1
|
Martins ALV, Annoni F, da Silva FA, Bolais-Ramos L, de Oliveira GC, Ribeiro RC, Diniz MML, Silva TGF, Pinheiro BD, Rodrigues NA, Dos Santos Matos AH, Motta-Santos D, Campagnole-Santos MJ, Verano-Braga T, Taccone FS, Santos RAS. Angiotensin-(1-7) infusion in COVID-19 patients admitted to the ICU: a seamless phase 1-2 randomized clinical trial. Ann Intensive Care 2024; 14:139. [PMID: 39231898 PMCID: PMC11374945 DOI: 10.1186/s13613-024-01369-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 08/17/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND The coronavirus-related disease (COVID-19) is mainly characterized by a respiratory involvement. The renin-angiotensin system (RAS) has a relevant role in the pathogenesis of COVID-19, as the virus enters host's cells via the angiotensin-converting enzyme 2 (ACE2). METHODS This investigator-initiated, seamless phase 1-2 randomized clinical trial was conceived to test the safety and efficacy of continuous short-term (up to 7 days) intravenous administration of Angiotensin-(1-7) in COVID-19 patients admitted to two intensive care units (ICU). In addition to standard of care, intravenous administration of Angiotensin-(1-7) was started at 5 mcg/Kg day and increased to 10 mcg/Kg day after 24 h (Phase 1; open label trial) or given at 10 mcg/Kg day and continued for a maximum of 7 days or until ICU discharge (Phase 2; double-blind randomized controlled trial). The rate of serious adverse events (SAEs) served as the primary outcome of the study for Phase 1, and the number of oxygen free days (OFDs) by day 28 for Phase 2. RESULTS Between August 2020 and July 2021, when the study was prematurely stopped due to low recruitment rate, 28 patients were included in Phase 1 and 79 patients in Phase 2. Of those, 78 were included in the intention to treat analysis, and the primary outcome was available for 77 patients. During Phase 1, one SAE (i.e., bradycardia) was considered possibly related to the infusion, justifying its discontinuation. In Phase 2, OFDs did not differ between groups (median 19 [0-21] vs. 14 [0-18] days; p = 0.15). When patients from both phases were analyzed in a pooled intention to treat approach (Phase 1-2 trial), OFDs were significantly higher in treated patients, when compared to controls (19 [0-21] vs. 14 [0-18] days; absolute difference -5 days, 95% CI [0-7] p = 0.04). CONCLUSIONS The main findings of our study indicate that continuous intravenous infusion of Angiotensin-(1-7) at 10 mcg/Kg day in COVID-19 patients admitted to the ICU with severe pneumonia is safe. In Phase II intention to treat analysis, there was no significant difference in OFD between groups. Trial Registration ClinicalTrials.gov Identifier: NCT04633772-Registro Brasileiro de Ensaios Clínicos, UTN number: U1111-1255-7167.
Collapse
Affiliation(s)
- Ana Luiza Valle Martins
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Filippo Annoni
- Department of Intensive Care Erasme Hospital, University Hospital of Brussels (HUB), Lennik Road 808, 1070, Brussels, Belgium
| | - Filipe Alex da Silva
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Lucas Bolais-Ramos
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Gisele Capanema de Oliveira
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Renata Cunha Ribeiro
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Mirella Monique Lana Diniz
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Thuanny Granato Fonseca Silva
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Beatriz Dias Pinheiro
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Natália Abdo Rodrigues
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Alana Helen Dos Santos Matos
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Daisy Motta-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Maria José Campagnole-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Thiago Verano-Braga
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil
| | - Fabio Silvio Taccone
- Department of Intensive Care Erasme Hospital, University Hospital of Brussels (HUB), Lennik Road 808, 1070, Brussels, Belgium
| | - Robson Augusto Souza Santos
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais Av. Antonio Carlos, 6627-ICB-UFMG, Belo Horizonte, 31270-901, Brazil.
| |
Collapse
|
2
|
Cerniello FM, Silva MG, Carretero OA, Gironacci MM. Mas receptor is translocated to the nucleus upon agonist stimulation in brainstem neurons from spontaneously hypertensive rats but not normotensive rats. Cardiovasc Res 2021; 116:1995-2008. [PMID: 31825460 DOI: 10.1093/cvr/cvz332] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 10/31/2019] [Accepted: 12/09/2019] [Indexed: 12/23/2022] Open
Abstract
AIMS Activation of the angiotensin (Ang)-(1-7)/Mas receptor (R) axis protects from sympathetic overactivity. Endocytic trafficking is an essential process that regulates receptor (R) function and its ultimate cellular responses. We investigated whether the blunted responses to Ang-(1-7) in hypertensive rats are associated to an alteration in MasR trafficking. METHODS AND RESULTS Brainstem neurons from Wistar-Kyoto (WKY) or spontaneously hypertensive rats (SHRs) were investigated for (i) Ang-(1-7) levels and binding and MasR expression, (ii) Ang-(1-7) responses (arachidonic acid and nitric oxide release and Akt and ERK1/2 phosphorylation), and (iii) MasR trafficking. Ang-(1-7) was determined by radioimmunoassay. MasR expression and functionality were evaluated by western blot and binding assays. MasR trafficking was evaluated by immunofluorescence. Ang-(1-7) treatment induced an increase in nitric oxide and arachidonic acid release and ERK1/2 and Akt phosphorylation in WKY neurons but did not have an effect in SHR neurons. Although SHR neurons showed greater MasR expression, Ang-(1-7)-elicited responses were substantially diminished presumably due to decreased Ang-(1-7) endogenous levels concomitant with impaired binding to its receptor. Through immunocolocalization studies, we evidenced that upon Ang-(1-7) stimulation MasRs were internalized through clathrin-coated pits and caveolae into early endosomes and slowly recycled back to the plasma membrane. However, the fraction of internalized MasRs into early endosomes was larger and the fraction of MasRs recycled back to the plasma membrane was smaller in SHR than in WKY neurons. Surprisingly, in SHR neurons but not in WKY neurons, Ang-(1-7) induced MasR translocation to the nucleus. Nuclear MasR expression and Ang-(1-7) levels were significantly greater in the nuclei of Ang-(1-7)-stimulated SHR neurons, indicating that the MasR is translocated with its ligand bound to it. CONCLUSION MasRs display differential trafficking in brainstem neurons from SHRs, which may contribute to the impaired responses to Ang-(1-7).
Collapse
Affiliation(s)
- Flavia M Cerniello
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, IQUIFIB (UBA-CONICET), Dpto. Química Biológica, Junín 956, 1113, Buenos Aires, Argentina
| | - Mauro G Silva
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, IQUIFIB (UBA-CONICET), Dpto. Química Biológica, Junín 956, 1113, Buenos Aires, Argentina
| | - Oscar A Carretero
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI, USA
| | - Mariela M Gironacci
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, IQUIFIB (UBA-CONICET), Dpto. Química Biológica, Junín 956, 1113, Buenos Aires, Argentina
| |
Collapse
|
3
|
Stoyell-Conti FF, Itty S, Abraham C, Rigatto K, West CA, Speth RC. 125I-Angiotensin 1-7 binds to a different site than angiotensin 1-7 in tissue membrane preparations. Endocrine 2021; 72:529-538. [PMID: 33415576 DOI: 10.1007/s12020-020-02572-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE To study the receptor for Angiotensin (Ang) 1-7 using a radioligand (125I-Ang 1-7)-binding assay. For more than a decade, Mas has been viewed as the receptor for Ang 1-7; however, Ang 1-7 binding has not been pharmacologically characterized in tissue membrane preparations. METHODS Radioligand-binding assays were carried out using tissue membrane preparations using radioiodinated Angiotensin 1-7 (125I-Ang 1-7) to characterize its binding site. Non-radioactive 127I-Ang 1-7 was used to test if the addition of an iodine to the tyrosine4 moiety of Ang 1-7 changes the ability of Ang 1-7 to competitively inhibit 125I-Ang 1-7 binding. RESULTS 125I-Ang 1-7 binds saturably, with moderately high affinity (10-20 nM) to a binding site in rat liver membranes that is displaceable by 127I-Ang 1-7 at nanomolar concentrations (IC50 = 62 nM) while Ang 1-7 displaces at micromolar concentrations (IC50 = 80 µM) at ~22 °C. This binding was also displaceable by inhibitors of metalloproteases at room temperature. This suggests that 125I-Ang 1-7 binds to MMPs and/or ADAMs as well as other liver membrane elements at ~ 22 °C. However, when 125I-Ang 1-7-binding assays were run at 0-4 °C, the same MMP inhibitors did not effectively compete for 125I-Ang 1-7. CONCLUSIONS The addition of an iodine molecule to the tyrosine in position 4 of Ang 1-7 drastically changes the binding characteristics of this peptide making it unsuitable for characterization of Ang 1-7 receptors.
Collapse
Affiliation(s)
- Filipe F Stoyell-Conti
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Sarin Itty
- Halmos College of Natural Science & Oceanography, Nova Southeastern University, Fort Lauderdale, FL, USA
- Kiran P. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Christy Abraham
- Halmos College of Natural Science & Oceanography, Nova Southeastern University, Fort Lauderdale, FL, USA
- Kiran P. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Katya Rigatto
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
- Laboratório de Fisiologia Translacional, Universidade Federal de Ciências da Saúde de Porto, Alegre, RS, Brazil
| | - Crystal A West
- Department of Biology, Appalachian State University, North Carolina Research Campus, Kannapolis, NC, USA
| | - Robert C Speth
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA.
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, DC, USA.
| |
Collapse
|
4
|
Patel S, Hussain T. Synergism between Angiotensin receptors ligands: Role of Angiotensin-(1-7) in modulating AT 2 R agonist response on nitric oxide in kidney cells. Pharmacol Res Perspect 2020; 8:e00667. [PMID: 33197136 PMCID: PMC7668194 DOI: 10.1002/prp2.667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 01/04/2023] Open
Abstract
Angiotensin-(1-7), an endogenous agonist for the MasR, has been shown to interact with ang-II AT1 R and AT2 R. Earlier we showed a physical and functional interaction between MasR and AT2 R in response to their respective agonists ang-(1-7) and C21. Moreover, ang-(1-7) is cardio-protective via AT1 R and alters ang-II function. Such complex nature of ang-(1-7) function is not clearly understood, particularly in relation to its functional interaction with these receptors. We tested how ang-(1-7) affects AT2 R function by utilizing HK-2 cells. The HK-2 cells were treated with a wide range of concentrations of angiotensin receptor agonists. The generation of NO• in response to agonists was determined as a readout and subjected to Bliss definition (δ score) to assess the nature of functional interaction between these receptors. Preincubation with ang-(1-7) followed by incubation with endogenous AT1 R/AT2 R agonist ang-II (δ = 162) or selective AT2 R agonist C21 (δ = 304) synergized NO• formation. The synergism was also observed when the order of incubation with ang-(1-7)/C21 was reversed (δ = 484), but not when the cells were simultaneously incubated with a mixture of ang-(1-7) and C21 (δ = 76). The synergism with nonpeptidic MasR agonist AVE0991 followed by C21 (δ = 45) was minimal. Ligand binding experiment suggested the binding of ang-(1-7) with these three receptors. However, the synergism observed with ang-(1-7) and ang-II/C21 was sensitive to the antagonists of AT2 R (PD123319) and AT1 R (candesartan), but not MasR (A779). Ang-(1-7) at lower concentrations synergies the AT2 R function in an AT1 R-dependent but MasR-independent manner. This phenomenon may have a physiological significance.
Collapse
Affiliation(s)
- Sanket Patel
- Department of Pharmacological and Pharmaceutical SciencesCollege of PharmacyUniversity of HoustonHoustonTXUSA
| | - Tahir Hussain
- Department of Pharmacological and Pharmaceutical SciencesCollege of PharmacyUniversity of HoustonHoustonTXUSA
| |
Collapse
|
5
|
Viana SD, Nunes S, Reis F. ACE2 imbalance as a key player for the poor outcomes in COVID-19 patients with age-related comorbidities - Role of gut microbiota dysbiosis. Ageing Res Rev 2020; 62:101123. [PMID: 32683039 PMCID: PMC7365123 DOI: 10.1016/j.arr.2020.101123] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 patients with pre-existing age-related comorbidities have poor outcomes. Gut microbiota dysbiosis is associated with ageing and age-related diseases. Viral-mediated ACE2 shedding favors poor outcomes by RAS-dependent mechanisms. Viral-mediated ACE2 shedding favors poor outcomes by RAS-independent gut dysbiosis. Potential of ACE2 and gut microbiota-based therapeutic opportunities for COVID-19.
Coronavirus disease 19 (COVID-19) is a pandemic condition caused by the new coronavirus SARS-CoV-2. The typical symptoms are fever, cough, shortness of breath, evolving to a clinical picture of pneumonia and, ultimately, death. Nausea and diarrhea are equally frequent, suggesting viral infection or transmission via the gastrointestinal-enteric system. SARS-CoV-2 infects human cells by using angiotensin converting enzyme 2 (ACE2) as a receptor, which is cleaved by transmembrane proteases during host cells infection, thus reducing its activities. ACE2 is a relevant player in the renin-angiotensin system (RAS), counterbalancing the deleterious effects of angiotensin II. Furthermore, intestinal ACE2 functions as a chaperone for the aminoacid transporter B0AT1. It has been suggested that B0AT1/ACE2 complex in the intestinal epithelium regulates gut microbiota (GM) composition and function, with important repercussions on local and systemic immune responses against pathogenic agents, namely virus. Notably, productive infection of SARS-CoV-2 in ACE2+ mature human enterocytes and patients’ GM dysbiosis was recently demonstrated. This review outlines the evidence linking abnormal ACE2 functions with the poor outcomes (higher disease severity and mortality rate) in COVID-19 patients with pre-existing age-related comorbidities and addresses a possible role for GM dysbiosis. The article culminates with the therapeutics opportunities based on these pathways.
Collapse
|
6
|
Involvement of ACE2/Ang-(1-7)/MAS1 Axis in the Regulation of Ovarian Function in Mammals. Int J Mol Sci 2020; 21:ijms21134572. [PMID: 32604999 PMCID: PMC7369927 DOI: 10.3390/ijms21134572] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
In addition to the classic, endocrine renin-angiotensin system, local renin-angiotensin system (RAS) has been documented in many tissues and organs, including the ovaries. The localization and functional activity of the two opposing axes of the system, viz. ACE1/Ang II/AT1 and ACE2/Ang-(1-7)/MAS1, differs between animal species and varied according to the stage of follicle development. It appears that the angiotensin peptides and their receptors participate in reproductive processes such as folliculogenesis, steroidogenesis, oocyte maturation, and ovulation. In addition, changes in the constituent compounds of local RAS may contribute to pathological conditions, such as polycystic ovary syndrome, ovarian hyperstimulation syndrome, and ovarian cancer. This review article examines the expression, localization, metabolism, and activity of individual elements of the ACE2/Ang-(1-7)/MAS1 axis in the ovaries of various animal species. The manuscript also presents the relationship between the secretion of gonadotropins and sex hormones and expression of Ang-(1-7) and MAS1 receptors. It also summarizes current knowledge regarding the positive and negative impact of ACE2/Ang-(1-7)/MAS1 axis on ovarian function.
Collapse
|
7
|
Exner EC, Geurts AM, Hoffmann BR, Casati M, Stodola T, Dsouza NR, Zimmermann M, Lombard JH, Greene AS. Interaction between Mas1 and AT1RA contributes to enhancement of skeletal muscle angiogenesis by angiotensin-(1-7) in Dahl salt-sensitive rats. PLoS One 2020; 15:e0232067. [PMID: 32324784 PMCID: PMC7179868 DOI: 10.1371/journal.pone.0232067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
The heptapeptide angiotensin-(1-7) (Ang-(1-7)) is protective in the cardiovascular system through its induction of vasodilator production and angiogenesis. Despite acting antagonistically to the effects of elevated, pathophysiological levels of angiotensin II (AngII), recent evidence has identified convergent and beneficial effects of low levels of both Ang-(1-7) and AngII. Previous work identified the AngII receptor type I (AT1R) as a component of the protein complex formed when Ang-(1-7) binds its receptor, Mas1. Importantly, pharmacological blockade of AT1R did not alter the effects of Ang-(1-7). Here, we use a novel mutation of AT1RA in the Dahl salt-sensitive (SS) rat to test the hypothesis that interaction between Mas1 and AT1R contributes to proangiogenic Ang-(1-7) signaling. In a model of hind limb angiogenesis induced by electrical stimulation, we find that the restoration of skeletal muscle angiogenesis in SS rats by Ang-(1-7) infusion is impaired in AT1RA knockout rats. Enhancement of endothelial cell (EC) tube formation capacity by Ang-(1-7) is similarly blunted in AT1RA mutant ECs. Transcriptional changes elicited by Ang-(1-7) in SS rat ECs are altered in AT1RA mutant ECs, and tandem mass spectrometry-based proteomics demonstrate that the protein complex formed upon binding of Ang-(1-7) to Mas1 is altered in AT1RA mutant ECs. Together, these data support the hypothesis that interaction between AT1R and Mas1 contributes to proangiogenic Ang-(1-7) signaling.
Collapse
MESH Headings
- Angiotensin I/metabolism
- Animals
- Electric Stimulation
- Male
- Mass Spectrometry
- Models, Animal
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/metabolism
- Mutation
- Neovascularization, Physiologic
- Peptide Fragments/metabolism
- Proteomics
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/metabolism
- Rats
- Rats, Inbred Dahl
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Eric C. Exner
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Aron M. Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Brian R. Hoffmann
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Bioengineering, Medical College of Wisconsin and Marquette University, Milwaukee, Wisconsin, United States of America
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Marc Casati
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Timothy Stodola
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Nikita R. Dsouza
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Michael Zimmermann
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Julian H. Lombard
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Andrew S. Greene
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| |
Collapse
|
8
|
Burghi V, Echeverría EB, Sosa MH, Quiroga DT, Muñoz MC, Davio C, Monczor F, Fernández NC, Dominici FP. Participation of Gα i-Adenylate Cyclase and ERK1/2 in Mas Receptor Signaling Pathways. Front Pharmacol 2019; 10:146. [PMID: 30853914 PMCID: PMC6395383 DOI: 10.3389/fphar.2019.00146] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/06/2019] [Indexed: 12/21/2022] Open
Abstract
The MasR receptor (MasR) is an orphan G protein-coupled receptor proposed as a candidate for mediating the angiotensin (Ang)-converting enzyme 2-Ang-(1–7) protective axis of renin-angiotensin system. This receptor has been suggested to participate in several physiological processes including cardio- and reno-protection and regulation of the central nervous system function. Although the knowledge of the signaling mechanisms associated with MasR is essential for therapeutic purposes, these are still poorly understood. Accordingly, in the current study we aimed to characterize the signaling pathways triggered by the MasR. To do that, we measured cAMP and Ca2+ levels in both naïve and MasR transfected cells in basal conditions and upon incubation with putative MasR ligands. Besides, we evaluated activation of ERK1/2 by Ang-(1–7) in MasR transfected cells. Results indicated the existence of a high degree of MasR constitutive activity toward cAMP modulation. This effect was not mediated by the PDZ-binding motif of the MasR but by receptor coupling to Gαi-adenylyl cyclase signaling pathway. Incubation of MasR transfected cells with Ang-(1–7) or the synthetic ligand AVE 0991 amplified MasR negative modulation of cAMP levels. On the other hand, we provided evidence for lack of MasR-associated modulation of Ca2+ levels by Ang-(1–7). Finally, it was determined that the MasR attenuated Ang-(1–7)-induced ERK1/2 phosphorylation mediated by AT1R. We provided further characterization of MasR signaling mechanisms regarding its constitutive activity and response to putative ligands. This information could prove useful to better describe MasR physiological role and development of therapeutic agents that could modulate its action.
Collapse
Affiliation(s)
- Valeria Burghi
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Emiliana B Echeverría
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Máximo H Sosa
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Diego T Quiroga
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Marina C Muñoz
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Carlos Davio
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Federico Monczor
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Natalia C Fernández
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Fernando P Dominici
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| |
Collapse
|
9
|
Ang-(1-7) is an endogenous β-arrestin-biased agonist of the AT 1 receptor with protective action in cardiac hypertrophy. Sci Rep 2017; 7:11903. [PMID: 28928410 PMCID: PMC5605686 DOI: 10.1038/s41598-017-12074-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 09/04/2017] [Indexed: 01/13/2023] Open
Abstract
The renin-angiotensin system (RAS) plays a key role in the control of vasoconstriction as well as sodium and fluid retention mediated mainly by angiotensin (Ang) II acting at the AT1 receptor (AT1R). Ang-(1-7) is another RAS peptide, identified as the endogenous ligand of the Mas receptor and known to counterbalance many of the deleterious effects of AngII. AT1R signaling triggered by β-arrestin-biased agonists has been associated to cardioprotection. Because position 8 in AngII is important for G protein activation, we hypothesized that Ang-(1-7) could be an endogenous β-arrestin-biased agonist of the AT1R. Here we show that Ang-(1-7) binds to the AT1R without activating Gq, but triggering β-arrestins 1 and 2 recruitment and activation. Using an in vivo model of cardiac hypertrophy, we show that Ang-(1-7) significantly attenuates heart hypertrophy by reducing both heart weight and ventricular wall thickness and the increased end-diastolic pressure. Whereas neither the single blockade of AT1 or Mas receptors with their respective antagonists prevented the cardioprotective action of Ang1-7, combination of the two antagonists partially impaired the effect of Ang-(1-7). Taken together, these data indicate that Ang-(1-7) mediates at least part of its cardioprotective effects by acting as an endogenous β-arrestin-biased agonist at the AT1R.
Collapse
|
10
|
Galandrin S, Denis C, Boularan C, Marie J, M'Kadmi C, Pilette C, Dubroca C, Nicaise Y, Seguelas MH, N'Guyen D, Banères JL, Pathak A, Sénard JM, Galés C. Cardioprotective Angiotensin-(1-7) Peptide Acts as a Natural-Biased Ligand at the Angiotensin II Type 1 Receptor. Hypertension 2016; 68:1365-1374. [PMID: 27698068 DOI: 10.1161/hypertensionaha.116.08118] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 07/14/2016] [Accepted: 08/25/2016] [Indexed: 12/14/2022]
Abstract
Hyperactivity of the renin-angiotensin-aldosterone system through the angiotensin II (Ang II)/Ang II type 1 receptor (AT1-R) axis constitutes a hallmark of hypertension. Recent findings indicate that only a subset of AT1-R signaling pathways is cardiodeleterious, and their selective inhibition by biased ligands promotes therapeutic benefit. To date, only synthetic biased ligands have been described, and whether natural renin-angiotensin-aldosterone system peptides exhibit functional selectivity at AT1-R remains unknown. In this study, we systematically determined efficacy and potency of Ang II, Ang III, Ang IV, and Ang-(1-7) in AT1-R-expressing HEK293T cells on the activation of cardiodeleterious G-proteins and cardioprotective β-arrestin2. Ang III and Ang IV fully activate similar G-proteins than Ang II, the prototypical AT1-R agonist, despite weaker potency of Ang IV. Interestingly, Ang-(1-7) that binds AT1-R fails to promote G-protein activation but behaves as a competitive antagonist for Ang II/Gi and Ang II/Gq pathways. Conversely, all renin-angiotensin-aldosterone system peptides act as agonists on the AT1-R/β-arrestin2 axis but display biased activities relative to Ang II as indicated by their differences in potency and AT1-R/β-arrestin2 intracellular routing. Importantly, we reveal Ang-(1-7) a known Mas receptor-specific ligand, as an AT1-R-biased agonist, selectively promoting β-arrestin activation while blocking the detrimental Ang II/AT1-R/Gq axis. This original pharmacological profile of Ang-(1-7) at AT1-R, similar to that of synthetic AT1-R-biased agonists, could, in part, contribute to its cardiovascular benefits. Accordingly, in vivo, Ang-(1-7) counteracts the phenylephrine-induced aorta contraction, which was blunted in AT1-R knockout mice. Collectively, these data suggest that Ang-(1-7) natural-biased agonism at AT1-R could fine-tune the physiology of the renin-angiotensin-aldosterone system.
Collapse
Affiliation(s)
- Ségolène Galandrin
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Colette Denis
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Cédric Boularan
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Jacky Marie
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Céline M'Kadmi
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Claire Pilette
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Caroline Dubroca
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Yvan Nicaise
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Marie-Hélène Seguelas
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Du N'Guyen
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Jean-Louis Banères
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Atul Pathak
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Jean-Michel Sénard
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France
| | - Céline Galés
- From the Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université de Toulouse, France (S.G., C.D., C.B., M.-H.S., D.N., A.P., J.-M.S., C.G.); Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier Cedex 05, France (J.M., C.M., J.-L.B.); Cardiomedex SAS, Toulouse, France (C.P., C.D.); and Département d'histopathologie (Y.N.) and Service de Pharmacologie Clinique, Faculté de médecine (D.N., A.P., J.-M.S.), Centre Hospitalier Universitaire de Toulouse, France.
| |
Collapse
|
11
|
Qaradakhi T, Apostolopoulos V, Zulli A. Angiotensin (1-7) and Alamandine: Similarities and differences. Pharmacol Res 2016; 111:820-826. [PMID: 27456244 DOI: 10.1016/j.phrs.2016.07.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 07/19/2016] [Accepted: 07/21/2016] [Indexed: 12/31/2022]
Abstract
A primary peptide of the renin angiotensin system (RAS), Angiotensin (Ang) II, is a vasoconstrictor and promotor of atherosclerosis. To counter this, the RAS also consists of peptides and receptors which increase nitric oxide release from the endothelium and decrease nicotinamide adenine dinucleotide phosphate oxidase-related superoxide production. Two peptides, Ang (1-7) and alamandine are vasodilators, by activating the nitric oxide pathway via different receptors in the endothelium. Thus, herein we focus on the similarities and differences between alamandine and Ang (1-7) and the counterbalancing hypothesis on Ang II during endothelial dysfunction and atherosclerosis.
Collapse
Affiliation(s)
- Tawar Qaradakhi
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Werribee Campus, 3030 Victoria, Australia.
| | - Vasso Apostolopoulos
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Werribee Campus, 3030 Victoria, Australia
| | - Anthony Zulli
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Werribee Campus, 3030 Victoria, Australia
| |
Collapse
|
12
|
Dominguez JM, Hu P, Caballero S, Moldovan L, Verma A, Oudit GY, Li Q, Grant MB. Adeno-Associated Virus Overexpression of Angiotensin-Converting Enzyme-2 Reverses Diabetic Retinopathy in Type 1 Diabetes in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1688-700. [PMID: 27178803 DOI: 10.1016/j.ajpath.2016.01.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 01/07/2016] [Accepted: 01/19/2016] [Indexed: 12/17/2022]
Abstract
Angiotensin-converting enzyme (ACE)-2 is the primary enzyme of the vasoprotective axis of the renin angiotensin system that regulates the classic renin angiotensin system axis. We aimed to determine whether local retinal overexpression of adenoassociated virus (AAV)-ACE2 prevents or reverses diabetic retinopathy. Green fluorescent protein (GFP)-chimeric mice were generated to distinguish resident (retinal) from infiltrating bone marrow-derived inflammatory cells and were made diabetic using streptozotocin injections. Retinal digestion using trypsin was performed and acellular capillaries enumerated. Capillary occlusion by GFP(+) cells was used to measure leukostasis. Overexpression of ACE2 prevented (prevention cohort: untreated diabetic, 11.3 ± 1.4; ACE2 diabetic, 6.4 ± 0.9 per mm(2)) and partially reversed (reversal cohort: untreated diabetic, 15.7 ± 1.9; ACE2 diabetic, 6.5 ± 1.2 per mm(2)) the diabetes-associated increase of acellular capillaries and the increase of infiltrating inflammatory cells into the retina (F4/80(+)) (prevention cohort: untreated diabetic, 24.2 ± 6.7; ACE2 diabetic, 2.5 ± 1.6 per mm(2); reversal cohort: untreated diabetic, 56.8 ± 5.2; ACE2 diabetic, 5.6 ± 2.3 per mm(2)). In both study cohorts, intracapillary bone marrow-derived cells, indicative of leukostasis, were only observed in diabetic animals receiving control AAV injections. These results indicate that diabetic retinopathy, and possibly other diabetic microvascular complications, can be prevented and reversed by locally restoring the balance between the classic and vasoprotective renin angiotensin system.
Collapse
Affiliation(s)
- James M Dominguez
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, University of Florida, Gainesville, Florida; Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ping Hu
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sergio Caballero
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Leni Moldovan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Amrisha Verma
- Department of Ophthalmology and Powell Gene Therapy Center, University of Florida College of Medicine, University of Florida, Gainesville, Florida
| | - Gavin Y Oudit
- Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Qiuhong Li
- Department of Ophthalmology and Powell Gene Therapy Center, University of Florida College of Medicine, University of Florida, Gainesville, Florida
| | - Maria B Grant
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
13
|
Jensen TW, Olsen NV. Angiotensin II during Experimentally Simulated Central Hypovolemia. Front Cardiovasc Med 2016; 3:6. [PMID: 26973842 PMCID: PMC4776081 DOI: 10.3389/fcvm.2016.00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/18/2016] [Indexed: 11/13/2022] Open
Abstract
Central hypovolemia, defined as diminished blood volume in the heart and pulmonary vascular bed, is still an unresolved problem from a therapeutic point of view. The development of pharmaceutical agents targeted at specific angiotensin II receptors, such as the non-peptidergic AT2-receptor agonist compound 21, is yielding many opportunities to uncover more knowledge about angiotensin II receptor profiles and possible therapeutic use. Cardiovascular, anti-inflammatory, and neuroprotective therapeutic use of compound 21 have been suggested. However, there has not yet been a focus on the use of these agents in a hypovolemic setting. We argue that the latest debates on the effect of angiotensin II during hypovolemia might guide for future studies, investigating the effect of such agents during experimentally simulated central hypovolemia. The purpose of this review is to examine the role of angiotensin II during episodes of central hypovolemia. To examine this, we reviewed results from studies with three experimental models of simulated hypovolemia: head up tilt table test, lower body negative pressure, and hemorrhage of animals. A systemic literature search was made with the use of PubMed/MEDLINE for studies that measured variables of the renin–angiotensin system or its effect during simulated hypovolemia. Twelve articles, using one of the three models, were included and showed a possible organ-protective effect and an effect on the sympathetic system of angiotensin II during hypovolemia. The results support the possible organ-protective vasodilatory role for the AT2-receptor during hypovolemia on both the kidney and the splanchnic tissue.
Collapse
Affiliation(s)
- Theo Walther Jensen
- Department of Neuroscience and Pharmacology, The Health Faculty, University of Copenhagen , Copenhagen , Denmark
| | - Niels Vidiendal Olsen
- Department of Neuroscience and Pharmacology, The Health Faculty, University of Copenhagen, Copenhagen, Denmark; Department of Neuroanaesthesia, The Neuroscience Centre, University Hospital of Copenhagen (Rigshospitalet), Copenhagen, Denmark
| |
Collapse
|
14
|
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]
|
15
|
Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, Liu FF, Zhang K, Zhang C. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat Rev Cardiol 2014; 11:413-26. [PMID: 24776703 PMCID: PMC7097196 DOI: 10.1038/nrcardio.2014.59] [Citation(s) in RCA: 293] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Angiotensin-converting enzyme (ACE) 2 and its product angiotensin 1–7 are thought to have effects that counteract the adverse actions of other, better-known renin–angiotensin system (RAS) components Numerous experimental studies have suggested that ACE2 and angiotensin 1–7 have notable protective effects in the heart and blood vessels ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the functional importance and signalling mechanisms of angiotensin-1–7-induced actions remain unclear New pharmacological interventions targeting ACE2 are expected to be useful in clinical treatment of cardiovascular disease, especially those associated with overactivation of the conventional RAS More studies, especially randomized controlled clinical trials, are needed to clearly delineate the benefits of therapies targeting angiotensin 1–7 actions
Angiotensin-converting enzyme 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of the better-known members of the renin–angiotensin system and might, therefore, be useful therapeutic targets in patients with cardiovascular disease. Professor Jiang and colleagues review the evidence for the potential roles of these proteins in various cardiovascular conditions, including hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes. The renin–angiotensin system (RAS) has pivotal roles in the regulation of normal physiology and the pathogenesis of cardiovascular disease. Angiotensin-converting enzyme (ACE) 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of other, better known and understood, members of the RAS. The physiological and pathological importance of ACE2 and angiotensin 1–7 in the cardiovascular system are not completely understood, but numerous experimental studies have indicated that these components have protective effects in the heart and blood vessels. Here, we provide an overview on the basic properties of ACE2 and angiotensin 1–7 and a summary of the evidence from experimental and clinical studies of various pathological conditions, such as hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes mellitus. ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the relevant functions and signalling mechanisms of actions induced by angiotensin 1–7 have not been conclusively determined. The ACE2–angiotensin 1–7 pathway, however, might provide a useful therapeutic target for the treatment of cardiovascular disease, especially in patients with overactive RAS.
Collapse
Affiliation(s)
- Fan Jiang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Jianmin Yang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Yongtao Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Mei Dong
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Shuangxi Wang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Fang Fang Liu
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Kai Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| |
Collapse
|
16
|
Perlot T, Penninger JM. ACE2 - from the renin-angiotensin system to gut microbiota and malnutrition. Microbes Infect 2013; 15:866-73. [PMID: 23962453 PMCID: PMC7110844 DOI: 10.1016/j.micinf.2013.08.003] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 08/06/2013] [Indexed: 02/07/2023]
Abstract
The renin-angiotensin system (RAS) is a complex network that regulates blood pressure, electrolyte and fluid homeostasis, as well as the function of several organs. Angiotensin-converting enzyme 2 (ACE2) was identified as an enzyme that negatively regulates the RAS by converting Ang II, the main bioactive molecule of the RAS, to Ang 1-7. Thus, ACE2 counteracts the role of angiotensin-converting enzyme (ACE) which generates Ang II from Ang I. ACE and ACE2 have been implicated in several pathologies such as cardiovascular and renal disease or acute lung injury. In addition, ACE2 has functions independent of the RAS: ACE2 is the receptor for the SARS coronavirus and ACE2 is essential for expression of neutral amino acid transporters in the gut. In this context, ACE2 modulates innate immunity and influences the composition of the gut microbiota, which can explain diarrhea and intestinal inflammation observed in Hartnup disorder, Pellagra, or under conditions of severe malnutrition. Here we review and discuss the diverse functions of ACE2 and its relevance to human pathologies.
Collapse
Affiliation(s)
- Thomas Perlot
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | | |
Collapse
|
17
|
Lee SH, Masuda T, Goto T, Oe T. MALDI-TOF/MS-based label-free binding assay for angiotensin II type 1 receptor: application for novel angiotensin peptides. Anal Biochem 2013; 437:10-6. [DOI: 10.1016/j.ab.2013.01.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 01/18/2013] [Accepted: 01/19/2013] [Indexed: 02/07/2023]
|
18
|
Santos RAS, Ferreira AJ, Verano-Braga T, Bader M. Angiotensin-converting enzyme 2, angiotensin-(1-7) and Mas: new players of the renin-angiotensin system. J Endocrinol 2013; 216:R1-R17. [PMID: 23092879 DOI: 10.1530/joe-12-0341] [Citation(s) in RCA: 363] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Angiotensin (Ang)-(1-7) is now recognized as a biologically active component of the renin-angiotensin system (RAS). Ang-(1-7) appears to play a central role in the RAS because it exerts a vast array of actions, many of them opposite to those attributed to the main effector peptide of the RAS, Ang II. The discovery of the Ang-converting enzyme (ACE) homolog ACE2 brought to light an important metabolic pathway responsible for Ang-(1-7) synthesis. This enzyme can form Ang-(1-7) from Ang II or less efficiently through hydrolysis of Ang I to Ang-(1-9) with subsequent Ang-(1-7) formation by ACE. In addition, it is now well established that the G protein-coupled receptor Mas is a functional binding site for Ang-(1-7). Thus, the axis formed by ACE2/Ang-(1-7)/Mas appears to represent an endogenous counterregulatory pathway within the RAS, the actions of which are in opposition to the vasoconstrictor/proliferative arm of the RAS consisting of ACE, Ang II, and AT(1) receptor. In this brief review, we will discuss recent findings related to the biological role of the ACE2/Ang-(1-7)/Mas arm in the cardiovascular and renal systems, as well as in metabolism. In addition, we will highlight the potential interactions of Ang-(1-7) and Mas with AT(1) and AT(2) receptors.
Collapse
Affiliation(s)
- Robson A S Santos
- Departments of Physiology and Biophysics Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | |
Collapse
|
19
|
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: 4.1] [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]
|
20
|
Cerrato BD, Frasch AP, Nakagawa P, Longo-Carbajosa N, Peña C, Höcht C, Gironacci MM. Angiotensin-(1–7) upregulates central nitric oxide synthase in spontaneously hypertensive rats. Brain Res 2012; 1453:1-7. [DOI: 10.1016/j.brainres.2012.03.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Revised: 02/17/2012] [Accepted: 03/08/2012] [Indexed: 11/15/2022]
|
21
|
Ognibene DT, Carvalho LC, Costa CA, Rocha APM, Moura RSD, Resende ÂC. Role of renin-angiotensin system and oxidative status on the maternal cardiovascular regulation in spontaneously hypertensive rats. Am J Hypertens 2012; 25:498-504. [PMID: 22258331 DOI: 10.1038/ajh.2011.253] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The purpose of this study was to investigate the contribution of renin-angiotensin system (RAS) and oxidative status on the maternal cardiovascular regulation at the end of pregnancy in normotensive and spontaneously hypertensive rats (SHR). METHODS Blood pressure (BP), mesenteric arterial bed (MAB) reactivity, mesenteric oxidative damage, protein expression, and antioxidant activities were compared between four groups: SHR (SHR-P) and normotensive Wistar controls (W-P) in the 20th day of pregnancy or age-matched nonpregnant rats (SHR-NP and W-NP). RESULTS BP in W-P and SHR-P was reduced at the end of pregnancy. The vasodilator effects of angiotensin II (Ang II) and angiotensin 1-7 (Ang-(1-7)) were higher in SHR-P than in other groups. Endothelial nitric oxide synthase (eNOS) expression was increased in W-P and SHR-P compared to nonpregnant groups. Angiotensin-converting enzyme (ACE) and AT(1) receptor expressions were increased in SHR-NP compared to normotensive groups and pregnancy reduced their expressions in SHR. No difference was observed in AT(2) receptor expression among the groups. ACE2 expression was higher in hypertensive than normotensive groups. The levels of thiobarbituric acid-reactive substances (TBARS) were reduced in pregnant compared to nonpregnant groups. Superoxide dismutase (SOD) activity was reduced in SHR-P compared to SHR-NP. However, pregnancy increased catalase (CAT) and glutathione peroxidase (GPx) activities in normotensive rats and SHR, respectively. CONCLUSIONS The results suggest that the reduction of BP to normal values at the end of pregnancy in SHR may be related to an increased NO production and vasorelaxation to Ang II and Ang-(1-7) associated with decreased expression of vascular ACE and AT(1) receptors and oxidative status.
Collapse
|
22
|
Liu GC, Oudit GY, Fang F, Zhou J, Scholey JW. Angiotensin-(1-7)-induced activation of ERK1/2 is cAMP/protein kinase A-dependent in glomerular mesangial cells. Am J Physiol Renal Physiol 2011; 302:F784-90. [PMID: 22189944 DOI: 10.1152/ajprenal.00455.2011] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The renin-angiotensin system (RAS) plays an important role in renal physiology and kidney injury. Although the cellular effects of the RAS activation are generally attributed to angiotensin II (ANG II), the recent identification of angiotensin-converting enzyme 2 has shifted the focus to other peptides including Ang-(1-7). The G protein-coupled receptor for Ang-(1-7), mas, is expressed by mesangial cells (MC) but the signal transduction pathways activated by Ang-(1-7) in MC have not been fully elucidated. Accordingly, we studied the effect of Ang-(1-7) on extracellular signal-related kinase (ERK)1/2 activation in rat MC. Ang-(1-7)-induced ERK1/2 phosphorylation in MC is time- and concentration-dependent. Pretreatment of MC with the mas receptor antagonist A-779 but not the AT(1) antagonist losartan or the AT(2) antagonist PD123319 abrogated ERK1/2 activation. Neither pretreatment with the NADPH oxidase inhibitors diphenyleneiodonium and apocynin nor pretreatment with the epidermal growth factor (EGF) receptor antagonists AG1478 and PD158780 attenuated Ang-(1-7)-induced activation of ERK1/2. Even though each of these compounds abolished ANG II-induced activation of ERK1/2. Ang-(1-7) increased intracellular cAMP levels and activated protein kinase A (PKA) and inhibition of either adenylyl cyclase or PKA activity attenuated Ang-(1-7)-induced ERK1/2 activation. In conclusion, Ang-(1-7)-induced activation of ERK1/2 is cAMP/PKA-dependent in MC, but independent of NADPH oxidase and the EGF receptor.
Collapse
Affiliation(s)
- George C Liu
- Department of Medicine and Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada M5G 2A8
| | | | | | | | | |
Collapse
|
23
|
Zhang T, Li Z, Dang H, Chen R, Liaw C, Tran TA, Boatman PD, Connolly DT, Adams JW. Inhibition of Mas G-protein signaling improves coronary blood flow, reduces myocardial infarct size, and provides long-term cardioprotection. Am J Physiol Heart Circ Physiol 2011; 302:H299-311. [PMID: 22003054 DOI: 10.1152/ajpheart.00723.2011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Mas receptor is a class I G-protein-coupled receptor that is expressed in brain, testis, heart, and kidney. The intracellular signaling pathways activated downstream of Mas are still largely unknown. In the present study, we examined the expression pattern and signaling of Mas in the heart and assessed the participation of Mas in cardiac ischemia-reperfusion injury. Mas mRNA and protein were present in all chambers of human hearts, with cardiomyocytes and coronary arteries being sites of enriched expression. Expression of Mas in either HEK293 cells or cardiac myocytes resulted in constitutive coupling to the G(q) protein, which in turn activated phospholipase C and caused inositol phosphate accumulation. To generate chemical tools for use in probing the function of Mas, we performed a library screen and chemistry optimization program to identify potent and selective nonpeptide agonists and inverse agonists. Mas agonists activated G(q) signaling in a dose-dependent manner and reduced coronary blood flow in isolated mouse and rat hearts. Conversely, treatment of isolated rat hearts with Mas inverse agonists improved coronary flow, reduced arrhythmias, and provided cardioprotection from ischemia-reperfusion injury, an effect that was due, at least in part, to decreased cardiomyocyte apoptosis. Participation of Mas in ischemia-reperfusion injury was confirmed in Mas knockout mice, which had reduced infarct size relative to mice with normal Mas expression. These results suggest that activation of Mas during myocardial infarction contributes to ischemia-reperfusion injury and further suggest that inhibition of Mas-G(q) signaling may provide a new therapeutic strategy directed at cardioprotection.
Collapse
Affiliation(s)
- Tong Zhang
- Arena Pharmaceuticals Incorporated, San Diego, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Gironacci MM, Adamo HP, Corradi G, Santos RA, Ortiz P, Carretero OA. Angiotensin (1-7) induces MAS receptor internalization. Hypertension 2011; 58:176-81. [PMID: 21670420 DOI: 10.1161/hypertensionaha.111.173344] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Angiotensin (Ang) (1-7) is the endogenous ligand for the G protein-coupled receptor Mas, a receptor associated with cardiac, renal, and cerebral protective responses. Physiological evidence suggests that Mas receptor (MasR) undergoes agonist-dependent desensitization, but the underlying molecular mechanism regulating receptor activity is unknown. We investigated the hypothesis that MasR desensitizes and internalizes on stimulation with Ang-(1-7). For this purpose, we generated a chimera between the MasR and the yellow fluorescent protein (YFP; MasR-YFP). MasR-YFP-transfected HEK 293T cells were incubated with Ang-(1-7), and the relative cellular distribution of MasR-YFP was observed by confocal microscopy. In resting cells, MasR-YFP was mostly localized to the cell membrane. Ang-(1-7) induced a redistribution of MasR-YFP to intracellular vesicles of various sizes after 5 minutes. Following the time course of [(125)I]Ang-(1-7) endocytosis, we observed that half of MasR-YFP underwent endocytosis after 10 minutes, and this was blocked by a MasR antagonist. MasR-YFP colocalized with Rab5, the early endosome antigen 1, and the adaptor protein complex 2, indicating that the R is internalized through a clathrin-mediated pathway and targeted to early endosomes after Ang-(1-7) stimulation. A fraction of MasR-YFP also colocalized with caveolin 1, suggesting that at some point MasR-YFP traverses caveolin 1-positive compartments. In conclusion, MasR undergoes endocytosis on stimulation with Ang-(1-7), and this event may explain the desensitization of MasR responsiveness. In this way, MasR activity and density may be tightly controlled by the cell.
Collapse
Affiliation(s)
- Mariela M Gironacci
- Instituto de Química y Fisicoquímica Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113 Buenos Aires, Argentina.
| | | | | | | | | | | |
Collapse
|
25
|
Lopez Verrilli MA, Pirola CJ, Pascual MM, Dominici FP, Turyn D, Gironacci MM. Angiotensin-(1-7) through AT2receptors mediates tyrosine hydroxylase degradation via the ubiquitin-proteasome pathway. J Neurochem 2009; 109:326-35. [DOI: 10.1111/j.1471-4159.2009.05912.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
26
|
Tang L, Carey LC, Bi J, Valego N, Sun X, Deibel P, Perrott J, Figueroa JP, Chappell MC, Rose JC. Gender differences in the effects of antenatal betamethasone exposure on renal function in adult sheep. Am J Physiol Regul Integr Comp Physiol 2009; 296:R309-17. [PMID: 19036827 PMCID: PMC2643986 DOI: 10.1152/ajpregu.90645.2008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Accepted: 11/20/2008] [Indexed: 11/22/2022]
Abstract
Exposure to clinically relevant doses of glucocorticoids during fetal life increases blood pressure in adult male and female sheep. The purpose of this study was to evaluate the effects of prenatal exposure to betamethasone at 80-81 days of gestation on renal function in ewes and rams at 1.5 yr of age. In prenatal betamethasone-exposed males, compared with the vehicle-exposed animals, basal glomerular filtration rate (GFR) (1.93 +/- 0.08 vs. 2.27 +/- 0.10 ml.min(-1).kg body wt(-1)) and the ability to excrete an acute Na+ load (37.1 +/- 4.4 vs. 53.7 +/- 9.7%) were reduced. (P < 0.03 and P = 0.03, respectively). In contrast, prenatal betamethasone exposure had no effect on basal GFR, Na+ excretion, or the percentage of the Na+ load excreted during the experiment in females. Systemic infusions of ANG-(1-7) at 9 ng.min(-1).kg(-1) for 2 h had minimal effects on basal GFR, renal plasma flow, and Na+ excretion in males but increased Na+ excretion in females. However, the percentage of Na+ load excreted during ANG-(1-7) infusion did not change in prenatal betamethasone-exposed females (113.1 +/- 14.2 vs. 98.1 +/- 12.2%) compared with the significant increase in vehicle females (139.2 +/- 22.3 vs. 92.2 +/- 7.5%) (P = 0.01). The data indicate that antenatal betamethasone exposure produces gender-specific alternations in renal function and thus suggest that different mechanisms underlie the antenatal steroid-induced elevations in blood pressure in male and female offspring.
Collapse
Affiliation(s)
- Lijun Tang
- Department of Obstetrics and Gynecology and Center of Research for Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zimpelmann J, Burns KD. Angiotensin-(1-7) activates growth-stimulatory pathways in human mesangial cells. Am J Physiol Renal Physiol 2008; 296:F337-46. [PMID: 19052102 DOI: 10.1152/ajprenal.90437.2008] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Angiotensin-(1-7) [Ang-(1-7)] is generated in part via ACE2-dependent degradation of angiotensin II (ANG II). In proximal tubular cells, Ang-(1-7) inhibits ANG II-stimulated phosphorylation of the mitogen-activated protein kinases (MAPKs) p38, extracellular signal-related kinase (ERK1/ERK2), and c-jun N-terminal kinase (JNK), suggesting that Ang-(1-7) protects against ANG II-mediated tubulointerstitial injury. We determined the effect of Ang-(1-7) on signaling and growth responses in cultured human mesangial cells. Ang-(1-7) increased phosphorylation of p38, ERK1/ERK2, and JNK MAPKs, which was blocked by the Ang-(1-7) antagonist A-779. Neither the AT(1) receptor antagonist losartan, nor the AT(2) antagonist PD123319 affected specific binding of [(125)I]Ang-(1-7) or Ang-(1-7)-stimulated p38 phosphorylation. Ang-(1-7) increased cell arachidonic acid release, an effect blocked by A-779. The p38 MAPK antagonist SB202190 completely prevented Ang-(1-7)-stimulated release of arachidonic acid, whereas inhibitors of ERK or JNK had no effect. Ang-(1-7) significantly enhanced DNA synthesis and increased production of transforming growth factor-beta1 (TGF-beta1), fibronectin, and collagen IV. Both A-779 and SB202190 blocked the Ang-(1-7)-stimulated increases in TGF-beta1, fibronectin, and collagen IV. These data indicate that Ang-(1-7) activates MAPK phosphorylation via binding to a specific receptor in human mesangial cells. Stimulation of p38 MAPK phosphorylation by Ang-(1-7) leads to release of arachidonic acid and production of TGF-beta1 and extracellular matrix proteins. We conclude that Ang-(1-7) exerts growth-stimulatory effects in human mesangial cells.
Collapse
Affiliation(s)
- Joseph Zimpelmann
- Div. of Nephrology, Dept. of Medicine, Kidney Research Centre, Ottawa Health Research Institute, Univ. of Ottawa, 1967 Riverside Dr., Rm. 535, Ottawa, Ontario, Canada K1H7W9
| | | |
Collapse
|
28
|
Iusuf D, Henning RH, van Gilst WH, Roks AJ. Angiotensin-(1–7): Pharmacological properties and pharmacotherapeutic perspectives. Eur J Pharmacol 2008; 585:303-12. [DOI: 10.1016/j.ejphar.2008.02.090] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Revised: 01/23/2008] [Accepted: 02/06/2008] [Indexed: 11/30/2022]
|
29
|
Höcht C, Gironacci MM, Mayer MA, Schuman M, Bertera FM, Taira CA. Involvement of angiotensin-(1-7) in the hypothalamic hypotensive effect of captopril in sinoaortic denervated rats. ACTA ACUST UNITED AC 2007; 146:58-66. [PMID: 17850902 DOI: 10.1016/j.regpep.2007.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Revised: 07/17/2007] [Accepted: 08/03/2007] [Indexed: 11/18/2022]
Abstract
The role of anterior hypothalamic angiotensin-(1-7) (Ang-(1-7)) on blood pressure regulation was studied in sinoaortic denervated (SAD) rats. Since angiotensin-converting enzyme inhibitors increase endogenous levels of Ang-(1-7), we addressed the involvement of Ang-(1-7) in the hypotensive effect induced by captopril in SAD rats. Wistar rats 7 days after SAD or sham operation (SO) were anaesthetized and the carotid artery was cannulated for monitoring mean arterial pressure (MAP). A needle was inserted into the anterior hypothalamus for drug administration. Intrahypothalamic administration of Ang-(1-7) (5 pmol) was without effect in SO rats but reduced MAP in SAD rats by 15.5+/-3.2 mm Hg and this effect was blocked by 250 pmol [D-Ala(7)]-Ang-(1-7), a Mas receptor antagonist. Angiotensin II (Ang II) induced an increase in MAP in both groups being the effect greater in SAD rats (DeltaMAP=15.8+/-1.4 mm Hg) than in SO rats (DeltaMAP=9.6+/-1.0 mm Hg). Ang-(1-7) partially abolished the pressor response caused by Ang II in SAD rats. Whilst the captopril intrahypothalamic injection did not affect MAP in SO animals, it significantly reduced MAP in SAD rats (DeltaMAP=-13.3+/-1.9 mm Hg). Either [D-Ala(7)]-Ang-(1-7) or an anti-Ang-(1-7) polyclonal antibody partially blocked the MAP reduction caused by captopril. In conclusion, whilst Ang-(1-7) does not contribute to hypothalamic blood pressure regulation in SO normotensive animals, in SAD rats the heptapeptide induces a reduction of blood pressure mediated by Mas receptor activation. Although Ang-(1-7) is not formed in enough amount in the AHA of SAD animals to exert cardiovascular effects in normal conditions, our results suggest that enhancement of hypothalamic Ang-(1-7) levels by administration of captopril is partially involved in the hypotensive effect of the ACE inhibitor.
Collapse
Affiliation(s)
- Christian Höcht
- Department of Pharmacology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, Junín 956, (C1113AAD) Buenos Aires, Argentina.
| | | | | | | | | | | |
Collapse
|
30
|
Giani JF, Gironacci MM, Muñoz MC, Peña C, Turyn D, Dominici FP. Angiotensin-(1–7) stimulates the phosphorylation of JAK2, IRS-1 and Akt in rat heart in vivo: role of the AT1 and Mas receptors. Am J Physiol Heart Circ Physiol 2007; 293:H1154-63. [PMID: 17496209 DOI: 10.1152/ajpheart.01395.2006] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Angiotensin (ANG) II exerts a negative modulation on insulin signal transduction that might be involved in the pathogenesis of hypertension and insulin resistance. ANG-(1–7), an endogenous heptapeptide hormone formed by cleavage of ANG I and ANG II, counteracts many actions of ANG II. In the current study, we have explored the role of ANG-(1–7) in the signaling crosstalk that exists between ANG II and insulin. We demonstrated that ANG-(1–7) stimulates the phosphorylation of Janus kinase 2 (JAK2) and insulin receptor substrate (IRS)-1 in rat heart in vivo. This stimulating effect was blocked by administration of the selective ANG type 1 (AT1) receptor blocker losartan. In contrast to ANG II, ANG-(1–7) stimulated cardiac Akt phosphorylation, and this stimulation was blunted in presence of the receptor Mas antagonist A-779 or the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin. The specific JAK2 inhibitor AG-490 blocked ANG-(1–7)-induced JAK2 and IRS-1 phosphorylation but had no effect on ANG-(1–7)-induced phosphorylation of Akt, indicating that activation of cardiac Akt by ANG-(1–7) appears not to involve the recruitment of JAK2 but proceeds through the receptor Mas and involves PI3K. Acute in vivo insulin-induced cardiac Akt phosphorylation was inhibited by ANG II. Interestingly, coadministration of insulin with an equimolar mixture of ANG II and ANG-(1–7) reverted this inhibitory effect. On the basis of our present results, we postulate that ANG-(1–7) could be a positive physiological contributor to the actions of insulin in heart and that the balance between ANG II and ANG-(1–7) could be relevant for the association among insulin resistance, hypertension, and cardiovascular disease.
Collapse
Affiliation(s)
- Jorge F Giani
- Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Tecnológicas, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
31
|
Pereyra-Alfonso S, Rodríguez de Lores Arnaiz G, Peña C. Phosphoinositide hydrolysis increase by angiotensin-(1–7) in neonatal rat brain. ACTA ACUST UNITED AC 2007; 140:162-7. [PMID: 17218025 DOI: 10.1016/j.regpep.2006.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 11/24/2006] [Accepted: 12/01/2006] [Indexed: 11/23/2022]
Abstract
Angiotensin (Ang)-(1-7) is an endogenous peptide hormone of the renin-angiotensin system which exerts diverse biological actions, some of them counterregulate Ang II effects. In the present study potential effect of Ang-(1-7) on phosphoinositide (PI) turnover was evaluated in neonatal rat brain. Cerebral cortex prisms of seven-day-old rats were preloaded with [(3)H]myoinositol, incubated with additions during 30 min and later [(3)H]inositol-phosphates (IPs) accumulation quantified. It was observed that PI hydrolysis enhanced 30% to 60% in the presence of 0.01 nM to 100 nM Ang-(1-7). Neither 10 nM [D-Ala(7)]Ang-(1-7), an Ang-(1-7) specific antagonist, nor 10 nM losartan, an angiotensin II type 1 (AT(1)) receptor antagonist, blocked the effect of 0.1 nM Ang-(1-7) on PI metabolism. The effect of 0.1 nM Ang-(1-7) on PI hydrolysis was not reduced but it was even significantly increased in the simultaneous presence of [D-Ala(7)]Ang-(1-7) or losartan. PI turnover enhancement achieved with 0.1 nM Ang-(1-7) decreased roughly 30% in the presence of 10 nM PD 123319, an angiotensin II type 2 (AT(2)) receptor antagonist. The antagonists alone also enhanced PI turnover. Present findings showing an increase in PI turnover by Ang-(1-7) represent a novel action for this peptide and suggest that it exerts a function in this signaling system in neonatal rat brain, an effect involving, at least partially, angiotensin AT(2) receptors.
Collapse
Affiliation(s)
- Susana Pereyra-Alfonso
- Instituto de Biología Celular y Neurociencias Prof. E. De Robertis, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, (1121) Buenos Aires, Argentina
| | | | | |
Collapse
|
32
|
Dharmani M, Mustafa MR, Achike FI, Sim MK. Effects of angiotensin 1-7 on the actions of angiotensin II in the renal and mesenteric vasculature of hypertensive and streptozotocin-induced diabetic rats. Eur J Pharmacol 2007; 561:144-50. [PMID: 17320855 DOI: 10.1016/j.ejphar.2007.01.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Revised: 01/09/2007] [Accepted: 01/12/2007] [Indexed: 11/17/2022]
Abstract
Angiotensin 1-7, a heptapeptide derived from metabolism of either angiotensin I or angiotensin II, is a biologically active peptide of the renin-angiotensin system. The present study investigated the effect of angiotensin 1-7 on the vasopressor action of angiotensin II in the renal and mesenteric vasculature of Wistar-Kyoto (WKY) rats, spontaneously hypertensive rats (SHR) and streptozotocin-induced diabetic rats. Angiotensin II-induced dose-dependent vasoconstrictions in the renal vasculature. The pressor response was enhanced in the SHR and reduced in the streptozotocin-diabetic rat compared to WKY rats. Angiotensin 1-7 attenuated the angiotensin II pressor responses in the renal vasculature of WKY and SHR rats. However, the ability to reduce angiotensin II response was diminished in diabetic-induced rat kidneys. The effect of angiotensin 1-7 was not inhibited by 1-[(4-(Dimethylamino)-3-methylphenyl] methyl]-5-(diphenylacetyl)-4,5,6,7-tetrahydro-1H-imidazo[4,5-c]pyridine-6-carboxylic acid ditrifluoroacetate (PD123319), an angiotensin AT(2) receptor antagonist. (D-ALA(7))-Angiotensin I/II (1-7) (D-ALA) (an angiotensin 1-7 receptor antagonist), indomethacin (a cyclo-oxygenase inhibitor), and N(omega)-Nitro-L-Arginine Methyl Ester (L-NAME)(a nitric oxide synthetase inhibitor) abolished the attenuation by angiotensin 1-7 in both WKY rats and SHR, indicating that its action is mediated by angiotensin 1-7 receptor that is either coupled to the release of prostaglandins and/or nitric oxide. The vasopressor responses to angiotensin II in mesenteric vasculature bed was also dose-dependent but smaller in magnitude compared to the renal vasculature. The responses to angiotensin II were relatively smaller in SHR but no significant difference was observed between WKY and streptozotocin-induced diabetic rats. Angiotensin 1-7 attenuated the angiotensin II pressor responses in WKY, SHR and diabetic-induced mesenteric bed. The attenuation was observed at the lower concentrations of angiotensin II in WKY and diabetic-induced rats but at higher concentrations in SHR. Similar observation as in the renal vasculature was seen with PD123319, D-ALA, and L-NAME. Indomethacin reversed the attenuation by angiotensin 1-7 only in the SHR mesenteric vascular bed. The present findings support the regulatory role of angiotensin 1-7 in the renal and mesenteric vasculature, which is differentially altered in hypertension and diabetes.
Collapse
Affiliation(s)
- Murugan Dharmani
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | | | | |
Collapse
|
33
|
Höcht C, Opezzo JAW, Gironacci MM, Peña C, Taira CA. Hypothalamic cardiovascular effects of angiotensin-(1–7) in spontaneously hypertensive rats. ACTA ACUST UNITED AC 2006; 135:39-44. [PMID: 16678284 DOI: 10.1016/j.regpep.2006.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Revised: 03/20/2006] [Accepted: 03/24/2006] [Indexed: 10/24/2022]
Abstract
The objective of the present work was to study the cardiovascular actions of the intrahypothalamic injection of Ang-(1-7) and its effects on the pressor response to Ang II in spontaneously hypertensive (SH) rats and Wistar Kyoto (WKY) animals. In anaesthetized SH and WKY rats, a carotid artery was cannulated for mean arterial pressure (MAP) measurement and a stainless-steel needle was inserted into the anterior hypothalamus for drug administration. The cardiovascular effects of the intrahypothalamic administration of Ang-(1-7) were determined in SH and WKY rats. In SH rats, the effect of irbesartan and D-Ala-Ang-(1-7) on Ang-(1-7) cardiovascular effect was also evaluated. Ang II was administered in the hypothalamus of SH and WKY rats and changes in blood pressure and heart rate were measured followed by the administration of Ang II, Ang II+Ang-(1-7) or Ang II+D-Ala-Ang-(1-7). Ang-(1-7) did not the change basal MAP in WKY rats, but induced a pressor response in SH animals. Whilst the co-administration of D-Ala-Ang-(1-7) did not affect the response to Ang-(1-7), the previous administration of irbesartan prevented the effect of the peptide. The intrahypothalamic injection of Ang II induced a significantly greater pressor response in SH animals compared to normotensive rats. The co-administration of Ang-(1-7) with Ang II did not affect the pressor response to Ang II in the WKY group. In SH rats, whilst the co-administration of Ang-(1-7) with Ang II reduced the pressor response to Ang II, the concomitant application of D-Ala-Ang-(1-7) with Ang II increased the pressor response to the octapeptide after 5 and 10 min of intrahypothalamic administration. In conclusion, our result demonstrated that the biologically active peptide Ang-(1-7) did not participate in the hypothalamic blood pressure regulation of WKY animals. In SH rats, Ang-(1-7) exerted pleiotropic effects on blood pressure regulation. High dose of the heptapeptide produced a pressor response because of an unspecific action by activation of AT1 receptors. The concomitant administration of lower doses of Ang-(1-7) with Ang II reduced the pressor response to the octapeptide. Finally, the effect of AT(1-7) antagonist on Ang II pressor response suggested that hypothalamic formed Ang-(1-7) are implicated in the regulation of the cardiovascular effects of Ang II.
Collapse
Affiliation(s)
- Christian Höcht
- Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, (C1113AAD) Buenos Aires, Argentina.
| | | | | | | | | |
Collapse
|
34
|
Gurzu B, Costuleanu M, Slatineanu SM, Ciobanu A, Petrescu G. Are multiple angiotensin receptor types involved in angiotensin (1-7) actions on isolated rat portal vein. J Renin Angiotensin Aldosterone Syst 2006; 6:90-5. [PMID: 16470488 DOI: 10.3317/jraas.2005.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Angiotensin (1-7) [Ang (1-7)] is a bioactive component of the renin angiotensin system. Ang (1-7) may interact with angiotensin type 1 (AT1) or type 2 (AT2) receptors and with Ang (1-7) - specific receptors. We examined the interactions between different doses of Ang (1-7) (1 nM-1 microM) and angiotensin II (Ang II) (10 and 100 nM) on isolated rat portal vein. In endothelium-denuded portal vein rings, Ang (1-7) inhibited contractile effects induced by Ang II. The effects of Ang (1-7) were modified by indomethacin, N(G)-nitro-L-arginine methyl ester (L-NAME), (D-Ala7)-Angiotensin (1-7) (H-2888) and losartan. Our results suggest that on rat isolated portal vein rings without endothelium, Ang (1-7) reduces Ang II-induced contractions by acting mostly on Ang (1-7) specific receptors, and this effect is mediated by vasodilatatory prostaglandins. At high concentrations, Ang (1-7) effects are mediated by AT1-receptors, though to a lesser extent than by Ang (1-7) specific receptors.
Collapse
Affiliation(s)
- Bogdan Gurzu
- Department of Physiology, Faculty of Dentistry, University of Medicine and Pharmacy "Gr. T. Popa", Iasi, RO-700115, Romania
| | | | | | | | | |
Collapse
|
35
|
Grobe JL, Katovich MJ. Alterations in aortic vascular reactivity to angiotensin 1–7 in 17-β-estradiol-treated female SD rats. ACTA ACUST UNITED AC 2006; 133:62-7. [PMID: 16219374 DOI: 10.1016/j.regpep.2005.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2005] [Revised: 09/09/2005] [Accepted: 09/12/2005] [Indexed: 12/01/2022]
Abstract
Estrogen's suggested cardio-protective effects have come into question following the results of recent clinical trials. Two major components of the renin-angiotensin system (RAS) that are modulated by estrogen are angiotensin converting enzyme, and the angiotensin II type 1 receptor. Further research has revealed several new components of the RAS, including angiotensin converting enzyme 2, its peptide product angiotensin 1-7 (Ang 1-7), and that peptide's receptor, Mas. These components appear to oppose the classical effects of the RAS, and may act to buffer the RAS in vivo. Recent work has shown that during pregnancy, when estradiol levels are elevated, renal and urinary Ang 1-7 are greatly increased. This study examined the effects of estradiol on the efficacy of Ang 1-7 in the rat aorta. Female Sprague-Dawley rats were ovariectomized and a subgroup was chronically treated with subcutaneous pellets of estradiol for 3 weeks. Thoracic aortas were harvested for assessment of in vitro vascular reactivity to Ang 1-7. The results demonstrated that increased estradiol exposure attenuated the relaxation response to Ang 1-7 in a dose-dependent manner. These findings are in contrast to recent work showing potentiated responses to Ang 1-7 in mesenteric arteries from estrogen-manipulated rats, and may suggest a regional specificity in estradiol-mediated changes in the RAS.
Collapse
Affiliation(s)
- Justin L Grobe
- Department of Pharmacodynamics, University of Florida College of Pharmacy, JHMHSC, PO Box 100487, Gainesville, FL 32610, USA
| | | |
Collapse
|
36
|
Oudot A, Vergely C, Ecarnot-Laubriet A, Rochette L. Pharmacological concentration of angiotensin-(1-7) activates NADPH oxidase after ischemia-reperfusion in rat heart through AT1 receptor stimulation. ACTA ACUST UNITED AC 2005; 127:101-10. [PMID: 15680476 DOI: 10.1016/j.regpep.2004.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2004] [Revised: 10/20/2004] [Accepted: 10/27/2004] [Indexed: 11/15/2022]
Abstract
The cardiovascular role of angiotensin-(1-7), especially in the functional and metabolic alterations associated with ischemia-reperfusion (IR), is still not clearly defined. Our objective was to evaluate the cardiac effects of angiotensin-(1-7), the receptors involved, and their relationships with NADPH oxidase activation under non-ischemic conditions and, during an ischemia-reperfusion sequence. Isolated perfused rat hearts underwent 45 min of non-ischemic perfusion, or 30 min of global ischemia followed by 30 min of reperfusion. Angiotensin-(1-7) and/or AT1 receptor blocker losartan or angiotensin-(1-7) receptor antagonist (D-Ala7)-angiotensin-(1-7) were perfused. Our results showed that angiotensin-(1-7) was without effect at low concentrations (10(-10) to 10(-7) M). At a pharmacological concentration, 0.5 microM angiotensin-(1-7) induced vasoconstriction, which was antagonised by losartan. After ischemia, we noted a partial recovery of functional parameters, which was not modified by any of the treatments. The expression of AT1 receptor mRNA was increased by ischemia-reperfusion, except in (D-Ala7)-angiotensin-(1-7) treated hearts. Angiotensin-(1-7) further increased the AT1 expression. NADPH oxidase activity was enhanced in 0.5 microM angiotensin-(1-7)-treated hearts subjected to ischemia-reperfusion, this effect was totally reversed by losartan. This is the first time that it has been shown that, in the heart, angiotensin-(1-7) at pharmacological concentration activates NADPH oxidase, an enzyme thought to be involved in several angiotensin II effects.
Collapse
Affiliation(s)
- Alexandra Oudot
- Laboratoire de Physiopathologie et Pharmacologie, Cardio-vasculaires Expérimentales, IFR no. 100, Facultés de Médecine et Pharmacie, 7, Boulevard Jeanne d'Arc-BP 87900, 21079 Dijon, France.
| | | | | | | |
Collapse
|
37
|
Matos de Moura M, Augusto Sousa dos Santos R, Antônio Peliky Fontes M. Evidence for a functional cardiac interaction between losartan and angiotensin-(1-7) receptors revealed by orthostatic tilting test in rats. Br J Pharmacol 2005; 144:755-60. [PMID: 15685215 PMCID: PMC1576057 DOI: 10.1038/sj.bjp.0706039] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. Studies have shown that the angiotensin II (Ang II) AT1 receptor antagonist, losartan, accentuates the orthostatic hypotensive response in anesthetized rats, and there is evidence indicating that this effect is not exclusively mediated by AT1 receptors. 2. We investigated whether the pronounced orthostatic cardiovascular response observed in losartan-treated rats involves an interference with angiotensin-(1-7) (Ang-(1-7)) receptors. 3. Urethane-anesthetized rats were submitted to orthostatic stress (90 degree head-up tilt for 2 min). Intravenous injection of losartan (1 mg kg(-1), n=9) significantly accentuated the decrease in mean arterial pressure (MAP) induced by head-up tilt (-33+/-6% after losartan vs -15+/-8% control tilt). This effect was accompanied by a significant bradycardia (-8+/-3% after losartan vs -3+/-3% control tilt). Another AT1 antagonist, candesartan, did not potentiate the decrease of MAP and did not change the cardiac response induced by head-up tilt. Strikingly, administration of the Ang-(1-7) antagonist, A-779 (10 nmol kg(-1), n=5), totally reversed the bradycardic effect caused by losartan and this effect was accompanied by a tendency towards attenuation of the hypotensive response caused by losartan. 4. These findings indicate that the marked orthostatic cardiovascular response is specific for losartan, and that it may be due, in part, to an interaction of this antagonist with Ang-(1-7) receptors, probably at the cardiac level.
Collapse
Affiliation(s)
- Marina Matos de Moura
- Department of Physiology and Biophysics, ICB, Universidade Federal de Minas Gerais, Av. Antonio Carlos 6627, Pampulha, Belo Horizonte, MG 31270 901, Brazil
| | - Robson Augusto Sousa dos Santos
- Department of Physiology and Biophysics, ICB, Universidade Federal de Minas Gerais, Av. Antonio Carlos 6627, Pampulha, Belo Horizonte, MG 31270 901, Brazil
| | - Marco Antônio Peliky Fontes
- Department of Physiology and Biophysics, ICB, Universidade Federal de Minas Gerais, Av. Antonio Carlos 6627, Pampulha, Belo Horizonte, MG 31270 901, Brazil
- Author for correspondence:
| |
Collapse
|
38
|
Bürgelová M, Kramer HJ, Teplan V, Thumová M, Cervenka L. Effects of angiotensin-(1–7) blockade on renal function in rats with enhanced intrarenal Ang II activity. Kidney Int 2005; 67:1453-61. [PMID: 15780097 DOI: 10.1111/j.1523-1755.2005.00222.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Increasing evidence suggests that angiotensin-(1-7) [Ang-(1-7)] acts as an endogenous antagonist of Ang II when the renin-angiotensin system (RAS) is activated. In the present study, we therefore compared the effects of acute intrarenal (i.r.) Ang-(1-7) receptor blockade on renal function under conditions of normal and increased intrarenal Ang II concentration. METHODS Salt-replete Hannover-Sprague Dawley rats (HanSD) served as control animals. As models with enhanced action of Ang II we first used transgenic rats harboring the Ren-2 renin gene (TGR), second, Ang II-infused rats, third, 2-kidney, 1-clip (2K1C) hypertensive rats on normal salt intake, and fourth, salt-depleted TGR and HanSD. RESULTS I.r. Ang-(1-7) receptor blockade elicited significant decreases in glomerular filtration rate (GFR), renal plasma flow (RPF), and sodium excretion in 2K1C rats, and in salt-depleted TGR and HanSD. In contrast, i.r. Ang-(1-7) receptor blockade did not significantly change GFR, RPF, and sodium excretion in salt-replete TGR and HanSD, or in Ang II-infused rats. CONCLUSION These findings suggest that under conditions of normal intrarenal RAS activity and increased intrarenal Ang II action by infusion of Ang II or by insertion of a renin gene in salt-replete conditions, Ang-(1-7) is not an important factor in the regulation of renal function. In contrast, under conditions of endogenous RAS activation due to clipping of the renal artery or to sodium restriction, Ang-(1-7) serves as opponent of the vasoconstrictor actions of Ang II.
Collapse
Affiliation(s)
- Marcela Bürgelová
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
39
|
Loot AE, van Buiten A, Roks AJM, Henning RH. The suitability of iodinated Angiotensin-(1–7) peptides as pharmacological tools. J Pharmacol Toxicol Methods 2005; 51:51-5. [PMID: 15596114 DOI: 10.1016/j.vascn.2004.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2004] [Accepted: 07/07/2004] [Indexed: 11/15/2022]
Abstract
INTRODUCTION The heptapeptide Angiotensin-(1-7) [(Ang-(1-7)] is a biologically active component of the Renin-Angiotensin System. Pharmacological studies often involve Ang-(1-7) radioactively labelled with (125)I. Given the small size of the original peptide, we investigated whether introduction of a rather bulky iodine label interferes with the biological activity of Ang-(1-7). METHODS Ang-(1-7) was labelled with nonradioactive iodine with the chloramine-T method. The reaction products were separated on HPLC and analysed with mass spectrometry. The products were tested for biological activity in two ways: The ability of labelled Ang-(1-7) to block Ang II-induced contraction in rat aortic rings was tested in an organ bath setup. The affinity of labelled angiotensin for ACE in rat plasma was examined in vitro. RESULTS Iodination of Angiotensin-(1-7) resulted in two main products: monoiodinated and diiodinated Ang-(1-7) that could be easily separated on HPLC. In an organ bath experiment, monoiodinated Ang-(1-7) blocked Ang II responses identical to the native compound, whereas diiodinated Ang-(1-7) had lost its ability to block Ang II responses. Likewise, monoiodinated Ang-(1-7) had retained its affinity for ACE, while the affinity of diiodinated Ang-(1-7) was greatly reduced. DISCUSSION Monoiodinated Ang-(1-7) has a biological activity identical to the native compound, whereas this is lost in diiodinated Ang-(1-7). Therefore, only the monoiodinated radioactive form seems suited for pharmacological studies.
Collapse
Affiliation(s)
- Annemarieke E Loot
- Department of Clinical Pharmacology, Groningen University Institute for Drug Evaluation (GUIDE), University of Groningen, PO Box 196, Groningen 9700 AD, The Netherlands.
| | | | | | | |
Collapse
|
40
|
Sampaio WO, Nascimento AAS, Santos RAS. Systemic and regional hemodynamic effects of angiotensin-(1-7) in rats. Am J Physiol Heart Circ Physiol 2003; 284:H1985-94. [PMID: 12573992 DOI: 10.1152/ajpheart.01145.2002] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The systemic and regional hemodynamics effects of ANG-(1-7) were examined in urethane-anesthetized rats. The blood flow distribution (kidneys, skin, mesentery, lungs, spleen, brain, muscle, and adrenals), cardiac output, and total peripheral resistance were investigated by using fluorescent microspheres. Blood pressure and heart rate were recorded from the brachial artery. ANG-(1-7) infusion (110 fmol x min(-1) x 10 min(-1) iv) significantly increased blood flow to the kidney (5.10 +/- 1.07 to 8.30 +/- 0.97 ml x min(-1) x g(-1)), mesentery (0.73 +/- 0.16 to 1.17 +/- 0.49 ml x min(-1) x g(-1)), brain (1.32 +/- 0.44 to 2.18 +/- 0.85 ml x min(-1) x g(-1)), and skin (0.07 +/- 0.02 to 0.18 +/- 0.07 ml x min(-1) x g(-1)) and the vascular conductance in these organs. ANG-(1-7) also produced a significant increase in cardiac index (30%) and a decrease in total peripheral resistance (2.90 +/- 0.55 to 2.15 +/- 0.28 mmHg x ml(-1) x min x 100 g). Blood flow to the spleen, muscle, lungs, and adrenals, as well as the blood pressure and heart rate, were not altered by the ANG-(1-7) infusion. The selective ANG-(1-7) antagonist A-779 reduced the blood flow in renal, cerebral, mesenteric, and cutaneous beds and blocked the ANG-(1-7)-induced vasodilatation in the kidney, mesentery, and skin, suggesting a significant role of endogenous ANG-(1-7) in these territories. The effects of ANG-(1-7) on the cerebral blood flow, cardiac index, systolic volume, and total peripheral resistance were partially attenuated by A-779. A high dose of ANG-(1-7) (11 pmol x min(-1) x 10 min(-1)) caused an opposite effect of that produced by the low dose. Our results show for the first time that ANG-(1-7) has a previously unsuspected potent effect in the blood flow distribution and systemic hemodynamics.
Collapse
Affiliation(s)
- Walkyria O Sampaio
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Brazil
| | | | | |
Collapse
|
41
|
Bürgelová M, Kramer HJ, Teplan V, Velicková G, Vítko S, Heller J, Malý J, Cervenka L. Intrarenal infusion of angiotensin-(1-7) modulates renal functional responses to exogenous angiotensin II in the rat. Kidney Blood Press Res 2003; 25:202-10. [PMID: 12424421 DOI: 10.1159/000066340] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In the present study we investigated the possible role of angiotensin-(1-7) [Ang-(1-7)] in modulating renal functional responses to intrarenal (i.e.) infusion of angiotensin II (ANG II) in normotensive anesthetized rats. ANG II (6 ng/min, n = 14) decreased glomerular filtration rate (GFR), renal plasma flow (RPF), absolute and fractional sodium excretion by -24 +/- 5, -25 +/- 6, -44 +/- 6 and -28 +/- 7%, respectively (p < 0.05). i.r. infusion of Ang-(1-7) (50 ng/min, n = 13) did not significantly alter GFR (+6 +/- 4%) but reduced RPF by -19 +/- 7% (p < 0.05). Ang-(1-7) increased absolute and fractional sodium excretion by +36 +/- 6 and +37 +/- 8%, respectively (p < 0.05). Infusion of Ang-(1-7) did not prevent the decreases in GFR and RPF but completely blunted the decreases in absolute (-2 +/- 2%) and fractional sodium excretion (-4 +/- 4%) induced by ANG II (n = 11). Blockade of the Ang-(1-7) receptor by [7-D-Ala]-Ang-(1-7) (5 microg/min, n = 11) significantly decreased GFR, RPF, absolute and fractional sodium excretion by -28 +/- 7, -20 +/- 5, -32 +/- 7 and -24 +/- 4%, respectively (p < 0.05), suggesting that the action of endogenous ANG II is unopposed by compensatory effect of endogenous Ang-(1-7). i.r. infusion of Ang-(1-7) (n = 10) did not alter the effect of Ang-(1-7) receptor blockade on RPF (-21 +/- 6%) but blunted its effects on GFR (+4 +/- 3%) and absolute (+7 +/- 5%) and fractional (+6 +/- 4%) urinary sodium excretion probably by displacing the receptor blocker. While exogenous ANG II during blockade of the Ang-(1-7) receptor and the AT(2) receptor (by PD 123319; 1 microg/min i.r., n = 9) resulted in the same decreases in absolute and fractional sodium excretion (-39 +/- 8 and -38 +/- 6%, respectively, p < 0.05) as did ANG II in the absence of Ang-(1-7) receptor blockade. These results suggest that in normotensive rats high i.r. Ang-(1-7) concentration attenuates the tubular, i.e. sodium reabsorptive effect, but not the vascular effect of exogenous i.r. ANG II. Results obtained during blockade of Ang-(1-7) and of AT(2) receptors imply that AT(2) receptors play a role in tubular sodium reabsorption in the presence of high ANG II concentration.
Collapse
Affiliation(s)
- Marcela Bürgelová
- Transplant Center, Department of Nephrology, Institute for Clinical and Experimental Medicine, 1858/9 Videnská, 140 00 Prague 4, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Vauquelin G, Michotte Y, Smolders I, Sarre S, Ebinger G, Dupont A, Vanderheyden P. Cellular targets for angiotensin II fragments: pharmacological and molecular evidence. J Renin Angiotensin Aldosterone Syst 2002; 3:195-204. [PMID: 12584663 DOI: 10.3317/jraas.2002.041] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Although angiotensin II has long been considered to represent the end product of the renin-angiotensin system (RAS), there is accumulating evidence that it encompasses additional effector peptides with diverse functions. In this respect, angiotensin IV (Ang IV) formed by deletion of the two N terminal amino acids, has sparked great interest because of its wide range of physiological effects. Among those, its facilitatory role in memory acquisition and retrieval is of special therapeutic relevance. High affinity binding sites for this peptide have been denoted as AT(4)- receptors and, very recently, they have been proposed to correspond to the membrane-associated OTase/ IRAP aminopeptidase. This offers new opportunities for examining physiological roles of Ang IV in the fields of cognition, cardiovascular and renal metabolism and pathophysiological conditions like diabetes and hypertension. Still new recognition sites may be unveiled for this and other angiotensin fragments. Recognition sites for Ang-(1-7) (deletion of the C terminal amino acid) are still elusive and some of the actions of angiotensin III (deletion of the N terminal amino acid) in the CNS are hard to explain on the basis of their interaction with AT(1)-receptors only. A more thorough cross-talk between in vitro investigations on native and transfected cell lines and in vivo investigations on healthy, diseased and transgenic animals may prove to be essential to further unravel the molecular basis of the physiological actions of these small endogenous angiotensin fragments.
Collapse
Affiliation(s)
- Georges Vauquelin
- Department of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel (VUB), Sint-Genesius, Rode, B-1640, Belgium
| | | | | | | | | | | | | |
Collapse
|
43
|
Kucharewicz I, Pawlak R, Matys T, Pawlak D, Buczko W. Antithrombotic effect of captopril and losartan is mediated by angiotensin-(1-7). Hypertension 2002; 40:774-9. [PMID: 12411476 DOI: 10.1161/01.hyp.0000035396.27909.40] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is well established that renin-angiotensin system blockers exert NO/prostacyclin-dependent antithrombotic effects. Because some beneficial effects of these drugs are mediated by angiotensin (Ang)-(1-7), in the present study we examined if their antithrombotic action could be mediated by Ang-(1-7). Intravenous infusion of Ang-(1-7) (1, 10, or 100 pmol/kg per minute for 2 hours) into rats developing venous thrombosis caused 50% to 70% reduction of the thrombus weight. This effect was dose-dependently reversed by cotreatment with A-779 (selective Ang-[1-7] receptor antagonist) or EXP 3174 (angiotensin type 1 receptor antagonist) but not by PD 123,319 (angiotensin type 2 receptor antagonist). Similarly, the antithrombotic effects of captopril (ACE inhibitor) and losartan (angiotensin type 1 receptor blocker) were attenuated by A-779 in a dose-dependent manner. The effect of Ang-(1-7) was completely abolished by concomitant administration of NO synthase inhibitor (N(G)-nitro-L-arginine methyl ester) and prostacyclin synthesis inhibitor (indomethacin), as has been shown previously for captopril and losartan. Thus, the antithrombotic effect of renin-angiotensin system blockers involves Ang-(1-7)-evoked release of NO and prostacyclin.
Collapse
Affiliation(s)
- Iwona Kucharewicz
- Department of Pharmacodynamics, Medical Academy of Bialystok, Bialystok, Poland
| | | | | | | | | |
Collapse
|
44
|
Gironacci MM, Fernández-Tomé MDC, Speziale E, Sterin-Speziale N, Peña C. Enhancement of phosphatidylcholine biosynthesis by angiotensin-(1-7) in the rat renal cortex. Biochem Pharmacol 2002; 63:507-14. [PMID: 11853701 DOI: 10.1016/s0006-2952(01)00920-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the present paper, we investigated the effect of angiotensin-(1-7) (Ang-(1-7)) on phospholipid biosynthesis in the rat renal cortex. A significant increase in phosphatidylcholine (PC) labeling was observed when cortical slices, prelabeled with [32P]orthophosphate, were incubated for 30 min in the presence of Ang-(1-7) (1 pM to 100 nM). Neither the phospholipase C inhibitors, neomycin or db-cAMP nor the protein kinase C inhibitors, chelerythrine or H7, modified the stimulatory effect induced by 0.1 nM Ang-(1-7). The enhancement of PC biosynthesis caused by 0.1 nM Ang-(1-7) was unmodified by either losartan, an AT(1) receptor antagonist, or (1-[[4-(dimethylamino)-3-methylphenyl]methyl]-5-(diphenylacetyl)-4,5,6,7-tetrahydro-1H-imidazol[4,5-c]pyridine-6-carboxylic acid ditrifluoroacetate) (PD 123319), an AT(2) receptor antagonist, but was partially blocked by [D-Ala(7)]Ang-(1-7), an Ang-(1-7) specific antagonist. However, losartan potentiated the effect of 100 nM Ang-(1-7) on PC biosynthesis. Losartan by itself increased the de novo synthesis of PC. These results suggest that the Ang-(1-7)-mediated increase in PC biosynthesis is independent of AT(1) and AT(2) receptor activation but mediated by a specific Ang-(1-7) receptor. This mechanism is independent of phospholipase C and PKC activation.
Collapse
Affiliation(s)
- Mariela M Gironacci
- Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
45
|
Souza Dos Santos RA, Passaglio KT, Pesquero JB, Bader M, Simões E Silva AC. Interactions between angiotensin-(1-7), kinins, and angiotensin II in kidney and blood vessels. Hypertension 2001; 38:660-4. [PMID: 11566951 DOI: 10.1161/01.hyp.38.3.660] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The heptapeptide angiotensin (Ang)-(1-7) is currently considered one of the biologically active end products of the renin-angiotensin system. The formation of Ang-(1-7) by pathways independent of Ang II generation, the selectivity of its actions, and its peculiar property of exhibiting effects that are partially opposite of those of the parent compound, Ang II, confer a unique biochemical and functional profile to this peptide. In this article, we will review novel aspects of the biological actions of Ang-(1-7), dealing with its interaction with Ang II and kinins, especially in the kidney and blood vessels.
Collapse
Affiliation(s)
- R A Souza Dos Santos
- Laboratório de Hipertensão, Instituto de Ciências Biológicas, Departamento de Pediatria, Fac. Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | | | | | | | | |
Collapse
|
46
|
Caruso-Neves C, Rangel LB, Lara LS, Lopes AG. Regulation of the renal proximal tubule second sodium pump by angiotensins. Braz J Med Biol Res 2001; 34:1079-84. [PMID: 11471048 DOI: 10.1590/s0100-879x2001000800015] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
For several years it was believed that angiotensin II (Ang II) alone mediated the effects of the renin-angiotensin system. However, it has been observed that other peptides of this system, such as angiotensin-(1-7) (Ang-(1-7)), present biological activity. The effect of Ang II and Ang-(1-7) on renal sodium excretion has been associated, at least in part, with modulation of proximal tubule sodium reabsorption. In the present review, we discuss the evidence for the involvement of Na+-ATPase, called the second sodium pump, as a target for the actions of these compounds in the regulation of proximal tubule sodium reabsorption.
Collapse
Affiliation(s)
- C Caruso-Neves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21949-900 Rio de Janeiro RJ, Brasil
| | | | | | | |
Collapse
|
47
|
Chansel D, Vandermeersch S, Oko A, Curat C, Ardaillou R. Effects of angiotensin IV and angiotensin-(1-7) on basal and angiotensin II-stimulated cytosolic Ca2+ in mesangial cells. Eur J Pharmacol 2001; 414:165-75. [PMID: 11239916 DOI: 10.1016/s0014-2999(01)00791-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This study analyzed the influence of two main metabolites of angiotensin II, angiotensin IV and angiotensin-(1-7), on basal and angiotensin II-dependent [Ca2+](i) in rat mesangial cells. Angiotensin IV behaved as a weak agonist. Its effects were abolished by angiotensin AT(1) receptor antagonists. Treatment with angiotensin II abolished the effect of a subsequent treatment with angiotensin IV whereas two successive angiotensin IV-dependent [Ca2+](i) peaks were obtained. Angiotensin II increased [Ca2+](i) in a Ca2+-free medium whereas angiotensin IV was inactive. Leucine-valine-valine-hemorphin 7, a hemorphin specific for the angiotensin AT(4) receptor, was devoid of any agonistic or antagonistic effect. In contrast, angiotensin-(1-7), if without influence on basal [Ca2+](i), inhibited angiotensin II- and angiotensin IV-dependent [Ca2+](i) increases. Total inhibition of the angiotensin IV effect was obtained whereas association of angiotensin-(1-7) to 8-(NN-diethylamino)-octyl-3,4,5-trimethoxybenzoate, an inhibitor of inositol phosphate-mediated Ca2+ release, was necessary to suppress the effect of angiotensin II. These results provide evidence that angiotensin II metabolites may participate in the control of [Ca2+](i) in mesangial cells at the initial stage of binding to the angiotensin AT(1) receptors.
Collapse
Affiliation(s)
- D Chansel
- INSERM U. 489, Hôpital Tenon, 4 rue de la Chine, 75020 Paris, France
| | | | | | | | | |
Collapse
|
48
|
Abstract
The renin-angiotensin system is a major physiological regulator of arterial pressure and hydro-electrolyte balance. Evidence has now been accumulated that in addition to angiotensin (Ang) II other Ang peptides [Ang III, Ang IV and Ang-(1-7)], formed in the limited proteolysis processing of angiotensinogen, are importantly involved in mediating several actions of the RAS. In this article we will review our knowledge of the biological actions of Ang-(1-7) with focus on the puzzling aspects of the mediation of its effects and the interaction Ang-(1-7)-kinins. In addition, we will attempt to summarize the evidence that Ang-(1-7) takes an important part of the mechanisms aimed to counteract the vasoconstrictor and proliferative effects of Ang II.
Collapse
Affiliation(s)
- R A Santos
- Departamento de Fisiologia e Biofísica, Av. Antonio Carlos, 6627 - Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG Brazil.
| | | | | |
Collapse
|