51
|
Chinnakkannu P, Reese C, Gaspar JA, Panneerselvam S, Pleasant-Jenkins D, Mukherjee R, Baicu C, Tourkina E, Hoffman S, Kuppuswamy D. Suppression of angiotensin II-induced pathological changes in heart and kidney by the caveolin-1 scaffolding domain peptide. PLoS One 2018; 13:e0207844. [PMID: 30576317 PMCID: PMC6303044 DOI: 10.1371/journal.pone.0207844] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/07/2018] [Indexed: 01/15/2023] Open
Abstract
Dysregulation of the renin-angiotensin system leads to systemic hypertension and maladaptive fibrosis in various organs. We showed recently that myocardial fibrosis and the loss of cardiac function in mice with transverse aortic constriction (TAC) could be averted by treatment with the caveolin-1 scaffolding domain (CSD) peptide. Here, we used angiotensin II (AngII) infusion (2.1 mg/kg/day for 2 wk) in mice as a second model to confirm and extend our observations on the beneficial effects of CSD on heart and kidney disease. AngII caused cardiac hypertrophy (increased heart weight to body weight ratio (HW/BW) and cardiomyocyte cross-sectional area); fibrosis in heart and kidney (increased levels of collagen I and heat shock protein-47 (HSP47)); and vascular leakage (increased levels of IgG in heart and kidney). Echocardiograms of AngII-infused mice showed increased left ventricular posterior wall thickness (pWTh) and isovolumic relaxation time (IVRT), and decreased ejection fraction (EF), stroke volume (SV), and cardiac output (CO). CSD treatment (i.p. injections, 50 μg/mouse/day) of AngII-infused mice significantly suppressed all of these pathological changes in fibrosis, hypertrophy, vascular leakage, and ventricular function. AngII infusion increased β1 and β3 integrin levels and activated Pyk2 in both heart and kidney. These changes were also suppressed by CSD. Finally, bone marrow cell (BMC) isolated from AngII-infused mice showed hyper-migration toward SDF1. When AngII-infused mice were treated with CSD, BMC migration was reduced to the basal level observed in cells from control mice. Importantly, CSD did not affect the AngII-induced increase in blood pressure (BP), indicating that the beneficial effects of CSD were not mediated via normalization of BP. These results strongly indicate that CSD suppresses AngII-induced pathological changes in mice, suggesting that CSD can be developed as a treatment for patients with hypertension and pressure overload-induced heart failure.
Collapse
Affiliation(s)
- Panneerselvam Chinnakkannu
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Charles Reese
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | | | - Saraswathi Panneerselvam
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Dorea Pleasant-Jenkins
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Rupak Mukherjee
- Division of Cardiothoracic Surgery, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Catalin Baicu
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Elena Tourkina
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Stanley Hoffman
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Dhandapani Kuppuswamy
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, South Carolina, United States of America
| |
Collapse
|
52
|
Sharma N, Anders HJ, Gaikwad AB. Fiend and friend in the renin angiotensin system: An insight on acute kidney injury. Biomed Pharmacother 2018; 110:764-774. [PMID: 30554115 DOI: 10.1016/j.biopha.2018.12.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 02/06/2023] Open
Abstract
Besides assisting the maintenance of blood pressure and sodium homeostasis, the renin-angiotensin system (RAS) plays a pivotal role in pathogenesis of acute kidney injury (AKI). The RAS is equipped with two arms i) the pressor arm composed of Angiotensin II (Ang II)/Angiotensin converting enzyme (ACE)/Angiotensin II type 1 receptor (AT1R) also called conventional RAS, and ii) the depressor arm consisting of Angiotensin (1-7) (Ang 1-7)/Angiotensin converting enzyme 2 (ACE2)/MasR known as non-conventional RAS. Activation of conventional RAS triggers oxidative stress, inflammatory, hypertrophic, apoptotic, and pro-fibrotic signaling cascades which promote AKI. The preclinical and clinical studies have reported beneficial as well as deleterious effects of RAS blockage either by angiotensin receptor blocker or ACE inhibitor in AKI. On the contrary, the depressor arm opposes the conventional RAS, has beneficial effects on the kidney but has been less explored in pathogenesis of AKI. This review focuses on significance of RAS in pathogenesis of AKI and provides better understanding of novel and possible therapeutic approaches to combat AKI.
Collapse
Affiliation(s)
- Nisha Sharma
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333 031, India
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Internal Medicine IV, University Hospital of the Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333 031, India.
| |
Collapse
|
53
|
Cardiac hypertrophy in chronic kidney disease—role of Aldosterone and FGF23. RENAL REPLACEMENT THERAPY 2018. [DOI: 10.1186/s41100-018-0152-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
54
|
Hao Q, Dong X, Chen X, Yan F, Wang X, Shi H, Dong B. Angiotensin-Converting Enzyme 2 Inhibits Angiotensin II-Induced Abdominal Aortic Aneurysms in Mice. Hum Gene Ther 2018; 29:1387-1395. [PMID: 28142259 DOI: 10.1089/hum.2016.144] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recent studies have demonstrated that angiotensin-converting enzyme 2 (ACE2) plays an important role in the pathogenesis of abdominal aortic aneurysms (AAAs). However, few studies have reported the direct effect of ACE2 overexpression on the aneurysm. This study hypothesized that the overexpression of ACE2 may prevent the pathogenesis of aneurysms by decreasing RAS activation. Thirty-nine mice were randomly assigned to three groups (n = 13 in each group): the Ad.ACE2 group, the Ad.EGFP group, and a control group. After 8 weeks of treatment, abdominal aortas with AAAs were obtained for hematoxylin and eosin staining, Verhoeff Van Gieson staining, immunohistochemistry, and Western blotting. The incidence and severity of AAAs, macrophage infiltration, and MMP protein expression were all recorded. The results showed that ACE2 gene transfer significantly decreased the occurrence of AAAs and inhibited AAA formation in ApoE-/- mice by inhibiting the inflammatory response and MMP activation, and the mechanisms may involve decreased ERK and Ang II-nuclear factor kappa B signaling pathways.
Collapse
Affiliation(s)
- QingQing Hao
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, P.R. China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Shandong University, Shandong, P.R. China.,Department of Physiopathology, Fenyang College Shanxi Medical University, Fenyang, P.R. China
| | - XueFei Dong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Xu Chen
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, P.R. China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Shandong University, Shandong, P.R. China
| | - Feng Yan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Shandong University, Shandong, P.R. China
| | - Xiaoyu Wang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, P.R. China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Shandong University, Shandong, P.R. China
| | - HaiShui Shi
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, P.R. China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Shandong University, Shandong, P.R. China
| |
Collapse
|
55
|
Zhang Q, Cong M, Wang N, Li X, Zhang H, Zhang K, Jin M, Wu N, Qiu C, Li J. Association of angiotensin-converting enzyme 2 gene polymorphism and enzymatic activity with essential hypertension in different gender: A case-control study. Medicine (Baltimore) 2018; 97:e12917. [PMID: 30335025 PMCID: PMC6211892 DOI: 10.1097/md.0000000000012917] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) plays an important role in the development of essential hypertension (EH). The aim of this study was to investigate the relationship of ACE2 gene polymorphisms and enzymatic activity with EH in the northeastern Chinese Han population. 34 single-nucleotide polymorphism (SNP) loci of ACE2 were detected in 1024 EH patients and 956 normotensive (NT) controls by Sequenom Mass-ARRAY RS1000. Five SNPs (rs1514283, rs4646155, rs4646176, rs2285666, and rs879922) in ACE2 gene were determined to significantly associate with EH in female participants, while no SNP locus was linked to male group. Specifically, it was the first time to report that rs4646155 was significantly associated with EH in females. Furthermore, the correlation between ACE2 activity and clinical parameters were performed by Pearson correlation analysis in EH patients. We found that the ACE2 activity level was negatively correlated with body mass index (BMI), DBP, and pulse pressure, and significantly positively with ACE2 concentration, blood glucose and estrogen level in female EH patients. These results demonstrated that the genetic variants of ACE2 played vital roles in the development of EH. And the serum ACE2 activity can predict the development of cardiac dysfunction in EH patients.
Collapse
Affiliation(s)
- Qi Zhang
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, Heilongjiang Province
| | - Mingyu Cong
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, Heilongjiang Province
| | - Ningning Wang
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, Heilongjiang Province
| | - Xueyan Li
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, Heilongjiang Province
| | - Hao Zhang
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, Heilongjiang Province
| | - Keyong Zhang
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, Heilongjiang Province
| | - Ming Jin
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, Heilongjiang Province
| | - Nan Wu
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, Heilongjiang Province
| | - Changchun Qiu
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, Heilongjiang Province
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, P. R. China
| | - Jingping Li
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, Heilongjiang Province
| |
Collapse
|
56
|
Micakovic T, Papagiannarou S, Clark E, Kuzay Y, Abramovic K, Peters J, Sticht C, Volk N, Fleming T, Nawroth P, Hammes HP, Alenina N, Gröne HJ, Hoffmann SC. The angiotensin II type 2 receptors protect renal tubule mitochondria in early stages of diabetes mellitus. Kidney Int 2018; 94:937-950. [PMID: 30190172 DOI: 10.1016/j.kint.2018.06.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 05/23/2018] [Accepted: 06/08/2018] [Indexed: 12/19/2022]
Abstract
Diabetic nephropathy correlates more closely to defective mitochondria and increased oxidative stress in the kidney than to hyperglycemia. A key driving factor of diabetic nephropathy is angiotensin II acting via the G-protein-coupled cell membrane type 1 receptor. The present study aimed to investigate the role of the angiotensin II type 2 receptor (AT2R) at the early stages of diabetic nephropathy. Using receptor binding studies and immunohistochemistry we found that the mitochondria in renal tubules contain high-affinity AT2Rs. Increased renal mitochondrial AT2R density by transgenic overexpression was associated with reduced superoxide production of isolated mitochondria from non-diabetic rats. Streptozotocin-induced diabetes (28 days) caused a drop in the ATP/oxygen ratio and an increase in the superoxide production of isolated renal mitochondria from wild-type diabetic rats. This correlated with changes in the renal expression profile and increased tubular epithelial cell proliferation. AT2R overexpression in tubular epithelial cells inhibited all diabetes-induced renal changes including a drop in mitochondrial bioenergetics efficiency, a rise in mitochondrial superoxide production, metabolic reprogramming, and increased proliferation. Thus, AT2Rs translocate to mitochondria and can contribute to reno-protective effects at early stages of diabetes. Hence, targeted AT2R overexpression in renal cells may open new avenues to develop novel types of drugs preventing diabetic nephropathy.
Collapse
Affiliation(s)
- Tamara Micakovic
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Stamatia Papagiannarou
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Euan Clark
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Yalcin Kuzay
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Katarina Abramovic
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Jörg Peters
- Institute of Physiology, University Medicine of Greifswald, Karlsburg, Germany
| | - Carsten Sticht
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Nadine Volk
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg Medical Faculty, University of Heidelberg, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Peter Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg Medical Faculty, University of Heidelberg, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Hans-Peter Hammes
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Natalia Alenina
- Max-Delbrück Center for Molecular Medicine, Cardiovascular Hormones - Berlin-Buch, Berlin, Germany
| | - Hermann-Josef Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Sigrid Christa Hoffmann
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
57
|
Liu P, Wysocki J, Souma T, Ye M, Ramirez V, Zhou B, Wilsbacher LD, Quaggin SE, Batlle D, Jin J. Novel ACE2-Fc chimeric fusion provides long-lasting hypertension control and organ protection in mouse models of systemic renin angiotensin system activation. Kidney Int 2018; 94:114-125. [PMID: 29691064 DOI: 10.1016/j.kint.2018.01.029] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 12/22/2017] [Accepted: 01/25/2018] [Indexed: 12/11/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a carboxypeptidase that potently degrades angiotensin II to angiotensin 1-7. Previous studies showed that injection of the enzymatic ectodomain of recombinant ACE2 (rACE2) markedly increases circulatory levels of ACE2 activity, and effectively lowered blood pressure in angiotensin II-induced hypertension. However, due to the short plasma half-life of rACE2, its therapeutic potential for chronic use is limited. To circumvent this, we generated a chimeric fusion of rACE2 and the immunoglobulin fragment Fc segment to increase its plasma stability. This rACE2-Fc fusion protein retained full peptidase activity and exhibited greatly extended plasma half-life in mice, from less than two hours of the original rACE2, to over a week. A single 2.5 mg/kg injection of rACE2-Fc increased the overall angiotensin II-conversion activities in blood by up to 100-fold and enhanced blood pressure recovery from acute angiotensin II induced hypertension seven days after administration. To assess rACE2-Fc given weekly on cardiac protection, we performed studies in mice continuously infused with angiotensin II for 28 days and in a Renin transgenic mouse model of hypertension. The angiotensin II infused mice achieved sustained blood pressure control and reduced cardiac hypertrophy and fibrosis. In chronic hypertensive transgenic mice, weekly injections of rACE2-Fc effectively lowered plasma angiotensin II and blood pressure. Additionally, rACE2-Fc ameliorated albuminuria, and reduced kidney and cardiac fibrosis. Thus, our chimeric fusion strategy for rACE2-Fc is suitable for future development of new renin angiotensin system-based inhibition therapies.
Collapse
Affiliation(s)
- Pan Liu
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jan Wysocki
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tomokazu Souma
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Minghao Ye
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Veronica Ramirez
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bisheng Zhou
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lisa D Wilsbacher
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Susan E Quaggin
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniel Batlle
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jing Jin
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
58
|
Oparil S, Acelajado MC, Bakris GL, Berlowitz DR, Cífková R, Dominiczak AF, Grassi G, Jordan J, Poulter NR, Rodgers A, Whelton PK. Hypertension. Nat Rev Dis Primers 2018; 4:18014. [PMID: 29565029 PMCID: PMC6477925 DOI: 10.1038/nrdp.2018.14] [Citation(s) in RCA: 567] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Systemic arterial hypertension is the most important modifiable risk factor for all-cause morbidity and mortality worldwide and is associated with an increased risk of cardiovascular disease (CVD). Fewer than half of those with hypertension are aware of their condition, and many others are aware but not treated or inadequately treated, although successful treatment of hypertension reduces the global burden of disease and mortality. The aetiology of hypertension involves the complex interplay of environmental and pathophysiological factors that affect multiple systems, as well as genetic predisposition. The evaluation of patients with hypertension includes accurate standardized blood pressure (BP) measurement, assessment of the patients' predicted risk of atherosclerotic CVD and evidence of target-organ damage, and detection of secondary causes of hypertension and presence of comorbidities (such as CVD and kidney disease). Lifestyle changes, including dietary modifications and increased physical activity, are effective in lowering BP and preventing hypertension and its CVD sequelae. Pharmacological therapy is very effective in lowering BP and in preventing CVD outcomes in most patients; first-line antihypertensive medications include angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, dihydropyridine calcium-channel blockers and thiazide diuretics.
Collapse
Affiliation(s)
- Suzanne Oparil
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, School of Medicine, The University of Alabama at Birmingham (UAB), 1720 2nd Avenue South, Birmingham, AL, 35294-0007, USA
| | | | | | - Dan R Berlowitz
- Center for Healthcare Organization and Implementation Research, Bedford Veteran Affairs Medical Center, Bedford, MA, USA
- Schools of Medicine and Public Health, Boston University, Boston, MA, USA
| | - Renata Cífková
- Center for Cardiovascular Prevention, Charles University in Prague, First Faculty of Medicine and Thomayer Hospital, Prague, Czech Republic
| | - Anna F Dominiczak
- Institute of Cardiovascular and Medical Science, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Guido Grassi
- Clinica Medica, University of Milano-Bicocca, Milan, Italy
- IRCCS Multimedica, Sesto San Giovanni, Milan, Italy
| | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center (DLR), University of Cologne, Cologne, Germany
| | - Neil R Poulter
- Imperial Clinical Trials Unit, School of Public Health, Imperial College London, London, UK
| | - Anthony Rodgers
- The George Institute for Global Health, Sydney, New South Wales, Australia
| | - Paul K Whelton
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| |
Collapse
|
59
|
Yisireyili M, Uchida Y, Yamamoto K, Nakayama T, Cheng XW, Matsushita T, Nakamura S, Murohara T, Takeshita K. Angiotensin receptor blocker irbesartan reduces stress-induced intestinal inflammation via AT1a signaling and ACE2-dependent mechanism in mice. Brain Behav Immun 2018; 69:167-179. [PMID: 29155324 DOI: 10.1016/j.bbi.2017.11.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 11/02/2017] [Accepted: 11/15/2017] [Indexed: 12/30/2022] Open
Abstract
Stress is associated with pathophysiology of both irritable bowel syndrome (IBS) and hypertension. Angiotensin receptor blockers (ARB) have anti-inflammatory properties via inhibition of angiotensin II (Ang II)/Ang II type I receptor axis (AT1). Inhibition of the classical RAS pathway is also involved in upregulation of angiotensin converting enzyme-2 (ACE2), which activates the Ang-(1-7)/Mas pathway to counteract inflammatory signaling and acts as a partner of the amino acid transporter, B0AT-1, to absorb tryptophan for regulation of microbiota-gut-brain axis. In this study, we determined the effects of ARB irbesartan on stress-induced intestinal inflammation. C57BL/6J mice were subjected to 2-week intermittent restraint stress. They were orally treated during the stress with either vehicle, 3 or 10 mg/kg/day irbesartan. Restraint stress resulted in colon inflammation with higher histological damage scores, increased expression of Nox4, TLR-4 and IL1-β, accumulation of reactive oxygen species (ROS), and activation of the ACE-angiotensin II-AT1 receptor axis. Stress also downregulated intestinal amino acid transporter, ACE2/B0AT-1, and activity of intestinal mammalian target of rapamycin (mTOR) and p70 S6 kinase (p70S6K), resulting in decrease in α-defensins, changes in intestinal microbial contents, and perturbation of tryptophan metabolism with activation of the kynurenine pathway. Administration of irbesartan inhibited activation of stress-induced AT1 pathway to reduce intestinal ROS accumulation and inflammation, restored expression of ACE2/B0AT-1, activity of mTOR and p70S6K, dysbiosis and tryptophan metabolism. Our results suggest that AT1 is a potentially suitable therapeutic target in stress-induced intestinal inflammation, and that irbesartan could be beneficially suitable for the treatment of stressed patients with IBS.
Collapse
Affiliation(s)
- Maimaiti Yisireyili
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Minimally Invasive Hernia and Abdominal Wall Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830000, Xinjiang Uygur Autonomous Region, China
| | - Yasuhiro Uchida
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koji Yamamoto
- Department of Transfusion Medicine and Cell Therapy, Saitama Medical Centre, Saitama Medical University, Kawagoe, Japan
| | - Takayuki Nakayama
- Department of Blood Transfusion, Aichi Medical University Hospital, Nagakute, Japan
| | - Xian Wu Cheng
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tadashi Matsushita
- Department of Clinical Laboratory, Nagoya University Hospital, Nagoya, Japan; Department of Blood Transfusion, Nagoya University Hospital, Nagoya, Japan
| | - Shigeo Nakamura
- Department of Pathology, Nagoya University Hospital, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kyosuke Takeshita
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Clinical Laboratory, Nagoya University Hospital, Nagoya, Japan.
| |
Collapse
|
60
|
Zheng J, Wang J, Pan H, Wu H, Ren D, Lu J. Effects of IQP, VEP and Spirulina platensis hydrolysates on the local kidney renin angiotensin system in spontaneously hypertensive rats. Mol Med Rep 2017; 16:8485-8492. [PMID: 28944898 DOI: 10.3892/mmr.2017.7602] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 06/14/2017] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the antihypertensive effects of the bioactive Spirulina platensis peptides Ile‑Gln‑Pro (IQP), Val‑Glu‑Pro (VEP), as well as Spirulina platensis hydrolysates (SH), and assessed whether the synthesis of components of the myocardial and renal local renin angiotensin system (RAS) are regulated differentially in spontaneously hypertensive rats (SHR). The SHR were administrated with IQP, VEP and SH respectively (10 mg/kg/day) for 6 weeks and received continuous monitoring of blood pressure (BP) for two more weeks. During the trial, the rats' kidney tissues were removed from these rats and collected at weeks 3, 6 and 8. The expression of the main components of local kidney RAS was measured at the mRNA levels by reverse transcription‑quantitative polymerase chain reaction, and at the protein levels by ELISA or western blotting. Oral administration of IQP, VEP and SH into SHR resulted in marked antihypertensive effects. IQP, VEP and SH decreased rats' BP by affecting the expression of local kidney RAS components via downregulating the angiotensin‑converting enzyme (ACE), Ang II and angiotensin II (Ang II) and angiotensin type‑1 receptor (AT 1), while upregulating ACE2, Ang (1‑7), Mas and AT 2. The comparisons of SH effects on local tissue RAS demonstrated that local kidney RAS regulated BP via the ACE‑Ang II‑AT 1/AT 2 axis and the ACE2‑Ang (1‑7)‑Mas axis primarily at the mRNA level, while the local myocardium RAS mainly at the protein level. This preliminary study suggests that the main components of local RAS presented different expression levels in myocardium and kidney, which is important to the development of bioactive peptides targeting for lowering BP by changing the levels of some components in local RAS in specific tissues.
Collapse
Affiliation(s)
- Jiahui Zheng
- Beijing Key Laboratory of Forest Food Process and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Jingyue Wang
- Beijing Key Laboratory of Forest Food Process and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Huanglei Pan
- Beijing Key Laboratory of Forest Food Process and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Hongli Wu
- Beijing Key Laboratory of Forest Food Process and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Difeng Ren
- Beijing Key Laboratory of Forest Food Process and Safety, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Jun Lu
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing 100015, P.R. China
| |
Collapse
|
61
|
Chen YY, Zhang P, Zhou XM, Liu D, Zhong JC, Zhang CJ, Jin LJ, Yu HM. Relationship between genetic variants of ACE2 gene and circulating levels of ACE2 and its metabolites. J Clin Pharm Ther 2017; 43:189-195. [PMID: 28895159 DOI: 10.1111/jcpt.12625] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 07/21/2017] [Accepted: 08/21/2017] [Indexed: 01/20/2023]
Abstract
WHAT IS KNOWN Angiotensin-converting enzyme 2 (ACE2) plays an important role in the development of essential hypertension (EH). Genetic factors remarkably influence circulating ACE2 level. OBJECTIVE Because heritability had remarkable effects on circulating ACE2, we designed this study to shed light on whether circulating levels of ACE2, angiotensin-(1-7) and angiotensin-(1-9) were linked to single nucleotide polymorphisms (SNPs) and haplotypes in ACE2 gene. METHODS A total of 213 patients with newly diagnosed mild to moderate EH were enrolled in the present study. Four ACE2 tag SNPs (rs2074192, rs4646171, rs4646155 and rs2106809) were genotyped, and major haplotypes consisting of these 4 SNPs were reconstructed for all subjects. Circulating levels of ACE2, angiotensin-(1-7) and angiotensin-(1-9) were measured using enzyme-linked immunosorbent assay. RESULTS In female subjects, linear regression analysis suggested that rare alleles of ACE2 rs2074192 and rs2106809 were associated with reduced circulating angiotensin-(1-7) levels (P=.007 and P=.006, respectively). ACE2 haplotype CAGC was associated with elevated circulating angiotensin-(1-7) levels (P=.03) whereas TAGT was associated with reduced circulating angiotensin-(1-7) levels in females (P<.001). Univariate linear regression analysis revealed that circulating ACE2 levels were positively associated with systolic blood pressure (P=.02), mean arterial pressure (P=.02) and serum creatinine (P<.001) in females whereas circulating ACE2 levels were positively associated with age (P<.001) and serum creatinine (P<.001) in males. WHAT IS NEW AND CONCLUSION ACE2 SNPs and haplotypes are associated with circulating angiotensin-(1-7) levels. ACE2 genetic variants may be the determinants of circulating angiotensin-(1-7) levels in hypertensive females.
Collapse
Affiliation(s)
- Y Y Chen
- Department of Cardiology, Shantou Central Hospital and Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, China
| | - P Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - X M Zhou
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - D Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - J C Zhong
- State Key Laboratory of Medical Genomics & Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - C J Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - L J Jin
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - H M Yu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
62
|
Lin S, Pan H, Wu H, Ren D, Lu J. Role of the ACE2‑Ang‑(1‑7)‑Mas axis in blood pressure regulation and its potential as an antihypertensive in functional foods (Review). Mol Med Rep 2017; 16:4403-4412. [PMID: 28791402 DOI: 10.3892/mmr.2017.7168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 06/08/2017] [Indexed: 11/05/2022] Open
Abstract
The renin‑angiotensin system (RAS) serves a critical role in blood pressure regulation and prevention of cardiovascular diseases. Efforts to develop functional foods that enhance the RAS have focused on inhibition of angiotensin‑converting enzyme (ACE) activity in the ACE‑angiotensin II (Ang II)‑Ang II type 1 receptor axis. ACE2 and the Mas receptor are important components of this axis. ACE2 catalyzes Ang II into Ang‑(1‑7), which then binds to the G‑protein‑coupled receptor Mas. In addition, it induces nitric oxide release from endothelial cells and exerts antiproliferative, vasodilatory and antihypertensive effects. The present review examined recent findings regarding the physiological and biological roles of the ACE2‑Ang‑(1‑7)‑Mas axis in the cardiovascular system, discussed potential food‑derived ACE2‑activating agents, and highlighted initiatives, based on this axis, that aim to develop functional foods for the treatment of hypertension.
Collapse
Affiliation(s)
- Shiqi Lin
- Beijing Key Laboratory of Forest Food Process and Safety, Department of Food Science and Engineering, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Huanglei Pan
- Beijing Key Laboratory of Forest Food Process and Safety, Department of Food Science and Engineering, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Hongli Wu
- Beijing Key Laboratory of Forest Food Process and Safety, Department of Food Science and Engineering, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Difeng Ren
- Beijing Key Laboratory of Forest Food Process and Safety, Department of Food Science and Engineering, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Jun Lu
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing 100015, P.R. China
| |
Collapse
|
63
|
Patel S, Rauf A, Khan H, Abu-Izneid T. Renin-angiotensin-aldosterone (RAAS): The ubiquitous system for homeostasis and pathologies. Biomed Pharmacother 2017; 94:317-325. [PMID: 28772209 DOI: 10.1016/j.biopha.2017.07.091] [Citation(s) in RCA: 348] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/10/2017] [Accepted: 07/19/2017] [Indexed: 12/22/2022] Open
Abstract
Renin-angiotensin-aldosterone system (RAAS) is a vital system of human body, as it maintains plasma sodium concentration, arterial blood pressure and extracellular volume. Kidney-secreted renin enzyme acts on its substrate to form angiotensin II, a versatile effector peptide hormone. Every organ is affected by RAAS activation and the resultant hypertension, cell proliferation, inflammation, and fibrosis. The imbalance of renin and angiotensin II can result in an overwhelming number of chronic and acute diseases. RAAS is influenced by other enzymes, hormones, pumps and signaling pathways, hence, this review discusses important facets of this system, its crosstalk with other crucial factors like estrogen, thyroid, cortisol, kallikrein-kinin system, Wnt/β-catenin signaling, and sodium-potassium pump. The nexus of RAAS with the above-discussed systems was scantily explored before. So, this review furnishes a new perspective in comprehension of inflammation diseases. It is followed by the formulation of hypotheses, which can contribute to better management of an array of pathologies plaguing mankind. Manipulation of RAAS, by bending it towards ACE2 expression can regulate endocrine functions, which can be critical for a number of pathological management. Dietary intervention can restore RAAS to normalcy.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, San Diego, 92182, USA.
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar-23561, Khyber Pakhtunkhwa, Pakistan.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan
| | - Tareq Abu-Izneid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah, P.O. Box 42, Saudi Arabia
| |
Collapse
|
64
|
Wang J, He W, Guo L, Zhang Y, Li H, Han S, Shen D. The ACE2-Ang (1-7)-Mas receptor axis attenuates cardiac remodeling and fibrosis in post-myocardial infarction. Mol Med Rep 2017; 16:1973-1981. [PMID: 28656296 PMCID: PMC5561970 DOI: 10.3892/mmr.2017.6848] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 04/19/2017] [Indexed: 12/12/2022] Open
Abstract
Myocardial remodeling serves an important role in the pathophysiology of coronary heart disease. The angiotensin-converting enzyme (ACE)2-angiotensin-(1–7) [Ang (1–7)]-Mas receptor (MasR) axis is a key regulator in myocardial remodeling and development of heart failure. To investigate how ACE2-Ang-(1–7)-MasR axis function on myocardial remodeling and cardiac fibrosis in post-myocardial infarction (MI), male Sprague-Dawley rats (weight, 200±20 g) were used to establish the model of myocardial infarction by ligating the left coronary artery. The present study suggests that telmisartan (Tel) and olmesartan (Olm) (5 mg/kg/d) can inhibit myocardial remodeling of post-myocardial infarction through the ACE2-Ang (1–7)-MasR pathway. Administration of Tel or Olm was demonstrated to significantly inhibit collagen deposition using Masson staining. In addition, telmisartan and olmesartan was indicated to antagonize angiotensin II (Ang II) and upregulate ACE2, MasR, Ang (1–7) expression in myocardial tissue using immunoassay and ELISA test, and the effect of Olm was more marked than that of Tel at the same dosage. Simultaneously, compared with the MI or Sham group, the mRNA and protein expression of ACE2, Ang II and MasR in myocardial tissue demonstrated a remarkable increase in the Olm group, when compared with the Tel group. Taken together, our data demonstrated that ACE2-Ang (1–7)-MasR axis may present a potential protective role in the development of myocardial remodeling and may provide a new target for drug development of cardiac fibrosis. In conclusion, Olm is superior to Tel in inhibiting myocardial local Ang II level reducing myocardial collagen deposition and improving myocardial remodeling by upregulating the expression of ACE2, Ang (1–7) and MasR.
Collapse
Affiliation(s)
- Juan Wang
- Department of Cardiology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830001, P.R. China
| | - Wen He
- Department of Cardiology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830001, P.R. China
| | - Liping Guo
- Department of Cardiology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830001, P.R. China
| | - Yin Zhang
- Department of Cardiology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830001, P.R. China
| | - Hui Li
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830001, P.R. China
| | - Suxia Han
- Department of Cardiology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830001, P.R. China
| | - Difei Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
65
|
Wysocki J, Ye M, Khattab AM, Fogo A, Martin A, David NV, Kanwar Y, Osborn M, Batlle D. Angiotensin-converting enzyme 2 amplification limited to the circulation does not protect mice from development of diabetic nephropathy. Kidney Int 2017; 91:1336-1346. [PMID: 27927599 PMCID: PMC5429993 DOI: 10.1016/j.kint.2016.09.032] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/23/2016] [Accepted: 09/15/2016] [Indexed: 12/14/2022]
Abstract
Blockers of the renin-angiotensin system are effective in the treatment of experimental and clinical diabetic nephropathy. An approach different from blocking the formation or action of angiotensin II (1-8) that could also be effective involves fostering its degradation. Angiotensin-converting enzyme 2 (ACE2) is a monocarboxypeptidase that cleaves angiotensin II (1-8) to form angiotensin (1-7). Therefore, we examined the renal effects of murine recombinant ACE2 in mice with streptozotocin-induced diabetic nephropathy as well as that of amplification of circulating ACE2 using minicircle DNA delivery prior to induction of experimental diabetes. This delivery resulted in a long-term sustained and profound increase in serum ACE2 activity and enhanced ability to metabolize an acute angiotensin II (1-8) load. In mice with streptozotocin-induced diabetes pretreated with minicircle ACE2, ACE2 protein in plasma increased markedly and this was associated with a more than 100-fold increase in serum ACE2 activity. However, minicircle ACE2 did not result in changes in urinary ACE2 activity as compared to untreated diabetic mice. In both diabetic groups, glomerular filtration rate increased significantly and to the same extent as compared to non-diabetic controls. Albuminuria, glomerular mesangial expansion, glomerular cellularity, and glomerular size were all increased to a similar extent in minicircle ACE2-treated and untreated diabetic mice, as compared to non-diabetic controls. Recombinant mouse ACE2 given for 4 weeks by intraperitoneal daily injections in mice with streptozotocin-induced diabetic nephropathy also failed to improve albuminuria or kidney pathology. Thus, a profound augmentation of ACE2 confined to the circulation failed to ameliorate the glomerular lesions and hyperfiltration characteristic of early diabetic nephropathy. These findings emphasize the importance of targeting the kidney rather than the circulatory renin angiotensin system to combat diabetic nephropathy.
Collapse
Affiliation(s)
- Jan Wysocki
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Minghao Ye
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ahmed M Khattab
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Agnes Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Aline Martin
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nicolae Valentin David
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yashpal Kanwar
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mark Osborn
- University of Minnesota, Division of Blood and Marrow Transplantation, Department of Pediatrics, Minneapolis, Minnesota, USA
| | - Daniel Batlle
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
66
|
Murine recombinant angiotensin-converting enzyme 2 attenuates kidney injury in experimental Alport syndrome. Kidney Int 2017; 91:1347-1361. [DOI: 10.1016/j.kint.2016.12.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 01/11/2023]
|
67
|
Simões e Silva AC, Miranda AS, Rocha NP, Teixeira AL. Renin angiotensin system in liver diseases: Friend or foe? World J Gastroenterol 2017; 23:3396-3406. [PMID: 28596676 PMCID: PMC5442076 DOI: 10.3748/wjg.v23.i19.3396] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 03/17/2017] [Accepted: 04/12/2017] [Indexed: 02/06/2023] Open
Abstract
In the last three decades, the understanding of the renin angiotensin system (RAS) has been changed by the discoveries of functional local systems, novel biologically active peptides, additional specific receptors, alternative pathways of angiotensin (Ang) II generation, and new roles for enzymes and precursor components other than those in Ang II synthesis. In this regard, the discovery that Ang-(1-7) opposes the pressor, proliferative, pro-fibrotic, and pro-inflammatory effects mediated by Ang II has contributed to the realization that the RAS is composed of two axes. The first axis consists of the angiotensin-converting enzyme (ACE), with Ang II as the end product, and the angiotensin type 1 (AT1) receptor as the main effector mediating the biological actions of Ang II. The second axis results from ACE2-mediated hydrolysis of Ang II, leading to the production of Ang-(1-7), with the Mas receptor as the main effector conveying the vasodilatory, anti-proliferative, anti-fibrotic, and anti-inflammatory effects of Ang-(1-7). Experimental and clinical studies have shown that both axes of the RAS may take part in the pathogenesis of liver diseases. In this manuscript, we summarize the current evidence regarding the role of RAS in hepatic cirrhosis and its complications, including hemodynamic changes and hepatorenal syndrome. The therapeutic potential of the modulation of RAS molecules in liver diseases is also discussed.
Collapse
|
68
|
A Fluorometric Method of Measuring Carboxypeptidase Activities for Angiotensin II and Apelin-13. Sci Rep 2017; 7:45473. [PMID: 28378780 PMCID: PMC5381230 DOI: 10.1038/srep45473] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/28/2017] [Indexed: 12/29/2022] Open
Abstract
Degradation of the biologically potent octapeptide angiotensin Ang II-(1-8) is mediated by the activities of several peptidases. The conversion of Ang II to the septapeptide Ang-(1-7) is of particular interest as the latter also confers organ protection. The conversion is catalyzed by angiotensin-converting enzyme 2 and other enzymes that selectively cleave the peptide bond between the proline and the phenylalanine at the carboxyl terminus of Ang II. The contribution of various enzyme activities that collectively lead to the formation of Ang-(1-7) from Ang II, in both normal conditions and in disease states, remains only partially understood. This is largely due to the lack of a reliable and sensitive method to detect these converting activities in complex samples, such as blood and tissues. Here, we report a fluorometric method to measure carboxypeptidase activities that cleave the proline-phenylalanine dipeptide bond in Ang II. This method is also suitable for measuring the conversion of apelin-13. The assay detects the release of phenylalanine amino acid in a reaction with the yeast enzyme of phenylalanine ammonia lyase (PAL). When used in cell and mouse organs, the assay can robustly measure endogenous Ang II and apelin-13-converting activities involved in the renin-angiotensin and the apelinergic systems, respectively.
Collapse
|
69
|
Cao G, Della Penna SL, Kouyoumdzian NM, Choi MR, Gorzalczany S, Fernández BE, Toblli JE, Rosón MI. Immunohistochemical expression of intrarenal renin angiotensin system components in response to tempol in rats fed a high salt diet. World J Nephrol 2017; 6:29-40. [PMID: 28101449 PMCID: PMC5215206 DOI: 10.5527/wjn.v6.i1.29] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/06/2016] [Accepted: 11/02/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To determine the effect of tempol in normal rats fed high salt on arterial pressure and the balance between antagonist components of the renal renin-angiotensin system.
METHODS Sprague-Dawley rats were fed with 8% NaCl high-salt (HS) or 0.4% NaCl (normal-salt, NS) diet for 3 wk, with or without tempol (T) (1 mmol/L, administered in drinking water). Mean arterial pressure (MAP), glomerular filtration rate (GFR), and urinary sodium excretion (UVNa) were measured. We evaluated angiotensin II (Ang II), angiotensin 1-7 (Ang 1-7), angiotensin converting enzyme 2 (ACE2), mas receptor (MasR), angiotensin type 1 receptor (AT1R) and angiotensin type 2 receptor (AT2R) in renal tissues by immunohistochemistry.
RESULTS The intake of high sodium produced a slight but significant increase in MAP and differentially regulated components of the renal renin-angiotensin system (RAS). This included an increase in Ang II and AT1R, and decrease in ACE-2 staining intensity using immunohistochemistry. Antioxidant supplementation with tempol increased natriuresis and GFR, prevented changes in blood pressure and reversed the imbalance of renal RAS components. This includes a decrease in Ang II and AT1R, as increase in AT2, ACE2, Ang (1-7) and MasR staining intensity using immunohistochemistry. In addition, the natriuretic effects of tempol were observed in NS-T group, which showed an increased staining intensity of AT2, ACE2, Ang (1-7) and MasR.
CONCLUSION These findings suggest that a high salt diet leads to changes in the homeostasis and balance between opposing components of the renal RAS in hypertension to favour an increase in Ang II. Chronic antioxidant supplementation can modulate the balance between the natriuretic and antinatriuretic components of the renal RAS.
Collapse
|
70
|
Fenofibrate Therapy Restores Antioxidant Protection and Improves Myocardial Insulin Resistance in a Rat Model of Metabolic Syndrome and Myocardial Ischemia: The Role of Angiotensin II. Molecules 2016; 22:molecules22010031. [PMID: 28036029 PMCID: PMC6155612 DOI: 10.3390/molecules22010031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/04/2016] [Accepted: 12/20/2016] [Indexed: 11/25/2022] Open
Abstract
Renin-angiotensin system (RAS) activation promotes oxidative stress which increases the risk of cardiac dysfunction in metabolic syndrome (MetS) and favors local insulin resistance. Fibrates regulate RAS improving MetS, type-2 diabetes and cardiovascular diseases. We studied the effect of fenofibrate treatment on the myocardic signaling pathway of Angiotensin II (Ang II)/Angiotensin II type 1 receptor (AT1) and its relationship with oxidative stress and myocardial insulin resistance in MetS rats under heart ischemia. Control and MetS rats were assigned to the following groups: (a) sham; (b) vehicle-treated myocardial infarction (MI) (MI-V); and (c) fenofibrate-treated myocardial infarction (MI-F). Treatment with fenofibrate significantly reduced triglycerides, non-high density lipoprotein cholesterol (non-HDL-C), insulin levels and insulin resistance index (HOMA-IR) in MetS animals. MetS and MI increased Ang II concentration and AT1 expression, favored myocardial oxidative stress (high levels of malondialdehyde, overexpression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4), decreased total antioxidant capacity and diminished expression of superoxide dismutase (SOD)1, SOD2 and catalase) and inhibited expression of the insulin signaling cascade: phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PkB, also known as Akt)/Glut-4/endothelial nitric oxide synthase (eNOS). In conclusion, fenofibrate treatment favors an antioxidant environment as a consequence of a reduction of the Ang II/AT1/NOX4 signaling pathway, reestablishing the cardiac insulin signaling pathway. This might optimize cardiac metabolism and improve the vasodilator function during myocardial ischemia.
Collapse
|
71
|
Froogh G, Pinto JT, Le Y, Kandhi S, Aleligne Y, Huang A, Sun D. Chymase-dependent production of angiotensin II: an old enzyme in old hearts. Am J Physiol Heart Circ Physiol 2016; 312:H223-H231. [PMID: 27815252 DOI: 10.1152/ajpheart.00534.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/17/2016] [Accepted: 10/27/2016] [Indexed: 02/08/2023]
Abstract
Age-dependent alteration of the renin-angiotensin system (RAS) and generation of angiotensin II (Ang II) are well documented. By contrast, RAS-independent generation of Ang II in aging and its responses to exercise have not been explored. To this end, we examined the effects of chymase, a secretory serine protease, on the angiotensin-converting enzyme (ACE)-independent conversion of Ang I to Ang II. We hypothesized that age-dependent alteration of cardiac Ang II formation is chymase dependent in nature and is prevented by exercise training. Experiments were conducted on hearts isolated from young (3 mo), aged sedentary (24 mo), and aged rats chronically exercised on a treadmill. In the presence of low Ang I levels and downregulation of ACE expression/activity, cardiac Ang II levels were significantly higher in aged than young rats, suggesting an ACE-independent response. Aged hearts also displayed significantly increased chymase expression and activity, as well as upregulation of tryptase, a biological marker of mast cells, confirming a mast cell-sourced increase in chymase. Coincidently, cardiac superoxide produced from NADPH oxidase (Nox) was significantly enhanced in aged rats and was normalized by exercise. Conversely, a significant reduction in cardiac expression of ACE2 followed by lower Ang 1-7 levels and downregulation of the Mas receptor (binding protein of Ang 1-7) in aged rats were completely reversed by exercise. In conclusion, local formation of Ang II is increased in aged hearts, and chymase is primarily responsible for this increase. Chronic exercise is able to normalize the age-dependent alterations via compromising chymase/Ang II/angiotensin type 1 receptor/Nox actions while promoting ACE2/Ang 1-7/MasR signaling. NEW & NOTEWORTHY Aging increases angiotensin-converting enzyme (ACE)-independent production of cardiac angiotensin II (Ang II), a response that is driven by chymase in an exercise-reversible manner. These findings highlight chymase, in addition to ACE, as an important therapeutic target in the treatment and prevention of Ang II-induced deterioration of cardiac function in the elderly.
Collapse
Affiliation(s)
- Ghezal Froogh
- Department of Physiology, New York Medical College, Valhalla, New York; and
| | - John T Pinto
- Department of Biochemistry, New York Medical College, Valhalla, New York
| | - Yicong Le
- Department of Physiology, New York Medical College, Valhalla, New York; and
| | - Sharath Kandhi
- Department of Physiology, New York Medical College, Valhalla, New York; and
| | - Yeabsra Aleligne
- Department of Physiology, New York Medical College, Valhalla, New York; and
| | - An Huang
- Department of Physiology, New York Medical College, Valhalla, New York; and
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, New York; and
| |
Collapse
|
72
|
Wang G, Zhang Q, Yuan W, Wu J, Li C. Enalapril protects against myocardial ischemia/reperfusion injury in a swine model of cardiac arrest and resuscitation. Int J Mol Med 2016; 38:1463-1473. [PMID: 27633002 PMCID: PMC5065301 DOI: 10.3892/ijmm.2016.2737] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/06/2016] [Indexed: 12/16/2022] Open
Abstract
There is strong evidence to suggest that angiotensin-converting enzyme inhibitors (ACEIs) protect against local myocardial ischemia/reperfusion (I/R) injury. This study was designed to explore whether ACEIs exert cardioprotective effects in a swine model of cardiac arrest (CA) and resuscitation. Male pigs were randomly assigned to three groups: sham-operated group, saline treatment group and enalapril treatment group. Thirty minutes after drug infusion, the animals in the saline and enalapril groups were subjected to ventricular fibrillation (8 min) followed by cardiopulmonary resuscitation (up to 30 min). Cardiac function was monitored, and myocardial tissue and blood were collected for analysis. Enalapril pre-treatment did not improve cardiac function or the 6-h survival rate after CA and resuscitation; however, this intervention ameliorated myocardial ultrastructural damage, reduced the level of plasma cardiac troponin I and decreased myocardial apoptosis. Plasma angiotensin (Ang) II and Ang-(1–7) levels were enhanced in the model of CA and resuscitation. Enalapril reduced the plasma Ang II level at 4 and 6 h after the return of spontaneous circulation whereas enalapril did not affect the plasma Ang-(1–7) level. Enalapril pre-treatment decreased the myocardial mRNA and protein expression of angiotensin-converting enzyme (ACE). Enalapril treatment also reduced the myocardial ACE/ACE2 ratio, both at the mRNA and the protein level. Enalapril pre-treatment did not affect the upregulation of ACE2, Ang II type 1 receptor (AT1R) and MAS after CA and resuscitation. Taken together, these findings suggest that enalapril protects against ischemic injury through the attenuation of the ACE/Ang II/AT1R axis after CA and resuscitation in pigs. These results suggest the potential therapeutic value of ACEIs in patients with CA.
Collapse
Affiliation(s)
- Guoxing Wang
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Qian Zhang
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation (NO.BZ0370), Beijing 100020, P.R. China
| | - Wei Yuan
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation (NO.BZ0370), Beijing 100020, P.R. China
| | - Junyuan Wu
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation (NO.BZ0370), Beijing 100020, P.R. China
| | - Chunsheng Li
- Department of Emergency Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation (NO.BZ0370), Beijing 100020, P.R. China
| |
Collapse
|
73
|
Cerrato BD, Carretero OA, Janic B, Grecco HE, Gironacci MM. Heteromerization Between the Bradykinin B2 Receptor and the Angiotensin-(1-7) Mas Receptor: Functional Consequences. Hypertension 2016; 68:1039-48. [PMID: 27550920 DOI: 10.1161/hypertensionaha.116.07874] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/26/2016] [Indexed: 12/20/2022]
Abstract
Bradykinin B2 receptor (B2R) and angiotensin-(1-7) Mas receptor (MasR)-mediated effects are physiologically interconnected. The molecular basis for such cross talk is unknown. It is hypothesized that the cross talk occurs at the receptor level. We investigated B2R-MasR heteromerization and the functional consequences of such interaction. B2R fused to the cyan fluorescent protein and MasR fused to the yellow fluorescent protein were transiently coexpressed in human embryonic kidney293T cells. Fluorescence resonance energy transfer analysis showed that B2R and MasR formed a constitutive heteromer, which was not modified by their agonists. B2R or MasR antagonists decreased fluorescence resonance energy transfer efficiency, suggesting that the antagonist promoted heteromer dissociation. B2R-MasR heteromerization induced an 8-fold increase in the MasR ligand-binding affinity. On agonist stimulation, the heteromer was internalized into early endosomes with a slower sequestration rate from the plasma membrane, compared with single receptors. B2R-MasR heteromerization induced a greater increase in arachidonic acid release and extracellular signal-regulated kinase phosphorylation after angiotensin-(1-7) stimulation, and this effect was blocked by the B2R antagonist. Concerning serine/threonine kinase Akt activity, a significant bradykinin-promoted activation was detected in B2R-MasR but not in B2R-expressing cells. Angiotensin-(1-7) and bradykinin elicited antiproliferative effects only in cells expressing B2R-MasR heteromers, but not in cells expressing each receptor alone. Proximity ligation assay confirmed B2R-MasR interaction in human glomerular endothelial cells supporting the interaction between both receptors in vivo. Our findings provide an explanation for the cross talk between bradykinin B2R and angiotensin-(1-7) MasR-mediated effects. B2R-MasR heteromerization induces functional changes in the receptor that may lead to long-lasting protective properties.
Collapse
Affiliation(s)
- Bruno D Cerrato
- From the Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina (B.D.C., M.M.G.); Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI (O.A.C., B.J.); and Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IFIBA-CONICET, Argentina (H.E.G)
| | - Oscar A Carretero
- From the Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina (B.D.C., M.M.G.); Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI (O.A.C., B.J.); and Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IFIBA-CONICET, Argentina (H.E.G)
| | - Brana Janic
- From the Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina (B.D.C., M.M.G.); Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI (O.A.C., B.J.); and Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IFIBA-CONICET, Argentina (H.E.G)
| | - Hernán E Grecco
- From the Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina (B.D.C., M.M.G.); Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI (O.A.C., B.J.); and Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IFIBA-CONICET, Argentina (H.E.G)
| | - Mariela M Gironacci
- From the Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina (B.D.C., M.M.G.); Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI (O.A.C., B.J.); and Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IFIBA-CONICET, Argentina (H.E.G).
| |
Collapse
|
74
|
Wang M, Zhang X, Song X, Zou X, Wu W, Wang Y, Lin B, Li R, Hu F, Zhao H. Nodular glomerulosclerosis and renin angiotensin system in Chinese patients with type 2 diabetes. Mol Cell Endocrinol 2016; 427:92-100. [PMID: 26973293 DOI: 10.1016/j.mce.2016.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/24/2016] [Accepted: 03/07/2016] [Indexed: 11/20/2022]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a multifactorial and polygenic disease with nodular glomerulosclerosis (NGS) pathognomonic for diabetes and hypertension. Patients with type 2 diabetes and hypertension have characteristic renin-angiotensin system (RAS) gene polymorphisms. METHODS AND RESULTS In this retrospective cohort study, we correlated the presence of NGS with renal function, angiotensin-converting enzyme (ACE) genotypes (DD, DI, and II), angiotensinogen (AGT) genotypes (MM, MT, and TT) and immunohistochemical staining characteristics of RAS components in 847 patients and 172 consecutive autopsy cases with type 2 diabetes. T allele of AGT was associated with macroalbuminuria (P = 0.040). Multitude regression analysis revealed ACE insertion (I)/deletion (D) polymorphism as an independent determinant for estimated glomerular filtration rate (eGFR) less than 60 mL min(-1)·1.73 m(-2) (DD carriers: odds ratio [OR] = 3.46, 95% confidence interval [CI] = 1.08-11.07; DI carriers: OR = 3.51, 95% CI = 1.63-7.56). A significant association between NGS and eGFR less than 60 mL min(-1)·1.73 m(-2) also persisted after adjusting for nonlinear relationship (P < 0.001). In NGS patients, immunoreactivity of angiotensin I converting enzyme 2 (ACE2) significantly decreased in glomeruli with mesangial nodules compared with glomeruli without the mesangial nodules. CONCLUSIONS These data suggest associations of ACE D allele with glomerular filtration impairment, and NGS with glomerular ACE2 down-regulation and reduced glomerular filtration in Chinese patients with type 2 diabetes.
Collapse
Affiliation(s)
- Min Wang
- Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China; Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Xiaoxi Zhang
- Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China; Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Xinnan Song
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Xia Zou
- Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China; Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Weijie Wu
- Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China; Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Yanchao Wang
- Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China; Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Bingjie Lin
- Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China; Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Rong Li
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Endocrinology and Toxicology Laboratory, Department of Biology, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Fang Hu
- Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Hailu Zhao
- Institute of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China; Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China.
| |
Collapse
|
75
|
Novel players in cardioprotection: Insulin like growth factor-1, angiotensin-(1–7) and angiotensin-(1–9). Pharmacol Res 2015; 101:41-55. [DOI: 10.1016/j.phrs.2015.06.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 06/27/2015] [Accepted: 06/28/2015] [Indexed: 12/14/2022]
|
76
|
Wang HB, Yang J. The role of renin-angiotensin aldosterone system related micro-ribonucleic acids in hypertension. Saudi Med J 2015; 36:1151-5. [PMID: 26446323 PMCID: PMC4621718 DOI: 10.15537/smj.2015.10.12458] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Micro-ribonucleic acids (miRNAs) are small (21-25 nucleotide) single-stranded, evolutionarily conserved non-protein-coding RNAs, which control diverse cellular functions by interacting with the 3' untranslated region of specific target messenger RNAs at the post-transcriptional level. Research shows that an aberrant expression profile of miRNAs has been linked to a series of diseases, including hypertension. In the past few decades, it has been demonstrated that excessive activation of the renin-angiotensin aldosterone system (RAAS) involves in the pathogenesis of hypertension. This article reviews the latest insights in the identification of RAAS-correlative miRNAs and the potential mechanisms for their roles in hypertension.
Collapse
Affiliation(s)
- Hui-Bo Wang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei Province, Hubei, China. E-mail.
| | | |
Collapse
|
77
|
Bernardi S, Toffoli B, Zennaro C, Bossi F, Losurdo P, Michelli A, Carretta R, Mulatero P, Fallo F, Veglio F, Fabris B. Aldosterone effects on glomerular structure and function. J Renin Angiotensin Aldosterone Syst 2015; 16:730-8. [PMID: 26283678 DOI: 10.1177/1470320315595568] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/11/2015] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Experimental evidence suggests that aldosterone directly contributes to organ damage by promoting cell growth, fibrosis, and inflammation. Based on these premises, this work aimed to assess the glomerular effects of aldosterone, alone and in combination with salt. METHODS After undergoing uninephrectomy, 75 rats were allocated to five groups: control, salt diet, aldosterone, aldosterone + salt diet, aldosterone + salt diet and eplerenone, and they were all studied for four weeks. We focused on glomerular structural, functional, and molecular changes, including slit diaphragm components, local renin-angiotensin system activation, as well as pro-oxidative and profibrotic changes. RESULTS Aldosterone significantly increased systolic blood pressure, led to glomerular hypertrophy, mesangial expansion, and it significantly increased the glomerular permeability to albumin and the albumin excretion rate, indicating the presence of glomerular damage. These effects were worsened by adding salt to aldosterone, while they were reduced by eplerenone. Aldosterone-induced glomerular damage was associated with glomerular angiotensin-converting enzyme (ACE) 2 downregulation, with ACE/ACE2 ratio increase, ANP decrease, as well as with glomerular pro-oxidative and profibrotic changes. CONCLUSIONS Aldosterone damages not only the structure but also the function of the glomerulus. ACE/ACE2 upregulation, ACE2 and ANP downregulation, and pro-oxidative and profibrotic changes are possible mechanisms accounting for aldosterone-induced glomerular injury.
Collapse
Affiliation(s)
- Stella Bernardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Italy
| | - Barbara Toffoli
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Italy Centre for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | - Cristina Zennaro
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Italy
| | - Fleur Bossi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Italy
| | - Pasquale Losurdo
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Italy
| | - Andrea Michelli
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Italy
| | - Renzo Carretta
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Italy
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension, University of Torino, Italy
| | - Francesco Fallo
- Department of Medical and Surgical Sciences, University of Padova, Italy
| | - Franco Veglio
- Division of Internal Medicine and Hypertension, University of Torino, Italy
| | - Bruno Fabris
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Italy
| |
Collapse
|
78
|
Abstract
Angiotensin (Ang) (1-7) is the main component of the depressor and protective arm of the renin-angiotensin system. Ang-(1-7) induces vasodilation, natriuresis and diuresis, cardioprotection, inhibits angiogenesis and cell growth and opposes the pressor, proliferative, profibrotic, and prothrombotic actions mediated by Ang II. Centrally, Ang-(1-7) induces changes in mean arterial pressure and this effect may be linked with its inhibitory neuromodulatory action on norepinephrine neurotransmission. The present review is focused on the role of Ang-(1-7) as a protective agent in the brain.
Collapse
Affiliation(s)
- Mariela M Gironacci
- Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 1113 Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
79
|
Mendoza-Torres E, Oyarzún A, Mondaca-Ruff D, Azocar A, Castro PF, Jalil JE, Chiong M, Lavandero S, Ocaranza MP. ACE2 and vasoactive peptides: novel players in cardiovascular/renal remodeling and hypertension. Ther Adv Cardiovasc Dis 2015; 9:217-37. [PMID: 26275770 DOI: 10.1177/1753944715597623] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The renin-angiotensin system (RAS) is a key component of cardiovascular physiology and homeostasis due to its influence on the regulation of electrolyte balance, blood pressure, vascular tone and cardiovascular remodeling. Deregulation of this system contributes significantly to the pathophysiology of cardiovascular and renal diseases. Numerous studies have generated new perspectives about a noncanonical and protective RAS pathway that counteracts the proliferative and hypertensive effects of the classical angiotensin-converting enzyme (ACE)/angiotensin (Ang) II/angiotensin type 1 receptor (AT1R) axis. The key components of this pathway are ACE2 and its products, Ang-(1-7) and Ang-(1-9). These two vasoactive peptides act through the Mas receptor (MasR) and AT2R, respectively. The ACE2/Ang-(1-7)/MasR and ACE2/Ang-(1-9)/AT2R axes have opposite effects to those of the ACE/Ang II/AT1R axis, such as decreased proliferation and cardiovascular remodeling, increased production of nitric oxide and vasodilation. A novel peptide from the noncanonical pathway, alamandine, was recently identified in rats, mice and humans. This heptapeptide is generated by catalytic action of ACE2 on Ang A or through a decarboxylation reaction on Ang-(1-7). Alamandine produces the same effects as Ang-(1-7), such as vasodilation and prevention of fibrosis, by interacting with Mas-related GPCR, member D (MrgD). In this article, we review the key roles of ACE2 and the vasoactive peptides Ang-(1-7), Ang-(1-9) and alamandine as counter-regulators of the ACE-Ang II axis as well as the biological properties that allow them to regulate blood pressure and cardiovascular and renal remodeling.
Collapse
Affiliation(s)
- Evelyn Mendoza-Torres
- Advanced Center for Chronic Diseases (ACCDiS), Centro de Estudios Moleculares de la Célula, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alejandra Oyarzún
- Advanced Center for Chronic Diseases (ACCDiS), Centro de Estudios Moleculares de la Célula, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - David Mondaca-Ruff
- Advanced Center for Chronic Diseases (ACCDiS), Centro de Estudios Moleculares de la Célula, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Andrés Azocar
- Advanced Center for Chronic Diseases (ACCDiS), Centro de Estudios Moleculares de la Célula, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pablo F Castro
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile Division Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge E Jalil
- Division Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Centro de Estudios Moleculares de la Célula, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Centro de Estudios Moleculares de la Célula, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - María Paz Ocaranza
- Advanced Center for Chronic Diseases(ACCDiS), Facultad de Medicina, PontificiaUniversidad Católica de Chile, Santiago, Chile.Division Enfermedades Cardiovasculares,Facultad de Medicina, Pontificia UniversidadCatólica de Chile, Santiago, Chile
| |
Collapse
|
80
|
Jin HY, Chen LJ, Zhang ZZ, Xu YL, Song B, Xu R, Oudit GY, Gao PJ, Zhu DL, Zhong JC. Deletion of angiotensin-converting enzyme 2 exacerbates renal inflammation and injury in apolipoprotein E-deficient mice through modulation of the nephrin and TNF-alpha-TNFRSF1A signaling. J Transl Med 2015; 13:255. [PMID: 26245758 PMCID: PMC4527357 DOI: 10.1186/s12967-015-0616-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/27/2015] [Indexed: 12/17/2022] Open
Abstract
Background The renin-angiotensin system (RAS) has been implicated in atherosclerotic lesions and progression to chronic kidney diseases. We examined regulatory roles of angiotensin-converting enzyme 2 (ACE2) in the apolipoprotein E (ApoE) knockout (KO) kidneys. Methods The 3-month-old wild-type, ApoEKO, ACE2KO and ApoE/ACE2 double-KO (DKO) mice in a C57BL/6 background were used. The ApoEKO mice were randomized to daily deliver either Ang II (1.5 mg/kg) and/or human recombinant ACE2 (rhACE2; 2 mg/kg) for 2 weeks. We examined changes in pro-inflammatory cytokines, renal ultrastructure, and pathological signaling in mouse kidneys. Results Downregulation of ACE2 and nephrin levels was observed in ApoEKO kidneys. Genetic ACE2 deletion resulted in modest elevations in systolic blood pressure levels and Ang II type 1 receptor expression and reduced nephrin expression in kidneys of the ApoE/ACE2 DKO mice with a decrease in renal Ang-(1-7) levels. These changes were linked with marked increases in renal superoxide generation, NADPH oxidase (NOX) 4 and proinflammatory factors levels, including interleukin (IL)-1beta, IL-6, IL-17A, RANTES, ICAM-1, Tumor necrosis factor-alpha (TNF-alpha) and TNFRSF1A. Renal dysfunction and ultrastructure injury were aggravated in the ApoE/ACE2 DKO mice and Ang II-infused ApoEKO mice with increased plasma levels of creatinine, blood urea nitrogen and enhanced levels of Ang II in plasma and kidneys. The Ang II-mediated reductions of renal ACE2 and nephrin levels in ApoEKO mice were remarkably rescued by rhACE2 supplementation, along with augmentation of renal Ang-(1-7) levels. More importantly, rhACE2 treatment significantly reversed Ang II-induced renal inflammation, superoxide generation, kidney dysfunction and adverse renal injury in ApoEKO mice with suppression of the NOX4 and TNF-alpha-TNFRSF1A signaling. However, rhACE2 had no effect on renal NOX2 and TNFRSF1B expression and circulating lipid levels. Conclusions ACE2 deficiency exacerbates kidney inflammation, oxidative stress and adverse renal injury in the ApoE-mutant mice through modulation of the nephrin, NOX4 and TNF-alpha-TNFRSF1A signaling. While rhACE2 supplementation alleviates inflammation, renal dysfunction and glomerulus injury in the ApoE-mutant mice associated with upregulations of Ang-(1-7) levels and nephrin expression and suppression of the TNF-alpha-TNFRSF1A signaling. Strategies aimed at enhancing the ACE2/Ang-(1-7) actions may have important therapeutic potential for atherosclerotic renal injury and kidney diseases.
Collapse
Affiliation(s)
- Hai-Yan Jin
- State Key Laboratory of Medical Genomics and Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China. .,Pôle Sino-Français de Recherches en Science du Vivant et Génomique, Department of Mental Health, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Lai-Jiang Chen
- State Key Laboratory of Medical Genomics and Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China. .,Institute of Health Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.
| | - Zhen-Zhou Zhang
- State Key Laboratory of Medical Genomics and Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China. .,Institute of Health Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.
| | - Ying-Le Xu
- State Key Laboratory of Medical Genomics and Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Bei Song
- State Key Laboratory of Medical Genomics and Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Ran Xu
- State Key Laboratory of Medical Genomics and Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Gavin Y Oudit
- Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, T6G 2S2, Canada.
| | - Ping-Jin Gao
- State Key Laboratory of Medical Genomics and Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China. .,Institute of Health Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.
| | - Ding-Liang Zhu
- State Key Laboratory of Medical Genomics and Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China. .,Pôle Sino-Français de Recherches en Science du Vivant et Génomique, Department of Mental Health, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China. .,Institute of Health Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.
| | - Jiu-Chang Zhong
- State Key Laboratory of Medical Genomics and Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China. .,Pôle Sino-Français de Recherches en Science du Vivant et Génomique, Department of Mental Health, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China. .,Institute of Health Sciences, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.
| |
Collapse
|
81
|
Murphy AM, Wong AL, Bezuhly M. Modulation of angiotensin II signaling in the prevention of fibrosis. FIBROGENESIS & TISSUE REPAIR 2015; 8:7. [PMID: 25949522 PMCID: PMC4422447 DOI: 10.1186/s13069-015-0023-z] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 03/06/2015] [Indexed: 12/20/2022]
Abstract
Over the last decade, it has become clear that the role of angiotensin II extends far beyond recognized renal and cardiovascular effects. The presence of an autologous renin-angiotensin system has been demonstrated in almost all tissues of the body. It is now known that angiotensin II acts both independently and in synergy with TGF-beta to induce fibrosis via the angiotensin type 1 receptor (AT1) in a multitude of tissues outside of the cardiovascular and renal systems, including pulmonary fibrosis, intra-abdominal fibrosis, and systemic sclerosis. Interestingly, recent studies have described a paradoxically regenerative effect of the angiotensin system via stimulation of the angiotensin type 2 receptor (AT2). Activation of AT2 has been shown to ameliorate fibrosis in animal models of skeletal muscle, gastrointestinal, and neurologic diseases. Clinical reports suggest a beneficial role for modulation of angiotensin II signaling in cutaneous scarring. This article reviews current knowledge on the role that angiotensin II plays in tissue fibrosis, as well as current and potential therapies targeting this system.
Collapse
Affiliation(s)
- Amanda M Murphy
- Division of Plastic and Reconstructive Surgery, Dalhousie University, 5850/5980 University Avenue, PO Box 9700, B3K 6R8 Halifax, NS Canada
| | - Alison L Wong
- Division of Plastic and Reconstructive Surgery, Dalhousie University, 5850/5980 University Avenue, PO Box 9700, B3K 6R8 Halifax, NS Canada
| | - Michael Bezuhly
- Division of Plastic and Reconstructive Surgery, Dalhousie University, 5850/5980 University Avenue, PO Box 9700, B3K 6R8 Halifax, NS Canada ; IWK Health Centre, Dalhousie University, 5850/5980 University Avenue, PO Box 9700, B3K 6R8 Halifax, NS Canada
| |
Collapse
|
82
|
Olkowicz M, Chlopicki S, Smolenski RT. Perspectives for angiotensin profiling with liquid chromatography/mass spectrometry to evaluate ACE/ACE2 balance in endothelial dysfunction and vascular pathologies. Pharmacol Rep 2015; 67:778-85. [PMID: 26321281 DOI: 10.1016/j.pharep.2015.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 03/22/2015] [Accepted: 03/25/2015] [Indexed: 02/07/2023]
Abstract
Vascular injury, characterized by endothelial dysfunction, inflammation, structural remodeling, thrombosis and calcification leads to cardiovascular diseases. Angiotensin (Ang) II (1-8) - synthesized mainly by angiotensin converting enzyme (ACE) is the best characterized mediator of the renin-angiotensin system (RAS). This peptide initially identified by its vasoactive properties was found to play a major role in vascular response to insult. However, recent discovery of angiotensin converting enzyme 2 (ACE2) that produces vasoprotective Ang-(1-7) peptide highlighted complexity of the system and suggested that balance between ACE/Ang II and ACE2/Ang-(1-7) is fundamental in maintaining vascular homeostasis and its disorders are associated with cardiovascular pathology. There is therefore a need to develop methods for comprehensive analysis of biologically active Ang peptides and their metabolites of ACE/Ang II and ACE2/Ang-(1-7) axes. Liquid chromatography/mass spectrometry (LC/MS) is an analytical technique that offers potential for specific, simultaneous analysis of Ang peptides. With sensitivity added by application of preconcentration nanochromatography reaching picomolar concentrations, practically all Ang peptides identified so far could be quantified in biological samples. Ang profiling is important not only for understanding their physiological or pathological role but could also serve as an early diagnostic biomarker of endothelial dysfunction and cardiovascular pathology. It could also be used for monitoring the efficacy of the RAS-targeted therapies. Although, the methodology requires further improvements to adopt it for routine application, Ang peptide profiling with targeted LC/MS analysis might assess functional balance between ACE/Ang II and ACE2/Ang-(1-7) axes, facilitate our understanding of the cardiovascular pathology and enhance biomarker portfolio in cardiovascular diseases.
Collapse
Affiliation(s)
- Mariola Olkowicz
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland; Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, Poznań, Poland.
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland; Department of Experimental Pharmacology, Jagiellonian University Medical College, Kraków, Poland
| | | |
Collapse
|
83
|
Short-term treatment with diminazene aceturate ameliorates the reduction in kidney ACE2 activity in rats with subtotal nephrectomy. PLoS One 2015; 10:e0118758. [PMID: 25786223 PMCID: PMC4364975 DOI: 10.1371/journal.pone.0118758] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 01/06/2015] [Indexed: 01/28/2023] Open
Abstract
Angiotensin converting enzyme (ACE) 2 is an important modulator of the renin angiotensin system (RAS) through its role to degrade angiotensin (Ang) II. Depletion of kidney ACE2 occurs following kidney injury due to renal mass reduction and may contribute to progressive kidney disease. This study assessed the effect of diminazine aceturate (DIZE), which has been described as an ACE2 activator, on kidney ACE2 mRNA and activity in rats with kidney injury due to subtotal nephrectomy (STNx). Sprague Dawley rats were divided into Control groups or underwent STNx; rats then received vehicle or the DIZE (s.c. 15 mg/kg/day) for 2 weeks. STNx led to hypertension (P<0.01), kidney hypertrophy (P<0.001) and impaired kidney function (P<0.001) compared to Control rats. STNx was associated with increased kidney cortical ACE activity, and reduced ACE2 mRNA in the cortex (P<0.01), with reduced cortical and medullary ACE2 activity (P<0.05), and increased urinary ACE2 excretion (P<0.05) compared to Control rats. Urinary ACE2 activity correlated positively with urinary protein excretion (P<0.001), and negatively with creatinine clearance (P=0.04). In STNx rats, DIZE had no effect on blood pressure or kidney function, but was associated with reduced cortical ACE activity (P<0.01), increased cortical ACE2 mRNA (P<0.05) and increased cortical and medullary ACE2 activity (P<0.05). The precise in vivo mechanism of action of DIZE is not clear, and its effects to increase ACE2 activity may be secondary to an increase in ACE2 mRNA abundance. In ex vivo studies, DIZE did not increase ACE2 activity in either Control or STNx kidney cortical membranes. It is not yet known if chronic administration of DIZE has long-term benefits to slow the progression of kidney disease.
Collapse
|
84
|
Clayton D, Hanchapola I, Thomas WG, Widdop RE, Smith AI, Perlmutter P, Aguilar MI. Structural determinants for binding to angiotensin converting enzyme 2 (ACE2) and angiotensin receptors 1 and 2. Front Pharmacol 2015; 6:5. [PMID: 25688208 PMCID: PMC4311625 DOI: 10.3389/fphar.2015.00005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/08/2015] [Indexed: 11/13/2022] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is a zinc carboxypeptidase involved in the renin–angiotensin system (RAS) and inactivates the potent vasopressive peptide angiotensin II (Ang II) by removing the C-terminal phenylalanine residue to yield Ang1–7. This conversion inactivates the vasoconstrictive action of Ang II and yields a peptide that acts as a vasodilatory molecule at the Mas receptor and potentially other receptors. Given the growing complexity of RAS and level of cross-talk between ligands and their corresponding enzymes and receptors, the design of molecules with selectivity for the major RAS binding partners to control cardiovascular tone is an on-going challenge. In previous studies we used single β-amino acid substitutions to modulate the structure of Ang II and its selectivity for ACE2, AT1R, and angiotensin type 2 (AT2R) receptor. We showed that modification at the C-terminus of Ang II generally resulted in more pronounced changes to secondary structure and ligand binding, and here, we further explore this region for the potential to modulate ligand specificity. In this study, (1) a library of 47 peptides derived from the C-terminal tetrapeptide sequence (-IHPF) of Ang II was synthesized and assessed for ACE2 binding, (2) the terminal group requirements for high affinity ACE2 binding were explored by and N- and C-terminal modification, (3) high affinity ACE2 binding chimeric AngII analogs were then synthesized and assessed, (4) the structure of the full-length Ang II analogs were assessed by circular dichroism, and (5) the Ang II analogs were assessed for AT1R/AT2R selectivity by cell-based assays. Studies on the C-terminus of Ang II demonstrated varied specificity at different residue positions for ACE2 binding and four Ang II chimeric peptides were identified as selective ligands for the AT2 receptor. Overall, these results provide insight into the residue and structural requirements for ACE2 binding and angiotensin receptor selectivity.
Collapse
Affiliation(s)
- Daniel Clayton
- Department of Biochemistry and Molecular Biology, Monash University Clayton, VIC, Australia
| | - Iresha Hanchapola
- Department of Biochemistry and Molecular Biology, Monash University Clayton, VIC, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, University of Queensland Brisbane, QLD, Australia
| | - Robert E Widdop
- Department of Pharmacology, Monash University Clayton, VIC, Australia
| | - Alexander I Smith
- Department of Biochemistry and Molecular Biology, Monash University Clayton, VIC, Australia
| | | | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Monash University Clayton, VIC, Australia
| |
Collapse
|
85
|
Červenka L, Bíbová J, Husková Z, Vaňourková Z, Kramer HJ, Herget J, Jíchová Š, Sadowski J, Hampl V. Combined suppression of the intrarenal and circulating vasoconstrictor renin-ACE-ANG II axis and augmentation of the vasodilator ACE2-ANG 1-7-Mas axis attenuates the systemic hypertension in Ren-2 transgenic rats exposed to chronic hypoxia. Physiol Res 2014; 64:11-24. [PMID: 25194129 DOI: 10.33549/physiolres.932842] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The aim of the present study was to test the hypothesis that chronic hypoxia would aggravate hypertension in Ren-2 transgenic rats (TGR), a well-defined monogenetic model of hypertension with increased activity of endogenous renin-angiotensin system (RAS). Systolic blood pressure (SBP) in conscious rats and mean arterial pressure (MAP) in anesthetized TGR and normotensive Hannover Sprague-Dawley (HanSD) rats were determined under normoxia that was either continuous or interrupted by two weeks´ hypoxia. Expression, activities and concentrations of individual components of RAS were studied in plasma and kidney of TGR and HanSD rats under normoxic conditions and after exposure to chronic hypoxia. In HanSD rats two weeks´ exposure to chronic hypoxia did not alter SBP and MAP. Surprisingly, in TGR it decreased markedly SBP and MAP; this was associated with substantial reduction in plasma and kidney renin activities and also of angiotensin II (ANG II) levels, without altering angiotensin-converting enzyme (ACE) activities. Simultaneously, in TGR the exposure to hypoxia increased kidney ACE type 2 (ACE2) activity and angiotensin 1-7 (ANG 1-7) concentrations as compared with TGR under continuous normoxia. Based on these results, we propose that suppression of the hypertensiogenic ACE-ANG II axis in the circulation and kidney tissue, combined with augmentation of the intrarenal vasodilator ACE2-ANG 1-7 axis, is the main mechanism responsible for the blood pressure-lowering effects of chronic hypoxia in TGR.
Collapse
Affiliation(s)
- L Červenka
- Department of Pathophysiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Gao X, Shi X, Fu X, Ge L, Zhang Y, Su C, Yang X, Silvennoinen O, Yao Z, He J, Wei M, Yang J. Human Tudor staphylococcal nuclease (Tudor-SN) protein modulates the kinetics of AGTR1-3'UTR granule formation. FEBS Lett 2014; 588:2154-61. [PMID: 24815690 DOI: 10.1016/j.febslet.2014.04.045] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 04/17/2014] [Accepted: 04/25/2014] [Indexed: 10/25/2022]
Abstract
Human Tudor staphylococcal nuclease (Tudor-SN) interacts with the G3BP protein and is recruited into stress granules (SGs), the main type of discrete RNA-containing cytoplasmic foci structure that is formed under stress conditions. Here, we further demonstrate that Tudor-SN binds and co-localizes with AGTR1-3'UTR (3'-untranslated region of angiotensin II receptor, type 1 mRNA) into SG. Tudor-SN plays an important role in the assembly of AGTR1-3'UTR granules. Moreover, endogenous Tudor-SN knockdown can decrease the recovery kinetics of AGTR1-3'UTR granules. Collectively, our data indicate that Tudor-SN modulates the kinetics of AGTR1-3'UTR granule formation, which provides an additional biological role of Tudor-SN in RNA metabolism during stress.
Collapse
Affiliation(s)
- Xingjie Gao
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Laboratory of Molecular Immunology, Research Center of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China
| | - Xuebin Shi
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China
| | - Xue Fu
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Laboratory of Molecular Immunology, Research Center of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China
| | - Lin Ge
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China
| | - Yi Zhang
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Laboratory of Molecular Immunology, Research Center of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China
| | - Chao Su
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Laboratory of Molecular Immunology, Research Center of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China
| | - Xi Yang
- Department of Immunology, University of Manitoba, 471 Apotex Centre, 750 McDermot Avenue, Winnipeg R3E 0T5, Canada
| | - Olli Silvennoinen
- Institute of Medical Technology, University of Tampere, Tampere University Hospital, Biokatu 8, FI-33014 Tampere, Finland
| | - Zhi Yao
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China
| | - Jinyan He
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China
| | - Minxin Wei
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin 300070, China.
| | - Jie Yang
- Department of Biochemistry and Molecular Biology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Laboratory of Molecular Immunology, Research Center of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Educational Ministry of China, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|