1
|
Jara ZP, Harford T, Singh KD, Desnoyer R, Kumar A, Srinivasan D, Karnik SS. Distinct Mechanisms of β-Arrestin-Biased Agonist and Blocker of AT1R in Preventing Aortic Aneurysm and Associated Mortality. Hypertension 2023; 80:385-402. [PMID: 36440576 PMCID: PMC9852074 DOI: 10.1161/hypertensionaha.122.19232] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 11/04/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Aortic aneurysm (AA) is a "silent killer" human disease with no effective treatment. Although the therapeutic potential of various pharmacological agents have been evaluated, there are no reports of β-arrestin-biased AT1R (angiotensin-II type-1 receptor) agonist (TRV027) used to prevent the progression of AA. METHODS We tested the hypothesis that TRV027 infusion in AngII (angiotensin II)-induced mouse model of AA prevents AA. High-fat-diet-fed ApoE (apolipoprotein E gene)-null mice were infused with AngII to induce AA and co-infused with TRV027 and a clinically used AT1R blocker Olmesartan to prevent AA. Aortas explanted from different ligand infusion groups were compared with assess different grades of AA or lack of AA. RESULTS AngII produced AA in ≈67% male mice with significant mortality associated with AA rupture. We observed ≈13% mortality due to aortic arch dissection without aneurysm in male mice. AngII-induced AA and mortality was prevented by co-infusion of TRV027 or Olmesartan, but through different mechanisms. In TRV027 co-infused mice aortic wall thickness, elastin content, new DNA, and protein synthesis were higher than untreated and Olmesartan co-infused mice. Co-infusion with both TRV027 and Olmesartan prevented endoplasmic reticulum stress, fibrosis, and vasomotor hyper responsiveness. CONCLUSIONS TRV027-engaged AT1R prevented AA and associated mortality by distinct molecular mechanisms compared with the AT1R blocker, Olmesartan. Developing novel β-arrestin-biased AT1R ligands may yield promising drugs to combat AA.
Collapse
Affiliation(s)
- Zaira Palomino Jara
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic
| | - Terri Harford
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic
| | | | - Russell Desnoyer
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic
| | - Avinash Kumar
- Pathobiology Department, Lerner Research Institute, Cleveland Clinic
| | | | - Sadashiva S. Karnik
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic
| |
Collapse
|
2
|
Maudsley S, Walter D, Schrauwen C, Van Loon N, Harputluoğlu İ, Lenaerts J, McDonald P. Intersection of the Orphan G Protein-Coupled Receptor, GPR19, with the Aging Process. Int J Mol Sci 2022; 23:ijms232113598. [PMID: 36362387 PMCID: PMC9653598 DOI: 10.3390/ijms232113598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
G protein-coupled receptors (GPCRs) represent one of the most functionally diverse classes of transmembrane proteins. GPCRs and their associated signaling systems have been linked to nearly every physiological process. They also constitute nearly 40% of the current pharmacopeia as direct targets of remedial therapies. Hence, their place as a functional nexus in the interface between physiological and pathophysiological processes suggests that GPCRs may play a central role in the generation of nearly all types of human disease. Perhaps one mechanism through which GPCRs can mediate this pivotal function is through the control of the molecular aging process. It is now appreciated that, indeed, many human disorders/diseases are induced by GPCR signaling processes linked to pathological aging. Here we discuss one such novel member of the GPCR family, GPR19, that may represent an important new target for novel remedial strategies for the aging process. The molecular signaling pathways (metabolic control, circadian rhythm regulation and stress responsiveness) associated with this recently characterized receptor suggest an important role in aging-related disease etiology.
Collapse
Affiliation(s)
- Stuart Maudsley
- Receptor Biology Lab, University of Antwerp, 2610 Antwerpen, Belgium
- Correspondence:
| | - Deborah Walter
- Receptor Biology Lab, University of Antwerp, 2610 Antwerpen, Belgium
| | - Claudia Schrauwen
- Receptor Biology Lab, University of Antwerp, 2610 Antwerpen, Belgium
| | - Nore Van Loon
- Receptor Biology Lab, University of Antwerp, 2610 Antwerpen, Belgium
| | - İrem Harputluoğlu
- Receptor Biology Lab, University of Antwerp, 2610 Antwerpen, Belgium
| | - Julia Lenaerts
- Receptor Biology Lab, University of Antwerp, 2610 Antwerpen, Belgium
| | | |
Collapse
|
3
|
Zhang X, Zhou W, Niu Y, Zhu S, Zhang Y, Li X, Yu C. Lysyl oxidase promotes renal fibrosis via accelerating collagen cross-link driving by β-arrestin/ERK/STAT3 pathway. FASEB J 2022; 36:e22427. [PMID: 35792886 PMCID: PMC9544652 DOI: 10.1096/fj.202200573r] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/25/2022] [Accepted: 06/10/2022] [Indexed: 11/27/2022]
Abstract
Lysyl oxidase (LOX) is a copper‐dependent monoamine oxidase whose primary function is the covalent cross‐linking of collagen in the extracellular matrix (ECM). Evidence has shown that LOX is associated with cancer and some fibrotic conditions. We recently found that serum LOX is a potential diagnostic biomarker for renal fibrosis, but the mechanism by which LOX is regulated and contributes to renal fibrosis remains unknown. The current study demonstrates the following: (1) LOX expression was increased in fibrotic kidneys including ischemia‐reperfusion injury‐(IRI‐), unilateral ureteral obstruction‐(UUO‐), and folic acid‐ (FA‐) induced fibrotic kidneys as well as in the paraffin‐embedded sections of human kidneys from the patients with renal fibrosis. (2) The increasing deposition and cross‐linking of collagen induced by LOX was observed in IRI‐, UUO‐ and FA‐kidneys. (3) LOX was regulated by the β‐arrestin‐ERK‐STAT3 pathway in renal fibrosis. STAT3 was the downstream of AT1R‐β‐arrestin‐ERK, ERK entered the nucleus and activated STAT3‐pY705 but not STAT3‐pS727. (4) STAT3 nuclear subtranslocation and binding to the LOX promoter may be responsible for the upregulation of LOX expression. (5) Pharmacologic inhibition of LOX with BAPN in vivo inhibited the upregulation of LOX, decreased collagen over cross‐linking and ameliorated renal fibrosis after ischemic injury. Collectively, these observations suggest that LOX plays an essential role in the development of renal fibrosis by catalyzing collagen over cross‐linking. Thus, strategies targeting LOX could be a new avenue in developing therapeutics against renal fibrosis.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenqian Zhou
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yangyang Niu
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Saiya Zhu
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yingying Zhang
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Dpartment of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Chen Yu
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
The CXCL12/CXCR4/ACKR3 Signaling Axis Regulates PKM2 and Glycolysis. Cells 2022; 11:cells11111775. [PMID: 35681470 PMCID: PMC9179862 DOI: 10.3390/cells11111775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/13/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023] Open
Abstract
In response to CXCL12, CXCR4 and ACKR3 both recruit β-arrestin 2, regulating the assembly of interacting proteins that drive signaling and contribute to the functions of both receptors in cancer and multiple other diseases. A prior proteomics study revealed that β-arrestin 2 scaffolds pyruvate kinase M2 (PKM2), an enzyme implicated in shifting cells to glycolytic metabolism and poor prognosis in cancer. We hypothesized that CXCL12 signaling regulates PKM2 protein interactions, oligomerization, and glucose metabolism. We used luciferase complementation in cell-based assays and a tumor xenograft model of breast cancer in NSG mice to quantify how CXCR4 and ACKR3 change protein interactions in the β-arrestin-ERK-PKM2 pathway. We also used mass spectrometry to analyze the effects of CXCL12 on glucose metabolism. CXCL12 signaling through CXCR4 and ACKR3 stimulated protein interactions among β-arrestin 2, PKM2, ERK2, and each receptor, leading to the dissociation of PKM2 from β-arrestin 2. The activation of both receptors reduced the oligomerization of PKM2, reflecting a shift from tetramers to dimers or monomers with low enzymatic activity. Mass spectrometry with isotopically labeled glucose showed that CXCL12 signaling increased intermediate metabolites in glycolysis and the pentose phosphate pathway, with ACKR3 mediating greater effects. These data establish how CXCL12 signaling regulates PKM2 and reprograms cellular metabolism.
Collapse
|
5
|
Reiter E. [β-arrestins, their mechanisms of action and multiple roles in the biology of G protein-coupled receptors]. Biol Aujourdhui 2022; 215:107-118. [PMID: 35275055 DOI: 10.1051/jbio/2021010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Indexed: 06/14/2023]
Abstract
The stimulation of G protein-coupled receptors (GPCRs) induces biological responses to a wide range of extracellular cues. The heterotrimeric G proteins, which are recruited to the active conformation of GPCRs, lead to the generation of various diffusible second messengers. Only two other families of proteins exhibit the remarkable characteristic of recognizing and binding to the active conformation of most GPCRs: GPCR kinases (GRKs) and β-arrestins. These two families of proteins were initially identified as key players in the desensitization of G protein activation by GPCRs. Over the years, β-arrestins have been implicated in an increasing number of interactions with non-receptor proteins, expanding the range of cellular functions in which they are involved. It is now well established that β-arrestins, by scaffolding and recruiting protein complexes in an agonist-dependent manner, directly regulate the trafficking and signaling of GPCRs. Remarkable advances have been made in recent years which have made it possible i) to identify biased ligands capable, by stabilizing particular conformations of a growing number of GPCRs, of activating or blocking the action of β-arrestins independently of that of G proteins, some of these ligands holding great therapeutic interest; ii) to demonstrate β-arrestins' role in the compartmentalization of GPCR signaling within the cell, and iii) to understand the molecular details of their interaction with GPCRs and of their activation through structural and biophysical approaches.
Collapse
Affiliation(s)
- Eric Reiter
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France - Inria, Centre de recherche Inria Saclay-Île-de-France, 91120 Palaiseau, France
| |
Collapse
|
6
|
Lino CA, Barreto-Chaves ML. Beta-arrestins in the context of cardiovascular diseases: Focusing on type 1 angiotensin II receptor (AT1R). Cell Signal 2022; 92:110253. [DOI: 10.1016/j.cellsig.2022.110253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/16/2022]
|
7
|
Sevastre AS, Costachi A, Tataranu LG, Brandusa C, Artene SA, Stovicek O, Alexandru O, Danoiu S, Sfredel V, Dricu A. Glioblastoma pharmacotherapy: A multifaceted perspective of conventional and emerging treatments (Review). Exp Ther Med 2021; 22:1408. [PMID: 34676001 PMCID: PMC8524703 DOI: 10.3892/etm.2021.10844] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022] Open
Abstract
Due to its localisation, rapid onset, high relapse rate and resistance to most currently available treatment methods, glioblastoma multiforme (GBM) is considered to be the deadliest type of all gliomas. Although surgical resection, chemotherapy and radiotherapy are among the therapeutic strategies used for the treatment of GBM, the survival rates achieved are not satisfactory, and there is an urgent need for novel effective therapeutic options. In addition to single-target therapy, multi-target therapies are currently under development. Furthermore, drugs are being optimised to improve their ability to cross the blood-brain barrier. In the present review, the main strategies applied for GBM treatment in terms of the most recent therapeutic agents and approaches that are currently under pre-clinical and clinical testing were discussed. In addition, the most recently reported experimental data following the testing of novel therapies, including stem cell therapy, immunotherapy, gene therapy, genomic correction and precision medicine, were reviewed, and their advantages and drawbacks were also summarised.
Collapse
Affiliation(s)
- Ani-Simona Sevastre
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Alexandra Costachi
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ligia Gabriela Tataranu
- Department of Neurosurgery, ‘Bagdasar-Arseni’ Emergency Clinical Hospital, 041915 Bucharest, Romania
| | - Corina Brandusa
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Stefan Alexandru Artene
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Olivian Stovicek
- Department of Pharmacology, Faculty of Nursing Targu Jiu, Titu Maiorescu University of Bucharest, 210106 Targu Jiu, Romania
| | - Oana Alexandru
- Department of Neurology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Suzana Danoiu
- Department of Pathophysiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Veronica Sfredel
- Department of Physiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Anica Dricu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
8
|
Lino CA, de Bortoli Teixeira L, Capelupe Simões S, de Oliveira Silva T, Diniz GP, da Costa-Neto CM, Barreto-Chaves MLM. Beta-arrestin 2 mediates cardiac hypertrophy induced by thyroid hormones via AT1R. J Cell Physiol 2021; 236:4640-4654. [PMID: 33345322 DOI: 10.1002/jcp.30187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 12/30/2022]
Abstract
We have previously reported that angiotensin II receptor type 1 (AT1R) contributes to the hypertrophic effects of thyroid hormones (TH) in cardiac cells. Even though evidence indicates crosstalks between TH and AT1R, the underlying mechanisms are poorly understood. Beta-arrestin (ARRB) signaling has been described as noncanonical signal transduction pathway that exerts important effects in the cardiovascular system through G-protein-coupled receptors, as AT1R. Herein, we investigated the contribution of ARRB signaling in TH-induced cardiomyocyte hypertrophy. Primary cardiomyocyte cultures were treated with Triiodothyronine (T3) to induce cell hypertrophy. T3 rapidly activates extracellular signal-regulated kinase 1/2 (ERK1/2) signaling, which was partially inhibited by AT1R blockade. Also, ERK1/2 inhibition attenuated the hypertrophic effects of T3. ARRB2 was upregulated by T3, and small interfering RNA assays revealed the role of ARRB2-but not ARRB1-on ERK1/2 activation and cardiomyocyte hypertrophy. Corroborating these findings, the ARRB2-overexpressed cells showed increased expression of hypertrophic markers, which were attenuated by ERK1/2 inhibition. Immunocytochemistry and immunoprecipitation assays revealed the increased expression of nuclear AT1R after T3 stimulation and the increased interaction of AT1R/ARRB2. The inhibition of endocytosis also attenuated the T3 effects on cardiac cells. Our results evidence the contribution of ARRB2 on ERK1/2 activation and cardiomyocyte hypertrophy induced by T3 via AT1R.
Collapse
Affiliation(s)
- Caroline Antunes Lino
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Larissa de Bortoli Teixeira
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Sarah Capelupe Simões
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Gabriela Placoná Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Claudio Miguel da Costa-Neto
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | | |
Collapse
|
9
|
van Gastel J, Leysen H, Boddaert J, Vangenechten L, Luttrell LM, Martin B, Maudsley S. Aging-related modifications to G protein-coupled receptor signaling diversity. Pharmacol Ther 2020; 223:107793. [PMID: 33316288 DOI: 10.1016/j.pharmthera.2020.107793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Aging is a highly complex molecular process, affecting nearly all tissue systems in humans and is the highest risk factor in developing neurodegenerative disorders such as Alzheimer's and Parkinson's disease, cardiovascular disease and Type 2 diabetes mellitus. The intense complexity of the aging process creates an incentive to develop more specific drugs that attenuate or even reverse some of the features of premature aging. As our current pharmacopeia is dominated by therapeutics that target members of the G protein-coupled receptor (GPCR) superfamily it may be prudent to search for effective anti-aging therapeutics in this fertile domain. Since the first demonstration of GPCR-based β-arrestin signaling, it has become clear that an enhanced appreciation of GPCR signaling diversity may facilitate the creation of therapeutics with selective signaling activities. Such 'biased' ligand signaling profiles can be effectively investigated using both standard molecular biological techniques as well as high-dimensionality data analyses. Through a more nuanced appreciation of the quantitative nature across the multiple dimensions of signaling bias that drugs possess, researchers may be able to further refine the efficacy of GPCR modulators to impact the complex aberrations that constitute the aging process. Identifying novel effector profiles could expand the effective pharmacopeia and assist in the design of precision medicines. This review discusses potential non-G protein effectors, and specifically their potential therapeutic suitability in aging and age-related disorders.
Collapse
Affiliation(s)
- Jaana van Gastel
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Hanne Leysen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Jan Boddaert
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, Antwerp, Belgium
| | - Laura Vangenechten
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Louis M Luttrell
- Division of Endocrinology, Diabetes & Medical Genetics, Medical University of South Carolina, USA
| | - Bronwen Martin
- Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
10
|
Abstract
GPCRs (G-protein [guanine nucleotide-binding protein]-coupled receptors) play a central physiological role in the regulation of cardiac function in both health and disease and thus represent one of the largest class of surface receptors targeted by drugs. Several antagonists of GPCRs, such as βARs (β-adrenergic receptors) and Ang II (angiotensin II) receptors, are now considered standard of therapy for a wide range of cardiovascular disease, such as hypertension, coronary artery disease, and heart failure. Although the mechanism of action for GPCRs was thought to be largely worked out in the 80s and 90s, recent discoveries have brought to the fore new and previously unappreciated mechanisms for GPCR activation and subsequent downstream signaling. In this review, we focus on GPCRs most relevant to the cardiovascular system and discuss traditional components of GPCR signaling and highlight evolving concepts in the field, such as ligand bias, β-arrestin-mediated signaling, and conformational heterogeneity.
Collapse
Affiliation(s)
- Jialu Wang
- From the Department of Medicine (J.W., C.G., H.A.R.)
| | | | - Howard A Rockman
- From the Department of Medicine (J.W., C.G., H.A.R.).,Department of Cell Biology (H.A.R.).,Department of Molecular Genetics and Microbiology (H.A.R.), Duke University Medical Center, Durham, NC
| |
Collapse
|
11
|
Nealon CM, Henderson-Redmond AN, Hale DE, Morgan DJ. Tolerance to WIN55,212-2 is delayed in desensitization-resistant S426A/S430A mice. Neuropharmacology 2019; 148:151-159. [PMID: 30629988 PMCID: PMC6535342 DOI: 10.1016/j.neuropharm.2018.12.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 12/05/2018] [Accepted: 12/21/2018] [Indexed: 02/08/2023]
Abstract
Tolerance to cannabinoid agonists can develop through desensitization of the cannabinoid receptor 1 (CB1) following prolonged administration. Desensitization results from phosphorylation of CB1 by a G protein-coupled receptor kinase (GRK), and subsequent association of the receptor with arrestin. Mice expressing a mutant form of CB1, in which the serine residues at two putative phosphorylation sites necessary for desensitization have been replaced by non-phosphorylatable alanines (S426A/S430A), display reduced tolerance to Δ9-tetrahydrocannabinol (Δ9-THC). Tolerance to the antinociceptive effects of WIN55,212-2 was delayed in S426A/S430A mutants using the tail-flick and formalin tests. However, tolerance to the antinociceptive effects of once daily CP55,940 injections was not significantly delayed in S426A/S430A mutant mice using either of these tests. Interestingly, the dose response curve shifts for the hypothermic and antinociceptive effects of CP55,940 that were induced by chronic treatment with this agonist in wild-type mice were blocked in S426A/S430A mutant mice. Assessment of mechanical allodynia in mice exhibiting chronic cisplatin-evoked neuropathic pain found that tolerance to the anti-allodynic effects WIN55,212-2 but not CP55,940 was delayed in S426A/S430A mice compared to wild-type littermates. Despite these deficits in tolerance, S426A/S430A mutant mice eventually developed tolerance to both WIN55,212-2 and CP55,940 for all pain assays that were examined, suggesting that other mechanisms likely contribute to tolerance for these cannabinoid agonists. These findings suggest that GRK- and βarrestin2-mediated desensitization of CB1 may strongly contribute to the rate of tolerance to the antinociceptive effects of WIN55,212-2, and raises the possibility of agonist-specific mechanisms of cannabinoid tolerance.
Collapse
Affiliation(s)
- Caitlin M Nealon
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Angela N Henderson-Redmond
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - David E Hale
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Daniel J Morgan
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA; Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, 17033, USA; Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
12
|
|
13
|
Turu G, Balla A, Hunyady L. The Role of β-Arrestin Proteins in Organization of Signaling and Regulation of the AT1 Angiotensin Receptor. Front Endocrinol (Lausanne) 2019; 10:519. [PMID: 31447777 PMCID: PMC6691095 DOI: 10.3389/fendo.2019.00519] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/15/2019] [Indexed: 12/30/2022] Open
Abstract
AT1 angiotensin receptor plays important physiological and pathophysiological roles in the cardiovascular system. Renin-angiotensin system represents a target system for drugs acting at different levels. The main effects of ATR1 stimulation involve activation of Gq proteins and subsequent IP3, DAG, and calcium signaling. It has become evident in recent years that besides the well-known G protein pathways, AT1R also activates a parallel signaling pathway through β-arrestins. β-arrestins were originally described as proteins that desensitize G protein-coupled receptors, but they can also mediate receptor internalization and G protein-independent signaling. AT1R is one of the most studied receptors, which was used to unravel the newly recognized β-arrestin-mediated pathways. β-arrestin-mediated signaling has become one of the most studied topics in recent years in molecular pharmacology and the modulation of these pathways of the AT1R might offer new therapeutic opportunities in the near future. In this paper, we review the recent advances in the field of β-arrestin signaling of the AT1R, emphasizing its role in cardiovascular regulation and heart failure.
Collapse
Affiliation(s)
- Gábor Turu
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- MTA-SE Laboratory of Molecular Physiology, Semmelweis University, Hungarian Academy of Sciences, Budapest, Hungary
| | - András Balla
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- MTA-SE Laboratory of Molecular Physiology, Semmelweis University, Hungarian Academy of Sciences, Budapest, Hungary
| | - László Hunyady
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- MTA-SE Laboratory of Molecular Physiology, Semmelweis University, Hungarian Academy of Sciences, Budapest, Hungary
- *Correspondence: László Hunyady
| |
Collapse
|
14
|
Laporte SA, Scott MGH. β-Arrestins: Multitask Scaffolds Orchestrating the Where and When in Cell Signalling. Methods Mol Biol 2019; 1957:9-55. [PMID: 30919345 DOI: 10.1007/978-1-4939-9158-7_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The β-arrestins (β-arrs) were initially appreciated for the roles they play in the desensitization and endocytosis of G protein-coupled receptors (GPCRs). They are now also known to act as multifunctional adaptor proteins binding many non-receptor protein partners to control multiple signalling pathways. β-arrs therefore act as key regulatory hubs at the crossroads of external cell inputs and functional outputs in cellular processes ranging from gene transcription to cell growth, survival, cytoskeletal regulation, polarity, and migration. An increasing number of studies have also highlighted the scaffolding roles β-arrs play in vivo in both physiological and pathological conditions, which opens up therapeutic avenues to explore. In this introductory review chapter, we discuss the functional roles that β-arrs exert to control GPCR function, their dynamic scaffolding roles and how this impacts signal transduction events, compartmentalization of β-arrs, how β-arrs are regulated themselves, and how the combination of these events culminates in cellular regulation.
Collapse
Affiliation(s)
- Stéphane A Laporte
- Department of Medicine, Research Institute of the McGill University Health Center (RI-MUHC), McGill University, Montreal, QC, Canada. .,Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada. .,RI-MUHC/Glen Site, Montréal, QC, Canada.
| | - Mark G H Scott
- Institut Cochin, INSERM U1016, Paris, France. .,CNRS, UMR 8104, Paris, France. .,Univ. Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
15
|
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: 18.9] [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
|
16
|
Grisanti LA, Schumacher SM, Tilley DG, Koch WJ. Designer Approaches for G Protein-Coupled Receptor Modulation for Cardiovascular Disease. JACC Basic Transl Sci 2018; 3:550-562. [PMID: 30175279 PMCID: PMC6115700 DOI: 10.1016/j.jacbts.2017.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 12/17/2022]
Abstract
The new horizon for cardiac therapy may lie beneath the surface, with the downstream mediators of G protein–coupled receptor (GPCR) activity. Targeted approaches have shown that receptor activation may be biased toward signaling through G proteins or through GPCR kinases (GRKs) and β-arrestins, with divergent functional outcomes. In addition to these canonical roles, numerous noncanonical activities of GRKs and β-arrestins have been demonstrated to modulate GPCR signaling at all levels of receptor activation and regulation. Further, research continues to identify novel GRK/effector and β-arrestin/effector complexes with distinct impacts on cardiac function in the normal heart and the diseased heart. Coupled with the identification of once orphan receptors and endogenous ligands with beneficial cardiovascular effects, this expands the repertoire of GPCR targets. Together, this research highlights the potential for focused therapeutic activation of beneficial pathways, with simultaneous exclusion or inhibition of detrimental signaling, and represents a new wave of therapeutic development.
Collapse
Key Words
- AR, adrenergic receptor
- AT1R, angiotensin II type 1A receptor
- CRF, corticotropin-releasing factor
- EGFR, epidermal growth factor receptor
- ERK1/2, extracellular signal-regulated kinase
- G protein–coupled receptor kinases
- G protein–coupled receptors
- GPCR, G protein–coupled receptor
- GRK, G protein–coupled receptor kinase
- HF, heart failure
- ICL, intracellular loop
- PI3K, phosphoinositide 3-kinase
- SERCA2a, sarco(endo)plasmic reticulum Ca2+-ATPase
- SII, [Sar(1), Ile (4), Ile(8)]-angiotensin II
- biased ligands
Collapse
Affiliation(s)
- Laurel A Grisanti
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Sarah M Schumacher
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania.,Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Douglas G Tilley
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Walter J Koch
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Marceau F, Bawolak MT, Fortin JP, Morissette G, Roy C, Bachelard H, Gera L, Charest-Morin X. Bifunctional ligands of the bradykinin B 2 and B 1 receptors: An exercise in peptide hormone plasticity. Peptides 2018; 105:37-50. [PMID: 29802875 DOI: 10.1016/j.peptides.2018.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/14/2018] [Accepted: 05/15/2018] [Indexed: 12/24/2022]
Abstract
Kinins are the small and fragile hydrophilic peptides related to bradykinin (BK) and derived from circulating kininogens via the action of kallikreins. Kinins bind to the preformed and widely distributed B2 receptor (B2R) and to the inducible B1 receptor (B1R). B2Rs and B1Rs are related G protein coupled receptors that possess natural agonist ligands of nanomolar affinity (BK and Lys BK for B2Rs, Lys-des-Arg9-BK for B1R). Decades of structure-activity exploration have resulted in the production of peptide analogs that are antagonists, one of which is clinically used (the B2R antagonist icatibant), and also non-peptide ligands for both receptor subtypes. The modification of kinin receptor ligands has made them resistant to extracellular or endosomal peptidases and/or produced bifunctional ligands, defined as agonist or antagonist peptide ligands conjugated with a chemical fluorophore (emitting in the whole spectrum, from the infrared to the ultraviolet), a drug-like moiety, an epitope, an isotope chelator/carrier, a cleavable sequence (thus forming a pro-drug) and even a fused protein. Dual molecular targets for specific modified peptides may be a source of side effects or of medically exploitable benefits. Biotechnological protein ligands for either receptor subtype have been produced: they are enhanced green fluorescent protein or the engineered peroxidase APEX2 fused to an agonist kinin sequence at their C-terminal terminus. Antibodies endowed with pharmacological actions (agonist, antagonist) at B2R have been reported, though not monoclonal antibodies. These findings define classes of alternative ligands of the kinin receptor of potential therapeutic and diagnostic value.
Collapse
Affiliation(s)
| | | | | | | | - Caroline Roy
- CHU de Québec - Université Laval, Québec, QC, G1 V 4G2, Canada
| | | | - Lajos Gera
- Department of Biochemistry, University of Colorado Denver, Aurora, CO, 80045, USA
| | | |
Collapse
|
18
|
Abstract
β-arrestin1 (or arrestin2) and β-arrestin2 (or arrestin3) are ubiquitously expressed cytosolic adaptor proteins that were originally discovered for their inhibitory role in G protein-coupled receptor (GPCR) signaling through heterotrimeric G proteins. However, further biochemical characterization revealed that β-arrestins do not just "block" the activated GPCRs, but trigger endocytosis and kinase activation leading to specific signaling pathways that can be localized on endosomes. The signaling pathways initiated by β-arrestins were also found to be independent of G protein activation by GPCRs. The discovery of ligands that blocked G protein activation but promoted β-arrestin binding, or vice-versa, suggested the exciting possibility of selectively activating intracellular signaling pathways. In addition, it is becoming increasingly evident that β-arrestin-dependent signaling is extremely diverse and provokes distinct cellular responses through different GPCRs even when the same effector kinase is involved. In this review, we summarize various signaling pathways mediated by β-arrestins and highlight the physiologic effects of β-arrestin-dependent signaling.
Collapse
|
19
|
Tréfier A, Guillou F, Crépieux P. [Investigation methods to explore G protein-coupled receptor-regulated translatome]. C R Biol 2018; 341:65-74. [PMID: 29326051 DOI: 10.1016/j.crvi.2017.12.002] [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: 10/26/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/26/2022]
Abstract
With the advent of next-generation sequencing technologies, identifying the translatome, which includes genome-wide ribosome-associated mRNAs, provides new opportunities to define faithfully the protein repertoire of a cell, as opposed to transcriptomic approaches. In addition, the role that extracellular signals such as hormonal modulations could play on the translatome remains to be deciphered. In particular, the regulation of the translatome by G protein-coupled receptors (GPCR) is still poorly described, albeit the trophic role that many receptors of this family play in their target cells. Here, we provide an overview of the current methods that are used to study the translatome, applied to the GPCR receptor family.
Collapse
Affiliation(s)
- Aurélie Tréfier
- Groupe Biologie et bioinformatique des systèmes de signalisation, Inra, UMR 85, unité Physiologie de la reproduction et des comportements, 37380 Nouzilly, France; CNRS, UMR 7247, 37380 Nouzilly, France; Université François-Rabelais, 37041 Tours, France; IFCE, 37380 Nouzilly, France
| | - Florian Guillou
- Plasticité génomique et expression phénotypique, Inra, UMR 85, unité Physiologie de la reproduction et des comportements, 37380 Nouzilly, France; CNRS, UMR 7247, 37380 Nouzilly, France; Université François-Rabelais, 37041 Tours, France; IFCE, 37380 Nouzilly, France
| | - Pascale Crépieux
- Groupe Biologie et bioinformatique des systèmes de signalisation, Inra, UMR 85, unité Physiologie de la reproduction et des comportements, 37380 Nouzilly, France; CNRS, UMR 7247, 37380 Nouzilly, France; Université François-Rabelais, 37041 Tours, France; IFCE, 37380 Nouzilly, France.
| |
Collapse
|
20
|
Tréfier A, Musnier A, Landomiel F, Bourquard T, Boulo T, Ayoub MA, León K, Bruneau G, Chevalier M, Durand G, Blache MC, Inoue A, Fontaine J, Gauthier C, Tesseraud S, Reiter E, Poupon A, Crépieux P. G protein-dependent signaling triggers a β-arrestin-scaffolded p70S6K/ rpS6 module that controls 5'TOP mRNA translation. FASEB J 2018; 32:1154-1169. [PMID: 29084767 DOI: 10.1096/fj.201700763r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Many interaction partners of β-arrestins intervene in the control of mRNA translation. However, how β-arrestins regulate this cellular process has been poorly explored. In this study, we show that β-arrestins constitutively assemble a p70S6K/ribosomal protein S6 (rpS6) complex in HEK293 cells and in primary Sertoli cells of the testis. We demonstrate that this interaction is direct, and experimentally validate the interaction interface between β-arrestin 1 and p70S6K predicted by our docking algorithm. Like most GPCRs, the biological function of follicle-stimulating hormone receptor (FSHR) is transduced by G proteins and β-arrestins. Upon follicle-stimulating hormone (FSH) stimulation, activation of G protein-dependent signaling enhances p70S6K activity within the β-arrestin/p70S6K/rpS6 preassembled complex, which is not recruited to the FSHR. In agreement, FSH-induced rpS6 phosphorylation within the β-arrestin scaffold was decreased in cells depleted of Gαs. Integration of the cooperative action of β-arrestin and G proteins led to the translation of 5' oligopyrimidine track mRNA with high efficacy within minutes of FSH input. Hence, this work highlights new relationships between G proteins and β-arrestins when acting cooperatively on a common signaling pathway, contrasting with their previously shown parallel action on the ERK MAP kinase pathway. In addition, this study provides insights into how GPCR can exert trophic effects in the cell.-Tréfier, A., Musnier, A., Landomiel, F., Bourquard, T., Boulo, T., Ayoub, M. A., León, K., Bruneau, G., Chevalier, M., Durand, G., Blache, M.-C., Inoue, A., Fontaine, J., Gauthier, C., Tesseraud, S., Reiter, E., Poupon, A., Crépieux, P. G protein-dependent signaling triggers a β-arrestin-scaffolded p70S6K/ rpS6 module that controls 5'TOP mRNA translation.
Collapse
Affiliation(s)
- Aurélie Tréfier
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Astrid Musnier
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Flavie Landomiel
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Thomas Bourquard
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Thomas Boulo
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Mohammed Akli Ayoub
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France.,Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kelly León
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Gilles Bruneau
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Manon Chevalier
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Guillaume Durand
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Marie-Claire Blache
- Plateau d'Imagerie Cellulaire (PIC), Unité Mixte de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan; and
| | - Joël Fontaine
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Christophe Gauthier
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Sophie Tesseraud
- Metabolism of Birds, Quality and Adaptation (MOQA) Group, Unité de Recherches 83, Unité de Recherches Avicoles, Institut National de la Recherche Agronomique (INRA), Nouzilly, France
| | - Eric Reiter
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Anne Poupon
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| | - Pascale Crépieux
- Biology and Bioinformatics of Signaling Systems (BIOS) Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique (INRA), Nouzilly, France.,Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 7247, Nouzilly, France.,Université François Rabelais, Tours, France.,Institut Français du Cheval et de l'Équitation (IFCE), Nouzilly, France
| |
Collapse
|
21
|
Peterson YK, Luttrell LM. The Diverse Roles of Arrestin Scaffolds in G Protein-Coupled Receptor Signaling. Pharmacol Rev 2017. [PMID: 28626043 DOI: 10.1124/pr.116.013367] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The visual/β-arrestins, a small family of proteins originally described for their role in the desensitization and intracellular trafficking of G protein-coupled receptors (GPCRs), have emerged as key regulators of multiple signaling pathways. Evolutionarily related to a larger group of regulatory scaffolds that share a common arrestin fold, the visual/β-arrestins acquired the capacity to detect and bind activated GPCRs on the plasma membrane, which enables them to control GPCR desensitization, internalization, and intracellular trafficking. By acting as scaffolds that bind key pathway intermediates, visual/β-arrestins both influence the tonic level of pathway activity in cells and, in some cases, serve as ligand-regulated scaffolds for GPCR-mediated signaling. Growing evidence supports the physiologic and pathophysiologic roles of arrestins and underscores their potential as therapeutic targets. Circumventing arrestin-dependent GPCR desensitization may alleviate the problem of tachyphylaxis to drugs that target GPCRs, and find application in the management of chronic pain, asthma, and psychiatric illness. As signaling scaffolds, arrestins are also central regulators of pathways controlling cell growth, migration, and survival, suggesting that manipulating their scaffolding functions may be beneficial in inflammatory diseases, fibrosis, and cancer. In this review we examine the structure-function relationships that enable arrestins to perform their diverse roles, addressing arrestin structure at the molecular level, the relationship between arrestin conformation and function, and sites of interaction between arrestins, GPCRs, and nonreceptor-binding partners. We conclude with a discussion of arrestins as therapeutic targets and the settings in which manipulating arrestin function might be of clinical benefit.
Collapse
Affiliation(s)
- Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (Y.K.P.), and Departments of Medicine and Biochemistry and Molecular Biology (L.M.L.), Medical University of South Carolina, Charleston, South Carolina; and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina (L.M.L.)
| | - Louis M Luttrell
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy (Y.K.P.), and Departments of Medicine and Biochemistry and Molecular Biology (L.M.L.), Medical University of South Carolina, Charleston, South Carolina; and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina (L.M.L.)
| |
Collapse
|
22
|
Stoppel LJ, Auerbach BD, Senter RK, Preza AR, Lefkowitz RJ, Bear MF. β-Arrestin2 Couples Metabotropic Glutamate Receptor 5 to Neuronal Protein Synthesis and Is a Potential Target to Treat Fragile X. Cell Rep 2017; 18:2807-2814. [PMID: 28329674 DOI: 10.1016/j.celrep.2017.02.075] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 01/31/2017] [Accepted: 02/24/2017] [Indexed: 10/19/2022] Open
Abstract
Synaptic protein synthesis is essential for modification of the brain by experience and is aberrant in several genetically defined disorders, notably fragile X (FX), a heritable cause of autism and intellectual disability. Neural activity directs local protein synthesis via activation of metabotropic glutamate receptor 5 (mGlu5), yet how mGlu5 couples to the intracellular signaling pathways that regulate mRNA translation is poorly understood. Here, we provide evidence that β-arrestin2 mediates mGlu5-stimulated protein synthesis in the hippocampus and show that genetic reduction of β-arrestin2 corrects aberrant synaptic plasticity and cognition in the Fmr1-/y mouse model of FX. Importantly, reducing β-arrestin2 does not induce psychotomimetic activity associated with full mGlu5 inhibitors and does not affect Gq signaling. Thus, in addition to identifying a key requirement for mGlu5-stimulated protein synthesis, these data suggest that β-arrestin2-biased negative modulators of mGlu5 offer significant advantages over first-generation inhibitors for the treatment of FX and related disorders.
Collapse
Affiliation(s)
- Laura J Stoppel
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Benjamin D Auerbach
- The Center for Hearing and Deafness, Department of Communicative Disorders and Sciences, The State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Rebecca K Senter
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anthony R Preza
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Robert J Lefkowitz
- Departments of Medicine and Biochemistry, Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Mark F Bear
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
23
|
Reiter E, Ayoub MA, Pellissier LP, Landomiel F, Musnier A, Tréfier A, Gandia J, De Pascali F, Tahir S, Yvinec R, Bruneau G, Poupon A, Crépieux P. β-arrestin signalling and bias in hormone-responsive GPCRs. Mol Cell Endocrinol 2017; 449:28-41. [PMID: 28174117 DOI: 10.1016/j.mce.2017.01.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 01/31/2017] [Accepted: 01/31/2017] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) play crucial roles in the ability of target organs to respond to hormonal cues. GPCRs' activation mechanisms have long been considered as a two-state process connecting the agonist-bound receptor to heterotrimeric G proteins. This view is now challenged as mounting evidence point to GPCRs being connected to large arrays of transduction mechanisms involving heterotrimeric G proteins as well as other players. Amongst the G protein-independent transduction mechanisms, those elicited by β-arrestins upon their recruitment to the active receptors are by far the best characterized and apply to most GPCRs. These concepts, in conjunction with remarkable advances made in the field of GPCR structural biology and biophysics, have supported the notion of ligand-selective signalling also known as pharmacological bias. Interestingly, recent reports have opened intriguing prospects to the way β-arrestins control GPCR-mediated signalling in space and time within the cells. In the present paper, we review the existing evidence linking endocrine-related GPCRs to β-arrestin recruitement, signalling, pathophysiological implications and selective activation by biased ligands and/or receptor modifications. Emerging concepts surrounding β-arrestin-mediated transduction are discussed in the light of the peculiarities of endocrine systems.
Collapse
Affiliation(s)
- Eric Reiter
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France.
| | - Mohammed Akli Ayoub
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France; LE STUDIUM(®) Loire Valley Institute for Advanced Studies, 45000, Orléans, France; Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | - Flavie Landomiel
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Astrid Musnier
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Aurélie Tréfier
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Jorge Gandia
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | | | - Shifa Tahir
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Romain Yvinec
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Gilles Bruneau
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Anne Poupon
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Pascale Crépieux
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| |
Collapse
|
24
|
Abstract
Cardiomyopathies represent a heterogeneous group of diseases that negatively affect heart function. Primary cardiomyopathies specifically target the myocardium, and may arise from genetic [hypertrophic cardiomyopathy (HCM), arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D), mitochondrial cardiomyopathy] or genetic and acquired [dilated cardiomyopathy (DCM), restrictive cardiomyopathy (RCM)] etiology. Modern genomics has identified mutations that are common in these populations, while in vitro and in vivo experimentation with these mutations have provided invaluable insight into the molecular mechanisms native to these diseases. For example, increased myosin heavy chain (MHC) binding and ATP utilization lead to the hypercontractile sarcomere in HCM, while abnormal protein–protein interaction and impaired Ca2+ flux underlie the relaxed sarcomere of DCM. Furthermore, expanded access to genetic testing has facilitated identification of potential risk factors that appear through inheritance and manifest sometimes only in the advanced stages of the disease. In this review, we discuss the genetic and molecular abnormalities unique to and shared between these primary cardiomyopathies and discuss some of the important advances made using more traditional basic science experimentation.
Collapse
|
25
|
Delgado-Peraza F, Ahn KH, Nogueras-Ortiz C, Mungrue IN, Mackie K, Kendall DA, Yudowski GA. Mechanisms of Biased β-Arrestin-Mediated Signaling Downstream from the Cannabinoid 1 Receptor. Mol Pharmacol 2016; 89:618-29. [PMID: 27009233 PMCID: PMC4885504 DOI: 10.1124/mol.115.103176] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 03/22/2016] [Indexed: 12/23/2022] Open
Abstract
Activation of G protein-coupled receptors results in multiple waves of signaling that are mediated by heterotrimeric G proteins and the scaffolding proteins β-arrestin 1/2. Ligands can elicit full or subsets of cellular responses, a concept defined as ligand bias or functional selectivity. However, our current understanding of β-arrestin-mediated signaling is still very limited. Here we provide a comprehensive view of β-arrestin-mediated signaling from the cannabinoid 1 receptor (CB1R). By using a signaling biased receptor, we define the cascades, specific receptor kinases, and molecular mechanism underlying β-arrestin-mediated signaling: We identify the interaction kinetics of CB1R and β-arrestin 1 during their endocytic trafficking as directly proportional to its efficacy. Finally, we demonstrate that signaling results in the control of genes clustered around prosurvival and proapoptotic functions among others. Together, these studies constitute a comprehensive description of β-arrestin-mediated signaling from CB1Rs and suggest modulation of receptor endocytic trafficking as a therapeutic approach to control β-arrestin-mediated signaling.
Collapse
Affiliation(s)
- Francheska Delgado-Peraza
- Department of Anatomy and Neurobiology (F.D.-P., G.A.Y.) and Institute of Neurobiology (F.D.-P., C.N.-O., G.A.Y.), University of Puerto Rico - Medical Sciences Campus, San Juan, Puerto Rico; Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (K.H.A., D.A.K.); Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana (I.N.M.); and Department of Psychological & Brain Sciences, Gill Center for Biomedical Sciences, Indiana University, Bloomington, Indiana (K.M.)
| | - Kwang H Ahn
- Department of Anatomy and Neurobiology (F.D.-P., G.A.Y.) and Institute of Neurobiology (F.D.-P., C.N.-O., G.A.Y.), University of Puerto Rico - Medical Sciences Campus, San Juan, Puerto Rico; Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (K.H.A., D.A.K.); Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana (I.N.M.); and Department of Psychological & Brain Sciences, Gill Center for Biomedical Sciences, Indiana University, Bloomington, Indiana (K.M.)
| | - Carlos Nogueras-Ortiz
- Department of Anatomy and Neurobiology (F.D.-P., G.A.Y.) and Institute of Neurobiology (F.D.-P., C.N.-O., G.A.Y.), University of Puerto Rico - Medical Sciences Campus, San Juan, Puerto Rico; Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (K.H.A., D.A.K.); Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana (I.N.M.); and Department of Psychological & Brain Sciences, Gill Center for Biomedical Sciences, Indiana University, Bloomington, Indiana (K.M.)
| | - Imran N Mungrue
- Department of Anatomy and Neurobiology (F.D.-P., G.A.Y.) and Institute of Neurobiology (F.D.-P., C.N.-O., G.A.Y.), University of Puerto Rico - Medical Sciences Campus, San Juan, Puerto Rico; Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (K.H.A., D.A.K.); Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana (I.N.M.); and Department of Psychological & Brain Sciences, Gill Center for Biomedical Sciences, Indiana University, Bloomington, Indiana (K.M.)
| | - Ken Mackie
- Department of Anatomy and Neurobiology (F.D.-P., G.A.Y.) and Institute of Neurobiology (F.D.-P., C.N.-O., G.A.Y.), University of Puerto Rico - Medical Sciences Campus, San Juan, Puerto Rico; Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (K.H.A., D.A.K.); Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana (I.N.M.); and Department of Psychological & Brain Sciences, Gill Center for Biomedical Sciences, Indiana University, Bloomington, Indiana (K.M.)
| | - Debra A Kendall
- Department of Anatomy and Neurobiology (F.D.-P., G.A.Y.) and Institute of Neurobiology (F.D.-P., C.N.-O., G.A.Y.), University of Puerto Rico - Medical Sciences Campus, San Juan, Puerto Rico; Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (K.H.A., D.A.K.); Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana (I.N.M.); and Department of Psychological & Brain Sciences, Gill Center for Biomedical Sciences, Indiana University, Bloomington, Indiana (K.M.)
| | - Guillermo A Yudowski
- Department of Anatomy and Neurobiology (F.D.-P., G.A.Y.) and Institute of Neurobiology (F.D.-P., C.N.-O., G.A.Y.), University of Puerto Rico - Medical Sciences Campus, San Juan, Puerto Rico; Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (K.H.A., D.A.K.); Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana (I.N.M.); and Department of Psychological & Brain Sciences, Gill Center for Biomedical Sciences, Indiana University, Bloomington, Indiana (K.M.)
| |
Collapse
|
26
|
Ikeda Y, Kumagai H, Motozawa Y, Suzuki JI, Komuro I. Biased Agonism of the Angiotensin II Type I Receptor. Int Heart J 2016; 56:485-8. [PMID: 26180022 DOI: 10.1536/ihj.15-256] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Angiotensin II (AngII) type I receptor (AT1R) recognizes AngII, a cardiovascular peptide hormone that acts as a terminal effector of the renin-angiotensin system (RAS). AT1R belongs to the rhodopsin-like peptidergic family of G protein-coupled receptors (GPCRs) and serves as a therapeutic target for the treatment of cardiovascular diseases, such as hypertension, cardiac hypertrophy and heart failure. Classically, AT1R was considered to signal only through G proteins. However, recent studies have revealed that AT1R is capable of activating G protein-independent signaling that is mediated by β-arrestins. β-arrestin is a cytosolic scaffold that is recruited to the activated GPCRs. In vitro and ex vivo studies have demonstrated that the activation of the AT1R-β-arrestin pathway stimulates contractility and exerts prosurvival effects in cardiomyocytes. TRV027, a potent synthetic β-arrestin-biased ligand for AT1R, specifically activates AT1R-β-arrestin signaling without stimulating G proteins. In preclinical studies, TRV027 not only produced vasodilation by antagonizing the AT1R-Gαq pathway but also enhanced cardiac performance by activating AT1R-β-arrestin signaling. Because of this unique pharmacological profile, TRV027 is now being evaluated in a phase II clinical trial as a novel therapeutic for acute heart failure (AHF).
Collapse
Affiliation(s)
- Yuichi Ikeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | | | | | | | | |
Collapse
|
27
|
Wu L, Mei L, Chong L, Huang Y, Li Y, Chu M, Yang X. Olmesartan ameliorates pressure overload-induced cardiac remodeling through inhibition of TAK1/p38 signaling in mice. Life Sci 2016; 145:121-6. [DOI: 10.1016/j.lfs.2015.12.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/29/2015] [Accepted: 12/14/2015] [Indexed: 11/30/2022]
|
28
|
Wisler JW, Harris EM, Raisch M, Mao L, Kim J, Rockman HA, Lefkowitz RJ. The role of β-arrestin2-dependent signaling in thoracic aortic aneurysm formation in a murine model of Marfan syndrome. Am J Physiol Heart Circ Physiol 2015; 309:H1516-27. [PMID: 26371162 PMCID: PMC4666970 DOI: 10.1152/ajpheart.00291.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 08/15/2015] [Indexed: 01/14/2023]
Abstract
Ang II type 1a receptor (AT1aR)-mediated activation of MAPKs contributes to thoracic aortic aneurysm (TAA) development in Marfan syndrome (MFS). β-Arrestin2 (βarr2) is known to mediate AT1aR-dependent MAPK activation, as well as proproliferative and profibrotic signaling in aortic vascular smooth muscle cells. Therefore, we investigated whether βarr2-dependent signaling contributes to TAA formation in MFS. We used a murine model of MFS [fibrillin (Fbn)(C1039G/+)] to generate an MFS murine model in combination with genetic βarr2 deletion (Fbn(C1039G/+)/βarr2(-/-)). Fbn(C1039G/+)/βarr2(-/-) mice displayed delayed aortic root dilation compared with Fbn(C1039G/+) mice. The mRNA and protein expression of several mediators of TAA formation, including matrix metalloproteinase (MMP)-2 and -9, was reduced in the aorta of Fbn(C1039G/+)/βarr2(-/-) mice relative to Fbn(C1039G/+) mice. Activation of ERK1/2 was also decreased in the aortas of Fbn(C1039G/+)/βarr2(-/-) mice compared with Fbn(C1039G/+) animals. Small interfering RNA targeting βarr2 inhibited angiotensin-stimulated expression of proaneurysmal signaling mediators in primary aortic root smooth muscle cells. Angiotensin-stimulated expression of the proaneurysmal signaling mediators MMP-2 and -9 was inhibited by blockade of ERK1/2 or the EGF receptor, whereas blockade of the transforming growth factor-β receptor had no effect. These results suggest that βarr2 contributes to TAA formation in MFS by regulating ERK1/2-dependent expression of proaneurysmal genes and proteins downstream of the AT1aR. Importantly, this demonstration of the unique signaling mechanism by which βarr2 contributes to aneurysm formation identifies multiple novel, potential therapeutic targets in MFS.
Collapse
MESH Headings
- Angiotensins/pharmacology
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/genetics
- Arrestins/genetics
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Disease Models, Animal
- ErbB Receptors/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/genetics
- Fibrillins
- Fibrosis
- MAP Kinase Signaling System
- Marfan Syndrome/genetics
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinase 9/genetics
- Mice
- Mice, Knockout
- Microfilament Proteins/genetics
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- RNA, Messenger/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Receptors, Transforming Growth Factor beta/antagonists & inhibitors
- Signal Transduction
- Transcriptome
- beta-Arrestins
Collapse
Affiliation(s)
- James W Wisler
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Emily M Harris
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Michael Raisch
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina
| | - Lan Mao
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Jihee Kim
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, Durham, North Carolina; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina; Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina
| | - Robert J Lefkowitz
- Department of Medicine, Duke University Medical Center, Durham, North Carolina; Department of Biochemistry, Duke University Medical Center, Durham, North Carolina; and Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
29
|
Karnik SS, Unal H, Kemp JR, Tirupula KC, Eguchi S, Vanderheyden PML, Thomas WG. International Union of Basic and Clinical Pharmacology. XCIX. Angiotensin Receptors: Interpreters of Pathophysiological Angiotensinergic Stimuli [corrected]. Pharmacol Rev 2015; 67:754-819. [PMID: 26315714 PMCID: PMC4630565 DOI: 10.1124/pr.114.010454] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The renin angiotensin system (RAS) produced hormone peptides regulate many vital body functions. Dysfunctional signaling by receptors for RAS peptides leads to pathologic states. Nearly half of humanity today would likely benefit from modern drugs targeting these receptors. The receptors for RAS peptides consist of three G-protein-coupled receptors—the angiotensin II type 1 receptor (AT1 receptor), the angiotensin II type 2 receptor (AT2 receptor), the MAS receptor—and a type II trans-membrane zinc protein—the candidate angiotensin IV receptor (AngIV binding site). The prorenin receptor is a relatively new contender for consideration, but is not included here because the role of prorenin receptor as an independent endocrine mediator is presently unclear. The full spectrum of biologic characteristics of these receptors is still evolving, but there is evidence establishing unique roles of each receptor in cardiovascular, hemodynamic, neurologic, renal, and endothelial functions, as well as in cell proliferation, survival, matrix-cell interaction, and inflammation. Therapeutic agents targeted to these receptors are either in active use in clinical intervention of major common diseases or under evaluation for repurposing in many other disorders. Broad-spectrum influence these receptors produce in complex pathophysiological context in our body highlights their role as precise interpreters of distinctive angiotensinergic peptide cues. This review article summarizes findings published in the last 15 years on the structure, pharmacology, signaling, physiology, and disease states related to angiotensin receptors. We also discuss the challenges the pharmacologist presently faces in formally accepting newer members as established angiotensin receptors and emphasize necessary future developments.
Collapse
Affiliation(s)
- Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Hamiyet Unal
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Jacqueline R Kemp
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Satoru Eguchi
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Patrick M L Vanderheyden
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Walter G Thomas
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| |
Collapse
|
30
|
Choudhary P, Loewen MC. Evidence of a role for S. cerevisiae α-arrestin Art1 (Ldb19) in mating projection and zygote formations. Cell Biol Int 2015; 40:83-90. [PMID: 26314564 DOI: 10.1002/cbin.10541] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/22/2015] [Indexed: 01/08/2023]
Abstract
The discovery of arrestin-mediated biased signalling mechanisms for mammalian G-protein coupled receptors (GPCRs) has revolutionized the field over the last decade. Now, with the recent demonstration of a role for α-arrestins in internalization of the yeast pheromone GPCR, Ste2p, the possibility of arrestin-mediated alternate GPCR functionalities in yeast also follows. Here, the effects of knockout and complementation of yeast α-arrestin expression during mating are reported. Although minor effects on classical pheromone-related signalling are noted for a few arrestins, much stronger effects were observed downstream of cell cycle arrest, in particular linking Ldb19 (Art1) to mediation of zygote formation. Subsequent phenotypic observations linked this activity to more pronounced projection formation in an Art1 complemented noncuple α-arrestin knockout line, compared to the knockout-line alone, or either of the Art3 or Art6 complemented lines. Together with the observation of ligand-stimulated localization of Art-GFP to the mating projection, a possible role for this arrestin-like protein in projection formation is supported. While leaving the full mechanism of alternate Art1 functionality to be elucidated, together these findings implicate Art1 in selective regulation of mating events downstream of receptor internalization and cell cycle arrest, leading to schmoo, and ultimately zygote formation.
Collapse
Affiliation(s)
- Pooja Choudhary
- Department of Biochemistry, University of Saskatchewan, 107 Wiggins Rd., Saskatoon, S7N 5E5, Saskatchewan, Canada
| | - Michele C Loewen
- Department of Biochemistry, University of Saskatchewan, 107 Wiggins Rd., Saskatoon, S7N 5E5, Saskatchewan, Canada.,National Research Council of Canada, 110 Gymnasium Place, Saskatoon, S7N 0W9, Saskatchewan, Canada
| |
Collapse
|
31
|
Shukla AK, Singh G, Ghosh E. Emerging structural insights into biased GPCR signaling. Trends Biochem Sci 2014; 39:594-602. [DOI: 10.1016/j.tibs.2014.10.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/24/2014] [Accepted: 10/01/2014] [Indexed: 01/04/2023]
|
32
|
Kendall RT, Lee MH, Pleasant DL, Robinson K, Kuppuswamy D, McDermott PJ, Luttrell LM. Arrestin-dependent angiotensin AT1 receptor signaling regulates Akt and mTor-mediated protein synthesis. J Biol Chem 2014; 289:26155-26166. [PMID: 25081544 DOI: 10.1074/jbc.m114.595728] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Control of protein synthesis is critical to both cell growth and proliferation. The mammalian target of rapamycin (mTOR) integrates upstream growth, proliferation, and survival signals, including those transmitted via ERK1/2 and Akt, to regulate the rate of protein translation. The angiotensin AT1 receptor has been shown to activate both ERK1/2 and Akt in arrestin-based signalsomes. Here, we examine the role of arrestin-dependent regulation of ERK1/2 and Akt in the stimulation of mTOR-dependent protein translation by the AT1 receptor using HEK293 and primary vascular smooth muscle cell models. Nascent protein synthesis stimulated by both the canonical AT1 receptor agonist angiotensin II (AngII), and the arrestin pathway-selective agonist [Sar(1)-Ile(4)-Ile(8)]AngII (SII), is blocked by shRNA silencing of βarrestin1/2 or pharmacological inhibition of Akt, ERK1/2, or mTORC1. In HEK293 cells, SII activates a discrete arrestin-bound pool of Akt and promotes Akt-dependent phosphorylation of mTOR and its downstream effector p70/p85 ribosomal S6 kinase (p70/85S6K). In parallel, SII-activated ERK1/2 helps promote mTOR and p70/85S6K phosphorylation, and is required for phosphorylation of the known ERK1/2 substrate p90 ribosomal S6 kinase (p90RSK). Thus, arrestins coordinate AT1 receptor regulation of ERK1/2 and Akt activity and stimulate protein translation via both Akt-mTOR-p70/85S6K and ERK1/2-p90RSK pathways. These results suggest that in vivo, arrestin pathway-selective AT1 receptor agonists may promote cell growth or hypertrophy through arrestin-mediated mechanisms despite their antagonism of G protein signaling.
Collapse
Affiliation(s)
- Ryan T Kendall
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425 and
| | - Mi-Hye Lee
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425 and
| | - Dorea L Pleasant
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425 and
| | - Katherine Robinson
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425 and
| | - Dhandapani Kuppuswamy
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425 and; Research Service of the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| | - Paul J McDermott
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425 and; Research Service of the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| | - Louis M Luttrell
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425 and; Research Service of the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401.
| |
Collapse
|
33
|
Zhou Y, Zhang Y, Guo Y, Zhang Y, Xu M, He B. β2-Adrenoceptor involved in smoking-induced airway mucus hypersecretion through β-arrestin-dependent signaling. PLoS One 2014; 9:e97788. [PMID: 24905583 PMCID: PMC4048185 DOI: 10.1371/journal.pone.0097788] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/22/2014] [Indexed: 12/11/2022] Open
Abstract
Progression of chronic obstructive pulmonary disease is associated with small airway obstruction by accumulation of inflammatory mucous exudates. However, the mechanism of mucin hypersecretion after exposure to cigarette smoke (CS) is still not clear. In this study, we explored the contribution of β2-adrenoceptor (β2-AR) signaling to CS extract (CSE)-induced mucus hypersecretion in vitro and examined the effect of a β-blocker on airway mucin hypersecretion in vivo. NCI-H292 epithelial cell line was used to determine the contribution of β2-AR signaling to CSE-induced MUC5AC production by treatment with β2-AR antagonists propranolol and ICI118551 and β2-AR-targeted small interfering RNA. The effect of propranolol on airway mucus hypersecretion was examined in a rat model exposed to CS. MUC5AC expression was assayed by real-time PCR, immunohistochemistry and ELISA. β2-AR and its downstream signaling were detected by western blot analysis. We found that pretreating NCI-H292 cells with propranolol, ICI118551 for 30 min or β2AR-targeted siRNA for 48 h reduced MUC5AC mRNA and protein levels stimulated by CSE. However,inhibiting the classical β2AR-cAMP-PKA pathway didn't attenuate CSE-induced MUC5AC production, while silencing β-arretin2 expression significantly decreased ERK and p38MAPK phosphorylation, thus reduced the CSE-stimulated MUC5AC production. In vivo, we found that administration of propranolol (25 mg kg(-1) d(-1)) for 28 days significantly attenuated the airway goblet cell metaplasia, mucus hypersecretion and MUC5AC expression of rats exposed to CS. From the study, β2-AR-β-arrestin2-ERK1/2 signaling was required for CS-induced airway MUC5AC expression. Chronic propranolol administration ameliorated airway mucus hypersecretion and MUC5AC expression in smoking rats. The exploration of these mechanisms may contribute to the optimization of β2-AR target therapy in chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Yujiao Zhou
- Department of Respiratory Medicine and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, People's Republic of China
| | - Yuan Zhang
- Department of Respiratory Medicine and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, People's Republic of China
| | - Yang Guo
- Department of Respiratory Medicine, Changji Renmin Hospital, Changji, Xinjiang, People's Republic of China
| | - Youyi Zhang
- Department of Respiratory Medicine and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, People's Republic of China
| | - Ming Xu
- Department of Respiratory Medicine and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, People's Republic of China
| | - Bei He
- Department of Respiratory Medicine and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, People's Republic of China
| |
Collapse
|
34
|
León K, Boulo T, Musnier A, Morales J, Gauthier C, Dupuy L, Heyne S, Backofen R, Poupon A, Cormier P, Reiter E, Crepieux P. Activation of a GPCR leads to eIF4G phosphorylation at the 5' cap and to IRES-dependent translation. J Mol Endocrinol 2014; 52:373-82. [PMID: 24711644 DOI: 10.1530/jme-14-0009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The control of mRNA translation has been mainly explored in response to activated tyrosine kinase receptors. In contrast, mechanistic details on the translational machinery are far less available in the case of ligand-bound G protein-coupled receptors (GPCRs). In this study, using the FSH receptor (FSH-R) as a model receptor, we demonstrate that part of the translational regulations occurs by phosphorylation of the translation pre-initiation complex scaffold protein, eukaryotic initiation factor 4G (eIF4G), in HEK293 cells stably expressing the FSH-R. This phosphorylation event occurred when eIF4G was bound to the mRNA 5' cap, and probably involves mammalian target of rapamycin. This regulation might contribute to cap-dependent translation in response to FSH. The cap-binding protein eIF4E also had its phosphorylation level enhanced upon FSH stimulation. We also show that FSH-induced signaling not only led to cap-dependent translation but also to internal ribosome entry site (IRES)-dependent translation of some mRNA. These data add detailed information on the molecular bases underlying the regulation of selective mRNA translation by a GPCR, and a topological model recapitulating these mechanisms is proposed.
Collapse
Affiliation(s)
- Kelly León
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Thomas Boulo
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Astrid Musnier
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Julia Morales
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, Germany
| | - Christophe Gauthier
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Laurence Dupuy
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Steffen Heyne
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, Germany
| | - Rolf Backofen
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, Germany
| | - Anne Poupon
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Patrick Cormier
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, Germany
| | - Eric Reiter
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Pascale Crepieux
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| |
Collapse
|
35
|
Biased ligands: pathway validation for novel GPCR therapeutics. Curr Opin Pharmacol 2014; 16:108-15. [PMID: 24834870 DOI: 10.1016/j.coph.2014.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/15/2014] [Accepted: 04/18/2014] [Indexed: 01/14/2023]
Abstract
G protein-coupled receptors (GPCRs), in recent years, have been shown to signal via multiple distinct pathways. Furthermore, biased ligands for some receptors can differentially stimulate or inhibit these pathways versus unbiased endogenous ligands or drugs. Biased ligands can be used to gain a deeper understanding of the molecular targets and cellular responses associated with a GPCR, and may be developed into therapeutics with improved efficacy, safety and/or tolerability. Here we review examples and approaches to pathway validation that establish the relevance and therapeutic potential of distinct pathways that can be selectively activated or blocked by biased ligands.
Collapse
|
36
|
Abstract
It is now established that agonists do not uniformly activate pleiotropic signaling mechanisms initiated by receptors but rather can bias signals according to the unique receptor conformations they stabilize. One of the important emerging signaling systems where this can occur is through β-arrestin. This chapter discusses biased signaling where emphasis or de-emphasis of β-arrestin signaling is postulated (or been shown) to be beneficial. The chapter specifically focuses on methods to quantify biased effects; these methods furnish scales that can be used in the process of optimizing biased agonism (and antagonism) for therapeutic benefit. Specifically, methods to derive ΔΔLog(τ/K A) or ΔΔLog(Relative Activity) values are described to do this.
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, 120 Mason Farm Road, Room 4042, Genetic Medicine Building, CB# 7365, Chapel Hill, NC, 27599-7365, USA,
| |
Collapse
|
37
|
Arrestin interaction with E3 ubiquitin ligases and deubiquitinases: functional and therapeutic implications. Handb Exp Pharmacol 2014; 219:187-203. [PMID: 24292831 DOI: 10.1007/978-3-642-41199-1_10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Arrestins constitute a small family of four homologous adaptor proteins (arrestins 1-4), which were originally identified as inhibitors of signal transduction elicited by the seven-transmembrane G protein-coupled receptors. Currently arrestins (especially arrestin2 and arrestin3; also called β-arrestin1 and β-arrestin2) are known to be activators of cell signaling and modulators of endocytic trafficking. Arrestins mediate these effects by binding to not only diverse cell-surface receptors but also by associating with a variety of critical signaling molecules in different intracellular compartments. Thus, the functions of arrestins are multifaceted and demand interactions with a host of proteins and require an array of selective conformations. Furthermore, receptor ligands that specifically induce signaling via arrestins are being discovered and their physiological roles are emerging. Recent evidence suggests that the activity of arrestin is regulated in space and time by virtue of its dynamic association with specific enzymes of the ubiquitination pathway. Ubiquitin-dependent, arrestin-mediated signaling could serve as a potential platform for developing novel therapeutic strategies to target transmembrane signaling and physiological responses.
Collapse
|
38
|
Abstract
The four members of the mammalian arrestin family, two visual and two nonvisual, share the property of stimulus-dependent docking to G protein-coupled receptors. This conformational selectivity permits them to function in receptor desensitization, as arrestin binding sterically inhibits G protein coupling. The two nonvisual arrestins further act as adapter proteins, linking receptors to the clathrin-dependent endocytic machinery and regulating receptor sequestration, intracellular trafficking, recycling, and degradation. Arrestins also function as ligand-regulated scaffolds, recruiting catalytically active proteins into receptor-based multiprotein "signalsome" complexes. Arrestin binding thus marks the transition from a transient G protein-coupled state on the plasma membrane to a persistent arrestin-coupled state that continues to signal as the receptor internalizes. Two of the earliest discovered and most studied arrestin-dependent signaling pathways involve regulation of Src family nonreceptor tyrosine kinases and the ERK1/2 mitogen-activated kinase cascade. In each case, arrestin scaffolding imposes constraints on kinase activity that dictate signal duration and substrate specificity. Evidence suggests that arrestin-bound ERK1/2 and Src not only play regulatory roles in receptor desensitization and trafficking but also mediate longer term effects on cell growth, migration, proliferation, and survival.
Collapse
Affiliation(s)
- Erik G Strungs
- Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | | |
Collapse
|
39
|
Violin JD, Soergel DG, Boerrigter G, Burnett JC, Lark MW. GPCR biased ligands as novel heart failure therapeutics. Trends Cardiovasc Med 2013; 23:242-9. [DOI: 10.1016/j.tcm.2013.01.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 01/22/2013] [Accepted: 01/23/2013] [Indexed: 01/14/2023]
|
40
|
Ghanemi A. Targeting G protein coupled receptor-related pathways as emerging molecular therapies. Saudi Pharm J 2013; 23:115-29. [PMID: 25972730 PMCID: PMC4420995 DOI: 10.1016/j.jsps.2013.07.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 07/29/2013] [Indexed: 12/20/2022] Open
Abstract
G protein coupled receptors (GPCRs) represent the most important targets in modern pharmacology because of the different functions they mediate, especially within brain and peripheral nervous system, and also because of their functional and stereochemical properties. In this paper, we illustrate, via a variety of examples, novel advances about the GPCR-related molecules that have been shown to play diverse roles in GPCR pathways and in pathophysiological phenomena. We have exemplified how those GPCRs’ pathways are, or might constitute, potential targets for different drugs either to stimulate, modify, regulate or inhibit the cellular mechanisms that are hypothesized to govern some pathologic, physiologic, biologic and cellular or molecular aspects both in vivo and in vitro. Therefore, influencing such pathways will, undoubtedly, lead to different therapeutical applications based on the related pharmacological implications. Furthermore, such new properties can be applied in different fields. In addition to offering fruitful directions for future researches, we hope the reviewed data, together with the elements found within the cited references, will inspire clinicians and researchers devoted to the studies on GPCR’s properties.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
41
|
Arresting inflammation: contributions of plasma membrane and endosomal signalling to neuropeptide-driven inflammatory disease. Biochem Soc Trans 2013; 41:137-43. [PMID: 23356273 DOI: 10.1042/bst20120343] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
GPCR (G-protein-coupled receptor) signalling at the plasma membrane is under tight control. In the case of neuropeptides such as SP (substance P), plasma membrane signalling is regulated by cell-surface endopeptidases (e.g. neprilysin) that degrade extracellular neuropeptides, and receptor interaction with β-arrestins, which uncouple receptors from heterotrimeric G-proteins and mediate receptor endocytosis. By recruiting GPCRs, kinases and phosphatases to endocytosed GPCRs, β-arrestins assemble signalosomes that can mediate a second wave of signalling by internalized receptors. Endosomal peptidases, such as ECE-1 (endothelin-converting enzyme-1), can degrade SP in acidified endosomes, which destabilizes signalosomes and allows receptors, freed from β-arrestins, to recycle and resensitize. By disassembling signalosomes, ECE-1 terminates β-arrestin-mediated endosomal signalling. These mechanisms have been studied in model cell systems, and the relative importance of plasma membrane and endosomal signalling to complex pathophysiological processes, such as inflammation, pain and proliferation, is unclear. However, deletion or inhibition of metalloendopeptidases that control neuropeptide signalling at the plasma membrane and in endosomes has marked effects on inflammation. Neprilysin deletion exacerbates inflammation because of diminished degradation of pro-inflammatory SP. Conversely, inhibition of ECE-1 attenuates inflammation by preventing receptor recycling/resensitization, which is required for sustained pro-inflammatory signals from the plasma membrane. β-Arrestin deletion also affects inflammation because of the involvement of β-arrestins in pro-inflammatory signalling and migration of inflammatory cells. Knowledge of GPCR signalling in specific subcellular locations provides insights into pathophysiological processes, and can provide new opportunities for therapy. Selective targeting of β-arrestin-mediated endosomal signalling or of mechanisms of receptor recycling/resensitization may offer more effective and selective treatments than global targeting of cell-surface signalling.
Collapse
|
42
|
Luttrell LM. Arrestin Pathways as Drug Targets. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:469-97. [DOI: 10.1016/b978-0-12-394440-5.00018-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
43
|
Arrestins as regulators of kinases and phosphatases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:115-47. [PMID: 23764052 DOI: 10.1016/b978-0-12-394440-5.00005-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The discovery that, in addition to mediating G protein-coupled receptor (GPCR) desensitization and endocytosis, arrestins bind to diverse catalytically active nonreceptor proteins and act as ligand-regulated signaling scaffolds led to a paradigm shift in the study of GPCR signal transduction. Research over the past decade has solidified the concept that arrestins confer novel GPCR-signaling capacity by recruiting protein and lipid kinase, phosphatase, phosphodiesterase, and ubiquitin ligase activity into receptor-based multiprotein "signalsome" complexes. Signalsomes regulate downstream pathways controlled by Src family nonreceptor tyrosine kinases, mitogen-activated protein kinases, protein kinase B (AKT), glycogen synthase kinase 3, protein phosphatase 2A, nuclear factor-κB, and several others, imposing spatial and temporal control on their function. While many arrestin-bound kinases and phosphatases are involved in the control of cytoskeletal rearrangement, vesicle endocytosis, exocytosis, and cell migration, other signals reach into the nucleus, affecting cell proliferation, apoptosis, and survival. Indeed, the kinase/phosphatase network regulated by arrestins may be fully as diverse as that regulated by heterotrimeric G proteins.
Collapse
|
44
|
Systems Analysis of Arrestin Pathway Functions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:431-67. [DOI: 10.1016/b978-0-12-394440-5.00017-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
Abstract
The very large G protein coupled receptor (Vlgr1) is a member of adhesion receptors or large N-terminal family B-7 transmembrane helixes (LNB7TM) receptors within the seven trans-membrane receptor superfamily. Vlgr1 is the largest GPCR identified to date; its mRNA spans 19 kb and encodes 6,300 amino acids. Vlgr1 is a core component of ankle-link complex in inner ear hair cells. Knock-out and mutation mouse models show that loss of Vlgr1 function leads to abnormal stereociliary development and hearing loss, indicating crucial roles of Vlgr1 in hearing transduction or auditory system development. Over the past 10 or so years, human genetics data suggested that Vlgr1 mutations cause Usher syndromes and seizures. Although significant progresses have been made, the details of Vlgr1's function in hair cells, its signaling cascade, and the mechanisms underlying causative effects of Vlgr1 mutations in human diseases remain elusive and ask for further investigation.
Collapse
|
46
|
Cavanaugh A, Huang Y, Breitwieser GE. Behind the curtain: cellular mechanisms for allosteric modulation of calcium-sensing receptors. Br J Pharmacol 2012; 165:1670-1677. [PMID: 21470201 DOI: 10.1111/j.1476-5381.2011.01403.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Calcium-sensing receptors (CaSR) are integral to regulation of systemic Ca(2+) homeostasis. Altered expression levels or mutations in CaSR cause Ca(2+) handling diseases. CaSR is regulated by both endogenous allosteric modulators and allosteric drugs, including the first Food and Drug Administration-approved allosteric agonist, Cinacalcet HCl (Sensipar®). Recent studies suggest that allosteric modulators not only alter function of plasma membrane-localized CaSR, but regulate CaSR stability at the endoplasmic reticulum. This brief review summarizes our current understanding of the role of membrane-permeant allosteric agonists in cotranslational stabilization of CaSR, and highlights additional, indirect, signalling-dependent role(s) for membrane-impermeant allosteric drugs. Overall, these studies suggest that allosteric drugs act at multiple cellular organelles to control receptor abundance and hence function, and that drug hydrophobicity can bias the relative contributions of plasma membrane and intracellular organelles to CaSR abundance and signalling.
Collapse
Affiliation(s)
- Alice Cavanaugh
- Weis Center for Research, Geisinger Clinic, Danville, PA, USACancer Drug Research Laboratory, McGill University/Royal Victoria Hospital, Montreal, QC, Canada
| | - Ying Huang
- Weis Center for Research, Geisinger Clinic, Danville, PA, USACancer Drug Research Laboratory, McGill University/Royal Victoria Hospital, Montreal, QC, Canada
| | - Gerda E Breitwieser
- Weis Center for Research, Geisinger Clinic, Danville, PA, USACancer Drug Research Laboratory, McGill University/Royal Victoria Hospital, Montreal, QC, Canada
| |
Collapse
|
47
|
β-Arrestin2 mediates the initiation and progression of myeloid leukemia. Proc Natl Acad Sci U S A 2012; 109:12532-7. [PMID: 22773819 DOI: 10.1073/pnas.1209815109] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
β-Arrestins were initially discovered as negative regulators of G protein-coupled receptor signaling. Although β-arrestins have more recently been implicated as scaffold proteins that interact with various mitogenic and developmental signals, the genetic role of β-arrestins in driving oncogenesis is not known. Here we have investigated the role of β-arrestin in hematologic malignancies and have found that although both β-arrestin1 and -2 are expressed in the hematopoietic system, loss of β-arrestin2 preferentially leads to a severe impairment in the establishment and propagation of the chronic and blast crisis phases of chronic myelogenous leukemia (CML). These defects are linked to a reduced frequency, as well as defective self-renewal capacity of the cancer stem-cell population, in mouse models and in human CML patient samples. At a molecular level, the loss of β-arrestin2 leads to a significant inhibition of β-catenin stabilization, and ectopic activation of Wnt signaling reverses the defects observed in the β-arrestin2 mutant cells. These data cumulatively show that β-arrestin2 is essential for CML disease propagation and indicate that β-arrestins and the Wnt/β-catenin pathway lie in a signaling hierarchy in the context of CML cancer stem cell maintenance.
Collapse
|
48
|
Christensen GL, Aplin M, Hansen JL. Therapeutic potential of functional selectivity in the treatment of heart failure. Trends Cardiovasc Med 2012; 20:221-7. [PMID: 22293022 DOI: 10.1016/j.tcm.2011.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Adrenergic and angiotensin receptors are prominent targets in pharmacological alleviation of cardiac remodeling and heart failure, but their use is associated with cardiodepressant side effects. Recent advances in our understanding of seven transmembrane receptor signaling show that it is possible to design ligands with "functional selectivity," acting as agonists on certain signaling pathways while antagonizing others. This represents a major pharmaceutical opportunity to separate desired from adverse effects governed by the same receptor. Accordingly, functionally selective ligands are currently pursued as next-generation drugs for superior treatment of heart failure.
Collapse
Affiliation(s)
- Gitte Lund Christensen
- Department of Clinical Biochemistry, Glostrup Research Institute, Glostrup Hospital, DK-2600 Glostrup, Denmark
| | | | | |
Collapse
|
49
|
Jeppesen PL, Christensen GL, Schneider M, Nossent AY, Jensen HB, Andersen DC, Eskildsen T, Gammeltoft S, Hansen JL, Sheikh SP. Angiotensin II type 1 receptor signalling regulates microRNA differentially in cardiac fibroblasts and myocytes. Br J Pharmacol 2012; 164:394-404. [PMID: 21449976 DOI: 10.1111/j.1476-5381.2011.01375.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The angiotensin II type 1 receptor (AT(1)R) is a key regulator of blood pressure and cardiac contractility and is profoundly involved in development of cardiac disease. Since several microRNAs (miRNAs) have been implicated in cardiac disease, we determined whether miRNAs might be regulated by AT(1)R signals in a Gαq/11-dependent or -independent manner. EXPERIMENTAL APPROACH We performed a global miRNA array analysis of angiotensin II (Ang II)-mediated miRNA regulation in HEK293N cells overexpressing the AT(1)R and focused on separating the role of Gαq/11-dependent and -independent pathways. MiRNA regulation was verified with quantitative PCR in both HEK293N cells and primary cardiac myocytes and fibroblasts. KEY RESULTS Our studies revealed five miRNAs (miR-29b, -129-3p, -132, -132* and -212) that were up-regulated by Ang II in HEK293N cells. In contrast, the biased Ang II analogue, [Sar1, Ile4, Ile8] Ang II (SII Ang II), which selectively activates Gαq/11-independent signalling, failed to regulate miRNAs in HEK293N cells. Furthermore, Ang II-induced miRNA regulation was blocked following Gαq/11 and Mek1 inhibition. The observed Ang II regulation of miRNA was confirmed in primary cultures of adult cardiac fibroblasts. Interestingly, Ang II did not regulate miRNA expression in cardiac myocytes, but SII Ang II significantly down-regulated miR-129-3p. CONCLUSIONS AND IMPLICATIONS Five miRNAs were regulated by Ang II through mechanisms depending on Gαq/11 and Erk1/2 activation. These miRNAs may be involved in Ang II-mediated cardiac biology and disease, as several of these miRNAs have previously been associated with cardiovascular disease and were found to be regulated in cardiac cells.
Collapse
Affiliation(s)
- Pia Lindgren Jeppesen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark and Department of Clinical Biochemistry & Pharmacology, Odense University Hospital, Odense, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bhakar AL, Dölen G, Bear MF. The pathophysiology of fragile X (and what it teaches us about synapses). Annu Rev Neurosci 2012; 35:417-43. [PMID: 22483044 DOI: 10.1146/annurev-neuro-060909-153138] [Citation(s) in RCA: 279] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fragile X is the most common known inherited cause of intellectual disability and autism, and it typically results from transcriptional silencing of FMR1 and loss of the encoded protein, FMRP (fragile X mental retardation protein). FMRP is an mRNA-binding protein that functions at many synapses to inhibit local translation stimulated by metabotropic glutamate receptors (mGluRs) 1 and 5. Recent studies on the biology of FMRP and the signaling pathways downstream of mGluR1/5 have yielded deeper insight into how synaptic protein synthesis and plasticity are regulated by experience. This new knowledge has also suggested ways that altered signaling and synaptic function can be corrected in fragile X, and human clinical trials based on this information are under way.
Collapse
Affiliation(s)
- Asha L Bhakar
- Howard Hughes Medical Institute, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | | | | |
Collapse
|