1
|
Montezano AC, Kuriakose J, Hood KY, Sin YY, Camargo LL, Namkung Y, Castro CH, Santos RA, Alves-Lopes R, Tejeda G, Passaglia P, Basheer S, Gallen E, Findlay JE, Awan FR, Laporte SA, MacLean MR, Baillie GS, Touyz RM. Ang-(1-7) and ET-1 Interplay Through Mas and ET B Receptor Interaction Defines a Novel Vasoprotective Mechanism. Hypertension 2025; 82:267-281. [PMID: 39633565 DOI: 10.1161/hypertensionaha.124.22693] [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: 01/10/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Ang-(1-7) (angiotensin (1-7)) via MasR (Mas receptor) opposes vaso-injurious actions of Ang II (angiotensin II) as shown in models of pulmonary hypertension. The underlying mechanisms remain unclear. We hypothesized cross talk between Ang-(1-7) and the protective arm of the ET-1 (endothelin-1) system involving MasR and ETBR (endothelin receptor type B). METHODS To address this, we studied multiple models: in vivo, in a mouse model of ET-1-associated vascular injury (hypoxia-induced pulmonary hypertension); ex vivo, in isolated mouse arteries; and in vitro, in human endothelial cells. RESULTS Pulmonary hypertension mice exhibited pulmonary vascular remodeling, endothelial dysfunction, and ET-1-induced hypercontractility. Ang-(1-7) treatment (14 days) ameliorated these effects and increased the expression of vascular ETBR. In human endothelial cells, Ang-(1-7)-induced activation of eNOS (endothelial NO synthase)/NO was attenuated by A779 (MasR antagonist) and BQ788 (ETBR antagonist). A779 inhibited ET-1-induced signaling. Coimmunoprecipitation and peptide array experiments demonstrated the interaction between MasR and ETBR. Binding sites for ETBR were mapped to MasR (amino acids 290-314). Binding sites for MasR on ETBR were identified (amino acids 176-200). Peptides that disrupt MasR:ETBR prevented Ang-(1-7) and ET-1 signaling. Using high-throughput screening, we identified compounds that enhance MasR:ETBR interaction, which we termed enhancers. Enhancers increased Ang-(1-7)-induced eNOS activity, NO production, and Ang-(1-7)-mediated vasorelaxation, and reduced contractile responses. CONCLUSIONS We identify cross talk between Ang-(1-7) and ET-1 through MasR:ETBR interaction as a novel network that is vasoprotective. Promoting coactivity between these systems amplifies Ang-(1-7) signaling, increases ET-1/ETBR-mediated vascular actions, and attenuates the injurious effects of ET-1. Enhancing Ang-(1-7)/MasR:ET-1/ETBR signaling may have therapeutic potential in conditions associated with vascular damage.
Collapse
Affiliation(s)
- Augusto C Montezano
- Research Institute of McGill University Health Centre, Montreal, Quebec, Canada (A.C.M., L.L.C., Y.N., S.A.L., R.M.T.)
| | - Jithin Kuriakose
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (J.K., K.Y.H., Y.Y.S., G.T., E.G., J.E.F., G.S.B.)
| | - Katie Y Hood
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (J.K., K.Y.H., Y.Y.S., G.T., E.G., J.E.F., G.S.B.)
| | - Yuan Yan Sin
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (J.K., K.Y.H., Y.Y.S., G.T., E.G., J.E.F., G.S.B.)
| | - Livia L Camargo
- Research Institute of McGill University Health Centre, Montreal, Quebec, Canada (A.C.M., L.L.C., Y.N., S.A.L., R.M.T.)
| | - Yoon Namkung
- Research Institute of McGill University Health Centre, Montreal, Quebec, Canada (A.C.M., L.L.C., Y.N., S.A.L., R.M.T.)
| | - Carlos H Castro
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology, Department of Physiological Sciences, Federal University of Goiás, Goiania, Brazil (C.H.C.)
| | - Robson A Santos
- School of Medicine, University of Aberdeen, United Kingdom (R.A.S.)
| | - Rheure Alves-Lopes
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Federal University of Minas Gerais, Belo Horizonte, Brazil (R.A.)
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Brazil (R.A.-L.)
| | - Gonzalo Tejeda
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (J.K., K.Y.H., Y.Y.S., G.T., E.G., J.E.F., G.S.B.)
| | - Patricia Passaglia
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan (P.P.)
| | - Sehrish Basheer
- Department of Pharmacology and Therapeutics (S.B., F.R.A., S.A.L.), McGill University, Montreal, Quebec, Canada
| | - Emily Gallen
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (J.K., K.Y.H., Y.Y.S., G.T., E.G., J.E.F., G.S.B.)
| | - Jane E Findlay
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (J.K., K.Y.H., Y.Y.S., G.T., E.G., J.E.F., G.S.B.)
| | - Fazli R Awan
- Department of Pharmacology and Therapeutics (S.B., F.R.A., S.A.L.), McGill University, Montreal, Quebec, Canada
| | - Stéphane A Laporte
- Research Institute of McGill University Health Centre, Montreal, Quebec, Canada (A.C.M., L.L.C., Y.N., S.A.L., R.M.T.)
- Department of Pharmacology and Therapeutics (S.B., F.R.A., S.A.L.), McGill University, Montreal, Quebec, Canada
- Department of Medicine (S.A.L., R.M.T.), McGill University, Montreal, Quebec, Canada
| | - Margaret R MacLean
- Department of Family Medicine (M.R.M., R.M.T.), McGill University, Montreal, Quebec, Canada
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasglow, Scotland, United Kingdom (M.M.)
| | - George S Baillie
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (J.K., K.Y.H., Y.Y.S., G.T., E.G., J.E.F., G.S.B.)
| | - Rhian M Touyz
- Research Institute of McGill University Health Centre, Montreal, Quebec, Canada (A.C.M., L.L.C., Y.N., S.A.L., R.M.T.)
- Department of Medicine (S.A.L., R.M.T.), McGill University, Montreal, Quebec, Canada
- Department of Family Medicine (M.R.M., R.M.T.), McGill University, Montreal, Quebec, Canada
| |
Collapse
|
2
|
Steckelings UM, Widdop RE, Sturrock ED, Lubbe L, Hussain T, Kaschina E, Unger T, Hallberg A, Carey RM, Sumners C. The Angiotensin AT 2 Receptor: From a Binding Site to a Novel Therapeutic Target. Pharmacol Rev 2022; 74:1051-1135. [PMID: 36180112 PMCID: PMC9553111 DOI: 10.1124/pharmrev.120.000281] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/19/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Discovered more than 30 years ago, the angiotensin AT2 receptor (AT2R) has evolved from a binding site with unknown function to a firmly established major effector within the protective arm of the renin-angiotensin system (RAS) and a target for new drugs in development. The AT2R represents an endogenous protective mechanism that can be manipulated in the majority of preclinical models to alleviate lung, renal, cardiovascular, metabolic, cutaneous, and neural diseases as well as cancer. This article is a comprehensive review summarizing our current knowledge of the AT2R, from its discovery to its position within the RAS and its overall functions. This is followed by an in-depth look at the characteristics of the AT2R, including its structure, intracellular signaling, homo- and heterodimerization, and expression. AT2R-selective ligands, from endogenous peptides to synthetic peptides and nonpeptide molecules that are used as research tools, are discussed. Finally, we summarize the known physiological roles of the AT2R and its abundant protective effects in multiple experimental disease models and expound on AT2R ligands that are undergoing development for clinical use. The present review highlights the controversial aspects and gaps in our knowledge of this receptor and illuminates future perspectives for AT2R research. SIGNIFICANCE STATEMENT: The angiotensin AT2 receptor (AT2R) is now regarded as a fully functional and important component of the renin-angiotensin system, with the potential of exerting protective actions in a variety of diseases. This review provides an in-depth view of the AT2R, which has progressed from being an enigma to becoming a therapeutic target.
Collapse
Affiliation(s)
- U Muscha Steckelings
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert E Widdop
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Edward D Sturrock
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Lizelle Lubbe
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Tahir Hussain
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Elena Kaschina
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Thomas Unger
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Anders Hallberg
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert M Carey
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Colin Sumners
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| |
Collapse
|
3
|
Delaitre C, Boisbrun M, Lecat S, Dupuis F. Targeting the Angiotensin II Type 1 Receptor in Cerebrovascular Diseases: Biased Signaling Raises New Hopes. Int J Mol Sci 2021; 22:ijms22136738. [PMID: 34201646 PMCID: PMC8269339 DOI: 10.3390/ijms22136738] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/17/2021] [Accepted: 06/20/2021] [Indexed: 12/20/2022] Open
Abstract
The physiological and pathophysiological relevance of the angiotensin II type 1 (AT1) G protein-coupled receptor no longer needs to be proven in the cardiovascular system. The renin–angiotensin system and the AT1 receptor are the targets of several classes of therapeutics (such as angiotensin converting enzyme inhibitors or angiotensin receptor blockers, ARBs) used as first-line treatments in cardiovascular diseases. The importance of AT1 in the regulation of the cerebrovascular system is also acknowledged. However, despite numerous beneficial effects in preclinical experiments, ARBs do not induce satisfactory curative results in clinical stroke studies. A better understanding of AT1 signaling and the development of biased AT1 agonists, able to selectively activate the β-arrestin transduction pathway rather than the Gq pathway, have led to new therapeutic strategies to target detrimental effects of AT1 activation. In this paper, we review the involvement of AT1 in cerebrovascular diseases as well as recent advances in the understanding of its molecular dynamics and biased or non-biased signaling. We also describe why these alternative signaling pathways induced by β-arrestin biased AT1 agonists could be considered as new therapeutic avenues for cerebrovascular diseases.
Collapse
Affiliation(s)
- Céline Delaitre
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France;
- Biotechnologie et Signalisation Cellulaire, UMR7242 CNRS/Université de Strasbourg, 300 Boulevard Sébastien Brant, CS 10413, CEDEX, 67412 Illkirch-Graffenstaden, France;
| | | | - Sandra Lecat
- Biotechnologie et Signalisation Cellulaire, UMR7242 CNRS/Université de Strasbourg, 300 Boulevard Sébastien Brant, CS 10413, CEDEX, 67412 Illkirch-Graffenstaden, France;
| | - François Dupuis
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France;
- Correspondence: ; Tel.: +33-372747272
| |
Collapse
|
4
|
González-Blázquez R, Alcalá M, Fernández-Alfonso MS, Steckelings UM, Lorenzo MP, Viana M, Boisvert WA, Unger T, Gil-Ortega M, Somoza B. C21 preserves endothelial function in the thoracic aorta from DIO mice: role for AT2, Mas and B2 receptors. Clin Sci (Lond) 2021; 135:1145-1163. [PMID: 33899912 DOI: 10.1042/cs20210049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023]
Abstract
Compound 21 (C21), a selective agonist of angiotensin II type 2 receptor (AT2R), induces vasodilation through NO release. Since AT2R seems to be overexpressed in obesity, we hypothesize that C21 prevents the development of obesity-related vascular alterations. The main goal of the present study was to assess the effect of C21 on thoracic aorta endothelial function in a model of diet-induced obesity (DIO) and to elucidate the potential cross-talk among AT2R, Mas receptor (MasR) and/or bradykinin type 2 receptor (B2R) in this response. Five-week-old male C57BL6J mice were fed a standard (CHOW) or a high-fat diet (HF) for 6 weeks and treated daily with C21 (1 mg/kg p.o) or vehicle, generating four groups: CHOW-C, CHOW-C21, HF-C, HF-C21. Vascular reactivity experiments were performed in thoracic aorta rings. Human endothelial cells (HECs; EA.hy926) were used to elucidate the signaling pathways, both at receptor and intracellular levels. Arteries from HF mice exhibited increased contractions to Ang II than CHOW mice, effect that was prevented by C21. PD123177, A779 and HOE-140 (AT2R, Mas and B2R antagonists) significantly enhanced Ang II-induced contractions in CHOW but not in HF-C rings, suggesting a lack of functionality of those receptors in obesity. C21 prevented those alterations and favored the formation of AT2R/MasR and MasR/B2R heterodimers. HF mice also exhibited impaired relaxations to acetylcholine (ACh) due to a reduced NO availability. C21 preserved NO release through PKA/p-eNOS and AKT/p-eNOS signaling pathways. In conclusion, C21 favors the interaction among AT2R, MasR and B2R and prevents the development of obesity-induced endothelial dysfunction by stimulating NO release through PKA/p-eNOS and AKT/p-eNOS signaling pathways.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Diet, High-Fat
- Drug Evaluation, Preclinical
- Endothelium, Vascular/drug effects
- Human Umbilical Vein Endothelial Cells
- Humans
- Imidazoles/pharmacology
- Imidazoles/therapeutic use
- Male
- Mice, Inbred C57BL
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Obesity/complications
- Obesity/metabolism
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor Cross-Talk
- Receptor, Angiotensin, Type 2/agonists
- Receptor, Angiotensin, Type 2/metabolism
- Receptor, Bradykinin B2/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Renin-Angiotensin System/drug effects
- Signal Transduction/drug effects
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Thiophenes/pharmacology
- Thiophenes/therapeutic use
- Vascular Diseases/etiology
- Vascular Diseases/metabolism
- Vascular Diseases/prevention & control
- Mice
Collapse
Affiliation(s)
- Raquel González-Blázquez
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid 28925, Spain
| | - Martín Alcalá
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid 28925, Spain
| | - María S Fernández-Alfonso
- Instituto Pluridisciplinar, Unidad de Cartografía Cerebral, Universidad Complutense de Madrid, Madrid 28040, Spain
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Ulrike Muscha Steckelings
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - M Paz Lorenzo
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid 28925, Spain
| | - Marta Viana
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid 28925, Spain
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, BSB311, Honolulu, HI 96813, U.S.A
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Str., Kazan 420008, Russia
| | - Thomas Unger
- CARIM - School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid 28925, Spain
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid 28925, Spain
| |
Collapse
|
5
|
Rivas-Santisteban R, Lillo J, Muñoz A, Rodríguez-Pérez AI, Labandeira-García JL, Navarro G, Franco R. Novel Interactions Involving the Mas Receptor Show Potential of the Renin-Angiotensin system in the Regulation of Microglia Activation: Altered Expression in Parkinsonism and Dyskinesia. Neurotherapeutics 2021; 18:998-1016. [PMID: 33474655 PMCID: PMC7817140 DOI: 10.1007/s13311-020-00986-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 12/30/2022] Open
Abstract
The renin-angiotensin system (RAS) not only plays an important role in controlling blood pressure but also participates in almost every process to maintain homeostasis in mammals. Interest has recently increased because SARS viruses use one RAS component (ACE2) as a target-cell receptor. The occurrence of RAS in the basal ganglia suggests that the system may be targeted to improve the therapy of neurodegenerative diseases. RAS-related data led to the hypothesis that RAS receptors may interact with each other. The aim of this paper was to find heteromers formed by Mas and angiotensin receptors and to address their functionality in neurons and microglia. Novel interactions were discovered by using resonance energy transfer techniques. The functionality of individual and interacting receptors was assayed by measuring levels of the second messengers cAMP and Ca2+ in transfected human embryonic kidney cells (HEK-293T) and primary cultures of striatal cells. Receptor complex expression was assayed by in situ proximity ligation assay. Functionality and expression were assayed in parallel in primary cultures of microglia treated or not with lipopolysaccharide and interferon-γ (IFN-γ). The proximity ligation assay was used to assess heteromer expression in parkinsonian and dyskinetic conditions. Complexes formed by Mas and the angiotensin AT1 or AT2 receptors were identified in both a heterologous expression system and in neural primary cultures. In the heterologous system, we showed that the three receptors-MasR, AT1R, and AT2R-can interact to form heterotrimers. The expression of receptor dimers (AT1R-MasR or AT2R-MasR) was higher in microglia than in neurons and was differentially affected upon microglial activation with lipopolysaccharide and IFN-γ. In all cases, agonist-induced signaling was reduced upon coactivation, and in some cases just by coexpression. Also, the blockade of signaling of two receptors in a complex by the action of a given (selective) receptor antagonist (cross-antagonism) was often observed. Differential expression of the complexes was observed in the striatum under parkinsonian conditions and especially in animals rendered dyskinetic by levodopa treatment. The negative modulation of calcium mobilization (mediated by AT1R activation), the multiplicity of possibilities on RAS affecting the MAPK pathway, and the disbalanced expression of heteromers in dyskinesia yield new insight into the operation of the RAS system, how it becomes unbalanced, and how a disbalanced RAS can be rebalanced. Furthermore, RAS components in activated microglia warrant attention in drug-development approaches to address neurodegeneration.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Corpus Striatum/drug effects
- Corpus Striatum/metabolism
- Dyskinesia, Drug-Induced/metabolism
- HEK293 Cells
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Microglia/drug effects
- Microglia/metabolism
- Oxidopamine/toxicity
- Parkinsonian Disorders/chemically induced
- Parkinsonian Disorders/metabolism
- Proto-Oncogene Mas/agonists
- Proto-Oncogene Mas/metabolism
- Rats
- Rats, Wistar
- Receptor, Angiotensin, Type 1/agonists
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/agonists
- Receptor, Angiotensin, Type 2/metabolism
- Renin-Angiotensin System/drug effects
- Renin-Angiotensin System/physiology
Collapse
Affiliation(s)
- Rafael Rivas-Santisteban
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Diagonal 643, Barcelona, Catalonia, 08028, Spain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
| | - Jaume Lillo
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Diagonal 643, Barcelona, Catalonia, 08028, Spain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
| | - Ana Muñoz
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Ana I Rodríguez-Pérez
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - José Luís Labandeira-García
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, University of Barcelona, Barcelona, Catalonia, 08028, Spain
| | - Rafael Franco
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Diagonal 643, Barcelona, Catalonia, 08028, Spain.
| |
Collapse
|
6
|
Sinphitukkul K, Manotham K, Eiam-Ong S, Eiam-Ong S. Aldosterone nongenomically induces angiotensin II receptor dimerization in rat kidney: role of mineralocorticoid receptor and NADPH oxidase. Arch Med Sci 2019; 15:1589-1598. [PMID: 31749889 PMCID: PMC6855162 DOI: 10.5114/aoms.2019.87135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/01/2017] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Previous in vitro studies demonstrated that aldosterone nongenomically induces transglutaminase (TG) and reactive oxygen species (ROS), which enhanced angiotensin II receptor (ATR) dimerization. There are no in vivo data in the kidney. MATERIAL AND METHODS Male Wistar rats were intraperitoneally injected with normal saline solution, or aldosterone (Aldo: 150 μg/kg BW); or received pretreatment with eplerenone (mineralocorticoid receptor (MR) blocker, Ep. + Aldo), or with apocynin (nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor, Apo. + Aldo) 30 min before aldosterone. Thirty minutes after aldosterone injection, protein abundances of dimeric and monomeric forms of AT1R and AT2R, and protein abundances and localizations of TG2 and p47phox, a cytosolic subunit of NADPH oxidase, were determined by Western blot analysis and immunohistochemistry, respectively. RESULTS Protein abundances of dimeric forms of AT1R and AT2R were enhanced by 170% and 70%, respectively. Apocynin could block dimeric forms of both receptors while eplerenone inhibited only AT2R. Monomeric protein levels of both receptors were maintained. Aldosterone significantly enhanced TG2 and p47phox protein abundances, which were blunted by eplerenone or apocynin. Aldosterone stimulated p47phox protein expression in both the cortex and the medulla while TG2 was induced mostly in the medulla. Eplerenone or apocynin normalized the immunoreactivity of both TG2 and p47phox. CONCLUSIONS This is the first in vivo study demonstrating that aldosterone nongenomically increases renal TG2 and p47phox protein expression and then activates AT1R and AT2R dimerizations. Aldosterone-stimulated AT1R and AT2R dimerizations are mediated through activation of NADPH oxidase. Aldosterone-induced AT1R dimer formation is an MR-independent pathway, whereas the formation of AT2R dimer is modulated in an MR-dependent manner.
Collapse
Affiliation(s)
| | - Krissanapong Manotham
- Molecular and Cell Biology Unit, Department of Medicine, Lerdsin General Hospital, Bangkok, Thailand
| | - Somchai Eiam-Ong
- Department of Medicine, Division of Nephrology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Somchit Eiam-Ong
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
7
|
Erol I, Cosut B, Durdagi S. Toward Understanding the Impact of Dimerization Interfaces in Angiotensin II Type 1 Receptor. J Chem Inf Model 2019; 59:4314-4327. [PMID: 31429557 DOI: 10.1021/acs.jcim.9b00294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Angiotensin II type 1 receptor (AT1R) is a prototypical class A G protein-coupled receptor (GPCR) that has an important role in cardiovascular pathologies and blood pressure regulation as well as in the central nervous system. GPCRs may exist and function as monomers; however, they can assemble to form higher order structures, and as a result of oligomerization, their function and signaling profiles can be altered. In the case of AT1R, the classical Gαq/11 pathway is initiated with endogenous agonist angiotensin II binding. A variety of cardiovascular pathologies such as heart failure, diabetic nephropathy, atherosclerosis, and hypertension are associated with this pathway. Recent findings reveal that AT1R can form homodimers and activate the noncanonical (β-arrestin-mediated) pathway. Nevertheless, the exact dimerization interface and atomic details of AT1R homodimerization have not been still elucidated. Here, six different symmetrical dimer interfaces of AT1R are considered, and homodimers were constructed using other published GPCR crystal dimer interfaces as template structures. These AT1R homodimers were then inserted into the model membrane bilayers and subjected to all-atom molecular dynamics simulations. Our simulation results along with the principal component analysis and water pathway analysis suggest four different interfaces as the most plausible: symmetrical transmembrane (TM)1,2,8; TM5; TM4; and TM4,5 AT1R dimer interfaces that consist of one inactive and one active protomer. Moreover, we identified ILE2386.33 as a hub residue in the stabilization of the inactive state of AT1R.
Collapse
Affiliation(s)
- Ismail Erol
- Department of Chemistry , Gebze Technical University , Gebze 41400 , Kocaeli , Turkey
| | - Bunyemin Cosut
- Department of Chemistry , Gebze Technical University , Gebze 41400 , Kocaeli , Turkey
| | | |
Collapse
|
8
|
Abstract
OBJECTIVE Epidemiological studies confirm that hypertensive patients respond differently to renin-angiotensin system (RAS) inhibition depending on their gender. The aim of present work is to focus on sex-dependent differences in RAS regulation under conditions of increased salt intake. METHOD To investigate RAS, we measured the expression of angiotensinogen (Agt) mRNA, angiotensin receptor type 1 (AT1) mRNA and mitochondria assembly receptor (MasR) in the liver of rats under control conditions and after feeding with a salt diet (2% NaCl). In parallel, vascular endothelial growth factor A (VEGF-A) mRNA was analyzed. RESULTS Regression analysis revealed sex-dependent differences in the correlation between mRNA expression of AT1 and that of Agt, MasR and VEGF-A in both groups. There was a significant negative correlation between AT1 and Agt mRNA expression in the male control group, but this correlation disappeared in males exposed to a salt diet. In females, AT1 and Agt expression correlated only in the group exposed to the salt diet. In control males, there was a borderline trend to correlation between AT1 and MasR mRNA expression. The correlation between AT1 and VEGF-A mRNA expression was significant only in the control females, however, after exposure to a salt diet, this correlation diminished. CONCLUSIONS We hypothesize that RAS components expression is compensated differently in males and females. The observed loss of compensatory relationships in RAS between AT1 and Agt and AT1 and MasR in male rats under a salt diet can contribute to the differences observed in human with hypertension associated with an unhealthy diet.
Collapse
|
9
|
Heteromerization fingerprints between bradykinin B2 and thromboxane TP receptors in native cells. PLoS One 2019; 14:e0216908. [PMID: 31086419 PMCID: PMC6516669 DOI: 10.1371/journal.pone.0216908] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/30/2019] [Indexed: 12/12/2022] Open
Abstract
Bradykinin (BK) and thromboxane-A2 (TX-A2) are two vasoactive mediators that modulate vascular tone and inflammation via binding to their cognate "class A" G-protein coupled receptors (GPCRs), BK-B2 receptors (B2R) and TX-prostanoid receptors (TP), respectively. Both BK and TX-A2 lead to ERK1/2-mediated vascular smooth muscle cell (VSMC) proliferation and/or hypertrophy. While each of B2R and TP could form functional dimers with various GPCRs, the likelihood that B2R-TP heteromerization could contribute to their co-regulation has never been investigated. The main objective of this study was to investigate the mode of B2R and TP interaction in VSMC, and its possible impact on downstream signaling. Our findings revealed synergistically activated ERK1/2 following co-stimulation of rat VSMC with a subthreshold dose of BK and effective doses of the TP stable agonist, IBOP, possibly involving biased agonist signaling. Single detection of each of B2R and TP in VSMC, using in-situ proximity ligation assay (PLA), provided evidence of the constitutive expression of nuclear and extranuclear B2R and TP. Moreover, inspection of B2R-TP PLA signals in VSMC revealed agonist-modulated nuclear and extranuclear proximity between B2R and TP, whose quantification varied substantially following single versus dual agonist stimulations. B2R-TP interaction was further verified by the findings of co-immunoprecipitation (co-IP) analysis of VSMC lysates. To our knowledge, this is the first study that provides evidence supporting the existence of B2R-TP heteromerization fingerprints in primary cultured VSMC.
Collapse
|
10
|
Zha D, Yao T, Bao L, Gao P, Wu X. Telmisartan attenuates diabetic nephropathy progression by inhibiting the dimerization of angiotensin type-1 receptor and adiponectin receptor-1. Life Sci 2019; 221:109-120. [DOI: 10.1016/j.lfs.2019.01.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/25/2019] [Accepted: 01/26/2019] [Indexed: 02/07/2023]
|
11
|
Nematbakhsh M, Mansouri A. Renal vascular response to angiotensin 1-7 in rats: the role of Mas receptor. Res Pharm Sci 2018; 13:177-180. [PMID: 29606972 PMCID: PMC5842489 DOI: 10.4103/1735-5362.223803] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recently a cross talk between angiotensin 1-7 (Ang1-7) receptor (MasR) and angiotensin II receptors types 1 and 2 (AT1R and AT2R) has been highlighted. The effects of MasR antagonist (A779) compared to the vehicle on the renal blood flow (RBF) and renal vascular resistance (RVR) responses to Ang1-7 (300 ng/kg/min) infusion in the absence of Ang II receptors in male and female rats were determined at controlled renal perfusion pressure. Ang1-7 infusion did not alter mean arterial pressure in male and female rats. However, A779 compared to vehicle increased RBF (18% vs 3%) and decreased RVR (13% vs 4%) responses to Ang1-7 infusion significantly (P < 0.05) in male when AngII receptors were blocked. Such observation was not occurred in female animals. Finally it was concluded that renal vascular responses to Ang1-7 administration may not be exerted by MasR in male rats, and these responses are not mediated with AngII receptors.
Collapse
Affiliation(s)
- Mehdi Nematbakhsh
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, I.R. Iran.,Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Azam Mansouri
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
12
|
Leonhardt J, Villela DC, Teichmann A, Münter LM, Mayer MC, Mardahl M, Kirsch S, Namsolleck P, Lucht K, Benz V, Alenina N, Daniell N, Horiuchi M, Iwai M, Multhaup G, Schülein R, Bader M, Santos RA, Unger T, Steckelings UM. Evidence for Heterodimerization and Functional Interaction of the Angiotensin Type 2 Receptor and the Receptor MAS. Hypertension 2017; 69:1128-1135. [PMID: 28461604 DOI: 10.1161/hypertensionaha.116.08814] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/21/2016] [Accepted: 04/06/2017] [Indexed: 11/16/2022]
Abstract
The angiotensin type 2 receptor (AT2R) and the receptor MAS are receptors of the protective arm of the renin-angiotensin system. They mediate strikingly similar actions. Moreover, in various studies, AT2R antagonists blocked the effects of MAS agonists and vice versa. Such cross-inhibition may indicate heterodimerization of these receptors. Therefore, this study investigated the molecular and functional interplay between MAS and the AT2R. Molecular interactions were assessed by fluorescence resonance energy transfer and by cross correlation spectroscopy in human embryonic kidney-293 cells transfected with vectors encoding fluorophore-tagged MAS or AT2R. Functional interaction of AT2R and MAS was studied in astrocytes with CX3C chemokine receptor-1 messenger RNA expression as readout. Coexpression of fluorophore-tagged AT2R and MAS resulted in a fluorescence resonance energy transfer efficiency of 10.8 ± 0.8%, indicating that AT2R and MAS are capable to form heterodimers. Heterodimerization was verified by competition experiments using untagged AT2R and MAS. Specificity of dimerization of AT2R and MAS was supported by lack of dimerization with the transient receptor potential cation channel, subfamily C-member 6. Dimerization of the AT2R was abolished when it was mutated at cysteine residue 35. AT2R and MAS stimulation with the respective agonists, Compound 21 or angiotensin-(1-7), significantly induced CX3C chemokine receptor-1 messenger RNA expression. Effects of each agonist were blocked by an AT2R antagonist (PD123319) and also by a MAS antagonist (A-779). Knockout of a single of these receptors made astrocytes unresponsive for both agonists. Our results suggest that MAS and the AT2R form heterodimers and that-at least in astrocytes-both receptors functionally depend on each other.
Collapse
Affiliation(s)
- Julia Leonhardt
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Daniel C Villela
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Anke Teichmann
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Lisa-Marie Münter
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Magnus C Mayer
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Maibritt Mardahl
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Sebastian Kirsch
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Pawel Namsolleck
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Kristin Lucht
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Verena Benz
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Natalia Alenina
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Nicholas Daniell
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Masatsugu Horiuchi
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Masaru Iwai
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Gerhard Multhaup
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Ralf Schülein
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Michael Bader
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Robson A Santos
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Thomas Unger
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Ulrike Muscha Steckelings
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.).
| |
Collapse
|
13
|
Pernomian L, Pernomian L, Gomes MS, da Silva CH. Pharmacological significance of the interplay between angiotensin receptors: MAS receptors as putative final mediators of the effects elicited by angiotensin AT1 receptors antagonists. Eur J Pharmacol 2015; 769:143-6. [DOI: 10.1016/j.ejphar.2015.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/04/2015] [Accepted: 11/04/2015] [Indexed: 11/28/2022]
|
14
|
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
|
15
|
Abstract
The angiotensin type 2 receptor (AT2R) and the receptor Mas are components of the protective arms of the renin-angiotensin system (RAS), i.e. they both mediate tissue protective and regenerative actions. The spectrum of actions of these two receptors and their signalling mechanisms display striking similarities. Moreover, in some instances, antagonists for one receptor are able to inhibit the action of agonists for the respective other receptor. These observations suggest that there may be a functional or even physical interaction of both receptors. This article discusses potential mechanisms underlying the phenomenon of blockade of angiotensin-(1-7) [Ang-(1-7)] actions by AT2R antagonists and vice versa. Such mechanisms may comprise dimerization of the receptors or dimerization-independent mechanisms such as lack of specificity of the receptor ligands used in the experiments or involvement of the Ang-(1-7) metabolite alamandine and its receptor MrgD in the observed effects. We conclude that evidence for a functional interaction of both receptors is strong, but that such an interaction may be species- and/or tissue-specific and that elucidation of the precise nature of the interaction is only at the very beginning.
Collapse
|
16
|
Nostramo R, Serova L, Laukova M, Tillinger A, Peddu C, Sabban EL. Regulation of nonclassical renin-angiotensin system receptor gene expression in the adrenal medulla by acute and repeated immobilization stress. Am J Physiol Regul Integr Comp Physiol 2015; 308:R517-29. [DOI: 10.1152/ajpregu.00130.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The involvement of the nonclassical renin-angiotensin system (RAS) in the adrenomedullary response to stress is unclear. Therefore, we examined basal and immobilization stress (IMO)-triggered changes in gene expression of the classical and nonclassical RAS receptors in the rat adrenal medulla, specifically the angiotensin II type 2 (AT2) and type 4 (AT4) receptors, (pro)renin receptor [(P)RR], and Mas receptor (MasR). All RAS receptors were identified, with AT2 receptor mRNA levels being the most abundant, followed by the (P)RR, AT1A receptor, AT4 receptor, and MasR. Following a single IMO, AT2 and AT4 receptor mRNA levels decreased by 90 and 50%, respectively. Their mRNA levels were also transiently decreased by repeated IMO. MasR mRNA levels displayed a 75% transient decrease as well. Conversely, (P)RR mRNA levels were increased by 50% following single or repeated IMO. Because of its abundance, the function of the (P)RR was explored in PC-12 cells. Prorenin activation of the (P)RR increased phosphorylation of extracellular signal-regulated kinase 1/2 and tyrosine hydroxylase at Ser31, likely increasing its enzymatic activity and catecholamine biosynthesis. Together, the broad and dynamic changes in gene expression of the nonclassical RAS receptors implicate their role in the intricate response of the adrenomedullary catecholaminergic system to stress.
Collapse
Affiliation(s)
- Regina Nostramo
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| | - Lidia Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| | - Marcela Laukova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| | - Andrej Tillinger
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| | - Chandana Peddu
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| | - Esther L. Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| |
Collapse
|
17
|
da Silva AR, Fraga-Silva RA, Stergiopulos N, Montecucco F, Mach F. Update on the role of angiotensin in the pathophysiology of coronary atherothrombosis. Eur J Clin Invest 2015; 45:274-87. [PMID: 25586671 DOI: 10.1111/eci.12401] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 01/10/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Coronary atherothrombosis due to atherosclerotic plaque rupture or erosion is frequently associated with acute coronary syndromes (ACS). Significant efforts have been made to elucidate the pathophysiological mechanisms underlying acute coronary events. MATERIALS AND METHODS This narrative review is based on the material searched for and obtained via PubMed up to August 2014. The search terms we used were as follows: 'angiotensin, acute coronary syndromes, acute myocardial infarction' in combination with 'atherosclerosis, vulnerability, clinical trial, ACE inhibitors, inflammation'. RESULTS Among several regulatory components, the renin-angiotensin system (RAS) was shown as a key pathway modulating coronary atherosclerotic plaque vulnerability. Indeed, these molecules are involved in all stages of atherogenesis. Classically, the RAS is composed by a series of enzymatic reactions leading to the angiotensin (Ang) II generation and activity. However, the knowledge of RAS has expanded and become more complex. The discovery of novel components and their functions has revealed additional pathways that contribute to or counterbalance the actions of Ang II. In this review, we discussed on recent findings concerning the role of different angiotensin peptides in the pathophysiology of ACS and coronary atherothrombosis, exploring the link between these molecules and atherosclerotic plaque vulnerability. CONCLUSIONS Treatments selectively targeting angiotensins (including Mas and AT2 agonists, ACE2 recombinant, or Ang-(1-7) and almandine in oral formulations) have been tested in animal studies or in small human subgroups, expanding the perspective in the ACS prevention. These novel strategies, especially in the counter-regulatory axis ACE2/Ang-(1-7)/Mas, might be promising to reduce plaque vulnerability and inflammation.
Collapse
Affiliation(s)
- Analina R da Silva
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
18
|
Wagenaar GTM, Sengers RMA, Laghmani EH, Chen X, Lindeboom MPHA, Roks AJM, Folkerts G, Walther FJ. Angiotensin II type 2 receptor ligand PD123319 attenuates hyperoxia-induced lung and heart injury at a low dose in newborn rats. Am J Physiol Lung Cell Mol Physiol 2014; 307:L261-72. [PMID: 24951776 DOI: 10.1152/ajplung.00345.2013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Intervening in angiotensin (Ang)-II type 2 receptor (AT2) signaling may have therapeutic potential for bronchopulmonary dysplasia (BPD) by attenuating lung inflammation and preventing arterial hypertension (PAH)-induced right ventricular hypertrophy (RVH). We first investigated the role of AT2 inhibition with PD123319 (0.5 and 2 mg·kg(-1)·day(-1)) on the beneficial effect of AT2 agonist LP2-3 (5 μg/kg twice a day) on RVH in newborn rats with hyperoxia-induced BPD. Next we determined the cardiopulmonary effects of PD123319 (0.1 mg·kg(-1)·day(-1)) in two models: early treatment during continuous exposure to hyperoxia for 10 days and late treatment starting on day 6 in rat pups exposed postnatally to hyperoxia for 9 days, followed by a 9-day recovery period in room air. Parameters investigated included lung and heart histopathology, fibrin deposition, vascular leakage, and differential mRNA expression. Ten days of coadministration of LP2-3 and PD123319 abolished the beneficial effects of LP2-3 on RVH in experimental BPD. In the early treatment model PD123319 attenuated cardiopulmonary injury by reducing alveolar septal thickness, pulmonary influx of inflammatory cells, including macrophages and neutrophils, medial wall thickness of small arterioles, and extravascular collagen III deposition, and by preventing RVH. In the late treatment model PD123319 diminished PAH and RVH, demonstrating that PAH is reversible in the neonatal period. At high concentrations PD123319 blocks the beneficial effects of the AT2-agonist LP2-3 on RVH. At low concentrations PD123319 attenuates cardiopulmonary injury by reducing pulmonary inflammation and fibrosis and preventing PAH-induced RVH but does not affect alveolar and vascular development in newborn rats with experimental BPD.
Collapse
Affiliation(s)
- Gerry T M Wagenaar
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands;
| | - Rozemarijn M A Sengers
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - El Houari Laghmani
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Xueyu Chen
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Melissa P H A Lindeboom
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anton J M Roks
- Division of Vascular Disease and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gert Folkerts
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; and
| | - Frans J Walther
- Department of Pediatrics, Division of Neonatology, Leiden University Medical Center, Leiden, The Netherlands; Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California
| |
Collapse
|
19
|
Lee LTO, Ng SYL, Chu JYS, Sekar R, Harikumar KG, Miller LJ, Chow BKC. Transmembrane peptides as unique tools to demonstrate the in vivo action of a cross-class GPCR heterocomplex. FASEB J 2014; 28:2632-44. [PMID: 24599969 DOI: 10.1096/fj.13-246868] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Angiotensin (ANGII) and secretin (SCT) share overlapping, interdependent osmoregulatory functions in brain, where SCT peptide/receptor function is required for ANGII action, yet the molecular basis is unknown. Since receptors for these peptides (AT1aR, SCTR) are coexpressed in osmoregulatory centers, a possible mechanism is formation of a cross-class receptor heterocomplex. Here, we demonstrate such a complex and its functional importance to modulate signaling. Association of AT1aR with SCTR reduced ability of SCT to stimulate cyclic adenosine monophosphate (cAMP), with signaling augmented in presence of ANGII or constitutively active AT1aR. Several transmembrane (TM) peptides of these receptors were able to affect their conformation within complexes, reducing receptor BRET signals. AT1aR TM1 affected only formation and activity of the heterocomplex, without effect on homomers of either receptor, and reduced SCT-stimulated cAMP responses in cells expressing both receptors. This peptide was active in vivo by injection into mouse lateral ventricle, thereby suppressing water-drinking behavior after hyperosmotic shock, similar to SCTR knockouts. This supports the interpretation that active conformation of AT1aR is a key modulator of cAMP responses induced by SCT stimulation of SCTR. The SCTR/AT1aR complex is physiologically important, providing differential signaling to SCT in settings of hyperosmolality or food intake, modulated by differences in levels of ANGII.
Collapse
Affiliation(s)
- Leo T O Lee
- School of Biological Sciences, University of Hong Kong, Hong Kong, China; and
| | - Stephanie Y L Ng
- School of Biological Sciences, University of Hong Kong, Hong Kong, China; and
| | - Jessica Y S Chu
- School of Biological Sciences, University of Hong Kong, Hong Kong, China; and
| | - Revathi Sekar
- School of Biological Sciences, University of Hong Kong, Hong Kong, China; and
| | - Kaleeckal G Harikumar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona, USA
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona, USA
| | - Billy K C Chow
- School of Biological Sciences, University of Hong Kong, Hong Kong, China; and
| |
Collapse
|
20
|
Nakamura Y, Ishii J, Kondo A. Rapid, Facile Detection of Heterodimer Partners for Target Human G-Protein-Coupled Receptors Using a Modified Split-Ubiquitin Membrane Yeast Two-Hybrid System. PLoS One 2013; 8:e66793. [PMID: 23805278 PMCID: PMC3689660 DOI: 10.1371/journal.pone.0066793] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 05/11/2013] [Indexed: 12/03/2022] Open
Abstract
Potentially immeasurable heterodimer combinations of human G-protein-coupled receptors (GPCRs) result in a great deal of physiological diversity and provide a new opportunity for drug discovery. However, due to the existence of numerous combinations, the sets of GPCR dimers are almost entirely unknown and thus their dominant roles are still poorly understood. Thus, the identification of GPCR dimer pairs has been a major challenge. Here, we established a specialized method to screen potential heterodimer partners of human GPCRs based on the split-ubiquitin membrane yeast two-hybrid system. We demonstrate that the mitogen-activated protein kinase (MAPK) signal-independent method can detect ligand-induced conformational changes and rapidly identify heterodimer partners for target GPCRs. Our data present the abilities to apply for the intermolecular mapping of interactions among GPCRs and to uncover potential GPCR targets for the development of new therapeutic agents.
Collapse
Affiliation(s)
- Yasuyuki Nakamura
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe, Japan
| | - Jun Ishii
- Organization of Advanced Science and Technology, Kobe University, Kobe, Japan
| | - Akihiko Kondo
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe, Japan
- * E-mail:
| |
Collapse
|
21
|
Pena-Silva RA, Faraci FM, Heistad DD. Response to letter regarding article, "Impact of ACE2 deficiency and oxidative stress on cerebrovascular function with aging". Stroke 2013; 44:e35. [PMID: 23646368 DOI: 10.1161/strokeaha.111.000481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
Wilson PC, Lee MH, Appleton KM, El-Shewy HM, Morinelli TA, Peterson YK, Luttrell LM, Jaffa AA. The arrestin-selective angiotensin AT1 receptor agonist [Sar1,Ile4,Ile8]-AngII negatively regulates bradykinin B2 receptor signaling via AT1-B2 receptor heterodimers. J Biol Chem 2013; 288:18872-84. [PMID: 23661707 DOI: 10.1074/jbc.m113.472381] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The renin-angiotensin and kallikrein-kinin systems are key regulators of vascular tone and inflammation. Angiotensin II, the principal effector of the renin-angiotensin system, promotes vasoconstriction by activating angiotensin AT1 receptors. The opposing effects of the kallikrein-kinin system are mediated by bradykinin acting on B1 and B2 bradykinin receptors. The renin-angiotensin and kallikrein-kinin systems engage in cross-talk at multiple levels, including the formation of AT1-B2 receptor heterodimers. In primary vascular smooth muscle cells, we find that the arrestin pathway-selective AT1 agonist, [Sar(1),Ile(4),Ile(8)]-AngII, but not the neutral AT1 antagonist, losartan, inhibits endogenous B2 receptor signaling. In a transfected HEK293 cell model that recapitulates this effect, we find that the actions of [Sar(1),Ile(4), Ile(8)]-AngII require the AT1 receptor and result from arrestin-dependent co-internalization of AT1-B2 heterodimers. BRET50 measurements indicate that AT1 and B2 receptors efficiently heterodimerize. In cells expressing both receptors, pretreatment with [Sar(1),Ile(4),Ile(8)]-AngII blunts B2 receptor activation of Gq/11-dependent intracellular calcium influx and Gi/o-dependent inhibition of adenylyl cyclase. In contrast, [Sar(1),Ile(4),Ile(8)]-AngII has no effect on B2 receptor ligand affinity or bradykinin-induced arrestin3 recruitment. Both radioligand binding assays and quantitative microscopy-based analysis demonstrate that [Sar(1),Ile(4),Ile(8)]-AngII promotes internalization of AT1-B2 heterodimers. Thus, [Sar(1),Ile(4),Ile(8)]-AngII exerts lateral allosteric modulation of B2 receptor signaling by binding to the orthosteric ligand binding site of the AT1 receptor and promoting co-sequestration of AT1-B2 heterodimers. Given the opposing roles of the renin-angiotensin and kallikrein-kinin systems in vivo, the distinct properties of arrestin pathway-selective and neutral AT1 receptor ligands may translate into different pharmacologic actions.
Collapse
Affiliation(s)
- Parker C Wilson
- Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Hansen JT, Lyngsø C, Speerschneider T, Hansen PBL, Galés C, Weiner DM, Sheikh SP, Burstein ES, Hansen JL. Functional enhancement of AT1R potency in the presence of the TPαR is revealed by a comprehensive 7TM receptor co-expression screen. PLoS One 2013; 8:e58890. [PMID: 23516570 PMCID: PMC3597553 DOI: 10.1371/journal.pone.0058890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 02/07/2013] [Indexed: 01/14/2023] Open
Abstract
Background Functional cross-talk between seven transmembrane (7TM) receptors can dramatically alter their pharmacological properties, both in vitro and in vivo. This represents an opportunity for the development of novel therapeutics that potentially target more specific biological effects while causing fewer adverse events. Although several studies convincingly have established the existence of 7TM receptor cross-talk, little is known about the frequencey and biological significance of this phenomenon. Methodology/Principal Findings To evaluate the extent of synergism in 7TM receptor signaling, we took a comprehensive approach and co-expressed 123 different 7TM receptors together with the angiotensin II type 1 receptor (AT1R) and analyzed how each receptor affected the angiotensin II (AngII) response. To monitor the effect we used integrative receptor activation/signaling assay called Receptor Selection and Amplification Technology (R-SAT). In this screen the thromboxane A2α receptor (TPαR) was the only receptor which significantly enhanced the AngII-mediated response. The TPαR-mediated enhancement of AngII signaling was significantly reduced when a signaling deficient receptor mutant (TPαR R130V) was co-expressed instead of the wild-type TPαR, and was completely blocked both by TPαR antagonists and COX inhibitors inhibiting formation of thromboxane A2 (TXA2). Conclusions/Significance We found a functional enhancement of AT1R only when co-expressed with TPαR, but not with 122 other 7TM receptors. In addition, the TPαR must be functionally active, indicating the AT1R enhancement is mediated by a paracrine mechanism. Since we only found one receptor enhancing AT1R potency, our results suggest that functional augmentation through 7TM receptor cross-talk is a rare event that may require specific conditions to occur.
Collapse
Affiliation(s)
- Jonas Tind Hansen
- Laboratory for Molecular Cardiology, Department of Biomedical Sciences and The Danish National Research Foundation Centre for Cardiac Arrhythmia, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Muñoz MC, Giani JF, Burghi V, Mayer MA, Carranza A, Taira CA, Dominici FP. The Mas receptor mediates modulation of insulin signaling by angiotensin-(1–7). ACTA ACUST UNITED AC 2012; 177:1-11. [DOI: 10.1016/j.regpep.2012.04.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 03/29/2012] [Accepted: 04/23/2012] [Indexed: 02/01/2023]
|
25
|
Impact of kinins in the treatment of cardiovascular diseases. Pharmacol Ther 2012; 135:94-111. [DOI: 10.1016/j.pharmthera.2012.04.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/02/2012] [Indexed: 12/24/2022]
|
26
|
Szalai B, Barkai L, Turu G, Szidonya L, Várnai P, Hunyady L. Allosteric interactions within the AT₁ angiotensin receptor homodimer: role of the conserved DRY motif. Biochem Pharmacol 2012; 84:477-85. [PMID: 22579851 DOI: 10.1016/j.bcp.2012.04.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 04/15/2012] [Accepted: 04/18/2012] [Indexed: 12/20/2022]
Abstract
G protein coupled receptor (GPCR) dimerization has a remarkable impact on the diversity of receptor signaling. Allosteric communication between the protomers of the dimer can alter ligand binding, receptor conformation and interactions with different effector proteins. In this study we investigated the allosteric interactions between wild type and mutant protomers of type 1 angiotensin receptor (AT₁R) dimers transiently expressed in CHO cells. In our experimental setup, one protomer of the dimer was selectively stimulated and the β-arrestin2 binding and conformation alteration of the other protomer was followed. The interaction between β-arrestin2 and the non-stimulated protomer was monitored through a bioluminescence resonance energy transfer (BRET) based method. To measure the conformational alterations in the non-stimulated protomer directly, we also used a BRET based intramolecular receptor biosensor, which was created by inserting yellow fluorescent protein (YFP) into the 3rd intracellular loop of AT₁R and fusing Renilla luciferase (RLuc) to its C terminal region. We have detected β-arrestin2 binding, and altered conformation of the non-stimulated protomer. The cooperative ligand binding of the receptor homodimer was also observed by radioligand dissociation experiments. Mutation of the conserved DRY sequence in the activated protomer, which is also required for G protein activation, abolished all the observed allosteric effects. These data suggest that allosteric interactions in the homodimers of AT₁R significantly affect the function of the non-stimulated protomer, and the conserved DRY motif has a crucial role in these interactions.
Collapse
Affiliation(s)
- Bence Szalai
- Department of Physiology, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
27
|
Jankowski V, Tölle M, Santos RAS, Günthner T, Krause E, Beyermann M, Welker P, Bader M, Pinheiro SVB, Sampaio WO, Lautner R, Kretschmer A, van der Giet M, Zidek W, Jankowski J. Angioprotectin: an angiotensin II-like peptide causing vasodilatory effects. FASEB J 2011; 25:2987-95. [PMID: 21628446 DOI: 10.1096/fj.11-185470] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The family of angiotensin peptides has been steadily growing in recent years. Most are fragments of angiotensin II (Ang II) with different affinities to the known angiotensin receptors. Here, we describe a novel endogenous Ang II-like octapeptide in plasma from healthy humans and patients with end-stage renal failure, which acts as a stronger agonist at Mas receptors than Ang 1-7. Chromatographic purification and structural analysis by matrix-assisted laser desorption/ionization time-of-flight/time-of-flight (MALDI-TOF/TOF) revealed an Ang II-like octapeptide, angioprotectin, with the sequence Pro-Glu-Val-Tyr-Ile-His-Pro-Phe, which differs from Ang II in Pro¹ and Glu² instead of Asp¹ and Arg². Pro-Glu-Val-Tyr-Ile-His-Pro-Phe in angioprotectin is most likely generated enzymatically from Ang II. Angioprotectin antagonized the contractile actions of Ang II on rat aortic rings. The physiological antagonism of vasoconstrictor actions of Ang II by angioprotectin is mediated by the Mas receptor. Angioprotectin has a stronger affinity to the Mas receptor than Ang-1-7. Plasma concentrations were ~15% of plasma Ang II concentrations in healthy volunteers and up to 50% in patients with renal failure. A commercially available Ang II antibody did not discriminate between angioprotectin and Ang II; thus, angioprotectin can contribute to Ang II concentrations measured by antibody-based assays. This novel peptide is likely to be a relevant component of the human renin-angiotensin-system.
Collapse
Affiliation(s)
- Vera Jankowski
- Medizinische Klinik IV, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Xu P, Sriramula S, Lazartigues E. ACE2/ANG-(1-7)/Mas pathway in the brain: the axis of good. Am J Physiol Regul Integr Comp Physiol 2010; 300:R804-17. [PMID: 21178125 DOI: 10.1152/ajpregu.00222.2010] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The last decade has seen the discovery of several new components of the renin-angiotensin system (RAS). Among them, angiotensin converting enzyme-2 (ACE2) and the Mas receptor have forced a reevaluation of the original cascade and led to the emergence of a new arm of the RAS: the ACE2/ANG-(1-7)/Mas axis. Accordingly, the new system is now seen as a balance between a provasoconstrictor, profibrotic, progrowth axis (ACE/ANG-II/AT(1) receptor) and a provasodilatory, antifibrotic, antigrowth arm (ACE2/ANG-(1-7)/Mas receptor). Already, this simplistic vision is evolving and new components are branching out upstream [ANG-(1-12) and (pro)renin receptor] and downstream (angiotensin-IV and other angiotensin peptides) of the classical cascade. In this review, we will summarize the role of the ACE2/ANG-(1-7)/Mas receptor, focusing on the central nervous system with respect to cardiovascular diseases such as hypertension, chronic heart failure, and stroke, as well as neurological diseases. In addition, we will discuss the new pharmacological (antagonists, agonists, activators) and genomic (knockout and transgenic animals) tools that are currently available. Finally, we will review the latest data regarding the various signaling pathways downstream of the Mas receptor.
Collapse
Affiliation(s)
- Ping Xu
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | |
Collapse
|
29
|
Abstract
For cancers to develop, sustain and spread, the appropriation of key homeostatic physiological systems that influence cell growth, migration and death, as well as inflammation and the expansion of vascular networks are required. There is accumulating molecular and in vivo evidence to indicate that the expression and actions of the renin-angiotensin system (RAS) influence malignancy and also predict that RAS inhibitors, which are currently used to treat hypertension and cardiovascular disease, might augment cancer therapies. To appreciate this potential hegemony of the RAS in cancer, an expanded comprehension of the cellular actions of this system is needed, as well as a greater focus on translational and in vivo research.
Collapse
Affiliation(s)
- Amee J George
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | | | | |
Collapse
|
30
|
Deng W, Baki L, Baumgarten CM. Endothelin signalling regulates volume-sensitive Cl- current via NADPH oxidase and mitochondrial reactive oxygen species. Cardiovasc Res 2010; 88:93-100. [PMID: 20444986 DOI: 10.1093/cvr/cvq125] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
AIMS We assessed regulation of volume-sensitive Cl(-) current (I(Cl,swell)) by endothelin-1 (ET-1) and characterized the signalling pathway responsible for its activation in rabbit atrial and ventricular myocytes. METHODS AND RESULTS ET-1 elicited I(Cl,swell) under isosmotic conditions. Outwardly rectified Cl(-) current was blocked by the I(Cl,swell)-selective inhibitor DCPIB or osmotic shrinkage and involved ET(A) but not ET(B) receptors. ET-1-induced current was abolished by inhibiting epidermal growth factor receptor (EGFR) kinase or phosphoinositide-3-kinase (PI-3K), indicating that these kinases were downstream. Regarding upstream events, activation of I(Cl,swell) by osmotic swelling or angiotensin II (AngII) was suppressed by ET(A) blockade, whereas AngII AT(1) receptor blockade failed to alter ET-1-induced current. Reactive oxygen species (ROS) produced by NADPH oxidase (NOX) stimulate I(Cl,swell). As expected, blockade of NOX suppressed ET-1-induced I(Cl,swell), but blockade of mitochondrial ROS production with rotenone also suppressed I(Cl,swell). I(Cl,swell) was activated by augmenting complex III ROS production with antimycin A or diazoxide; in this case, I(Cl,swell) was insensitive to NOX inhibitors, indicating that mitochondria were downstream from NOX. ROS generation in HL-1 cardiomyocytes measured by flow cytometry confirmed the electrophysiological findings. ET-1-induced ROS production was inhibited by blocking either NOX or mitochondrial complex I, whereas complex III-induced ROS production was insensitive to NOX blockade. CONCLUSION ET-1-ET(A) signalling activated I(Cl,swell) via EGFR kinase, PI-3K, and NOX ROS production, which triggered mitochondrial ROS production. ET(A) receptors were downstream effectors when I(Cl,swell) was elicited by osmotic swelling or AngII. These data suggest that ET-1-induced ROS-dependent I(Cl,swell) is likely to participate in multiple physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Wu Deng
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | | | | |
Collapse
|
31
|
Christensen GL, Kelstrup CD, Lyngsø C, Sarwar U, Bøgebo R, Sheikh SP, Gammeltoft S, Olsen JV, Hansen JL. Quantitative phosphoproteomics dissection of seven-transmembrane receptor signaling using full and biased agonists. Mol Cell Proteomics 2010; 9:1540-53. [PMID: 20363803 DOI: 10.1074/mcp.m900550-mcp200] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Seven-transmembrane receptors (7TMRs) signal through the well described heterotrimeric G proteins but can also activate G protein-independent signaling pathways of which the impact and complexity are less understood. The angiotensin II type 1 receptor (AT(1)R) is a prototypical 7TMR and an important drug target in cardiovascular diseases. "Biased agonists" with intrinsic "functional selectivity" that simultaneously blocks Galpha(q) protein activity and activates G protein-independent pathways of the AT(1)R confer important perspectives in treatment of cardiovascular diseases. In this study, we performed a global quantitative phosphoproteomics analysis of the AT(1)R signaling network. We analyzed ligand-stimulated SILAC (stable isotope labeling by amino acids in cell culture) cells by high resolution (LTQ-Orbitrap) MS and compared the phosphoproteomes of the AT(1)R agonist angiotensin II and the biased agonist [Sar(1),Ile(4),Ile(8)]angiotensin II (SII angiotensin II), which only activates the Galpha(q) protein-independent signaling. We quantified more than 10,000 phosphorylation sites of which 1183 were regulated by angiotensin II or its analogue SII angiotensin II. 36% of the AT(1)R-regulated phosphorylations were regulated by SII angiotensin II. Analysis of phosphorylation site patterns showed a striking distinction between protein kinases activated by Galpha(q) protein-dependent and -independent mechanisms, and we now place protein kinase D as a key protein involved in both Galpha(q)-dependent and -independent AT(1)R signaling. This study provides substantial novel insight into angiotensin II signal transduction and is the first study dissecting the differences between a full agonist and a biased agonist from a 7TMR on a systems-wide scale. Importantly, it reveals a previously unappreciated diversity and quantity of Galpha(q) protein-independent signaling and uncovers novel signaling pathways. We foresee that the amount and diversity of G protein-independent signaling may be more pronounced than previously recognized for other 7TMRs as well. Quantitative mass spectrometry is a promising tool for evaluation of the signaling properties of biased agonists to other receptors in the future.
Collapse
Affiliation(s)
- Gitte L Christensen
- Laboratory for Molecular Cardiology, Department of Biomedical Sciences, Danish National Research Foundation Centre for Cardiac Arrhythmia, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Albrecht D. Physiological and pathophysiological functions of different angiotensins in the brain. Br J Pharmacol 2010. [DOI: 10.1111/j.1476-5381.2010.00648.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
33
|
Cui XL, Chang B, Myatt L. Expression and distribution of NADPH oxidase isoforms in human myometrium--role in angiotensin II-induced hypertrophy. Biol Reprod 2009; 82:305-12. [PMID: 19812300 DOI: 10.1095/biolreprod.109.080275] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The renin-angiotensin system is upregulated in pregnant women and may play a role in myometrial hypertrophy during pregnancy. We examined whether angiotensin II could induce myometrial protein synthesis as determined by (3)H-leucine incorporation in an immortalized human myometrial smooth muscle cell line (ULTR cells). The effects of angiotensin II were mediated by NADPH oxidase because diphenylene iodonium abolished angiotensin II-induced protein synthesis. We investigated gene expression and cellular localization of NADPH oxidase isoforms in ULTR cells and confirmed expression of NOX1, NOX4, and NOX5 in myometrial tissue. Angiotensin II induced a cellular redistribution and upregulation of NOX5 protein without altering NOX1 and NOX4 expression. It seems the effect of angiotensin II relies on the type 1 receptor (AT1), because losartan significantly blocked angiotensin II-induced increase in (3)H-leucine incorporation. We conclude that NADPH oxidase mediates angiotensin II-stimulated protein synthesis downstream of AT1 in myometrium smooth muscle cells.
Collapse
Affiliation(s)
- Xiao-Lan Cui
- Department of Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267-0524, USA.
| | | | | |
Collapse
|