1
|
Faraco G. Dietary salt, vascular dysfunction, and cognitive impairment. Cardiovasc Res 2025; 120:2349-2359. [PMID: 39429024 PMCID: PMC11976728 DOI: 10.1093/cvr/cvae229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/24/2024] [Accepted: 09/15/2024] [Indexed: 10/22/2024] Open
Abstract
Excessive salt consumption is a major health problem worldwide leading to serious cardiovascular events including hypertension, heart disease, and stroke. Additionally, high-salt diet has been increasingly associated with cognitive impairment in animal models and late-life dementia in humans. High-salt consumption is harmful for the cerebral vasculature, disrupts blood supply to the brain, and could contribute to Alzheimer's disease pathology. Although animal models have advanced our understanding of the cellular and molecular mechanisms, additional studies are needed to further elucidate the effects of salt on brain function. Furthermore, the association between excessive salt intake and cognitive impairment will have to be more thoroughly investigated in humans. Since the harmful effects of salt on the brain are independent by its effect on blood pressure, in this review, I will specifically discuss the evidence, available in experimental models and humans, on the effects of salt on vascular and cognitive function in the absence of changes in blood pressure. Given the strong effects of salt on the function of immune cells, I will also discuss the evidence linking salt consumption to gut immunity dysregulation with particular attention to the ability of salt to disrupt T helper 17 (Th17) cell homeostasis. Lastly, I will briefly discuss the data implicating IL-17A, the major cytokine produced by Th17 cells, in vascular dysfunction and cognitive impairment.
Collapse
Affiliation(s)
- Giuseppe Faraco
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| |
Collapse
|
2
|
Meier M, Becker S, Levine E, DuFresne O, Foster K, Moore J, Burnett FN, Hermanns VC, Heath SP, Abdelsaid M, Coucha M. Timing matters in the use of renin-angiotensin system modulators and COVID-related cognitive and cerebrovascular dysfunction. PLoS One 2024; 19:e0304135. [PMID: 39074114 DOI: 10.1371/journal.pone.0304135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 05/07/2024] [Indexed: 07/31/2024] Open
Abstract
Renin-angiotensin system (RAS) modulators, including Angiotensin receptor blockers (ARB) and angiotensin-converting enzyme inhibitors (ACEI), are effective medications for controlling blood pressure. Cognitive deficits, including lack of concentration, memory loss, and confusion, were reported after COVID-19 infection. ARBs or ACEI increase the expression of angiotensin-converting enzyme-2 (ACE-2), a functional receptor that allows binding of SARS-CoV-2 spike protein for cellular invasion. To date, the association between the use of RAS modulators and the severity of COVID-19 cognitive dysfunction is still controversial. PURPOSE This study addressed the following questions: 1) Does prior treatment with RAS modulator worsen COVID-19-induced cerebrovascular and cognitive dysfunction? 2) Can post-treatment with RAS modulator improve cognitive performance and cerebrovascular function following COVID-19? We hypothesize that pre-treatment exacerbates COVID-19-induced detrimental effects while post-treatment displays protective effects. METHODS Clinical study: Patients diagnosed with COVID-19 between May 2020 and December 2022 were identified through the electronic medical record system. Inclusion criteria comprised a documented medical history of hypertension treated with at least one antihypertensive medication. Subsequently, patients were categorized into two groups: those who had been prescribed ACEIs or ARBs before admission and those who had not received such treatment before admission. Each patient was evaluated on admission for signs of neurologic dysfunction. Pre-clinical study: Humanized ACE-2 transgenic knock-in mice received the SARS-CoV-2 spike protein via jugular vein injection for 2 weeks. One group had received Losartan (10 mg/kg), an ARB, in their drinking water for two weeks before the injection, while the other group began Losartan treatment after the spike protein injection. Cognitive functions, cerebral blood flow, and cerebrovascular density were determined in all experimental groups. Moreover, vascular inflammation and cell death were assessed. RESULTS Signs of neurological dysfunction were observed in 97 out of 177 patients (51%) taking ACEIs/ARBs prior to admission, compared to 32 out of 118 patients (27%) not receiving ACEI or ARBs. In animal studies, spike protein injection increased vascular inflammation, increased endothelial cell apoptosis, and reduced cerebrovascular density. In parallel, spike protein decreased cerebral blood flow and cognitive function. Our results showed that pretreatment with Losartan exacerbated these effects. However, post-treatment with Losartan prevented spike protein-induced vascular and neurological dysfunctions. CONCLUSION Our clinical data showed that the use of RAS modulators before encountering COVID-19 can initially exacerbate vascular and neurological dysfunctions. Similar findings were demonstrated in the in-vivo experiments; however, the protective effects of targeting the RAS become apparent in the animal model when the treatment is initiated after spike protein injection.
Collapse
Affiliation(s)
- Mackenzi Meier
- Department of Pharmacy Practice, School of Pharmacy, South University, Savannah, Georgia, United States of America
| | - Sara Becker
- Department of Pharmacy Practice, School of Pharmacy, South University, Savannah, Georgia, United States of America
| | - Erica Levine
- Department of Pharmacy Practice, School of Pharmacy, South University, Savannah, Georgia, United States of America
| | - Oriana DuFresne
- Department of Pharmacy Practice, School of Pharmacy, South University, Savannah, Georgia, United States of America
| | - Kaleigh Foster
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Joshua Moore
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Faith N Burnett
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Veronica C Hermanns
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Stan P Heath
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Mohammed Abdelsaid
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Maha Coucha
- Department of Pharmaceutical Sciences, School of Pharmacy, South University, Savannah, Georgia, United States of America
| |
Collapse
|
3
|
Hershberger C, Mariam A, Pantalone KM, Buse JB, Motsinger-Reif AA, Rotroff DM. Polygenic subtype identified in ACCORD trial displays a favorable type 2 diabetes phenotype in the UKBiobank population. Hum Genomics 2024; 18:70. [PMID: 38909264 PMCID: PMC11193210 DOI: 10.1186/s40246-024-00639-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/12/2024] [Indexed: 06/24/2024] Open
Abstract
INTRODUCTION We previously identified a genetic subtype (C4) of type 2 diabetes (T2D), benefitting from intensive glycemia treatment in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial. Here, we characterized the population of patients that met the C4 criteria in the UKBiobank cohort. RESEARCH DESIGN AND METHODS Using our polygenic score (PS), we identified C4 individuals in the UKBiobank and tested C4 status with risk of developing T2D, cardiovascular disease (CVD) outcomes, and differences in T2D medications. RESULTS C4 individuals were less likely to develop T2D, were slightly older at T2D diagnosis, had lower HbA1c values, and were less likely to be prescribed T2D medications (P < .05). Genetic variants in MAS1 and IGF2R, major components of the C4 PS, were associated with fewer overall T2D prescriptions. CONCLUSION We have confirmed C4 individuals are a lower risk subpopulation of patients with T2D.
Collapse
Affiliation(s)
- Courtney Hershberger
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Arshiya Mariam
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Kevin M Pantalone
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH, 44195, USA
- Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - John B Buse
- Division of Endocrinology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alison A Motsinger-Reif
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institute of Health, Durham, NC, USA
| | - Daniel M Rotroff
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
4
|
Xu FF, Zheng F, Chen Y, Wang Y, Ma SB, Ding W, Zhang LS, Guo JZ, Zheng CB, Shen B. Role of thrombospondin-1 in high-salt-induced mesenteric artery endothelial impairment in rats. Acta Pharmacol Sin 2024; 45:545-557. [PMID: 37932403 PMCID: PMC10834453 DOI: 10.1038/s41401-023-01181-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 10/08/2023] [Indexed: 11/08/2023]
Abstract
The matrix glycoprotein thrombospondin-1 (THBS1) modulates nitric oxide (NO) signaling in endothelial cells. A high-salt diet induces deficiencies of NO production and bioavailability, thereby leading to endothelial dysfunction. In this study we investigated the changes of THBS1 expression and its pathological role in the dysfunction of mesenteric artery endothelial cells (MAECs) induced by a high-salt diet. Wild-type rats, and wild-type and Thbs1-/- mice were fed chow containing 8% w/w NaCl for 4 weeks. We showed that a high salt diet significantly increased THBS1 expression and secretion in plasma and MAECs, and damaged endothelium-dependent vasodilation of mesenteric resistance arteries in wild-type animals, but not in Thbs1-/- mice. In rat MAECs, we demonstrated that a high salt environment (10-40 mM) dose-dependently increased THBS1 expression accompanied by suppressed endothelial nitric oxide synthase (eNOS) and phospho-eNOS S1177 production as well as NO release. Blockade of transforming growth factor-β1 (TGF-β1) activity by a TGF-β1 inhibitor SB 431542 reversed THBS1 up-regulation, rescued the eNOS decrease, enhanced phospho-eNOS S1177 expression, and inhibited Smad4 translocation to the nucleus. By conducting dual-luciferase reporter experiments in HEK293T cells, we demonstrated that Smad4, a transcription promoter, upregulated Thbs1 transcription. We conclude that THBS1 contributes to endothelial dysfunction in a high-salt environment and may be a potential target for treatment of high-salt-induced endothelium dysfunction.
Collapse
Affiliation(s)
- Fang-Fang Xu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Fan Zheng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Ye Chen
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yang Wang
- Department of Otolaryngology-Head and Neck Surgery, Lu'an People's Hospital, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, 237000, China
| | - Shao-Bo Ma
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Weng Ding
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Le-Sha Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Ji-Zheng Guo
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Chang-Bo Zheng
- School of Pharmaceutical Science, Kunming Medical University, Kunming, 650500, China.
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
5
|
Deng Y, Ding W, Peng Q, Wang W, Duan R, Zhang Y. Advancement in Beneficial Effects of AVE 0991: A Brief Review. Mini Rev Med Chem 2024; 24:139-158. [PMID: 36998128 DOI: 10.2174/1389557523666230328134932] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 04/01/2023]
Abstract
AVE 0991, a non-peptide analogue of Angiotensin-(1-7) [Ang-(1-7)], is orally active and physiologically well tolerated. Several studies have demonstrated that AVE 0991 improves glucose and lipid metabolism, and contains anti-inflammatory, anti-apoptotic, anti-fibrosis, and anti-oxidant effects. Numerous preclinical studies have also reported that AVE 0991 appears to have beneficial effects on a variety of systemic diseases, including cardiovascular, liver, kidney, cancer, diabetes, and nervous system diseases. This study searched multiple literature databases, including PubMed, Web of Science, EMBASE, Google Scholar, Cochrane Library, and the ClinicalTrials.gov website from the establishment to October 2022, using AVE 0991 as a keyword. This literature search revealed that AVE 0991 could play different roles via various signaling pathways. However, the potential mechanisms of these effects need further elucidation. This review summarizes the benefits of AVE 0991 in several medical problems, including the COVID-19 pandemic. The paper also describes the underlying mechanisms of AVE 0991, giving in-depth insights and perspectives on the pharmaceutical value of AVE 0991 in drug discovery and development.
Collapse
Affiliation(s)
- Yang Deng
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Wangli Ding
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Qiang Peng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Wei Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Rui Duan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yingdong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| |
Collapse
|
6
|
Lin X, Wang X, Feng W, Wan Y, Chai J, Li F, Xu M. The Counteracting Effects of Ang II and Ang-(1-7) on the Function andGrowth of Insulin-secreting NIT-1 Cells. Curr Diabetes Rev 2024; 20:e010124225112. [PMID: 38173074 DOI: 10.2174/0115733998276291231204115314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 01/05/2024]
Abstract
INTRODUCTION China now has the highest number of diabetes in the world. Angiotensin II (Ang II) causes insulin resistance by acting on the insulin signaling pathway of peripheral target tissues. However, its effect on islet β-cells remains unclear. The possible role of Angiotensin-( 1-7) [Ang-(1-7)] as an antagonist to the effects of Ang II and in treating diabetes needs to be elucidated. OBJECTIVES To assess the effects of Ang II and Ang-(1-7) on the function and growth of islet β cell line NIT-1, which is derived from the islets of non-obese diabetic/large T-antigen (NOD/LT) mice with insulinoma. METHODS NIT-1 cells were treated with Ang II, Ang-(1-7) and their respective receptor antagonists. The impact on cell function and growth was then evaluated. RESULTS Ang II significantly reduced insulin-stimulated IR-β-Tyr and Akt-Ser; while Ang-(1-7), saralasin (an Ang II receptor antagonist), and diphenyleneiodonium [DPI, a nicotinamide adenine dinucleotide phosphate oxidase (NOX) antagonist] reversed the inhibiting effect. Conversely, Ang II significantly increased insulin-stimulated intracellular H2O2 and P47 phox, while saralasin and DPI reverted the effect. Furthermore, Ang-(1-7) reduced the elevated concentrations of ROS and MDA while increasing the proliferation rate that was reduced by high glucose, all of which were reversed by A-779, an antagonist of the Mas receptor (MasR). CONCLUSION Angiotensin II poses a negative regulatory effect on insulin signal transduction, increases oxidative stress, and may inhibit the transcription of insulin genes stimulated by insulin in NIT-1 cells. Meanwhile, angiotensin-(1-7) blocked these effects via MasR. These results corroborate the rising potential of the renin-angiotensin system (RAS) in treating diabetes.
Collapse
Affiliation(s)
- Xiuhong Lin
- Department of Clinical Nutrition, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoyun Wang
- Department of Endocrinology, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, People's Republic of China
| | - Weilian Feng
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yan Wan
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jiani Chai
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Feng Li
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Mingtong Xu
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
7
|
Wagenaar GTM, Moll GN. Evolving views on the first two ligands of the angiotensin II type 2 receptor. From putative antagonists to potential agonists? Eur J Pharmacol 2023; 961:176189. [PMID: 37951489 DOI: 10.1016/j.ejphar.2023.176189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/29/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
The renin-angiotensin system is one of the most complex regulatory systems that controls multiple organ functions. One of its key components, angiotensin II (Ang II), stimulates two G-protein coupled class A receptors: the Ang II type 1 (AT1) receptor and the Ang II type 2 (AT2) receptor. While stimulation of the AT1 receptor causes G-protein-dependent signaling and arrestin recruitment, the AT2 receptor seems to have a constitutively active-like conformation and appears to act via G-protein-dependent and -independent pathways. Overstimulation of the AT1 receptor may lead to unwanted effects like inflammation and fibrosis. In contrast, stimulation of the AT2 receptor leads to opposite effects thus restoring the balance. However, the role of the AT2 receptor has become controversial due to beneficial effects of putative AT2 receptor antagonists. The two first synthetic AT2 receptor-selective ligands, peptide CGP42112 and small molecule PD123319, were initially both considered antagonists. CGP42112 was subsequently considered a partial agonist and it was recently demonstrated to be a full agonist. Based on the search-term PD123319 in Pubmed, 1652 studies have investigated putative AT2 receptor antagonist PD123319. Here, we put forward literature that shows beneficial effects of PD123319 alone, even at doses too low for antagonist efficacy. These beneficial effects appear compatible with agonist-like activity via the AT2 receptor. Taken together, a more consistent image of a therapeutic role of stimulated AT2 receptor emerges which may clarify current controversies.
Collapse
Affiliation(s)
| | - Gert N Moll
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG, Groningen, the Netherlands.
| |
Collapse
|
8
|
Cohen-Segev R, Nativ O, Kinaneh S, Aronson D, Kabala A, Hamoud S, Karram T, Abassi Z. Effects of Angiotensin 1-7 and Mas Receptor Agonist on Renal System in a Rat Model of Heart Failure. Int J Mol Sci 2023; 24:11470. [PMID: 37511227 PMCID: PMC10380355 DOI: 10.3390/ijms241411470] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Congestive heart failure (CHF) is often associated with impaired kidney function. Over- activation of the renin-angiotensin-aldosterone system (RAAS) contributes to avid salt/water retention and cardiac hypertrophy in CHF. While the deleterious effects of angiotensin II (Ang II) in CHF are well established, the biological actions of angiotensin 1-7 (Ang 1-7) are not fully characterized. In this study, we assessed the acute effects of Ang 1-7 (0.3, 3, 30 and 300 ng/kg/min, IV) on urinary flow (UF), urinary Na+ excretion (UNaV), glomerular filtration rate (GFR) and renal plasma flow )RPF) in rats with CHF induced by the placement of aortocaval fistula. Additionally, the chronic effects of Ang 1-7 (24 µg/kg/h, via intra-peritoneally implanted osmotic minipumps) on kidney function, cardiac hypertrophy and neurohormonal status were studied. Acute infusion of either Ang 1-7 or its agonist, AVE 0991, into sham controls, but not CHF rats, increased UF, UNaV, GFR, RPF and urinary cGMP. In the chronic protocols, untreated CHF rats displayed lower cumulative UF and UNaV than their sham controls. Chronic administration of Ang 1-7 and AVE 0991 exerted significant diuretic, natriuretic and kaliuretic effects in CHF rats, but not in sham controls. Serum creatinine and aldosterone levels were significantly higher in vehicle-treated CHF rats as compared with controls. Treatment with Ang 1-7 and AVE 0991 reduced these parameters to comparable levels observed in sham controls. Notably, chronic administration of Ang 1-7 to CHF rats reduced cardiac hypertrophy. In conclusion, Ang 1-7 exerts beneficial renal and cardiac effects in rats with CHF. Thus, we postulate that ACE2/Ang 1-7 axis represents a compensatory response to over-activity of ACE/AngII/AT1R system characterizing CHF and suggest that Ang 1-7 may be a potential therapeutic agent in this disease state.
Collapse
Affiliation(s)
- Ravit Cohen-Segev
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Omri Nativ
- Department of Urology, Rambam Health Center, Haifa 3109601, Israel
| | - Safa Kinaneh
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Doron Aronson
- Cardiology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Aviva Kabala
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Shadi Hamoud
- Department of Internal Medicine E, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Tony Karram
- Vascular Surgery, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Zaid Abassi
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
- Laboratory Medicine, Rambam Health Care Campus, Haifa 31096, Israel
| |
Collapse
|
9
|
Molaei A, Molaei E, Hayes AW, Karimi G. Mas receptor: a potential strategy in the management of ischemic cardiovascular diseases. Cell Cycle 2023; 22:1654-1674. [PMID: 37365840 PMCID: PMC10361149 DOI: 10.1080/15384101.2023.2228089] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/10/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
MasR is a critical element in the RAS accessory pathway that protects the heart against myocardial infarction, ischemia-reperfusion injury, and pathological remodeling by counteracting the effects of AT1R. This receptor is mainly stimulated by Ang 1-7, which is a bioactive metabolite of the angiotensin produced by ACE2. MasR activation attenuates ischemia-related myocardial damage by facilitating vasorelaxation, improving cell metabolism, reducing inflammation and oxidative stress, inhibiting thrombosis, and stabilizing atherosclerotic plaque. It also prevents pathological cardiac remodeling by suppressing hypertrophy- and fibrosis-inducing signals. In addition, the potential of MasR in lowering blood pressure, improving blood glucose and lipid profiles, and weight loss has made it effective in modulating risk factors for coronary artery disease including hypertension, diabetes, dyslipidemia, and obesity. Considering these properties, the administration of MasR agonists offers a promising approach to the prevention and treatment of ischemic heart disease.Abbreviations: Acetylcholine (Ach); AMP-activated protein kinase (AMPK); Angiotensin (Ang); Angiotensin receptor (ATR); Angiotensin receptor blocker (ARB); Angiotensin-converting enzyme (ACE); Angiotensin-converting enzyme inhibitor (ACEI); Anti-PRD1-BF1-RIZ1 homologous domain containing 16 (PRDM16); bradykinin (BK); Calcineurin (CaN); cAMP-response element binding protein (CREB); Catalase (CAT); C-C Motif Chemokine Ligand 2 (CCL2); Chloride channel 3 (CIC3); c-Jun N-terminal kinases (JNK); Cluster of differentiation 36 (CD36); Cocaine- and amphetamine-regulated transcript (CART); Connective tissue growth factor (CTGF); Coronary artery disease (CAD); Creatine phosphokinase (CPK); C-X-C motif chemokine ligand 10 (CXCL10); Cystic fibrosis transmembrane conductance regulator (CFTR); Endothelial nitric oxide synthase (eNOS); Extracellular signal-regulated kinase 1/2 (ERK 1/2); Fatty acid transport protein (FATP); Fibroblast growth factor 21 (FGF21); Forkhead box protein O1 (FoxO1); Glucokinase (Gk); Glucose transporter (GLUT); Glycogen synthase kinase 3β (GSK3β); High density lipoprotein (HDL); High sensitive C-reactive protein (hs-CRP); Inositol trisphosphate (IP3); Interleukin (IL); Ischemic heart disease (IHD); Janus kinase (JAK); Kruppel-like factor 4 (KLF4); Lactate dehydrogenase (LDH); Left ventricular end-diastolic pressure (LVEDP); Left ventricular end-systolic pressure (LVESP); Lipoprotein lipase (LPL); L-NG-Nitro arginine methyl ester (L-NAME); Low density lipoprotein (LDL); Mammalian target of rapamycin (mTOR); Mas-related G protein-coupled receptors (Mrgpr); Matrix metalloproteinase (MMP); MAPK phosphatase-1 (MKP-1); Mitogen-activated protein kinase (MAPK); Monocyte chemoattractant protein-1 (MCP-1); NADPH oxidase (NOX); Neuropeptide FF (NPFF); Neutral endopeptidase (NEP); Nitric oxide (NO); Nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB); Nuclear-factor of activated T-cells (NFAT); Pancreatic and duodenal homeobox 1 (Pdx1); Peroxisome proliferator- activated receptor γ (PPARγ); Phosphoinositide 3-kinases (PI3k); Phospholipase C (PLC); Prepro-orexin (PPO); Prolyl-endopeptidase (PEP); Prostacyclin (PGI2); Protein kinase B (Akt); Reactive oxygen species (ROS); Renin-angiotensin system (RAS); Rho-associated protein kinase (ROCK); Serum amyloid A (SAA); Signal transducer and activator of transcription (STAT); Sirtuin 1 (Sirt1); Slit guidance ligand 3 (Slit3); Smooth muscle 22α (SM22α); Sterol regulatory element-binding protein 1 (SREBP-1c); Stromal-derived factor-1a (SDF); Superoxide dismutase (SOD); Thiobarbituric acid reactive substances (TBARS); Tissue factor (TF); Toll-like receptor 4 (TLR4); Transforming growth factor β1 (TGF-β1); Tumor necrosis factor α (TNF-α); Uncoupling protein 1 (UCP1); Ventrolateral medulla (VLM).
Collapse
Affiliation(s)
- Ali Molaei
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Emad Molaei
- PharmD, Assistant of Clinical Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - A. Wallace Hayes
- University of South Florida College of Public Health, Tampa, Florida, USA
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Park BM, Li W, Kim SH. Cardio-protective effects of angiotensin-(1-5) via mas receptor in rats against ischemic-perfusion injury. Peptides 2021; 139:170516. [PMID: 33582209 DOI: 10.1016/j.peptides.2021.170516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/04/2021] [Accepted: 02/07/2021] [Indexed: 11/28/2022]
Abstract
Angiotensin-(1-5) [Ang-(1-5)], which is a metabolite of Ang-(1-7) catalyzed by angiotensin-converting enzyme, is a novel pentapeptide of the renin-angiotensin system. Ang-(1-7), Ang III and Ang IV have a cardio-protective effect via Mas receptor, Ang II type 2 receptor (AT2R) and AT4R, respectively. However, it is not clear whether Ang-(1-5) has cardio-protective effects. The aim of this study is to investigate whether Ang-(1-5) protects the heart against ischemia-reperfusion (I/R) injury. After sacrificing Sprague-Dawley rats, the hearts were perfused with Krebs-Henseleit buffer for a 20 min pre-ischemic period with and without Ang-(1-5) followed by 20 min global ischemia and 50 min reperfusion. Ang-(1-5) (1 μM) improved changes in post-ischemic left ventricular developed pressure (LVDP), ±dP/dt, and post-ischemic left ventricular end-diastolic pressure (LVEDP) induced by reperfusion compared to control hearts. Ang-(1-5) decreased myocardial infarct size and LDH activity, and increased coronary flow and the amount of atrial natriuretic peptide (ANP) in coronary effluent during reperfusion compared to control hearts. Pretreatment with Mas receptor antagonist but not with AT1R or AT2R antagonist attenuated the improvement of changes in I/R-induced ventricular hemodynamics by Ang-(1-5). Ang-(1-5) treatment decreased Bax, caspase-3 and caspase-9 protein levels, and increased Bcl-2 protein level, which were attenuated by A779 pretreatment. Ang-(1-5) treatment increased Mn-superoxide dismutase, catalase, and heme oxygenase-1 protein levels, which was attenuated by A779 pretreatment. These results suggest that the cardio-protective effects of Ang-(1-5) against I/R injury may be partly related to activating anti-oxidant and anti-apoptotic enzymes via Mas receptor.
Collapse
Affiliation(s)
- Byung Mun Park
- Department of Physiology, Research Institute for Endocrine Sciences, Jeonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeollabuk-do 54907, Republic of Korea
| | - Weijian Li
- Department of Physiology, Research Institute for Endocrine Sciences, Jeonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeollabuk-do 54907, Republic of Korea
| | - Suhn Hee Kim
- Department of Physiology, Research Institute for Endocrine Sciences, Jeonbuk National University Medical School, 20 Geonji-ro, Deokjin-gu, Jeonju, Jeollabuk-do 54907, Republic of Korea.
| |
Collapse
|
11
|
Mechanisms of Dietary Sodium-Induced Impairments in Endothelial Function and Potential Countermeasures. Nutrients 2021; 13:nu13010270. [PMID: 33477837 PMCID: PMC7832854 DOI: 10.3390/nu13010270] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 01/11/2023] Open
Abstract
Despite decades of efforts to reduce sodium intake, excess dietary sodium remains commonplace, and contributes to increased cardiovascular morbidity and mortality independent of its effects on blood pressure. An increasing amount of research suggests that high-sodium diets lead to reduced nitric oxide-mediated endothelial function, even in the absence of a change in blood pressure. As endothelial dysfunction is an early step in the progression of cardiovascular diseases, the endothelium presents a target for interventions aimed at reducing the impact of excess dietary sodium. In this review, we briefly define endothelial function and present the literature demonstrating that excess dietary sodium results in impaired endothelial function. We then discuss the mechanisms through which sodium impairs the endothelium, including increased reactive oxygen species, decreased intrinsic antioxidant defenses, endothelial cell stiffening, and damage to the endothelial glycocalyx. Finally, we present selected research findings suggesting that aerobic exercise or increased intake of dietary potassium may counteract the deleterious vascular effects of a high-sodium diet.
Collapse
|
12
|
Dos Reis Costa DEF, Silveira ALM, Campos GP, Nóbrega NRC, de Araújo NF, de Figueiredo Borges L, Dos Santos Aggum Capettini L, Ferreira AVM, Bonaventura D. High-Carbohydrate Diet Enhanced the Anticontractile Effect of Perivascular Adipose Tissue Through Activation of Renin-Angiotensin System. Front Physiol 2021; 11:628101. [PMID: 33519529 PMCID: PMC7845559 DOI: 10.3389/fphys.2020.628101] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The perivascular adipose tissue (PVAT) is an active endocrine organ responsible for release several substances that influence on vascular tone. Increasing evidence suggest that hyperactivation of the local renin-angiotensin system (RAS) in the PVAT plays a pivotal role in the pathogenesis of cardiometabolic diseases. However, the local RAS contribution to the PVAT control of vascular tone during obesity is still not clear. Since the consumption of a high-carbohydrate diet (HC diet) contributes to obesity inducing a rapid and sustained increase in adiposity, so that the functional activity of PVAT could be modulated, we aimed to evaluate the effect of HC diet on the PVAT control of vascular tone and verify the involvement of RAS in this effect. For that, male Balb/c mice were fed standard or HC diet for 4 weeks. Vascular reactivity, histology, fluorescence, and immunofluorescence analysis were performed in intact thoracic aorta in the presence or absence of PVAT. The results showed that HC diet caused an increase in visceral adiposity and also in the PVAT area. Phenylephrine-induced vasoconstriction was significantly reduced in the HC group only in the presence of PVAT. The anticontractile effect of PVAT induced by HC diet was lost when aortic rings were previously incubated with angiotensin-converting enzyme inhibitor, Mas, and AT2 receptors antagonists, PI3K, nNOS, and iNOS inhibitors, hydrogen peroxide (H2O2) decomposing enzyme or non-selective potassium channels blocker. Immunofluorescence assays showed that both Mas and AT2 receptors as well as nNOS and iNOS isoforms were markedly expressed in the PVAT of the HC group. Furthermore, the PVAT from HC group also exhibited higher nitric oxide (NO) and hydrogen peroxide bioavailability. Taken together, these findings suggest that the anticontractile effect of PVAT induced by HC diet involves the signaling cascade triggered by the renin-angiotensin system through the activation of Mas and AT2 receptors, PI3K, nNOS, and iNOS, leading to increased production of nitric oxide and hydrogen peroxide, and subsequently opening of potassium channels. The contribution of PVAT during HC diet-induced obesity could be a compensatory adaptive characteristic in order to preserve the vascular function.
Collapse
Affiliation(s)
| | - Ana Letícia Malheiros Silveira
- Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gianne Paul Campos
- Department of Pharmacology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Natália Ferreira de Araújo
- Department of Pharmacology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luciano de Figueiredo Borges
- Department of Biological Sciences, Morphophysiology & Pathology Sector, Federal University of São Paulo, São Paulo, Brazil
| | | | - Adaliene Versiani Matos Ferreira
- Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Department of Nutrition, Nursing School, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Daniella Bonaventura
- Department of Pharmacology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
13
|
ACE2, angiotensin 1-7 and skeletal muscle: review in the era of COVID-19. Clin Sci (Lond) 2020; 134:3047-3062. [PMID: 33231620 PMCID: PMC7687025 DOI: 10.1042/cs20200486] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Angiotensin converting enzyme-2 (ACE2) is a multifunctional transmembrane protein recently recognised as the entry receptor of the virus causing COVID-19. In the renin–angiotensin system (RAS), ACE2 cleaves angiotensin II (Ang II) into angiotensin 1-7 (Ang 1-7), which is considered to exert cellular responses to counteract the activation of the RAS primarily through a receptor, Mas, in multiple organs including skeletal muscle. Previous studies have provided abundant evidence suggesting that Ang 1-7 modulates multiple signalling pathways leading to protection from pathological muscle remodelling and muscle insulin resistance. In contrast, there is relatively little evidence to support the protective role of ACE2 in skeletal muscle. The potential contribution of endogenous ACE2 to the regulation of Ang 1-7-mediated protection of these muscle pathologies is discussed in this review. Recent studies have suggested that ACE2 protects against ageing-associated muscle wasting (sarcopenia) through its function to modulate molecules outside of the RAS. Thus, the potential association of sarcopenia with ACE2 and the associated molecules outside of RAS is also presented herein. Further, we introduce the transcriptional regulation of muscle ACE2 by drugs or exercise, and briefly discuss the potential role of ACE2 in the development of COVID-19.
Collapse
|
14
|
Verma A, Zhu P, Xu K, Du T, Liao S, Liang Z, Raizada MK, Li Q. Angiotensin-(1-7) Expressed From Lactobacillus Bacteria Protect Diabetic Retina in Mice. Transl Vis Sci Technol 2020; 9:20. [PMID: 33344064 PMCID: PMC7735952 DOI: 10.1167/tvst.9.13.20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/03/2020] [Indexed: 01/04/2023] Open
Abstract
Purpose A multitude of animal studies substantiates the beneficial effects of Ang-(1-7), a peptide hormone in the protective axis of the renin angiotensin system, in diabetes and its associated complications including diabetic retinopathy (DR). However, the clinical application of Ang-(1-7) is limited due to unfavorable pharmacological properties. As emerging evidence implicates gut dysbiosis in pathogenesis of diabetes and supports beneficial effects of probiotics, we sought to develop probiotics-based expression and delivery system to enhance Ang-(1-7) and evaluate the efficacy of engineered probiotics expressing Ang-(1-7) in attenuation of DR in animal models. Methods Ang-(1-7) was expressed in the Lactobacillus species as a secreted fusion protein with a trans-epithelial carrier to allow uptake into circulation. To evaluate the effects of Ang-(1-7) expressed from Lactobacillus paracasei (LP), adult diabetic eNOS-/- and Akita mice were orally gavaged with either 1 × 109 CFU of LP secreting Ang-(1-7) (LP-A), LP alone or vehicle, 3 times/week, for 8 and 12 weeks, respectively. Results Ang-(1-7) is efficiently expressed from different Lactobacillus species and secreted into circulation in mice fed with LP-A. Oral administration of LP-A significantly reduced diabetes-induced loss of retinal vascular capillaries. LP-A treatment also prevented loss of retinal ganglion cells, and significantly decreased retinal inflammatory cytokine expression in both diabetic eNOS-/- and Akita mice. Conclusions These results provide proof-of-concept for feasibility and efficacy of using engineered probiotic species as live vector for delivery of Ang-(1-7) with enhanced bioavailability. Translational Relevance Probiotics-based delivery of Ang-(1-7) may hold important therapeutic potential for the treatment of DR and other diabetic complications.
Collapse
Affiliation(s)
- Amrisha Verma
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ping Zhu
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Kang Xu
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Tao Du
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Shengquan Liao
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Zhibing Liang
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Mohan K. Raizada
- Physiology & Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Qiuhong Li
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
15
|
Raupp D, Fernandes RS, Antunes KH, Perin FA, Rigatto K. Impact of angiotensin II type 1 and G-protein-coupled Mas receptor expression on the pulmonary performance of patients with idiopathic pulmonary fibrosis. Peptides 2020; 133:170384. [PMID: 32777324 PMCID: PMC7411382 DOI: 10.1016/j.peptides.2020.170384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/24/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe interstitial disease with a mean survival of about 2.5-5 years after diagnosis. Its pathophysiology is still a major challenge for science. It is known that angiotensin II (Ang-II) binds AT1 receptor (AT1R) and its overactivation induces fibrosis, inflammation and oxidative stress. In contrast, activation of the Mas receptor (Mas-R) by angiotensin 1-7 opposes the harmful effects induced by Ang-II. Thus, our innovative objective was to analyze, in patients' lung with IPF, the balance between AT1R and Mas-R expression and their possible association with pulmonary spirometric parameters: forced expiratory volume in the first second (FEV1%) and forced vital capacity (FVC%). One cubic centimeter of lung tissue was obtained from IPF patients (n = 6) and from patients without IPF (n = 6) who underwent bronchial carcinoma resection. Receptor expression was quantified using western blot. AT1R expression was significantly higher (34 %) in patients with IPF (P = 0.006), whereas Mas-R was significantly less expressed (54 %) in these patients' lungs (P = 0.046). There was also a positive correlation between Mas-R expression and FEV1% (r = 0.62, P = 0.03) and FVC% (r = 0.58, P = 0.05). Conversely, AT1R expression was negatively correlated with FEV1% (r = 0.80, P = 0.002) and FVC% (r = 0.74, P = 0.006). In conclusion, our results demonstrated an increased expression of AT1R and reduced expression of Mas-R in the lung of patients with IPF. The dominance of AT1R expression is associated with reduced lung function, highlighting the role of the renin-angiotensin system peptides in the pathophysiology of IPF.
Collapse
Affiliation(s)
- Débora Raupp
- Laboratório de Fisiologia Translacional, Curso de Pós-Graduaçao em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Renata Streck Fernandes
- Laboratório de Fisiologia Translacional, Curso de Pós-Graduaçao em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil
| | - Krist Helen Antunes
- Laboratório de Imunologia Clínica e Experimental da Pontifícia, Universidade Católica do Rio Grande do Sul, Brazil
| | - Fabíola Adélia Perin
- Complexo Hospitalar da Irmandade Santa Casa de Misericórdia de Porto Alegre, Brazil
| | - Katya Rigatto
- Laboratório de Fisiologia Translacional, Curso de Pós-Graduaçao em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Brazil.
| |
Collapse
|
16
|
Liao W, Wu J. The ACE2/Ang (1-7)/MasR axis as an emerging target for antihypertensive peptides. Crit Rev Food Sci Nutr 2020; 61:2572-2586. [PMID: 32551837 DOI: 10.1080/10408398.2020.1781049] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Food protein-derived bioactive peptides, particularly antihypertensive peptides, are important constituents of functional foods or nutraceuticals. Most antihypertensive are identified as the inhibitors of angiotensin converting enzyme (ACE), a key enzyme responsible for the generation of angiotensin II (Ang II), which is a vasoconstricting peptide. Hence, ACE has long been used as a universal target to identify antihypertensive peptides. Angiotensin converting enzyme 2 (ACE2), is a homolog of ACE but uses Ang II as its key substrate to produce angiotensin (1-7), exerting vasodilatory activity via the mas receptor (MasR). Therefore, ACE2 functions in the opposite way as ACE and is an emerging novel target for cardiovascular therapy. The potential of food protein-derived bioactive peptides in targeting ACE2 has been rarely explored. While, recently we found that IRW, an egg white ovotransferrin-derived antihypertensive peptide, reduced blood pressure in spontaneously hypertensive rats via the ACE2/Ang (1-7)/MasR axis, indicating a new mechanism of food protein-derived bioactive peptides in reducing blood pressure. The objectives of this review are to summarize the functions of the ACE2/Ang (1-7)/MasR axis and to examine its potential roles in the actions of food protein-derived antihypertensive peptides. The interaction between antihypertensive peptides and the ACE2/Ang (1-7)/MasR axis will also be discussed.
Collapse
Affiliation(s)
- Wang Liao
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jianping Wu
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
17
|
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: 0.8] [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
|
18
|
Zou X, Wang J, Chen C, Tan X, Huang Y, Jose PA, Yang J, Zeng C. Secreted Monocyte miR-27a, via Mesenteric Arterial Mas Receptor-eNOS Pathway, Causes Hypertension. Am J Hypertens 2020; 33:31-42. [PMID: 31328772 DOI: 10.1093/ajh/hpz112] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/09/2019] [Accepted: 07/12/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Essential hypertension is associated with increased plasma concentrations of extracellular vesicles (EVs). We aimed to determine the role of monocyte miR-27a in EVs on arterial Mas receptor expression, and its involvement in the pathogenesis of hypertension. METHODS THP-1 cells were transfected with miR-27a mimic and miR-27a inhibitor, and EVs were collected. Mas receptor expression and endothelial nitric oxide synthase (eNOS) phosphorylation were determined by immunoblotting. Sprague-Dawley (SD) rats received EVs via tail-vein injection. Blood pressure (BP) was measured with the tail-cuff method. The vasodilatory response of mesenteric arteries was measured using a small vessel myograph. RESULTS EVs from THP-1 cells increased rat BP by impairing Ang-(1-7)-mediated vasodilation in mesenteric arteries, which was further exaggerated by EVs from lipopolysaccharides-treated THP-1 cells. As the receptor and key signaling of Ang-(1-7), next experiments found that Mas receptor expression and eNOS phosphorylation were decreased in mesenteric arteries from EVs-treated SD rats. Screening studies found miR-27a in EVs may be involved in this process. Through transfection with miR-27a inhibitor or miR-27a mimic, we found that miR-27a downregulates Mas receptor expression in endothelial cells. Injection of EVs from miR-27a-transfected HEK-293 cells decreased Mas receptor and eNOS phosphorylation in mesenteric arteries, impaired Ang-(1-7)-mediated vasodilation and increased BP. Earlier effects were reversed using cells with downregulation of miR-27 in EVs. CONCLUSIONS Monocyte miR-27a in EVs decreases Mas receptor expression and eNOS phosphorylation in endothelium, impairs Ang-(1-7)-mediated vasodilation, and causes hypertension. Understanding the contributions of EVs in the pathogenesis of hypertension may facilitate their use as a diagnostic biomarker.
Collapse
Affiliation(s)
- Xue Zou
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Chongqing Institute of Cardiology and Chongqing Key Laboratory for Hypertension Research, Chongqing, P.R. China
| | - Jialiang Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Chongqing Institute of Cardiology and Chongqing Key Laboratory for Hypertension Research, Chongqing, P.R. China
| | - Caiyu Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Chongqing Institute of Cardiology and Chongqing Key Laboratory for Hypertension Research, Chongqing, P.R. China
| | - Xiaorong Tan
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Chongqing Institute of Cardiology and Chongqing Key Laboratory for Hypertension Research, Chongqing, P.R. China
| | - Yu Huang
- Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, P.R. China
| | - Pedro A Jose
- Division of Renal Disease and Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Chongqing Institute of Cardiology and Chongqing Key Laboratory for Hypertension Research, Chongqing, P.R. China
| |
Collapse
|
19
|
Effects of Angiotensin-(1-7) and Angiotensin II on Acetylcholine-Induced Vascular Relaxation in Spontaneously Hypertensive Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6512485. [PMID: 31827689 PMCID: PMC6886389 DOI: 10.1155/2019/6512485] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 01/31/2023]
Abstract
Endothelial dysfunction of small arteries occurs in patients with hypertension and in various hypertensive models. Endothelial function is usually evaluated by the degree of acetylcholine- (ACh-) induced vascular relaxation. Our previous study has found that compared to Wistar-Kyoto rats (WKY), ACh-induced vasodilatation was attenuated significantly in the mesenteric artery (MA), coronary artery (CA), and pulmonary artery (PA) of spontaneously hypertensive rats (SHR). This study investigated the influence of angiotensin- (Ang-) (1-7) and Ang II on blood pressure and ACh-induced vascular relaxation, as well as their interactive roles and downstream signal pathways in SHR and WKY. Intravenous injection of Ang II significantly increased, while Ang-(1-7) decreased the mean arterial pressure (MAP) in SHR. Ang-(1-7) improved ACh-induced relaxation in the MA, CA, and PA of SHR, while Ang II further attenuated it, which were inhibited by pretreatment with Mas receptor antagonist A-779 or AT1 receptor antagonist losartan, respectively. Ang-(1-7) decreased the basal arterial tension, and Ang II induced great vasoconstriction in SHR. Pretreatment with Ang-(1-7) inhibited the Ang II-induced pressor response, vasoconstriction, and the effects on ACh-induced relaxation in SHR. AT1 receptor expression was higher, while nitric oxide (NO), cGMP, and protein kinase G (PKG) levels of arteries were lower in SHR than in WKY. Ang II decreased, while Ang-(1-7) increased, the levels of NO, cGMP, and PKG of arteries. In addition, pretreatment with Ang-(1-7) inhibited the Ang II-induced reduction of NO, cGMP, and PKG in SHR. These results indicate that the activation of the Mas receptor by Ang-(1-7) can improve endothelial function and decrease MAP in SHR and inhibit the deteriorative effect of Ang II on endothelial function through the NO-cGMP-PKG pathway.
Collapse
|
20
|
Angiotensin-(1-7) induced vascular relaxation in spontaneously hypertensive rats. Nitric Oxide 2019; 88:1-9. [PMID: 30880106 DOI: 10.1016/j.niox.2019.03.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/10/2019] [Accepted: 03/11/2019] [Indexed: 11/20/2022]
Abstract
Enhanced vasoconstriction and decreased vasodilatation due to endothelial dysfunction contribute to the progression of hypertension. Angiotensin (Ang)-(1-7) plays important roles in regulating the cardiovascular activity. The current study aimed to investigate the roles of Ang-(1-7) in modulating blood pressure, vascular tension and its signal pathway in spontaneously hypertensive rats (SHR). The effects of intravenous injection of drugs were determined in rats with anesthesia in vivo. Mesenteric artery (MA), coronary artery (CA) and pulmonary artery (PA) were isolated from rats and isometric tension measurements in arteries were performed. Compared with Wistar-Kyoto rats (WKY), the high K+ induced vasoconstriction was enhanced and acetylcholine-induced vasodilatation were attenuated in the MA, CA and PA in SHR. Intravenous injection of Ang-(1-7) decreased, while A-779 increased mean arterial pressure and abolished the hypotensive effect of Ang-(1-7) in SHR. Ang-(1-7) caused dose-dependent relaxation in MA, CA and PA in SHR, which was inhibited by pretreatment with Mas receptor antagonist A-779, nitric oxide (NO) synthase inhibitor l-NAME, guanylate cyclase inhibitor ODQ and protein kinase G (PKG) inhibitor DT-2. The Mas receptor expression, NO, cGMP and PKG levels of the three above arteries of SHR were lower than that of WKY. Ang-(1-7) increased the NO, cGMP and PKG levels in arteries from SHR, which was blocked by A-779. Activation of the Mas receptor by Ang-(1-7) relaxes the MA, CA, and PA through the NO-cGMP-PKG pathway, which contributes to the decrease of arterial pressure in SHR.
Collapse
|
21
|
Chen QF, Hao H, Kuang XD, Hu QD, Huang YH, Zhou XY. BML-111, a lipoxin receptor agonist, protects against acute injury via regulating the renin angiotensin-aldosterone system. Prostaglandins Other Lipid Mediat 2018; 140:9-17. [PMID: 30412790 DOI: 10.1016/j.prostaglandins.2018.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 09/30/2018] [Accepted: 11/05/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The renin angiotensin-aldosterone system (RAAS) and lipoxins (LXs) have similar roles in many processes. We previously reported that BML-111, a Lipoxin receptor agonist, inhibited chronic injury hepatic fibrosis by regulating RAAS, but whether LXs are involved in BML-111-mediated protection from acute injury is unclear still. METHODS We established models of acute liver/lung injury and confirmed them with histopathology and myeloperoxidase (MPO) measurements. BML-111, a lipoxin receptor agonist, was applied to mimic the effects of LXs. The contents and activities of angiotensin converting enzyme(ACE) and angiotensinconverting enzyme 2 (ACE2) were measured through ELISA and activity assay kits respectively. Angiotensin II (AngII), angiotensin-(1-7) (Ang-1-7), AngII type 1 receptor (AT1R), and Mas receptor were quantified with ELISA and Western blot. RESULTS Models of acute injury were established successfully and BML-111 protected LPS-induced acute lung injury and LPS/D-GalN-induced acute liver injury. BML-111 repressed the activity of ACE, but increased the activity of ACE2. BML-111 decreased the expression levels of ACE, AngII, and AT1R, meanwhile increased the levels of ACE2, Ang-(1-7), and Mas. Furthermore, BOC-2, an inhibitor of lipoxin receptor, reversed all the effects. CONCLUSION BML-111 could protect against acute injury via regulation RAAS.
Collapse
Affiliation(s)
- Qiong-Feng Chen
- Department of Pathophysiology, Medical College of Nanchang University, Nanchang 330006, China
| | - Hua Hao
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiao-Dong Kuang
- Department of Pathology, Medical College of Nanchang University, Nanchang 330006, China
| | - Quan-Dong Hu
- Department of Pathophysiology, Medical College of Nanchang University, Nanchang 330006, China
| | - Yong-Hong Huang
- Department of Pathophysiology, Medical College of Nanchang University, Nanchang 330006, China; Jiangxi Province Key Laboratory of Tumor Etiology and Molecular Pathology, Nanchang 330006, China
| | - Xiao-Yan Zhou
- Department of Pathophysiology, Medical College of Nanchang University, Nanchang 330006, China; Jiangxi Province Key Laboratory of Tumor Etiology and Molecular Pathology, Nanchang 330006, China.
| |
Collapse
|
22
|
de Souza-Neto FP, Carvalho Santuchi M, de Morais E Silva M, Campagnole-Santos MJ, da Silva RF. Angiotensin-(1-7) and Alamandine on Experimental Models of Hypertension and Atherosclerosis. Curr Hypertens Rep 2018. [PMID: 29541937 DOI: 10.1007/s11906-018-0798-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review was to summarize the current knowledge on the role of angiotensin-(1-7) [Ang-(1-7)] and alamandine in experimental hypertension and atherosclerosis. RECENT FINDINGS The renin-angiotensin system (RAS) is a very complex system, composed of a cascade of enzymes, peptides, and receptors, known to be involved in the pathogenesis of hypertension and atherosclerosis. Ang-(1-7), identified and characterized in 1987, and alamandine, discovered 16 years after, are the newest two main effector molecules from the RAS, protecting the vascular system against hypertension and atherosclerosis. While the beneficial effects of Ang-(1-7) have been widely studied in several experimental models of hypertension, much less studies were performed in experimental models of atherosclerosis. Alamandine has shown similar vascular effects to Ang-(1-7), namely, endothelial-dependent vasorelaxation mediated by nitric oxide and hypotensive effects in experimental hypertension. There are few studies on the effects of alamandine on atherosclerosis.
Collapse
Affiliation(s)
- Fernando Pedro de Souza-Neto
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos. 6627, Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Melissa Carvalho Santuchi
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos. 6627, Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Mario de Morais E Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos. 6627, Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Maria José Campagnole-Santos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos. 6627, Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Rafaela Fernandes da Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos. 6627, Pampulha, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
23
|
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: 89] [Impact Index Per Article: 11.1] [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
|
24
|
Nehme A, Zibara K. Cellular distribution and interaction between extended renin-angiotensin-aldosterone system pathways in atheroma. Atherosclerosis 2017; 263:334-342. [PMID: 28600074 DOI: 10.1016/j.atherosclerosis.2017.05.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 04/14/2017] [Accepted: 05/24/2017] [Indexed: 01/06/2023]
Abstract
The importance of the renin-angiotensin-aldosterone system (RAAS) in the development of atherosclerotic has been experimentally documented. In fact, RAAS components have been shown to be locally expressed in the arterial wall and to be differentially regulated during atherosclerotic lesion progression. RAAS transcripts and proteins were shown to be differentially expressed and to interact in the 3 main cells of atheroma: endothelial cells, vascular smooth muscle cells, and macrophages. This review describes the local expression and cellular distribution of extended RAAS components in the arterial wall and their differential regulation during atherosclerotic lesion development.
Collapse
Affiliation(s)
- Ali Nehme
- EA4173, Functional Genomics of Arterial Hypertension, Hôpital Nord-Ouest, Villefranche-sur-Saône, Université Lyon1, Lyon, France; ER045, Laboratory of Stem Cells, Department of Biology, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- ER045, Laboratory of Stem Cells, Department of Biology, Faculty of Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
25
|
Castelo-Branco RC, Leite-Dellova DCA, Fernandes FB, Malnic G, de Mello-Aires M. The effects of angiotensin-(1-7) on the exchanger NHE3 and on [Ca 2+] i in the proximal tubules of spontaneously hypertensive rats. Am J Physiol Renal Physiol 2017; 313:F450-F460. [PMID: 28490531 DOI: 10.1152/ajprenal.00557.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 11/22/2022] Open
Abstract
The acute effects of angiotensin-1-7 [ANG-(1-7)] on the reabsorptive bicarbonate flow (J[Formula: see text]) were evaluated using stationary microperfusion in vivo in the proximal tubules of spontaneously hypertensive rats (SHR) and their normotensive controls, Wistar-Kyoto (WKY) rats, using a microelectrode sensitive to H+ In WKY rats, the control J[Formula: see text] was 2.40 ± 0.10 nmol·cm-2·s-1 (n = 120); losartan (10-7 M) or A779 (10-6 M, a specific Mas antagonist), alone or in combination with losartan, decreased the J[Formula: see text] ANG-(1-7) had biphasic effects on J[Formula: see text]: at 10-9 M, it inhibited, and at 10-6, it stimulated the flow. S3226 [10-6 M, a specific Na+-H+ exchanger 3 (NHE3) antagonist] decreased J[Formula: see text] and changed the stimulatory effect of ANG-(1-7) to an inhibitory one but did not alter the inhibitory action of ANG-(1-7). In SHR, the control J[Formula: see text] was 2.04 ± 0.13 nmol·cm-2·s-1 (n = 56), and A779 and/or losartan reduced the flow. ANG-(1-7) at 10-9 M increased J[Formula: see text], and ANG-(1-7) at 10-6 M reduced it. The effects of A779, losartan, and S3226 on the J[Formula: see text] were similar to those found in WKY rats, which indicated that in SHR, the ANG-(1-7) action on the NHE3 was via Mas and ANG II type 1. The cytosolic calcium in the WKY or SHR rats was ~100 nM and was increased by ANG-(1-7) at 10-9 or 10-6 M. In hypertensive animals, a high plasma level of ANG-(1-7) inhibited NHE3 in the proximal tubule, which mitigated the hypertension caused by the high plasma level of ANG II.
Collapse
Affiliation(s)
| | - Deise C A Leite-Dellova
- Department of Basic Sciences, Faculdade de Zootecnia e Engenharia de Alimentos, University of São Paulo, Pirassununga, Brazil; and
| | - Fernanda Barrinha Fernandes
- Presbiteriana Mackenzie University of São Paulo and Department of Nephrology, Federal University of São Paulo-Universidade Estadual Paulista, São Paulo, Brazil
| | - Gerhard Malnic
- Department of Physiology and Biophysics, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Margarida de Mello-Aires
- Department of Physiology and Biophysics, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Leonhardt J, Villela DC, Teichmann A, Münter LM, Mayer MC, Mardahl M, Kirsch S, Namsolleck P, Lucht K, Benz V, Alenina N, Daniell N, Horiuchi M, Iwai M, Multhaup G, Schülein R, Bader M, Santos RA, Unger T, Steckelings UM. Evidence for Heterodimerization and Functional Interaction of the Angiotensin Type 2 Receptor and the Receptor MAS. Hypertension 2017; 69:1128-1135. [PMID: 28461604 DOI: 10.1161/hypertensionaha.116.08814] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/21/2016] [Accepted: 04/06/2017] [Indexed: 11/16/2022]
Abstract
The angiotensin type 2 receptor (AT2R) and the receptor MAS are receptors of the protective arm of the renin-angiotensin system. They mediate strikingly similar actions. Moreover, in various studies, AT2R antagonists blocked the effects of MAS agonists and vice versa. Such cross-inhibition may indicate heterodimerization of these receptors. Therefore, this study investigated the molecular and functional interplay between MAS and the AT2R. Molecular interactions were assessed by fluorescence resonance energy transfer and by cross correlation spectroscopy in human embryonic kidney-293 cells transfected with vectors encoding fluorophore-tagged MAS or AT2R. Functional interaction of AT2R and MAS was studied in astrocytes with CX3C chemokine receptor-1 messenger RNA expression as readout. Coexpression of fluorophore-tagged AT2R and MAS resulted in a fluorescence resonance energy transfer efficiency of 10.8 ± 0.8%, indicating that AT2R and MAS are capable to form heterodimers. Heterodimerization was verified by competition experiments using untagged AT2R and MAS. Specificity of dimerization of AT2R and MAS was supported by lack of dimerization with the transient receptor potential cation channel, subfamily C-member 6. Dimerization of the AT2R was abolished when it was mutated at cysteine residue 35. AT2R and MAS stimulation with the respective agonists, Compound 21 or angiotensin-(1-7), significantly induced CX3C chemokine receptor-1 messenger RNA expression. Effects of each agonist were blocked by an AT2R antagonist (PD123319) and also by a MAS antagonist (A-779). Knockout of a single of these receptors made astrocytes unresponsive for both agonists. Our results suggest that MAS and the AT2R form heterodimers and that-at least in astrocytes-both receptors functionally depend on each other.
Collapse
Affiliation(s)
- Julia Leonhardt
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Daniel C Villela
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Anke Teichmann
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Lisa-Marie Münter
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Magnus C Mayer
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Maibritt Mardahl
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Sebastian Kirsch
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Pawel Namsolleck
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Kristin Lucht
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Verena Benz
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Natalia Alenina
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Nicholas Daniell
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Masatsugu Horiuchi
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Masaru Iwai
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Gerhard Multhaup
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Ralf Schülein
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Michael Bader
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Robson A Santos
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Thomas Unger
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.)
| | - Ulrike Muscha Steckelings
- From the Center for Cardiovascular Research, Charité-Medical Faculty Berlin, Germany (J.L., D.C.V., M.M., S.K., P.N., K.L., V.B., N.D., T.U., U.M.S.); The Integrated Research and Treatment Center for Sepsis Control and Care (CSCC) and Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Germany (J.L.); Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil (D.C.V., R.A.S.); Faculty of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil (D.C.V.); Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany (A.T., R.S.); Institut für Chemie und Biochemie, Free University Berlin, Germany (L.-M.M., M.C.M., G.M.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada (L.-M.M., G.M.); CARIM, Maastricht University, The Netherlands (P.N., T.U.); Max-Delbrück-Center for Molecular Medicine, Berlin-Buch, Germany (N.A., M.B.); Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University Graduate School of Medicine, Japan (M.H., M.I.); and IMM-Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense (U.M.S.).
| |
Collapse
|
27
|
Abstract
Salt resistance/sensitivity refers specifically to the effect of dietary sodium chloride (salt) intake on BP. Increased dietary salt intake promotes an early and uniform expansion of extracellular fluid volume and increased cardiac output. To compensate for these hemodynamic changes and maintain constant BP in salt resistance, renal and peripheral vascular resistance falls and is associated with an increase in production of nitric oxide. In contrast, the decline in peripheral vascular resistance and the increase in nitric oxide are impaired or absent in salt sensitivity, promoting an increase in BP in these individuals. Endothelial dysfunction may pose a particularly significant risk factor in the development of salt sensitivity and subsequent hypertension. Vulnerable salt-sensitive populations may have in common underlying endothelial dysfunction due to genetic or environmental influences. These individuals may be very sensitive to the hemodynamic stress of increased effective blood volume, setting in motion untoward molecular and biochemical events that lead to overproduction of TGF-β, oxidative stress, and limited bioavailable nitric oxide. Finally, chronic high-salt ingestion produces endothelial dysfunction, even in salt-resistant subjects. Thus, the complex syndrome of salt sensitivity may be a function of the endothelium, which is integrally involved in the vascular responses to high salt intake.
Collapse
Affiliation(s)
| | - Louis J Dell'Italia
- Departments of Medicine and
- Department of Medicine, Veterans Affairs Medical Center, Birmingham, Alabama
| | - Paul W Sanders
- Departments of Medicine and
- Department of Medicine, Veterans Affairs Medical Center, Birmingham, Alabama
- Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
28
|
Skiba DS, Nosalski R, Mikolajczyk TP, Siedlinski M, Rios FJ, Montezano AC, Jawien J, Olszanecki R, Korbut R, Czesnikiewicz-Guzik M, Touyz RM, Guzik TJ. Anti-atherosclerotic effect of the angiotensin 1-7 mimetic AVE0991 is mediated by inhibition of perivascular and plaque inflammation in early atherosclerosis. Br J Pharmacol 2017; 174:4055-4069. [PMID: 27935022 PMCID: PMC5659999 DOI: 10.1111/bph.13685] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/28/2016] [Accepted: 11/30/2016] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE Inflammation plays a key role in atherosclerosis. The protective role of angiotensin 1-7 (Ang-(1-7)) in vascular pathologies suggested the therapeutic use of low MW, non-peptide Ang-(1-7) mimetics, such as AVE0991. The mechanisms underlying the vaso-protective effects of AVE0991, a Mas receptor agonist, remain to be explored. EXPERIMENTAL APPROACH We investigated the effects of AVE0991 on the spontaneous atherosclerosis in apolipoprotein E (ApoE)-/- mice, in the context of vascular inflammation and plaque stability. KEY RESULTS AVE0991 has significant anti-atherosclerotic properties in ApoE-/- mice and increases plaque stability, by reducing plaque macrophage content, without effects on collagen. Using the descending aorta of chow-fed ApoE-/- mice, before significant atherosclerotic plaque develops, we gained insight to early events in atherosclerosis. Interestingly, perivascular adipose tissue (PVAT) and adventitial infiltration with macrophages and T-cells precedes atherosclerotic plaque or the impairment of endothelium-dependent NO bioavailability (a measure of endothelial function). AVE0991 inhibited perivascular inflammation, by reducing chemokine expression in PVAT and through direct actions on monocytes/macrophages inhibiting their activation, characterized by production of IL-1β, TNF-α, CCL2 and CXCL10, and differentiation to M1 phenotype. Pretreatment with AVE0991 inhibited migration of THP-1 monocytes towards supernatants of activated adipocytes (SW872). Mas receptors were expressed in PVAT and in THP-1 cells in vitro, and the anti-inflammatory effects of AVE0991 were partly Mas dependent. CONCLUSIONS AND IMPLICATIONS The selective Mas receptor agonist AVE0991 exhibited anti-atherosclerotic and anti-inflammatory actions, affecting monocyte/macrophage differentiation and recruitment to the perivascular space during early stages of atherosclerosis in ApoE-/- mice. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- D S Skiba
- Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - R Nosalski
- Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - T P Mikolajczyk
- Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - M Siedlinski
- Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland
| | - F J Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - A C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - J Jawien
- Department of Pharmacology, Jagiellonian University School of Medicine, Krakow, Poland
| | - R Olszanecki
- Department of Pharmacology, Jagiellonian University School of Medicine, Krakow, Poland
| | - R Korbut
- Department of Pharmacology, Jagiellonian University School of Medicine, Krakow, Poland
| | - M Czesnikiewicz-Guzik
- Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland
| | - R M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - T J Guzik
- Department of Internal and Agricultural Medicine, Jagiellonian University School of Medicine, Krakow, Poland.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| |
Collapse
|
29
|
Hoffmann BR, Stodola TJ, Wagner JR, Didier DN, Exner EC, Lombard JH, Greene AS. Mechanisms of Mas1 Receptor-Mediated Signaling in the Vascular Endothelium. Arterioscler Thromb Vasc Biol 2017; 37:433-445. [PMID: 28082260 DOI: 10.1161/atvbaha.116.307787] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/02/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Angiotensin II (AngII) has been shown to regulate angiogenesis and at high pathophysiological doses to cause vasoconstriction through the AngII receptor type 1. Angiotensin 1 to 7 (Ang-(1-7)) acting through the Mas1 receptor can act antagonistically to high pathophysiological levels of AngII by inducing vasodilation, whereas the effects of Ang-(1-7) signaling on angiogenesis are less defined. To complicate the matter, there is growing evidence that a subpressor dose of AngII produces phenotypes similar to Ang-(1-7). APPROACH AND RESULTS This study shows that low-dose Ang-(1-7), acting through the Mas1 receptor, promotes angiogenesis and vasodilation similar to a low, subpressor dose of AngII acting through AngII receptor type 1. In addition, we show through in vitro tube formation that Ang-(1-7) augments the angiogenic response in rat microvascular endothelial cells. Using proteomic and genomic analyses, downstream components of Mas1 receptor signaling were identified, including Rho family of GTPases, phosphatidylinositol 3-kinase, protein kinase D1, mitogen-activated protein kinase, and extracellular signal-related kinase signaling. Further experimental antagonism of extracellular signal-related kinases 1/2 and p38 mitogen-activated protein kinase signaling inhibited endothelial tube formation and vasodilation when stimulated with equimolar, low doses of either AngII or Ang-(1-7). CONCLUSIONS These results significantly expand the known Ang-(1-7)/Mas1 receptor signaling pathway and demonstrate an important distinction between the pathological effects of elevated and suppressed AngII compared with the beneficial effects of AngII normalization and Ang-(1-7) administration. The observed convergence of Ang-(1-7)/Mas1 and AngII/AngII receptor type 1 signaling at low ligand concentrations suggests a nuanced regulation in vasculature. These data also reinforce the importance of mitogen-activated protein kinase/extracellular signal-related kinase signaling in maintaining vascular function.
Collapse
Affiliation(s)
- Brian R Hoffmann
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Timothy J Stodola
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Jordan R Wagner
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Daniela N Didier
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Eric C Exner
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Julian H Lombard
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee
| | - Andrew S Greene
- From the Department of Medicine, Division of Cardiology (B.R.H.), the Department of Biomedical Engineering (B.R.H., A.S.G.), and the Department of Physiology (T.J.S., J.R.W., J.H.L., D.N.D., E.C.E., A.S.G.), Cardiovascular Center (B.R.H.), Medical College of Wisconsin, Milwaukee.
| |
Collapse
|
30
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 628] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
31
|
Yu L, Yuan K, Phuong HTA, Park BM, Kim SH. Angiotensin-(1-5), an active mediator of renin-angiotensin system, stimulates ANP secretion via Mas receptor. Peptides 2016; 86:33-41. [PMID: 27660028 DOI: 10.1016/j.peptides.2016.09.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/17/2016] [Accepted: 09/19/2016] [Indexed: 01/29/2023]
Abstract
Angiotensin-(1-5) [Ang-(1-5)], which is a metabolite of Angiotensin-(1-7) [Ang-(1-7)] catalyzed by angiotensin-converting enzyme (ACE), is a pentapeptide of the renin-angiotensin system (RAS). It has been reported that Ang-(1-7) and Ang-(1-9) stimulate the secretion of atrial natriuretic peptide (ANP) via Mas receptor (Mas R) and Ang II type 2 receptor (AT2R), respectively. However, it still remains unknown whether Ang-(1-5) has a similar function to Ang-(1-7). We investigated the effect of Ang-(1-5) on ANP secretion and to define its signaling pathway using isolated perfused beating rat atria. Ang-(1-5) (0.3, 3, 10μM) stimulated high pacing frequency-induced ANP secretion in a dose-dependent manner. Ang-(1-5)-induced ANP secretion (3μM) was attenuated by the pretreatment with an antagonist of Mas R (A-779) but not by an antagonist of AT1R (losartan) or AT2R (PD123,319). An inhibitor for phosphatidylinositol 3-kinase (PI3K; wortmannin), protein kinase B (Akt; API-2), or nitric oxide synthase (NOS; L-NAME) also attenuated the augmentation of ANP secretion induced by Ang-(1-5). Ang-(1-5)-induced ANP secretion was markedly attenuated in isoproterenol-treated hypertrophied atria. The secretagogue effect of Ang-(1-5) on ANP secretion was similar to those induced by Ang-(1-9) and Ang-(1-7). These results suggest that Ang-(1-5) is an active mediator of renin-angiotensin system to stimulate ANP secretion via Mas R and PI3K-Akt-NOS pathway.
Collapse
Affiliation(s)
- Lamei Yu
- Department of Physiology, Research Institute for Endocrine Sciences Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Kuichang Yuan
- Department of Internal Medicine, Yanbian University, China
| | - Hoang Thi Ai Phuong
- Department of Physiology, Research Institute for Endocrine Sciences Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Byung Mun Park
- Department of Physiology, Research Institute for Endocrine Sciences Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Suhn Hee Kim
- Department of Physiology, Research Institute for Endocrine Sciences Chonbuk National University Medical School, Jeonju, Republic of Korea.
| |
Collapse
|
32
|
Raffai G, Lombard JH. Angiotensin-(1-7) Selectively Induces Relaxation and Modulates Endothelium-Dependent Dilation in Mesenteric Arteries of Salt-Fed Rats. J Vasc Res 2016; 53:105-118. [PMID: 27676088 DOI: 10.1159/000448714] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/26/2016] [Indexed: 12/19/2022] Open
Abstract
This study investigated the acute effects of angiotensin-(1-7) and AVE0991 on active tone and vasodilator responses to bradykinin and acetylcholine in isolated mesenteric arteries from Sprague-Dawley rats fed a high-salt (HS; 4% NaCl) versus a normal salt (NS; 0.4% NaCl) diet. Angiotensin-(1-7) and AVE0991 elicited relaxation, and angiotensin-(1-7) unmasked vasodilator responses to bradykinin in arteries from HS-fed rats. These effects of angiotensin-(1-7) and AVE0991 were inhibited by endothelium removal, A779, PD123319, HOE140 and L-NAME. Angiotensin-(1-7) also restored the acetylcholine-induced relaxation that was suppressed by the HS diet. Vasodilator responses to bradykinin and acetylcholine in the presence of angiotensin-(1-7) were mimicked by captopril and the AT2 receptor agonist CGP42112 in arteries from HS-fed rats. Thus, in contrast to salt-induced impairment of vascular relaxation in response to vasodilator stimuli, angiotensin-(1-7) induces endothelium-dependent and NO-mediated relaxation, unmasks bradykinin responses via activation of mas and AT2 receptors, and restores acetylcholine-induced vasodilation in HS-fed rats. AT2 receptor activation and angiotensin-converting enzyme (ACE) inhibition shared the ability of angiotensin-(1-7) to enhance bradykinin and acetylcholine responses in HS-fed rats. These findings suggest a therapeutic potential for mas and/or AT2 receptor activation and ACE inhibition in restoring endothelial function impaired by elevated dietary salt intake or other pathological conditions.
Collapse
Affiliation(s)
- Gábor Raffai
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wis., USA
| | | |
Collapse
|
33
|
Endoplasmic reticulum stress inhibition reduces hypertension through the preservation of resistance blood vessel structure and function. J Hypertens 2016; 34:1556-69. [DOI: 10.1097/hjh.0000000000000943] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
34
|
Gromotowicz-Poplawska A, Szoka P, Kolodziejczyk P, Kramkowski K, Wojewodzka-Zelezniakowicz M, Chabielska E. New agents modulating the renin-angiotensin-aldosterone system-Will there be a new therapeutic option? Exp Biol Med (Maywood) 2016; 241:1888-1899. [PMID: 27439538 DOI: 10.1177/1535370216660211] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/22/2016] [Indexed: 12/19/2022] Open
Abstract
The renin-angiotensin-aldosterone system (RAAS) is more complex than it was originally regarded. According to the current subject knowledge, there are two main axes of the RAAS: (1) angiotensin-converting enzyme (ACE)-angiotensin II-AT1 receptor axis and (2) ACE2-angiotensin-(1-7)-Mas receptor axis. The activation of the first axis leads to deleterious effects, including vasoconstriction, endothelial dysfunction, thrombosis, inflammation, and fibrosis; therefore, blocking the components of this axis is a highly rational and commonly used therapeutic procedure. The ACE2-Ang-(1-7)-Mas receptor axis has a different role, since it often opposes the effects induced by the classical ACE-Ang II-AT1 axis. Once the positive effects of the ACE2-Ang-(1-7)-Mas axis were discovered, the alternative ways of pharmacotherapy activating this axis of RAAS appeared. This article briefly describes new molecules affecting the RAAS, namely: recombinant human ACE2, ACE2 activators, angiotensin-(1-7) peptide and non-peptide analogs, aldosterone synthase inhibitors, and the third and fourth generation of mineralocorticoid receptor antagonists. The results of the experimental and clinical studies are encouraging, which leads us to believe that these new molecules can support the treatment of cardiovascular diseases as well as cardiometabolic disorders.
Collapse
Affiliation(s)
| | - Piotr Szoka
- Department of Biopharmacy, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Patrycjusz Kolodziejczyk
- Department of Pharmaceutical Analysis, Medical University of Bialystok, 15-522 Bialystok, Poland
| | - Karol Kramkowski
- Department of Biopharmacy, Medical University of Bialystok, 15-089 Bialystok, Poland
| | | | - Ewa Chabielska
- Department of Biopharmacy, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
35
|
Durand MJ, Zinkevich NS, Riedel M, Gutterman DD, Nasci VL, Salato VK, Hijjawi JB, Reuben CF, North PE, Beyer AM. Vascular Actions of Angiotensin 1-7 in the Human Microcirculation: Novel Role for Telomerase. Arterioscler Thromb Vasc Biol 2016; 36:1254-62. [PMID: 27079876 DOI: 10.1161/atvbaha.116.307518] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/28/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVE This study examined vascular actions of angiotensin 1-7 (ANG 1-7) in human atrial and adipose arterioles. APPROACH AND RESULTS The endothelium-derived hyperpolarizing factor of flow-mediated dilation (FMD) switches from antiproliferative nitric oxide (NO) to proatherosclerotic hydrogen peroxide in arterioles from humans with coronary artery disease (CAD). Given the known vasoprotective properties of ANG 1-7, we tested the hypothesis that overnight ANG 1-7 treatment restores the NO component of FMD in arterioles from patients with CAD. Endothelial telomerase activity is essential for preserving the NO component of vasodilation in the human microcirculation; thus, we also tested whether telomerase activity was necessary for ANG 1-7-mediated vasoprotection by treating separate arterioles with ANG 1-7±the telomerase inhibitor 2-[[(2E)-3-(2-naphthalenyl)-1-oxo-2-butenyl1-yl]amino]benzoic acid. ANG 1-7 dilated arterioles from patients without CAD, whereas dilation was significantly reduced in arterioles from patients with CAD. In atrial arterioles from patients with CAD incubated with ANG 1-7 overnight, the NO synthase inhibitor NG-nitro-l-arginine methyl ester abolished FMD, whereas the hydrogen peroxide scavenger polyethylene glycol catalase had no effect. Conversely, in vessels incubated with ANG 1-7+2-[[(2E)-3-(2-naphthalenyl)-1-oxo-2-butenyl1-yl]amino]benzoic acid, NG-nitro-l-arginine methyl ester had no effect on FMD, but polyethylene glycol catalase abolished dilation. In cultured human coronary artery endothelial cells, ANG 1-7 significantly increased telomerase activity. These results indicate that ANG 1-7 dilates human microvessels, and dilation is abrogated in the presence of CAD. Furthermore, ANG 1-7 treatment is sufficient to restore the NO component of FMD in arterioles from patients with CAD in a telomerase-dependent manner. CONCLUSIONS ANG 1-7 exerts vasoprotection in the human microvasculature via modulation of telomerase activity.
Collapse
Affiliation(s)
- Matthew J Durand
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Natalya S Zinkevich
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Michael Riedel
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - David D Gutterman
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Victoria L Nasci
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Valerie K Salato
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - John B Hijjawi
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Charles F Reuben
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Paula E North
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Andreas M Beyer
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.).
| |
Collapse
|
36
|
Abstract
Angiotensin (Ang)-(1-7) is recognized as a new bioactive peptide in renin-angiotensin system (RAS). Ang-(1-7) is a counter-regulatory mediator of Ang-II which appears to be protective against cardiovascular disease. Recent studies have found that Ang-(1-7) played an important role in reducing smooth muscle cell proliferation and migration, improving endothelial function and regulating lipid metabolism, leading to inhibition of atherosclerotic lesions and increase of plaque stability. Although clinical application of Ang-(1-7) is restricted due to its pharmacokinetic properties, identification of stabilized compounds, including more stable analogues and specific delivery compounds, has enabled clinical application of Ang-(1-7). In this review, we discussed recent findings concerning the biological role of Ang-(1-7) and related mechanism during atherosclerosis development. In addition, we highlighted the perspective to develop therapeutic strategies using Ang-(1-7) to treat atherosclerosis.
Collapse
|
37
|
Boegehold MA, Drenjancevic I, Lombard JH. Salt, Angiotensin II, Superoxide, and Endothelial Function. Compr Physiol 2015; 6:215-54. [PMID: 26756632 DOI: 10.1002/cphy.c150008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Proper function of the vascular endothelium is essential for cardiovascular health, in large part due to its antiproliferative, antihypertrophic, and anti-inflammatory properties. Crucial to the protective role of the endothelium is the production and liberation of nitric oxide (NO), which not only acts as a potent vasodilator, but also reduces levels of reactive oxygen species, including superoxide anion (O2•-). Superoxide anion is highly injurious to the vasculature because it not only scavenges NO molecules, but has other damaging effects, including direct oxidative disruption of normal signaling mechanisms in the endothelium and vascular smooth muscle cells. The renin-angiotensin system plays a crucial role in the maintenance of normal blood pressure. This function is mediated via the peptide hormone angiotensin II (ANG II), which maintains normal blood volume by regulating Na+ excretion. However, elevation of ANG II above normal levels increases O2•- production, promotes oxidative stress and endothelial dysfunction, and plays a major role in multiple disease conditions. Elevated dietary salt intake also leads to oxidant stress and endothelial dysfunction, but these occur in the face of salt-induced ANG II suppression and reduced levels of circulating ANG II. While the effects of abnormally high levels of ANG II have been extensively studied, far less is known regarding the mechanisms of oxidant stress and endothelial dysfunction occurring in response to chronic exposure to abnormally low levels of ANG II. The current article focuses on the mechanisms and consequences of this less well understood relationship among salt, superoxide, and endothelial function.
Collapse
Affiliation(s)
| | - Ines Drenjancevic
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Julian H Lombard
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
38
|
Zhang Y, Liu J, Luo JY, Tian XY, Cheang WS, Xu J, Lau CW, Wang L, Wong WT, Wong CM, Lan HY, Yao X, Raizada MK, Huang Y. Upregulation of Angiotensin (1-7)-Mediated Signaling Preserves Endothelial Function Through Reducing Oxidative Stress in Diabetes. Antioxid Redox Signal 2015; 23:880-92. [PMID: 25867182 PMCID: PMC4617412 DOI: 10.1089/ars.2014.6070] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS Angiotensin-converting enzyme 2 (ACE2)-angiotensin (1-7) [Ang (1-7)]-Mas constitutes the vasoprotective axis and is demonstrated to antagonize the vascular pathophysiological effects of the classical renin-angiotensin system. We sought to study the hypothesis that upregulation of ACE2-Ang (1-7) signaling protects endothelial function through reducing oxidative stress that would result in beneficial outcome in diabetes. RESULTS Ex vivo treatment with Ang (1-7) enhanced endothelium-dependent relaxation (EDR) in renal arteries from diabetic patients. Both Ang (1-7) infusion via osmotic pump (500 ng/kg/min) for 2 weeks and exogenous ACE2 overexpression mediated by adenoviral ACE2 via tail vein injection (10(9) pfu/mouse) rescued the impaired EDR and flow-mediated dilatation (FMD) in db/db mice. Diminazene aceturate treatment (15 mg/kg/day) activated ACE2, increased the circulating Ang (1-7) level, and augmented EDR and FMD in db/db mouse arteries. In addition, activation of the ACE2-Ang (1-7) axis reduced reactive oxygen species (ROS) overproduction determined by dihydroethidium staining, CM-H2DCFDA fluorescence imaging, and chemiluminescence assay in db/db mouse aortas and also in high-glucose-treated endothelial cells. Pharmacological benefits of ACE2-Ang (1-7) upregulation on endothelial function were confirmed in ACE2 knockout (ACE2 KO) mice both ex vivo and in vitro. INNOVATION We elucidate that the ACE2-Ang (1-7)-Mas axis serves as an important signal pathway in endothelial cell protection in diabetic mice, especially in diabetic human arteries. CONCLUSION Endogenous ACE2-Ang (1-7) activation or ACE2 overexpression preserves endothelial function in diabetic mice through increasing nitric oxide bioavailability and inhibiting oxidative stress, suggesting the therapeutic potential of ACE2-Ang(1-7) axis activation against diabetic vasculopathy. Antioxid.
Collapse
Affiliation(s)
- Yang Zhang
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China .,2 Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Jian Liu
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China .,2 Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Jiang-Yun Luo
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China .,2 Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Xiao Yu Tian
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Wai San Cheang
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China .,2 Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Jian Xu
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China .,3 School of Life Sciences, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Chi Wai Lau
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Li Wang
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China .,2 Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Wing Tak Wong
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China .,4 Department of Cardiovascular Sciences, Houston Methodist Research Institute , Houston, Texas
| | - Chi Ming Wong
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China .,2 Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Hui Yao Lan
- 5 Department of Medicine and Therapeutics, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Xiaoqiang Yao
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China
| | - Mohan K Raizada
- 6 Department of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Yu Huang
- 1 Institute of Vascular Medicine, Chinese University of Hong Kong , Hong Kong SAR, China .,2 Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong , Hong Kong SAR, China
| |
Collapse
|
39
|
Feng W, Ying WZ, Aaron KJ, Sanders PW. Transforming growth factor-β mediates endothelial dysfunction in rats during high salt intake. Am J Physiol Renal Physiol 2015; 309:F1018-25. [PMID: 26447221 DOI: 10.1152/ajprenal.00328.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/06/2015] [Indexed: 01/08/2023] Open
Abstract
Endothelial dysfunction has been shown to be predictive of subsequent cardiovascular events and death. Through a mechanism that is incompletely understood, increased dietary salt intake promotes endothelial dysfunction in healthy, salt-resistant humans. The present study tested the hypothesis that dietary salt-induced transforming growth factor (TGF)-β promoted endothelial dysfunction and salt-dependent changes in blood pressure (BP). Sprague-Dawley rats that received diets containing 0.3% NaCl [low salt (LS)] or 8.0% NaCl [high salt (HS)] were treated with vehicle or SB-525334, a specific inhibitor of TGF-β receptor I/activin receptor-like kinase 5, beginning on day 5. BP was monitored using radiotelemetry in four groups of rats (LS, LS + SB-525334, HS, and HS + SB-525334) for up to 14 days. By day 14 of the study, mean daytime systolic BP and mean pulse pressure of the HS group treated with vehicle was greater than those in the other three groups; mean daytime systolic BP and pulse pressure of the HS + SB-525334 group did not differ from the LS and LS + SB-525334-treated groups. Whereas mean systolic BP, mean diastolic BP, and mean arterial pressure did not differ among the groups on the seventh day of the study, endothelium-dependent vasorelaxation was impaired specifically in the HS group; treatment with the activin receptor-like kinase 5 inhibitor prevented the dietary HS intake-induced increases in phospho-Smad2 (Ser(465/467)) and NADPH oxidase-4 in endothelial lysates and normalized endothelial function. These findings suggest that HS-induced endothelial dysfunction and the development of salt-dependent increases in BP were related to endothelial TGF-β signaling.
Collapse
Affiliation(s)
- Wenguang Feng
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wei-Zhong Ying
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kristal J Aaron
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Paul W Sanders
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
40
|
AVE 0991 attenuates cardiac hypertrophy through reducing oxidative stress. Biochem Biophys Res Commun 2015; 474:621-625. [PMID: 26403967 DOI: 10.1016/j.bbrc.2015.09.050] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 09/09/2015] [Indexed: 11/21/2022]
Abstract
AVE 0991, the nonpeptide angiotensin-(1-7) (Ang-(1-7)) analog, is recognized as having beneficial cardiovascular effects. However, the mechanisms have not been fully elucidated. This study was designed to investigate the effects of AVE 0991 on cardiac hypertrophy and the mechanisms involved. Mice were underwent aortic banding to induce cardiac hypertrophy followed by the administration of AVE 0991 (20 mg kg·day (-1)) for 4 weeks. It was shown that AVE 0991 reduced left ventricular hypertrophy and improved heart function, characterized by decreases in left ventricular weight and left ventricular end-diastolic diameter, and increases in ejection fraction. Moreover, AVE 0991 significantly down-regulated mean myocyte diameter and attenuate the gene expression of the hypertrophic markers. Furthermore, AVE 0991 inhibited the expression of NOX 2 and NOX 4, meaning that AVE 0991 reduced oxidative stress of cardiac hypertrophy mice. Our data showed that AVE 0991 treatment could attenuate cardiac hypertrophy and improve heart function, which may be due to reduce oxidative stress.
Collapse
|
41
|
Carver KA, Smith TL, Gallagher PE, Tallant EA. Angiotensin-(1-7) prevents angiotensin II-induced fibrosis in cremaster microvessels. Microcirculation 2015; 22:19-27. [PMID: 25079175 DOI: 10.1111/micc.12159] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/29/2014] [Indexed: 01/19/2023]
Abstract
OBJECTIVE The effect of the heptapeptide hormone Ang-(1-7) on microvascular fibrosis in rats with Ang II-induced hypertension was investigated, since vascular fibrosis/remodeling plays a prominent role in hypertension-induced end-organ damage and Ang-(1-7) inhibits vascular growth and fibrosis. METHODS Fibrosis of cremaster microvessels was studied in male Lewis rats infused with Ang II and/or Ang-(1-7). RESULTS Ang II elevated systolic blood pressure by approximately 40 mmHg, while blood pressure was not changed by Ang-(1-7). Ang II increased perivascular fibrosis surrounding 20-50 μm arterioles as well as interstitial fibrosis; coadministration of Ang-(1-7) prevented the increases in fibrosis. The fibrotic factor CTGF and phospho-Smad 2/3, which upregulates CTGF, were increased by Ang II; this effect was prevented by coadministration of Ang-(1-7). Although TGF-β phosphorylates Smad 2/3, TGF-β was no different among treatment groups. In contrast, Ang II increased the MAP kinase phospho-ERK1/2, which also phosphorylates Smad; p-ERK was reduced by Ang-(1-7). Ang-(1-7), in the presence or absence of Ang II, upregulated the MAP kinase phosphatase DUSP1. CONCLUSIONS These results suggest that Ang-(1-7) increases DUSP1 to reduce MAP kinase/Smad/CTGF signaling and decrease fibrosis in resistance arterioles, to attenuate end-organ damage associated with chronic hypertension.
Collapse
Affiliation(s)
- Kyle A Carver
- Hypertension and Vascular Research Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | | | | |
Collapse
|
42
|
Cabello-Verrugio C, Morales MG, Rivera JC, Cabrera D, Simon F. Renin-angiotensin system: an old player with novel functions in skeletal muscle. Med Res Rev 2015; 35:437-63. [PMID: 25764065 DOI: 10.1002/med.21343] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Skeletal muscle is a tissue that shows the most plasticity in the body; it can change in response to physiological and pathological stimuli. Among the diseases that affect skeletal muscle are myopathy-associated fibrosis, insulin resistance, and muscle atrophy. A common factor in these pathologies is the participation of the renin-angiotensin system (RAS). This system can be functionally separated into the classical and nonclassical RAS axis. The main components of the classical RAS pathway are angiotensin-converting enzyme (ACE), angiotensin II (Ang-II), and Ang-II receptors (AT receptors), whereas the nonclassical axis is composed of ACE2, angiotensin 1-7 [Ang (1-7)], and the Mas receptor. Hyperactivity of the classical axis in skeletal muscle has been associated with insulin resistance, atrophy, and fibrosis. In contrast, current evidence supports the action of the nonclassical RAS as a counter-regulator axis of the classical RAS pathway in skeletal muscle. In this review, we describe the mechanisms involved in the pathological effects of the classical RAS, advances in the use of pharmacological molecules to inhibit this axis, and the beneficial effects of stimulation of the nonclassical RAS pathway on insulin resistance, atrophy, and fibrosis in skeletal muscle.
Collapse
Affiliation(s)
- Claudio Cabello-Verrugio
- Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas & Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | | | | | | | | |
Collapse
|
43
|
Mendonça L, Mendes-Ferreira P, Bento-Leite A, Cerqueira R, Amorim MJ, Pinho P, Brás-Silva C, Leite-Moreira AF, Castro-Chaves P. Angiotensin-(1–7) Modulates Angiotensin II-Induced Vasoconstriction in Human Mammary Artery. Cardiovasc Drugs Ther 2014; 28:513-22. [DOI: 10.1007/s10557-014-6555-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
44
|
The Ang-(1-7)/Mas-1 axis attenuates the expression and signalling of TGF-β1 induced by AngII in mouse skeletal muscle. Clin Sci (Lond) 2014; 127:251-64. [PMID: 24588264 DOI: 10.1042/cs20130585] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
AngII (angiotensin II) induces pathological conditions such as fibrosis in skeletal muscle. In this process, AngII increases ROS (reactive oxygen species) and induces a biphasic phosphorylation of p38 MAPK (mitogen-activated protein kinase). In addition, AngII stimulates the expression and production of TGF (transforming growth factor)-β1 via a mechanism dependent on ROS production mediated by NADPH oxidase (NOX) and p38 MAPK activation. In the present study, we investigated whether Ang-(1-7) [angiotensin-(1-7)], through the Mas-1 receptor, can counteract the signalling induced by AngII in mouse skeletal muscle and cause a decrease in the expression and further activity of TGF-β1 in skeletal muscle cells. Our results show that Ang-(1-7) decreased the expression of TGF-β1 induced by AngII in a dose-dependent manner. In addition, we observed that Ang-(1-7) prevented the increase in TGF-β1 expression induced by AngII, ROS production dependent on NOX and the early phase of p38 MAPK phosphorylation. Interestingly, Ang-(1-7) also prevented the late phase of p38 MAPK phosphorylation, Smad-2 phosphorylation and Smad-4 nuclear translocation, an increase in transcriptional activity, as determined using the p3TP-lux reporter, and fibronectin levels, all of which are dependent on the TGF-β1 levels induced by AngII. We also demonstrated that Ang-(1-7) prevented the increase in TGF-β1, fibronectin and collagen content in the diaphragm of mice infused with AngII. All of these effects were reversed by the administration of A779, indicating the participation of Mas-1. In conclusion, our findings support the hypothesis that Ang-(1-7) decreases the expression and further biological activity of TGF-β1 induced by AngII in vitro and in vivo.
Collapse
|
45
|
Affiliation(s)
- Robson Augusto Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, CEP 31270-910, Brazil.
| |
Collapse
|
46
|
Haber PK, Ye M, Wysocki J, Maier C, Haque SK, Batlle D. Angiotensin-converting enzyme 2-independent action of presumed angiotensin-converting enzyme 2 activators: studies in vivo, ex vivo, and in vitro. Hypertension 2014; 63:774-82. [PMID: 24446061 DOI: 10.1161/hypertensionaha.113.02856] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Angiotensin (Ang)-converting enzyme 2 (ACE2) is a key enzyme in the metabolism of Ang II. XNT (1-[(2-dimethylamino)ethylamino]-4-(hydroxymethyl)-7-[(4-methylphenyl) sulfonyl oxy]-9H-xanthene-9-one) and diminazene have been reported to exert various organ-protective effects, which are attributed to the activation of ACE2. To test the effect of these compounds, we studied Ang II degradation in vivo and in vitro as well as their effect on ACE2 activity in vivo and in vitro. In a model of Ang II-induced acute hypertension, blood pressure (BP) recovery was markedly enhanced by XNT (slope with XNT, -3.26±0.2 versus -1.6±0.2 mm Hg/min without XNT; P<0.01). After Ang II infusion, neither plasma nor kidney ACE2 activity was affected by XNT. Plasma Ang II and Ang (1-7) levels also were not significantly affected by XNT. The BP-lowering effect of XNT seen in wild-type animals was also observed in ACE2 knockout mice (slope with XNT, -3.09±0.30 versus -1.28±0.22 mm Hg/min without XNT; P<0.001). These findings show that the BP-lowering effect of XNT in Ang II-induced hypertension cannot be because of the activation of ACE2. In vitro and ex vivo experiments in both mice and rat kidney confirmed a lack of enhancement of ACE2 enzymatic activity by XNT and diminazene. Moreover, Ang II degradation in vitro and ex vivo was unaffected by XNT and diminazene. We conclude that the biological effects of these compounds are ACE2-independent and should not be attributed to the activation of this enzyme.
Collapse
Affiliation(s)
- Philipp K Haber
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, 320 E Superior, Chicago, IL 60611.
| | | | | | | | | | | |
Collapse
|
47
|
Beyer AM, Raffai G, Weinberg BD, Fredrich K, Rodgers MS, Geurts AM, Jacob HJ, Dwinell MR, Lombard JH. Amelioration of salt-induced vascular dysfunction in mesenteric arteries of Dahl salt-sensitive rats by missense mutation of extracellular superoxide dismutase. Am J Physiol Heart Circ Physiol 2013; 306:H339-47. [PMID: 24322611 DOI: 10.1152/ajpheart.00619.2012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Superoxide dismutase (SOD) enzymes, including extracellular SOD (ecSOD), are important for scavenging superoxide radicals (O2(·-)) in the vasculature. This study investigated vascular control in rats [SS-Sod(3m1Mcwi) (ecSOD(E124D))] with a missense mutation that alters a single amino acid (E124D) of ecSOD that produces a malfunctioning protein in the salt-sensitive (Dahl SS) genetic background. We hypothesized that this mutation would exacerbate endothelial dysfunction due to elevated vascular O2(·-) levels in SS, even under normal salt (NS; 0.4% NaCl) conditions. Aortas of ecSOD(E124D) rats fed standard rodent chow showed enhanced sensitivity to phenylephrine and reduced relaxation to acetylcholine (ACh) vs. SS rats. Endothelium-dependent dilation to ACh was unaffected by the mutation in small mesenteric arteries of ecSOD(E124D) rats fed NS diet, and mesenteric arteries of ecSOD(E124D) rats were protected from endothelial dysfunction during short-term (3-5 days) high-salt (HS; 4% NaCl) diet. ACh-induced dilation of mesenteric arteries of ecSOD(E124D) rats and SS rats fed NS diet was inhibited by N(G)-nitro-l-arginine methyl ester and/or by H2O2 scavenging with polyethylene glycol-catalase at higher concentrations of ACh. Total SOD activity was significantly higher in ecSOD(E124D) rats vs. SS controls fed HS diet, most likely reflecting a compensatory response to loss of a functional ecSOD isoform. These findings indicate that, contrary to its effect in the aorta, this missense mutation of ecSOD in the SS rat genome has no negative effect on vascular function in small resistance arteries, but instead protects against salt-induced endothelial dysfunction, most likely via compensatory mechanisms involving an increase in total SOD activity.
Collapse
Affiliation(s)
- Andreas M Beyer
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Boegehold MA. The effect of high salt intake on endothelial function: reduced vascular nitric oxide in the absence of hypertension. J Vasc Res 2013; 50:458-67. [PMID: 24192502 DOI: 10.1159/000355270] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 08/21/2013] [Indexed: 11/19/2022] Open
Abstract
Within the last 25 years, it has become increasingly clear that high dietary salt intake represents a risk factor for the development of cardiovascular disease that is independent of its well-known ability to increase arterial pressure in some individuals. Studies in normotensive experimental animals and human subjects have revealed that a key feature of this pressure-independent effect of dietary salt is a profound reduction in vascular nitric oxide (NO) bioavailability that limits endothelium-dependent dilation. This reduction in NO is strongly associated with increased levels of reactive oxygen species (ROS) generated by NAD(P)H oxidase, xanthine oxidase or uncoupled endothelial NO synthase within the vascular wall, leading not only to scavenging of NO but also to disruption of some signaling pathways that mediate its production. The mechanistic link between high salt intake and elevated levels of enzymatically generated ROS in the peripheral vasculature is not clear, but a reduction in circulating angiotensin II may play a key role in this regard. Additional studies are needed to further elucidate the mechanisms, both at the systemic level and within the vascular wall, that trigger these salt-induced deficits in endothelial function, and to further clarify how the attendant loss of NO may disrupt tissue blood flow regulation and ultimately lead to adverse cardiovascular events.
Collapse
Affiliation(s)
- Matthew A Boegehold
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, W.Va., USA
| |
Collapse
|
49
|
Lu J, Jiang T, Wu L, Gao L, Wang Y, Zhou F, Zhang S, Zhang Y. The expression of angiotensin-converting enzyme 2-angiotensin-(1-7)-Mas receptor axis are upregulated after acute cerebral ischemic stroke in rats. Neuropeptides 2013; 47:289-95. [PMID: 24090950 DOI: 10.1016/j.npep.2013.09.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 08/10/2013] [Accepted: 09/09/2013] [Indexed: 01/23/2023]
Abstract
There is now unequivocal evidence that the angiotensin-converting enzyme 2(ACE2)-Ang-(1-7)-Mas axis is a key component of the renin-angiotensin system (RAS) cascade, which is closely correlated with ischemic insult occurrence. Our previous studies demonstrated that the Ang-(1-7), was an active member of the brain RAS. However, the ACE2-Ang-(1-7)-Mas axis expression after cerebral ischemic injury are currently unclear. In the present study, we investigated the time course of ACE2-Ang-(1-7) and Mas receptor expression in the acute stage of cerebral ischemic stroke. The content of Ang-(1-7) in ischemic tissues and blood serum was measured by specific EIA kits. Real-time PCR and western blot were used to determine messenger RNA (mRNA) and protein levels of the ACE2 and Mas. The cerebral ischemic lesion resulted in a significant increase of regional cerebral and circulating Ang-(1-7) at 6-48 h compared with sham operation group following focal ischemic stroke (12h: 7.276±0.320 ng/ml vs. 2.466±0.410 ng/ml, serum; 1.024±0.056 ng/mg vs. 0.499±0.032, brain) (P<0.05). Both ACE2 and Mas expression were markedly enhanced compared to the control in the ischemic tissues (P<0.05). Mas immunopositive neurons were also seen stronger expression in the ischemic cortex (19.167±2.858 vs. 7.833±2.483) (P<0.05). The evidence collected in our present study will indicate that, ACE2-Ang-(1-7)-Mas axis are upregulated after acute ischemic stroke and would play a pivotal role in the regulation of acute neuron injury in ischemic cerebrovascular diseases.
Collapse
Affiliation(s)
- Jie Lu
- Department of Neurology, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, PR China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Varagic J, Ahmad S, VonCannon JL, Moniwa N, Brosnihan KB, Wysocki J, Batlle D, Ferrario CM. Predominance of AT(1) blockade over mas-mediated angiotensin-(1-7) mechanisms in the regulation of blood pressure and renin-angiotensin system in mRen2.Lewis rats. Am J Hypertens 2013; 26:583-90. [PMID: 23459599 DOI: 10.1093/ajh/hps090] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND We investigated whether the antihypertensive actions of the angiotensin II (Ang II) receptor (AT(1)-R) blocker, olmesartan medoxomil, may in part be mediated by increased Ang-(1-7) in the absence of significant changes in plasma Ang II. METHODS mRen2.Lewis congenic hypertensive rats were administered either a vehicle (n = 14) or olmesartan (0.5 mg/kg/day; n = 14) by osmotic minipumps. Two weeks later, rats from both groups were further randomized to receive either the mas receptor antagonist A-779 (0.5 mg/kg/day; n = 7 per group) or its vehicle (n = 7 per group) for the next 4 weeks. Blood pressure was monitored by telemetry, and circulating and tissue components of the renin-angiotensin system (RAS) were measured at the completion of the experiments. RESULTS Antihypertensive effects of olmesartan were associated with an increase in plasma renin concentration, plasma Ang I, Ang II, and Ang-(1-7), whereas serum aldosterone levels and kidney Ang II content were reduced. Preserved Ang-(1-7) content in kidneys was associated with increases of ACE2 protein but not activity and no changes on serum and kidney ACE activity. There was no change in cardiac peptide levels after olmesartan treatment. The antihypertensive effects of olmesartan were not altered by concomitant administration of the Ang-(1-7) receptor antagonist except for a mild further increase in plasma renin concentration. CONCLUSIONS Our study highlights the independent regulation of RAS among plasma, heart, and kidney tissue in response to AT(1)-R blockade. Ang-(1-7) through the mas receptor does not mediate long-term effects of olmesartan besides counterbalancing renin release in response to AT(1)-R blockade.
Collapse
Affiliation(s)
- Jasmina Varagic
- Hypertension and Vascular Research Center, Wake Forest University, Winston-Salem, NC, USA.
| | | | | | | | | | | | | | | |
Collapse
|