1
|
Guo GX, Qiu YH, Liu Y, Yu LL, Zhang X, Tsim KWK, Qin QW, Hu WH. Fucoxanthin Attenuates Angiogenesis by Blocking the VEGFR2-Mediated Signaling Pathway through Binding the Vascular Endothelial Growth Factor. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21610-21623. [PMID: 39292861 DOI: 10.1021/acs.jafc.4c05464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Fucoxanthin, a dietary carotenoid, is predominantly found in edible brown algae and is commonly consumed worldwide. Fucoxanthin has been shown to possess beneficial health activities such as antidiabetic, anti-inflammatory, antimutagenic, and antiobesity; however, the effects of fucoxanthin on VEGF-mediated angiogenesis and its possible binding with VEGF are unknown. Here, different lines of evidence supported the suppressive roles of fucoxanthin in VEGF-mediated angiogenesis. In human umbilical vein endothelial cells, fucoxanthin remarkedly suppressed VEGF-mediated cell proliferative, migration, and invasive abilities, as well as tube formation, without cytotoxicity. In addition, fucoxanthin inhibited the subintestinal vessel formation of zebrafish in vivo. In signaling cascades, fucoxanthin was proposed to interact with VEGF, thus attenuating VEGF's functions in activating the VEGF receptor and its related downstream signaling, i.e., phosphorylations of MEK and Erk. Fucoxanthin also significantly blocked VEGF-triggered ROS formation. Furthermore, the outcomes of applying fucoxanthin in cancer cells were identified, which included (i) inhibiting VEGF-mediated cell proliferation and migration and (ii) inhibiting NF-κB translocation via limiting MMP2 expression. These lines of investigations supported the antiangiogenic roles of fucoxanthin, as well as reviewing its signaling mechanisms, in blocking the VEGF-triggered responses. The results would benefit the potential development of fucoxanthin for the prevention and treatment of angiogenesis-related diseases.
Collapse
Affiliation(s)
- Guo-Xia Guo
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Yu-Huan Qiu
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
- Nansha-South China Agricultural University Fishery Research Institute, Guangzhou 511464, China
| | - Yang Liu
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Le-Le Yu
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoyong Zhang
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Karl Wah-Keung Tsim
- Nansha-South China Agricultural University Fishery Research Institute, Guangzhou 511464, China
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Qi-Wei Qin
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
- Nansha-South China Agricultural University Fishery Research Institute, Guangzhou 511464, China
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Wei-Hui Hu
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
- Nansha-South China Agricultural University Fishery Research Institute, Guangzhou 511464, China
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
2
|
Tawengi M, Al-Dali Y, Tawengi A, Benter IF, Akhtar S. Targeting the epidermal growth factor receptor (EGFR/ErbB) for the potential treatment of renal pathologies. Front Pharmacol 2024; 15:1394997. [PMID: 39234105 PMCID: PMC11373609 DOI: 10.3389/fphar.2024.1394997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Epidermal growth factor receptor (EGFR), which is referred to as ErbB1/HER1, is the prototype of the EGFR family of receptor tyrosine kinases which also comprises ErbB2 (Neu, HER2), ErbB3 (HER3), and ErbB4 (HER4). EGFR, along with other ErbBs, is expressed in the kidney tubules and is physiologically involved in nephrogenesis and tissue repair, mainly following acute kidney injury. However, its sustained activation is linked to several kidney pathologies, including diabetic nephropathy, hypertensive nephropathy, glomerulonephritis, chronic kidney disease, and renal fibrosis. This review aims to provide a summary of the recent findings regarding the consequences of EGFR activation in several key renal pathologies. We also discuss the potential interplay between EGFR and the reno-protective angiotensin-(1-7) (Ang-(1-7), a heptapeptide member of the renin-angiotensin-aldosterone system that counter-regulates the actions of angiotensin II. Ang-(1-7)-mediated inhibition of EGFR transactivation might represent a potential mechanism of action for its renoprotection. Our review suggests that there is a significant body of evidence supporting the potential inhibition of EGFR/ErbB, and/or administration of Ang-(1-7), as potential novel therapeutic strategies in the treatment of renal pathologies. Thus, EGFR inhibitors such as Gefitinib and Erlinotib that have an acceptable safety profile and have been clinically used in cancer chemotherapy since their FDA approval in the early 2000s, might be considered for repurposing in the treatment of renal pathologies.
Collapse
Affiliation(s)
- Mohamed Tawengi
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Yazan Al-Dali
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Ibrahim F Benter
- Faculty of Pharmacy, Final International University, Kyrenia, Cyprus
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
3
|
Galis P, Bartosova L, Farkasova V, Bartekova M, Ferenczyova K, Rajtik T. Update on clinical and experimental management of diabetic cardiomyopathy: addressing current and future therapy. Front Endocrinol (Lausanne) 2024; 15:1451100. [PMID: 39140033 PMCID: PMC11319149 DOI: 10.3389/fendo.2024.1451100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a severe secondary complication of type 2 diabetes mellitus (T2DM) that is diagnosed as a heart disease occurring in the absence of any previous cardiovascular pathology in diabetic patients. Although it is still lacking an exact definition as it combines aspects of both pathologies - T2DM and heart failure, more evidence comes forward that declares DCM as one complex disease that should be treated separately. It is the ambiguous pathological phenotype, symptoms or biomarkers that makes DCM hard to diagnose and screen for its early onset. This re-view provides an updated look on the novel advances in DCM diagnosis and treatment in the experimental and clinical settings. Management of patients with DCM proposes a challenge by itself and we aim to help navigate and advice clinicians with early screening and pharmacotherapy of DCM.
Collapse
Affiliation(s)
- Peter Galis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
| | - Linda Bartosova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
| | - Veronika Farkasova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| | - Kristina Ferenczyova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomas Rajtik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovakia
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
4
|
Wang Z, Fan H, Wu J. Food-Derived Up-Regulators and Activators of Angiotensin Converting Enzyme 2: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12896-12914. [PMID: 38810024 PMCID: PMC11181331 DOI: 10.1021/acs.jafc.4c01594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a key enzyme in the renin-angiotensin system (RAS), also serving as an amino acid transporter and a receptor for certain coronaviruses. Its primary role is to protect the cardiovascular system via the ACE2/Ang (1-7)/MasR cascade. Given the critical roles of ACE2 in regulating numerous physiological functions, molecules that can upregulate or activate ACE2 show vast therapeutic value. There are only a few ACE2 activators that have been reported, a wide range of molecules, including food-derived compounds, have been reported as ACE2 up-regulators. Effective doses of bioactive peptides range from 10 to 50 mg/kg body weight (BW)/day when orally administered for 1 to 7 weeks. Protein hydrolysates require higher doses at 1000 mg/kg BW/day for 20 days. Phytochemicals and vitamins are effective at doses typically ranging from 10 to 200 mg/kg BW/day for 3 days to 6 months, while Traditional Chinese Medicine requires doses of 1.25 to 12.96 g/kg BW/day for 4 to 8 weeks. ACE2 activation is linked to its hinge-bending region, while upregulation involves various signaling pathways, transcription factors, and epigenetic modulators. Future studies are expected to explore novel roles of ACE2 activators or up-regulators in disease treatments and translate the discovery to bedside applications.
Collapse
Affiliation(s)
- Zihan Wang
- Department
of Agricultural, Food and Nutritional Science, 4-10 Ag/For Building, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
- Cardiovascular
Research Centre, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| | - Hongbing Fan
- Department
of Animal and Food Sciences, University
of Kentucky, Lexington, Kentucky 40546, United States
| | - Jianping Wu
- Department
of Agricultural, Food and Nutritional Science, 4-10 Ag/For Building, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
- Cardiovascular
Research Centre, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| |
Collapse
|
5
|
Hu W, Tan J, Lin Y, Tao Y, Zhou Q. Bibliometric and visual analysis of ACE2/Ang 1-7/MasR axis in diabetes and its microvascular complications from 2000 to 2023. Heliyon 2024; 10:e31405. [PMID: 38807880 PMCID: PMC11130665 DOI: 10.1016/j.heliyon.2024.e31405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024] Open
Abstract
Background The pathogenesis of diabetes and its microvascular complications are intimately associated with renin angiotensin system dysregulation. Evidence suggests the angiotensin converting enzyme 2 (ACE2)/angiotensin 1-7 (Ang 1-7)/Mas receptor (MasR) axis regulates metabolic imbalances, inflammatory responses, reduces oxidative stress, and sustains microvascular integrity, thereby strengthening defences against diabetic conditions. This study aims to conduct a comprehensive analysis of the ACE2/Ang 1-7/MasR axis in diabetes and its microvascular complications over the past two decades, focusing on key contributors, research hotspots, and thematic trends. Methods This cross-sectional bibliometric analysis of 349 English-language publications was performed using HistCite, VOSviewer, CiteSpace, and Bibliometrix R for visualization and metric analysis. Primary analytical metrics included publication count and keyword trend dynamics. Results The United States, contributing 105 articles, emerged as the most productive country, with the University of Florida leading institutions with 18 publications. Benter IF was the most prolific author with 14 publications, and Clinical Science was the leading journal with 13 articles. A total of 151 of the 527 author's keywords with two or more occurrences clustered into four major clusters: diabetic microvascular pathogenesis, metabolic systems, type 2 diabetes, and coronavirus infections. Keywords such as "SARS", "ACE2", "coronavirus", "receptor" and "infection" displayed the strongest citation bursts. The thematic evolution in this field expanded from focusing on the renin angiotensin system (2002-2009) to incorporating ACE2 and diabetes metabolism (2010-2016). The latter period (2017-2023) witnessed a significant surge in diabetes research, reflecting the impact of COVID-19 and associated conditions such as diabetic retinopathy and cardiomyopathy. Conclusions This scientometric study offers a detailed analysis of the ACE2/Ang 1-7/MasR axis in diabetes and its microvascular complications, providing valuable insights for future research directions.
Collapse
Affiliation(s)
- Weiwen Hu
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Jian Tan
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Yeting Lin
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Yulin Tao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Qiong Zhou
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| |
Collapse
|
6
|
Yang DR, Wang MY, Zhang CL, Wang Y. Endothelial dysfunction in vascular complications of diabetes: a comprehensive review of mechanisms and implications. Front Endocrinol (Lausanne) 2024; 15:1359255. [PMID: 38645427 PMCID: PMC11026568 DOI: 10.3389/fendo.2024.1359255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/08/2024] [Indexed: 04/23/2024] Open
Abstract
Diabetic vascular complications are prevalent and severe among diabetic patients, profoundly affecting both their quality of life and long-term prospects. These complications can be classified into macrovascular and microvascular complications. Under the impact of risk factors such as elevated blood glucose, blood pressure, and cholesterol lipids, the vascular endothelium undergoes endothelial dysfunction, characterized by increased inflammation and oxidative stress, decreased NO biosynthesis, endothelial-mesenchymal transition, senescence, and even cell death. These processes will ultimately lead to macrovascular and microvascular diseases, with macrovascular diseases mainly characterized by atherosclerosis (AS) and microvascular diseases mainly characterized by thickening of the basement membrane. It further indicates a primary contributor to the elevated morbidity and mortality observed in individuals with diabetes. In this review, we will delve into the intricate mechanisms that drive endothelial dysfunction during diabetes progression and its associated vascular complications. Furthermore, we will outline various pharmacotherapies targeting diabetic endothelial dysfunction in the hope of accelerating effective therapeutic drug discovery for early control of diabetes and its vascular complications.
Collapse
Affiliation(s)
- Dong-Rong Yang
- Department of Endocrinology and Metabolism, Shenzhen University General Hospital, Shenzhen, Guangdong, China
- Department of Pathophysiology, Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Meng-Yan Wang
- Department of Pathophysiology, Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Cheng-Lin Zhang
- Department of Pathophysiology, Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Yu Wang
- Department of Endocrinology and Metabolism, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Molaei A, Molaei E, Hayes AW, Karimi G. Mas receptor: a potential strategy in the management of ischemic cardiovascular diseases. Cell Cycle 2023; 22:1654-1674. [PMID: 37365840 PMCID: PMC10361149 DOI: 10.1080/15384101.2023.2228089] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/10/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
MasR is a critical element in the RAS accessory pathway that protects the heart against myocardial infarction, ischemia-reperfusion injury, and pathological remodeling by counteracting the effects of AT1R. This receptor is mainly stimulated by Ang 1-7, which is a bioactive metabolite of the angiotensin produced by ACE2. MasR activation attenuates ischemia-related myocardial damage by facilitating vasorelaxation, improving cell metabolism, reducing inflammation and oxidative stress, inhibiting thrombosis, and stabilizing atherosclerotic plaque. It also prevents pathological cardiac remodeling by suppressing hypertrophy- and fibrosis-inducing signals. In addition, the potential of MasR in lowering blood pressure, improving blood glucose and lipid profiles, and weight loss has made it effective in modulating risk factors for coronary artery disease including hypertension, diabetes, dyslipidemia, and obesity. Considering these properties, the administration of MasR agonists offers a promising approach to the prevention and treatment of ischemic heart disease.Abbreviations: Acetylcholine (Ach); AMP-activated protein kinase (AMPK); Angiotensin (Ang); Angiotensin receptor (ATR); Angiotensin receptor blocker (ARB); Angiotensin-converting enzyme (ACE); Angiotensin-converting enzyme inhibitor (ACEI); Anti-PRD1-BF1-RIZ1 homologous domain containing 16 (PRDM16); bradykinin (BK); Calcineurin (CaN); cAMP-response element binding protein (CREB); Catalase (CAT); C-C Motif Chemokine Ligand 2 (CCL2); Chloride channel 3 (CIC3); c-Jun N-terminal kinases (JNK); Cluster of differentiation 36 (CD36); Cocaine- and amphetamine-regulated transcript (CART); Connective tissue growth factor (CTGF); Coronary artery disease (CAD); Creatine phosphokinase (CPK); C-X-C motif chemokine ligand 10 (CXCL10); Cystic fibrosis transmembrane conductance regulator (CFTR); Endothelial nitric oxide synthase (eNOS); Extracellular signal-regulated kinase 1/2 (ERK 1/2); Fatty acid transport protein (FATP); Fibroblast growth factor 21 (FGF21); Forkhead box protein O1 (FoxO1); Glucokinase (Gk); Glucose transporter (GLUT); Glycogen synthase kinase 3β (GSK3β); High density lipoprotein (HDL); High sensitive C-reactive protein (hs-CRP); Inositol trisphosphate (IP3); Interleukin (IL); Ischemic heart disease (IHD); Janus kinase (JAK); Kruppel-like factor 4 (KLF4); Lactate dehydrogenase (LDH); Left ventricular end-diastolic pressure (LVEDP); Left ventricular end-systolic pressure (LVESP); Lipoprotein lipase (LPL); L-NG-Nitro arginine methyl ester (L-NAME); Low density lipoprotein (LDL); Mammalian target of rapamycin (mTOR); Mas-related G protein-coupled receptors (Mrgpr); Matrix metalloproteinase (MMP); MAPK phosphatase-1 (MKP-1); Mitogen-activated protein kinase (MAPK); Monocyte chemoattractant protein-1 (MCP-1); NADPH oxidase (NOX); Neuropeptide FF (NPFF); Neutral endopeptidase (NEP); Nitric oxide (NO); Nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB); Nuclear-factor of activated T-cells (NFAT); Pancreatic and duodenal homeobox 1 (Pdx1); Peroxisome proliferator- activated receptor γ (PPARγ); Phosphoinositide 3-kinases (PI3k); Phospholipase C (PLC); Prepro-orexin (PPO); Prolyl-endopeptidase (PEP); Prostacyclin (PGI2); Protein kinase B (Akt); Reactive oxygen species (ROS); Renin-angiotensin system (RAS); Rho-associated protein kinase (ROCK); Serum amyloid A (SAA); Signal transducer and activator of transcription (STAT); Sirtuin 1 (Sirt1); Slit guidance ligand 3 (Slit3); Smooth muscle 22α (SM22α); Sterol regulatory element-binding protein 1 (SREBP-1c); Stromal-derived factor-1a (SDF); Superoxide dismutase (SOD); Thiobarbituric acid reactive substances (TBARS); Tissue factor (TF); Toll-like receptor 4 (TLR4); Transforming growth factor β1 (TGF-β1); Tumor necrosis factor α (TNF-α); Uncoupling protein 1 (UCP1); Ventrolateral medulla (VLM).
Collapse
Affiliation(s)
- Ali Molaei
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Emad Molaei
- PharmD, Assistant of Clinical Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - A. Wallace Hayes
- University of South Florida College of Public Health, Tampa, Florida, USA
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Rysz J, Gluba-Brzózka A, Rokicki R, Franczyk B. Oxidative Stress-Related Susceptibility to Aneurysm in Marfan's Syndrome. Biomedicines 2021; 9:biomedicines9091171. [PMID: 34572356 PMCID: PMC8467736 DOI: 10.3390/biomedicines9091171] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 01/01/2023] Open
Abstract
The involvement of highly reactive oxygen-derived free radicals (ROS) in the genesis and progression of various cardiovascular diseases, including arrhythmias, aortic dilatation, aortic dissection, left ventricular hypertrophy, coronary arterial disease and congestive heart failure, is well-established. It has also been suggested that ROS may play a role in aortic aneurysm formation in patients with Marfan's syndrome (MFS). This syndrome is a multisystem disorder with manifestations including cardiovascular, skeletal, pulmonary and ocular systems, however, aortic aneurysm and dissection are still the most life-threatening manifestations of MFS. In this review, we will concentrate on the impact of oxidative stress on aneurysm formation in patients with MFS as well as on possible beneficial effects of some agents with antioxidant properties. Mechanisms responsible for oxidative stress in the MFS model involve a decreased expression of superoxide dismutase (SOD) as well as enhanced expression of NAD(P)H oxidase, inducible nitric oxide synthase (iNOS) and xanthine oxidase. The results of studies have indicated that reactive oxygen species may be involved in smooth muscle cell phenotype switching and apoptosis as well as matrix metalloproteinase activation, resulting in extracellular matrix (ECM) remodeling. The progression of the thoracic aortic aneurysm was suggested to be associated with markedly impaired aortic contractile function and decreased nitric oxide-mediated endothelial-dependent relaxation.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
- Correspondence: or ; Tel.: +48-42-639-3750
| | - Robert Rokicki
- Clinic of Hand Surgery, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| |
Collapse
|
9
|
Shraim BA, Moursi MO, Benter IF, Habib AM, Akhtar S. The Role of Epidermal Growth Factor Receptor Family of Receptor Tyrosine Kinases in Mediating Diabetes-Induced Cardiovascular Complications. Front Pharmacol 2021; 12:701390. [PMID: 34408653 PMCID: PMC8365470 DOI: 10.3389/fphar.2021.701390] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus is a major debilitating disease whose global incidence is progressively increasing with currently over 463 million adult sufferers and this figure will likely reach over 700 million by the year 2045. It is the complications of diabetes such as cardiovascular, renal, neuronal and ocular dysfunction that lead to increased patient morbidity and mortality. Of these, cardiovascular complications that can result in stroke and cardiomyopathies are 2- to 5-fold more likely in diabetes but the underlying mechanisms involved in their development are not fully understood. Emerging research suggests that members of the Epidermal Growth Factor Receptor (EGFR/ErbB/HER) family of tyrosine kinases can have a dual role in that they are beneficially required for normal development and physiological functioning of the cardiovascular system (CVS) as well as in salvage pathways following acute cardiac ischemia/reperfusion injury but their chronic dysregulation may also be intricately involved in mediating diabetes-induced cardiovascular pathologies. Here we review the evidence for EGFR/ErbB/HER receptors in mediating these dual roles in the CVS and also discuss their potential interplay with the Renin-Angiotensin-Aldosterone System heptapeptide, Angiotensin-(1-7), as well the arachidonic acid metabolite, 20-HETE (20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid). A greater understanding of the multi-faceted roles of EGFR/ErbB/HER family of tyrosine kinases and their interplay with other key modulators of cardiovascular function could facilitate the development of novel therapeutic strategies for treating diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Bara A Shraim
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Moaz O Moursi
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ibrahim F Benter
- Faculty of Medicine, Eastern Mediterranean University, Famagusta, North Cyprus
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
10
|
Rao H, Jalali JA, Johnston TP, Koulen P. Emerging Roles of Dyslipidemia and Hyperglycemia in Diabetic Retinopathy: Molecular Mechanisms and Clinical Perspectives. Front Endocrinol (Lausanne) 2021; 12:620045. [PMID: 33828528 PMCID: PMC8020813 DOI: 10.3389/fendo.2021.620045] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Diabetic retinopathy (DR) is a significant cause of vision loss and a research subject that is constantly being explored for new mechanisms of damage and potential therapeutic options. There are many mechanisms and pathways that provide numerous options for therapeutic interventions to halt disease progression. The purpose of the present literature review is to explore both basic science research and clinical research for proposed mechanisms of damage in diabetic retinopathy to understand the role of triglyceride and cholesterol dysmetabolism in DR progression. This review delineates mechanisms of damage secondary to triglyceride and cholesterol dysmetabolism vs. mechanisms secondary to diabetes to add clarity to the pathogenesis behind each proposed mechanism. We then analyze mechanisms utilized by both triglyceride and cholesterol dysmetabolism and diabetes to elucidate the synergistic, additive, and common mechanisms of damage in diabetic retinopathy. Gathering this research adds clarity to the role dyslipidemia has in DR and an evaluation of the current peer-reviewed basic science and clinical evidence provides a basis to discern new potential therapeutic targets.
Collapse
Affiliation(s)
- Hussain Rao
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Jonathan A. Jalali
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Thomas P. Johnston
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri – Kansas City, Kansas City, MO, United States
| | - Peter Koulen
- Department of Ophthalmology, School of Medicine, Vision Research Center, University of Missouri – Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, United States
- *Correspondence: Peter Koulen,
| |
Collapse
|
11
|
Li J, Zhu R, Liu Y, Yang J, Wang X, Geng L, Xu T, He J. Angiotensin-(1-7) Improves Islet Function in a Rat Model of Streptozotocin- Induced Diabetes Mellitus by Up-Regulating the Expression of Pdx1/Glut2. Endocr Metab Immune Disord Drug Targets 2021; 21:156-162. [PMID: 32679026 DOI: 10.2174/1871530320666200717161538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To observe the effects of angiotensin-(1-7) (Ang-(1-7)) on glucose metabolism, islet function and insulin resistance in a rat model of streptozotocin-induced diabetes mellitus (DM) and investigate its mechanism. METHODS Thirty-four male Wistar rats were randomly divided into 3 groups: control group, which was fed a standard diet, DM group, high-fat diet and injected with streptozotocin, and Ang-(1-7) group receiving an injection of streptozotocin followed by Ang-(1-7) treatment. Blood glucose level, fasting serum Ang II and insulin levels, and homeostasis model assessment of insulin resistance (HOMA-IR) were measured. The pancreases were collected for histological examination, protein and gene expression analysis. RESULTS Compared with the control group, fasting blood glucose, serum angiotensin II level, and HOMA-IR value increased, while serum insulin level decreased in the DM group. Moreover, islet structure was damaged, β cells were irregularly arranged, the cytoplasm was loose in the DM group. Expressions of Pancreatic duodenal homeobox-1 (Pdx1), glucose transporter-2 (Glut2) and glucokinase (Gk) were significantly decreased in the DM group compared with the control group. However, the DM-associated changes were dramatically reversed following Ang-(1-7) treatment. CONCLUSION Ang-(1-7) protects against streptozotocin-induced DM through the improvement of insulin secretion, insulin resistance and islet fibrosis, which is associated with the upregulation of Pdx1, Glut2 and Gk expressions.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruifang Zhu
- School of Nursing, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yalin Liu
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinhui Yang
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoyan Wang
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lisha Geng
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tingting Xu
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Junhua He
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
12
|
Akhtar S, Benter IF, Danjuma MI, Doi SAR, Hasan SS, Habib AM. Pharmacotherapy in COVID-19 patients: a review of ACE2-raising drugs and their clinical safety. J Drug Target 2020; 28:683-699. [PMID: 32700580 DOI: 10.1080/1061186x.2020.1797754] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic is caused by the severe acute-respiratory-syndrome-coronavirus-2 that uses ACE2 as its receptor. Drugs that raise serum/tissue ACE2 levels include ACE inhibitors (ACEIs) and angiotensin-II receptor blockers (ARBs) that are commonly used in patients with hypertension, cardiovascular disease and/or diabetes. These comorbidities have adverse outcomes in COVID-19 patients that might result from pharmacotherapy. Increasing ACE2 could potentially increase the risk of infection, severity or mortality in COVID-19 or it might be protective as it forms angiotensin-(1-7) which exhibits anti-inflammatory/anti-oxidative effects and prevents diabetes- and/or hypertension-induced end-organ damage. Thus, there existed clinical uncertainty. Here, we review studies implicating 15 classes of drugs in increasing ACE2 levels in vivo and the available literature on the clinical safety of these drugs in COVID-19 patients. Further, in a re-analysis of clinical data from a meta-analysis of 9 studies, we show that ACEIs/ARBs usage was not associated with an increased risk of all-cause mortality. Literature suggests that ACEIs/ARBs usage generally appears to be clinically safe though their use in severe COVID-19 patients might increase the risk of acute renal injury. For definitive clarity, further clinical and mechanistic studies are needed in assessing the safety of all classes of ACE2 raising medications.
Collapse
Affiliation(s)
- Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ibrahim F Benter
- Faculty of Medicine, Eastern Mediterranean University, Famagusta, North Cyprus
| | - Mohammed I Danjuma
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Division of Internal Medicine, Hamad Medical Corporation Hospital, Doha, Qatar
| | - Suhail A R Doi
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Syed S Hasan
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
13
|
Mohammed El Tabaa M, Mohammed El Tabaa M. Targeting Neprilysin (NEP) pathways: A potential new hope to defeat COVID-19 ghost. Biochem Pharmacol 2020; 178:114057. [PMID: 32470547 PMCID: PMC7250789 DOI: 10.1016/j.bcp.2020.114057] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023]
Abstract
COVID-19 is an ongoing viral pandemic disease that is caused by SARS-CoV2, inducing severe pneumonia in humans. However, several classes of repurposed drugs have been recommended, no specific vaccines or effective therapeutic interventions for COVID-19 are developed till now. Viral dependence on ACE-2, as entry receptors, drove the researchers into RAS impact on COVID-19 pathogenesis. Several evidences have pointed at Neprilysin (NEP) as one of pulmonary RAS components. Considering the protective effect of NEP against pulmonary inflammatory reactions and fibrosis, it is suggested to direct the future efforts towards its potential role in COVID-19 pathophysiology. Thus, the review aimed to shed light on the potential beneficial effects of NEP pathways as a novel target for COVID-19 therapy by summarizing its possible molecular mechanisms. Additional experimental and clinical studies explaining more the relationships between NEP and COVID-19 will greatly benefit in designing the future treatment approaches.
Collapse
Affiliation(s)
- Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute, University of Sadat City, Egypt.
| | | |
Collapse
|
14
|
Comparison of Candesartan and Angiotensin-(1-7) Combination to Mito-TEMPO Treatment for Normalizing Blood Pressure and Sympathovagal Balance in (mREN2)27 Rats. J Cardiovasc Pharmacol 2020; 73:143-148. [PMID: 30540685 DOI: 10.1097/fjc.0000000000000645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Hypertensive transgenic (mRen2)27 rats exhibit impaired baroreflex sensitivity (BRS) for control of heart rate (HR). Intracerebroventricular infusion of Ang-(1-7) improves indices of vagal BRS independent of lowering mean arterial pressure (MAP), whereas AT1 receptor blockade normalizes MAP and indices of sympathetic tone without correcting the vagal BRS. Scavenging cellular reactive oxygen species (ROS) with tempol in brain fails to correct either hypertension or sympathovagal balance in these animals, despite reports that mitochondrial ROS contributes to Ang II-infusion hypertension. To examine effects of a putative preferential mitochondrial ROS scavenger in the brain of (mRen2)27 rats, ICV infusions of Mito-TEMPO (3.2 μg/2.5 μL/h) were compared with artificial cerebrospinal fluid (aCSF; 2.5 μL/h) and combination AT1 receptor antagonist candesartan (CAN: 4 μg/2.5 μL/h) plus Ang-(1-7) (0.1 μg/2.5 μL/h) treatment. MAP was lower after CAN + Ang-(1-7) treatment, and both vagal and sympathetic components of BRS and sympathovagal balance were improved. By contrast, Mito-TEMPO improved sympathetic components of BRS and tended to improve overall sympathovagal balance but failed to alter MAP in this model of hypertension. Although further studies are required to determine whether Mito-TEMPO or CAN + Ang-(1-7) treatment at the doses used altered mitochondrial ROS, optimal therapeutic benefits are achieved by shifting the balance from Ang II toward Ang-(1-7) in this model of chronic RAS-dependent hypertension.
Collapse
|
15
|
Rosiglitazone Suppresses Calcium Oxalate Crystal Binding and Oxalate-Induced Oxidative Stress in Renal Epithelial Cells by Promoting PPAR- γ Activation and Subsequent Regulation of TGF- β1 and HGF Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4826525. [PMID: 31781338 PMCID: PMC6875173 DOI: 10.1155/2019/4826525] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/11/2019] [Accepted: 09/17/2019] [Indexed: 12/21/2022]
Abstract
Peroxisome proliferator-activated receptor- (PPAR-) γ is a ligand-dependent transcription factor, and it has become evident that PPAR-γ agonists have renoprotective effects, but their influence and mechanism during the development of calcium oxalate (CaOx) nephrolithiasis remain unknown. Rosiglitazone (RSG) was used as a representative PPAR-γ agonist in our experiments. The expression of transforming growth factor-β1 (TGF-β1), hepatocyte growth factor (HGF), c-Met, p-Met, PPAR-γ, p-PPAR-γ (Ser112), Smad2, Smad3, pSmad2/3, and Smad7 was examined in oxalate-treated Madin-Darby canine kidney (MDCK) cells and a stone-forming rat model. A CCK-8 assay was used to evaluate the effects of RSG on cell viability. In addition, intracellular reactive oxygen species (ROS) levels were monitored, and lipid peroxidation in renal tissue was detected according to superoxide dismutase and malondialdehyde levels. Moreover, the location and extent of CaOx crystal deposition were evaluated by Pizzolato staining. Our results showed that, both in vitro and in vivo, oxalate impaired PPAR-γ expression and phosphorylation, and then accumulative ROS production was observed, accompanied by enhanced TGF-β1 and reduced HGF. These phenomena could be reversed by the addition of RSG. RSG also promoted cell viability and proliferation and decreased oxidative stress damage and CaOx crystal deposition. However, these protective effects of RSG were abrogated by the PPAR-γ-specific inhibitor GW9662. Our results revealed that the reduction of PPAR-γ activity played a critical role in oxalate-induced ROS damage and CaOx stone formation. RSG can regulate TGF-β1 and HGF/c-Met through PPAR-γ to exert antioxidant effects against hyperoxaluria and alleviate crystal deposition. Therefore, PPAR-γ agonists may be expected to be a novel therapy for nephrolithiasis, and this effect is related to PPAR-γ-dependent suppression of oxidative stress.
Collapse
|
16
|
Kim SM, Jung JI, Chai C, Imm JY. Characteristics and Glucose Uptake Promoting Effect of Chrysin-Loaded Phytosomes Prepared with Different Phospholipid Matrices. Nutrients 2019; 11:E2549. [PMID: 31652637 PMCID: PMC6835247 DOI: 10.3390/nu11102549] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022] Open
Abstract
Chrysin-loaded phytosomes (CP) were prepared using either soya phosphatidylcholine (SPC) or egg phospholipid (EPL) by the solvent evaporation method. Different phospholipid matrices resulted in significant differences in size, mechanical property and solubility of the CP. The most stable CP was obtained with EPL at a molar ratio of 1:3 (chrysin: EPL, CEP-1:3). CEP-1:3 displayed an average size of 117 nm with uniform size distribution (polydispersity index: 0.30) and zeta potential of -31 mV. A significantly greater elastic modulus of CEP-1:3 (2.7-fold) indicated tighter packing and strong molecular bonding than those of CP prepared with SPC (CSP-1:3). X-ray diffraction and Fourier transform infrared spectroscopic analysis of CEP-1:3 confirmed molecular complexation. CEP-1:3 displayed a greater glucose uptake promoting effect than free chrysin and CSP-1:3 in muscle cells by stimulating gene expression of peroxisome proliferator-activated receptor γ and glucose transporter type 4. The results of the present study suggest that the phospholipid matrix used for the preparation of phytosomes critically influences their performance.
Collapse
Affiliation(s)
- Seong-Min Kim
- Department of Foods and Nutrition, Kookmin University, Seoul 02707, Korea.
| | - Jae-In Jung
- Department of Foods and Nutrition, Kookmin University, Seoul 02707, Korea.
| | - Changhoon Chai
- Department of Applied Animal Science, Kangwon National University, Chuncheon 24341, Korea.
| | - Jee-Young Imm
- Department of Foods and Nutrition, Kookmin University, Seoul 02707, Korea.
| |
Collapse
|
17
|
El-Saka MH, Madi NM, Ibrahim RR, Alghazaly GM, Elshwaikh S, El-Bermawy M. The ameliorative effect of angiotensin 1-7 on experimentally induced-preeclampsia in rats: Targeting the role of peroxisome proliferator-activated receptors gamma expression & asymmetric dimethylarginine. Arch Biochem Biophys 2019; 671:123-129. [PMID: 31295432 DOI: 10.1016/j.abb.2019.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/02/2019] [Accepted: 07/07/2019] [Indexed: 01/10/2023]
Abstract
This study was designed to explore the effect of angiotensin 1-7 (Ang 1-7) on experimentally induced-preeclampsia in Wistar rats targeting the role of peroxisome proliferator-activated receptors gamma expression (PPARs-γ) & asymmetric dimethylarginine (ADMA). 30 female Wistar rats were divided into three groups: Normal pregnant (NP), preeclampsia (PE), and preeclampsia treated with Ang 1-7 (PE + Ang 1-7) groups. Reduced uterine perfusion pressure (RUPP) model was induced on GD14. On GD18, protein in urine, urine volume and urinary sodium excretion were determined. On GD19, the systolic blood pressure (SBP) was measured, and the gene expression of PPARs-γ were determined. The serum samples were separated for determination of Ang 1-7, ADMA, soluble fms-like tyrosine kinase (sFlt-1), vascular endothelial growth factor (VEGF), nitric oxide (NO) products, endothelial nitric oxide synthase (eNOS) activity, interleukin-6 (IL-6), interleukin-10 (IL-10), malondialdehyde (MDA), and total anti-oxidant capacity (T-AOC). Compared to NP group, SBP, urine protein, serum levels of ADMA, sFlt-1, IL-6 and MDA significantly increased, while expression of PPARs-γ, serum levels of Ang 1-7, VEGF, NO products, eNOS, IL-10 and T-AOC significantly decreased in PE group, while treatment of Ang 1-7 significantly ameliorated all these studied parameters as compared to PE group. We concluded that Ang 1-7 attenuated the symptoms of preeclampsia, which might be via increasing the expression of PPARs-γ and reduction of ADMA levels which could explain its anti-hypertensive, anti-angiogenic, anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Mervat H El-Saka
- The Department of Physiology, Faculty of Medicine, Tanta University, Egypt.
| | - Nermin M Madi
- The Department of Physiology, Faculty of Medicine, Tanta University, Egypt
| | - Rowida Raafat Ibrahim
- The Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Tanta University, Egypt
| | | | - Shereef Elshwaikh
- The Department of Gynecology and Obstetric, Faculty of Medicine, Tanta University, Egypt
| | - Manal El-Bermawy
- The Department of Anatomy, Faculty of Medicine, Tanta University, Egypt
| |
Collapse
|
18
|
Chen Y, Zhao W, Liu C, Meng W, Zhao T, Bhattacharya SK, Sun Y. Molecular and Cellular Effect of Angiotensin 1-7 on Hypertensive Kidney Disease. Am J Hypertens 2019; 32:460-467. [PMID: 30715105 DOI: 10.1093/ajh/hpz009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/25/2018] [Accepted: 01/15/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Studies implicate that angiotensin 1-7 (Ang1-7) imparts protective effects in the kidney. However, its relevance in hypertensive kidney disease is not fully understood. The purpose of this study was to explore the role of Ang1-7 on renal damage/remodeling during hypertension and its potential underlying molecular-cellular mechanisms. METHODS Hypertension was induced in adult Sprague-Dawley rats by infusion of aldosterone (ALDO; 0.75 μg/hour) for 4 weeks with or without co-treatment of Ang1-7 (1 mg/kg/day). Untreated rats served as controls. Systolic blood pressure was monitored by tail-cuff technique. Renal fibrosis was evaluated by picrosirius red staining and renal collagen volume fraction was quantitated using imaging analyzing system. The expression of profibrotic factors [transforming growth factor-β1 (TGF-β1), platelet-derived growth factor-D (PDGF-D), fibroblast growth factor-1 (FGF-1), vascular endothelial growth factor-D (VEGF-D), and tissue inhibitors of metalloproteinases (TIMPs)] and free radical producing enzymes (inducible nitric oxide synthase and nicotinamide adenine dinucleotide phosphate [NADPH] oxidase) in the kidney were examined by reverse transcription-polymerase chain reaction and western blot. Renal oxidative stress was assessed by malondialdehyde (MDA) measurement. RESULTS Chronic ALDO infusion caused hypertension and hypertensive renal disease represented as glomerular damage/sclerosis. Ang1-7 co-treatment did not affect blood pressure in ALDO-treated rats, but significantly attenuated the glomerular damage/fibrosis. ALDO treatment significantly elevated renal expression of profibrogenic factors, including TGF-β1, TIMP-1/TIMP-2, FGF-1, PDGF-D, and VEGF-D, whereas Ang1-7 co-treatment significantly reduced renal TGF-β1, TIMP-1/TIMP-2, and FGF-1, but not PDGF-D and VEGF-D. Furthermore, ALDO infusion elevated NADPH oxidase (gp91phox) and MDA in the kidney, which was attenuated by Ang1-7 co-treatment. CONCLUSIONS Ang1-7 plays a protective role in the hypertensive kidney disease independent of blood pressure. The beneficial effects of Ang1-7 are likely mediated via suppressing TGF-β/FGF-1 pathways and oxidative stress.
Collapse
Affiliation(s)
- Yuanjian Chen
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Wenyuan Zhao
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Chang Liu
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Weixin Meng
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Tieqiang Zhao
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Syamal K Bhattacharya
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Yao Sun
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
19
|
Long-Term Administration of Angiotensin (1-7) to db/db Mice Reduces Oxidative Stress Damage in the Kidneys and Prevents Renal Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1841046. [PMID: 30425780 PMCID: PMC6218718 DOI: 10.1155/2018/1841046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/04/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023]
Abstract
Aims The goal of this study was to evaluate the effects of long-term (16 weeks) administration of angiotensin (1–7) [A(1–7)] on kidney function in db/db mice and to identify the protective mechanisms of this therapy. Methods db/db mice and heterozygous controls were treated with A(1–7) or vehicle daily, subcutaneously for up to 16 weeks. Kidney injury was assessed by measuring blood flow in renal arteries, plasma creatinine levels, and proteinuria. Effects of treatment on oxidative stress were evaluated by histological staining and gene expression. Results 16 weeks of daily administration of A(1–7) to a mouse model of severe type 2 diabetes (db/db) prevented the progression of kidney damage. Treatment with A(1–7) improved blood flow in the renal arteries, as well as decreased plasma creatinine levels and proteinuria in diabetic mice. Reduction of oxidative stress was identified as one of the mechanisms of the renoprotective action of A(1–7). Treatment prevented formation of nitrotyrosine residues, a marker of oxidative stress damage. A(1–7) also reduced the expression of two enzymes involved in formation of nitrotyrosine, namely, eNOS and NOX-4. A(1–7) regulated the phosphorylation pattern of eNOS to enhance production of NO in diabetic animals, possibly through the Akt pathway. However, these elevated levels of NO did not result in increased nitrosylation, possibly due to reduced NOX-4 levels. Conclusions Long-term administration of A(1–7) improved kidney function and reduced oxidative stress damage in db/db mice.
Collapse
|
20
|
Santos RAS, Sampaio WO, Alzamora AC, Motta-Santos D, Alenina N, Bader M, Campagnole-Santos MJ. The ACE2/Angiotensin-(1-7)/MAS Axis of the Renin-Angiotensin System: Focus on Angiotensin-(1-7). Physiol Rev 2018; 98:505-553. [PMID: 29351514 PMCID: PMC7203574 DOI: 10.1152/physrev.00023.2016] [Citation(s) in RCA: 772] [Impact Index Per Article: 110.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 05/09/2017] [Accepted: 06/18/2017] [Indexed: 12/16/2022] Open
Abstract
The renin-angiotensin system (RAS) is a key player in the control of the cardiovascular system and hydroelectrolyte balance, with an influence on organs and functions throughout the body. The classical view of this system saw it as a sequence of many enzymatic steps that culminate in the production of a single biologically active metabolite, the octapeptide angiotensin (ANG) II, by the angiotensin converting enzyme (ACE). The past two decades have revealed new functions for some of the intermediate products, beyond their roles as substrates along the classical route. They may be processed in alternative ways by enzymes such as the ACE homolog ACE2. One effect is to establish a second axis through ACE2/ANG-(1-7)/MAS, whose end point is the metabolite ANG-(1-7). ACE2 and other enzymes can form ANG-(1-7) directly or indirectly from either the decapeptide ANG I or from ANG II. In many cases, this second axis appears to counteract or modulate the effects of the classical axis. ANG-(1-7) itself acts on the receptor MAS to influence a range of mechanisms in the heart, kidney, brain, and other tissues. This review highlights the current knowledge about the roles of ANG-(1-7) in physiology and disease, with particular emphasis on the brain.
Collapse
Affiliation(s)
- Robson Augusto Souza Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Walkyria Oliveira Sampaio
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Andreia C Alzamora
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Daisy Motta-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Natalia Alenina
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Michael Bader
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Maria Jose Campagnole-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
21
|
Jayakumar V, Ahmed SSSJ, Ebenezar KK. Multivariate analysis and molecular interaction of curcumin with PPARγ in high fructose diet induced insulin resistance in rats. SPRINGERPLUS 2016; 5:1732. [PMID: 27777867 PMCID: PMC5053957 DOI: 10.1186/s40064-016-3364-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/22/2016] [Indexed: 11/13/2022]
Abstract
To investigate the effect of curcumin on the multivariate and docking analysis on peroxisome proliferator activated receptor-γ, the rats were fed with high fructose diet (Group 2) to induce insulin resistance and curcumin was co-administered orally (Group 4) for a period of 8 weeks and measured the biochemical parameters in blood, kidney and liver tissues. The results showed a significant (p ≤ 0.05) increase in the level of creatinine, glucose, insulin, low density lipoprotein, total cholesterol, triglyceride, urea, uric acid, very low density lipoprotein and decreased albumin, high density lipoprotein and total protein level in the blood of Group 2 when compared with Group 1 control rats. Further, analysis on liver and kidney tissues showed a significant decrease in antioxidants, hexokinase and increased glucose 6-phosphatase and fructose 1,6-bisphosphatase, hydroperoxides and TBARS in Group 2 rats. Furthermore, the multivariate and loading coefficient analysis showed that albumin, HDL, catalase, glutathione reductase, hexokinase and vitamin E are the most contributing factors in blood, liver and kidney. Subsequently, molecular docking was carried out to determine the binding efficiency of curcumin as agonist of PPARγ showed high affinity compared to pioglitazone. The histology of liver and kidney were also studied and the administration of curcumin along with fructose protects the organs from the abnormal changes and also prevents the fat accumulation. Overall, these results demonstrate the preventive role of curcumin on diet induced insulin resistant in rats by ameliorating the altered levels of metabolic changes and potential binding of curcumin with PPARγ as agonist in the treatment of insulin resistance.
Collapse
Affiliation(s)
- Vasanthi Jayakumar
- Molecular Physiology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Rajiv Gandhi Salai, Kelambakkam, Tamil Nadu 603103 India
| | - Shiek S. S. J. Ahmed
- Drug Discovery Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu 603103 India
| | - Kesavarao Kumar Ebenezar
- Molecular Physiology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Rajiv Gandhi Salai, Kelambakkam, Tamil Nadu 603103 India
| |
Collapse
|
22
|
Apocynin influence on oxidative stress and cardiac remodeling of spontaneously hypertensive rats with diabetes mellitus. Cardiovasc Diabetol 2016; 15:126. [PMID: 27585437 PMCID: PMC5009715 DOI: 10.1186/s12933-016-0442-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/18/2016] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Although increased oxidative stress is a major component of diabetic hypertensive cardiomyopathy, research into the effects of antioxidants on cardiac remodeling remains scarce. The actions of antioxidant apocynin include inhibiting reactive oxygen species (ROS) generation by nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and ROS scavenging. We evaluated the effects of apocynin on cardiac remodeling in spontaneously hypertensive rats (SHR) with diabetes mellitus (DM). METHODS Male SHR were divided into four groups: control (SHR, n = 16); SHR treated with apocynin (SHR-APO; 16 mg/kg/day, added to drinking water; n = 16); diabetic SHR (SHR-DM, n = 13); and SHR-DM treated with apocynin (SHR-DM-APO, n = 14), for eight weeks. DM was induced by streptozotocin (40 mg/kg, single dose). Statistical analyzes: ANOVA and Tukey or Mann-Whitney. RESULTS Echocardiogram in diabetic groups showed higher left ventricular and left atrium diameters indexed for body weight, and higher isovolumetric relaxation time than normoglycemic rats; systolic function did not differ between groups. Isolated papillary muscle showed impaired contractile and relaxation function in diabetic groups. Developed tension was lower in SHR-APO than SHR. Myocardial hydroxyproline concentration was higher in SHR-DM than SHR, interstitial collagen fraction was higher in SHR-DM-APO than SHR-APO, and type III collagen protein expression was lower in SHR-DM and SHR-DM-APO than their controls. Type I collagen and lysyl oxidase expression did not differ between groups. Apocynin did not change collagen tissue. Myocardial lipid hydroperoxide concentration was higher in SHR-DM than SHR and SHR-DM-APO. Glutathione peroxidase activity was lower and catalase higher in SHR-DM than SHR. Apocynin attenuated antioxidant enzyme activity changes in SHR-DM-APO. Advanced glycation end-products and NADPH oxidase activity did not differ between groups. CONCLUSION Apocynin reduces oxidative stress independently of NADPH oxidase activity and does not change ventricular or myocardial function in spontaneously hypertensive rats with diabetes mellitus. The apocynin-induced myocardial functional impairment in SHR shows that apocynin actions need to be clarified during sustained chronic pressure overload.
Collapse
|
23
|
Gromotowicz-Poplawska A, Szoka P, Kolodziejczyk P, Kramkowski K, Wojewodzka-Zelezniakowicz M, Chabielska E. New agents modulating the renin-angiotensin-aldosterone system-Will there be a new therapeutic option? Exp Biol Med (Maywood) 2016; 241:1888-1899. [PMID: 27439538 DOI: 10.1177/1535370216660211] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/22/2016] [Indexed: 12/19/2022] Open
Abstract
The renin-angiotensin-aldosterone system (RAAS) is more complex than it was originally regarded. According to the current subject knowledge, there are two main axes of the RAAS: (1) angiotensin-converting enzyme (ACE)-angiotensin II-AT1 receptor axis and (2) ACE2-angiotensin-(1-7)-Mas receptor axis. The activation of the first axis leads to deleterious effects, including vasoconstriction, endothelial dysfunction, thrombosis, inflammation, and fibrosis; therefore, blocking the components of this axis is a highly rational and commonly used therapeutic procedure. The ACE2-Ang-(1-7)-Mas receptor axis has a different role, since it often opposes the effects induced by the classical ACE-Ang II-AT1 axis. Once the positive effects of the ACE2-Ang-(1-7)-Mas axis were discovered, the alternative ways of pharmacotherapy activating this axis of RAAS appeared. This article briefly describes new molecules affecting the RAAS, namely: recombinant human ACE2, ACE2 activators, angiotensin-(1-7) peptide and non-peptide analogs, aldosterone synthase inhibitors, and the third and fourth generation of mineralocorticoid receptor antagonists. The results of the experimental and clinical studies are encouraging, which leads us to believe that these new molecules can support the treatment of cardiovascular diseases as well as cardiometabolic disorders.
Collapse
Affiliation(s)
| | - Piotr Szoka
- Department of Biopharmacy, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Patrycjusz Kolodziejczyk
- Department of Pharmaceutical Analysis, Medical University of Bialystok, 15-522 Bialystok, Poland
| | - Karol Kramkowski
- Department of Biopharmacy, Medical University of Bialystok, 15-089 Bialystok, Poland
| | | | - Ewa Chabielska
- Department of Biopharmacy, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
24
|
Zhang F, Liu C, Wang L, Cao X, Wang YY, Yang JK. Antioxidant effect of angiotensin (1‑7) in the protection of pancreatic β cell function. Mol Med Rep 2016; 14:1963-9. [PMID: 27430410 PMCID: PMC4991744 DOI: 10.3892/mmr.2016.5514] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 03/22/2016] [Indexed: 12/28/2022] Open
Abstract
It is well known that the local renin-angiotensin system (RAS) is activated in the diabetic state, which results in an increase in the level of oxidative stress injury to pancreatic β cells. The angiotensin-converting enzyme 2 (ACE2)/angiotensin (1-7) [Ang (1-7)]/Mas axis is a negative regulator of the classical renin-angiotensin system. In order to investigate the antioxidant effect of Ang (1-7) on pancreatic β cells, INS-1 cells were cultured and oxidative stress was induced by treatment with H2O2. Glucose-stimulated insulin secretion (GSIS), the generation of reactive oxygen species (ROS), mitochondrial membrane potential (MMP) and glucose-stimulated calcium (GSCa) responses in β cells were determined following treatment with Ang (1-7). It was observed that H2O2 significantly impaired the insulin secreting function, increased the production of ROS, and also decreased the levels of GSCa and MMP. Pre-treatment with Ang (1-7) alleviated these effects and treatment with A779 [antagonist of Ang (1-7)] prevented the effects of Ang (1-7). Based on these findings, it was concluded that Ang (1-7) can protect pancreatic β cells from oxidative injury and such protection can be blocked by its antagonist A779.
Collapse
Affiliation(s)
- Fen Zhang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Chang Liu
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Lei Wang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Xi Cao
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Ying Ying Wang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Jin Kui Yang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| |
Collapse
|
25
|
Durand MJ, Zinkevich NS, Riedel M, Gutterman DD, Nasci VL, Salato VK, Hijjawi JB, Reuben CF, North PE, Beyer AM. Vascular Actions of Angiotensin 1-7 in the Human Microcirculation: Novel Role for Telomerase. Arterioscler Thromb Vasc Biol 2016; 36:1254-62. [PMID: 27079876 DOI: 10.1161/atvbaha.116.307518] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/28/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVE This study examined vascular actions of angiotensin 1-7 (ANG 1-7) in human atrial and adipose arterioles. APPROACH AND RESULTS The endothelium-derived hyperpolarizing factor of flow-mediated dilation (FMD) switches from antiproliferative nitric oxide (NO) to proatherosclerotic hydrogen peroxide in arterioles from humans with coronary artery disease (CAD). Given the known vasoprotective properties of ANG 1-7, we tested the hypothesis that overnight ANG 1-7 treatment restores the NO component of FMD in arterioles from patients with CAD. Endothelial telomerase activity is essential for preserving the NO component of vasodilation in the human microcirculation; thus, we also tested whether telomerase activity was necessary for ANG 1-7-mediated vasoprotection by treating separate arterioles with ANG 1-7±the telomerase inhibitor 2-[[(2E)-3-(2-naphthalenyl)-1-oxo-2-butenyl1-yl]amino]benzoic acid. ANG 1-7 dilated arterioles from patients without CAD, whereas dilation was significantly reduced in arterioles from patients with CAD. In atrial arterioles from patients with CAD incubated with ANG 1-7 overnight, the NO synthase inhibitor NG-nitro-l-arginine methyl ester abolished FMD, whereas the hydrogen peroxide scavenger polyethylene glycol catalase had no effect. Conversely, in vessels incubated with ANG 1-7+2-[[(2E)-3-(2-naphthalenyl)-1-oxo-2-butenyl1-yl]amino]benzoic acid, NG-nitro-l-arginine methyl ester had no effect on FMD, but polyethylene glycol catalase abolished dilation. In cultured human coronary artery endothelial cells, ANG 1-7 significantly increased telomerase activity. These results indicate that ANG 1-7 dilates human microvessels, and dilation is abrogated in the presence of CAD. Furthermore, ANG 1-7 treatment is sufficient to restore the NO component of FMD in arterioles from patients with CAD in a telomerase-dependent manner. CONCLUSIONS ANG 1-7 exerts vasoprotection in the human microvasculature via modulation of telomerase activity.
Collapse
Affiliation(s)
- Matthew J Durand
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Natalya S Zinkevich
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Michael Riedel
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - David D Gutterman
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Victoria L Nasci
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Valerie K Salato
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - John B Hijjawi
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Charles F Reuben
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Paula E North
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.)
| | - Andreas M Beyer
- From the Department of Physical Medicine and Rehabilitation (M.J.D.), Department of Medicine, Cardiovascular Center (M.J.D., N.S.Z., M.R., D.D.G., V.L.N., A.M.B.), Department of Pathology, Division of Pediatric Pathology (V.K.S., P.E.N.), Department of Plastic Surgery (J.B.H.), Department of Cardiothoracic Surgery (C.F.R.), and Department of Physiology (A.M.B.), Medical College of Wisconsin, Milwaukee; and Department of Health and Medicine, Carroll University, Waukesha, WI (N.S.Z.).
| |
Collapse
|
26
|
Agarwal A, Ingham SA, Harkins KA, Do DV, Nguyen QD. The role of pharmacogenetics and advances in gene therapy in the treatment of diabetic retinopathy. Pharmacogenomics 2016; 17:309-20. [PMID: 26807609 DOI: 10.2217/pgs.15.173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Diabetic retinopathy (DR) and its complications such as diabetic macular edema continue to remain a major cause for legal blindness in the developed world. While the introduction of anti-tVEGF agents has significantly improved visual outcomes of patients with DR, unpredictable response, largely due to genetic polymorphisms, appears to be a challenge with this therapy. With advances in identification of various genetic biomarkers, novel therapeutic strategies consisting of gene transfer are being developed and tested for patients with DR. Application of pharmacogenetic principles appears to be a promising futuristic strategy to attenuate diabetes-mediated retinal vasculopathy. In this comprehensive review, data from recent studies in the field of pharmacogenomics for the treatment of DR have been provided.
Collapse
Affiliation(s)
- Aniruddha Agarwal
- Ocular Imaging Research & Reading Center (OIRRC), Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA
| | - Sally A Ingham
- College of Medicine, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA
| | - Keegan A Harkins
- Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA
| | - Diana V Do
- Ocular Imaging Research & Reading Center (OIRRC), Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA.,Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA
| | - Quan Dong Nguyen
- Ocular Imaging Research & Reading Center (OIRRC), Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA.,Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA
| |
Collapse
|
27
|
Yang HY, Bian YF, Xiao CS, Liang B, Zhang N, Gao F, Yang ZM. Angiotensin-(1-7) stimulates cholesterol efflux from angiotensin II-treated cholesterol-loaded THP-1 macrophages through the suppression of p38 and c-Jun N-terminal kinase signaling. Mol Med Rep 2015; 12:1387-1392. [PMID: 25779847 DOI: 10.3892/mmr.2015.3484] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 01/23/2015] [Indexed: 11/06/2022] Open
Abstract
Angiotensin II (Ang II) and Ang-(1-7) are key effector peptides of the renin-angiotensin system. The present study aimed to investigate the effects of Ang-(1-7) on Ang II-stimulated cholesterol efflux and the associated molecular mechanisms. Differentiated THP-1 macrophages were treated with Ang II (1 µM) and/or Ang-(1-7) (10 and 100 nM) for 24 h and the cholesterol efflux and gene expression levels were assessed. Pharmacological inhibition of peroxisome proliferator-activated receptor (PPAR)γ and mitogen-activated protein kinases (MAPKs) were performed to identify the signaling pathways involved. The results demonstrated that Ang II significantly inhibited the cholesterol efflux from cholesterol-loaded THP-1 macrophages. Treatment with Ang-(1-7) led to a dose-dependent restoration of cholesterol efflux in the Ang II-treated cells. The co-treatment with Ang-(1-7) and Ang II significantly increased the expression levels of adenosine triphosphate (ATP)-binding cassette (ABC)A1 and ABCG1 compared with treatment with Ang II alone. This was coupled with increased expression levels of PPARγ and liver X receptor (LXR)α. The pharmacological inhibition of PPARγ significantly (P<0.05) eliminated the Ang-(1-7)-mediated induction of ABCA1 and ABCG1 mRNA expression. Treatment with Ang-(1-7) caused the inactivation of c-Jun N-terminal kinases (JNK) and p38 MAPK signaling in the Ang II-treated THP-1 macrophages. In addition, the inhibition of JNK or p38 MAPK signaling using specific pharmacological inhibitors mimicked the Ang-(1-7)-induced expression of PPARγ and LXRα. In conclusion, the data demonstrated that treatment with Ang-(1-7) promoted cholesterol efflux in Ang II-treated THP-1 macrophages, partly through inactivation of p38 and JNK signaling and by inducing the expression of PPARγ and LXRα. Ang (1-7) may, therefore, have therapeutic benefits for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Hui-Yu Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yun-Fei Bian
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Chuan-Shi Xiao
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Bin Liang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Nana Zhang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Fen Gao
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Zhi-Ming Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
28
|
Angiotensin-(1-7) prevents systemic hypertension, attenuates oxidative stress and tubulointerstitial fibrosis, and normalizes renal angiotensin-converting enzyme 2 and Mas receptor expression in diabetic mice. Clin Sci (Lond) 2015; 128:649-63. [PMID: 25495544 DOI: 10.1042/cs20140329] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We investigated the relationship between Ang-(1-7) [angiotensin-(1-7)] action, sHTN (systolic hypertension), oxidative stress, kidney injury, ACE2 (angiotensin-converting enzyme-2) and MasR [Ang-(1-7) receptor] expression in Type 1 diabetic Akita mice. Ang-(1-7) was administered daily [500 μg/kg of BW (body weight) per day, subcutaneously] to male Akita mice from 14 weeks of age with or without co-administration of an antagonist of the MasR, A779 (10 mg/kg of BW per day). The animals were killed at 20 weeks of age. Age-matched WT (wild-type) mice served as controls. Ang-(1-7) administration prevented sHTN and attenuated kidney injury (reduced urinary albumin/creatinine ratio, glomerular hyperfiltration, renal hypertrophy and fibrosis, and tubular apoptosis) without affecting blood glucose levels in Akita mice. Ang-(1-7) also attenuated renal oxidative stress and the expression of oxidative stress-inducible proteins (NADPH oxidase 4, nuclear factor erythroid 2-related factor 2, haem oxygenase 1), pro-hypertensive proteins (angiotensinogen, angiotensin-converting enzyme, sodium/hydrogen exchanger 3) and profibrotic proteins (transforming growth factor-β1 and collagen IV), and increased the expression of anti-hypertensive proteins (ACE2 and MasR) in Akita mouse kidneys. These effects were reversed by A779. Our data suggest that Ang-(1-7) plays a protective role in sHTN and RPTC (renal proximal tubular cell) injury in diabetes, at least in part, through decreasing renal oxidative stress-mediated signalling and normalizing ACE2 and MasR expression.
Collapse
|
29
|
Padda RS, Shi Y, Lo CS, Zhang SL, Chan JSD. Angiotensin-(1-7): A Novel Peptide to Treat Hypertension and Nephropathy in Diabetes? ACTA ACUST UNITED AC 2015; 6. [PMID: 26793405 DOI: 10.4172/2155-6156.1000615] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The renin-angiotensin system (RAS) plays a pivotal role in mammalian homeostasis physiology. The RAS can be delineated into a classical RAS (the pressor arm) including angiotensinogen (Agt), renin, angiotensin-converting enzyme (ACE), angiotensin II (Ang II) and angiotensin type 1 receptor (AT1R), and a counterbalancing novel RAS (the depressor arm) including Agt, renin, angiotensin-converting enzyme-2 (ACE-2), angiotensin-(1-7) (Ang 1-7) and Ang 1-7 receptor (or Mas receptor (MasR)). Hyperglycemia (diabetes) induces severe tissue oxidative stress, which stimulates the pressor arm of the renal RAS axis and leads to an increase in ACE/ACE-2 ratio, with excessive formation of Ang II. There is a growing body of evidence for beneficial effects of the depressor arm of RAS (ACE-2/Ang 1-7/MasR) axis in diabetes, hypertension and several other diseased conditions. Evidence from in vitro, in vivo and clinical studies reflects anti-oxidant, anti-fibrotic, and anti-inflammatory properties of Ang 1-7. Most of the currently available therapies only target suppression of the pressor arm of RAS with angiotensin receptor blockers (ARBs) and ACE inhibitors (ACEi). However, it is time to consider simultaneous activation of the depressor arm for more effective outcomes. This review summarizes the recent updates on the protective role of Ang 1-7 in hypertension and kidney injury in diabetes, as well as the possible underlying mechanism(s) of Ang 1-7 action, suggesting that the ACE-2/Ang 1-7/MasR axis can be developed as a therapeutic target for the treatment of diabetes-induced hypertension and renal damage.
Collapse
Affiliation(s)
- Ranjit Singh Padda
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 Saint Denis Street, Montreal, Quebec, Canada H2X 0A9
| | - Yixuan Shi
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 Saint Denis Street, Montreal, Quebec, Canada H2X 0A9
| | - Chao-Sheng Lo
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 Saint Denis Street, Montreal, Quebec, Canada H2X 0A9
| | - Shao-Ling Zhang
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 Saint Denis Street, Montreal, Quebec, Canada H2X 0A9
| | - John S D Chan
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 Saint Denis Street, Montreal, Quebec, Canada H2X 0A9
| |
Collapse
|
30
|
Zhang K, Meng X, Li D, Yang J, Kong J, Hao P, Guo T, Zhang M, Zhang Y, Zhang C. Angiotensin(1-7) attenuates the progression of streptozotocin-induced diabetic renal injury better than angiotensin receptor blockade. Kidney Int 2014; 87:359-69. [PMID: 25075768 PMCID: PMC4317508 DOI: 10.1038/ki.2014.274] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/16/2014] [Accepted: 06/19/2014] [Indexed: 01/10/2023]
Abstract
To explore the potential therapeutic effects of angiotensin(1–7) (Ang(1–7)), an endogenous ligand of the Mas receptor, on streptozotocin-induced diabetic nephropathy, male Wistar rats were randomly divided into two groups: a control group and a diabetic model group. After 12 weeks, the diabetic rats were divided into subgroups for 4-week treatments consisting of no-treatment group, small-, moderate-, and large-dose Ang(1–7) groups, a valsartan group, a large-dose Ang(1–7) plus valsartan group, and an A779 (antagonist of the Mas receptor) group, each with 15 rats. Ang(1–7) improved renal function, attenuated glomeruli sclerosis, oxidative stress, and cell proliferation, decreased the expression of collagen IV, TGF-β1, VEGF, NOX4, p47phox, PKCα, and PKCβ1, and the phosphorylation of Smad3. In the rat mesangial HBZY-1 cell line, Ang(1–7) decreased high-glucose-induced oxidative stress, the proliferation and expression of NOX4, p47phox, and TGF-β1, the phosphorylation of Smad3, collagen IV, and VEGF, and the membrane translocation of PKCα and PKCβ1. A779 blocked the effects of Ang(1–7) both in vivo and in vitro. The effects of large-dose Ang(1–7) alone and in combination with valsartan were superior to valsartan alone, but the combination had no significant synergistic effect compared with Ang(1–7) alone. Thus, Ang(1–7) ameliorated streptozotocin-induced diabetic renal injury. Large-dose treatment was superior to valsartan in reducing oxidative stress and inhibiting TGFβ1/Smad3- and VEGF-mediated pathways.
Collapse
Affiliation(s)
- Kai Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao Meng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Dongmei Li
- Department of Otolaryngology, Qilu Children's Hospital of Shandong University, Jinan, China
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Kong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Panpan Hao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Tao Guo
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
31
|
Rubio-Ruíz ME, Del Valle-Mondragón L, Castrejón-Tellez V, Carreón-Torres E, Díaz-Díaz E, Guarner-Lans V. Angiotensin II and 1-7 during aging in Metabolic Syndrome rats. Expression of AT1, AT2 and Mas receptors in abdominal white adipose tissue. Peptides 2014; 57:101-8. [PMID: 24819472 DOI: 10.1016/j.peptides.2014.04.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 04/29/2014] [Accepted: 04/29/2014] [Indexed: 11/18/2022]
Abstract
Renin-Angiotensin System (RAS) plays an important role in the development of Metabolic Syndrome (MS) and in aging. Angiotensin 1-7 (Ang 1-7) has opposite effects to Ang II. All of the components of RAS are expressed locally in adipose tissue and there is over-activation of adipose RAS in obesity and hypertension. We determined serum and abdominal adipose tissue Ang II and Ang 1-7 in control and MS rats during aging and the expression of AT1, AT2 and Mas in white adipose tissue. MS was induced by sucrose ingestion during 6, 12 and 18 months. During aging, an increase in body weight, abdominal fat and dyslipidemia were found but increases in aging MS rats were higher. Control and MS concentrations of serum Ang II from 6-month old rats were similar. Aging did not modify Ang II seric concentration in control rats but decreased it in MS rats. Ang II levels increased in WAT from both groups of rats. Serum and adipose tissue Ang 1-7 increased during aging in MS rats. Western blot analysis revealed that AT1 expression increased in the control group during aging while AT2 and Mas remained unchanged. In MS rats, AT1 and AT2 expression decreased significantly in aged rats. The high concentration of Ang 1-7 and adiponectin in old MS rats might be associated to an increased expression of PPAR-γ. PPAR-γ was increased in adipose tissue from MS rats. It decreased with aging in control rats and showed no changes during aging in MS rats. Ang 1-7/Mas axis was the predominant pathway in WAT from old MS animals and could represent a potential target for therapeutical strategies in the treatment of MS during aging.
Collapse
Affiliation(s)
- M E Rubio-Ruíz
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico D.F., Mexico
| | - L Del Valle-Mondragón
- Department of Pharmacology, Instituto Nacional de Cardiología ``Ignacio Chávez'' Mexico D.F., Mexico
| | - V Castrejón-Tellez
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico D.F., Mexico
| | - E Carreón-Torres
- Department of Molecular Biology, Instituto Nacional de Cardiología ``Ignacio Chávez'' Mexico D.F., Mexico
| | - E Díaz-Díaz
- Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y de la Nutrición "Salvador Zubirán", Mexico D.F., Mexico
| | - V Guarner-Lans
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico D.F., Mexico.
| |
Collapse
|
32
|
Soto ME, Soria-Castro E, Lans VG, Ontiveros EM, Mejía BIH, Hernandez HJM, García RB, Herrera V, Pérez-Torres I. Analysis of oxidative stress enzymes and structural and functional proteins on human aortic tissue from different aortopathies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:760694. [PMID: 25101153 PMCID: PMC4102031 DOI: 10.1155/2014/760694] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/28/2014] [Accepted: 05/28/2014] [Indexed: 01/15/2023]
Abstract
The role of oxidative stress in different aortopathies is evaluated. Thirty-two tissue samples from 18 men and 14 women were divided into: 4 control (C) subjects, 11 patients with systemic arterial hypertension (SAH), 4 with variants of Marfan's syndrome (MV), 9 with Marfan's syndrome (M), 2 with Turner's syndrome, and 2 with Takayasu's arteritis (TA). Aorta fragments were homogenized. Lipoperoxidation (LPO), copper-zinc and manganese superoxide dismutase (Mn and Cu-Zn-SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione S-transferase (GST), endothelial nitric oxide synthase (eNOS), nitrates and nitrites (NO3(-)/NO2(-)), and type IV collagen, and laminin were evaluated. There was an increase in Mn- and Cu-Zn-SOD activity in SAH, MV, M, and Turner's syndrome. There was also an increase in CAT activity in M and Turner' syndrome. GPx and GST activity decreased and LPO increased in all groups. eNOS was decreased in SAH, MV, and M and NO3 (-)/NO2 (-) were increased in SAH and TA. Type IV collagen was decreased in Turner's syndrome and TA. Laminin γ-1 was decreased in MV and increased in M. In conclusion, similarities and differences in oxidative stress in the different aortopathies studied including pathologies with aneurysms were found with alterations in SOD, CAT, GPx, GST, and eNOS activity that modify subendothelial basement membrane proteins.
Collapse
Affiliation(s)
- María Elena Soto
- Immunology Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Elizabeth Soria-Castro
- Pathology Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Verónica Guarner Lans
- Physiology Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Eleazar Muruato Ontiveros
- Cardiovascular Surgery Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Benjamín Iván Hernández Mejía
- Cardiovascular Surgery Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Humberto Jorge Martínez Hernandez
- Cardiovascular Surgery Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Rodolfo Barragán García
- Cardiovascular Surgery Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Valentín Herrera
- Cardiovascular Surgery Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Israel Pérez-Torres
- Pathology Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| |
Collapse
|
33
|
Abstract
The prevalence of Type 2 diabetes mellitus is predicted to increase dramatically over the coming years and the clinical implications and healthcare costs from this disease are overwhelming. In many cases, this pathological condition is linked to a cluster of metabolic disorders, such as obesity, systemic hypertension and dyslipidaemia, defined as the metabolic syndrome. Insulin resistance has been proposed as the key mediator of all of these features and contributes to the associated high cardiovascular morbidity and mortality. Although the molecular mechanisms behind insulin resistance are not completely understood, a negative cross-talk between AngII (angiotensin II) and the insulin signalling pathway has been the focus of great interest in the last decade. Indeed, substantial evidence has shown that anti-hypertensive drugs that block the RAS (renin-angiotensin system) may also act to prevent diabetes. Despite its long history, new components within the RAS continue to be discovered. Among them, Ang-(1-7) [angiotensin-(1-7)] has gained special attention as a counter-regulatory hormone opposing many of the AngII-related deleterious effects. Specifically, we and others have demonstrated that Ang-(1-7) improves the action of insulin and opposes the negative effect that AngII exerts at this level. In the present review, we provide evidence showing that insulin and Ang-(1-7) share a common intracellular signalling pathway. We also address the molecular mechanisms behind the beneficial effects of Ang-(1-7) on AngII-mediated insulin resistance. Finally, we discuss potential therapeutic approaches leading to modulation of the ACE2 (angiotensin-converting enzyme 2)/Ang-(1-7)/Mas receptor axis as a very attractive strategy in the therapy of the metabolic syndrome and diabetes-associated diseases.
Collapse
|
34
|
Silveira KD, Barroso LC, Vieira AT, Cisalpino D, Lima CX, Bader M, Arantes RME, dos Santos RAS, Simões-e-Silva AC, Teixeira MM. Beneficial effects of the activation of the angiotensin-(1-7) MAS receptor in a murine model of adriamycin-induced nephropathy. PLoS One 2013; 8:e66082. [PMID: 23762470 PMCID: PMC3676359 DOI: 10.1371/journal.pone.0066082] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 05/06/2013] [Indexed: 12/16/2022] Open
Abstract
Angiotensin-(1–7) [Ang-(1–7)] is a biologically active heptapeptide that may counterbalance the physiological actions of angiotensin II (Ang II) within the renin-angiotensin system (RAS). Here, we evaluated whether activation of the Mas receptor with the oral agonist, AVE 0991, would have renoprotective effects in a model of adriamycin (ADR)-induced nephropathy. We also evaluated whether the Mas receptor contributed for the protective effects of treatment with AT1 receptor blockers. ADR (10 mg/kg) induced significant renal injury and dysfunction that was maximal at day 14 after injection. Treatment with the Mas receptor agonist AVE 0991 improved renal function parameters, reduced urinary protein loss and attenuated histological changes. Renoprotection was associated with reduction in urinary levels of TGF-β. Similar renoprotection was observed after treatment with the AT1 receptor antagonist, Losartan. AT1 and Mas receptor mRNA levels dropped after ADR administration and treatment with losartan reestablished the expression of Mas receptor and increased the expression of ACE2. ADR-induced nephropathy was similar in wild type (Mas+/+) and Mas knockout (Mas−/−) mice, suggesting there was no endogenous role for Mas receptor activation. However, treatment with Losartan was able to reduce renal injury only in Mas+/+, but not in Mas−/− mice. Therefore, these findings suggest that exogenous activation of the Mas receptor protects from ADR-induced nephropathy and contributes to the beneficial effects of AT1 receptor blockade. Medications which target specifically the ACE2/Ang-(1–7)/Mas axis may offer new therapeutic opportunities to treat human nephropathies.
Collapse
Affiliation(s)
- Kátia Daniela Silveira
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lívia Corrêa Barroso
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Angélica Thomáz Vieira
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Cisalpino
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cristiano Xavier Lima
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Pediatria da Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Michael Bader
- Max Delbrück Center for Molecular Medicin, Berlin Buch, Germany
| | - Rosa Maria Esteves Arantes
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Ana Cristina Simões-e-Silva
- Departamento de Pediatria da Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail: (ACSS); (MMT)
| | - Mauro Martins Teixeira
- Imunofarmacologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Pediatria da Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail: (ACSS); (MMT)
| |
Collapse
|
35
|
Kumar BNP, Rajput S, Dey KK, Parekh A, Das S, Mazumdar A, Mandal M. Celecoxib alleviates tamoxifen-instigated angiogenic effects by ROS-dependent VEGF/VEGFR2 autocrine signaling. BMC Cancer 2013; 13:273. [PMID: 23731702 PMCID: PMC3681557 DOI: 10.1186/1471-2407-13-273] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/31/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Tamoxifen (TAM) is widely used in the chemotherapy of breast cancer and as a preventive agent against recurrence after surgery. However, extended TAM administration for breast cancer induces increased VEGF levels in patients, promoting new blood vessel formation and thereby limiting its efficacy. Celecoxib (CXB), a selective COX-2 inhibitor, suppresses VEGF gene expression by targeting the VEGF promoter responsible for its inhibitory effect. For this study, we had selected CXB as non-steroidal anti-inflammatory drug in combination with TAM for suppressing VEGF expression and simultaneously reducing doses of both the drugs. METHODS The effects of CXB combined with TAM were examined in two human breast cancer cell lines in culture, MCF7 and MDA-MB-231. Assays of proliferation, apoptosis, angiogenesis, metastasis, cell cycle distribution, and receptor signaling were performed. RESULTS Here, we elucidated how the combination of TAM and CXB at nontoxic doses exerts anti-angiogenic effects by specifically targeting VEGF/VEGFR2 autocrine signaling through ROS generation. At the molecular level, TAM-CXB suppresses VHL-mediated HIF-1α activation, responsible for expression of COX-2, MMP-2 and VEGF. Besides low VEGF levels, TAM-CXB also suppresses VEGFR2 expression, confirmed through quantifying secreted VEGF levels, luciferase and RT-PCR studies. Interestingly, we observed that TAM-CXB was effective in blocking VEGFR2 promoter induced expression and further 2 fold decrease in VEGF levels was observed in combination than TAM alone in both cell lines. Secondly, TAM-CXB regulated VEGFR2 inhibits Src expression, responsible for tumor progression and metastasis. FACS and in vivo enzymatic studies showed significant increase in the reactive oxygen species upon TAM-CXB treatment. CONCLUSIONS Taken together, our experimental results indicate that this additive combination shows promising outcome in anti-metastatic and apoptotic studies. In a line, our preclinical studies evidenced that this additive combination of TAM and CXB is a potential drug candidate for treatment of breast tumors expressing high levels of VEGF and VEGFR2. This ingenious combination might be a better tailored clinical regimen than TAM alone for breast cancer treatment.
Collapse
Affiliation(s)
- B N Prashanth Kumar
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur-721302, West Bengal PIN-721302, India
| | | | | | | | | | | | | |
Collapse
|
36
|
Jiang T, Gao L, Zhu XC, Yu JT, Shi JQ, Tan MS, Lu J, Tan L, Zhang YD. Angiotensin-(1-7) inhibits autophagy in the brain of spontaneously hypertensive rats. Pharmacol Res 2013; 71:61-8. [PMID: 23499735 DOI: 10.1016/j.phrs.2013.03.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 03/05/2013] [Accepted: 03/06/2013] [Indexed: 12/19/2022]
Abstract
Autophagy is an important cellular process that mediates lysosomal degradation of damaged organelles, which is activated in response to a variety of stress-related diseases, including hypertension. The basal level of autophagy plays an important role in the maintenance of cellular homeostasis, whereas excessive autophagic activity leads to cell death and is considered as a contributing factor to several disorders. Recent works have demonstrated that Angiotensin-(1-7) [Ang-(1-7)] exerted its neuroprotective effects by modulating classic components of renin-angiotensin system associated with reducing oxidative stress and apoptosis in brains of spontaneously hypertensive rats (SHRs). However, the effect of Ang-(1-7) on autophagic activity in brain of hypertensive individual remains unclear. In this study, Wistar-Kyoto rats received intracerebroventricular (I.C.V.) infusion of artificial cerebrospinal fluid (aCSF) while SHRs received I.C.V. infusion of aCSF, Ang-(1-7), Mas receptor antagonist A-779, or angiotensin II type 2 receptor antagonist PD123319 for 4 weeks. Brain tissues were collected and analyzed by western blotting analysis, immunofluorescence assay, and transmission electron microscopic examination. Our study showed that infusion of Ang-(1-7) for 4 weeks inhibited the increase of microtubule-associated protein 1 light chain 3 (LC3)-II and Beclin-1 levels, as well as the autophagosome formation in SHR brain. Meanwhile, the reduction of p62 expression in SHR brain was also reversed by Ang-(1-7). Of note, the anti-autophagic effects of Ang-(1-7) were independent of blood pressure reduction and can be inhibited by A-779 and PD123319. These findings suggest that treatment with Ang-(1-7) may be useful to prevent hypertension-induced excessive autophagic activation in brain.
Collapse
Affiliation(s)
- Teng Jiang
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
The brain Renin-Angiotensin system and mitochondrial function: influence on blood pressure and baroreflex in transgenic rat strains. Int J Hypertens 2013; 2013:136028. [PMID: 23401750 PMCID: PMC3564433 DOI: 10.1155/2013/136028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 12/23/2012] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial dysfunction is implicated in many cardiovascular diseases, including hypertension, and may be associated with an overactive renin-angiotensin system (RAS). Angiotensin (Ang) II, a potent vasoconstrictor hormone of the RAS, also impairs baroreflex and mitochondrial function. Most deleterious cardiovascular actions of Ang II are thought to be mediated by NADPH-oxidase- (NOX-) derived reactive oxygen species (ROS) that may also stimulate mitochondrial oxidant release and alter redox-sensitive signaling pathways in the brain. Within the RAS, the actions of Ang II are counterbalanced by Ang-(1–7), a vasodilatory peptide known to mitigate against increased oxidant stress. A balance between Ang II and Ang-(1–7) within the brain dorsal medulla contributes to maintenance of normal blood pressure and proper functioning of the arterial baroreceptor reflex for control of heart rate. We propose that Ang-(1–7) may negatively regulate the redox signaling pathways activated by Ang II to maintain normal blood pressure, baroreflex, and mitochondrial function through attenuating ROS (NOX-generated and/or mitochondrial).
Collapse
|
38
|
Chrysin and Luteolin Attenuate Diabetes-Induced Impairment in Endothelial-Dependent Relaxation: Effect on Lipid Profile, AGEs and NO Generation. Phytother Res 2013; 27:1678-84. [DOI: 10.1002/ptr.4917] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 11/27/2012] [Accepted: 11/28/2012] [Indexed: 12/11/2022]
|
39
|
Abstract
The renin-angiotensin system (RAS) constitutes one of the most important hormonal systems in the physiological regulation of blood pressure through renal and nonrenal mechanisms. Indeed, dysregulation of the RAS is considered a major factor in the development of cardiovascular pathologies, including kidney injury, and blockade of this system by the inhibition of angiotensin converting enzyme (ACE) or blockade of the angiotensin type 1 receptor (AT1R) by selective antagonists constitutes an effective therapeutic regimen. It is now apparent with the identification of multiple components of the RAS within the kidney and other tissues that the system is actually composed of different angiotensin peptides with diverse biological actions mediated by distinct receptor subtypes. The classic RAS can be defined as the ACE-Ang II-AT1R axis that promotes vasoconstriction, water intake, sodium retention, and other mechanisms to maintain blood pressure, as well as increase oxidative stress, fibrosis, cellular growth, and inflammation in pathological conditions. In contrast, the nonclassical RAS composed primarily of the AngII/Ang III-AT2R pathway and the ACE2-Ang-(1-7)-AT7R axis generally opposes the actions of a stimulated Ang II-AT1R axis through an increase in nitric oxide and prostaglandins and mediates vasodilation, natriuresis, diuresis, and reduced oxidative stress. Moreover, increasing evidence suggests that these non-classical RAS components contribute to the therapeutic blockade of the classical system to reduce blood pressure and attenuate various indices of renal injury, as well as contribute to normal renal function.
Collapse
Affiliation(s)
- Mark C Chappell
- The Hypertension & Vascular Disease Center, Department of Physiology & Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
40
|
Akhtar S, Yousif MHM, Dhaunsi GS, Chandrasekhar B, Al-Farsi O, Benter IF. Angiotensin-(1-7) inhibits epidermal growth factor receptor transactivation via a Mas receptor-dependent pathway. Br J Pharmacol 2012; 165:1390-400. [PMID: 21806601 DOI: 10.1111/j.1476-5381.2011.01613.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE The transactivation of the epidermal growth factor (EGF) receptor appears to be an important central transduction mechanism in mediating diabetes-induced vascular dysfunction. Angiotensin-(1-7) [Ang-(1-7)] via its Mas receptor can prevent the development of hyperglycaemia-induced cardiovascular complications. Here, we investigated whether Ang-(1-7) can inhibit hyperglycaemia-induced EGF receptor transactivation and its classical signalling via ERK1/2 and p38 MAPK in vivo and in vitro. EXPERIMENTAL APPROACH Streptozotocin-induced diabetic rats were chronically treated with Ang-(1-7) or AG1478, a selective EGF receptor inhibitor, for 4 weeks and mechanistic studies performed in the isolated mesenteric vasculature bed as well as in primary cultures of vascular smooth muscle cells (VSMCs). KEY RESULTS Diabetes significantly enhanced phosphorylation of EGF receptor at tyrosine residues Y992, Y1068, Y1086, Y1148, as well as ERK1/2 and p38 MAPK in the mesenteric vasculature bed whereas these changes were significantly attenuated upon Ang-(1-7) or AG1478 treatment. In VSMCs grown in conditions of high glucose (25 mM), an Src-dependent elevation in EGF receptor phosphorylation was observed. Ang-(1-7) inhibited both Ang II- and glucose-induced transactivation of EGF receptor. The inhibition of high glucose-mediated Src-dependant transactivation of EGF receptor by Ang-(1-7) could be prevented by a selective Mas receptor antagonist, D-Pro7-Ang-(1-7). CONCLUSIONS AND IMPLICATIONS These results show for the first time that Ang-(1-7) inhibits EGF receptor transactivation via a Mas receptor/Src-dependent pathway and might represent a novel general mechanism by which Ang-(1-7) exerts its beneficial effects in many disease states including diabetes-induced vascular dysfunction.
Collapse
Affiliation(s)
- Saghir Akhtar
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Safat, Kuwait.
| | | | | | | | | | | |
Collapse
|
41
|
Sharma S, Sun X, Rafikov R, Kumar S, Hou Y, Oishi PE, Datar SA, Raff G, Fineman JR, Black SM. PPAR-γ regulates carnitine homeostasis and mitochondrial function in a lamb model of increased pulmonary blood flow. PLoS One 2012; 7:e41555. [PMID: 22962578 PMCID: PMC3433474 DOI: 10.1371/journal.pone.0041555] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 06/27/2012] [Indexed: 12/11/2022] Open
Abstract
Objective Carnitine homeostasis is disrupted in lambs with endothelial dysfunction secondary to increased pulmonary blood flow (Shunt). Our recent studies have also indicated that the disruption in carnitine homeostasis correlates with a decrease in PPAR-γ expression in Shunt lambs. Thus, this study was carried out to determine if there is a causal link between loss of PPAR-γ signaling and carnitine dysfunction, and whether the PPAR-γ agonist, rosiglitazone preserves carnitine homeostasis in Shunt lambs. Methods and Results siRNA-mediated PPAR-γ knockdown significantly reduced carnitine palmitoyltransferases 1 and 2 (CPT1 and 2) and carnitine acetyltransferase (CrAT) protein levels. This decrease in carnitine regulatory proteins resulted in a disruption in carnitine homeostasis and induced mitochondrial dysfunction, as determined by a reduction in cellular ATP levels. In turn, the decrease in cellular ATP attenuated NO signaling through a reduction in eNOS/Hsp90 interactions and enhanced eNOS uncoupling. In vivo, rosiglitazone treatment preserved carnitine homeostasis and attenuated the development of mitochondrial dysfunction in Shunt lambs maintaining ATP levels. This in turn preserved eNOS/Hsp90 interactions and NO signaling. Conclusion Our study indicates that PPAR-γ signaling plays an important role in maintaining mitochondrial function through the regulation of carnitine homeostasis both in vitro and in vivo. Further, it identifies a new mechanism by which PPAR-γ regulates NO signaling through Hsp90. Thus, PPAR-γ agonists may have therapeutic potential in preventing the endothelial dysfunction in children with increased pulmonary blood flow.
Collapse
Affiliation(s)
- Shruti Sharma
- Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Ang-(1-7) [angiotensin-(1-7)] is a biologically active heptapeptide component of the RAS (renin-angiotensin system), and is generated in the kidney at relatively high levels, via enzymatic pathways that include ACE2 (angiotensin-converting enzyme 2). The biological effects of Ang-(1-7) in the kidney are primarily mediated by interaction with the G-protein-coupled receptor Mas. However, other complex effects have been described that may involve receptor-receptor interactions with AT(1) (angiotensin II type 1) or AT(2) (angiotensin II type 2) receptors, as well as nuclear receptor binding. In the renal vasculature, Ang-(1-7) has vasodilatory properties and it opposes growth-stimulatory signalling in tubular epithelial cells. In several kidney diseases, including hypertensive and diabetic nephropathy, glomerulonephritis, tubulointerstitial fibrosis, pre-eclampsia and acute kidney injury, a growing body of evidence supports a role for endogenous or exogenous Ang-(1-7) as an antagonist of signalling mediated by AT(1) receptors and thereby as a protector against nephron injury. In certain experimental conditions, Ang-(1-7) appears to paradoxically exacerbate renal injury, suggesting that dose or route of administration, state of activation of the local RAS, cell-specific signalling or non-Mas receptor-mediated pathways may contribute to the deleterious responses. Although Ang-(1-7) has promise as a potential therapeutic agent in humans with kidney disease, further studies are required to delineate its signalling mechanisms in the kidney under physiological and pathophysiological conditions.
Collapse
|
43
|
Harrison-Bernard LM, Chappell MC. Unraveling the glomerular RAS: one peptidase at a time. Am J Physiol Renal Physiol 2012; 303:F373-4. [PMID: 22573374 PMCID: PMC3433875 DOI: 10.1152/ajprenal.00234.2012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
44
|
El-Bassossy HM, Abo-Warda SM, Fahmy A. Rosiglitazone, a peroxisome proliferator-activated receptor γ stimulant, abrogates diabetes-evoked hypertension by rectifying abnormalities in vascular reactivity. Clin Exp Pharmacol Physiol 2012; 39:643-9. [DOI: 10.1111/j.1440-1681.2012.05724.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | - Shaymaa M Abo-Warda
- Department of Pharmacology; Faculty of Pharmacy; Zagazig University; Zagazig; Egypt
| | - Ahmed Fahmy
- Department of Pharmacology; Faculty of Pharmacy; Zagazig University; Zagazig; Egypt
| |
Collapse
|
45
|
Characterization of Angiotensin-(1-7) effects on the cardiovascular system in an experimental model of type-1 diabetes. Pharmacol Res 2012; 66:269-75. [PMID: 22580236 DOI: 10.1016/j.phrs.2012.05.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 04/05/2012] [Accepted: 05/01/2012] [Indexed: 11/24/2022]
Abstract
Although exogenous administration of Angiotensin-(1-7) [Ang-(1-7)] can prevent development of diabetes induced end-organ damage, little is known about the role of endogenous Ang-(1-7) in diabetes and requires further characterization. Here, we studied the effects of chronically inhibiting endogenous Ang-(1-7) formation with DX600, a selective angiotensin converting enzyme-2 (ACE2) inhibitor, on renal and cardiac NADPH oxidase (NOX) activity, vascular reactivity and cardiac function in a model of Type-1 diabetes. The contribution of endogenous Ang-(1-7) to the protective effects of Losartan and Captopril and that of prostaglandins to the cardiovascular effects of exogenous Ang-(1-7) were also examined. Cardiac and renal NOX activity, vascular reactivity to endothelin-1 (ET-1) and cardiac recovery from ischemia/reperfusion (I/R) injury were evaluated in streptozotocin-treated rats. Chronic treatment with DX600 exacerbated diabetes-induced increase in cardiac and renal NOX activity. Diabetes-induced abnormal vascular reactivity to ET-1 and cardiac dysfunction were improved by treatment with Ang-(1-7) and worsened by treatment with DX600 or A779, a Mas receptor antagonist. Ang-(1-7)-mediated improvement in cardiac recovery or vascular reactivity was attenuated by Indomethacin. Captopril and Losartan-induced improvement in cardiovascular function was attenuated when these drugs were co-administered with A779. Ang-(1-7)-mediated decrease in renal NOX activity was prevented by indomethacin. Losartan also decreased renal NOX activity that could be attenuated with A779 co-treatment. In conclusion, endogenous Ang-(1-7) inhibits diabetes-induced cardiac/renal NOX activity and end-organ damage, and mediates the actions of Captopril and Losartan. Further, prostaglandins are important intermediaries in the beneficial effects of Ang-(1-7) in diabetes. Combining either Losartan or Captopril with Ang-(1-7) had additional beneficial effects in preventing diabetes-induced cardiac dysfunction and this may represent a novel therapeutic strategy. Collectively, these data shed new insights into the likely mechanism of action through which the ACE2/Ang-(1-7)/Mas receptor axis prevents Type 1 diabetes-induced cardiovascular dysfunction.
Collapse
|
46
|
Singh K, Sharma K, Singh M, Sharma PL. Possible mechanism of the cardio-renal protective effects of AVE-0991, a non-peptide Mas-receptor agonist, in diabetic rats. J Renin Angiotensin Aldosterone Syst 2012; 13:334-40. [PMID: 22345112 DOI: 10.1177/1470320311435534] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
HYPOTHESIS This study was designed to investigate the cardio-renal protective effect of AVE-0991, a non-peptide Mas-receptor agonist, and A-779, a Mas-receptor antagonist, in diabetic rats. MATERIALS AND METHODS Wistar rats treated with streptozotocin (50 mg/kg, i.p., once), developed diabetes mellitus after 1 week. After 8 weeks, myocardial functions were assessed by measuring left ventricular developed pressure (LVDP), rate of left ventricular pressure development (dp/dt (max)), rate of left ventricular pressure decay (dp/dt (min)) and left ventricular end diastolic pressure (LVEDP) on an isolated Langendorff's heart preparation. Further, mean arterial blood pressure (MABP) was measured by using the tail-cuff method. Assessment of renal functions and lipid profile was carried out using a spectrophotometer. RESULTS The administration of streptozotocin to rats produced persistent hyperglycaemia, dyslipidaemia and hypertension which consequently produced cardiac and renal dysfunction in 8 weeks. AVE0991 treatment produced cardio-renal protective effects, as evidenced by a significant increase in LVDP, dp/dt (max), dp/dt (min) and a significant decrease in LVEDP, BUN, and protein urea. Further, AVE-0991 treatment for the first time has been shown to reduce dyslipidaemia and produced antihyperglycaemic activity in streptozotocin-treated rats. However, MABP and creatinine clearance remained unaffected with AVE-0991 treatment. CONCLUSIONS AVE-0991 produced cardio-renal protection possibly by improving glucose and lipid metabolism in diabetic rats, independent of its blood pressure lowering action.
Collapse
|
47
|
Mario EG, Santos SHS, Ferreira AVM, Bader M, Santos RAS, Botion LM. Angiotensin-(1-7) Mas-receptor deficiency decreases peroxisome proliferator-activated receptor gamma expression in adipocytes. Peptides 2012; 33:174-7. [PMID: 22119778 DOI: 10.1016/j.peptides.2011.11.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 11/14/2011] [Accepted: 11/14/2011] [Indexed: 01/01/2023]
Abstract
The renin-angiotensin system is an important link between metabolic syndrome and cardiovascular diseases. Besides angiotensin II, other angiotensin peptides such as angiotensin-(1-7), have important biological activities. It has been demonstrated that angiotensin-(1-7), acting through the G protein-coupled receptor encoded by the Mas protooncogene have important actions on the cardiovascular system. However, the role of angiotensin-(1-7)-Mas axis in lipidic profile is not well established. In the present study, the adipocyte metabolism was investigated in wild type and FVB/N Mas-deficient male mice. The gene expression of peroxisome proliferator-activated receptor gamma, acetyl-CoA carboxylase and the amount of fatty acid synthase protein were reduced in the Mas-knockout mice. Serum nonesterified fatty acids of Mas-knockout showed a 50% increase in relation to wild type group. Basal and isoproterenol-stimulated lipolysis was similar between the groups, however, a significant decrease of the glycerol release (lipolytic index) in response to insulin was observed in wild type animals, while no effect of the insulin action was observed in a Mas-knockout group. The data suggest that the lack of angiotensin-(1-7) action through Mas receptor alters the response of adipocytes to insulin action. These effects might be related to decreased expression of PPARγ.
Collapse
Affiliation(s)
- Erica Guilhen Mario
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | | | | |
Collapse
|
48
|
Raffai G, Durand MJ, Lombard JH. Acute and chronic angiotensin-(1-7) restores vasodilation and reduces oxidative stress in mesenteric arteries of salt-fed rats. Am J Physiol Heart Circ Physiol 2011; 301:H1341-52. [PMID: 21803946 DOI: 10.1152/ajpheart.00202.2011] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study determined the effect of ANG-(1-7) on salt-induced suppression of endothelium-dependent vasodilatation in the mesenteric arteries of male Sprague-Dawley rats. Chronic intravenous infusion of ANG-(1-7), oral administration of the nonpeptide mas receptor agonist AVE-0991, and acute preincubation of the arteries with ANG-(1-7) and AVE-0991 all restored vasodilator responses to both ACh and histamine that were absent in the arteries of rats fed a high-salt (4% NaCl) diet. The protective effects of ANG-(1-7) and AVE-0991 were inhibited by acute or chronic administration of the mas receptor antagonist A-779, the ANG II type 2 (AT(2)) receptor blocker PD-123319, or N-nitro-l-arginine methyl ester, but not the ANG II type 1 receptor antagonist losartan. Preincubation with the antioxidant tempol or the nitric oxide (NO) donor diethylenetriamine NONOate and acute and chronic administration of the AT(2) receptor agonist CGP-42112 mimicked the protective effect of ANG-(1-7) to restore vascular relaxation. Acute preincubation with ANG-(1-7) and chronic infusion of ANG-(1-7) ameliorated the elevated superoxide levels in rats fed a high-salt diet, but the expression of Cu/Zn SOD and Mn SOD enzyme proteins in the vessel wall was unaffected by ANG-(1-7) infusion. These results indicate that both acute and chronic systemic administration of ANG-(1-7) or AVE-0991 restore endothelium-dependent vascular relaxation in salt-fed Sprague-Dawley rats by reducing vascular oxidant stress and enhancing NO availability via mas and AT(2) receptors. These findings suggest a therapeutic potential for mas/AT(2) receptor activation in preventing the vascular oxidant stress and endothelial dysfunction associated with elevated dietary salt intake.
Collapse
Affiliation(s)
- Gábor Raffai
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | |
Collapse
|
49
|
Wong YC, Sim MK, Lee KO. Des-aspartate-angiotensin-I and angiotensin IV improve glucose tolerance and insulin signalling in diet-induced hyperglycaemic mice. Biochem Pharmacol 2011; 82:1198-208. [PMID: 21803028 DOI: 10.1016/j.bcp.2011.07.080] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 07/13/2011] [Accepted: 07/13/2011] [Indexed: 11/29/2022]
Abstract
Although clinical studies suggested that blockade of the renin-angiotensin system may prevent diabetes, the mechanism is uncertain. As a follow-up to an earlier study, we investigated how des-aspartate-angiotensin-1 (DAA-1) and its metabolite, angiotensin IV (Ang-IV) improved glucose tolerance in diet-induced hyperglycaemic mice. Male C57BL/6J mice were fed a high-fat-high-sucrose (HFD) or normal (ND) diet for 52 weeks. HFD animals were orally administered either DAA-I (600nmol/kg/day), Ang-IV (400nmol/kg/day) or distilled water. Body weight, blood glucose and insulin were measured fortnightly. Inflammatory and insulin signalling transducers that are implicated in hyperglycaemia were analyzed in skeletal muscles at 52 weeks. HFD animals developed hyperglycemia, hyperinsulinemia and obesity. DAA-I and Ang-IV improved glucose tolerance but had no effect on hyperinsulinemia and obesity. Skeletal muscles of HFD animals showed increased level of ROS, gp91 of NADPH oxidase, pJNK and AT(1)R-JAK-2-IRS-1 complex. Both DAA-I and Ang-IV attenuated these increases. Insulin-induced activation of IR, IRS-1, IRS-1-PI3K coupling, phosphorylation of Akt, and GLUT4 translocation were attenuated in skeletal muscles of HFD animals. The attenuation was significantly ameliorated in DAA-I-treated HFD animals. In corresponding Ang-IV treated animals, insulin induced IRAP and PI3K interaction, activation of pAkt and GLUT4 translocation, but no corresponding activation of IR, IRS-1 and IRS-1-PI3K coupling were observed. DAA-I and Ang-IV improved glucose tolerance, insulin signalling, and para-inflammatory processes linked to hyperglycaemia. DAA-I acts via the angiotensin AT(1) receptor and activates the insulin pathway. Ang-IV acts via IRAP, which couples PI3K and activates the later part of the insulin pathway.
Collapse
Affiliation(s)
- Yong-Chiat Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Previous concepts regarding the pathways involved in the generation of angiotensin II (Ang II) have been challenged by studies showing the existence of a peptide acting as an endogenous antagonist of Ang II. The discovery that angiotensin-(1-7) [Ang-(1-7)] opposes the pressor, proliferative, profibrotic, and prothrombotic actions mediated by Ang II has contributed to the realization that the renin-angiotensin system is composed of two opposing arms: the pressor arm constituted by the enzyme angiotensin-converting enzyme (ACE), Ang II as the product, and the Ang II type 1 (AT1) receptor as the main protein mediating the biological actions of Ang II; the second arm is composed of the monocarboxypeptidase angiotensin-converting enzyme 2 (ACE2), Ang-(1-7) produced through hydrolysis of Ang II, and the Mas receptor as the protein conveying the vasodilator, antiproliferative, antifibrotic, and antithrombotic effects of Ang-(1-7). RECENT FINDINGS Experimental and clinical studies demonstrate a role for the Ang-(1-7)/ACE2/Mas axis in the evolution of hypertension, the regulation of renal function, and the progression of renal disease including diabetic nephropathy. Additional evidence suggests that a reduction in the expression and activity of this vasodepressor component may be a critical factor in mediating the progression of cardiovascular disease. SUMMARY Further research on the contribution of the Ang-(1-7)/ACE2/Mas axis to cardiovascular pathology will lead to the development of new pharmacological approaches resulting in the design of molecular or genetic means to increase the expression of ACE2, allow for increased tissue levels of Ang-(1-7), or both.
Collapse
|