51
|
Vascular Damage and Kidney Transplant Outcomes: An Unfriendly and Harmful Link. Am J Med Sci 2017; 354:7-16. [DOI: 10.1016/j.amjms.2017.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/20/2016] [Accepted: 01/09/2017] [Indexed: 12/31/2022]
|
52
|
Nehme A, Zibara K. Cellular distribution and interaction between extended renin-angiotensin-aldosterone system pathways in atheroma. Atherosclerosis 2017; 263:334-342. [PMID: 28600074 DOI: 10.1016/j.atherosclerosis.2017.05.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 04/14/2017] [Accepted: 05/24/2017] [Indexed: 01/06/2023]
Abstract
The importance of the renin-angiotensin-aldosterone system (RAAS) in the development of atherosclerotic has been experimentally documented. In fact, RAAS components have been shown to be locally expressed in the arterial wall and to be differentially regulated during atherosclerotic lesion progression. RAAS transcripts and proteins were shown to be differentially expressed and to interact in the 3 main cells of atheroma: endothelial cells, vascular smooth muscle cells, and macrophages. This review describes the local expression and cellular distribution of extended RAAS components in the arterial wall and their differential regulation during atherosclerotic lesion development.
Collapse
Affiliation(s)
- Ali Nehme
- EA4173, Functional Genomics of Arterial Hypertension, Hôpital Nord-Ouest, Villefranche-sur-Saône, Université Lyon1, Lyon, France; ER045, Laboratory of Stem Cells, Department of Biology, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- ER045, Laboratory of Stem Cells, Department of Biology, Faculty of Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
53
|
Lipotoxicity-Related Hematological Disorders in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:469-487. [DOI: 10.1007/978-3-319-48382-5_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
54
|
Ruddy JM, Akerman AW, Kimbrough D, Nadeau EK, Stroud RE, Mukherjee R, Ikonomidis JS, Jones JA. Differential hypertensive protease expression in the thoracic versus abdominal aorta. J Vasc Surg 2016; 66:1543-1552. [PMID: 28034583 DOI: 10.1016/j.jvs.2016.07.120] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 07/24/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Hypertension (HTN), which is a major risk factor for cardiovascular morbidity and mortality, can drive pathologic remodeling of the macro- and microcirculation. Patterns of aortic pathology differ, however, suggesting regional heterogeneity of the pressure-sensitive protease systems triggering extracellular matrix remodeling in the thoracic (TA) and abdominal aortas (AA). This study tested the hypothesis that the expression of two major protease systems (matrix metalloproteinases [MMPs] and cathepsins) in the TA and AA would be differentially affected with HTN. METHODS Normotensive (BPN3) mice at 14-16 weeks of age underwent implantation of osmotic infusion pumps for 28-day angiotensin II (AngII) delivery (1.46 mg/kg/day; BPN3+AngII; n = 8) to induce HTN. The TA and AA were harvested to determine levels of MMP-2, MMP-9, and membrane type 1-MMP, and cathepsins S, K, and L were evaluated in age-matched BPN3 (n = 8) control and BPH2 spontaneously hypertensive mice (non-AngII pathway; n = 7). Blood pressure was monitored via CODA tail cuff plethysmography (Kent Scientific Corporation, Torrington, Conn). Quantitative real-time polymerase chain reaction and immunoblotting/zymography were used to measure MMP and cathepsin messenger RNA expression and protein abundance, respectively. Target protease values were compared within each aortic region via analysis of variance. RESULTS Following 28 days infusion, the BPN3+AngII mice had a 17% increase in systolic blood pressure, matching that of the BPH2 spontaneously hypertensive mice (both P < .05 vs BPN3). MMP-2 gene expression demonstrated an AngII-dependent increase in the TA (P < .05), but MMP-9 was not altered with HTN. Expression of tissue inhibitor of metalloproteinases-1 was markedly increased in TA of BPN3+AngII mice, but tissue inhibitor of metalloproteinases-2 demonstrated decreased expression in the AA of both hypertensive groups (P < .05). Only cathepsin K responded to AngII-induced HTN with significant elevation in the TA of those mice, but expression of cathepsin L and cystatin C was inhibited in AA of both hypertensive groups (P < .05). Apoptotic markers were not significantly elevated in any experimental group. CONCLUSIONS By using two different models of HTN, this study has identified pressure-dependent as well as AngII-dependent regional alterations in aortic gene expression of MMPs and cathepsins that may lead to differential remodeling responses in each of the aortic regions. Further studies will delineate mechanisms and may provide targeted therapies to attenuate down-stream aortic pathology based on demonstrated regional heterogeneity.
Collapse
Affiliation(s)
- Jean Marie Ruddy
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC; Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC.
| | - Adam W Akerman
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| | - Denise Kimbrough
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| | - Elizabeth K Nadeau
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| | - Robert E Stroud
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| | - Rupak Mukherjee
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| | - John S Ikonomidis
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC; Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC
| | - Jeffrey A Jones
- Division of Cardiothoracic Research, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
55
|
Kendrick J, Zelnick L, Chonchol M, Siscovick D, Hoofnagle AN, Ix JH, Sarnak M, Shlipak M, Kestenbaum B, de Boer IH. Serum Bicarbonate Is Associated with Heart Failure in the Multi-Ethnic Study of Atherosclerosis. Am J Nephrol 2016; 45:118-126. [PMID: 27941322 PMCID: PMC5296282 DOI: 10.1159/000454783] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/05/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Low serum bicarbonate concentrations are associated with mortality and kidney disease progression. Data regarding associations between bicarbonate and cardiovascular disease (CVD) are scarce. METHODS We performed a cohort study of 6,229 adult participants from the Multi-Ethnic Study of Atherosclerosis, a community-based cohort free of CVD at baseline. Serum bicarbonate was measured at baseline. Cardiovascular outcomes were defined as: (1) subclinical CVD (left ventricular mass [LVM] and aortic pulse pressure [PP] measured at baseline), (2) incident atherosclerotic cardiovascular events (CVE; composite of myocardial infarction, resuscitated cardiac arrest, stroke, coronary heart disease death, and stroke death), and (3) incident heart failure. RESULTS During a median (interquartile range) follow-up of 8.5 (7.7-8.6) years, 331 (5.3%) participants had an incident CVE and 174 (2.8%) developed incident heart failure. We stratified analyses by use of diuretics because we observed a significant interaction between diuretic use and bicarbonate with study outcomes. Among diuretic nonusers, with adjustment, bicarbonate ≥25 mEq/L was associated with an estimated 3.0 g greater LVM (95% CI 0.5-5.0) and 1.0 mm Hg higher aortic PP (95% CI 0.4-2.0) compared to bicarbonate 23-24 mEq/L. Each 1 mEq/L of bicarbonate increase was associated with a 13% higher risk of incident heart failure (hazards ratio 1.13, 95% CI 1.01-2.11). Among diuretic users, higher bicarbonate was not associated with CVD. Bicarbonate was not associated with incident atherosclerotic CVE irrespective of diuretic use. CONCLUSION Among nonusers of diuretics in a large community-based study, higher serum bicarbonate concentrations are associated with subclinical CVD and new heart failure.
Collapse
Affiliation(s)
- Jessica Kendrick
- Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, CO
- Denver Health Medical Center, Denver, CO
| | - Leila Zelnick
- Division of Nephrology, Kidney Research Institute, University of Washington, Seattle WA
| | - Michel Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, CO
| | | | | | - Joachim H. Ix
- Division of Nephrology and Hypertension, Department of Medicine, University of California, San Diego, CA
| | - Mark Sarnak
- Division of Nephrology, Tufts Medical Center, Boston, MA
| | - Michael Shlipak
- Division of General Internal Medicine, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA
| | - Bryan Kestenbaum
- Division of Nephrology, Kidney Research Institute, University of Washington, Seattle WA
| | - Ian H. de Boer
- Division of Nephrology, Kidney Research Institute, University of Washington, Seattle WA
| |
Collapse
|
56
|
Tunçdemir M, Öztürk M. Regulation of the Ku70 and apoptosis-related proteins in experimental diabetic nephropathy. Metabolism 2016; 65:1466-77. [PMID: 27621182 DOI: 10.1016/j.metabol.2016.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Apoptosis contributes nephropathy pathogenesis in diabetes. However, its mechanisms still remain unclear. We examined the extent to which the angiotensin-II type 1 receptor blocker (AT1RB) irbesartan and the angiotensin converting enzyme inhibitor (ACEI) perindopril affected the apoptosis-related proteins Bcl-2, Bax, caspase-3, cytochrome-c and Ku70 in streptozotocin (STZ)-diabetic rats. MATERIALS AND METHODS Animals were divided into five groups of eight each, four of which received STZ (60mg/kg in a single dose, i.p.) to induce diabetes. The groups were performed as untreated diabetic; non-diabetic control; daily irbesartan (15mg/kg/day) or perindopril (6mg/kg/day) and also combined irbesartan and perindopril (respectively, 5mg/kg/day, 3mg/kg/day) were applied by gavage for 30days to STZ-diabetic rats. The kidney tissue analysis was performed by using immunohistochemical staining with Bcl-2, Bax, caspase-3, cytochrome-c and Ku70 antibodies and by using Western blot analysis with caspase-3 and cytochrome-c antibodies. RESULTS Immunoreactivity of Bax, caspase-3, cytochrome-c and Ku70 was increased in the tubuli and glomeruli of the untreated diabetic group, but decreased in all treated diabetic groups. In the irbesartan and perindopril treated diabetic groups Bcl-2 immunoreactivity was higher than that of the untreated diabetic group. Caspase-3 and cytoplasmic cytochrome-c protein levels increased in the untreated diabetic group. CONCLUSIONS We conclude that the increased expression of Bax and caspase-3, and the increased level of cytoplasmic cytochrome-c relate to renal tissue injury. This case is also seen in the early stages of diabetes as a result of the damage caused by local increased expression of renin angiotensin system (RAS) in the renal tissue, which is induced by hyperglycemia. The increase of the cytosolic cytochrome-c, caspase-3 and Ku70 expression in the tubuli is suggestive of apoptosis. Overall, our results show that treatments of irbesartan and perindopril are effective and efficient in preventing renal tissue injury and apoptosis by blocking the RAS in experimental diabetic nephropathy and reducing the expression of proteins associated with apoptosis.
Collapse
Affiliation(s)
- Matem Tunçdemir
- Istanbul University, Cerrahpaşa Faculty of Medicine, Department of Medical Biology, Istanbul, Turkey.
| | - Melek Öztürk
- Istanbul University, Cerrahpaşa Faculty of Medicine, Department of Medical Biology, Istanbul, Turkey.
| |
Collapse
|
57
|
Li RQ, Wu Y, Zhi Y, Yang X, Li Y, Xua FJ, Du J. PGMA-Based Star-Like Polycations with Plentiful Hydroxyl Groups Act as Highly Efficient miRNA Delivery Nanovectors for Effective Applications in Heart Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:7204-7212. [PMID: 27297033 DOI: 10.1002/adma.201602319] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 05/17/2016] [Indexed: 06/06/2023]
Abstract
Poly(glycidyl methacrylate)-based star-like polycations with rich hydrophilic hydroxyl groups can efficiently transfer miRNA into primary cardiac fibroblasts for effective applications in cardiac diseases, such as inhibition of cardiac fibrosis and hypertrophy.
Collapse
Affiliation(s)
- Rui-Quan Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Yina Wu
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), Beijing Institute of Heart, Lung, Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, P. R. China
| | - Ying Zhi
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), Beijing Institute of Heart, Lung, Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, P. R. China
| | - Xinchao Yang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Yulin Li
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), Beijing Institute of Heart, Lung, Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, P. R. China
| | - Fu-Jian Xua
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Jie Du
- Key Laboratory of Remodeling-Related Cardiovascular Diseases (Ministry of Education), Beijing Institute of Heart, Lung, Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, 100029, P. R. China
| |
Collapse
|
58
|
Li S, Wang Q, Tao Y, Liu C. Swertiamarin Attenuates Experimental Rat Hepatic Fibrosis by Suppressing Angiotensin II–Angiotensin Type 1 Receptor–Extracellular Signal-Regulated Kinase Signaling. J Pharmacol Exp Ther 2016; 359:247-255. [DOI: 10.1124/jpet.116.234179] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 08/17/2016] [Indexed: 12/14/2022] Open
|
59
|
Huang H, Luo Y, Liang Y, Long X, Peng Y, Liu Z, Wen X, Jia M, Tian R, Bai C, Li C, He F, Lin Q, Wang X, Dong X. CD4(+)CD25(+) T Cells in primary malignant hypertension related kidney injury. Sci Rep 2016; 6:27659. [PMID: 27278520 PMCID: PMC4899787 DOI: 10.1038/srep27659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/18/2016] [Indexed: 01/10/2023] Open
Abstract
CD4+CD25+ T cells are critical for maintenance of immunologic self-tolerance. We measured the number of CD4+CD25+ cells in the patients with primary malignant hypertension related kidney injury, to explore the molecular pathogenesis of this disease. We selected 30 patients with primary malignant hypertension related kidney injury and 30 healthy volunteers. Information on clinical characteristics and laboratory tests was obtained from each subject. The number of CD4+CD25+ cells and glomerular injury were assessed by flow cytometry and histopathology, respectively. Both serum IL-2, IL-4, and IL-6 and endothelial cell markers were analyzed by ELISA. ADAMTS13 antibody was detected by Western blotting. CD4+CD25+ cells were significantly reduced in patients with primary malignant hypertension related kidney injury compared to controls (P < 0.05). The number of CD4+CD25+ cells was negatively related to blood urea nitrogen, serum uric acid, proteinuria, and supernatant IL-4; whereas positively associated with estimated glomerular filtration rate in patients. Gradually decreasing CD4+CD25+ cells were also found as increasing renal injury. Additionally, patients exhibited increasing supernatant IL-4, serum IL-2 and IL-6, endothelial cell markers, and anti-ADAMTS13 antibody compared with controls (all P < 0.05). CD4+CD25+ cells may play a key role in the pathogenesis of primary malignant hypertension related kidney injury.
Collapse
Affiliation(s)
- Hongdong Huang
- Division of Nephrology, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, P.R. China.,Division of Nephrology, Hunan Normal University, Hunan Provincial People's Hospital of China, Changsha, Hunan Province, P.R. China
| | - Yang Luo
- Division of Nephrology, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, P.R. China
| | - Yumei Liang
- Division of Nephrology, Hunan Normal University, Hunan Provincial People's Hospital of China, Changsha, Hunan Province, P.R. China
| | - Xidai Long
- Department of Liver Surgery, the Affiliated Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Youming Peng
- Hunan Key Laboratory of Nephrology and Hemoperfusion, Division of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan Province, P.R. China
| | - Zhihua Liu
- Division of Nephrology, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, P.R. China
| | - Xiaojun Wen
- Division of Nephrology, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, P.R. China
| | - Meng Jia
- Division of Nephrology, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, P.R. China
| | - Ru Tian
- Division of Nephrology, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, P.R. China
| | - Chengli Bai
- Division of Nephrology, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, P.R. China
| | - Cui Li
- Division of Nephrology, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, P.R. China
| | - Fuliang He
- Department of Interventional Therapy, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, P.R. China.,Section of Hematology/Oncology, Section of Gastroenterology, Stephenson Cancer Center, Department of Internal Medicine, College of Medicine, The University of Oklahoma Health Sciences Center, USA
| | - Qiushi Lin
- Section of Hematology/Oncology, Section of Gastroenterology, Stephenson Cancer Center, Department of Internal Medicine, College of Medicine, The University of Oklahoma Health Sciences Center, USA
| | - Xueyan Wang
- Center of Allergy, Beijing Shijitan Hospital, Capital Medical University, Beijing, P.R. China
| | - Xiaoqun Dong
- Department of Interventional Therapy, Beijing Shijitan Hospital, Capital Medical University, The 9th Affiliated Hospital of Peking University, Beijing, P.R. China.,Section of Hematology/Oncology, Section of Gastroenterology, Stephenson Cancer Center, Department of Internal Medicine, College of Medicine, The University of Oklahoma Health Sciences Center, USA
| |
Collapse
|
60
|
Peng K, Lu X, Wang F, Nau A, Chen R, Zhou SF, Yang T. Collecting duct (pro)renin receptor targets ENaC to mediate angiotensin II-induced hypertension. Am J Physiol Renal Physiol 2016; 312:F245-F253. [PMID: 27122543 DOI: 10.1152/ajprenal.00178.2016] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/21/2016] [Indexed: 01/09/2023] Open
Abstract
The (pro)renin receptor (PRR) is abundantly expressed in the collecting duct (CD) and the expression is further induced by angiotensin II (ANG II). The present study was conducted to investigate the role of CD PRR during ANG II-induced hypertension and to further explore the underlying mechanism. Radiotelemetry demonstrated that a 1-wk ANG II infusion gradually and significantly induced hypertensive response in floxed mice and this response was significantly attenuated in mice lacking PRR in the CD (termed CD PRR KO). ANG II infusion in floxed mice increased urinary renin activity and selectively induced renal medullary α-epithelial sodium channel (α-ENaC) mRNA and protein expression, all of which were blunted in the null mice. In cultured mpkCCD cells grown in Transwells, transepithelial Na+ transport as measured by using a volt-ohmmeter was transiently stimulated by acute ANG II treatment, which was abolished by a PRR antagonist, PRO20. In a chronic setting, ANG II treatment induced α-ENaC mRNA expression in mpkCCD cells, which was similarly blocked by PRO20. Chronic intramedullary infusion of an ENaC inhibitor amiloride in rats significantly attenuated ANG II-induced hypertension. Overall, the present study suggests that CD PRR contributes to ANG II-induced hypertension at least partially via activation of renal medullary ENaC.
Collapse
Affiliation(s)
- Kexin Peng
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China.,Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Xiaohan Lu
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China.,Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Fei Wang
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China.,Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Adam Nau
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China
| | - Ren Chen
- Department of Internal Medicine, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, China; and
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida
| | - Tianxin Yang
- Institute of Hypertension, Sun Yat-Sen University School of Medicine, Guangzhou, China; .,Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| |
Collapse
|
61
|
“Angiotensin II memory” contributes to the development of hypertension and vascular injury via activation of NADPH oxidase. Life Sci 2016; 149:18-24. [DOI: 10.1016/j.lfs.2016.02.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/09/2016] [Indexed: 02/07/2023]
|
62
|
Oyagbemi AA, Omobowale TO, Adedapo AA, Yakubu MA. Kolaviron, Biflavonoid Complex from the Seed of Garcinia kola Attenuated Angiotensin II- and Lypopolysaccharide-induced Vascular Smooth Muscle Cell Proliferation and Nitric Oxide Production. Pharmacognosy Res 2016; 8:S50-5. [PMID: 27114693 PMCID: PMC4821108 DOI: 10.4103/0974-8490.178647] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION Kolaviron (KV), a biflavonoid extract from Garcinia kola seeds has been reported to possess anti-inflammatory, anti-oxidant, hepato-protective, cardio-protective, nephro-protective and other arrays of chemopreventive capabilities but the mechanism of action is still not completely understood. MATERIALS AND METHODS In this study, we investigated the anti-proliferative, anti-inflammatory and anti-oxidative potential of KV in cultured Vascular Smooth Muscle Cells (VSMCs). Effects of KV (25-100 μg/mL) on VSMC proliferation alone or following treatments with mitogen and proinflammatory agents Angiotensin II (Ag II, 10(-6) M) and lipopolysaccharide (LPS, 100 μg/mL) and effects on NO production were determined. Cellular proliferations were determined by MTT assay, nitric oxide (NO) level was determined by Griess assay. KV dose-and time dependently attenuated VSMC growth. RESULTS Treatment of VSMCs with Ag II and LPS significantly enhanced proliferation of the cell which was significantly attenuated by the treatment with KV. Treatment of VSMC with LPS significantly increased nitric oxide (NO) level in the media which was attenuated by KV. These results demonstrated anti-proliferative anti-inflammatory properties of KV as it clearly inhibited cellular proliferation induced by mitogens as well as LPS-induced inflammatory processes. CONCLUSION Therefore, KV may mitigate cardiovascular conditions that involve cell proliferation, free radical generation and inflammatory processes such as hypertension, diabetes and stroke. However, the molecular mechanism of action of KV needs to be investigated. SUMMARY Angiotensin-induced cell proliferationKolaviron mitigates angiotensin-induced cell proliferationKolaviron ameliorates nitric oxide productionKolaviron offers antioxidant activity. Abbreviations Used: VSMCs: Vascular Smooth Muscle Cells, Ag II: Angiotensin II, KV: Kolaviron, LPS: lypopolysaccharide, NO: Nitric Oxide, DMEM: Dulbecco's modified Eagle's medium, MTT: (3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide), DMSO: Dimethylsulfoxide, GB1: Garcinia kola biflavonoid-1, GB2: Garcinia kola biflavonoid-2, ROS: Reactive oxygen species, ET-1: Endothelin-1, NF-κB: Nuclear factor-kappa beta, COX-2: Cyclooxygenase-2.
Collapse
Affiliation(s)
- Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temidayo Olutayo Omobowale
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeolu Alex Adedapo
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Momoh Audu Yakubu
- Department of Environmental and Interdisciplinary Sciences, College of Science, Engineering, and Technology, Texas Southern University, Houston, TX 77004, USA
- Vascular Biology Unit, Center for Cardiovascular Diseases, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX 77004, USA
| |
Collapse
|
63
|
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: 1.1] [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
|
64
|
Horton RE, Yadid M, McCain ML, Sheehy SP, Pasqualini FS, Park SJ, Cho A, Campbell P, Parker KK. Angiotensin II Induced Cardiac Dysfunction on a Chip. PLoS One 2016; 11:e0146415. [PMID: 26808388 PMCID: PMC4725954 DOI: 10.1371/journal.pone.0146415] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/16/2015] [Indexed: 11/29/2022] Open
Abstract
In vitro disease models offer the ability to study specific systemic features in isolation to better understand underlying mechanisms that lead to dysfunction. Here, we present a cardiac dysfunction model using angiotensin II (ANG II) to elicit pathological responses in a heart-on-a-chip platform that recapitulates native laminar cardiac tissue structure. Our platform, composed of arrays of muscular thin films (MTF), allows for functional comparisons of healthy and diseased tissues by tracking film deflections resulting from contracting tissues. To test our model, we measured gene expression profiles, morphological remodeling, calcium transients, and contractile stress generation in response to ANG II exposure and compared against previous experimental and clinical results. We found that ANG II induced pathological gene expression profiles including over-expression of natriuretic peptide B, Rho GTPase 1, and T-type calcium channels. ANG II exposure also increased proarrhythmic early after depolarization events and significantly reduced peak systolic stresses. Although ANG II has been shown to induce structural remodeling, we control tissue architecture via microcontact printing, and show pathological genetic profiles and functional impairment precede significant morphological changes. We assert that our in vitro model is a useful tool for evaluating tissue health and can serve as a platform for studying disease mechanisms and identifying novel therapeutics.
Collapse
Affiliation(s)
- Renita E. Horton
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
- Department of Agriculture and Biological Engineering, James Worth Bagley College of Engineering, College of Agriculture and Life Sciences, Mississippi State University, Starkville, Mississippi, United States of America
| | - Moran Yadid
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Megan L. McCain
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Sean P. Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Francesco S. Pasqualini
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Sung-Jin Park
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Alexander Cho
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Patrick Campbell
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
65
|
Ko EJ, Kim BH, Jeong HY, Soe SU, Yang DH, Lee SY. Serum 25-hydroxyvitamin D as a predictor of hospitalization-free survival in predialysis and dialysis patients with chronic kidney disease: a single-center prospective observational analysis. Kidney Res Clin Pract 2016; 35:22-8. [PMID: 27069854 PMCID: PMC4811977 DOI: 10.1016/j.krcp.2015.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 12/11/2015] [Accepted: 12/18/2015] [Indexed: 12/16/2022] Open
Abstract
Background Vitamin D has pleiotropic effects important for the proper functioning of multiple organ systems. We investigated whether serum 25-hydroxyvitamin D [25(OH)D] levels influenced hospitalization-free survival in patients with chronic kidney disease (CKD). Methods In this prospective study, serum levels of 25(OH)D were measured in 210 patients with CKD in the winter of 2009. Data regarding hospitalizations were collected over the subsequent 3 years. Results Vitamin D deficiency, as defined by a serum 25(OH)D level below 15 ng/mL, was observed in 76.7% of the patients. The mean 25(OH)D serum level was 13.6 ± 7.8 ng/mL in predialysis patients (n = 62) and 11.3 ± 6.7 ng/mL in dialysis patients (n = 148). During the follow-up, 107 patients (28 predialysis and 79 dialysis) were hospitalized because of infectious (33.6%) or cardiovascular diseases (23.4%). Predialysis and dialysis groups were divided into 2 subgroups based on the median 25(OH)D serum level. Kaplan–Meier analysis revealed that the risk of hospitalization was significantly lower in both predialysis and dialysis patients with above-median serum 25(OH)D levels (log-rank test; P = 0.043 and 0.002, respectively). Multivariate Cox proportional hazards models also demonstrated that the risk of hospitalization was significantly lower for patients with higher serum 25(OH)D levels in both the predialysis (hazard ratio, 0.963; 95% confidence interval, 0.93–0.99) and dialysis groups (hazard ratio, 0.955; 95% confidence interval, 0.91–0.99). Conclusion A lower serum 25(OH)D level predicted poorer hospitalization-free survival in both predialysis and dialysis CKD patients.
Collapse
Affiliation(s)
- Eun-Jung Ko
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Bo Hye Kim
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Hye Yun Jeong
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Sung Un Soe
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Dong Ho Yang
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - So-Young Lee
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| |
Collapse
|
66
|
Castellon X, Bogdanova V. Chronic Inflammatory Diseases and Endothelial Dysfunction. Aging Dis 2016; 7:81-9. [PMID: 26815098 DOI: 10.14336/ad.2015.0803] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 08/03/2015] [Indexed: 01/10/2023] Open
Abstract
Chronic inflammatory diseases are associated with increases in cardiovascular diseases (CVD) and subclinical atherosclerosis as well as early-stage endothelial dysfunction screening using the FMD method (Flow Mediated Dilation). This phenomenon, referred to as accelerated pathological remodeling of arterial wall, could be attributed to traditional risk factors associated with atherosclerosis. Several new non-invasive techniques have been used to study arterial wall's structural and functional alterations. These techniques (based of Radio Frequency, RF) allow for an assessment of artery age through calculations of intima-media thickness (RF- QIMT), pulse wave rate (RF- QAS) and endothelial dysfunction degree (FMD). The inflammatory and autoimmune diseases should now be considered as new cardiovascular risk factors, result of the major consequences of oxidative stress and RAS (Renin Angiotensin System) imbalance associated with the deleterious effect of known risk factors that lead to the alteration of the arterial wall. Inflammation plays a key role in all stages of the formation of vascular lesions maintained and exacerbated by the risk factors. The consequence of chronic inflammation is endothelial dysfunction that sets in and we can define it as an integrated marker of the damage to arterial walls by classic risk factors. The atherosclerosis, which develops among these patients, is the main cause for cardiovascular morbi-mortality and uncontrolled chronic biological inflammation, which quickly favors endothelial dysfunction. These inflammatory and autoimmune diseases should now be considered as new cardiovascular risk factors.
Collapse
Affiliation(s)
| | - Vera Bogdanova
- Privat Hospital, Athis Mons, 91200 Athis-Mons, Paris, France
| |
Collapse
|
67
|
Steely AM, Callas PW, Bertges DJ. Renin-angiotensin-aldosterone-system inhibition is safe in the preoperative period surrounding carotid endarterectomy. J Vasc Surg 2015; 63:715-21. [PMID: 26603543 DOI: 10.1016/j.jvs.2015.09.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/28/2015] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Discontinuation of angiotensin-converting enzyme inhibitor (ACEI) and angiotensin receptor blocker (ARB) medications before surgery has been suggested because of the potentially deleterious effects of hypotension. We investigated the effect of preoperative ACEI and/or ARB use on early outcomes after carotid endarterectomy (CEA). METHODS We examined 3752 consecutive CEA patients within the Vascular Study Group of New England from September 2012 to September 2014 and compared outcomes for patients treated (n = 1772) or not treated (n = 1980) with ACEI and/or ARB preoperatively. Outcomes included perioperative need for intravenous vasoactive medication (IVBPmed) for hypotension or hypertension (HTN), major adverse cardiac events (MACEs), and the combined outcome of stroke or death. Adjusted analysis was performed using multivariable logistic regression of the crude cohort and by constructing a propensity score matched cohort (n = 1441). RESULTS ACEI and/or ARB users were more likely to be male (64% vs 59%; P = .001), with a higher prevalence of diabetes (41% vs 28%; P < .0001), HTN (97% vs 82%; P < .0001), coronary artery disease (31% vs 25%; P = .0001), congestive heart failure (10% vs 8%; P = .02), and asymptomatic carotid disease (59% vs 54%; P = .004). Patients who received ACEI and/or ARB preoperatively were more likely to be treated with aspirin (92% vs 88%; P = .0002) and statins (89% vs 85%; P = .001) preoperatively. In the unadjusted analysis, no significant differences were identified in hypotension that required IVBPmed (12% vs 11%; odds ratio [OR], 1.1; 95% confidence interval [CI], 0.9-1.4; P = .22), MACE (3% vs 2%; OR, 1.3; 95% CI, 0.8-1.9; P = .32), or stroke or death (3% vs 3%; OR, 1.0; 95% CI, 0.7-1.6; P = .89) for preoperative ACEI and/or ARB treated and nontreated patients, respectively. Preoperative ACEI and/or ARB usage was, however, associated with HTN that required IVBPmed (13% vs 10%; OR, 1.3; 95% CI, 1.1-1.6; P = .01). Analysis of the propensity score matched cohort revealed no significant differences in hypotension that required IVBPmed (12% vs 12%; OR, 1.0; 95% CI, 0.8-1.3; P = .86), MACE (3% vs 2%; OR, 1.1; 95% CI, 0.7-1.8; P = .62; ), or stroke or death (3% vs 3%; OR, 1.0; 95% CI, 0.7-1.6; P = .91) for patients treated or not treated with preoperative ACEI and/or ARB, respectively. ACEI and/or ARB remained associated with HTN that required IVBPmed (13% vs 10%; OR, 1.3; 95% CI, 1.0-1.7; P = .02). Results were similar after adjustment using logistic regression. The incidence of hospital length of stay >1 day was similar between ACEI and/or ARB treated and not treated patients (29% vs 32%; OR, 0.9; 95% CI, 0.8-1.1; P = .21). CONCLUSIONS Preoperative ACEI and/or ARB use was associated with marginally increased use of IVBPmed for HTN but not for hypotension and was not associated with increased MACE, stroke, or death. On the basis of these metrics, the use of preoperative ACEI and/or ARB appears safe before CEA.
Collapse
Affiliation(s)
- Andrea M Steely
- Division of Vascular Surgery, The University of Vermont Medical Center, Burlington, Vt
| | - Peter W Callas
- Division of Vascular Surgery, The University of Vermont Medical Center, Burlington, Vt
| | - Daniel J Bertges
- Division of Vascular Surgery, The University of Vermont Medical Center, Burlington, Vt.
| |
Collapse
|
68
|
Rasekh M, Young C, Roldo M, Lancien F, Le Mével JC, Hafizi S, Ahmad Z, Barbu E, Gorecki D. Hollow-layered nanoparticles for therapeutic delivery of peptide prepared using electrospraying. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:256. [PMID: 26449446 DOI: 10.1007/s10856-015-5588-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 09/25/2015] [Indexed: 06/05/2023]
Abstract
The viability of single and coaxial electrospray techniques to encapsulate model peptide-angiotensin II into near mono-dispersed spherical, nanocarriers comprising N-octyl-O-sulphate chitosan and tristearin, respectively, was explored. The stability of peptide under controlled electric fields (during particle generation) was evaluated. Resulting nanocarriers were analysed using dynamic light scattering and electron microscopy. Cell toxicity assays were used to determine optimal peptide loading concentration (~1 mg/ml). A trout model was used to assess particle behaviour in vivo. A processing limit of 20 kV was determined. A range of electrosprayed nanoparticles were formed (between 100 and 300 nm) and these demonstrated encapsulation efficiencies of ~92 ± 1.8%. For the single needle process, particles were in matrix form and for the coaxial format particles demonstrated a clear core-shell encapsulation of peptide. The outcomes of in vitro experiments demonstrated triphasic activity. This included an initial slow activity period, followed by a rapid and finally a conventional diffusive phase. This was in contrast to results from in vivo cardiovascular activity in the trout model. The results are indicative of the substantial potential for single/coaxial electrospray techniques. The results also clearly indicate the need to investigate both in vitro and in vivo models for emerging drug delivery systems.
Collapse
Affiliation(s)
- Manoochehr Rasekh
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St. Michael's Building, White Swan Road, Portsmouth, PO1 2DT, UK.
| | - Christopher Young
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St. Michael's Building, White Swan Road, Portsmouth, PO1 2DT, UK
| | - Marta Roldo
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St. Michael's Building, White Swan Road, Portsmouth, PO1 2DT, UK
| | - Frédéric Lancien
- Neurophysiology Laboratory, LaTIM UMR 1101, University of Brest, 29238, Brest Cedex 3, France
| | - Jean-Claude Le Mével
- Neurophysiology Laboratory, LaTIM UMR 1101, University of Brest, 29238, Brest Cedex 3, France
| | - Sassan Hafizi
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St. Michael's Building, White Swan Road, Portsmouth, PO1 2DT, UK
| | - Zeeshan Ahmad
- Leicester School of Pharmacy, De Montfort University, Leicester, LE1 9BH, UK
| | - Eugen Barbu
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St. Michael's Building, White Swan Road, Portsmouth, PO1 2DT, UK
| | - Darek Gorecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St. Michael's Building, White Swan Road, Portsmouth, PO1 2DT, UK
| |
Collapse
|
69
|
Karnik SS, Unal H, Kemp JR, Tirupula KC, Eguchi S, Vanderheyden PML, Thomas WG. International Union of Basic and Clinical Pharmacology. XCIX. Angiotensin Receptors: Interpreters of Pathophysiological Angiotensinergic Stimuli [corrected]. Pharmacol Rev 2015; 67:754-819. [PMID: 26315714 PMCID: PMC4630565 DOI: 10.1124/pr.114.010454] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The renin angiotensin system (RAS) produced hormone peptides regulate many vital body functions. Dysfunctional signaling by receptors for RAS peptides leads to pathologic states. Nearly half of humanity today would likely benefit from modern drugs targeting these receptors. The receptors for RAS peptides consist of three G-protein-coupled receptors—the angiotensin II type 1 receptor (AT1 receptor), the angiotensin II type 2 receptor (AT2 receptor), the MAS receptor—and a type II trans-membrane zinc protein—the candidate angiotensin IV receptor (AngIV binding site). The prorenin receptor is a relatively new contender for consideration, but is not included here because the role of prorenin receptor as an independent endocrine mediator is presently unclear. The full spectrum of biologic characteristics of these receptors is still evolving, but there is evidence establishing unique roles of each receptor in cardiovascular, hemodynamic, neurologic, renal, and endothelial functions, as well as in cell proliferation, survival, matrix-cell interaction, and inflammation. Therapeutic agents targeted to these receptors are either in active use in clinical intervention of major common diseases or under evaluation for repurposing in many other disorders. Broad-spectrum influence these receptors produce in complex pathophysiological context in our body highlights their role as precise interpreters of distinctive angiotensinergic peptide cues. This review article summarizes findings published in the last 15 years on the structure, pharmacology, signaling, physiology, and disease states related to angiotensin receptors. We also discuss the challenges the pharmacologist presently faces in formally accepting newer members as established angiotensin receptors and emphasize necessary future developments.
Collapse
Affiliation(s)
- Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Hamiyet Unal
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Jacqueline R Kemp
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Satoru Eguchi
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Patrick M L Vanderheyden
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Walter G Thomas
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| |
Collapse
|
70
|
Qiu Y, Tao L, Lei C, Wang J, Yang P, Li Q, Lei B. Downregulating p22phox ameliorates inflammatory response in Angiotensin II-induced oxidative stress by regulating MAPK and NF-κB pathways in ARPE-19 cells. Sci Rep 2015; 5:14362. [PMID: 26415877 PMCID: PMC4586461 DOI: 10.1038/srep14362] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/24/2015] [Indexed: 11/24/2022] Open
Abstract
Oxidative stress and inflammation are two interrelated biological events implicated in the pathogenesis of many diseases. Reactive oxygen species (ROS) produced under oxidative stress play a key role in pathological conditions. Inhibition of p22phox, an indispensable component of the NADPH oxidase (NOX) complex comprising the main source of ROS, plays a protective role in many ocular conditions by inhibiting the activation of NOXs and the generation of ROS. However, little is understood regarding the role of p22phox in oxidative stress-related inflammation in the eye. We used a p22phox small interfering RNA (siRNA) to transfect the retinal pigment epithelium (RPE)-derived cell line ARPE-19, and human primary RPE (hRPE) cells, then stimulated with Ang II. We observed a potent anti-inflammatory effect and studied the underlying mechanism. Downregulating p22phox resulted in decreased ROS generation, a reduction of NOXs (NOX1, 2, 4) and a decrease in inflammatory cytokine. In addition, p22phox downregulation reduced the activation of the MAPK and NF-κB signaling pathways. We conclude that inhibition of p22phox has an anti-inflammatory effect in Ang II-induced oxidative stress. Suppressing the MAPK and NF-κB pathways is involved in this protective effect. These results suggest that p22phox may provide a promising therapeutic target for oxidative stress-induced ocular inflammation
Collapse
Affiliation(s)
- Yiguo Qiu
- Department of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Lifei Tao
- Department of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Chunyan Lei
- Department of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Jiaming Wang
- Department of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Peizeng Yang
- Department of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Qiuhong Li
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Bo Lei
- Department of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| |
Collapse
|
71
|
Carver KA, Smith TL, Gallagher PE, Tallant EA. Angiotensin-(1-7) prevents angiotensin II-induced fibrosis in cremaster microvessels. Microcirculation 2015; 22:19-27. [PMID: 25079175 DOI: 10.1111/micc.12159] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/29/2014] [Indexed: 01/19/2023]
Abstract
OBJECTIVE The effect of the heptapeptide hormone Ang-(1-7) on microvascular fibrosis in rats with Ang II-induced hypertension was investigated, since vascular fibrosis/remodeling plays a prominent role in hypertension-induced end-organ damage and Ang-(1-7) inhibits vascular growth and fibrosis. METHODS Fibrosis of cremaster microvessels was studied in male Lewis rats infused with Ang II and/or Ang-(1-7). RESULTS Ang II elevated systolic blood pressure by approximately 40 mmHg, while blood pressure was not changed by Ang-(1-7). Ang II increased perivascular fibrosis surrounding 20-50 μm arterioles as well as interstitial fibrosis; coadministration of Ang-(1-7) prevented the increases in fibrosis. The fibrotic factor CTGF and phospho-Smad 2/3, which upregulates CTGF, were increased by Ang II; this effect was prevented by coadministration of Ang-(1-7). Although TGF-β phosphorylates Smad 2/3, TGF-β was no different among treatment groups. In contrast, Ang II increased the MAP kinase phospho-ERK1/2, which also phosphorylates Smad; p-ERK was reduced by Ang-(1-7). Ang-(1-7), in the presence or absence of Ang II, upregulated the MAP kinase phosphatase DUSP1. CONCLUSIONS These results suggest that Ang-(1-7) increases DUSP1 to reduce MAP kinase/Smad/CTGF signaling and decrease fibrosis in resistance arterioles, to attenuate end-organ damage associated with chronic hypertension.
Collapse
Affiliation(s)
- Kyle A Carver
- Hypertension and Vascular Research Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | | | | |
Collapse
|
72
|
Liu LH, Fan X, Xia ZK, An XX, Yang RY. Angiotensin II stimulates melanogenesis via the protein kinase C pathway. Exp Ther Med 2015; 10:1528-1532. [PMID: 26622519 DOI: 10.3892/etm.2015.2682] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 06/16/2015] [Indexed: 11/06/2022] Open
Abstract
Melanogenesis is a physiological process that results in the synthesis of melanin pigments, which serve a crucial function in hyperpigmentation. The aim of the present study was to determine the effects of angiotensin II (Ang II) on melanogenesis and to elucidate the molecular events of Ang II-induced melanogenesis. Experiments were performed on human melanocytes to elucidate the pigmenting effect of Ang II and the underlying mechanisms. The elements involved in melanogenesis, including melanin content, tyrosinase (TYR) activity, and microphthalmia-associated transcription factor (MITF) and TYR expression at the mRNA and protein levels were evaluated. Melanin content and TYR activity increased in response to Ang II treatment in a concentration-dependent manner. MITF and TYR mRNA and protein expression levels were increased significantly in response to Ang II in a concentration-dependent manner. The Ang II-induced increase in melanin synthesis was reduced significantly in response to co-treatment with Ro-32-0432, a protein kinase C (PKC) inhibitor, whereas co-treatment with H-89, a PKA inhibitor, did not attenuate the Ang II-induced increase in melanin levels. These results suggest that PKC is required for Ang II-induced pigmentation in human melanocytes and that the mechanism involves the PKC pathway and MITF upregulation.
Collapse
Affiliation(s)
- Li-Hong Liu
- Graduate School, The Third Military Medical University, Chongqing 400038, P.R. China ; Department of Dermatology, General Hospital of Beijing Military Region of PLA, Beijing 100125, P.R. China
| | - Xin Fan
- Department of Dermatology, General Hospital of Beijing Military Region of PLA, Beijing 100125, P.R. China
| | - Zhi-Kuan Xia
- Department of Dermatology, General Hospital of Beijing Military Region of PLA, Beijing 100125, P.R. China
| | - Xu-Xi An
- Department of Dermatology, General Hospital of Beijing Military Region of PLA, Beijing 100125, P.R. China
| | - Rong-Ya Yang
- Department of Dermatology, General Hospital of Beijing Military Region of PLA, Beijing 100125, P.R. China
| |
Collapse
|
73
|
Increase in Vascular Injury of Sodium Overloaded Mice May be Related to Vascular Angiotensin Modulation. PLoS One 2015; 10:e0128141. [PMID: 26030299 PMCID: PMC4451144 DOI: 10.1371/journal.pone.0128141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 04/22/2015] [Indexed: 01/07/2023] Open
Abstract
This study aimed to analyzing the effect of chronic sodium overload upon carotid and femoral injury, and its relation to vascular angiotensin modulation. Male C57Bl6 mice were divided in: control (cont), receiving 1% NaCl solution for 2 weeks (salt-2) or 12 weeks (salt-12). Two-weeks before the end of the study, a 2mm catheter was implanted around the left femoral and carotid arteries to induce injury. Blood pressure (BP) and heart rate (HR) were measured at the end of the study by tail plethysmography. Arteries were collected and prepared for histological analysis to determine arterial thickening and perivascular collagen deposition. Angiotensin II and Ang(1-7) were quantified in fresh arteries using the HPLC method. There were no differences in body weight, BP and HR. Intima/media ratio had a similar increase in both injured arteries of cont and salt-2 mice, but a more pronounced increase was observed in salt-12 mice (31.1±6%). On the other hand, sodium overload modified perivascular collagen deposition, increasing thick fibers (cont: 0.5%; salt-2: 3.4%; salt-12: 0.6%) and decreasing thin fibers (cont: 7.4%; salt-2: 0.5%; salt-12: 6.8%) in non-injured arteries. Injured arteries presented similar collagen fiber distribution. Angiotensin quantification showed increased Ang(1-7) in salt treated mice (salt-2: +72%; salt-12: +45%) with a concomitant decrease in Ang II (salt-2: -54%; salt-12: -60%). Vascular injury increased significantly Ang(1-7) in salt-12 mice (+80%), maintaining Ang II reduction similar to that of a non-injured artery. The lack of changes in BP and HR suggests that the structural changes observed may be due to non-hemodynamic mechanisms such as local renin-angiotensin system. Collagen evaluation suggests that sodium overload induces time-related changes in vascular remodeling. The increase of artery injury with concomitant increase in Ang(1-7) in 12-week treated mice shows a direct association between the duration of salt treatment and the magnitude of vascular injury.
Collapse
|
74
|
Luo R, Zhang W, Zhao C, Zhang Y, Wu H, Jin J, Zhang W, Grenz A, Eltzschig HK, Tao L, Kellems RE, Xia Y. Elevated Endothelial Hypoxia-Inducible Factor-1α Contributes to Glomerular Injury and Promotes Hypertensive Chronic Kidney Disease. Hypertension 2015; 66:75-84. [PMID: 25987665 DOI: 10.1161/hypertensionaha.115.05578] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/20/2015] [Indexed: 12/28/2022]
Abstract
Hypertensive chronic kidney disease is one of the most prevalent medical conditions with high morbidity and mortality in the United States and worldwide. However, early events initiating the progression to hypertensive chronic kidney disease are poorly understood. We hypothesized that elevated endothelial hypoxia-inducible factor-1α (HIF-1α) is a common early insult triggering initial glomerular injury leading to hypertensive chronic kidney disease. To test our hypothesis, we used an angiotensin II infusion model of hypertensive chronic kidney disease to determine the specific cell type and mechanisms responsible for elevation of HIF-1α and its role in the progression of hypertensive chronic kidney disease. Genetic studies coupled with reverse transcription polymerase chain reaction profiling revealed that elevated endothelial HIF-1α is essential to initiate glomerular injury and progression to renal fibrosis by the transcriptional activation of genes encoding multiple vasoactive proteins. Mechanistically, we found that endothelial HIF-1α gene expression was induced by angiotensin II in a nuclear factor-κB-dependent manner. Finally, we discovered reciprocal positive transcriptional regulation of endothelial Hif-1α and Nf-κb genes is a key driving force for their persistent activation and disease progression. Overall, our findings revealed that the stimulation of HIF-1α gene expression in endothelial cells is detrimental to induce kidney injury, hypertension, and disease progression. Our findings highlight early diagnostic opportunities and therapeutic approaches for hypertensive chronic kidney disease.
Collapse
Affiliation(s)
- Renna Luo
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Weiru Zhang
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Cheng Zhao
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Yujin Zhang
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Hongyu Wu
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Jianping Jin
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Wenzheng Zhang
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Almut Grenz
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Holger K Eltzschig
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Lijian Tao
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Rodney E Kellems
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.)
| | - Yang Xia
- From the Departments of Nephrology (R.L., L.T., Y.X.) and Urology (C.Z.), Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China; Departments of Biochemistry and Molecular Biology (R.L., W.Z., C.Z., Y.Z., H.W., J.J., R.E.K., Y.X.) and Internal Medicine (W.Z.), University of Texas Medical School at Houston; Program in Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, University of Texas, Houston (J.J., W.Z., R.E.K., Y.X.); and Department of Anesthesiology, University of Colorado-Medical School, Denver (A.G., H.K.E.).
| |
Collapse
|
75
|
Olkowicz M, Chlopicki S, Smolenski RT. Perspectives for angiotensin profiling with liquid chromatography/mass spectrometry to evaluate ACE/ACE2 balance in endothelial dysfunction and vascular pathologies. Pharmacol Rep 2015; 67:778-85. [PMID: 26321281 DOI: 10.1016/j.pharep.2015.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 03/22/2015] [Accepted: 03/25/2015] [Indexed: 02/07/2023]
Abstract
Vascular injury, characterized by endothelial dysfunction, inflammation, structural remodeling, thrombosis and calcification leads to cardiovascular diseases. Angiotensin (Ang) II (1-8) - synthesized mainly by angiotensin converting enzyme (ACE) is the best characterized mediator of the renin-angiotensin system (RAS). This peptide initially identified by its vasoactive properties was found to play a major role in vascular response to insult. However, recent discovery of angiotensin converting enzyme 2 (ACE2) that produces vasoprotective Ang-(1-7) peptide highlighted complexity of the system and suggested that balance between ACE/Ang II and ACE2/Ang-(1-7) is fundamental in maintaining vascular homeostasis and its disorders are associated with cardiovascular pathology. There is therefore a need to develop methods for comprehensive analysis of biologically active Ang peptides and their metabolites of ACE/Ang II and ACE2/Ang-(1-7) axes. Liquid chromatography/mass spectrometry (LC/MS) is an analytical technique that offers potential for specific, simultaneous analysis of Ang peptides. With sensitivity added by application of preconcentration nanochromatography reaching picomolar concentrations, practically all Ang peptides identified so far could be quantified in biological samples. Ang profiling is important not only for understanding their physiological or pathological role but could also serve as an early diagnostic biomarker of endothelial dysfunction and cardiovascular pathology. It could also be used for monitoring the efficacy of the RAS-targeted therapies. Although, the methodology requires further improvements to adopt it for routine application, Ang peptide profiling with targeted LC/MS analysis might assess functional balance between ACE/Ang II and ACE2/Ang-(1-7) axes, facilitate our understanding of the cardiovascular pathology and enhance biomarker portfolio in cardiovascular diseases.
Collapse
Affiliation(s)
- Mariola Olkowicz
- Department of Biochemistry, Medical University of Gdansk, Gdańsk, Poland; Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, Poznań, Poland.
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland; Department of Experimental Pharmacology, Jagiellonian University Medical College, Kraków, Poland
| | | |
Collapse
|
76
|
Gong Y, Cheng X, Li C, Li J, Jin M, Sun B, Tian H. Metabolic diseases and sex hormone levels affect differently in the development of cardiovascular and cerebrovascular diseases in elderly males. Aging Male 2015; 18:27-33. [PMID: 25657081 DOI: 10.3109/13685538.2014.1002082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE This single-centre cross-sectional study aimed to investigate the metabolic and gonadal risk factors of vascular diseases in elderly males. METHODS After screening, 337 subjects aged 60-90 were found to be qualified. Odds ratios (ORs) in cross-table analyses and exp(B) in logistic regression analyses were used to evaluate the vascular risk of dependent factors. R(2) of logistic regression equation was used to estimate the goodness-of-fit of vascular diseases logistic regression models. RESULTS Hypertension increased the risk of cardiovascular disease (CAVD) in elderly men approximately 3-fold. The number of metabolic diseases also correlated with incremental risks of CAVD; presence of one abnormality approximately increases the risk approximately 62%. Cerebrovascular disease (CEVD) development was closely associated with both metabolic syndrome and sex hormone levels; their explanation effects of single action and combined action were 13.2%, 12.55% and 28.5%. C-peptide might be the underlying mechanism of the metabolic syndrome's effect on CEVD. C-peptide = 2.43 U/L and FE(2) = 0.66 were the tangent points in receiver operating characteristic (ROC) analyses. CONCLUSIONS Metabolic diseases and sex hormones play different roles in the development of CAVD and CEVD, the methods for vascular protection in elderly men should be promoted differently according to the their risks of CAVD and CEVD.
Collapse
Affiliation(s)
- Yanping Gong
- Department of Geriatric Endocrinology, The Chinese People's Liberation Army General Hospital , Beijing , China
| | | | | | | | | | | | | |
Collapse
|
77
|
Grobe N, Leiva O, Morris M, Elased KM. Loss of prolyl carboxypeptidase in two-kidney, one-clip goldblatt hypertensive mice. PLoS One 2015; 10:e0117899. [PMID: 25706121 PMCID: PMC4338234 DOI: 10.1371/journal.pone.0117899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 01/05/2015] [Indexed: 11/19/2022] Open
Abstract
It is well documented that angiotensin (Ang) II contributes to kidney disease progression. The protease prolyl carboxypeptidase (PRCP) is highly expressed in the kidney and may be renoprotective by degrading Ang II to Ang-(1-7). The aim of the study was to investigate whether renal PRCP protein expression and activity are altered in two-kidney, one-clip (2K1C) Goldblatt hypertensive mice. Left renal artery was constricted by using 0.12 mm silver clips. Blood pressure was measured using telemetry over the eleven weeks of study period and revealed an immediate increase in 2K1C animals during the first week of clip placement which was followed by a gradual decrease to baseline blood pressure. Similarly, urinary albumin excretion was significantly increased one week after 2K1C and returned to baseline levels during the following weeks. At 2 weeks and at the end of the study, renal pathologies were exacerbated in the 2K1C model as revealed by a significant increase in mesangial expansion and renal fibrosis. Renal PRCP expression and activity were significantly reduced in clipped kidneys. Immunofluorescence revealed the loss of renal tubular PRCP but not glomerular PRCP. In contrast, expression of prolyl endopeptidase, another enzyme capable of converting Ang II into Ang-(1-7), was not affected, while angiotensin converting enzyme was elevated in unclipped kidneys and renin was increased in clipped kidneys. Results suggest that PRCP is suppressed in 2K1C and that this downregulation may attenuate renoprotective effects via impaired Ang II degradation by PRCP.
Collapse
Affiliation(s)
- Nadja Grobe
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, United States of America
- * E-mail:
| | - Orly Leiva
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, United States of America
| | - Mariana Morris
- College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, United States of America
| | - Khalid M. Elased
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, United States of America
| |
Collapse
|
78
|
Yang T. Crosstalk between (Pro)renin receptor and COX-2 in the renal medulla during angiotensin II-induced hypertension. Curr Opin Pharmacol 2015; 21:89-94. [PMID: 25681793 DOI: 10.1016/j.coph.2014.12.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 12/16/2014] [Accepted: 12/21/2014] [Indexed: 01/13/2023]
Abstract
Angiotensin II (AngII) is an octapeptide hormone that plays a central role in regulation of sodium balance, plasma volume, and blood pressure. Its role in the pathogenesis of hypertension is highlighted by the wide use of inhibitors of the renin-angiotensin system (RAS) as the first-line antihypertensive therapy. However, despite intensive investigation, the mechanism of AngII-induced hypertension is still incompletely understood. Although diverse pathways are likely involved, increasing evidence suggests that the activation of intrarenal RAS may represent a dominant mechanism of AngII-induced hypertension. (Pro)renin receptor (PRR), a potential regulator of intrarenal RAS, is expressed in the intercalated cells of the collecting duct (CD) and induced by AngII, in parallel with increased renin in the principal cells of the CD. Activation of PRR elevated PGE2 release and COX-2 expression in renal inner medullary cells whereas COX-2-derived PGE2via the EP4 receptor mediates the upregulation of PRR during AngII infusion, thus forming a vicious cycle. The mutually stimulatory relationship between PRR and COX-2 in the distal nephron may play an important role in mediating AngII-induced hypertension.
Collapse
Affiliation(s)
- Tianxin Yang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China; Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, UT, United States.
| |
Collapse
|
79
|
Shenoy V, Ferreira AJ, Katovich M, Raizada MK. Angiotensin-Converting Enzyme 2/Angiotensin-(1-7)/Mas Receptor Axis. THE PROTECTIVE ARM OF THE RENIN ANGIOTENSIN SYSTEM (RAS) 2015. [PMCID: PMC7149638 DOI: 10.1016/b978-0-12-801364-9.00038-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Experimental and clinical evidence supports an active role of the renin–angiotensin system (RAS) in the pathogenesis and progression of lung diseases. Angiotensin II (Ang II), a key vasoactive peptide of the RAS, has been implicated in pulmonary disorders such as pulmonary arterial hypertension, lung fibrosis, chronic obstructive pulmonary disease, and acute respiratory distress syndrome. Over the past few years, the classical concept of the RAS has undergone substantial changes to include several new active components. Among them, the identification of angiotensin-converting enzyme 2 (ACE2), its metabolic product angiotensin-(1-7) (Ang-(1-7)), and the Mas receptor has been of biological significance since these components form a counterregulatory axis (ACE2/Ang-(1-7)/Mas) that opposes the detrimental actions of Ang II. In this chapter, we will discuss the role of the ACE2/Ang-(1-7)/Mas axis in lung diseases and describe novel therapeutic approaches to activate this axis for the treatment of pulmonary disorders.
Collapse
|
80
|
Munshi A, Das S, Kaul S. Genetic determinants in ischaemic stroke subtypes: Seven year findings and a review. Gene 2015; 555:250-9. [DOI: 10.1016/j.gene.2014.11.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/06/2014] [Accepted: 11/09/2014] [Indexed: 02/06/2023]
|
81
|
Garcia RA, Yan M, Search D, Zhang R, Carson NL, Ryan CS, Smith-Monroy C, Zheng J, Chen J, Kong Y, Tang H, Hellings SE, Wardwell-Swanson J, Dinchuk JE, Psaltis GC, Gordon DA, Glunz PW, Gargalovic PS. P2Y6 receptor potentiates pro-inflammatory responses in macrophages and exhibits differential roles in atherosclerotic lesion development. PLoS One 2014; 9:e111385. [PMID: 25360548 PMCID: PMC4216081 DOI: 10.1371/journal.pone.0111385] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/23/2014] [Indexed: 12/15/2022] Open
Abstract
Background P2Y6, a purinergic receptor for UDP, is enriched in atherosclerotic lesions and is implicated in pro-inflammatory responses of key vascular cell types and macrophages. Evidence for its involvement in atherogenesis, however, has been lacking. Here we use cell-based studies and three murine models of atherogenesis to evaluate the impact of P2Y6 deficiency on atherosclerosis. Methodology/Principal Findings Cell-based studies in 1321N1 astrocytoma cells, which lack functional P2Y6 receptors, showed that exogenous expression of P2Y6 induces a robust, receptor- and agonist-dependent secretion of inflammatory mediators IL-8, IL-6, MCP-1 and GRO1. P2Y6-mediated inflammatory responses were also observed, albeit to a lesser extent, in macrophages endogenously expressing P2Y6 and in acute peritonitis models of inflammation. To evaluate the role of P2Y6 in atherosclerotic lesion development, we used P2Y6-deficient mice in three mouse models of atherosclerosis. A 43% reduction in aortic arch plaque was observed in high fat-fed LDLR knockout mice lacking P2Y6 receptors in bone marrow-derived cells. In contrast, no effect on lesion development was observed in fat-fed whole body P2Y6xLDLR double knockout mice. Interestingly, in a model of enhanced vascular inflammation using angiotensin II, P2Y6 deficiency enhanced formation of aneurysms and exhibited a trend towards increased atherosclerosis in the aorta of LDLR knockout mice. Conclusions P2Y6 receptor augments pro-inflammatory responses in macrophages and exhibits a pro-atherogenic role in hematopoietic cells. However, the overall impact of whole body P2Y6 deficiency on atherosclerosis appears to be modest and could reflect additional roles of P2Y6 in vascular disease pathophysiologies, such as aneurysm formation.
Collapse
Affiliation(s)
- Ricardo A. Garcia
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Mujing Yan
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Debra Search
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Rongan Zhang
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Nancy L. Carson
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Carol S. Ryan
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Constance Smith-Monroy
- Applied Genomics, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Joanna Zheng
- Pharmaceutical Compound Optimization: Discovery Toxicology, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Jian Chen
- Applied Genomics, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Yan Kong
- Lead Evaluation, Bristol-Myers Squibb Company, Lawrenceville, New Jersey, United States of America
| | - Huaping Tang
- Lead Evaluation, Bristol-Myers Squibb Company, Lawrenceville, New Jersey, United States of America
| | - Samuel E. Hellings
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Judith Wardwell-Swanson
- Applied Genomics, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Joseph E. Dinchuk
- Applied Genomics, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - George C. Psaltis
- Veterinary Sciences, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - David A. Gordon
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Peter W. Glunz
- Discovery Chemistry, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Peter S. Gargalovic
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
82
|
Zhang Z, Chen L, Zhong J, Gao P, Oudit GY. ACE2/Ang-(1–7) signaling and vascular remodeling. SCIENCE CHINA-LIFE SCIENCES 2014; 57:802-8. [DOI: 10.1007/s11427-014-4693-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 05/20/2014] [Indexed: 02/06/2023]
|
83
|
Kim SY, Lee J. Exercise Training suppresses vascular fibrosis in aging obesity induced rats. J Exerc Nutrition Biochem 2014; 18:175-80. [PMID: 25566453 PMCID: PMC4241917 DOI: 10.5717/jenb.2014.18.2.175] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 05/13/2014] [Accepted: 05/21/2014] [Indexed: 11/24/2022] Open
Abstract
[Purpose] The aim of this study was to investigate the effects of exercise training (ET) on vascular fibrosis in aging model rats with diet-induced obesity. [Methods] Twenty-four male Sprague-Dawley rats were divided into 3 groups: Aging control (A-C), A-C with high fat diet (AHF), AHF with ET (AHF + ET). Aging was induced by D-galactose (D-gal) and obesity was induced by HFD (60% fat) for 9 weeks. The experimental rats performed swimming (60 min/day, 5 days/week) for 8 weeks. All rat aorta samples were harvested for RT-PCR and morphologic analyses. [Results] The exercise training significantly decreased levels of AT-1, TGF-ß and Coll-1 gene expression compared to AHF group. The AHF + ET group showed a reduced collagen accumulation in the aorta media compared to AHF group. [Conclusion] These results suggest that ET could protect the aging obesity aorta against down-regulation of fibrotic factors (AT-1, TGF-ß and Coll-1 gene) and fibrosis by inhibition of collagen accumulation in the aorta media.
Collapse
Affiliation(s)
- Shin Young Kim
- Department of Anatomy and Cell Biology, Collage of Medicine, Han-Yang University, Seoul, 133-791, Korea
| | - Jin Lee
- Department of Anatomy and Cell Biology, Collage of Medicine, Han-Yang University, Seoul, 133-791, Korea
| |
Collapse
|
84
|
Haschke M, Schuster M, Poglitsch M, Loibner H, Salzberg M, Bruggisser M, Penninger J, Krähenbühl S. Pharmacokinetics and pharmacodynamics of recombinant human angiotensin-converting enzyme 2 in healthy human subjects. Clin Pharmacokinet 2014; 52:783-92. [PMID: 23681967 DOI: 10.1007/s40262-013-0072-7] [Citation(s) in RCA: 288] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVES Angiotensin-converting enzyme 2 (ACE2) converts angiotensin II (Ang1-8) to angiotensin 1-7 (Ang1-7), a functional antagonist of Ang1-8, with vasodilatory, antiproliferative, antiangiogenic, and anti-inflammatory properties. In conditions with an unbalanced renin-angiotensin-aldosterone system with elevated Ang1-8, administration of ACE2 has shown promising effects in a variety of animal models. Enhancing ACE2 activity by exogenous administration of ACE2 might also be beneficial in human diseases with pathologically elevated Ang1-8. As a first step we performed a first-in-man study to determine pharmacokinetics, pharmacodynamics, safety, and tolerability of recombinant ACE2 in healthy volunteers. METHODS Recombinant human ACE2 (rhACE2) was administered intravenously to healthy human subjects in a randomized, double-blind, placebo-controlled, single-dose, dose-escalation study followed by an open-label multiple-dose study. ACE2 concentrations were determined by quantifying ACE2 activity and ACE2 content in plasma samples. Concentrations of the angiotensin system effector peptides Ang1-8, Ang1-7, and Ang1-5 were determined using a liquid chromatography-tandem mass spectrometry method. RESULTS Single rhACE2 doses of 100-1,200 μg/kg caused a dose-dependent increase of systemic exposure with biphasic elimination and a dose-independent terminal half-life of 10 h. In all single-dose cohorts, Ang1-8 decreased within 30 min postinfusion, angiotensin 1-7 (Ang1-7) either increased (100 and 200 μg/kg doses), decreased, or remained unchanged (400-1,200 μg/kg doses), whereas angiotensin 1-5 (Ang1-5) transiently increased for all doses investigated. With the exception of the lowest rhACE2 dose, the decrease in Ang1-8 levels lasted for at least 24 h. Repeated dosing (400 μg/kg for 3 or 6 days) caused only minimal accumulation of ACE2, and Ang1-8 levels were suppressed over the whole application period. CONCLUSIONS Administration of rhACE2 was well tolerated by healthy human subjects. Exposure was dose dependent with a dose-independent terminal elimination half-life in the range of 10 h. Despite marked changes in angiotensin system peptide concentrations, cardiovascular effects were absent, suggesting the presence of effective compensatory mechanisms in healthy volunteers.
Collapse
Affiliation(s)
- Manuel Haschke
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel, 4031, Basel, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Increased negative impact of donor HLA-specific together with non-HLA-specific antibodies on graft outcome. Transplantation 2014; 97:595-601. [PMID: 24162250 DOI: 10.1097/01.tp.0000436927.08026.a8] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND De novo donor HLA-specific (dnDSA) and non-HLA antibodies including antiangiotensin type 1 receptor antibodies (AT1R-abs) have been associated with antibody-mediated rejection (AMR) and decreased graft survival as well as cellular-mediated rejection (CMR) and early onset of microvasculopathy in heart transplantation. The aim of our study was to determine the impact of anti-AT1R-ab and anti-donor HLA-specific antibody (DSA) on clinical outcomes. METHODS Pretransplant and posttransplant sera from 200 recipients transplanted between May 2007 and August 2011 were tested for DSA (Luminex-based single antigen bead assay) and AT1R-ab (enzyme-linked immunosorbent assay). Two cutoff levels (≥ 17 and ≥ 12 units) were used to define high and intermediate binding of AT1R-ab. Clinical parameters examined were 5-year AMR/CMR (≥ grade 2), coronary artery vasculopathy, and survival. RESULTS At 2 years after transplant, freedom from AMR and/or CMR was 95.4% for those with no DSA (n=175), 66.9% for those with dnDSA (n=19), and 25% for those with DSA at transplant (n=6) (P<0.0001). Neither ≥ 17 nor ≥ 12 units of pretransplant levels indicated a significant difference in freedom from AMR and/or CMR. When both dnDSA and AT1R-ab ≥ 17 or ≥ 12 units were considered, freedom from AMR and/or CMR decreased to 50% and 45% (P<0.0001), respectively. Coronary artery vasculopathy and survival were not significantly impacted. CONCLUSIONS These results show the increased negative impact of dnDSA and AT1R-ab on freedom from AMR and/or CMR and an increased hazard ratio when both parameters are considered. Both HLA- and non-HLA-specific antibodies seem to impact graft outcome in heart transplantation.
Collapse
|
86
|
Angiotensin II induces mitochondrial dysfunction and promotes apoptosis via JNK signalling pathway in primary mouse calvaria osteoblast. Arch Oral Biol 2014; 59:513-23. [PMID: 24632094 DOI: 10.1016/j.archoralbio.2014.02.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 12/20/2013] [Accepted: 02/25/2014] [Indexed: 02/05/2023]
Abstract
OBJECTIVES This present study was designed to investigate the effects of Angiotensin II on mitochondrial functions, ROS generation and c-jun N-terminal kinases (JNK) signalling pathway-mediated cell apoptosis in mouse calvaria osteoblasts. METHODS Calvaria osteoblast were isolated and cultured. The cells were separated into two groups-control and treated groups-where the latter was stimulated with angiotensin II (Ang II). Mitochondrial reactive oxygen species (ROS) and superoxide production were measured. Intracellular ATP levels were also detected. The cell proliferation rate was determined for the two groups. Protein production such as Anti-Bax, Bcl-2, COX IV and activation of c-jun N-terminal kinases signal (JNK) pathway was measured by enzyme-linked immunosorbent assay (ELISA) methods and Western blotting in this study. RESULTS Ang II treated cells showed significantly higher levels of superoxide production compared to the control group (p<0.05). Conversely, Ang II induced inhibitory effects on mitochondrial respiratory enzyme complexes, cause membrane potential dissipation, ATP loss and promote ROS generation, cell apoptosis in cultured osteoblasts. In addition, JNK phosphorylations were involved in activating the mitochondria-dependent apoptotic pathway following Ang II stimulation, as pre-treatment of JNK-specific inhibitor SP600125 could rescue osteoblast cells from apoptosis by enhancing the anti-apoptotic protein Bcl-2 expressions, suppressing the translocation of Bax from cytosol into mitochondria, blocking cytochrome C release and caspase-3 activation. CONCLUSIONS Ang II stimulates osteoblast apoptosis via suppression of the mitochondrial respiratory enzymes, membrane potential and cellular ATP productions. Clinical application with Ang II-stimulated osteoblast could be used for modelling or bone resorption in the oral region.
Collapse
|
87
|
|
88
|
Alghamri MS, Morris M, Meszaros JG, Elased KM, Grobe N. Novel role of aminopeptidase-A in angiotensin-(1-7) metabolism post myocardial infarction. Am J Physiol Heart Circ Physiol 2014; 306:H1032-40. [PMID: 24464749 DOI: 10.1152/ajpheart.00911.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aminopeptidase-A (APA) is a less well-studied enzyme of the renin-angiotensin system. We propose that it is involved in cardiac angiotensin (ANG) metabolism and its pathologies. ANG-(1-7) can ameliorate remodeling after myocardial injury. The aims of this study are to (1) develop mass spectrometric (MS) approaches for the assessment of ANG processing by APA within the myocardium; and (2) investigate the role of APA in cardiac ANG-(1-7) metabolism after myocardial infarction (MI) using sensitive MS techniques. MI was induced in C57Bl/6 male mice by ligating the left anterior descending (LAD) artery. Frozen mouse heart sections (in situ assay) or myocardial homogenates (in vitro assay) were incubated with the endogenous APA substrate, ANG II. Results showed concentration- and time-dependent cardiac formation of ANG III from ANG II, which was inhibited by the specific APA inhibitor, 4-amino-4-phosphonobutyric acid. Myocardial APA activity was significantly increased 24 h after LAD ligation (0.82 ± 0.02 vs. 0.32 ± 0.02 ρmol·min(-1)·μg(-1), MI vs. sham, P < 0.01). Both MS enzyme assays identified the presence of a new peptide, ANG-(2-7), m/z 784, which accumulated in the MI (146.45 ± 6.4 vs. 72.96 ± 7.0%, MI vs. sham, P < 0.05). Use of recombinant APA enzyme revealed that APA is responsible for ANG-(2-7) formation from ANG-(1-7). APA exhibited similar substrate affinity for ANG-(1-7) compared with ANG II {Km (ANG II) = 14.67 ± 1.6 vs. Km [ANG-(1-7)] = 6.07 ± 1.12 μmol/l, P < 0.05}. Results demonstrate a novel role of APA in ANG-(1-7) metabolism and suggest that the upregulation of APA, which occurs after MI, may deprive the heart of cardioprotective ANG-(1-7). Thus APA may serve as a potentially novel therapeutic target for management of tissue remodeling after MI.
Collapse
Affiliation(s)
- Mahmoud S Alghamri
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | | | | | | | | |
Collapse
|
89
|
Tocci G, Sciarretta S, Facciolo C, Volpe M. Antihypertensive strategy based on angiotensin II receptor blockers: a new gateway to reduce risk in hypertension. Expert Rev Cardiovasc Ther 2014; 5:767-76. [PMID: 17605654 DOI: 10.1586/14779072.5.4.767] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Effective treatment of high blood pressure levels represents a key strategy for reducing global cardiovascular risk. Other factors, beyond blood pressure control, however, appear to be of potential relevance in reducing the risk related to hypertension. Recent clinical trials have demonstrated that those pharmacological agents that counteract the renin-angiotensin system may confer additional clinical benefits across the spectrum of cardiovascular disease, beyond their blood pressure-lowering properties. These studies are largely based on the use of an antihypertensive strategy, based on the association between angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers (ARBs) and low-dose thiazide diuretics or calcium channel blockers. Over the last few decades, clinical trials have also tested the potential effects of combination therapy based on the association between angiotensin-converting enzyme inhibitors or ARBs and other renin-angiotensin system-blocking agents, including mineralocorticoid receptor antagonists and, more recently, renin inhibitors. This review highlights the evidence derived from recent clinical trials, supporting a role for pharmacological strategies based on ARBs in primary and secondary prevention of cardiovascular and renal disease.
Collapse
Affiliation(s)
- Giuliano Tocci
- University La Sapienza of Rome, Cardiology, II Faculty of Medicine, Sant'Andrea Hospital, Rome, Italy.
| | | | | | | |
Collapse
|
90
|
Campbell DJ. Clinical relevance of local Renin Angiotensin systems. Front Endocrinol (Lausanne) 2014; 5:113. [PMID: 25071727 PMCID: PMC4095645 DOI: 10.3389/fendo.2014.00113] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/30/2014] [Indexed: 12/12/2022] Open
Affiliation(s)
- Duncan J. Campbell
- St. Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Medicine, University of Melbourne, St. Vincent’s Hospital, Fitzroy, VIC, Australia
- *Correspondence:
| |
Collapse
|
91
|
Liu CC, Karimi Galougahi K, Weisbrod RM, Hansen T, Ravaie R, Nunez A, Liu YB, Fry N, Garcia A, Hamilton EJ, Sweadner KJ, Cohen RA, Figtree GA. Oxidative inhibition of the vascular Na+-K+ pump via NADPH oxidase-dependent β1-subunit glutathionylation: implications for angiotensin II-induced vascular dysfunction. Free Radic Biol Med 2013; 65:563-572. [PMID: 23816524 PMCID: PMC4474148 DOI: 10.1016/j.freeradbiomed.2013.06.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 02/07/2023]
Abstract
Glutathionylation of the Na(+)-K(+) pump's β1-subunit is a key molecular mechanism of physiological and pathophysiological pump inhibition in cardiac myocytes. Its contribution to Na(+)-K(+) pump regulation in other tissues is unknown, and cannot be assumed given the dependence on specific β-subunit isoform expression and receptor-coupled pathways. As Na(+)-K(+) pump activity is an important determinant of vascular tone through effects on [Ca(2+)]i, we have examined the role of oxidative regulation of the Na(+)-K(+) pump in mediating angiotensin II (Ang II)-induced increases in vascular reactivity. β1-subunit glutathione adducts were present at baseline and increased by exposure to Ang II in rabbit aortic rings, primary rabbit aortic vascular smooth muscle cells (VSMCs), and human arterial segments. In VSMCs, Ang II-induced glutathionylation was associated with marked reduction in Na(+)-K(+)ATPase activity, an effect that was abolished by the NADPH oxidase inhibitory peptide, tat-gp91ds. In aortic segments, Ang II-induced glutathionylation was associated with decreased K(+)-induced vasorelaxation, a validated index of pump activity. Ang II-induced oxidative inhibition of Na(+)-K(+) ATPase and decrease in K(+)-induced relaxation were reversed by preincubation of VSMCs and rings with recombinant FXYD3 protein that is known to facilitate deglutathionylation of β1-subunit. Knock-out of FXYD1 dramatically decreased K(+)-induced relaxation in a mouse model. Attenuation of Ang II signaling in vivo by captopril (8 mg/kg/day for 7 days) decreased superoxide-sensitive DHE levels in the media of rabbit aorta, decreased β1-subunit glutathionylation, and enhanced K(+)-induced vasorelaxation. Ang II inhibits the Na(+)-K(+) pump in VSMCs via NADPH oxidase-dependent glutathionylation of the pump's β1-subunit, and this newly identified signaling pathway may contribute to altered vascular tone. FXYD proteins reduce oxidative inhibition of the Na(+)-K(+) pump and may have an important protective role in the vasculature under conditions of oxidative stress.
Collapse
Affiliation(s)
- Chia-Chi Liu
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Keyvan Karimi Galougahi
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia; Department of Cardiology, Royal North Shore Hospital, Sydney, Australia
| | - Robert M Weisbrod
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Thomas Hansen
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Ramtin Ravaie
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Andrea Nunez
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Yi B Liu
- Laboratory Membrane Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Natasha Fry
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Alvaro Garcia
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Elisha J Hamilton
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Kathleen J Sweadner
- Laboratory Membrane Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Richard A Cohen
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Gemma A Figtree
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia; Department of Cardiology, Royal North Shore Hospital, Sydney, Australia.
| |
Collapse
|
92
|
Rodriguez-Perez AI, Dominguez-Meijide A, Lanciego JL, Guerra MJ, Labandeira-Garcia JL. Dopaminergic degeneration is enhanced by chronic brain hypoperfusion and inhibited by angiotensin receptor blockage. AGE (DORDRECHT, NETHERLANDS) 2013; 35:1675-90. [PMID: 22986582 PMCID: PMC3776095 DOI: 10.1007/s11357-012-9470-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 08/30/2012] [Indexed: 05/24/2023]
Abstract
The possible interaction between brain hypoperfusion related to aging and/or vascular disease, vascular parkinsonism and Parkinson's disease, as well as the possible contribution of aging-related chronic brain hypoperfusion in the development or severity of Parkinson's disease are largely unknown. We used a rat model of chronic cerebral hypoperfusion to study the long-term effects of hypoperfusion on dopaminergic neurons and the possible synergistic effects between chronic hypoperfusion and factors that are deleterious to dopaminergic neurons, such as the dopaminergic neurotoxin 6-hydroxydopamine. Chronic hypoperfusion induced significant loss of dopaminergic neurons and striatal dopaminergic terminals and a reduction in striatal dopamine levels. Furthermore, intrastriatal administration of 6-hydroxydopamine in rats subjected to chronic hypoperfusion induced a significantly greater loss of dopaminergic neurons than in sham-operated control rats. The dopaminergic neuron loss was significantly reduced by oral treatment with angiotensin type 1 receptor antagonist candesartan (3 mg/kg/day). The levels of angiotensin type 2 receptors were lower and the levels of angiotensin type 1 receptors, interleukin-1 β and nicotinamide adenine dinucleotide phosphate oxidase activity were higher in the substantia nigra of rats subjected to chronic hypoperfusion than in control rats; this was significantly reduced by treatment with candesartan. The results suggest that early treatment of vascular disease should be considered in the treatment of aged Parkinson's disease patients and Parkinson's disease patients with cerebrovascular risk factors. The findings also suggest that inhibition of brain renin-angiotensin activity may be useful as a neuroprotective strategy.
Collapse
Affiliation(s)
- Ana I. Rodriguez-Perez
- />Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- />Networking Research Centre on Neurodegenerative Diseases (CIBERNED), Santiago de Compostela, Spain
| | - Antonio Dominguez-Meijide
- />Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- />Networking Research Centre on Neurodegenerative Diseases (CIBERNED), Santiago de Compostela, Spain
| | - Jose L. Lanciego
- />Neurosciences Division, CIMA, University of Navarra, Pamplona, Spain
- />Networking Research Centre on Neurodegenerative Diseases (CIBERNED), Santiago de Compostela, Spain
| | - Maria J. Guerra
- />Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- />Networking Research Centre on Neurodegenerative Diseases (CIBERNED), Santiago de Compostela, Spain
| | - Jose L. Labandeira-Garcia
- />Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- />Networking Research Centre on Neurodegenerative Diseases (CIBERNED), Santiago de Compostela, Spain
| |
Collapse
|
93
|
Lan TH, Huang XQ, Tan HM. Vascular fibrosis in atherosclerosis. Cardiovasc Pathol 2013; 22:401-7. [DOI: 10.1016/j.carpath.2013.01.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 12/10/2012] [Accepted: 01/08/2013] [Indexed: 01/10/2023] Open
|
94
|
Reinsmoen NL. Role of angiotensin II type 1 receptor-activating antibodies in solid organ transplantation. Hum Immunol 2013; 74:1474-7. [PMID: 23831255 DOI: 10.1016/j.humimm.2013.06.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 06/04/2013] [Accepted: 06/14/2013] [Indexed: 02/03/2023]
Abstract
Angiotensin type I receptor (AT1R) mediates physiologic and pathophysiologic actions of its ligand, angiotensin II. Overactivity of the AT1R and angiotensin II interaction results in hypertension and vascular remodeling. Antibodies to AT1R have been implicated in several vascular pathologies. In renal transplantation, elevated levels of anti-AT1R antibodies have been associated with antibody mediated rejection (AMR) in the absence of donor HLA specific antibodies. In heart transplantation, increased levels of anti-AT1R antibodies have been associated with cellular and AMR as well as an early onset of microvasculopathy. This review summarizes the current investigations regarding the impact of anti-AT1R antibodies in solid organ transplantation and provides insight into the humoral response in the presence of non-HLA and HLA specific antibodies.
Collapse
Affiliation(s)
- Nancy L Reinsmoen
- HLA Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States.
| |
Collapse
|
95
|
Kuneš J, Kadlecová M, Vaněčková I, Zicha J. Critical developmental periods in the pathogenesis of hypertension. Physiol Res 2013; 61:S9-17. [PMID: 22827878 DOI: 10.33549/physiolres.932364] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Hypertension is one of the major risk factor of cardiovascular diseases, but after a century of clinical and basic research, the discrete etiology of this disease is still not fully understood. One reason is that blood pressure is a quantitative trait with multifactorial determination. Numerous genes, environmental factors as well as epigenetic factors should be considered. There is no doubt that although the full manifestation of hypertension and other cardiovascular diseases usually occurs predominantly in adulthood and/or senescence, the roots can be traced back to early ontogeny. The detailed knowledge of the ontogenetic changes occurring in the cardiovascular system of experimental animals during particular critical periods (developmental windows) could help to solve this problem in humans and might facilitate the age-specific prevention of human hypertension. We thus believe that this approach might contribute to the reduction of cardiovascular morbidity among susceptible individuals in the future.
Collapse
Affiliation(s)
- J Kuneš
- Centre for Cardiovascular Research, Prague, Czech Republic.
| | | | | | | |
Collapse
|
96
|
Campagnaro BP, Tonini CL, Doche LM, Nogueira BV, Vasquez EC, Meyrelles SS. Renovascular hypertension leads to DNA damage and apoptosis in bone marrow cells. DNA Cell Biol 2013; 32:458-66. [PMID: 23786322 DOI: 10.1089/dna.2013.2065] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Angiotensin II (Ang II), which plays a pivotal role in the pathophysiology of the two-kidney, one-clip (2K1C) Goldblatt hypertension, has been associated with augmented generation of reactive oxygen species (ROS) in some cells and tissues. In the present study, we evaluated the influence of 2K1C hypertension on oxidative stress, DNA fragmentation, and apoptosis of bone marrow (BM) cells. Two weeks after the renal artery clipping or Sham operation, flow cytometry analysis showed a higher production of superoxide anions (approximately sixfold) and hydrogen peroxide (approximately twofold) in 2K1C hypertensive than in Sham normotensive mice. 2K1C mice also showed an augmented DNA fragmentation (54%) and apoptotic cells (21%). Our data show that the 2K1C renovascular hypertension is characterized by an increased production of ROS, DNA damage, and apoptosis of BM, which is a fundamental source of the cells involved in tissue repair.
Collapse
Affiliation(s)
- Bianca P Campagnaro
- Laboratory of Transgenes, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | | | | | | | | | |
Collapse
|
97
|
Castellon X, Bogdanova V. Screening for subclinical atherosclerosis by noninvasive methods in asymptomatic patients with risk factors. Clin Interv Aging 2013; 8:573-80. [PMID: 23761967 PMCID: PMC3673861 DOI: 10.2147/cia.s40150] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular death due to the increasing prevalence of the disease and the impact of risk factors such as diabetes, obesity or smoking. Sudden cardiac death is the primary consequence of coronary artery disease in 50% of men and 64% of women. Currently the only available strategy to reduce mortality in the at-risk population is primary prevention; the target population must receive screening for atherosclerosis. The value of screening for subclinical atherosclerosis is still relevant, it has become standard clinical practice with the emergence of new noninvasive techniques (radio frequency [RF] measurement of intima-media thickness [RFQIMT] and arterial stiffness [RFQAS], and flow-mediated vasodilatation [FMV]), which have been used by our team since 2007 and are based on detection marker integrators which reflect the deleterious effect of risk factors on arterial remodeling before the onset of clinical events. These techniques allow the study of values according to age and diagnosis of the pathological value, the thickness of the intima media (RFQIMT), the speed of the pulse wave (RFQAS), and the degree of endothelial dysfunction (FMV). This screening is justified in asymptomatic patients with cardiovascular risk factors (hypertension, diabetes, obesity, dyslipidemia, and tobacco smoking). Studies conducted by RF coupled with two-dimensional echo since 2007 have led to a more detailed analysis of the state of the arterial wall. The various examinations allow an assessment of the degree of subclinical atherosclerosis and its impact on arterial remodeling and endothelial function. The use of noninvasive imaging in screening and early detection of subclinical atherosclerosis is reliable and reproducible and allows us to assess the susceptibility of our patients with risk factors and ensures better monitoring of atherosclerosis, thus reducing the occurrence of cardiovascular events in the long term.
Collapse
Affiliation(s)
- Xavier Castellon
- Department of Cardiology, Private Hospital Athis Mons, Paris, France.
| | | |
Collapse
|
98
|
DNA damage and augmented oxidative stress in bone marrow mononuclear cells from Angiotensin-dependent hypertensive mice. Int J Hypertens 2013; 2013:305202. [PMID: 23476745 PMCID: PMC3586517 DOI: 10.1155/2013/305202] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 01/16/2013] [Indexed: 02/07/2023] Open
Abstract
It has been proposed that the nonhemodynamic effects of angiotensin II are important for the damage observed in the two-kidney, one-clip (2K1C) renovascular hypertension model. Much evidence confirms that angiotensin II is directly involved in NAD(P)H oxidase activation and consequent superoxide anion production, which can damage DNA. The current study was performed to examine the effects of angiotensin-II-dependent hypertension in bone marrow mononuclear cells (BM-MNC); dihydroethidium staining was used to assess reactive oxygen species (ROS) production, and the comet assay was used to assess DNA fragmentation in 2K1C hypertensive mice 14 days after renal artery clipping. In this study we demonstrated that 2K1C hypertensive mice have an elevated lymphocyte count, while undifferentiated BM-MNC counts were diminished. 2K1C mice also showed an augmented ROS production and marked BM-MNC DNA fragmentation. In conclusion, endogenous renin angiotensin system activation-induced arterial hypertension is characterized by excessive ROS production in BM-MNC, which might cause marked DNA damage.
Collapse
|
99
|
Bahramali E, Firouzabadi N, Jonaidi-Jafari N, Shafiei M. Renin-angiotensin system genetic polymorphisms: lack of association with CRP levels in patients with coronary artery disease. J Renin Angiotensin Aldosterone Syst 2013; 15:559-65. [PMID: 23392788 DOI: 10.1177/1470320312474051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Angiotensin (Ang) II is believed to be a potential pro-inflammatory factor. The capability of Ang II to stimulate C-reactive protein (CRP) production has recently been described. Genetic polymorphisms of renin angiotensin system (RAS) components have been described to be associated with the development of coronary artery disease (CAD). This study investigated the association between six different genetic polymorphisms of RAS and serum CRP levels in a sample of CAD patients. Genotyping of RAS genes polymorphisms in 176 patients with documented CAD was performed by a modified polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Measurement of high-sensitivity (hs)-CRP was performed using standard immunoturbidimetric methods. Results show no significant differences in serum CRP regarding different variants of the six polymorphisms studied (p = 0.41, 0.24, 0.25, 0.19, 0.29, and 0.05 for Ang-converting enzyme (ACE) insertion/deletion (I/D), A-240T and A2350G, angiotensinogen M235T, AT1 receptor A1166C, and AT2 receptor C3123A polymorphisms, respectively). In conclusion, genetic polymorphisms of RAS are not associated with increased serum CRP in CAD. Compensation of an increased activity of ACE through counter-regulation and the secretion of CRP under the influence of Ang II in the vessel being local could explain the lack of association between the studied polymorphisms and CRP levels in CAD patients.
Collapse
Affiliation(s)
- Ehsan Bahramali
- Health Research Center, Baqiyatallah University of Medical Sciences, Iran
| | - Negar Firouzabadi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Iran
| | | | - Massoumeh Shafiei
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Iran
| |
Collapse
|
100
|
Wang L, Hu X, Zhang W, Tian F. Angiotensin (1–7) ameliorates angiotensin II-induced inflammation by inhibiting LOX-1 expression. Inflamm Res 2012; 62:219-28. [DOI: 10.1007/s00011-012-0571-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 10/15/2012] [Accepted: 10/22/2012] [Indexed: 01/13/2023] Open
|