trafficking is less well understood. Here, we use the mu opioid receptor (MOR), the primary target for opioid analgesics, to define a signaling pathway that dynamically regulates postendocytic receptor recycling. By directly visualizing individual MOR recycling events, we show that agonist increases MOR recycling. Inhibition of G βγ, phospholipase C, or protein kinase C mimicked agonist removal, whereas activation of G βγ increased recycling even after agonist removal. Phosphorylation of serine 363 on the C-terminal tail of MOR was required and sufficient for agonist-mediated regulation of MOR recycling. Our results identify a feedback loop that regulates MOR recycling via G βγ , protein kinase C, and receptor phosphorylation. This could serve as a general model for how signaling regulates postendocytic trafficking of G protein-coupled receptors. SIGNIFICANCE STATEMENT: G protein-coupled receptor (GPCR) localization in the endosome is being increasingly recognized as an important and distinct component of GPCR signaling and physiology. This study identifies a G protein-dependent and protein kinase C-dependent signaling pathway that dynamically regulates the endosomal localization of the mu opioid receptor, the primary target of opioid analgesics and abused drugs. This pathway could provide a mechanism to manipulate spatial encoding of opioid signaling and physiology.
Collapse
Affiliation(s)
- Jennifer M Kunselman
- Cellular and Molecular Biology Program (J.M.K., M.A.P.) and Department of Pharmacology (J.M.K., Z.Y.W., M.A.P.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania (A.S.Z., M.A.P.)
| | - Amanda S Zajac
- Cellular and Molecular Biology Program (J.M.K., M.A.P.) and Department of Pharmacology (J.M.K., Z.Y.W., M.A.P.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania (A.S.Z., M.A.P.)
| | - Zara Y Weinberg
- Cellular and Molecular Biology Program (J.M.K., M.A.P.) and Department of Pharmacology (J.M.K., Z.Y.W., M.A.P.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania (A.S.Z., M.A.P.)
| | - Manojkumar A Puthenveedu
- Cellular and Molecular Biology Program (J.M.K., M.A.P.) and Department of Pharmacology (J.M.K., Z.Y.W., M.A.P.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania (A.S.Z., M.A.P.)
| |
Collapse
|
34
|
Gaylo-Moynihan A, Prizant H, Popović M, Fernandes NRJ, Anderson CS, Chiou KK, Bell H, Schrock DC, Schumacher J, Capece T, Walling BL, Topham DJ, Miller J, Smrcka AV, Kim M, Hughson A, Fowell DJ. Programming of Distinct Chemokine-Dependent and -Independent Search Strategies for Th1 and Th2 Cells Optimizes Function at Inflamed Sites. Immunity 2019; 51:298-309.e6. [PMID: 31399281 PMCID: PMC6904228 DOI: 10.1016/j.immuni.2019.06.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/18/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023]
Abstract
T-helper (Th) cell differentiation drives specialized gene programs that dictate effector T cell function at sites of infection. Here, we have shown Th cell differentiation also imposes discrete motility gene programs that shape Th1 and Th2 cell navigation of the inflamed dermis. Th1 cells scanned a smaller tissue area in a G protein-coupled receptor (GPCR) and chemokine-dependent fashion, while Th2 cells scanned a larger tissue area independent of GPCR signals. Differential chemokine reliance for interstitial migration was linked to STAT6 transcription-factor-dependent programming of integrin αVβ3 expression: Th2 cell differentiation led to high αVβ3 expression relative to Th1 cells. Th1 and Th2 cell modes of motility could be switched simply by manipulating the amount of αVβ3 on the cell surface. Deviating motility modes from those established during differentiation impaired effector function. Thus, programmed expression of αVβ3 tunes effector T cell reliance on environmental cues for optimal exploration of inflamed tissues.
Collapse
Affiliation(s)
- Alison Gaylo-Moynihan
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hen Prizant
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Milan Popović
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ninoshka R J Fernandes
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Christopher S Anderson
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Kevin K Chiou
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hannah Bell
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Dillon C Schrock
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Justin Schumacher
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Tara Capece
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Brandon L Walling
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jim Miller
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Alan V Smrcka
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Minsoo Kim
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Angela Hughson
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Deborah J Fowell
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
35
|
Stott JB, Barrese V, Suresh M, Masoodi S, Greenwood IA. Investigating the Role of G Protein βγ in Kv7-Dependent Relaxations of the Rat Vasculature. Arterioscler Thromb Vasc Biol 2019; 38:2091-2102. [PMID: 30002060 DOI: 10.1161/atvbaha.118.311360] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Objective- In renal arteries, inhibitors of G protein βγ subunits (Gβγ) reduce Kv7 activity and inhibit Kv7-dependent receptor-mediated vasorelaxations. However, the mechanisms underlying receptor-mediated relaxation are artery specific. Consequently, the aim of this study was to ascertain the role of Gβγ in Kv7-dependent vasorelaxations of the rat vasculature. Approach and Results- Isometric tension recording was performed in isolated rat renal, mesenteric, and cerebral arteries to study isoproterenol and calcitonin gene-related peptide relaxations. Kv7.4 was knocked down via morpholino transfection while inhibition of Gβγ was investigated with gallein and M119K. Proximity ligation assay was performed on isolated myocytes to study the association between Kv7.4 and G protein β subunits or signaling intermediaries. Isoproterenol or calcitonin gene-related peptide-induced relaxations were attenuated by Kv7.4 knockdown in all arteries studied. Inhibition of Gβγ with gallein or M119K had no effect on isoproterenol-mediated relaxations in mesenteric artery but had a marked effect on calcitonin gene-related peptide-induced responses in mesenteric artery and cerebral artery and isoproterenol responses in renal artery. Isoproterenol increased association with Kv7.4 and Rap1a in mesenteric artery which were not sensitive to gallein, whereas in renal artery, isoproterenol increased Kv7.4-AKAP (A-kinase anchoring protein) associations in a gallein-sensitive manner. Conclusions- The Gβγ-Kv7 relationship differs between vessels and is an essential requirement for AKAP, but not Rap-mediated regulation of the channel.
Collapse
Affiliation(s)
- Jennifer B Stott
- From the Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| | - Vincenzo Barrese
- From the Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| | - Malavika Suresh
- From the Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| | - Shirou Masoodi
- From the Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| | - Iain A Greenwood
- From the Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| |
Collapse
|
36
|
Abstract
G protein-coupled receptors (GPCRs) are critical cellular sensors that mediate numerous physiological processes. In the heart, multiple GPCRs are expressed on various cell types, where they coordinate to regulate cardiac function by modulating critical processes such as contractility and blood flow. Under pathological settings, these receptors undergo aberrant changes in expression levels, localization and capacity to couple to downstream signalling pathways. Conventional therapies for heart failure work by targeting GPCRs, such as β-adrenergic receptor and angiotensin II receptor antagonists. Although these treatments have improved patient survival, heart failure remains one of the leading causes of mortality worldwide. GPCR kinases (GRKs) are responsible for GPCR phosphorylation and, therefore, desensitization and downregulation of GPCRs. In this Review, we discuss the GPCR signalling pathways and the GRKs involved in the pathophysiology of heart disease. Given that increased expression and activity of GRK2 and GRK5 contribute to the loss of contractile reserve in the stressed and failing heart, inhibition of overactive GRKs has been proposed as a novel therapeutic approach to treat heart failure.
Collapse
|
37
|
Emery MA, Eitan S. Members of the same pharmacological family are not alike: Different opioids, different consequences, hope for the opioid crisis? Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:428-449. [PMID: 30790677 DOI: 10.1016/j.pnpbp.2019.02.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 01/14/2023]
Abstract
Pain management is the specialized medical practice of modulating pain perception and thus easing the suffering and improving the life quality of individuals suffering from painful conditions. Since this requires the modulation of the activity of endogenous systems involved in pain perception, and given the large role that the opioidergic system plays in pain perception, opioids are currently the most effective pain treatment available and are likely to remain relevant for the foreseeable future. This contributes to the rise in opioid use, misuse, and overdose death, which is currently characterized by public health officials in the United States as an epidemic. Historically, the majority of preclinical rodent studies were focused on morphine. This has resulted in our understanding of opioids in general being highly biased by our knowledge of morphine specifically. However, recent in vitro studies suggest that direct extrapolation of research findings from morphine to other opioids is likely to be flawed. Notably, these studies suggest that different opioid analgesics (opioid agonists) engage different downstream signaling effects within the cell, despite binding to and activating the same receptors. This recognition implies that, in contrast to the historical status quo, different opioids cannot be made equivalent by merely dose adjustment. Notably, even at equianalgesic doses, different opioids could result in different beneficial and risk outcomes. In order to foster further translational research regarding drug-specific differences among opioids, here we review basic research elucidating differences among opioids in pharmacokinetics, pharmacodynamics, their capacity for second messenger pathway activation, and their interactions with the immune system and the dopamine D2 receptors.
Collapse
Affiliation(s)
- Michael A Emery
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA; Interdisciplinary Program in Neuroscience, Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA; Interdisciplinary Program in Neuroscience, Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, USA.
| |
Collapse
|
38
|
Zhao P, Pattison LA, Jensen DD, Jimenez-Vargas NN, Latorre R, Lieu T, Jaramillo JO, Lopez-Lopez C, Poole DP, Vanner SJ, Schmidt BL, Bunnett NW. Protein kinase D and Gβγ mediate sustained nociceptive signaling by biased agonists of protease-activated receptor-2. J Biol Chem 2019; 294:10649-10662. [PMID: 31142616 DOI: 10.1074/jbc.ra118.006935] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Proteases sustain hyperexcitability and pain by cleaving protease-activated receptor-2 (PAR2) on nociceptors through distinct mechanisms. Whereas trypsin induces PAR2 coupling to Gαq, Gαs, and β-arrestins, cathepsin-S (CS) and neutrophil elastase (NE) cleave PAR2 at distinct sites and activate it by biased mechanisms that induce coupling to Gαs, but not to Gαq or β-arrestins. Because proteases activate PAR2 by irreversible cleavage, and activated PAR2 is degraded in lysosomes, sustained extracellular protease-mediated signaling requires mobilization of intact PAR2 from the Golgi apparatus or de novo synthesis of new receptors by incompletely understood mechanisms. We found here that trypsin, CS, and NE stimulate PAR2-dependent activation of protein kinase D (PKD) in the Golgi of HEK293 cells, in which PKD regulates protein trafficking. The proteases stimulated translocation of the PKD activator Gβγ to the Golgi, coinciding with PAR2 mobilization from the Golgi. Proteases also induced translocation of a photoconverted PAR2-Kaede fusion protein from the Golgi to the plasma membrane of KNRK cells. After incubation of HEK293 cells and dorsal root ganglia neurons with CS, NE, or trypsin, PAR2 responsiveness initially declined, consistent with PAR2 cleavage and desensitization, and then gradually recovered. Inhibitors of PKD, Gβγ, and protein translation inhibited recovery of PAR2 responsiveness. PKD and Gβγ inhibitors also attenuated protease-evoked mechanical allodynia in mice. We conclude that proteases that activate PAR2 by canonical and biased mechanisms stimulate PKD in the Golgi; PAR2 mobilization and de novo synthesis repopulate the cell surface with intact receptors and sustain nociceptive signaling by extracellular proteases.
Collapse
Affiliation(s)
- Peishen Zhao
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Luke A Pattison
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Dane D Jensen
- the Departments of Surgery and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Nestor N Jimenez-Vargas
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Rocco Latorre
- the Departments of Surgery and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - TinaMarie Lieu
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Josue O Jaramillo
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Cintya Lopez-Lopez
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Daniel P Poole
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Stephen J Vanner
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Brian L Schmidt
- the Bluestone Center for Clinical Research, New York University College of Dentistry, New York, New York 10010
| | - Nigel W Bunnett
- the Departments of Surgery and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032,
| |
Collapse
|
39
|
Seyedabadi M, Ghahremani MH, Albert PR. Biased signaling of G protein coupled receptors (GPCRs): Molecular determinants of GPCR/transducer selectivity and therapeutic potential. Pharmacol Ther 2019; 200:148-178. [PMID: 31075355 DOI: 10.1016/j.pharmthera.2019.05.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Abstract
G protein coupled receptors (GPCRs) convey signals across membranes via interaction with G proteins. Originally, an individual GPCR was thought to signal through one G protein family, comprising cognate G proteins that mediate canonical receptor signaling. However, several deviations from canonical signaling pathways for GPCRs have been described. It is now clear that GPCRs can engage with multiple G proteins and the line between cognate and non-cognate signaling is increasingly blurred. Furthermore, GPCRs couple to non-G protein transducers, including β-arrestins or other scaffold proteins, to initiate additional signaling cascades. Receptor/transducer selectivity is dictated by agonist-induced receptor conformations as well as by collateral factors. In particular, ligands stabilize distinct receptor conformations to preferentially activate certain pathways, designated 'biased signaling'. In this regard, receptor sequence alignment and mutagenesis have helped to identify key receptor domains for receptor/transducer specificity. Furthermore, molecular structures of GPCRs bound to different ligands or transducers have provided detailed insights into mechanisms of coupling selectivity. However, receptor dimerization, compartmentalization, and trafficking, receptor-transducer-effector stoichiometry, and ligand residence and exposure times can each affect GPCR coupling. Extrinsic factors including cell type or assay conditions can also influence receptor signaling. Understanding these factors may lead to the development of improved biased ligands with the potential to enhance therapeutic benefit, while minimizing adverse effects. In this review, evidence for ligand-specific GPCR signaling toward different transducers or pathways is elaborated. Furthermore, molecular determinants of biased signaling toward these pathways and relevant examples of the potential clinical benefits and pitfalls of biased ligands are discussed.
Collapse
Affiliation(s)
- Mohammad Seyedabadi
- Department of Pharmacology, School of Medicine, Bushehr University of Medical Sciences, Iran; Education Development Center, Bushehr University of Medical Sciences, Iran
| | | | - Paul R Albert
- Ottawa Hospital Research Institute, Neuroscience, University of Ottawa, Canada.
| |
Collapse
|
40
|
Dela Paz NG, Frangos JA. Rapid flow-induced activation of Gα q/11 is independent of Piezo1 activation. Am J Physiol Cell Physiol 2019; 316:C741-C752. [PMID: 30811222 PMCID: PMC6580164 DOI: 10.1152/ajpcell.00215.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 02/08/2019] [Accepted: 02/25/2019] [Indexed: 12/22/2022]
Abstract
Endothelial cell (EC) mechanochemical transduction is the process by which mechanical stimuli are sensed by ECs and transduced into biochemical signals and ultimately into physiological responses. Identifying the mechanosensor/mechanochemical transducer(s) and describing the mechanism(s) by which they receive and transmit the signals has remained a central focus within the field. The heterotrimeric G protein, Gαq/11, is proposed to be part of a macromolecular complex together with PECAM-1 at EC junctions and may constitute the mechanochemical transducer as it is rapidly activated within seconds of flow onset. The mechanically activated cation channel Piezo1 has recently been implicated due to its involvement in mediating early responses, such as calcium and ATP release. Here, we investigate the role of Piezo1 in rapid shear stress-induced Gαq/11 activation. We show that flow-induced dissociation of Gαq/11 from PECAM-1 in ECs at 15 s is abrogated by BIM-46187, a selective inhibitor of Gαq/11 activation, suggesting that Gαq/11 activation is required for PECAM-1/Gαq/11 dissociation. Although siRNA knockdown of Piezo1 caused a dramatic decrease in PECAM-1/Gαq/11 association in the basal condition, it had no effect on flow-induced dissociation. Interestingly, siRNA knockdown of Piezo1 caused a marked decrease in PECAM-1 expression. Additionally, selective blockade of Piezo1 with ion channel inhibitors had no effect on flow-induced PECAM-1/Gαq/11 dissociations. Lastly, flow onset caused increased association of Gβ1 with Piezo1 as well as with the p101 subunit of phosphoinositide 3-kinase, which were both blocked by the Gβγ inhibitor gallein. Together, our results indicate that flow-induced activation of Piezo1 is not upstream of G protein activation.
Collapse
Affiliation(s)
| | - John A Frangos
- La Jolla Bioengineering Institute , La Jolla, California
| |
Collapse
|
41
|
Marivin A, Morozova V, Walawalkar I, Leyme A, Kretov DA, Cifuentes D, Dominguez I, Garcia-Marcos M. GPCR-independent activation of G proteins promotes apical cell constriction in vivo. J Cell Biol 2019; 218:1743-1763. [PMID: 30948426 PMCID: PMC6504902 DOI: 10.1083/jcb.201811174] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/19/2019] [Accepted: 03/12/2019] [Indexed: 01/21/2023] Open
Abstract
Heterotrimeric G proteins are signaling switches that control organismal morphogenesis across metazoans. In invertebrates, specific GPCRs instruct G proteins to promote collective apical cell constriction in the context of epithelial tissue morphogenesis. In contrast, tissue-specific factors that instruct G proteins during analogous processes in vertebrates are largely unknown. Here, we show that DAPLE, a non-GPCR protein linked to human neurodevelopmental disorders, is expressed specifically in the neural plate of Xenopus laevis embryos to trigger a G protein signaling pathway that promotes apical cell constriction during neurulation. DAPLE localizes to apical cell-cell junctions in the neuroepithelium, where it activates G protein signaling to drive actomyosin-dependent apical constriction and subsequent bending of the neural plate. This function is mediated by a Gα-binding-and-activating (GBA) motif that was acquired by DAPLE in vertebrates during evolution. These findings reveal that regulation of tissue remodeling during vertebrate development can be driven by an unconventional mechanism of heterotrimeric G protein activation that operates in lieu of GPCRs.
Collapse
Affiliation(s)
- Arthur Marivin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Veronika Morozova
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Isha Walawalkar
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Anthony Leyme
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Dmitry A Kretov
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Daniel Cifuentes
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Isabel Dominguez
- Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Mikel Garcia-Marcos
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| |
Collapse
|
42
|
Murga C, Arcones AC, Cruces-Sande M, Briones AM, Salaices M, Mayor F. G Protein-Coupled Receptor Kinase 2 (GRK2) as a Potential Therapeutic Target in Cardiovascular and Metabolic Diseases. Front Pharmacol 2019; 10:112. [PMID: 30837878 PMCID: PMC6390810 DOI: 10.3389/fphar.2019.00112] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptor kinase 2 (GRK2) is a central signaling node involved in the modulation of many G protein-coupled receptors (GPCRs) and also displaying regulatory functions in other cell signaling routes. GRK2 levels and activity have been reported to be enhanced in patients or in preclinical models of several relevant pathological situations, such as heart failure, cardiac hypertrophy, hypertension, obesity and insulin resistance conditions, or non-alcoholic fatty liver disease (NAFLD), and to contribute to disease progression by a variety of mechanisms related to its multifunctional roles. Therefore, targeting GRK2 by different strategies emerges as a potentially relevant approach to treat cardiovascular disease, obesity, type 2 diabetes, or NAFLD, pathological conditions which are frequently interconnected and present as co-morbidities.
Collapse
Affiliation(s)
- Cristina Murga
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Alba C Arcones
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Marta Cruces-Sande
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Ana M Briones
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Mercedes Salaices
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Federico Mayor
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| |
Collapse
|
43
|
Malfacini D, Patt J, Annala S, Harpsøe K, Eryilmaz F, Reher R, Crüsemann M, Hanke W, Zhang H, Tietze D, Gloriam DE, Bräuner-Osborne H, Strømgaard K, König GM, Inoue A, Gomeza J, Kostenis E. Rational design of a heterotrimeric G protein α subunit with artificial inhibitor sensitivity. J Biol Chem 2019; 294:5747-5758. [PMID: 30745359 PMCID: PMC6463727 DOI: 10.1074/jbc.ra118.007250] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/28/2019] [Indexed: 12/31/2022] Open
Abstract
Transmembrane signals initiated by a range of extracellular stimuli converge on members of the Gq family of heterotrimeric G proteins, which relay these signals in target cells. Gq family G proteins comprise Gq, G11, G14, and G16, which upon activation mediate their cellular effects via inositol lipid–dependent and –independent signaling to control fundamental processes in mammalian physiology. To date, highly specific inhibition of Gq/11/14 signaling can be achieved only with FR900359 (FR) and YM-254890 (YM), two naturally occurring cyclic depsipeptides. To further development of FR or YM mimics for other Gα subunits, we here set out to rationally design Gα16 proteins with artificial FR/YM sensitivity by introducing an engineered depsipeptide-binding site. Thereby we permit control of G16 function through ligands that are inactive on the WT protein. Using CRISPR/Cas9-generated Gαq/Gα11-null cells and loss- and gain-of-function mutagenesis along with label-free whole-cell biosensing, we determined the molecular coordinates for FR/YM inhibition of Gq and transplanted these to FR/YM-insensitive G16. Intriguingly, despite having close structural similarity, FR and YM yielded biologically distinct activities: it was more difficult to perturb Gq inhibition by FR and easier to install FR inhibition onto G16 than perturb or install inhibition with YM. A unique hydrophobic network utilized by FR accounted for these unexpected discrepancies. Our results suggest that non-Gq/11/14 proteins should be amenable to inhibition by FR scaffold–based inhibitors, provided that these inhibitors mimic the interaction of FR with Gα proteins harboring engineered FR-binding sites.
Collapse
Affiliation(s)
- Davide Malfacini
- From the Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Julian Patt
- From the Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Suvi Annala
- From the Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Kasper Harpsøe
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Funda Eryilmaz
- From the Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Raphael Reher
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Max Crüsemann
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Wiebke Hanke
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Hang Zhang
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Daniel Tietze
- Eduard Zintl Institute of Inorganic and Physical Chemistry, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Jesus Gomeza
- From the Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Evi Kostenis
- From the Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany.
| |
Collapse
|
44
|
Pierce ML, Mehrotra S, Mustoe AC, French JA, Murray TF. A Comparison of the Ability of Leu 8- and Pro 8-Oxytocin to Regulate Intracellular Ca 2+ and Ca 2+-Activated K + Channels at Human and Marmoset Oxytocin Receptors. Mol Pharmacol 2019; 95:376-385. [PMID: 30739093 DOI: 10.1124/mol.118.114744] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/30/2019] [Indexed: 02/02/2023] Open
Abstract
The neurohypophyseal hormone oxytocin (OT) regulates biologic functions in both peripheral tissues and the central nervous system. In the central nervous system, OT influences social processes, including peer relationships, maternal-infant bonding, and affiliative social relationships. In mammals, the nonapeptide OT structure is highly conserved with leucine in the eighth position (Leu8-OT). In marmosets (Callithrix), a nonsynonymous nucleotide substitution in the OXT gene codes for proline in the eighth residue position (Pro8-OT). OT binds to its cognate G protein-coupled receptor (OTR) and exerts diverse effects, including stimulation (Gs) or inhibition (Gi/o) of adenylyl cyclase, stimulation of potassium channel currents (Gi), and activation of phospholipase C (Gq). Chinese hamster ovary cells expressing marmoset or human oxytocin receptors (mOTRs or hOTRs, respectively) were used to characterize OT signaling. At the mOTR, Pro8-OT was more efficacious than Leu8-OT in measures of Gq activation, with both peptides displaying subnanomolar potencies. At the hOTR, neither the potency nor efficacy of Pro8-OT and Leu8-OT differed with respect to Gq signaling. In both mOTR- and hOTR-expressing cells, Leu8-OT was more potent and modestly more efficacious than Pro8-OT in inducing hyperpolarization. In mOTR cells, Leu8-OT-induced hyperpolarization was modestly inhibited by pretreatment with pertussis toxin (PTX), consistent with a minor role for Gi/o activation; however, the Pro8-OT response in mOTR and hOTR cells was PTX insensitive. These findings are consistent with membrane hyperpolarization being largely mediated by a Gq signaling mechanism leading to Ca2+-dependent activation of K+ channels. Evaluation of the influence of apamin, charybdotoxin, paxilline, and TRAM-34 demonstrated involvement of both intermediate and large conductance Ca2+-activated K+ channels.
Collapse
Affiliation(s)
- Marsha L Pierce
- Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska (M.L.P., S.M., T.F.M.); and Department of Psychology, University of Nebraska, Omaha, Nebraska (A.C.M., J.A.F.)
| | - Suneet Mehrotra
- Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska (M.L.P., S.M., T.F.M.); and Department of Psychology, University of Nebraska, Omaha, Nebraska (A.C.M., J.A.F.)
| | - Aaryn C Mustoe
- Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska (M.L.P., S.M., T.F.M.); and Department of Psychology, University of Nebraska, Omaha, Nebraska (A.C.M., J.A.F.)
| | - Jeffrey A French
- Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska (M.L.P., S.M., T.F.M.); and Department of Psychology, University of Nebraska, Omaha, Nebraska (A.C.M., J.A.F.)
| | - Thomas F Murray
- Department of Pharmacology, Creighton University School of Medicine, Omaha, Nebraska (M.L.P., S.M., T.F.M.); and Department of Psychology, University of Nebraska, Omaha, Nebraska (A.C.M., J.A.F.)
| |
Collapse
|
45
|
Druggable Targets in Cyclic Nucleotide Signaling Pathways in Apicomplexan Parasites and Kinetoplastids against Disabling Protozoan Diseases in Humans. Int J Mol Sci 2019; 20:ijms20010138. [PMID: 30609697 PMCID: PMC6337498 DOI: 10.3390/ijms20010138] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 12/19/2018] [Accepted: 12/24/2018] [Indexed: 12/20/2022] Open
Abstract
Cell signaling in eukaryotes is an evolutionarily conserved mechanism to respond and adapt to various environmental changes. In general, signal sensation is mediated by a receptor which transfers the signal to a cascade of effector proteins. The cyclic nucleotides 3′,5′-cyclic adenosine monophosphate (cAMP) and 3′,5′-cyclic guanosine monophosphate (cGMP) are intracellular messengers mediating an extracellular stimulus to cyclic nucleotide-dependent kinases driving a change in cell function. In apicomplexan parasites and kinetoplastids, which are responsible for a variety of neglected, tropical diseases, unique mechanisms of cyclic nucleotide signaling are currently identified. Collectively, cyclic nucleotides seem to be essential for parasitic proliferation and differentiation. However, there is no a genomic evidence for canonical G-proteins in these parasites while small GTPases and secondary effector proteins with structural differences to host orthologues occur. Database entries encoding G-protein-coupled receptors (GPCRs) are still without functional proof. Instead, signals from the parasite trigger GPCR-mediated signaling in the host during parasite invasion and egress. The role of cyclic nucleotide signaling in the absence of G-proteins and GPCRs, with a particular focus on small GTPases in pathogenesis, is reviewed here. Due to the absence of G-proteins, apicomplexan parasites and kinetoplastids may use small GTPases or their secondary effector proteins and host canonical G-proteins during infection. Thus, the feasibility of targeting cyclic nucleotide signaling pathways in these parasites, will be an enormous challenge for the identification of selective, pharmacological inhibitors since canonical host proteins also contribute to pathogenesis.
Collapse
|
46
|
Abstract
The nociceptin/orphanin FQ (N/OFQ) peptide receptor (NOP) is a G protein-coupled receptor involved in the regulation of several physiological functions and pathological conditions. Thus, researchers from academia and industry are pursuing NOP to discover and study novel pharmacological entities. In a multidisciplinary effort of pharmacologists, medicinal chemists, and molecular and structural biologists the mechanisms of NOP activation and inhibition have been, at least partially, disentangled. Here, we review the in vitro methodologies employed, which have contributed to our understanding of this target. We hope this chapter guides the reader through the mostly established assay platforms to investigate NOP pharmacology, and gives some hints taking advantage from what has already illuminated the function of other GPCRs. We analyzed the pharmacological results obtained with a large panel of NOP ligands investigated in several assays including receptor binding, stimulation of GTPγS binding, decrease of cAMP levels, calcium flux stimulation via chimeric G proteins, NOP/G protein and NOP/β-arrestin interaction, label-free assays such as dynamic mass redistribution, and bioassays such as the electrically stimulated mouse vas deferens.
Collapse
Affiliation(s)
- Davide Malfacini
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Girolamo Caló
- Section of Pharmacology, Department of Medical Sciences, National Institute of Neurosciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
47
|
Porcu A, Melis M, Turecek R, Ullrich C, Mocci I, Bettler B, Gessa GL, Castelli MP. Rimonabant, a potent CB1 cannabinoid receptor antagonist, is a Gα i/o protein inhibitor. Neuropharmacology 2018; 133:107-120. [PMID: 29407764 DOI: 10.1016/j.neuropharm.2018.01.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 01/10/2018] [Accepted: 01/18/2018] [Indexed: 12/25/2022]
Abstract
Rimonabant is a potent and selective cannabinoid CB1 receptor antagonist widely used in animal and clinical studies. Besides its antagonistic properties, numerous studies have shown that, at micromolar concentrations rimonabant behaves as an inverse agonist at CB1 receptors. The mechanism underpinning this activity is unclear. Here we show that micromolar concentrations of rimonabant inhibited Gαi/o-type G proteins, resulting in a receptor-independent block of G protein signaling. Accordingly, rimonabant decreased basal and agonist stimulated [35S]GTPγS binding to cortical membranes of CB1- and GABAB-receptor KO mice and Chinese Hamster Ovary (CHO) cell membranes stably transfected with GABAB or D2 dopamine receptors. The structural analog of rimonabant, AM251, decreased basal and baclofen-stimulated GTPγS binding to rat cortical and CHO cell membranes expressing GABAB receptors. Rimonabant prevented G protein-mediated GABAB and D2 dopamine receptor signaling to adenylyl cyclase in Human Embryonic Kidney 293 cells and to G protein-coupled inwardly rectifying K+ channels (GIRK) in midbrain dopamine neurons of CB1 KO mice. Rimonabant suppressed GIRK gating induced by GTPγS in CHO cells transfected with GIRK, consistent with a receptor-independent action. Bioluminescent resonance energy transfer (BRET) measurements in living CHO cells showed that, in presence or absence of co-expressed GABAB receptors, rimonabant stabilized the heterotrimeric Gαi/o-protein complex and prevented conformational rearrangements induced by GABAB receptor activation. Rimonabant failed to inhibit Gαs-mediated signaling, supporting its specificity for Gαi/o-type G proteins. The inhibition of Gαi/o protein provides a new site of rimonabant action that may help to understand its pharmacological and toxicological effects occurring at high concentrations.
Collapse
Affiliation(s)
- Alessandra Porcu
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy; Department of Biomedicine, University of Basel, Klingelbergstrasse 50-70, CH-4056, Basel, Switzerland
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - Rostislav Turecek
- Department of Biomedicine, University of Basel, Klingelbergstrasse 50-70, CH-4056, Basel, Switzerland
| | - Celine Ullrich
- Department of Biomedicine, University of Basel, Klingelbergstrasse 50-70, CH-4056, Basel, Switzerland
| | - Ignazia Mocci
- Institute of Translational Pharmacology, National Research Council of Italy (CNR) U.O.S. of Cagliari, 09010, Pula, Italy
| | - Bernhard Bettler
- Department of Biomedicine, University of Basel, Klingelbergstrasse 50-70, CH-4056, Basel, Switzerland
| | - Gian Luigi Gessa
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy; Guy Everett Laboratory, University of Cagliari, 09042, Monserrato, Italy; Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy; Center of Excellence "Neurobiology of Addiction", University of Cagliari, 09042, Monserrato, Italy
| | - M Paola Castelli
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy; Center of Excellence "Neurobiology of Addiction", University of Cagliari, 09042, Monserrato, Italy.
| |
Collapse
|
48
|
Translating biased signaling in the ghrelin receptor system into differential in vivo functions. Proc Natl Acad Sci U S A 2018; 115:E10255-E10264. [PMID: 30301804 DOI: 10.1073/pnas.1804003115] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Biased signaling has been suggested as a means of selectively modulating a limited fraction of the signaling pathways for G-protein-coupled receptor family members. Hence, biased ligands may allow modulation of only the desired physiological functions and not elicit undesired effects associated with pharmacological treatments. The ghrelin receptor is a highly sought antiobesity target, since the gut hormone ghrelin in humans has been shown to increase both food intake and fat accumulation. However, it also modulates mood, behavior, growth hormone secretion, and gastric motility. Thus, blocking all pathways of this receptor may give rise to potential side effects. In the present study, we describe a highly promiscuous signaling capacity for the ghrelin receptor. We tested selected ligands for their ability to regulate the various pathways engaged by the receptor. Among those, a biased ligand, YIL781, was found to activate the Gαq/11 and Gα12 pathways selectively without affecting the engagement of β-arrestin or other G proteins. YIL781 was further characterized for its in vivo physiological functions. In combination with the use of mice in which Gαq/11 was selectively deleted in the appetite-regulating AgRP neurons, this biased ligand allowed us to demonstrate that selective blockade of Gαq/11, without antagonism at β-arrestin or other G-protein coupling is sufficient to decrease food intake.
Collapse
|
49
|
Birchall K, Merritt A, Sattikar A, Kettleborough C, Saxty B. Design of the LifeArc Index Set and Retrospective Review of Its Performance: A Collection for Sharing. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2018; 24:332-345. [PMID: 30290126 DOI: 10.1177/2472555218803696] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Building, curating, and maintaining a compound collection is an expensive operation, beyond the scope of most academic organizations. Here we describe the selection criteria used to compile the LifeArc diversity set from commercial suppliers and the process we undertook to generate our representative LifeArc index set. The aim was to avoid a "junk in, junk out" screen collection to increase chemical tractability going forward, while maximizing diversity. Using historical LifeArc screening data, we demonstrate that the index set was predictive of ligandability and that progressable hits could be identified by mining associated clusters within our larger diversity set. Indeed, a higher percentage of index-derived hit clusters were found to have been progressed into hit-to-lead programs, reflecting better drug-likeness. In practice, the library has been shared widely with academic groups and used routinely within LifeArc to assess the ligandability of novel targets. Its small size is well suited to meet the needs of medium-throughput screening in labs with either limited automation, limited precious or expensive reagents, or complex cellular assays. The strategy of screening a small set in combination with rapid hit analog follow-up has demonstrated the utility of finding active clusters for potential development against challenging targets.
Collapse
Affiliation(s)
| | - Andy Merritt
- 1 LifeArc, Centre for Therapeutic Discovery, Stevenage, UK
| | - Afrah Sattikar
- 1 LifeArc, Centre for Therapeutic Discovery, Stevenage, UK
| | | | - Barbara Saxty
- 1 LifeArc, Centre for Therapeutic Discovery, Stevenage, UK
| |
Collapse
|
50
|
Abstract
G protein-coupled receptors (GPCRs) are the largest class of drug targets, largely owing to their druggability, diversity and physiological efficacy. Many drugs selectively target specific subtypes of GPCRs, but high specificity for individual GPCRs may not be desirable in complex multifactorial disease states in which multiple receptors may be involved. One approach is to target G protein subunits rather than the GPCRs directly. This approach has the potential to achieve broad efficacy by blocking pathways shared by multiple GPCRs. Additionally, because many GPCRs couple to multiple G protein signalling pathways, blocking specific G protein subunits can 'bias' GPCR signals by inhibiting only a subset of these signals. Molecules that target G protein α or βγ-subunits have been developed and show strong efficacy in multiple preclinical disease models and biased inhibition of G protein signalling. In this Review, we discuss the development and characterization of G protein α and βγ-subunit ligands and the preclinical evidence that this exciting new approach has potential for therapeutic efficacy in a number of indications, such as pain, thrombosis, asthma and heart failure.
Collapse
|