1
|
Cilek N, Ugurel E, Goksel E, Yalcin O. Signaling mechanisms in red blood cells: A view through the protein phosphorylation and deformability. J Cell Physiol 2024; 239:e30958. [PMID: 36748950 DOI: 10.1002/jcp.30958] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 01/07/2023] [Accepted: 01/19/2023] [Indexed: 02/08/2023]
Abstract
Intracellular signaling mechanisms in red blood cells (RBCs) involve various protein kinases and phosphatases and enable rapid adaptive responses to hypoxia, metabolic requirements, oxidative stress, or shear stress by regulating the physiological properties of the cell. Protein phosphorylation is a ubiquitous mechanism for intracellular signal transduction, volume regulation, and cytoskeletal organization in RBCs. Spectrin-based cytoskeleton connects integral membrane proteins, band 3 and glycophorin C to junctional proteins, ankyrin and Protein 4.1. Phosphorylation leads to a conformational change in the protein structure, weakening the interactions between proteins in the cytoskeletal network that confers a more flexible nature for the RBC membrane. The structural organization of the membrane and the cytoskeleton determines RBC deformability that allows cells to change their ability to deform under shear stress to pass through narrow capillaries. The shear stress sensing mechanisms and oxygenation-deoxygenation transitions regulate cell volume and mechanical properties of the membrane through the activation of ion transporters and specific phosphorylation events mediated by signal transduction. In this review, we summarize the roles of Protein kinase C, cAMP-Protein kinase A, cGMP-nitric oxide, RhoGTPase, and MAP/ERK pathways in the modulation of RBC deformability in both healthy and disease states. We emphasize that targeting signaling elements may be a therapeutic strategy for the treatment of hemoglobinopathies or channelopathies. We expect the present review will provide additional insights into RBC responses to shear stress and hypoxia via signaling mechanisms and shed light on the current and novel treatment options for pathophysiological conditions.
Collapse
Affiliation(s)
- Neslihan Cilek
- Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
- School of Medicine, Koc University, Istanbul, Turkey
- Graduate School of Health Sciences, Koc University, Istanbul, Turkey
| | - Elif Ugurel
- Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
- School of Medicine, Koc University, Istanbul, Turkey
| | - Evrim Goksel
- Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
- School of Medicine, Koc University, Istanbul, Turkey
- Graduate School of Health Sciences, Koc University, Istanbul, Turkey
| | - Ozlem Yalcin
- Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
- School of Medicine, Koc University, Istanbul, Turkey
| |
Collapse
|
2
|
Brosens N, Lesuis SL, Rao-Ruiz P, van den Oever MC, Krugers HJ. Shaping Memories Via Stress: A Synaptic Engram Perspective. Biol Psychiatry 2023:S0006-3223(23)01720-1. [PMID: 37977215 DOI: 10.1016/j.biopsych.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 10/09/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Stress modulates the activity of various memory systems and can thereby guide behavioral interaction with the environment in an adaptive or maladaptive manner. At the cellular level, a large body of evidence indicates that (nor)adrenaline and glucocorticoid release induced by acute stress exposure affects synapse function and synaptic plasticity, which are critical substrates for learning and memory. Recent evidence suggests that memories are supported in the brain by sparsely distributed neurons within networks, termed engram cell ensembles. While the physiological and molecular effects of stress on the synapse are increasingly well characterized, how these synaptic modifications shape the multiscale dynamics of engram cell ensembles is still poorly understood. In this review, we discuss and integrate recent information on how acute stress affects synapse function and how this may alter engram cell ensembles and their synaptic connectivity to shape memory strength and memory precision. We provide a mechanistic framework of a synaptic engram under stress and put forward outstanding questions that address knowledge gaps in our understanding of the mechanisms that underlie stress-induced memory modulation.
Collapse
Affiliation(s)
- Niek Brosens
- Brain Plasticity Group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands.
| | - Sylvie L Lesuis
- Brain Plasticity Group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Cellular and Cognitive Neuroscience group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Del Vecchio V, Mele L, Panda SK, Sanchez-Pajares IR, Mosca L, Tirino V, Barbieri M, Bruzzese F, Luciano A, Marino FZ, Accardo M, Nicoletti GF, Papaccio G, Barbieri A, Desiderio V. β 2-AR inhibition enhances EGFR antibody efficacy hampering the oxidative stress response machinery. Cell Death Dis 2023; 14:613. [PMID: 37723219 PMCID: PMC10507049 DOI: 10.1038/s41419-023-06129-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 08/11/2023] [Accepted: 09/06/2023] [Indexed: 09/20/2023]
Abstract
The β2-Adrenergic receptor (β2-ARs) is a cell membrane-spanning G protein-coupled receptors (GPCRs) physiologically involved in stress-related response. In many cancers, the β2-ARs signaling drives the tumor development and transformation, also promoting the resistance to the treatments. In HNSCC cell lines, the β2-AR selective inhibition synergistically amplifies the cytotoxic effect of the MEK 1/2 by affecting the p38/NF-kB oncogenic pathway and contemporary reducing the NRF-2 mediated antioxidant cell response. In this study, we aimed to validate the anti-tumor effect of β2-AR blockade and the synergism with MEK/ERK and EGFR pathway inhibition in a pre-clinical orthotopic mouse model of HNSCC. Interestingly, we found a strong β2-ARs expression in the tumors that were significantly reduced after prolonged treatment with β2-Ars inhibitor (ICI) and EGFR mAb Cetuximab (CTX) in combination. The β2-ARs down-regulation correlated in mice with a significant tumor growth delay, together with the MAPK signaling switch-off caused by the blockade of the MEK/ERK phosphorylation. We also demonstrated that the administration of ICI and CTX in combination unbalanced the cell ROS homeostasis by blocking the NRF-2 nuclear translocation with the relative down-regulation of the antioxidant enzyme expression. Our findings highlighted for the first time, in a pre-clinical in vivo model, the efficacy of the β2-ARs inhibition in the treatment of the HNSCC, remarkably in combination with CTX, which is the standard of care for unresectable HNSCC.
Collapse
Affiliation(s)
- Vitale Del Vecchio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luigi Mele
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Sameer Kumar Panda
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Laura Mosca
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Virginia Tirino
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Massimiliano Barbieri
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Francesca Bruzzese
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Antonio Luciano
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Federica Zito Marino
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marina Accardo
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Francesco Nicoletti
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania "L. Vanvitelli", Via L. de Crecchio 6, 80138, Naples, Italy
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Antonio Barbieri
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Vincenzo Desiderio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
4
|
Huang SM, Xiong MY, Liu L, Mu J, Wang MW, Jia YL, Cai K, Tie L, Zhang C, Cao S, Wen X, Wang JL, Guo SC, Li Y, Qu CX, He QT, Cai BY, Xue C, Gan S, Xie Y, Cong X, Yang Z, Kong W, Li S, Li Z, Xiao P, Yang F, Yu X, Guan YF, Zhang X, Liu Z, Yang BX, Du Y, Sun JP. Single hormone or synthetic agonist induces G s/G i coupling selectivity of EP receptors via distinct binding modes and propagating paths. Proc Natl Acad Sci U S A 2023; 120:e2216329120. [PMID: 37478163 PMCID: PMC10372679 DOI: 10.1073/pnas.2216329120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/18/2023] [Indexed: 07/23/2023] Open
Abstract
To accomplish concerted physiological reactions, nature has diversified functions of a single hormone at at least two primary levels: 1) Different receptors recognize the same hormone, and 2) different cellular effectors couple to the same hormone-receptor pair [R.P. Xiao, Sci STKE 2001, re15 (2001); L. Hein, J. D. Altman, B.K. Kobilka, Nature 402, 181-184 (1999); Y. Daaka, L. M. Luttrell, R. J. Lefkowitz, Nature 390, 88-91 (1997)]. Not only these questions lie in the heart of hormone actions and receptor signaling but also dissecting mechanisms underlying these questions could offer therapeutic routes for refractory diseases, such as kidney injury (KI) or X-linked nephrogenic diabetes insipidus (NDI). Here, we identified that Gs-biased signaling, but not Gi activation downstream of EP4, showed beneficial effects for both KI and NDI treatments. Notably, by solving Cryo-electron microscope (cryo-EM) structures of EP3-Gi, EP4-Gs, and EP4-Gi in complex with endogenous prostaglandin E2 (PGE2)or two synthetic agonists and comparing with PGE2-EP2-Gs structures, we found that unique primary sequences of prostaglandin E2 receptor (EP) receptors and distinct conformational states of the EP4 ligand pocket govern the Gs/Gi transducer coupling selectivity through different structural propagation paths, especially via TM6 and TM7, to generate selective cytoplasmic structural features. In particular, the orientation of the PGE2 ω-chain and two distinct pockets encompassing agonist L902688 of EP4 were differentiated by their Gs/Gi coupling ability. Further, we identified common and distinct features of cytoplasmic side of EP receptors for Gs/Gi coupling and provide a structural basis for selective and biased agonist design of EP4 with therapeutic potential.
Collapse
Affiliation(s)
- Shen-Ming Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Meng-Yao Xiong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Lei Liu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Jianqiang Mu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong518055, China
| | - Ming-Wei Wang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Ying-Li Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Kui Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Lu Tie
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Chao Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Sheng Cao
- School of Medicine, Kobilka Institute of Innovative Drug Discovery, Chinese University of Hong Kong, Shenzhen, Guangdong518172, China
| | - Xin Wen
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Jia-Le Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Sheng-Chao Guo
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong250012, China
| | - Yu Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Chang-Xiu Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Qing-Tao He
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong250012, China
| | - Bo-Yang Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Chenyang Xue
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong518055, China
| | - Shiyi Gan
- School of Medicine, Kobilka Institute of Innovative Drug Discovery, Chinese University of Hong Kong, Shenzhen, Guangdong518172, China
| | - Yihe Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Xin Cong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Shuo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Zijian Li
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital, Research, Beijing100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing100191, P. R. China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong250012, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong250012, China
| | - You-Fei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian116044, China
| | - Xiaoyan Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian116044, China
| | - Zhongmin Liu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong518055, China
| | - Bao-Xue Yang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
| | - Yang Du
- School of Medicine, Kobilka Institute of Innovative Drug Discovery, Chinese University of Hong Kong, Shenzhen, Guangdong518172, China
| | - Jin-Peng Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing100191, China
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing100191, P. R. China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, China
| |
Collapse
|
5
|
Akhtar MM, Cammann VL, Templin C, Ghadri JR, Lüscher TF. Takotsubo syndrome: getting closer to its causes. Cardiovasc Res 2023:7161872. [PMID: 37183265 DOI: 10.1093/cvr/cvad053] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 01/18/2023] [Accepted: 02/07/2023] [Indexed: 05/16/2023] Open
Abstract
Takotsubo syndrome (TTS) accounts for between 1 and 4% of cases presenting clinically as an acute coronary syndrome. It typically presents as a transient cardiac phenotype of left ventricular dysfunction with spontaneous recovery. More dramatic presentations may include cardiogenic shock or cardiac arrest. Despite progress in the understanding of the condition since its first description in 1990, considerable questions remain into understanding underlying pathomechanisms. In this review article, we describe the current published data on potential underlying mechanisms associated with the onset of TTS including sympathetic nervous system over-stimulation, structural and functional alterations in the central nervous system, catecholamine secretion, alterations in the balance and distribution of adrenergic receptors, the additive impact of hormones including oestrogen, epicardial coronary or microvascular spasm, endothelial dysfunction, and genetics as potentially contributing to the cascade of events leading to the onset. These pathomechanisms provide suggestions for novel potential therapeutic strategies in patients with TTS including the role of cognitive behavioural therapy, beta-blockers, and endothelin-A antagonists. The underlying mechanism of TTS remains elusive. In reality, physical or emotional stressors likely trigger through the amygdala and hippocampus a central neurohumoral activation with the local and systemic secretion of excess catecholamine and other neurohormones, which exert its effect on the myocardium through a metabolic switch, altered cellular signalling, and endothelial dysfunction. These complex pathways exert a regional activation in the myocardium through the altered distribution of adrenoceptors and density of autonomic innervation as a protective mechanism from myocardial apoptosis. More research is needed to understand how these different complex mechanisms interact with each other to bring on the TTS phenotype.
Collapse
Affiliation(s)
- Mohammed Majid Akhtar
- Royal Brompton and Harefield Hospitals, Imperial College and King's College, London SW3 6NP, UK
| | - Victoria L Cammann
- University Heart Center, Department of Cardiology, University Hospital Zürich, Zürich 8091, Switzerland
| | - Christian Templin
- University Heart Center, Department of Cardiology, University Hospital Zürich, Zürich 8091, Switzerland
| | - Jelena R Ghadri
- University Heart Center, Department of Cardiology, University Hospital Zürich, Zürich 8091, Switzerland
| | - Thomas F Lüscher
- Royal Brompton and Harefield Hospitals, Imperial College and King's College, London SW3 6NP, UK
- Center for Molecular Cardiology, University of Zürich, Zürich 8952, Switzerland
| |
Collapse
|
6
|
Kaur S, Sokrat B, Capozzi ME, El K, Bai Y, Jazic A, Han B, Krishnakumar K, D'Alessio DA, Campbell JE, Bouvier M, Shenoy SK. The Ubiquitination Status of the Glucagon Receptor determines Signal Bias. J Biol Chem 2023; 299:104690. [PMID: 37037304 DOI: 10.1016/j.jbc.2023.104690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023] Open
Abstract
The pancreatic hormone glucagon activates the glucagon receptor (GCGR), a class B seven-transmembrane G protein-coupled receptor (GPCR) that couples to the stimulatory heterotrimeric Gs protein and provokes protein kinase A-dependent signaling cascades vital to hepatic glucose metabolism and islet insulin secretion. Glucagon-stimulation also initiates recruitment of the endocytic adaptors, β-arrestin1 and β-arrestin2, which regulate desensitization and internalization of the GCGR. Unlike many other GPCRs, the GCGR expressed at the plasma membrane is constitutively ubiquitinated and upon agonist-activation, internalized GCGRs are deubiquitinated at early endosomes and recycled via Rab4-containing vesicles. Herein we report a novel link between the ubiquitination status and signal transduction mechanism of the GCGR. In the deubiquitinated state, coupling of the GCGR to Gs is diminished, while binding to β-arrestin is enhanced with signaling biased to a β-arrestin1-dependent p38 mitogen activated protein kinase (MAPK) pathway. This ubiquitin-dependent signaling bias arises through the modification of lysine333 (K333) on the cytoplasmic face of transmembrane helix V. Compared with the GCGR-WT, the mutant GCGR-K333R has impaired ubiquitination, diminished G protein coupling and protein kinase A signaling, but unimpaired potentiation of glucose-stimulated-insulin secretion in response to agonist-stimulation, which involves p38 MAPK signaling. Both WT and GCGR-K333R promote the formation of glucagon-induced β-arrestin1-dependent p38 signaling scaffold that requires canonical upstream MAPK-Kinase3, but is independent of Gs, Gi and β-arrestin2. Thus ubiquitination/deubiquitination at K333 in the GCGR defines the activation of distinct transducers with the potential to influence various facets of glucagon signaling in health and disease.
Collapse
Affiliation(s)
- Suneet Kaur
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Badr Sokrat
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, H3T 1J4 Canada; Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, H3T 1J4 Canada
| | - Megan E Capozzi
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Kimberley El
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Yushi Bai
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Aeva Jazic
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Bridgette Han
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Kaavya Krishnakumar
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford CA 94305
| | - David A D'Alessio
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Jonathan E Campbell
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, H3T 1J4 Canada; Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, H3T 1J4 Canada
| | - Sudha K Shenoy
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
7
|
Xu R, Feng S, Ao Z, Chen Y, Su C, Feng X, Fu Q, Yang X. Long-Acting β2 Adrenergic Receptor Agonist Ameliorates Imiquimod-Induced Psoriasis-Like Skin Lesion by Regulating Keratinocyte Proliferation and Apoptosis. Front Pharmacol 2022; 13:865715. [PMID: 35795567 PMCID: PMC9250983 DOI: 10.3389/fphar.2022.865715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 06/01/2022] [Indexed: 11/25/2022] Open
Abstract
Psoriasis is a chronic inflammatory disease that affects approximately 1%–5% of the population worldwide. Considering frequent relapse, adverse drug reactions, and large costs of treatment, it is urgent to identify new medications for psoriasis. Keratinocytes play an essential role during psoriasis development, and they express high levels of β2-Adrenergic receptor (β2-AR), which increases intracellular cAMP levels when activated. Increased level of cAMP is associated with the inhibition of epidermal cell proliferation. In the present study, we observed the effect of salmeterol, a long-acting β2-AR agonist, on the proliferation and apoptosis of keratinocytes as well as imiquimod-induced psoriasis-like skin lesions in mice. As phosphodiesterase 4 (PDE4) inhibitors increases intracellular cAMP concentration by inhibiting its inactivation, we further explored the synergetic effect of a PDE4 inhibitor and salmeterol on psoriasis-like skin lesions in mice. Our results indicated that salmeterol effectively inhibited the proliferation of HaCaT cells induced by TNF-α and serum, and this effect was accompanied by significantly increased apoptosis and CREB phosphorylation, which were reversed by the PKA inhibitor, H89. Salmeterol ameliorated imiquimod-induced psoriasis-like skin lesions in mice, but salmeterol combined with a PDE4 inhibitor had no synergetic effect in improving skin lesions in mice. Of note, the synergistic effects of anti-proliferation and induction of apoptosis in HaCaT cells appeared by inhibiting ERK signaling. In summary, salmeterol, a long-acting β2-AR agonist, alleviates the severity of psoriasis via inhibiting the proliferation and promoting apoptosis of keratinocytes, partially by activating the cAMP/PKA signaling pathway.
Collapse
Affiliation(s)
- Rui Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Shi Feng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Zhou Ao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Yingxiang Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Congping Su
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Xiuling Feng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
- *Correspondence: Qin Fu, ; Xiaoyan Yang,
| | - Xiaoyan Yang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
- *Correspondence: Qin Fu, ; Xiaoyan Yang,
| |
Collapse
|
8
|
Mani A. PDE4DIP in health and diseases. Cell Signal 2022; 94:110322. [PMID: 35346821 PMCID: PMC9618167 DOI: 10.1016/j.cellsig.2022.110322] [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: 02/16/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 11/15/2022]
Abstract
Cyclic-AMP (cAMP), the first second messenger to be identified, is synthesized, and is universally utilized as a second messenger, and plays important roles in integrity, and function of organs, including heart. Through its coupling with other intracellular messengers, cAMP facilitates excitation-contraction coupling, increases heart rate and conduction velocity. It is degraded by a class of enzymes called cAMP-dependent phosphodiesterase (PDE), with PDE3 and PDE4 being the predominant isoforms in the heart. This highly diverse class of enzymes degrade cAMP and through anchoring proteins generates dynamic microdomains to target specific proteins and control specific cell functions in response to various stimuli. The impaired function of the anchoring protein either by inherited genetic mutations or acquired injuries results in altered intracellular targeting, and blunted responsiveness to stimulating pathways and contributes to pathological cardiac remodeling, cardiac arrhythmias and reduced cell survival. Recent genetic studies provide compelling evidence for an association between the variants in the anchoring protein PDE4DIP and atrial fibrillation, stroke, and heart failure.
Collapse
Affiliation(s)
- Arya Mani
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
9
|
Convergent selective signaling impairment exposes the pathogenicity of latrophilin-3 missense variants linked to inheritable ADHD susceptibility. Mol Psychiatry 2022; 27:2425-2438. [PMID: 35393556 PMCID: PMC9135631 DOI: 10.1038/s41380-022-01537-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 12/13/2022]
Abstract
Latrophilin-3 (Lphn3; also known as ADGRL3) is a member of the adhesion G Protein Coupled Receptor subfamily, which participates in the stabilization and maintenance of neuronal networks by mediating intercellular adhesion through heterophilic interactions with transmembrane ligands. Polymorphisms modifying the Lphn3 gene are associated with attention-deficit/hyperactivity disorder (ADHD) in children and its persistence into adulthood. How these genetic alterations affect receptor function remains unknown. Here, we conducted the functional validation of distinct ADHD-related Lphn3 variants bearing mutations in the receptor's adhesion motif-containing extracellular region. We found that all variants tested disrupted the ability of Lphn3 to stabilize intercellular adhesion in a manner that was distinct between ligands classes, but which did not depend on ligand-receptor interaction parameters, thus pointing to altered intrinsic receptor signaling properties. Using G protein signaling biosensors, we determined that Lphn3 couples to Gαi1, Gαi2, Gαs, Gαq, and Gα13. However, all ADHD-related receptor variants consistently lacked intrinsic as well as ligand-dependent Gα13 coupling efficiency while maintaining unaltered coupling to Gαi, Gαs, and Gαq. Consistent with these alterations, actin remodeling functions as well as actin-relevant RhoA signaling normally displayed by the constitutively active Lphn3 receptor were impeded by select receptor variants, thus supporting additional signaling defects. Taken together, our data point to Gα13 selective signaling impairments as representing a disease-relevant pathogenicity pathway that can be inherited through Lphn3 gene polymorphisms. This study highlights the intricate interplay between Lphn3 GPCR functions and the actin cytoskeleton in modulating neurodevelopmental cues related to ADHD etiology.
Collapse
|
10
|
Harkes R, Kukk O, Mukherjee S, Klarenbeek J, van den Broek B, Jalink K. Dynamic FRET-FLIM based screening of signal transduction pathways. Sci Rep 2021; 11:20711. [PMID: 34671065 PMCID: PMC8528867 DOI: 10.1038/s41598-021-00098-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/15/2021] [Indexed: 12/28/2022] Open
Abstract
Fluorescence Lifetime Imaging (FLIM) is an intrinsically quantitative method to screen for protein-protein interactions and is frequently used to record the outcome of signal transduction events. With new highly sensitive and photon efficient FLIM instrumentation, the technique also becomes attractive to screen, with high temporal resolution, for fast changes in Förster Resonance Energy Transfer (FRET), such as those occurring upon activation of cell signaling. The second messenger cyclic adenosine monophosphate (cAMP) is rapidly formed following activation of certain cell surface receptors. cAMP is subsequently degraded by a set of phosphodiesterases (PDEs) which display cell-type specific expression and may also affect baseline levels of the messenger. To study which specific PDEs contribute most to cAMP regulation, we knocked down individual PDEs and recorded breakdown rates of cAMP levels following transient stimulation in HeLa cells stably expressing the FRET/FLIM sensor, Epac-SH189. Many hundreds of cells were recorded at 5 s intervals for each condition. FLIM time traces were calculated for every cell, and decay kinetics were obtained. cAMP clearance was significantly slower when PDE3A and, to a lesser amount, PDE10A were knocked down, identifying these isoforms as dominant in HeLa cells. However, taking advantage of the quantitative FLIM data, we found that knockdown of individual PDEs has a very limited effect on baseline cAMP levels. By combining photon-efficient FLIM instrumentation with optimized sensors, systematic gene knockdown and an automated open-source analysis pipeline, our study demonstrates that dynamic screening of transient cell signals has become feasible. The quantitative platform described here provides detailed kinetic analysis of cellular signals in individual cells with unprecedented throughput.
Collapse
Affiliation(s)
- Rolf Harkes
- Cell Biophysics Group, Department of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Olga Kukk
- Cell Biophysics Group, Department of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sravasti Mukherjee
- Cell Biophysics Group, Department of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeffrey Klarenbeek
- Cell Biophysics Group, Department of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bram van den Broek
- Cell Biophysics Group, Department of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- BioImaging Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kees Jalink
- Cell Biophysics Group, Department of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Mitra A, Sarkar A, Borics A. Universal Properties and Specificities of the β 2-Adrenergic Receptor-G s Protein Complex Activation Mechanism Revealed by All-Atom Molecular Dynamics Simulations. Int J Mol Sci 2021; 22:10423. [PMID: 34638767 PMCID: PMC8508748 DOI: 10.3390/ijms221910423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 11/16/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are transmembrane proteins of high pharmacological relevance. It has been proposed that their activity is linked to structurally distinct, dynamically interconverting functional states and the process of activation relies on an interconnecting network of conformational switches in the transmembrane domain. However, it is yet to be uncovered how ligands with different extents of functional effect exert their actions. According to our recent hypothesis, based on indirect observations and the literature data, the transmission of the external stimulus to the intracellular surface is accompanied by the shift of macroscopic polarization in the transmembrane domain, furnished by concerted movements of highly conserved polar motifs and the rearrangement of polar species. In this follow-up study, we have examined the β2-adrenergic receptor (β2AR) to see if our hypothesis drawn from an extensive study of the μ-opioid receptor (MOP) is fundamental and directly transferable to other class A GPCRs. We have found that there are some general similarities between the two receptors, in agreement with previous studies, and there are some receptor-specific differences that could be associated with different signaling pathways.
Collapse
Affiliation(s)
- Argha Mitra
- Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre, 62. Temesvári krt., H-6726 Szeged, Hungary; (A.M.); (A.S.)
- Theoretical Medicine Doctoral School, Faculty of Medicine, University of Szeged, 97. Tisza L. krt., H-6722 Szeged, Hungary
| | - Arijit Sarkar
- Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre, 62. Temesvári krt., H-6726 Szeged, Hungary; (A.M.); (A.S.)
- Theoretical Medicine Doctoral School, Faculty of Medicine, University of Szeged, 97. Tisza L. krt., H-6722 Szeged, Hungary
| | - Attila Borics
- Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre, 62. Temesvári krt., H-6726 Szeged, Hungary; (A.M.); (A.S.)
| |
Collapse
|
12
|
Pharmacological Characterization of Low Molecular Weight Biased Agonists at the Follicle Stimulating Hormone Receptor. Int J Mol Sci 2021; 22:ijms22189850. [PMID: 34576014 PMCID: PMC8469697 DOI: 10.3390/ijms22189850] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 01/14/2023] Open
Abstract
Follicle-stimulating hormone receptor (FSHR) plays a key role in reproduction through the activation of multiple signaling pathways. Low molecular weight (LMW) ligands composed of biased agonist properties are highly valuable tools to decipher complex signaling mechanisms as they allow selective activation of discrete signaling cascades. However, available LMW FSHR ligands have not been fully characterized yet. In this context, we explored the pharmacological diversity of three benzamide and two thiazolidinone derivatives compared to FSH. Concentration/activity curves were generated for Gαs, Gαq, Gαi, β-arrestin 2 recruitment, and cAMP production, using BRET assays in living cells. ERK phosphorylation was analyzed by Western blotting, and CRE-dependent transcription was assessed using a luciferase reporter assay. All assays were done in either wild-type, Gαs or β-arrestin 1/2 CRISPR knockout HEK293 cells. Bias factors were calculated for each pair of read-outs by using the operational model. Our results show that each ligand presented a discrete pharmacological efficacy compared to FSH, ranging from super-agonist for β-arrestin 2 recruitment to pure Gαs bias. Interestingly, LMW ligands generated kinetic profiles distinct from FSH (i.e., faster, slower or transient, depending on the ligand) and correlated with CRE-dependent transcription. In addition, clear system biases were observed in cells depleted of either Gαs or β-arrestin genes. Such LMW properties are useful pharmacological tools to better dissect the multiple signaling pathways activated by FSHR and assess their relative contributions at the cellular and physio-pathological levels.
Collapse
|
13
|
Abou Ziki MD, Bhat N, Neogi A, Driscoll TP, Ugwu N, Liu Y, Smith E, Abboud JM, Chouairi S, Schwartz MA, Akar JG, Mani A. Epistatic interaction of PDE4DIP and DES mutations in familial atrial fibrillation with slow conduction. Hum Mutat 2021; 42:1279-1293. [PMID: 34289528 DOI: 10.1002/humu.24265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/29/2021] [Accepted: 07/06/2021] [Indexed: 11/09/2022]
Abstract
The genetic causes of atrial fibrillation (AF) with slow conduction are unknown. Eight kindreds with familial AF and slow conduction, including a family affected by early-onset AF, heart block, and incompletely penetrant nonischemic dilated cardiomyopathy (DCM) underwent whole exome sequencing. A known pathogenic mutation in the desmin (DES) gene resulting in p.S13F substitution (NM_001927.3:c.38C>T) at a PKC phosphorylation site was identified in all four members of the kindred with early-onset AF and heart block, while only two developed DCM. Higher penetrance for AF and heart block prompted a genetic screening for DES modifier(s). A deleterious mutation in the phosphodiesterase-4D-interacting-protein (PDE4DIP) gene resulting in p.A123T substitution (NM_001002811:c.367G>A) was identified that segregated with early-onset AF, heart block, and the DES mutation. Three additional novel deleterious PDE4DIP mutations were identified in four other unrelated kindreds. Characterization of PDE4DIPA123T in vitro suggested impaired compartmentalization of PKA and PDE4D characterized by reduced colocalization with PDE4D, increased cAMP activation leading to higher PKA phosphorylation of the β2-adrenergic-receptor, and decreased PKA phosphorylation of desmin after isoproterenol stimulation. Our findings identify PDE4DIP as a novel gene for slow AF and unravel its epistatic interaction with DES mutations in development of conduction disease and arrhythmia.
Collapse
Affiliation(s)
- Maen D Abou Ziki
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Neha Bhat
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Arpita Neogi
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tristan P Driscoll
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, Tallahassee, Florida, USA
| | - Nelson Ugwu
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ya Liu
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emily Smith
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Johny M Abboud
- Saint George Hospital University Medical Center, Beirut, Lebanon
| | - Salah Chouairi
- Saint George Hospital University Medical Center, Beirut, Lebanon
| | - Martin A Schwartz
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Joseph G Akar
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Arya Mani
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
14
|
Chakrabarti S, Liu NJ, Gintzler AR. Relevance of Mu-Opioid Receptor Splice Variants and Plasticity of Their Signaling Sequelae to Opioid Analgesic Tolerance. Cell Mol Neurobiol 2021; 41:855-862. [PMID: 32804312 DOI: 10.1007/s10571-020-00934-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/01/2020] [Indexed: 10/23/2022]
Abstract
Opioid dose escalation to effectively control pain is often linked to the current prescription opioid abuse epidemic. This creates social as well as medical imperatives to better understand the mechanistic underpinnings of opioid tolerance to develop interventions that minimize it, thereby maximizing the analgesic effectiveness of opioids. Profound opioid analgesic tolerance can be observed in the absence of mu-opioid receptor (MOR) downregulation, aggregate MOR G protein uncoupling, and MOR desensitization, in the absence of impaired G protein coupled receptor kinase phosphorylation, arrestin binding, or endocytosis. Thus, we have explored alternative biochemical sequelae that might better account for opioid analgesic tolerance. Our findings indicate that substantial plasticity among upstream and downstream components of opioid receptor signaling and the emergence of alternative signaling pathways are major contributors to opioid analgesic tolerance. An exemplar of this plasticity is our findings that chronic morphine upregulates the MOR variants MOR-1B2 and MOR-1C1 and phosphorylation of their C-terminal sites not present in MOR-1, events causally associated with the chronic morphine-induced shift in MOR G protein coupling from predominantly Gi/Go inhibitory to Gs-stimulatory adenylyl cyclase signaling. The unique feature(s) of these variants that underlies their susceptibility to adapting to chronic morphine by altering the nature of their G protein coupling reveals the richness and pliability of MOR signaling that is enabled by generating a wide diversity of MOR variants. Furthermore, given differential anatomical expression patterns of MOR variants, MOR splice variant-dependent adaptations to chronic morphine could enable mechanistic underpinnings of tolerance and dependence that are CNS region- and cell-specific.
Collapse
Affiliation(s)
- Sumita Chakrabarti
- Department Obstetrics and Gynecology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY, 11203, USA
| | - Nai-Jiang Liu
- Department Obstetrics and Gynecology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY, 11203, USA
| | - Alan R Gintzler
- Department Obstetrics and Gynecology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY, 11203, USA.
| |
Collapse
|
15
|
Patwardhan A, Cheng N, Trejo J. Post-Translational Modifications of G Protein-Coupled Receptors Control Cellular Signaling Dynamics in Space and Time. Pharmacol Rev 2021; 73:120-151. [PMID: 33268549 PMCID: PMC7736832 DOI: 10.1124/pharmrev.120.000082] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family comprising >800 signaling receptors that regulate numerous cellular and physiologic responses. GPCRs have been implicated in numerous diseases and represent the largest class of drug targets. Although advances in GPCR structure and pharmacology have improved drug discovery, the regulation of GPCR function by diverse post-translational modifications (PTMs) has received minimal attention. Over 200 PTMs are known to exist in mammalian cells, yet only a few have been reported for GPCRs. Early studies revealed phosphorylation as a major regulator of GPCR signaling, whereas later reports implicated a function for ubiquitination, glycosylation, and palmitoylation in GPCR biology. Although our knowledge of GPCR phosphorylation is extensive, our knowledge of the modifying enzymes, regulation, and function of other GPCR PTMs is limited. In this review we provide a comprehensive overview of GPCR post-translational modifications with a greater focus on new discoveries. We discuss the subcellular location and regulatory mechanisms that control post-translational modifications of GPCRs. The functional implications of newly discovered GPCR PTMs on receptor folding, biosynthesis, endocytic trafficking, dimerization, compartmentalized signaling, and biased signaling are also provided. Methods to detect and study GPCR PTMs as well as PTM crosstalk are further highlighted. Finally, we conclude with a discussion of the implications of GPCR PTMs in human disease and their importance for drug discovery. SIGNIFICANCE STATEMENT: Post-translational modification of G protein-coupled receptors (GPCRs) controls all aspects of receptor function; however, the detection and study of diverse types of GPCR modifications are limited. A thorough understanding of the role and mechanisms by which diverse post-translational modifications regulate GPCR signaling and trafficking is essential for understanding dysregulated mechanisms in disease and for improving and refining drug development for GPCRs.
Collapse
Affiliation(s)
- Anand Patwardhan
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| | - Norton Cheng
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| | - JoAnn Trejo
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
16
|
Gurevich EV, Gurevich VV. GRKs as Modulators of Neurotransmitter Receptors. Cells 2020; 10:cells10010052. [PMID: 33396400 PMCID: PMC7823573 DOI: 10.3390/cells10010052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 01/08/2023] Open
Abstract
Many receptors for neurotransmitters, such as dopamine, norepinephrine, acetylcholine, and neuropeptides, belong to the superfamily of G protein-coupled receptors (GPCRs). A general model posits that GPCRs undergo two-step homologous desensitization: the active receptor is phosphorylated by kinases of the G protein-coupled receptor kinase (GRK) family, whereupon arrestin proteins specifically bind active phosphorylated receptors, shutting down G protein-mediated signaling, facilitating receptor internalization, and initiating distinct signaling pathways via arrestin-based scaffolding. Here, we review the mechanisms of GRK-dependent regulation of neurotransmitter receptors, focusing on the diverse modes of GRK-mediated phosphorylation of receptor subtypes. The immediate signaling consequences of GRK-mediated receptor phosphorylation, such as arrestin recruitment, desensitization, and internalization/resensitization, are equally diverse, depending not only on the receptor subtype but also on phosphorylation by GRKs of select receptor residues. We discuss the signaling outcome as well as the biological and behavioral consequences of the GRK-dependent phosphorylation of neurotransmitter receptors where known.
Collapse
|
17
|
Ippolito M, Benovic JL. Biased agonism at β-adrenergic receptors. Cell Signal 2020; 80:109905. [PMID: 33385503 DOI: 10.1016/j.cellsig.2020.109905] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/10/2020] [Accepted: 12/27/2020] [Indexed: 12/19/2022]
Abstract
The β-adrenergic receptors (βARs) include three subtypes, β1, β2 and β3. These receptors are widely expressed and regulate numerous physiological processes including cardiovascular and metabolic functions and airway tone. The βARs are also important targets in the treatment of many diseases including hypertension, heart failure and asthma. In some cases, the use of current βAR ligands to treat a disease is suboptimal and can lead to severe side effects. One strategy to potentially improve such treatments is the development of biased agonists that selectively regulate a subset of βAR signaling pathways and responses. Here we discuss the compounds identified to date that preferentially activate a Gs- or β-arrestin-mediated signaling pathway through βARs. Mechanistic insight on how these compounds bias signaling sheds light on the potential development of even more selective compounds that should have increased utility in treating disease.
Collapse
Affiliation(s)
- Michael Ippolito
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, United States of America.
| |
Collapse
|
18
|
Chakrabarti S, Liu NJ, Gintzler AR. Phosphorylation of unique C-terminal sites of the mu-opioid receptor variants 1B2 and 1C1 influences their Gs association following chronic morphine. J Neurochem 2019; 152:449-467. [PMID: 31479519 DOI: 10.1111/jnc.14863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/14/2019] [Accepted: 08/11/2019] [Indexed: 11/27/2022]
Abstract
We recently demonstrated in rat spinal cord that a regimen of escalating doses of systemic morphine, analogous to regimens used clinically for chronic pain management, selectively up-regulates the mu-opioid receptor (MOR) splice variants MOR-1B2 and MOR-1C1 mRNA and functional protein. This study investigated the potential relevance of up-regulating MOR-1B2 and MOR-1C1 to the ability of chronic morphine to shift MOR signaling from predominantly Gi /Go inhibitory to Gs stimulatory. Specifically, we tested the hypotheses that chronic morphine induces phosphorylation of carboxyl terminal sites unique to MOR-1B2 and MOR-1C1, and that this phosphorylation is causally related to augmented association of these variants with Gs α. Hypotheses were validated by (i) abolition of the chronic morphine-induced increment in MOR-1C1 and MOR-1B2 association with Gs α by inhibitors of protein kinase A and Casein kinase 2, respectively; (ii) failure of chronic morphine to augment MOR variant Gs α interactions in Chinese hamster ovary cells transiently transfected with either rat MOR-1C1 or MOR-1B2 in which targeted protein kinase A and Casein kinase 2 serine phosphorylation sites, respectively, were mutated to alanine; (iii) abrogation of chronic morphine-induced augmented MOR Gs α association in spinal cord of male rats following intrathecal administration of dicer substrate small interfering RNAs targeting MOR-1B2/MOR-1C1 mRNA. The ability of chronic morphine to not only up-regulate-specific MOR variants but also their carboxyl terminal phosphorylation and consequent augmented association with Gs α may represent a novel component of opioid tolerance mechanisms, suggesting novel potential targets for tolerance abatement.
Collapse
Affiliation(s)
- Sumita Chakrabarti
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| | - Nai-Jiang Liu
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| | - Alan R Gintzler
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| |
Collapse
|
19
|
Shen A, Chen D, Kaur M, Bartels P, Xu B, Shi Q, Martinez JM, Man KNM, Nieves-Cintron M, Hell JW, Navedo MF, Yu XY, Xiang YK. β-blockers augment L-type Ca 2+ channel activity by targeting spatially restricted β 2AR signaling in neurons. eLife 2019; 8:49464. [PMID: 31609201 PMCID: PMC6813027 DOI: 10.7554/elife.49464] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/13/2019] [Indexed: 01/02/2023] Open
Abstract
G protein-coupled receptors (GPCRs) transduce pleiotropic intracellular signals in mammalian cells. Here, we report neuronal excitability of β-blockers carvedilol and alprenolol at clinically relevant nanomolar concentrations. Carvedilol and alprenolol activate β2AR, which promote G protein signaling and cAMP/PKA activities without action of G protein receptor kinases (GRKs). The cAMP/PKA activities are restricted within the immediate vicinity of activated β2AR, leading to selectively enhance PKA-dependent phosphorylation and stimulation of endogenous L-type calcium channel (LTCC) but not AMPA receptor in rat hippocampal neurons. Moreover, we have engineered a mutant β2AR that lacks the catecholamine binding pocket. This mutant is preferentially activated by carvedilol but not the orthosteric agonist isoproterenol. Carvedilol activates the mutant β2AR in mouse hippocampal neurons augmenting LTCC activity through cAMP/PKA signaling. Together, our study identifies a mechanism by which β-blocker-dependent activation of GPCRs promotes spatially restricted cAMP/PKA signaling to selectively target membrane downstream effectors such as LTCC in neurons.
Collapse
Affiliation(s)
- Ao Shen
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Pharmacology, University of California Davis, Davis, United States
| | - Dana Chen
- Department of Pharmacology, University of California Davis, Davis, United States
| | - Manpreet Kaur
- Department of Pharmacology, University of California Davis, Davis, United States
| | - Peter Bartels
- Department of Pharmacology, University of California Davis, Davis, United States
| | - Bing Xu
- Department of Pharmacology, University of California Davis, Davis, United States.,VA Northern California Health Care System, Mather, United States
| | - Qian Shi
- Department of Pharmacology, University of California Davis, Davis, United States
| | - Joseph M Martinez
- Department of Pharmacology, University of California Davis, Davis, United States
| | - Kwun-Nok Mimi Man
- Department of Pharmacology, University of California Davis, Davis, United States
| | | | - Johannes W Hell
- Department of Pharmacology, University of California Davis, Davis, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California Davis, Davis, United States
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yang K Xiang
- Department of Pharmacology, University of California Davis, Davis, United States.,VA Northern California Health Care System, Mather, United States
| |
Collapse
|
20
|
Ovando-Zambrano JC, Arias-Montaño JA, Boucard AA. Alternative splicing event modifying ADGRL1/latrophilin-1 cytoplasmic tail promotes both opposing and dual cAMP signaling pathways. Ann N Y Acad Sci 2019; 1456:168-185. [PMID: 31339586 DOI: 10.1111/nyas.14198] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/11/2019] [Accepted: 07/02/2019] [Indexed: 12/31/2022]
Abstract
The adhesion G protein-coupled receptor ADGRL1/latrophilin-1 (LPHN1) stabilizes synapse formation through heterophilic interactions. A growing consensus is pointing to the role of LPHN1 in modulating intracellular levels of cAMP, although conflicting data exist. Variants of LPHN1 resulting from alternative splicing differ at multiple sites, two of which, designated as SSA and SSB, modify extracellular and intracellular receptor regions, respectively. While SSA splicing modulates receptor-ligand affinity, the function of SSB splicing remains elusive. Here, we explored the role of SSB in an attempt to unify current findings on LPHN1 signaling pathways by testing SSB-containing and SSB-deficient receptor variants in signaling paradigms involving interaction with their ligands neurexin and FLRT. cAMP competitive binding assays revealed that cells expressing either receptor variant exhibited a ligand-dependent decrease in the forskolin-induced cAMP accumulation. Surprisingly, the expression of SSB-containing LPHN1 promoted both constitutive and ligand-dependent cAMP production, whereas SSB-deficient LPHN1 did not. Pertussis toxin treatment unveiled a constitutive coupling to Gαi/o for SSB-containing LPHN1 while abrogating the ligand-mediated activation of Gαs . Importantly, neither receptor variant increased the intracellular concentration of Ca2+ nor MAP kinase activation in the presence of ligands. These results suggest that SSB splicing selectively affects the duality of LPHN1 signaling toward opposing cAMP pathways.
Collapse
Affiliation(s)
- José-Carlos Ovando-Zambrano
- Departamento de Fisiología, Biofísica, y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| | - José-Antonio Arias-Montaño
- Departamento de Fisiología, Biofísica, y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| | - Antony A Boucard
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| |
Collapse
|
21
|
Chavez-Abiega S, Goedhart J, Bruggeman FJ. Physical biology of GPCR signalling dynamics inferred from fluorescence spectroscopy and imaging. Curr Opin Struct Biol 2019; 55:204-211. [PMID: 31319372 DOI: 10.1016/j.sbi.2019.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/30/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022]
Abstract
The physical biology of G protein-coupled receptor (GPCR) signalling can be inferred from imaging of single molecules and single living cells. In this opinion paper, we highlight recent developments in technologies to study GPCR signalling in vitro and in cyto. We start from mobility and localisation characteristics of single receptors in membranes. Subsequently, we discuss the kinetics of shifts in receptor-conformation equilibrium due to allosteric binding events and G protein activation. We continue with recent insights into downstream signalling and the role of delayed negative feedback to suppress GPCR signalling. Finally, we discuss new strategies to reveal how the multiplex signalling responses of cells to ligand mixtures, mediated by their entire receptor arsenal, can be disentangled, using single-cell data.
Collapse
Affiliation(s)
- Sergei Chavez-Abiega
- Section Molecular Cytology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, NL-1098 XH, Amsterdam, The Netherlands; Section Systems Bioinformatics, Amsterdam Institute for Molecules, Medicines and Systems, VU University, De Boelelaan 1085, NL-1081 HV, Amsterdam, The Netherlands
| | - Joachim Goedhart
- Section Molecular Cytology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, NL-1098 XH, Amsterdam, The Netherlands
| | - Frank Johannes Bruggeman
- Section Systems Bioinformatics, Amsterdam Institute for Molecules, Medicines and Systems, VU University, De Boelelaan 1085, NL-1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Toledo C, Andrade DC, Díaz HS, Inestrosa NC, Del Rio R. Neurocognitive Disorders in Heart Failure: Novel Pathophysiological Mechanisms Underpinning Memory Loss and Learning Impairment. Mol Neurobiol 2019; 56:8035-8051. [PMID: 31165973 DOI: 10.1007/s12035-019-01655-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 05/20/2019] [Indexed: 01/01/2023]
Abstract
Heart failure (HF) is a major public health issue affecting more than 26 million people worldwide. HF is the most common cardiovascular disease in elder population; and it is associated with neurocognitive function decline, which represent underlying brain pathology diminishing learning and memory faculties. Both HF and neurocognitive impairment are associated with recurrent hospitalization episodes and increased mortality rate in older people, but particularly when they occur simultaneously. Overall, the published studies seem to confirm that HF patients display functional impairments relating to attention, memory, concentration, learning, and executive functioning compared with age-matched controls. However, little is known about the molecular mechanisms underpinning neurocognitive decline in HF. The present review round step recent evidence related to the possible molecular mechanism involved in the establishment of neurocognitive disorders during HF. We will make a special focus on cerebral ischemia, neuroinflammation and oxidative stress, Wnt signaling, and mitochondrial DNA alterations as possible mechanisms associated with cognitive decline in HF. Also, we provide an integrative mechanism linking pathophysiological hallmarks of altered cardiorespiratory control and the development of cognitive dysfunction in HF patients. Graphical Abstract Main molecular mechanisms involved in the establishment of cognitive impairment during heart failure. Heart failure is characterized by chronic activation of brain areas responsible for increasing cardiac sympathetic load. In addition, HF patients also show neurocognitive impairment, suggesting that the overall mechanisms that underpin cardiac sympathoexcitation may be related to the development of cognitive disorders in HF. In low cardiac output, HF cerebral infarction due to cardiac mural emboli and cerebral ischemia due to chronic or intermittent cerebral hypoperfusion has been described as a major mechanism related to the development of CI. In addition, while acute norepinephrine (NE) release may be relevant to induce neural plasticity in the hippocampus, chronic or tonic release of NE may exert the opposite effects due to desensitization of the adrenergic signaling pathway due to receptor internalization. Enhanced chemoreflex drive is a major source of sympathoexcitation in HF, and this phenomenon elevates brain ROS levels and induces neuroinflammation through breathing instability. Importantly, both oxidative stress and neuroinflammation can induce mitochondrial dysfunction and vice versa. Then, this ROS inflammatory pathway may propagate within the brain and potentially contribute to the development of cognitive impairment in HF through the activation/inhibition of key molecular pathways involved in neurocognitive decline such as the Wnt signaling pathway.
Collapse
Affiliation(s)
- C Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Center for Aging and Regeneration (CARE-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - D C Andrade
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de investigación en fisiología del ejercicio, Universidad Mayor, Santiago, Chile
| | - H S Díaz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - N C Inestrosa
- Center for Aging and Regeneration (CARE-UC), Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - R Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Center for Aging and Regeneration (CARE-UC), Pontificia Universidad Católica de Chile, Santiago, Chile. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
23
|
Local membrane charge regulates β 2 adrenergic receptor coupling to G i3. Nat Commun 2019; 10:2234. [PMID: 31110175 PMCID: PMC6527575 DOI: 10.1038/s41467-019-10108-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 04/18/2019] [Indexed: 11/13/2022] Open
Abstract
The β2 adrenergic receptor (β2AR) signals through both Gs and Gi in cardiac myocytes, and the Gi pathway counteracts the Gs pathway. However, Gi coupling is much less efficient than Gs coupling in most cell-based and biochemical assays, making it difficult to study β2AR−Gi interactions. Here we investigate the role of phospholipid composition on Gs and Gi coupling. While negatively charged phospholipids are known to enhance agonist affinity and stabilize an active state of the β2AR, we find that they impair coupling to Gi3 and facilitate coupling to Gs. Positively charged Ca2+ and Mg2+, known to interact with the negative charge on phospholipids, facilitates Gi3 coupling. Mutational analysis suggests that Ca2+ coordinates an interaction between phospholipid and the negatively charged EDGE motif on the amino terminal helix of Gi3. Taken together, our observations suggest that local membrane charge modulates the interaction between β2AR and competing G protein subtypes. In the healthy heart, the β2 adrenergic receptor (β2AR) signals through Gs and Gi proteins but the mechanism underlying G protein selectivity is not fully understood. Here, the authors show that membrane charge and intracellular cations modulate the β2AR−Gi3 interaction.
Collapse
|
24
|
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: 17.8] [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
|
25
|
Hu J, Wang Y, Le Q, Yu N, Cao X, Kuang S, Zhang M, Gu W, Sun Y, Yang Y, Yan X. Transcriptome sequencing of olfactory-related genes in olfactory transduction of large yellow croaker ( Larimichthy crocea) in response to bile salts. PeerJ 2019; 7:e6627. [PMID: 30918761 PMCID: PMC6431138 DOI: 10.7717/peerj.6627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/15/2019] [Indexed: 11/20/2022] Open
Abstract
Fish produce and release bile salts as chemical signalling substances that act as sensitive olfactory stimuli. To investigate how bile salts affect olfactory signal transduction in large yellow croaker (Larimichthy crocea), deep sequencing of olfactory epithelium was conducted to analyse olfactory-related genes in olfactory transduction. Sodium cholates (SAS) have typical bile salt chemical structures, hence we used four different concentrations of SAS to stimulate L. crocea, and the fish displayed a significant behavioural preference for 0.30% SAS. We then sequenced olfactory epithelium tissues, and identified 9938 unigenes that were significantly differentially expressed between SAS-stimulated and control groups, including 9055 up-regulated and 883 down-regulated unigenes. Subsequent Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses found eight categories linked to the olfactory transduction pathway that was highly enriched with some differentially expressed genes (DEGs), including the olfactory receptor (OR), Adenylate cyclase type 3 (ADCY3) and Calmodulin (CALM). Genes in these categories were analysed by RT-qPCR, which revealed aspects of the pathway transformation between odor detection, and recovery and adaptation. The results provide new insight into the effects of bile salt stimulation in olfactory molecular mechanisms in fishes, and expands our knowledge of olfactory transduction, and signal generation and decline.
Collapse
Affiliation(s)
- Jiabao Hu
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China
| | - Yajun Wang
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China
| | - Qijun Le
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China.,Ningbo Entry-Exit Inspection and Quarantine Bureau Technical Centre, Ningbo, China
| | - Na Yu
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China
| | - Xiaohuan Cao
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China
| | - Siwen Kuang
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China
| | - Man Zhang
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China
| | - Weiwei Gu
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China
| | - Yibo Sun
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China
| | - Yang Yang
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China
| | - Xiaojun Yan
- Key Laboratory of Applied Marine Biotechnology, Ningbo University, Ministry of Education, Ningbo, China.,Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Ningbo, China.,College of Marine Sciences, Ningbo University, Ningbo, China
| |
Collapse
|
26
|
Luttrell LM, Wang J, Plouffe B, Smith JS, Yamani L, Kaur S, Jean-Charles PY, Gauthier C, Lee MH, Pani B, Kim J, Ahn S, Rajagopal S, Reiter E, Bouvier M, Shenoy SK, Laporte SA, Rockman HA, Lefkowitz RJ. Manifold roles of β-arrestins in GPCR signaling elucidated with siRNA and CRISPR/Cas9. Sci Signal 2018; 11:11/549/eaat7650. [PMID: 30254056 DOI: 10.1126/scisignal.aat7650] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) use diverse mechanisms to regulate the mitogen-activated protein kinases ERK1/2. β-Arrestins (βArr1/2) are ubiquitous inhibitors of G protein signaling, promoting GPCR desensitization and internalization and serving as scaffolds for ERK1/2 activation. Studies using CRISPR/Cas9 to delete βArr1/2 and G proteins have cast doubt on the role of β-arrestins in activating specific pools of ERK1/2. We compared the effects of siRNA-mediated knockdown of βArr1/2 and reconstitution with βArr1/2 in three different parental and CRISPR-derived βArr1/2 knockout HEK293 cell pairs to assess the effect of βArr1/2 deletion on ERK1/2 activation by four Gs-coupled GPCRs. In all parental lines with all receptors, ERK1/2 stimulation was reduced by siRNAs specific for βArr2 or βArr1/2. In contrast, variable effects were observed with CRISPR-derived cell lines both between different lines and with activation of different receptors. For β2 adrenergic receptors (β2ARs) and β1ARs, βArr1/2 deletion increased, decreased, or had no effect on isoproterenol-stimulated ERK1/2 activation in different CRISPR clones. ERK1/2 activation by the vasopressin V2 and follicle-stimulating hormone receptors was reduced in these cells but was enhanced by reconstitution with βArr1/2. Loss of desensitization and receptor internalization in CRISPR βArr1/2 knockout cells caused β2AR-mediated stimulation of ERK1/2 to become more dependent on G proteins, which was reversed by reintroducing βArr1/2. These data suggest that βArr1/2 function as a regulatory hub, determining the balance between mechanistically different pathways that result in activation of ERK1/2, and caution against extrapolating results obtained from βArr1/2- or G protein-deleted cells to GPCR behavior in native systems.
Collapse
Affiliation(s)
- Louis M Luttrell
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA.,Research Service of the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Jialu Wang
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Bianca Plouffe
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C IJ4, Canada
| | - Jeffrey S Smith
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Lama Yamani
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Suneet Kaur
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Christophe Gauthier
- Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, CNRS, Université de Tours, 37380 Nouzilly, France
| | - Mi-Hye Lee
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Biswaranjan Pani
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Jihee Kim
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Seungkirl Ahn
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Sudarshan Rajagopal
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Eric Reiter
- Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, CNRS, Université de Tours, 37380 Nouzilly, France
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C IJ4, Canada
| | - Sudha K Shenoy
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Stéphane A Laporte
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Robert J Lefkowitz
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA. .,Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA.,Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
27
|
Discovery of novel antagonists on β2-adrenoceptor from natural products using a label-free cell phenotypic assay. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:1411-1420. [DOI: 10.1007/s00210-018-1555-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 08/15/2018] [Indexed: 10/28/2022]
|
28
|
Gao ZG, Inoue A, Jacobson KA. On the G protein-coupling selectivity of the native A 2B adenosine receptor. Biochem Pharmacol 2018; 151:201-213. [PMID: 29225130 PMCID: PMC5899946 DOI: 10.1016/j.bcp.2017.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/05/2017] [Indexed: 12/14/2022]
Abstract
A2B adenosine receptor (A2BAR) activation induces Gs-dependent cyclic AMP accumulation. However, A2BAR G protein-coupling to other signaling events, e.g. ERK1/2 and calcium, is not well documented. We explored Gi, Gq/11 and Gs coupling in 1321 N1 astrocytoma, HEK293, and T24 bladder cancer cells endogenously expressing human A2BAR, using NECA or nonnucleoside BAY60-6583 as agonist, selective Gi, Gs and Gq/11 blockers, and CRISPR/Cas9-based Gq- and Gs-null HEK293 cells. In HEK293 cells, A2BAR-mediated ERK1/2 activity occurred via both Gi and Gs, but not Gq/11. However, HEK293 cell calcium mobilization was completely blocked by Gq/11 inhibitor UBO-QIC and by Gq/11 knockout. In T24 cells, Gi was solely responsible for A2BAR-mediated ERK1/2 stimulation, and Gs suppressed ERK1/2 activity. A2BAR-mediated intracellular calcium mobilization in T24 cells was mainly via Gi, although Gs may also play a role, but Gq/11 is not involved. In 1321 N1 astrocytoma cells A2BAR activation suppressed rather than stimulated ERK1/2 activity. The ERK1/2 activity decrease was reversed by Gs downregulation using cholera toxin, but potentiated by Gi inhibitor pertussis toxin, and UBO-QIC had no effect. EPACs played an important role in A2BAR-mediated ERK1/2 signaling in all three cells. Thus, A2BAR may: couple to the same downstream pathway via different G proteins in different cell types; activate different downstream events via different G proteins in the same cell type; activate Gi and Gs, which have opposing or synergistic roles in different cell types/signaling pathways. The findings, relevant to drug discovery, address some reported controversial roles of A2BAR and could apply to signaling mechanisms in other GPCRs.
Collapse
Affiliation(s)
- Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|
29
|
Abstract
Diabetes is a major risk factor for the development of heart failure. One of the hallmarks of diabetes is insulin resistance associated with hyperinsulinemia. The literature shows that insulin and adrenergic signaling is intimately linked to each other; however, whether and how insulin may modulate cardiac adrenergic signaling and cardiac function remains unknown. Notably, recent studies have revealed that insulin receptor and β2 adrenergic receptor (β2AR) forms a membrane complex in animal hearts, bringing together the direct contact between 2 receptor signaling systems, and forming an integrated and dynamic network. Moreover, insulin can drive cardiac adrenergic desensitization via protein kinase A and G protein-receptor kinases phosphorylation of the β2AR, which compromises adrenergic regulation of cardiac contractile function. In this review, we will explore the current state of knowledge linking insulin and G protein-coupled receptor signaling, especially β-adrenergic receptor signaling in the heart, with emphasis on molecular insights regarding its role in diabetic cardiomyopathy.
Collapse
|
30
|
Kawai S, Yamada T, Matsuura T, Funao T, Nishikawa K. Neuropathic pain attenuates ischemia reperfusion injury through β2-adrenergic pathway. Life Sci 2017; 187:9-16. [PMID: 28827152 DOI: 10.1016/j.lfs.2017.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/31/2017] [Accepted: 08/02/2017] [Indexed: 12/23/2022]
Abstract
AIMS The relationship between neuropathic pain and myocardial infarction (MI) was uncertain because of some medication or underlying diseases. This study investigated the impact of neuropathic pain on ischemia reperfusion injury using isolated rat hearts and cardiomyocytes. MAIN METHODS Male Sprague-Dawley rats were assigned to the control and allodynia (AL) groups, with the latter subjected to the fifth lumbar spinal-nerve ligation. First, isolated hearts underwent 25-min ischemia and 90-min reperfusion to assess hemodynamic changes and MI area. Second, isolated cardiomyocytes underwent 10-min laser illumination to assess the opening of mitochondrial permeability transition pore (mPTP) and cellular hypercontraction. Lastly, expression of pro-survival kinases was measured in another cardiomyocytes using flow cytometry. AL-treated hearts were concomitantly examined regarding the involvement of β-adrenergic pathways by esmolol (ESM), β1-blocker (100μM, AL+ESM), and ICI118551 (ICI), β2-blocker (50nM, AL+ICI). KEY FINDINGS All hemodynamic variables did not change significantly in between-group comparisons except at 30min of reperfusion. MI area decreased remarkably in the AL and AL+ESM groups after 90-min reperfusion. The AL+ICI group significantly increased it as compared with the AL and AL+ESM groups. Similarly, the AL and AL+ESM groups significantly inhibited mPTP opening and cellular hypercontraction, whereas the AL+ICI group reversed these effects. Enhanced expression of pro-survival kinases was observed in the AL and AL+ESM groups, but the AL+ICI group abolished this enhancement. SIGNIFICANCE Our findings suggested that neuropathic pain possessed cardioprotective effects through inhibiting mPTP opening. The underlying mechanisms were possibly regulated by β2-adrenergic activation and pro-survival kinase expression in cardiomyocytes.
Collapse
Affiliation(s)
- Shigeaki Kawai
- Department of Anesthesiology, Osaka City University Graduate School of Medicine, Japan
| | - Tokuhiro Yamada
- Department of Anesthesiology, Osaka City University Graduate School of Medicine, Japan.
| | - Tadashi Matsuura
- Department of Anesthesiology, Osaka City University Graduate School of Medicine, Japan
| | - Tomoharu Funao
- Department of Anesthesiology, Osaka City University Graduate School of Medicine, Japan
| | - Kiyonobu Nishikawa
- Department of Anesthesiology, Osaka City University Graduate School of Medicine, Japan
| |
Collapse
|
31
|
β 2 -adrenoceptor-induced modulation of transglutaminase 2 transamidase activity in cardiomyoblasts. Eur J Pharmacol 2017; 813:105-121. [DOI: 10.1016/j.ejphar.2017.07.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/24/2017] [Accepted: 07/24/2017] [Indexed: 12/12/2022]
|
32
|
Shi Q, Li M, Mika D, Fu Q, Kim S, Phan J, Shen A, Vandecasteele G, Xiang YK. Heterologous desensitization of cardiac β-adrenergic signal via hormone-induced βAR/arrestin/PDE4 complexes. Cardiovasc Res 2017; 113:656-670. [PMID: 28339772 PMCID: PMC5852637 DOI: 10.1093/cvr/cvx036] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 10/20/2017] [Accepted: 02/17/2017] [Indexed: 12/22/2022] Open
Abstract
AIMS Cardiac β-adrenergic receptor (βAR) signalling is susceptible to heterologous desensitization by different neurohormonal stimuli in clinical conditions associated with heart failure. We aim to examine the underlying mechanism of cross talk between βARs and a set of G-protein coupled receptors (GPCRs) activated by hormones/agonists. METHODS AND RESULTS Rat ventricular cardiomyocytes were used to determine heterologous phosphorylation of βARs under a series of GPCR agonists. Activation of Gs-coupled dopamine receptor, adenosine receptor, relaxin receptor and prostaglandin E2 receptor, and Gq-coupled α1 adrenergic receptor and angiotensin II type 1 receptor promotes phosphorylation of β1AR and β2AR at putative protein kinase A (PKA) phosphorylation sites; but activation of Gi-coupled α2 adrenergic receptor and activation of protease-activated receptor does not. The GPCR agonists that promote β2AR phosphorylation effectively inhibit βAR agonist isoproterenol-induced PKA phosphorylation of phospholamban and contractile function in ventricular cardiomyocytes. Heterologous GPCR stimuli have minimal to small effect on isoproterenol-induced β2AR activation and G-protein coupling for cyclic adenosine monophosphate (cAMP) production. However, these GPCR stimuli significantly promote phosphorylation of phosphodiesterase 4D (PDE4D), and recruit PDE4D to the phosphorylated β2AR in a β-arrestin 2 dependent manner without promoting β2AR endocytosis. The increased binding between β2AR and PDE4D effectively hydrolyzes cAMP signal generated by subsequent stimulation with isoproterenol. Mutation of PKA phosphorylation sites in β2AR, inhibition of PDE4, or genetic ablation of PDE4D or β-arrestin 2 abolishes this heterologous inhibitory effect. Ablation of β-arrestin 2 or PDE4D gene also rescues β-adrenergic stimuli-induced myocyte contractile function. CONCLUSIONS These data reveal essential roles of β-arrestin 2 and PDE4D in a common mechanism for heterologous desensitization of cardiac βARs under hormonal stimulation, which is associated with impaired cardiac function during the development of pathophysiological conditions.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 4/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism
- Hormones/pharmacology
- Male
- Mice, Knockout
- Myocardial Contraction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Phosphorylation
- Protein Kinase C/metabolism
- Rats
- Receptor Cross-Talk
- Receptors, Adrenergic, beta-1/drug effects
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-1/metabolism
- Receptors, Adrenergic, beta-2/drug effects
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Signal Transduction/drug effects
- Time Factors
- beta-Arrestin 1/genetics
- beta-Arrestin 1/metabolism
- beta-Arrestin 2/genetics
- beta-Arrestin 2/metabolism
Collapse
Affiliation(s)
- Qian Shi
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
| | - Minghui Li
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210001, China
| | - Delphine Mika
- INSERM UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Qin Fu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Technology and Sciences, Wuhan 430030, China
| | - Sungjin Kim
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
| | - Jason Phan
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
| | - Ao Shen
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
| | | | - Yang K. Xiang
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
- VA Northern California Health care system, Mather, CA 95655, USA
| |
Collapse
|
33
|
Rozier K, Bondarenko VE. Distinct physiological effects of β1- and β2-adrenoceptors in mouse ventricular myocytes: insights from a compartmentalized mathematical model. Am J Physiol Cell Physiol 2017; 312:C595-C623. [DOI: 10.1152/ajpcell.00273.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/03/2017] [Accepted: 01/18/2017] [Indexed: 01/08/2023]
Abstract
The β1- and β2-adrenergic signaling systems play different roles in the functioning of cardiac cells. Experimental data show that the activation of the β1-adrenergic signaling system produces significant inotropic, lusitropic, and chronotropic effects in the heart, whereas the effects of the β2-adrenergic signaling system is less apparent. In this paper, a comprehensive compartmentalized experimentally based mathematical model of the combined β1- and β2-adrenergic signaling systems in mouse ventricular myocytes is developed to simulate the experimental findings and make testable predictions of the behavior of the cardiac cells under different physiological conditions. Simulations describe the dynamics of major signaling molecules in different subcellular compartments; kinetics and magnitudes of phosphorylation of ion channels, transporters, and Ca2+ handling proteins; modifications of action potential shape and duration; and [Ca2+]i and [Na+]i dynamics upon stimulation of β1- and β2-adrenergic receptors (β1- and β2-ARs). The model reveals physiological conditions when β2-ARs do not produce significant physiological effects and when their effects can be measured experimentally. Simulations demonstrated that stimulation of β2-ARs with isoproterenol caused a marked increase in the magnitude of the L-type Ca2+ current, [Ca2+]i transient, and phosphorylation of phospholamban only upon additional application of pertussis toxin or inhibition of phosphodiesterases of type 3 and 4. The model also made testable predictions of the changes in magnitudes of [Ca2+]i and [Na+]i fluxes, the rate of decay of [Na+]i concentration upon both combined and separate stimulation of β1- and β2-ARs, and the contribution of phosphorylation of PKA targets to the changes in the action potential and [Ca2+]i transient.
Collapse
Affiliation(s)
- Kelvin Rozier
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia; and
| | - Vladimir E. Bondarenko
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia; and
- Neuroscience Institute, Georgia State University, Atlanta, Georgia
| |
Collapse
|
34
|
Pabbidi MR, Ji X, Maxwell JT, Mignery GA, Samarel AM, Lipsius SL. Inhibition of cAMP-Dependent PKA Activates β2-Adrenergic Receptor Stimulation of Cytosolic Phospholipase A2 via Raf-1/MEK/ERK and IP3-Dependent Ca2+ Signaling in Atrial Myocytes. PLoS One 2016; 11:e0168505. [PMID: 27977772 PMCID: PMC5158063 DOI: 10.1371/journal.pone.0168505] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 12/01/2016] [Indexed: 11/19/2022] Open
Abstract
We previously reported in atrial myocytes that inhibition of cAMP-dependent protein kinase (PKA) by laminin (LMN)-integrin signaling activates β2-adrenergic receptor (β2-AR) stimulation of cytosolic phospholipase A2 (cPLA2). The present study sought to determine the signaling mechanisms by which inhibition of PKA activates β2-AR stimulation of cPLA2. We therefore determined the effects of zinterol (0.1 μM; zint-β2-AR) to stimulate ICa,L in atrial myocytes in the absence (+PKA) and presence (-PKA) of the PKA inhibitor (1 μM) KT5720 and compared these results with atrial myocytes attached to laminin (+LMN). Inhibition of Raf-1 (10 μM GW5074), phospholipase C (PLC; 0.5 μM edelfosine), PKC (4 μM chelerythrine) or IP3 receptor (IP3R) signaling (2 μM 2-APB) significantly inhibited zint-β2-AR stimulation of ICa,L in-PKA but not +PKA myocytes. Western blots showed that zint-β2-AR stimulation increased ERK1/2 phosphorylation in-PKA compared to +PKA myocytes. Adenoviral (Adv) expression of dominant negative (dn) -PKCα, dn-Raf-1 or an IP3 affinity trap, each inhibited zint-β2-AR stimulation of ICa,L in + LMN myocytes compared to control +LMN myocytes infected with Adv-βgal. In +LMN myocytes, zint-β2-AR stimulation of ICa,L was enhanced by adenoviral overexpression of wild-type cPLA2 and inhibited by double dn-cPLA2S505A/S515A mutant compared to control +LMN myocytes infected with Adv-βgal. In-PKA myocytes depletion of intracellular Ca2+ stores by 5 μM thapsigargin failed to inhibit zint-β2-AR stimulation of ICa,L via cPLA2. However, disruption of caveolae formation by 10 mM methyl-β-cyclodextrin inhibited zint-β2-AR stimulation of ICa,L in-PKA myocytes significantly more than in +PKA myocytes. We conclude that inhibition of PKA removes inhibition of Raf-1 and thereby allows β2-AR stimulation to act via PKCα/Raf-1/MEK/ERK1/2 and IP3-mediated Ca2+ signaling to stimulate cPLA2 signaling within caveolae. These findings may be relevant to the remodeling of β-AR signaling in failing and/or aging heart, both of which exhibit decreases in adenylate cyclase activity.
Collapse
MESH Headings
- Animals
- Calcium Signaling/drug effects
- Calcium Signaling/genetics
- Carbazoles/pharmacology
- Cats
- Cells, Cultured
- Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Extracellular Signal-Regulated MAP Kinases/genetics
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Female
- Group IV Phospholipases A2/genetics
- Group IV Phospholipases A2/metabolism
- Heart Atria/cytology
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Male
- Mitogen-Activated Protein Kinase Kinases/genetics
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Proto-Oncogene Proteins c-raf/genetics
- Proto-Oncogene Proteins c-raf/metabolism
- Pyrroles/pharmacology
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Signal Transduction/genetics
- Signal Transduction/physiology
Collapse
Affiliation(s)
- M. R. Pabbidi
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS, United States of America
- * E-mail:
| | - X. Ji
- Department of Physiology, Loyola University Medical Center, Maywood, IL, United States of America
| | - J. T. Maxwell
- Department of Physiology, Loyola University Medical Center, Maywood, IL, United States of America
| | - G. A. Mignery
- Department of Physiology, Loyola University Medical Center, Maywood, IL, United States of America
| | - A. M. Samarel
- Department of Medicine, Loyola University Medical Center, Maywood, IL, United States of America
| | - S. L. Lipsius
- Department of Physiology, Loyola University Medical Center, Maywood, IL, United States of America
| |
Collapse
|
35
|
Khwaja YH, Tai JM. Takotsubo cardiomyopathy with use of salbutamol nebulisation and aminophylline infusion in a patient with acute asthma exacerbation. BMJ Case Rep 2016; 2016:bcr-2016-217364. [PMID: 27793870 DOI: 10.1136/bcr-2016-217364] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Takotsubo cardiomyopathy, apical ballooning syndrome or stress-induced cardiomyopathy is characterised by transient left ventricular dysfunction, mimicking myocardial infarction in the absence of obstructive coronary artery disease or acute plaque rupture on coronary angiography. The exact mechanism of myocardial dysfunction in Takotsubo cardiomyopathy is unknown; however, due to its association with physical and emotional stress, it is postulated that catecholamines play a central role in its pathogenesis. We present a case of a patient who was admitted with acute asthma exacerbation and was treated with β-2 agonist nebulisation and intravenous aminophylline. During her hospital stay she developed Takotsubo cardiomyopathy.
Collapse
Affiliation(s)
| | - Javed Majid Tai
- Department of Medicine, The Aga Khan University Hospital, Karachi, Pakistan
| |
Collapse
|
36
|
Bowman SL, Shiwarski DJ, Puthenveedu MA. Distinct G protein-coupled receptor recycling pathways allow spatial control of downstream G protein signaling. J Cell Biol 2016; 214:797-806. [PMID: 27646272 PMCID: PMC5037407 DOI: 10.1083/jcb.201512068] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 08/12/2016] [Indexed: 12/22/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are recycled via a sequence-dependent pathway that is spatially and biochemically distinct from bulk recycling. Why there are two distinct recycling pathways from the endosome is a fundamental question in cell biology. In this study, we show that the separation of these two pathways is essential for normal spatial encoding of GPCR signaling. The prototypical β-2 adrenergic receptor (B2AR) activates Gα stimulatory protein (Gαs) on the endosome exclusively in sequence-dependent recycling tubules marked by actin/sorting nexin/retromer tubular (ASRT) microdomains. B2AR was detected in an active conformation in bulk recycling tubules, but was unable to activate Gαs. Protein kinase A phosphorylation of B2AR increases the fraction of receptors localized to ASRT domains and biases the downstream transcriptional effects of B2AR to genes controlled by endosomal signals. Our results identify the physiological relevance of separating GPCR recycling from bulk recycling and suggest a mechanism to tune downstream responses of GPCR signaling by manipulating the spatial origin of G protein signaling.
Collapse
Affiliation(s)
- Shanna Lynn Bowman
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | | | | |
Collapse
|
37
|
Adachi N, Hess DT, McLaughlin P, Stamler JS. S-Palmitoylation of a Novel Site in the β2-Adrenergic Receptor Associated with a Novel Intracellular Itinerary. J Biol Chem 2016; 291:20232-46. [PMID: 27481942 DOI: 10.1074/jbc.m116.725762] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Indexed: 01/04/2023] Open
Abstract
We report here that a population of human β2-adrenergic receptors (β2AR), a canonical G protein-coupled receptor, traffics along a previously undescribed intracellular itinerary via the Golgi complex that is associated with the sequential S-palmitoylation and depalmitoylation of a previously undescribed site of modification, Cys-265 within the third intracellular loop. Basal S-palmitoylation of Cys-265 is negligible, but agonist-induced β2AR activation results in enhanced S-palmitoylation, which requires phosphorylation by the cAMP-dependent protein kinase of Ser-261/Ser-262. Agonist-induced turnover of palmitate occurs predominantly on Cys-265. Cys-265 S-palmitoylation is mediated by the Golgi-resident palmitoyl transferases zDHHC9/14/18 and is followed by depalmitoylation by the plasma membrane-localized acyl-protein thioesterase APT1. Inhibition of depalmitoylation reveals that S-palmitoylation of Cys-265 may stabilize the receptor at the plasma membrane. In addition, β2AR S-palmitoylated at Cys-265 are selectively preserved under a sustained adrenergic stimulation, which results in the down-regulation and degradation of βAR. Cys-265 is not conserved in β1AR, and S-palmitoylation of Cys-265 may thus be associated with functional differences between β2AR and β1AR, including relative resistance of β2AR to down-regulation in multiple pathophysiologies. Trafficking via the Golgi complex may underlie new roles in G protein-coupled receptor biology.
Collapse
Affiliation(s)
- Naoko Adachi
- From the Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106, the Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, and
| | - Douglas T Hess
- From the Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106, the Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, and
| | - Precious McLaughlin
- From the Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106, the Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, and
| | - Jonathan S Stamler
- From the Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine and University Hospitals Case Medical Center, Cleveland, Ohio 44106, the Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, and the Harrington Discovery Institute, University Hospitals Case Medical Center, Cleveland, Ohio 44106
| |
Collapse
|
38
|
Huang HW, Zuo C, Chen X, Peng YP, Qiu YH. Effect of tyrosine hydroxylase overexpression in lymphocytes on the differentiation and function of T helper cells. Int J Mol Med 2016; 38:635-42. [PMID: 27315039 DOI: 10.3892/ijmm.2016.2639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 06/03/2016] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to examine the effect of the overexpression of tyrosine hydroxylase (TH), a rate-limiting enzyme for the synthesis of catecholamines (CAs), in lymphocytes on the differentiation and function of T helper (Th) cells. A recombinant TH overexpression plasmid (pEGFP-N1-TH) was constructed and transfected into mesenteric lymphocytes using nucleofection technology. These cells were stimulated with concanavalin A (Con A) for 48 h and then examined for TH expression and CA content, as well as for the percentage of Th1 and Th2 cells, cytokine concentrations and for the levels of signaling molecules. The lymphocytes overexpressing TH also expressed higher mRNA and protein levels of TH, and synthesized more CAs, including norepinephrine (NE), epinephrine (E) and dopamine (DA) than the mock-transfected control cells. TH gene overexpression in the lymphocytes reduced the percentage of interferon-γ (IFN-γ)-producing CD4+ cells and the ratio of CD4+IFN-γ+/CD4+IL-4+ cells, as well as the percentages of CD4+CD26+ and CD4+CD30+ cells and the ratio of CD4+CD26+/CD4+CD30+ cells. TH overexpression also reduced the secretion of IFN-γ and tumor necrosis factor (TNF) from lymphocytes. Moreover, NE inhibited the Con A-induced lymphocyte proliferation and decreased both cyclic adenosine monophosphate (cAMP) levels and p38 mitogen-activated protein kinase (MAPK) expression in the lymphocytes. Our findings thus indicate that TH gene overexpression promotes the polarization and differentiation of CD4+ cells towards Th2 cells, and this effect is mediated by the cAMP and p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Hui-Wei Huang
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Cong Zuo
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiao Chen
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
39
|
Yang Z, Lu ZQ, Zhang YJ, Li YB, Wang ZY, Zhang YL, Zhuang PW, Bai G. Looking for agonists of β2 adrenergic receptor from Fuzi and Chuanwu by virtual screening and dual-luciferase reporter assay. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2016; 18:550-561. [PMID: 26700061 DOI: 10.1080/10286020.2015.1123692] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 11/17/2015] [Indexed: 06/05/2023]
Abstract
More and more studies demonstrated that β2 adrenergic receptor (β2-AR) plays a crucial role for the treatment of heart failure. Chuanwu and Fuzi have been used over thousands of years in China for the treatment of heart failure. Considering the effects of these herbs are very similar to β2-AR agonists, we presume whether β2-AR agonists can be found from Fuzi and Chuanwu. Fuzi and Chuanwu decoction were used to receive the luciferase reporter activity assay to verify the hypothesis, and the result is positive and encouraging. For it is very difficult to get all of the monomer compounds of Fuzi and Chuanwu, virtual screening was used to find potential β2-AR agonists and a cell-based β2-AR agonist functional evaluation model, combined with a luciferase reporter assay system, was used to confirm the final result. In this research, 45 compounds were identified as β2-AR agonists, and four compounds were verified and the rest need further experiment.
Collapse
Affiliation(s)
- Zhen Yang
- a Tianjin State Key Laboratory of Modern Chinese Medicine , Tianjin University of Traditional Chinese Medicine , Tianjin , China
| | - Zhi-Qiang Lu
- a Tianjin State Key Laboratory of Modern Chinese Medicine , Tianjin University of Traditional Chinese Medicine , Tianjin , China
| | - Yan-Jun Zhang
- a Tianjin State Key Laboratory of Modern Chinese Medicine , Tianjin University of Traditional Chinese Medicine , Tianjin , China
| | - Yu-Bo Li
- a Tianjin State Key Laboratory of Modern Chinese Medicine , Tianjin University of Traditional Chinese Medicine , Tianjin , China
| | - Zeng-Yong Wang
- b State Key Laboratory of Medicinal Chemical Biology , Nankai University , Tianjin 300071 , China
| | - Yan-Ling Zhang
- c School of Chinese Pharmacy , Beijing University of Traditional Chinese Medicine , Beijing 100102 , China
| | - Peng-Wei Zhuang
- a Tianjin State Key Laboratory of Modern Chinese Medicine , Tianjin University of Traditional Chinese Medicine , Tianjin , China
- d Tianjin JF-Pharmaland Technology Development Co., Ltd. , Tianjin 300457 , China
| | - Gang Bai
- b State Key Laboratory of Medicinal Chemical Biology , Nankai University , Tianjin 300071 , China
| |
Collapse
|
40
|
Chay A, Zamparo I, Koschinski A, Zaccolo M, Blackwell KT. Control of βAR- and N-methyl-D-aspartate (NMDA) Receptor-Dependent cAMP Dynamics in Hippocampal Neurons. PLoS Comput Biol 2016; 12:e1004735. [PMID: 26901880 PMCID: PMC4763502 DOI: 10.1371/journal.pcbi.1004735] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/05/2016] [Indexed: 11/18/2022] Open
Abstract
Norepinephrine, a neuromodulator that activates β-adrenergic receptors (βARs), facilitates learning and memory as well as the induction of synaptic plasticity in the hippocampus. Several forms of long-term potentiation (LTP) at the Schaffer collateral CA1 synapse require stimulation of both βARs and N-methyl-D-aspartate receptors (NMDARs). To understand the mechanisms mediating the interactions between βAR and NMDAR signaling pathways, we combined FRET imaging of cAMP in hippocampal neuron cultures with spatial mechanistic modeling of signaling pathways in the CA1 pyramidal neuron. Previous work implied that cAMP is synergistically produced in the presence of the βAR agonist isoproterenol and intracellular calcium. In contrast, we show that when application of isoproterenol precedes application of NMDA by several minutes, as is typical of βAR-facilitated LTP experiments, the average amplitude of the cAMP response to NMDA is attenuated compared with the response to NMDA alone. Models simulations suggest that, although the negative feedback loop formed by cAMP, cAMP-dependent protein kinase (PKA), and type 4 phosphodiesterase may be involved in attenuating the cAMP response to NMDA, it is insufficient to explain the range of experimental observations. Instead, attenuation of the cAMP response requires mechanisms upstream of adenylyl cyclase. Our model demonstrates that Gs-to-Gi switching due to PKA phosphorylation of βARs as well as Gi inhibition of type 1 adenylyl cyclase may underlie the experimental observations. This suggests that signaling by β-adrenergic receptors depends on temporal pattern of stimulation, and that switching may represent a novel mechanism for recruiting kinases involved in synaptic plasticity and memory. Noradrenaline is a stress related molecule that facilitates learning and memory when released in the hippocampus. The facilitation of memory is related to modulation of synaptic plasticity, but the mechanisms underlying this modulation are not well understood. We utilize a combination of live cell imaging and computational modeling to discover how noradrenergic receptor stimulation interacts with other molecules, such as calcium, required for synaptic plasticity and memory storage. Though prior work has shown that noradrenergic receptors and calcium interact synergistically to elevate intracellular second messengers when combined simultaneously, our results demonstrate that prior stimulation of noradrenergic receptors inhibits the elevation of intracellular second messengers. Our results further demonstrate that the inhibition may be caused by the noradrenergic receptor switching signaling pathways, thereby recruiting a different set of memory kinases. This switching represents a novel mechanism for recruiting molecules involved in synaptic plasticity and memory.
Collapse
Affiliation(s)
- Andrew Chay
- Molecular Neuroscience Department, Krasnow Institute, George Mason University, Fairfax, Virginia, United States of America
| | | | - Andreas Koschinski
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, United Kingdom
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, United Kingdom
| | - Kim T. Blackwell
- Molecular Neuroscience Department, Krasnow Institute, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
41
|
Black JB, Premont RT, Daaka Y. Feedback regulation of G protein-coupled receptor signaling by GRKs and arrestins. Semin Cell Dev Biol 2016; 50:95-104. [PMID: 26773211 PMCID: PMC4779377 DOI: 10.1016/j.semcdb.2015.12.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 12/19/2015] [Indexed: 12/16/2022]
Abstract
GPCRs are ubiquitous in mammalian cells and present intricate mechanisms for cellular signaling and communication. Mechanistically, GPCR signaling was identified to occur vectorially through heterotrimeric G proteins that are negatively regulated by GRK and arrestin effectors. Emerging evidence highlights additional roles for GRK and Arrestin partners, and establishes the existence of interconnected feedback pathways that collectively define GPCR signaling. GPCRs influence cellular dynamics and can mediate pathologic development, such as cancer and cardiovascular remolding. Hence, a better understanding of their overall signal regulation is of great translational interest and research continues to exploit the pharmacologic potential for modulating their activity.
Collapse
Affiliation(s)
- Joseph B Black
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Richard T Premont
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Yehia Daaka
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, United States.
| |
Collapse
|
42
|
Chakrabarti S, Madia PA, Gintzler AR. Selective up-regulation of functional mu-opioid receptor splice variants by chronic opioids. J Neurochem 2016; 136:1119-1130. [PMID: 26718622 DOI: 10.1111/jnc.13519] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/15/2015] [Accepted: 12/18/2015] [Indexed: 11/28/2022]
Abstract
We recently reported (Verzillo, et al. J. Neurochem: 130, 790-796, 2014) that chronic systemic morphine selectively up-regulates mRNA encoding two C-terminal μ-opioid receptor (MOR) splice variants, MOR-1C1 and MOR-1B2 (MOR-1B2/-1C1). Given the known disconnects between changes in levels of mRNA and corresponding protein, it is essential to directly demonstrate that chronic opioid treatment elevates functional MOR-1B2/-1C1 protein prior to inferring relevance of these MOR variants to spinal opioid tolerance mechanisms. Accordingly, we investigated the ability of chronic opioid exposure to up-regulate MOR protein in Chinese hamster ovary cells stably transfected with rat MOR variants MOR-1B2, MOR-1C1, or MOR-1 (considered to be the predominant MOR). Findings revealed that chronic treatment with the clinically relevant opioids morphine, oxycodone and hydrocodone substantially up-regulated membrane MOR-1B2/-1C1 protein. This up-regulation was abolished by the protein synthesis inhibitor cycloheximide, eliminating contributions from receptor redistribution. The increment in MOR-1B2/-1C1 protein was paralleled by a significant increment in opioid agonist-stimulated GTPγS-binding (reflective of increased aggregate MOR G protein coupling) indicating that up-regulated MOR-1B2/-1C1 represented functional receptors. Strikingly, these tolerance-associated adaptations of MOR-1B2/-1C1 differed considerably from those of MOR-1. Antithetical regulation of MOR-1B2/-1C1 and MOR-1 by chronic opioids has significant implications for the design of new therapeutic agents to counteract opioid analgesic tolerance and accompanying addiction. Since chronic opioids induce MOR-1B2/-1C1 up-regulation in spinal cord of males, but not females, elucidating cellular compartments and intracellular pathways mediating MOR-1B2/-1C1 up-regulation and defining their unique signaling attributes would enable a precision medicinal approach to pain management and addiction therapy. In the spinal cord of males, but not females, chronic morphine up-regulates mRNA encoding two mu-opioid receptor (MOR) variants, MOR-1B2 and MOR-1C1 (MOR-1B2/-1C1). We now demonstrate that chronic treatment with the clinically relevant opioids morphine, hydrocodone or oxycodone up-regulates MOR-1B2/-1C1 functional protein, which is dependent on de novo protein synthesis. Findings underscore the importance of unique signaling attributes of MOR variants to sexually dimorphic tolerance mechanisms.
Collapse
Affiliation(s)
- Sumita Chakrabarti
- Department of Obstetrics and Gynecology, State University of New York, Brooklyn, New York, USA
| | - Priyanka A Madia
- Department of Obstetrics and Gynecology, State University of New York, Brooklyn, New York, USA
| | - Alan R Gintzler
- Department of Obstetrics and Gynecology, State University of New York, Brooklyn, New York, USA
| |
Collapse
|
43
|
Biased signalling: the instinctive skill of the cell in the selection of appropriate signalling pathways. Biochem J 2015; 470:155-67. [DOI: 10.1042/bj20150358] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
GPCRs (G-protein-coupled receptors) are members of a family of proteins which are generally regarded as the largest group of therapeutic drug targets. Ligands of GPCRs do not usually activate all cellular signalling pathways linked to a particular seven-transmembrane receptor in a uniform manner. The fundamental idea behind this concept is that each ligand has its own ability, while interacting with the receptor, to activate different signalling pathways (or a particular set of signalling pathways) and it is this concept which is known as biased signalling. The importance of biased signalling is that it may selectively activate biological responses to favour therapeutically beneficial signalling pathways and to avoid adverse effects. There are two levels of biased signalling. First, bias can arise from the ability of GPCRs to couple to a subset of the available G-protein subtypes: Gαs, Gαq/11, Gαi/o or Gα12/13. These subtypes produce the diverse effects of GPCRs by targeting different effectors. Secondly, biased GPCRs may differentially activate G-proteins or β-arrestins. β-Arrestins are ubiquitously expressed and function to terminate or inhibit classic G-protein signalling and initiate distinct β-arrestin-mediated signalling processes. The interplay of G-protein and β-arrestin signalling largely determines the cellular consequences of the administration of GPCR-targeted drugs. In the present review, we highlight the particular functionalities of biased signalling and discuss its biological effects subsequent to GPCR activation. We consider that biased signalling is potentially allowing a choice between signalling through ‘beneficial’ pathways and the avoidance of ‘harmful’ ones.
Collapse
|
44
|
Cao X, Zhou C, Chong J, Fu L, Zhang L, Sun D, Hou H, Zhang Y, Li D, Sun H. Estrogen resisted stress-induced cardiomyopathy through increasing the activity of β2AR–Gαs signal pathway in female rats. Int J Cardiol 2015; 187:377-86. [DOI: 10.1016/j.ijcard.2015.02.113] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 02/16/2015] [Accepted: 02/21/2015] [Indexed: 02/08/2023]
|
45
|
Trafficking of β-Adrenergic Receptors: Implications in Intracellular Receptor Signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:151-88. [PMID: 26055058 DOI: 10.1016/bs.pmbts.2015.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
β-Adrenergic receptors (βARs), prototypical G-protein-coupled receptors, play a pivotal role in regulating neuronal and cardiovascular responses to catecholamines during stress. Agonist-induced receptor endocytosis is traditionally considered as a primary mechanism to turn off the receptor signaling (or receptor desensitization). However, recent progress suggests that intracellular trafficking of βAR presents a mean to translocate receptor signaling machinery to intracellular organelles/compartments while terminating the signaling at the cell surface. Moreover, the apparent multidimensionality of ligand efficacy in space and time in a cell has forecasted exciting pathophysiological implications, which are just beginning to be explored. As we begin to understand how these pathways impact downstream cellular programs, this will have significant implications for a number of pathophysiological conditions in heart and other systems, that in turn open up new therapeutic opportunities.
Collapse
|
46
|
Jiang L, Chen SH, Chu CH, Wang SJ, Oyarzabal E, Wilson B, Sanders V, Xie K, Wang Q, Hong JS. A novel role of microglial NADPH oxidase in mediating extra-synaptic function of norepinephrine in regulating brain immune homeostasis. Glia 2015; 63:1057-72. [PMID: 25740080 DOI: 10.1002/glia.22801] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 01/15/2015] [Indexed: 12/30/2022]
Abstract
Although the peripheral anti-inflammatory effect of norepinephrine (NE) is well documented, the mechanism by which this neurotransmitter functions as an anti-inflammatory/neuroprotective agent in the central nervous system (CNS) is unclear. This article aimed to determine the anti-inflammatory/neuroprotective effects and underlying mechanisms of NE in inflammation-based dopaminergic neurotoxicity models. In mice, NE-depleting toxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) was injected at 6 months of lipopolysaccharide (LPS)-induced neuroinflammation. It was found that NE depletion enhanced LPS-induced dopaminergic neuron loss in the substantia nigra. This piece of in vivo data prompted us to conduct a series of studies in an effort to elucidate the mechanism as to how NE affects dopamine neuron survival by using primary midbrain neuron/glia cultures. Results showed that submicromolar concentrations of NE dose-dependently protected dopaminergic neurons from LPS-induced neurotoxicity by inhibiting microglia activation and subsequent release of pro-inflammatory factors. However, NE-elicited neuroprotection was not totally abolished in cultures from β2-adrenergic receptor (β2-AR)-deficient mice, suggesting that novel pathways other than β2-AR are involved. To this end, It was found that submicromolar NE dose-dependently inhibited NADPH oxidase (NOX2)-generated superoxide, which contributes to the anti-inflammatory and neuroprotective effects of NE. This novel mechanism was indeed adrenergic receptors independent since both (+) and (-) optic isomers of NE displayed the same potency. We further demonstrated that NE inhibited LPS-induced NOX2 activation by blocking the translocation of its cytosolic subunit to plasma membranes. In summary, we revealed a potential physiological role of NE in maintaining brain immune homeostasis and protecting neurons via a novel mechanism.
Collapse
Affiliation(s)
- Lulu Jiang
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China; Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, 27709
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wypijewski KJ, Tinti M, Chen W, Lamont D, Ashford MLJ, Calaghan SC, Fuller W. Identification of caveolar resident proteins in ventricular myocytes using a quantitative proteomic approach: dynamic changes in caveolar composition following adrenoceptor activation. Mol Cell Proteomics 2015; 14:596-608. [PMID: 25561500 PMCID: PMC4349980 DOI: 10.1074/mcp.m114.038570] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The lipid raft concept proposes that membrane environments enriched in cholesterol and sphingolipids cluster certain proteins and form platforms to integrate cell signaling. In cardiac muscle, caveolae concentrate signaling molecules and ion transporters, and play a vital role in adrenergic regulation of excitation–contraction coupling, and consequently cardiac contractility. Proteomic analysis of cardiac caveolae is hampered by the presence of contaminants that have sometimes, erroneously, been proposed to be resident in these domains. Here we present the first unbiased analysis of the proteome of cardiac caveolae, and investigate dynamic changes in their protein constituents following adrenoreceptor (AR) stimulation. Rat ventricular myocytes were treated with methyl-β-cyclodextrin (MβCD) to deplete cholesterol and disrupt caveolae. Buoyant caveolin-enriched microdomains (BCEMs) were prepared from MβCD-treated and control cell lysates using a standard discontinuous sucrose gradient. BCEMs were harvested, pelleted, and resolubilized, then alkylated, digested, and labeled with iTRAQ reagents, and proteins identified by LC-MS/MS on a LTQ Orbitrap Velos Pro. Proteins were defined as BCEM resident if they were consistently depleted from the BCEM fraction following MβCD treatment. Selective activation of α-, β1-, and β2-AR prior to preparation of BCEMs was achieved by application of agonist/antagonist pairs for 10 min in populations of field-stimulated myocytes. We typically identified 600–850 proteins per experiment, of which, 249 were defined as high-confidence BCEM residents. Functional annotation clustering indicates cardiac BCEMs are enriched in integrin signaling, guanine nucleotide binding, ion transport, and insulin signaling clusters. Proteins possessing a caveolin binding motif were poorly enriched in BCEMs, suggesting this is not the only mechanism that targets proteins to caveolae. With the notable exception of the cavin family, very few proteins show altered abundance in BCEMs following AR activation, suggesting signaling complexes are preformed in BCEMs to ensure a rapid and high fidelity response to adrenergic stimulation in cardiac muscle.
Collapse
Affiliation(s)
- Krzysztof J Wypijewski
- From the ‡Division of Cardiovascular and Diabetes Medicine, College of Medicine, Dentistry and Nursing, University of Dundee, Dundee, United Kingdom
| | - Michele Tinti
- §College of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Wenzhang Chen
- §College of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Douglas Lamont
- §College of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Michael L J Ashford
- From the ‡Division of Cardiovascular and Diabetes Medicine, College of Medicine, Dentistry and Nursing, University of Dundee, Dundee, United Kingdom
| | - Sarah C Calaghan
- ¶School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - William Fuller
- From the ‡Division of Cardiovascular and Diabetes Medicine, College of Medicine, Dentistry and Nursing, University of Dundee, Dundee, United Kingdom;
| |
Collapse
|
48
|
Abstract
The interaction between the sympathetic nervous system and the immune system has been documented over the last several decades. In this review, the neuroanatomical, cellular, and molecular evidence for neuroimmune regulation in the maintenance of immune homeostasis will be discussed, as well as the potential impact of neuroimmune dysregulation in health and disease.
Collapse
Affiliation(s)
- Caroline J Padro
- The Biomedical Sciences Graduate Program, The Ohio State University Wexner College of Medicine, Columbus, OH 43210, United States.
| | - Virginia M Sanders
- The Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University Wexner College of Medicine, Columbus, OH 43210, United States; The Institute of Behavioral Medicine Research, The Ohio State University Wexner College of Medicine, Columbus, OH 43210, United States.
| |
Collapse
|
49
|
|
50
|
Verzillo V, Madia PA, Liu NJ, Chakrabarti S, Gintzler AR. Mu-opioid receptor splice variants: sex-dependent regulation by chronic morphine. J Neurochem 2014; 130:790-6. [PMID: 24848866 DOI: 10.1111/jnc.12768] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/06/2014] [Accepted: 05/20/2014] [Indexed: 11/29/2022]
Abstract
The gene encoding the mu-opioid receptor (MOR) generates a remarkable diversity of subtypes, the functional significance of which remains largely unknown. The structure of MOR could be a critical determinant of MOR functionality and its adaptations to chronic morphine exposure. As MOR antinociception has sexually dimorphic dimensions, we determined the influence of sex, stage of estrus cycle, and chronic systemic morphine on levels of MOR splice variant mRNA in rat spinal cord. Chronic systemic morphine influenced the spinal expression of mRNA encoding rMOR-1B2 and rMOR-1C1 in a profoundly sex-dependent fashion. In males, chronic morphine resulted in a twofold increase in expression levels of rMOR-1B2 and rMOR-1C1 mRNA. This effect of chronic morphine was completely absent in females. Increased density of MOR protein in spinal cord of males accompanied the chronic morphine-induced increase in MOR variant mRNA, suggesting that it reflected an increase in corresponding receptor protein. These results suggest that tolerance/dependence results, at least in part, from different adaptational strategies in males and females. The signaling consequences of the unique composition of the C-terminus tip of rMOR-1C1 and rMOR-1B2 could point the way to defining the molecular components of sex-dependent tolerance and withdrawal mechanisms. Chronic systemic morphine increases levels of mRNA encoding two splice variants of mu-opioid receptor (MOR), MOR-1B2 and MOR-1C1, variants differing from rMOR-1 in their C-terminal (and phosphorylation sites therein) and thus possibly signaling sequelae. This adaptation is sex-specific. It occurs in the spinal cord of males, but not females, indicating the importance of sex-specific mechanisms for and treatments of tolerance and addiction.
Collapse
Affiliation(s)
- Vittorio Verzillo
- Department of Obstetrics and Gynecology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| | | | | | | | | |
Collapse
|