1
|
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
|
2
|
Moreira JD, Nist KM, Carmichael CY, Kuwabara JT, Wainford RD. Sensory Afferent Renal Nerve Activated Gαi 2 Subunit Proteins Mediate the Natriuretic, Sympathoinhibitory and Normotensive Responses to Peripheral Sodium Challenges. Front Physiol 2021; 12:771167. [PMID: 34916958 PMCID: PMC8669768 DOI: 10.3389/fphys.2021.771167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/09/2021] [Indexed: 11/15/2022] Open
Abstract
We have previously reported that brain Gαi2 subunit proteins are required to maintain sodium homeostasis and are endogenously upregulated in the hypothalamic paraventricular nucleus (PVN) in response to increased dietary salt intake to maintain a salt resistant phenotype in rats. However, the origin of the signal that drives the endogenous activation and up-regulation of PVN Gαi2 subunit protein signal transduction pathways is unknown. By central oligodeoxynucleotide (ODN) administration we show that the pressor responses to central acute administration and central infusion of sodium chloride occur independently of brain Gαi2 protein pathways. In response to an acute volume expansion, we demonstrate, via the use of selective afferent renal denervation (ADNX) and anteroventral third ventricle (AV3V) lesions, that the sensory afferent renal nerves, but not the sodium sensitive AV3V region, are mechanistically involved in Gαi2 protein mediated natriuresis to an acute volume expansion [peak natriuresis (μeq/min) sham AV3V: 43 ± 4 vs. AV3V 45 ± 4 vs. AV3V + Gαi2 ODN 25 ± 4, p < 0.05; sham ADNX: 43 ± 4 vs. ADNX 23 ± 6, AV3V + Gαi2 ODN 25 ± 3, p < 0.05]. Furthermore, in response to chronically elevated dietary sodium intake, endogenous up-regulation of PVN specific Gαi2 proteins does not involve the AV3V region and is mediated by the sensory afferent renal nerves to counter the development of the salt sensitivity of blood pressure (MAP [mmHg] 4% NaCl; Sham ADNX 124 ± 4 vs. ADNX 145 ± 4, p < 0.05; Sham AV3V 125 ± 4 vs. AV3V 121 ± 5). Additionally, the development of the salt sensitivity of blood pressure following central ODN-mediated Gαi2 protein down-regulation occurs independently of the actions of the brain angiotensin II type 1 receptor. Collectively, our data suggest that in response to alterations in whole body sodium the peripheral sensory afferent renal nerves, but not the central AV3V sodium sensitive region, evoke the up-regulation and activation of PVN Gαi2 protein gated pathways to maintain a salt resistant phenotype. As such, both the sensory afferent renal nerves and PVN Gαi2 protein gated pathways, represent potential targets for the treatment of the salt sensitivity of blood pressure.
Collapse
Affiliation(s)
- Jesse D. Moreira
- Whitaker Cardiovascular Institute, School of Medicine, Boston University, Boston, MA, United States
- Department of Medicine, School of Medicine, Boston University, Boston, MA, United States
| | - Kayla M. Nist
- Whitaker Cardiovascular Institute, School of Medicine, Boston University, Boston, MA, United States
- Department of Anatomy & Neurobiology, School of Medicine, Boston University, Boston, MA, United States
| | - Casey Y. Carmichael
- Whitaker Cardiovascular Institute, School of Medicine, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, School of Medicine, Boston University, Boston, MA, United States
| | - Jill T. Kuwabara
- Whitaker Cardiovascular Institute, School of Medicine, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, School of Medicine, Boston University, Boston, MA, United States
| | - Richard D. Wainford
- Whitaker Cardiovascular Institute, School of Medicine, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, School of Medicine, Boston University, Boston, MA, United States
| |
Collapse
|
3
|
Mohammed M, Johnson DN, Wang LA, Harden SW, Sheng W, Spector EA, Elsaafien K, Bader M, Steckelings UM, Scott KA, Frazier CJ, Sumners C, Krause EG, de Kloet AD. Targeting angiotensin type 2 receptors located on pressor neurons in the nucleus of the solitary tract to relieve hypertension in mice. Cardiovasc Res 2021; 118:883-896. [PMID: 33723600 DOI: 10.1093/cvr/cvab085] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 03/10/2021] [Indexed: 12/24/2022] Open
Abstract
AIMS These studies evaluate whether angiotensin type-2 receptors (AT2Rs) that are expressed on γ-aminobutyric acid (GABA) neurons in the nucleus of the solitary tract (NTS) represent a novel endogenous blood pressure lowering mechanism. METHODS AND RESULTS Experiments combined advanced genetic and neuroanatomical techniques, pharmacology, electrophysiology and optogenetics in mice to define the structure and cardiovascular-related function of NTS neurons that contain AT2R. Using mice with Cre-recombinase directed to the AT2R gene, we discovered that optogenetic stimulation of AT2R-expressing neurons in the NTS increases GABA release and blood pressure. To evaluate the role of the receptor, per se, in cardiovascular regulation, we chronically delivered C21, a selective AT2R agonist, into the brains of normotensive mice and found that central AT2R activation reduces GABA-related gene expression and blunts the pressor responses induced by optogenetic excitation of NTS AT2R neurons. Next, using in situ hybridization, we found that the levels of Agtr2 mRNAs in GABAergic NTS neurons rise during experimentally-induced hypertension, and we hypothesized that this increased expression may be exploited to ameliorate the disease. Consistent with this, final experiments revealed that central administration of C21 attenuates hypertension, an effect that is abolished in mice lacking AT2R in GABAergic NTS neurons. CONCLUSIONS These studies unveil novel hindbrain circuits that maintain arterial blood pressure, and reveal a specific population of AT2R that can be engaged to alleviate hypertension. The implication is that these discrete receptors may serve as an access point for activating an endogenous depressor circuit. TRANSLATIONAL PERSPECTIVE Hypertension is a widespread health problem and risk factor for cardiovascular disease and stroke. Although treatment options exist, many patients suffer from resistant hypertension, which is associated with enhanced sympathetic drive. Thus, many available therapeutics focus on dampening pressor mechanisms. The present studies take the alternative approach of treating hypertension by exploiting an endogenous depressor mechanism.
Collapse
Affiliation(s)
- Mazher Mohammed
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL
| | - Dominique N Johnson
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL
| | - Lei A Wang
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL
| | - Scott W Harden
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL
| | - Wanhui Sheng
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL
| | - Eliot A Spector
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL
| | - Khalid Elsaafien
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125 Berlin-Buch, Germany.,University of Lübeck, Institute for Biology, Ratzeburger Allee 160, 23562 Lübeck, Germany.,Charité University Medicine, Charitéplatz 1, 10117 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - U Muscha Steckelings
- Institute of Molecular Medicine, Department of Cardiovascular & Renal Research, University of Southern Denmark, Odense, Denmark
| | | | - Charles J Frazier
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL.,Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Colin Sumners
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL.,Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL.,Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Eric G Krause
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL.,Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Annette D de Kloet
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL.,Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL.,Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL
| |
Collapse
|
4
|
Chaudhary M. Anti-Hypertensive Potential and Epigenetics of Angiotensin II type 2 Receptor (AT2R). Curr Hypertens Rev 2020; 17:176-180. [PMID: 33302839 DOI: 10.2174/1573402116999201209203015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/29/2020] [Accepted: 10/07/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Renin angiotensin system (RAS) is a critical pathway involved in blood pressure regulation. Octapeptide, angiotensin II (Ang II), is a biologically active compound of RAS pathway which mediates its action by binding to either angiotensin II type 1 receptor (AT1R) or angiotensin II type 2 receptor (AT2R). Binding of Ang II to AT1R facilitates blood pressure regulation, whereas AT2R is primarily involved in wound healing and tissue remodeling. OBJECTIVES Recent studies have highlighted the additional role of AT2R to counterbalance the detrimental effects of AT1R. Activation of angiotensin II type 2 receptor using AT2R agonist has shown the effect on natriuresis and release of nitric oxide. Additionally, AT2R activation has been found to inhibit angiotensin converting enzyme (ACE) and enhance angiotensin receptor blocker (ARB) activity. These findings highlight the potential of AT2R as a novel therapeutic target against hypertension. CONCLUSION The potential role of AT2R highlights the importance of exploring additional mechanisms that might be crucial for AT2R expression. Epigenetic mechanisms, including DNA methylation and histone modification, have been explored vastly with relation to cancer, but the role of such mechanisms in the expression of AT2R has recently gained interest.
Collapse
Affiliation(s)
- Mayank Chaudhary
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala-133207, Haryana, India
| |
Collapse
|
5
|
McFall A, Nicklin SA, Work LM. The counter regulatory axis of the renin angiotensin system in the brain and ischaemic stroke: Insight from preclinical stroke studies and therapeutic potential. Cell Signal 2020; 76:109809. [PMID: 33059037 PMCID: PMC7550360 DOI: 10.1016/j.cellsig.2020.109809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 01/01/2023]
Abstract
Stroke is the 2nd leading cause of death worldwide and the leading cause of physical disability and cognitive issues. Although we have made progress in certain aspects of stroke treatment, the consequences remain substantial and new treatments are needed. Hypertension has long been recognised as a major risk factor for stroke, both haemorrhagic and ischaemic. The renin angiotensin system (RAS) plays a key role in blood pressure regulation and this, plus local expression and signalling of RAS in the brain, both support the potential for targeting this axis therapeutically in the setting of stroke. While historically, focus has been on suppressing classical RAS signalling through the angiotensin type 1 receptor (AT1R), the identification of a counter-regulatory axis of the RAS signalling via the angiotensin type 2 receptor (AT2R) and Mas receptor has renewed interest in targeting the RAS. This review describes RAS signalling in the brain and the potential of targeting the Mas receptor and AT2R in preclinical models of ischaemic stroke. The animal and experimental models, and the route and timing of intervention, are considered from a translational perspective.
Collapse
Affiliation(s)
- Aisling McFall
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Stuart A Nicklin
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Lorraine M Work
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
6
|
Elsaafien K, de Kloet AD, Krause EG, Sumners C. Brain Angiotensin Type-1 and Type-2 Receptors in Physiological and Hypertensive Conditions: Focus on Neuroinflammation. Curr Hypertens Rep 2020; 22:48. [PMID: 32661792 PMCID: PMC7780348 DOI: 10.1007/s11906-020-01062-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW To review recent data that suggest opposing effects of brain angiotensin type-1 (AT1R) and type-2 (AT2R) receptors on blood pressure (BP). Here, we discuss recent studies that suggest pro-hypertensive and pro-inflammatory actions of AT1R and anti-hypertensive and anti-inflammatory actions of AT2R. Further, we propose mechanisms for the interplay between brain angiotensin receptors and neuroinflammation in hypertension. RECENT FINDINGS The renin-angiotensin system (RAS) plays an important role in regulating cardiovascular physiology. This includes brain AT1R and AT2R, both of which are expressed in or adjacent to brain regions that control BP. Activation of AT1R within those brain regions mediate increases in BP and cause neuroinflammation, which augments the BP increase in hypertension. The fact that AT1R and AT2R have opposing actions on BP suggests that AT1R and AT2R may have similar opposing actions on neuroinflammation. However, the mechanisms by which brain AT1R and AT2R mediate neuroinflammatory responses remain unclear. The interplay between brain angiotensin receptor subtypes and neuroinflammation exacerbates or protects against hypertension.
Collapse
Affiliation(s)
- Khalid Elsaafien
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL, USA
| | - Annette D de Kloet
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Eric G Krause
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, FL, USA
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Colin Sumners
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA.
- Center for Integrative Cardiovascular and Metabolic Diseases, University of Florida, Gainesville, FL, USA.
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
7
|
Dupont AG, Légat L. GABA is a mediator of brain AT 1 and AT 2 receptor-mediated blood pressure responses. Hypertens Res 2020; 43:995-1005. [PMID: 32451494 DOI: 10.1038/s41440-020-0470-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023]
Abstract
The nucleus tractus solitarius (NTS), paraventricular nucleus (PVN), and rostral ventrolateral medulla (RVLM) are the most targeted regions of central blood pressure control studies. Glutamate and gamma-aminobutyric acid (GABA) interact within these brain regions to modulate blood pressure. The brain renin-angiotensin system also participates in central blood pressure control. Angiotensin II increases blood pressure through the stimulation of angiotensin II type 1 (AT1) receptors within the PVN and RVLM and attenuates baroreceptor sensitivity, resulting in elevated blood pressure within the NTS. Angiotensin II type 2 (AT2) receptors in cardiovascular control centers in the brain also appear to be involved in blood pressure control and counteract AT1 receptor-mediated effects. The current review is focused on the interaction of GABA with AT1 and AT2 receptors in the control of blood pressure within the RVLM, PVN and NTS. Within the NTS, GABA is released from local GABAergic interneurons that are stimulated by local AT1 receptors and mediates a hypertensive response. In contrast, the local increase in GABA levels observed after AT2 receptor stimulation within the RVLM, likely from GABAergic nerve endings originating in the caudal ventrolateral medulla, is important in the mediation of the hypotensive response. Preliminary results suggest that the hypertensive response to AT1 receptor stimulation within the RVLM is associated with a reduction in GABA release. The current experimental evidence therefore indicates that GABA is an important mediator of brainstem responses to AT1 and AT2 receptor stimulation and that increased GABA release may play a role in hypertensive and hypotensive responses, depending on the site of action.
Collapse
Affiliation(s)
- Alain G Dupont
- Department of Pharmacology, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences, (C4N) Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Laura Légat
- Department of Pharmacology, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences, (C4N) Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium.
| |
Collapse
|
8
|
Assersen KB, Sumners C, Steckelings UM. The Renin-Angiotensin System in Hypertension, a Constantly Renewing Classic: Focus on the Angiotensin AT 2-Receptor. Can J Cardiol 2020; 36:683-693. [PMID: 32389341 DOI: 10.1016/j.cjca.2020.02.095] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 02/07/2023] Open
Abstract
It is common knowledge that the renin-angiotensin system (RAS), in particular angiotensin II acting through the angiotensin AT1-receptor (AT1R), is pivotal for the regulation of blood pressure (BP) and extracellular volume. More recent findings have revealed that the RAS is far more complex than initially thought and that it harbours additional mediators and receptors, which are able to counteract and thereby fine-tune AT1R-mediated actions. This review will focus on the angiotensin AT2-receptor (AT2R), which is one of the "counter-regulatory" receptors within the RAS. It will review and discuss data related to the role of the AT2R in regulation of BP and focus on the following 3 questions: Do peripheral AT2R have an impact on BP regulation, and, if so, does this effect become apparent only under certain conditions? Are central nervous system AT2R involved in regulation of BP, and, if so, which brain areas are involved and what are the mechanisms? Does dysfunction of AT2R contribute to the pathogenesis of hypertension in preeclampsia?
Collapse
Affiliation(s)
- Kasper B Assersen
- Institute for Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Colin Sumners
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - U Muscha Steckelings
- Institute for Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
9
|
Légat L, Smolders I, Dupont AG. Angiotensin-II-mediated AT1 receptor stimulation increases glutamate release within the rostral ventrolateral medulla of normotensive rats. Hypertens Res 2020; 43:848-850. [DOI: 10.1038/s41440-020-0419-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/09/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023]
|
10
|
Sumners C, Alleyne A, Rodríguez V, Pioquinto DJ, Ludin JA, Kar S, Winder Z, Ortiz Y, Liu M, Krause EG, de Kloet AD. Brain angiotensin type-1 and type-2 receptors: cellular locations under normal and hypertensive conditions. Hypertens Res 2019; 43:281-295. [PMID: 31853042 DOI: 10.1038/s41440-019-0374-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/25/2019] [Accepted: 11/02/2019] [Indexed: 12/15/2022]
Abstract
Brain angiotensin-II (Ang-II) type-1 receptors (AT1Rs), which exert profound effects on normal cardiovascular, fluid, and metabolic homeostasis, are overactivated in and contribute to chronic sympathoexcitation and hypertension. Accumulating evidence indicates that the activation of Ang-II type-2 receptors (AT2Rs) in the brain exerts effects that are opposite to those of AT1Rs, lowering blood pressure, and reducing hypertension. Thus, it would be interesting to understand the relative cellular localization of AT1R and AT2R in the brain under normal conditions and whether this localization changes during hypertension. Here, we developed a novel AT1aR-tdTomato reporter mouse strain in which the location of brain AT1aR was largely consistent with that determined in the previous studies. This AT1aR-tdTomato reporter mouse strain was crossed with our previously described AT2R-eGFP reporter mouse strain to yield a novel dual AT1aR/AT2R reporter mouse strain, which allowed us to determine that AT1aR and AT2R are primarily localized to different populations of neurons in brain regions controlling cardiovascular, fluid, and metabolic homeostasis. Using the individual AT1aR-tdTomato reporter mice, we also demonstrated that during hypertension induced by the administration of deoxycorticosterone acetate-salt, there was no shift in the expression of AT1aR from neurons to microglia or astrocytes in the paraventricular nucleus, a brain area important for sympathetic regulation. Using AT2R-eGFP reporter mice under similar hypertensive conditions, we demonstrated that the same was true of AT2R expression in the nucleus of the solitary tract (NTS), an area critical for baroreflex control. Collectively, these findings provided a novel means to assess the colocalization of AT1R and AT2R in the brain and a novel view of their cellular localization in hypertension.
Collapse
Affiliation(s)
- Colin Sumners
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Amy Alleyne
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Vermalí Rodríguez
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - David J Pioquinto
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Jacob A Ludin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Shormista Kar
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Zachary Winder
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA.,Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Yuma Ortiz
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Meng Liu
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Eric G Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Annette D de Kloet
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
11
|
Moreira JD, Chaudhary P, Frame AA, Puleo F, Nist KM, Abkin EA, Moore TL, George JC, Wainford RD. Inhibition of microglial activation in rats attenuates paraventricular nucleus inflammation in Gαi 2 protein-dependent, salt-sensitive hypertension. Exp Physiol 2019; 104:1892-1910. [PMID: 31631436 PMCID: PMC6884700 DOI: 10.1113/ep087924] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS • What is the central question of this study? We hypothesized that central inflammatory processes that involve activation of microglia and astrocytes contribute to the development of Gαi2 protein-dependent, salt-sensitive hypertension. • What is the main finding and its importance? The main finding is that PVN-specific inflammatory processes, driven by microglial activation, appear to be linked to the development of Gαi2 protein-dependent, salt-sensitive hypertension in Sprague-Dawley rats. This finding might reveal new mechanistic targets in the treatment of hypertension. ABSTRACT The central mechanisms underlying salt-sensitive hypertension, a significant public health issue, remain to be established. Researchers in our laboratory have reported that hypothalamic paraventricular nucleus (PVN) Gαi2 proteins mediate the sympathoinhibitory and normotensive responses to high sodium intake in salt-resistant rats. Given the recent evidence of central inflammation in animal models of hypertension, we hypothesized that PVN inflammation contributes to Gαi2 protein-dependent, salt-sensitive hypertension. Male Sprague-Dawley rats received chronic intracerebroventricular infusions of a targeted Gαi2 or control scrambled oligodeoxynucleotide (ODN) and were maintained for 7 days on a normal-salt (NS; 0.6% NaCl) or high-salt (HS; 4% NaCl) diet; in subgroups on HS, intracerebroventricular minocycline (microglial inhibitor) was co-infused with ODNs. Radiotelemetry was used in subgroups of rats to measure mean arterial pressure (MAP) chronically. In a separate group of rats, plasma noradrenaline, plasma renin activity, urinary angiotensinogen and mRNA levels of the PVN pro-inflammatory cytokines TNFα, IL-1β and IL-6 and the anti-inflammatory cytokine IL-10 were assessed. In additional groups, immunohistochemistry was performed for markers of PVN and subfornical organ microglial activation and cytokine levels and PVN astrocyte activation. High salt intake evoked salt-sensitive hypertension, increased plasma noradrenaline, PVN pro-inflammatory cytokine mRNA upregulation, anti-inflammatory cytokine mRNA downregulation and PVN-specific microglial activation in rats receiving a targeted Gαi2 but not scrambled ODN. Minocycline co-infusion significantly attenuated the increase in MAP and abolished the increase in plasma noradrenaline and inflammation in Gαi2 ODN-infused animals on HS. Our data suggest that central Gαi2 protein prevents microglial-mediated PVN inflammation and the development of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Jesse D. Moreira
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Health SciencesBoston University Sargent CollegeBostonMAUSA
| | - Parul Chaudhary
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
| | - Alissa A. Frame
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
| | - Franco Puleo
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
| | - Kayla M. Nist
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Anatomy & NeurobiologyBoston University School of MedicineBostonMAUSA
| | - Eric A. Abkin
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Health SciencesBoston University Sargent CollegeBostonMAUSA
| | - Tara L. Moore
- Department of Anatomy & NeurobiologyBoston University School of MedicineBostonMAUSA
| | - Jonique C. George
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
| | - Richard D. Wainford
- The Whitaker Cardiovascular InstituteBoston UniversityBostonMAUSA
- Department of Health SciencesBoston University Sargent CollegeBostonMAUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMAUSA
| |
Collapse
|
12
|
Légat L, Smolders IJ, Dupont AG. Investigation of the Role of AT2 Receptors in the Nucleus Tractus Solitarii of Normotensive Rats in Blood Pressure Control. Front Neurosci 2019; 13:589. [PMID: 31231188 PMCID: PMC6560158 DOI: 10.3389/fnins.2019.00589] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/23/2019] [Indexed: 11/25/2022] Open
Abstract
Aim The nucleus tractus solitarii (NTS) densely expresses angiotensin II type 2 receptors (AT2R), which are mainly located on inhibitory gamma-aminobutyric acid (GABA) neurons. Central AT2R stimulation reduces blood pressure, and AT2R stimulation in the rostral ventrolateral medulla (RVLM), mediates a hypotensive response through a GABAergic mechanism. We aimed to test the hypothesis that an AT2R mediated inhibition of the GABA release within the NTS might be involved in this hypotensive response, by assessing possible alterations in blood pressure and heart rate, as well as in GABA levels in normotensive Wistar rats. Methods In vivo microdialysis was used for measurement of extracellular GABA levels and for perfusion of the selective AT2R agonist, Compound 21, within the NTS. Our set-up allowed to determine simultaneously the excitatory glutamate dialysate levels. The mean arterial pressure and heart rate responses were monitored with a pressure transducer. Results Local perfusion of Compound 21 into the NTS did not modify blood pressure and heart rate, nor glutamate and GABA levels compared to baseline concentrations. A putative effect was also not unmasked by concomitant angiotensin II type 1 receptor blockade with candesartan. Positive control experiments confirmed that the experimental set up had enough sensitivity to detect a reduction in GABA dialysate levels and blood pressure. Conclusion The results did not provide evidence for a role of the AT2R within the NTS in the control of blood pressure, nor for an interaction with local GABAergic signaling in normotensive rats.
Collapse
Affiliation(s)
- Laura Légat
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium.,Cardiovascular Center, Universitair Ziekenhuis Brussel, Brussels, Belgium.,Department of Clinical Pharmacology and Clinical Pharmacy (KFAR), Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ilse J Smolders
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Alain G Dupont
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium.,Cardiovascular Center, Universitair Ziekenhuis Brussel, Brussels, Belgium.,Department of Clinical Pharmacology and Clinical Pharmacy (KFAR), Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
13
|
Frame AA, Carmichael CY, Kuwabara JT, Cunningham JT, Wainford RD. Role of the afferent renal nerves in sodium homeostasis and blood pressure regulation in rats. Exp Physiol 2019; 104:1306-1323. [PMID: 31074108 PMCID: PMC6675646 DOI: 10.1113/ep087700] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/09/2019] [Indexed: 12/11/2022]
Abstract
New Findings What is the central question of this study? What are the differential roles of the mechanosensitive and chemosensitive afferent renal nerves in the reno‐renal reflex that promotes natriuresis, sympathoinhibition and normotension during acute and chronic challenges to sodium homeostasis? What is the main finding and its importance? The mechanosensitive afferent renal nerves contribute to an acute natriuretic sympathoinhibitory reno‐renal reflex that may be integrated within the paraventricular nucleus of the hypothalamus. Critically, the afferent renal nerves are required for the maintenance of salt resistance in Sprague–Dawley and Dahl salt‐resistant rats and attenuate the development of Dahl salt‐sensitive hypertension.
Abstract These studies tested the hypothesis that in normotensive salt‐resistant rat phenotypes the mechanosensitive afferent renal nerve (ARN) reno‐renal reflex promotes natriuresis, sympathoinhibition and normotension during acute and chronic challenges to fluid and electrolyte homeostasis. Selective ARN ablation was conducted prior to (1) an acute isotonic volume expansion (VE) or 1 m NaCl infusion in Sprague–Dawley (SD) rats and (2) chronic high salt intake in SD, Dahl salt‐resistant (DSR), and Dahl salt‐sensitive (DSS) rats. ARN responsiveness following high salt intake was assessed ex vivo in response to noradrenaline and sodium concentration (SD, DSR and DSS) and via in vivo manipulation of renal pelvic pressure and sodium concentration (SD and DSS). ARN ablation attenuated the natriuretic and sympathoinhibitory responses to an acute VE [peak natriuresis (µeq min−1) sham 52 ± 5 vs. ARN ablation 28 ± 3, P < 0.05], but not a hypertonic saline infusion in SD rats. High salt (HS) intake enhanced ARN reno‐renal reflex‐mediated natriuresis in response to direct increases in renal pelvic pressure (mechanoreceptor stimulus) in vivo and ARN responsiveness to noradrenaline ex vivo in SD, but not DSS, rats. In vivo and ex vivo ARN responsiveness to increased renal pelvic sodium concentration (chemoreceptor stimulus) was unaltered during HS intake. ARN ablation evoked sympathetically mediated salt‐sensitive hypertension in SD rats [MAP (mmHg): sham normal salt 102 ± 2 vs. sham HS 104 ± 2 vs. ARN ablation normal salt 103 ± 2 vs. ARN ablation HS 121 ± 2, P < 0.05] and DSR rats and exacerbated DSS hypertension. The mechanosensitive ARNs mediate an acute sympathoinhibitory natriuretic reflex and counter the development of salt‐sensitive hypertension.
Collapse
Affiliation(s)
- Alissa A Frame
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Casey Y Carmichael
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Jill T Kuwabara
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Richard D Wainford
- Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
14
|
Mirabito Colafella KM, Bovée DM, Danser AHJ. The renin-angiotensin-aldosterone system and its therapeutic targets. Exp Eye Res 2019; 186:107680. [PMID: 31129252 DOI: 10.1016/j.exer.2019.05.020] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 12/22/2022]
Abstract
The renin-angiotensin-aldosterone system (RAAS) plays a pivotal role in the regulation of blood pressure and body fluid homeostasis and is a mainstay for the treatment of cardiovascular and renal diseases. Angiotensin II and aldosterone are the two most powerful biologically active products of the RAAS, inducing all of the classical actions of the RAAS including vasoconstriction, sodium retention, tissue remodeling and pro-inflammatory and pro-fibrotic effects. In recent years, new components of the RAAS have been discovered beyond the classical pathway that have led to the identification of depressor or so-called protective RAAS pathways and the development of novel therapies targeting this system. Moreover, dual inhibitors which block the RAAS and other systems involved in the regulation of blood pressure or targeting upstream of angiotensin II by selectively deleting liver-derived angiotensinogen, the precursor to all angiotensins, may provide superior treatment for cardiovascular and renal diseases and revolutionize RAAS-targeting therapy.
Collapse
Affiliation(s)
- Katrina M Mirabito Colafella
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia
| | - Dominique M Bovée
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|
15
|
Légat L, Smolders I, Dupont AG. AT1 Receptor Mediated Hypertensive Response to Ang II in the Nucleus Tractus Solitarii of Normotensive Rats Involves NO Dependent Local GABA Release. Front Pharmacol 2019; 10:460. [PMID: 31130861 PMCID: PMC6509664 DOI: 10.3389/fphar.2019.00460] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/11/2019] [Indexed: 11/30/2022] Open
Abstract
Aim It is well-established that angiotensin II exerts a dampening effect on the baroreflex within the nucleus tractus solitarii (NTS), the principal brainstem site for termination of baroreceptor afferents and which is densely populated with gamma-aminobutyric acid (GABA)ergic neurons and nerve terminals. The present study was designed to investigate whether local release of GABA is involved in the effects mediated by local angiotensin II within the NTS. Methods In vivo microdialysis was used for measurement of extracellular glutamate and GABA levels and for infusion of angiotensin II within the NTS of conscious normotensive Wistar rats. The mean arterial pressure (MAP) and heart rate response to local infusion of angiotensin II were subsequently monitored with a pressure transducer under anesthesia. The angiotensin II type 1 receptor (AT1R) antagonist, candesartan, was used to assess whether responses were AT1R dependent and the nitric oxide (NO) synthase inhibitor, N(ω)-nitro-L-arginine methyl ester (L-NAME), was used to assess the involvement of NO in the evoked responses by infusion of angiotensin II. The MAP and heart rate responses were monitored with a pressure transducer. Results Local infusion into the NTS of angiotensin II induced a significant to ninefold significantly increase in extracellular GABA levels; as well as MAP was increased by 15 mmHg. These responses were both abolished by co-infusion of either, the angiotensin II type 1 receptor antagonist, candesartan, or the NO synthase inhibitor, L-NAME, demonstrating that the effect is not only AT1R dependent but also NO dependent. The pressor response to angiotensin II was reversed by co-infusion with the GABAA receptor antagonist, bicuculline. Local blockade of NO synthase decreased both, GABA and glutamate concentrations. Conclusion Our results suggest that the AT1R mediated hypertensive response to angiotensin II within the NTS in normotensive rats is GABA and NO dependent. Nitric oxide produced within the NTS tonically potentiates local GABA and glutamate release.
Collapse
Affiliation(s)
- Laura Légat
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium.,Cardiovascular Center, Universitair Ziekenhuis Brussel, Brussels, Belgium.,Department of Clinical Pharmacology and Clinical Pharmacy, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Alain G Dupont
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium.,Cardiovascular Center, Universitair Ziekenhuis Brussel, Brussels, Belgium.,Department of Clinical Pharmacology and Clinical Pharmacy, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
16
|
Légat L, Smolders I, Dupont AG. GABAergic Signaling Mediates Central Cardiovascular Angiotensin II Type 2 Receptor Effects. Trends Endocrinol Metab 2018; 29:605-606. [PMID: 29739704 DOI: 10.1016/j.tem.2018.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 11/22/2022]
Affiliation(s)
- Laura Légat
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Belgium; Research Group Clinical Pharmacology and Clinical Pharmacy, Vrije Universiteit Brussel, Belgium; Department of Clinical Pharmacology, Universitair Ziekenhuis Brussel, Belgium.
| | - Ilse Smolders
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Belgium
| | - Alain G Dupont
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Belgium; Research Group Clinical Pharmacology and Clinical Pharmacy, Vrije Universiteit Brussel, Belgium; Department of Clinical Pharmacology, Universitair Ziekenhuis Brussel, Belgium
| |
Collapse
|
17
|
Neuroprotection via AT2 receptor agonists in ischemic stroke. Clin Sci (Lond) 2018; 132:1055-1067. [DOI: 10.1042/cs20171549] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/30/2018] [Accepted: 05/01/2018] [Indexed: 12/12/2022]
Abstract
Stroke is a devastating disease that afflicts millions of people each year worldwide. Ischemic stroke, which accounts for ~88% of cases, occurs when blood supply to the brain is decreased, often because of thromboembolism or atherosclerotic occlusion. This deprives the brain of oxygen and nutrients, causing immediate, irreversible necrosis within the core of the ischemic area, but more delayed and potentially reversible neuronal damage in the surrounding brain tissue, the penumbra. The only currently approved therapies for ischemic stroke, the thrombolytic agent recombinant tissue plasminogen activator (rtPA) and the endovascular clot retrieval/destruction processes, are aimed at restoring blood flow to the infarcted area, but are only available for a minority of patients and are not able in most cases to completely restore neurological deficits. Consequently, there remains a need for agents that will protect neurones against death following ischemic stroke. Here, we evaluate angiotensin II (Ang II) type 2 (AT2) receptor agonists as a possible therapeutic target for this disease. We first provide an overview of stroke epidemiology, pathophysiology, and currently approved therapies. We next review the large amount of preclinical evidence, accumulated over the past decade and a half, which indicates that AT2 receptor agonists exert significant neuroprotective effects in various animal models, and discuss the potential mechanisms involved. Finally, after discussing the challenges of delivering blood–brain barrier (BBB) impermeable AT2 receptor agonists to the infarcted areas of the brain, we summarize the evidence for and against the development of these agents as a promising therapeutic strategy for ischemic stroke.
Collapse
|
18
|
Winklewski PJ, Radkowski M, Demkow U. Neuroinflammatory mechanisms of hypertension: potential therapeutic implications. Curr Opin Nephrol Hypertens 2018; 25:410-6. [PMID: 27490783 DOI: 10.1097/mnh.0000000000000250] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Inflammation of forebrain and hindbrain nuclei has recently been highlighted as an emerging factor in the pathogenesis of neurogenic hypertension. The aim of this review is to summarize the state of the art in this field and to discuss recently discovered pathophysiological mechanisms, opening new perspectives for therapeutic application. RECENT FINDINGS Microglia Toll-like receptor 4 causally links angiotensin II (AngII)-mediated microglia cell activation and oxidative stress within the hypothalamic paraventricular nucleus (PVN). Toll-like receptor 4 can also be activated by lipopolysaccharides. PVN infusion of nuclear factor κB inhibitor lowers the blood pressure and ameliorates cardiac hypertrophy. Ang-(1-7) exerts direct effects on microglia, causing a reduction in both baseline and prorenin-induced release of proinflammatory cytokines. A compromised blood-brain barrier (BBB) constitutes a complementary mechanism that exacerbates AngII-driven neurohumoral activation, contributing to the development of hypertension. SUMMARY PVN and BBB seem to be pivotal targets for therapeutic intervention in hypertension. Recent advances in imaging techniques enable visualization of the inflammatory state in microglia and BBB integrity in humans. AngII type I receptor blockers and AngII-converting enzyme inhibitors are the most likely candidates for controlled randomized trials in humans aimed at amelioration of brain inflammation in the forthcoming years.
Collapse
Affiliation(s)
- Pawel J Winklewski
- aInstitute of Human Physiology, Medical University of Gdansk, GdanskbDepartment of Immunopathology of Infectious and Parasitic DiseasescDepartment of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, Poland
| | | | | |
Collapse
|
19
|
de Kloet AD, Steckelings UM, Sumners C. Protective Angiotensin Type 2 Receptors in the Brain and Hypertension. Curr Hypertens Rep 2017; 19:46. [PMID: 28488048 DOI: 10.1007/s11906-017-0746-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW The goal of this review is to assess the evidence that activation of angiotensin type 2 receptors (AT2R) in the brain can lower blood pressure and possibly constitute an endogenous anti-hypertensive mechanism. RECENT FINDINGS Recent studies that detail the location of AT2R in the brain, particularly within or near cardiovascular control centers, mesh well with findings from pharmacological and gene transfer studies which demonstrate that activation of central AT2R can influence cardiovascular regulation. Collectively, these studies indicate that selective activation of brain AT2R causes moderate decreases in blood pressure in normal animals and more profound anti-hypertensive effects, along with restoration of baroreflex function, in rodent models of neurogenic hypertension. These findings have opened the door to studies that can (i) assess the role of specific AT2R neuron populations in depressing blood pressure, (ii) determine the relevance of such mechanisms, and (iii) investigate interactions between AT2R and depressor angiotensin-(1-7)/Mas mechanisms in the brain.
Collapse
Affiliation(s)
- Annette D de Kloet
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, 1600 SW Archer Road, Box 100274, Gainesville, FL, 32610-0274, USA
| | - Ulrike M Steckelings
- IMM - Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Colin Sumners
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, 1600 SW Archer Road, Box 100274, Gainesville, FL, 32610-0274, USA.
| |
Collapse
|
20
|
Steckelings UM, Kloet AD, Sumners C. Centrally Mediated Cardiovascular Actions of the Angiotensin II Type 2 Receptor. Trends Endocrinol Metab 2017; 28:684-693. [PMID: 28733135 PMCID: PMC5563271 DOI: 10.1016/j.tem.2017.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/06/2017] [Accepted: 06/08/2017] [Indexed: 02/07/2023]
Abstract
Sustained increases in the activity of the sympathetic neural pathways that exit the brain and which increase blood pressure (BP) are a major underlying factor in resistant hypertension. Recently available information on the occurrence of angiotensin II type 2 receptors (AT2Rs) within or adjacent to brain cardiovascular control centers is consistent with findings that stimulation of these receptors lowers BP, particularly during hypertension of neurogenic origin. Until recently brain AT2R had not been considered by many to play a role in the central control of BP. Demonstration of these powerful antihypertensive effects of brain AT2R opens the door to reconsideration of their role in BP regulation, and their consideration as a novel therapeutic avenue for resistant hypertension.
Collapse
Affiliation(s)
- U Muscha Steckelings
- Institute of Molecular Medicine (IMM), Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Annette de Kloet
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, USA
| | - Colin Sumners
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, USA.
| |
Collapse
|
21
|
Légat L, Brouwers S, Smolders IJ, Dupont AG. Hypotensive Response to Angiotensin II Type 2 Receptor Stimulation in the Rostral Ventrolateral Medulla Requires Functional GABA-A Receptors. Front Neurosci 2017; 11:346. [PMID: 28674483 PMCID: PMC5474467 DOI: 10.3389/fnins.2017.00346] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 06/02/2017] [Indexed: 11/16/2022] Open
Abstract
Objectives: Angiotensin II, glutamate and gamma-aminobutyric acid (GABA) interact within the rostral ventrolateral medulla (RVLM) and the paraventricular nucleus (PVN) modulating the central regulation of blood pressure and sympathetic tone. Our aim was to assess the effects of local angiotensin II type 2 receptor stimulation within the RVLM and the PVN on neurotransmitter concentrations and mean arterial pressure (MAP). Methods:In vivo microdialysis was used for measurement of extracellular glutamate and GABA levels and for local infusion of the angiotensin II type 2 receptor agonist Compound 21 in the RVLM and the PVN of conscious normotensive Wistar rats. The MAP response to local Compound 21 was monitored with a pressure transducer under anaesthesia. Angiotensin II type 2 receptor selectivity was assessed using the angiotensin II type 2 receptor antagonist PD123319; the GABA-A receptor antagonist bicuculline was used to assess the involvement of GABA-A receptors. Results: Infusion of Compound 21 (0.05 μg/μl/h) in the RVLM significantly increased GABA levels and lowered blood pressure. These effects were abolished by co-infusion with PD123319. No changes in neurotransmitter levels or effects on blood pressure were seen with PD123319 infusion alone. Co-infusion of bicuculline abolished the Compound 21 evoked decrease in MAP. Infusion of Compound 21 within the PVN did not change extracellular neurotransmitter levels nor MAP. Conclusion: Selective stimulation of angiotensin II type 2 receptor within the RVLM by local Compound 21 infusion reduces blood pressure and increases local GABA levels in normotensive rats. This hypotensive response requires functional GABA-A receptors, suggesting that GABAergic neurons are involved in the sympatho-inhibitory action underlying this hypotensive response.
Collapse
Affiliation(s)
- Laura Légat
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit BrusselBrussels, Belgium.,Cardiovascular Center, Universitair Ziekenhuis BrusselBrussels, Belgium.,Department of Clinical Pharmacology and Clinical Pharmacy, Universitair Ziekenhuis BrusselBrussels, Belgium
| | - Sofie Brouwers
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit BrusselBrussels, Belgium.,Cardiovascular Center, Universitair Ziekenhuis BrusselBrussels, Belgium.,Department of Clinical Pharmacology and Clinical Pharmacy, Universitair Ziekenhuis BrusselBrussels, Belgium
| | - Ilse J Smolders
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit BrusselBrussels, Belgium
| | - Alain G Dupont
- Laboratory of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit BrusselBrussels, Belgium.,Cardiovascular Center, Universitair Ziekenhuis BrusselBrussels, Belgium.,Department of Clinical Pharmacology and Clinical Pharmacy, Universitair Ziekenhuis BrusselBrussels, Belgium
| |
Collapse
|
22
|
Tamargo M, Tamargo J. Future drug discovery in renin-angiotensin-aldosterone system intervention. Expert Opin Drug Discov 2017; 12:827-848. [PMID: 28541811 DOI: 10.1080/17460441.2017.1335301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Renin-angiotensin-aldosterone system inhibitors (RAASIs), including angiotensin-converting enzyme inhibitors, angiotensin AT1 receptor blockers and mineralocorticoid receptor antagonists (MRAs), are the cornerstone for the treatment of cardiovascular and renal diseases. Areas covered: The authors searched MEDLINE, PubMed and ClinicalTrials.gov to identify eligible full-text English language papers. Herein, the authors discuss AT2-receptor agonists and ACE2/angiotensin-(1-7)/Mas-receptor axis modulators, direct renin inhibitors, brain aminopeptidase A inhibitors, biased AT1R blockers, chymase inhibitors, multitargeted drugs, vaccines and aldosterone receptor antagonists as well as aldosterone synthase inhibitors. Expert opinion: Preclinical studies have demonstrated that activation of the protective axis of the RAAS represents a novel therapeutic strategy for treating cardiovascular and renal diseases, but there are no clinical trials supporting our expectations. Non-steroidal MRAs might become the third-generation of MRAs for the treatment of heart failure, diabetes mellitus and chronic kidney disease. The main challenge for these new drugs is that conventional RAASIs are safe, effective and cheap generics. Thus, the future of new RAASIs will be directed by economical/strategic reasons.
Collapse
Affiliation(s)
- Maria Tamargo
- a Department of Cardiology , Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV , Madrid , Spain
| | - Juan Tamargo
- b Department of Pharmacology , School of Medicine, University Complutense, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV , Madrid , Spain
| |
Collapse
|
23
|
Tomasova L, Drapala A, Jurkowska H, Wróbel M, Ufnal M. Na 2S, a fast-releasing H 2S donor, given as suppository lowers blood pressure in rats. Pharmacol Rep 2017; 69:971-977. [PMID: 28822959 DOI: 10.1016/j.pharep.2017.03.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/22/2017] [Accepted: 03/31/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Hydrogen sulfide (H2S) is involved in blood pressure control. The available slow-releasing H2S-donors are poorly soluble in water and their ability to release H2S in biologically relevant amounts under physiological conditions is questionable. Therefore, new slow-releasing donors or new experimental approaches to fast-releasing H2S donors are needed. METHODS Hemodynamics and ECG were recorded in male, anesthetized Wistar Kyoto rats (WKY) and in Spontaneously hypertensive rats (SHR) at baseline and after: 1) intravenous (iv) infusion of vehicle or Na2S; 2) administration of vehicle suppositories or Na2S suppositories. RESULTS Intravenously administered vehicle and vehicle suppositories did not affect mean arterial blood pressure (MABP) and heart rate (HR). Na2S administered iv caused a significant, but transient (2-5min) decrease in MABP. Na2S suppositories produced a dose-dependent hypotensive response that lasted ∼45min in WKY and ∼75-80min in SHR. It was accompanied by a decrease in HR in WKY, and an increase in HR in SHR. Na2S suppositories did not produce a significant change in corrected QT, an indicator of cardiotoxicity. Na2S suppositories increased blood level of thiosulfates, products of H2S oxidation. CONCLUSIONS Na2S administered in suppositories exerts a prolonged hypotensive effect in rats, with no apparent cardiotoxic effect. SHR and WKY differ in hemodynamic response to the H2S donor. Suppository formulation of fast-releasing H2S donors may be useful in research, if a reference slow-releasing H2S donor is not available.
Collapse
Affiliation(s)
- Lenka Tomasova
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warszawa, Poland; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic; Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Adrian Drapala
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warszawa, Poland
| | - Halina Jurkowska
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kraków, Poland
| | - Maria Wróbel
- Chair of Medical Biochemistry, Jagiellonian University Medical College, Kraków, Poland
| | - Marcin Ufnal
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warszawa, Poland.
| |
Collapse
|
24
|
Stoop W, De Geyter D, Verachtert S, Brouwers S, Verdood P, De Keyser J, Kooijman R. Post-stroke treatment with 17β-estradiol exerts neuroprotective effects in both normotensive and hypertensive rats. Neuroscience 2017; 348:335-345. [DOI: 10.1016/j.neuroscience.2017.02.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 12/01/2022]
|
25
|
Carey RM. AT2 Receptors: Potential Therapeutic Targets for Hypertension. Am J Hypertens 2017; 30:339-347. [PMID: 27664954 DOI: 10.1093/ajh/hpw121] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/07/2016] [Indexed: 12/15/2022] Open
Abstract
The renin-angiotensin system (RAS) is arguably the most important and best studied hormonal system in the control of blood pressure (BP) and the pathogenesis of hypertension. The RAS features its main effector angiotensin II (Ang II) acting via its 2 major receptors, angiotensin type-1(AT1R) and type-2 (AT2R). In general, AT2Rs oppose the detrimental actions of Ang II via AT1Rs. AT2R activation induces vasodilation and natriuresis, but its effects to lower BP in hypertension have not been as clear as anticipated. Recent studies, however, have demonstrated that acute and chronic AT2R stimulation can induce natriuresis and lower BP in the Ang II infusion model of experimental hypertension. AT2R activation induces receptor recruitment from intracellular sites to the apical plasma membranes of renal proximal tubule cells via a bradykinin, nitric oxide, and cyclic guanosine 3',5' monophosphate signaling pathway that results in internalization and inactivation of sodium (Na+) transporters Na+-H+ exchanger-3 and Na+/K+ATPase. These responses do not require the presence of concurrent AT1R blockade and are effective both in the prevention and reversal of hypertension. This review will address the role of AT2Rs in the control of BP and Na+ excretion and the case for these receptors as potential therapeutic targets for hypertension in humans.
Collapse
Affiliation(s)
- Robert M Carey
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, USA
| |
Collapse
|
26
|
Paulis L, Foulquier S, Namsolleck P, Recarti C, Steckelings UM, Unger T. Combined Angiotensin Receptor Modulation in the Management of Cardio-Metabolic Disorders. Drugs 2016; 76:1-12. [PMID: 26631237 PMCID: PMC4700059 DOI: 10.1007/s40265-015-0509-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cardiovascular and metabolic disorders, such as hypertension, insulin resistance, dyslipidemia or obesity are linked with chronic low-grade inflammation and dysregulation of the renin–angiotensin system (RAS). Consequently, RAS inhibition by ACE inhibitors or angiotensin AT1 receptor (AT1R) blockers is the evidence-based standard for cardiovascular risk reduction in high-risk patients, including diabetics with albuminuria. In addition, RAS inhibition reduces the new onset of diabetes mellitus. Yet, the high and increasing prevalence of metabolic disorders, and the high residual risk even in properly treated patients, calls for additional means of pharmacological intervention. In the past decade, the stimulation of the angiotensin AT2 receptor (AT2R) has been shown to reduce inflammation, improve cardiac and vascular remodeling, enhance insulin sensitivity and increase adiponectin production. Therefore, a concept of dual AT1R/AT2R modulation emerges as a putative means for risk reduction in cardio-metabolic diseases. The approach employing simultaneous RAS blockade (AT1R) and RAS stimulation (AT2R) is distinct from previous attempts of double intervention in the RAS by dual blockade. Dual blockade abolishes the AT1R-linked RAS almost completely with subsequent risk of hypotension and hypotension-related events, i.e. syncope or renal dysfunction. Such complications might be especially prominent in patients with renal impairment or patients with isolated systolic hypertension and normal-to-low diastolic blood pressure values. In contrast to dual RAS blockade, the add-on of AT2R stimulation does not exert significant blood pressure effects, but it may complement and enhance the anti-inflammatory and antifibrotic/de-stiffening effects of the AT1R blockade and improve the metabolic profile. Further studies will have to investigate these putative effects in particular for settings in which blood pressure reduction is not primarily desired.
Collapse
Affiliation(s)
- Ludovit Paulis
- Faculty of Medicine, Institute of Pathological Physiology, Comenius University in Bratislava, Sasinkova 4, 81108, Bratislava, Slovak Republic.,Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Sienkiewiczova 1, 81371, Bratislava, Slovak Republic
| | - Sébastien Foulquier
- CARIM-School for Cardiovascular Diseases, Maastricht University, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - Pawel Namsolleck
- CARIM-School for Cardiovascular Diseases, Maastricht University, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - Chiara Recarti
- CARIM-School for Cardiovascular Diseases, Maastricht University, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - Ulrike Muscha Steckelings
- Institute of Molecular Medicine-Department of Cardiovascular and Renal Research, University of Southern Denmark, 5000, Odense, Denmark
| | - Thomas Unger
- CARIM-School for Cardiovascular Diseases, Maastricht University, PO Box 616, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|
27
|
de Kloet AD, Pitra S, Wang L, Hiller H, Pioquinto DJ, Smith JA, Sumners C, Stern JE, Krause EG. Angiotensin Type-2 Receptors Influence the Activity of Vasopressin Neurons in the Paraventricular Nucleus of the Hypothalamus in Male Mice. Endocrinology 2016; 157:3167-80. [PMID: 27267713 PMCID: PMC4967126 DOI: 10.1210/en.2016-1131] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It is known that angiotensin-II acts at its type-1 receptor to stimulate vasopressin (AVP) secretion, which may contribute to angiotensin-II-induced hypertension. Less well known is the impact of angiotensin type-2 receptor (AT2R) activation on these processes. Studies conducted in a transgenic AT2R enhanced green fluorescent protein reporter mouse revealed that although AT2R are not themselves localized to AVP neurons within the paraventricular nucleus of the hypothalamus (PVN), they are localized to neurons that extend processes into the PVN. In the present set of studies, we set out to characterize the origin, phenotype, and function of nerve terminals within the PVN that arise from AT2R-enhanced green fluorescent protein-positive neurons and synapse onto AVP neurons. Initial experiments combined genetic and neuroanatomical techniques to determine that γ-aminobutyric acid (GABA)ergic neurons derived from the peri-PVN area containing AT2R make appositions onto AVP neurons within the PVN, thereby positioning AT2R to negatively regulate neuroendocrine secretion. Subsequent patch-clamp electrophysiological experiments revealed that selective activation of AT2R in the peri-PVN area using compound 21 facilitates inhibitory (ie, GABAergic) neurotransmission and leads to reduced activity of AVP neurons within the PVN. Final experiments determined the functional impact of AT2R activation by testing the effects of compound 21 on plasma AVP levels. Collectively, these experiments revealed that AT2R expressing neurons make GABAergic synapses onto AVP neurons that inhibit AVP neuronal activity and suppress baseline systemic AVP levels. These findings have direct implications in the targeting of AT2R for disorders of AVP secretion and also for the alleviation of high blood pressure.
Collapse
|
28
|
Central Infusion of Angiotensin II Type 2 Receptor Agonist Compound 21 Attenuates DOCA/NaCl-Induced Hypertension in Female Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3981790. [PMID: 26783414 PMCID: PMC4691472 DOI: 10.1155/2016/3981790] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/11/2015] [Accepted: 09/13/2015] [Indexed: 12/20/2022]
Abstract
The present study investigated whether central activation of angiotensin II type 2 receptor (AT2-R) attenuates deoxycorticosterone acetate (DOCA)/NaCl-induced hypertension in intact and ovariectomized (OVX) female rats and whether female sex hormone status has influence on the effects of AT2-R activation. DOCA/NaCl elicited a greater increase in blood pressure in OVX females than that in intact females. Central infusion of compound 21, a specific AT2-R agonist, abolished DOCA/NaCl pressor effect in intact females, whereas same treatment in OVX females produced an inhibitory effect. Real-time RT-PCR analysis revealed that DOCA/NaCl enhanced the mRNA expression of hypertensive components including AT1-R, ACE-1, and TNF-α in the paraventricular nucleus of hypothalamus in both intact and OVX females. However, the mRNA expressions of antihypertensive components such as AT2-R, ACE-2, and IL-10 were increased only in intact females. Central AT2-R agonist reversed the changes in the hypertensive components in all females, while this agonist further upregulated the expression of ACE2 and IL-10 in intact females, but only IL-10 in OVX females. These results indicate that brain AT2-R activation plays an inhibitory role in the development of DOCA/NaCl-induced hypertension in females. This beneficial effect of AT2-R activation involves regulation of renin-angiotensin system and proinflammatory cytokines.
Collapse
|
29
|
de Kloet AD, Liu M, Rodríguez V, Krause EG, Sumners C. Role of neurons and glia in the CNS actions of the renin-angiotensin system in cardiovascular control. Am J Physiol Regul Integr Comp Physiol 2015; 309:R444-58. [PMID: 26084692 DOI: 10.1152/ajpregu.00078.2015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/15/2015] [Indexed: 02/07/2023]
Abstract
Despite tremendous research efforts, hypertension remains an epidemic health concern, leading often to the development of cardiovascular disease. It is well established that in many instances, the brain plays an important role in the onset and progression of hypertension via activation of the sympathetic nervous system. Further, the activity of the renin-angiotensin system (RAS) and of glial cell-mediated proinflammatory processes have independently been linked to this neural control and are, as a consequence, both attractive targets for the development of antihypertensive therapeutics. Although it is clear that the predominant effector peptide of the RAS, ANG II, activates its type-1 receptor on neurons to mediate some of its hypertensive actions, additional nuances of this brain RAS control of blood pressure are constantly being uncovered. One of these complexities is that the RAS is now thought to impact cardiovascular control, in part, via facilitating a glial cell-dependent proinflammatory milieu within cardiovascular control centers. Another complexity is that the newly characterized antihypertensive limbs of the RAS are now recognized to, in many cases, antagonize the prohypertensive ANG II type 1 receptor (AT1R)-mediated effects. That being said, the mechanism by which the RAS, glia, and neurons interact to regulate blood pressure is an active area of ongoing research. Here, we review the current understanding of these interactions and present a hypothetical model of how these exchanges may ultimately regulate cardiovascular function.
Collapse
Affiliation(s)
- Annette D de Kloet
- Department of Physiology and Functional Genomics, and McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida; and
| | - Meng Liu
- Department of Physiology and Functional Genomics, and McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida; and
| | - Vermalí Rodríguez
- Department of Physiology and Functional Genomics, and McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida; and
| | - Eric G Krause
- Department of Pharmacodynamics, University of Florida College of Pharmacy, Gainesville, Florida
| | - Colin Sumners
- Department of Physiology and Functional Genomics, and McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida; and
| |
Collapse
|