51
|
Kostyunin AE, Ovcharenko EA, Barbarash OL. [The renin-angiotensin-aldosterone system as a potential target for therapy in patients with calcific aortic stenosis: a literature review]. ACTA ACUST UNITED AC 2019; 59:4-17. [PMID: 31884936 DOI: 10.18087/cardio.n328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/14/2019] [Indexed: 11/18/2022]
Abstract
Calcific aortic valve stenosis (CAVS) is a serious socio-economic problem in developed countries because this disease is the most common indication for aortic valve replacement. Currently, there are no methods for non-invasive treatment of CAVS. Nevertheless, it is assumed that effective drug therapy for CAVS can be developed on the basis of modulators of the renin-angiotensin-aldosterone system (RAAS), which is involved in the pathogenesis of this disease. The purpose of this paper is to compile and analyze current information on the role of RAAS in the CAVS pathophysiology. Recent data on the effectiveness of RAAS inhibition are reviewed.
Collapse
Affiliation(s)
- A E Kostyunin
- Research Institute for Complex Issues of Cardiovascular Diseases
| | - E A Ovcharenko
- Research Institute for Complex Issues of Cardiovascular Diseases
| | - O L Barbarash
- Research Institute for Complex Issues of Cardiovascular Diseases
| |
Collapse
|
52
|
Aboud HM, Mahmoud MO, Abdeltawab Mohammed M, Shafiq Awad M, Sabry D. Preparation and appraisal of self-assembled valsartan-loaded amalgamated Pluronic F127/Tween 80 polymeric micelles: Boosted cardioprotection via regulation of Mhrt/Nrf2 and Trx1 pathways in cisplatin-induced cardiotoxicity. J Drug Target 2019; 28:282-299. [DOI: 10.1080/1061186x.2019.1650053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Heba M. Aboud
- Department of Pharmaceutics, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed O. Mahmoud
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | | | - Mohammad Shafiq Awad
- Department of Cardiology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
53
|
Oliveira SHP, Brito VGB, Frasnelli SCT, Ribeiro BDS, Ferreira MN, Queiroz DP, Beltan CT, Lara VS, Santos CF. Aliskiren Attenuates the Inflammatory Response and Wound Healing Process in Diabetic Mice With Periodontal Disease. Front Pharmacol 2019; 10:708. [PMID: 31333451 PMCID: PMC6620569 DOI: 10.3389/fphar.2019.00708] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/31/2019] [Indexed: 01/08/2023] Open
Abstract
The aim of this study was to characterize the role of local RAS (renin–angiotensin system) in the inflammatory response of normal (N) and diabetic (D) mice with periodontal disease (PD). Diabetes Mellitus (DM) was induced by peritoneal injection of streptozotocin in Balb/c mice. PD was induced by ligature around the first molar in both N and D, irrespective of whether they were treated with aliskiren (50 mg/kg, Alisk). Mandibles were harvested for histomorphometric analyses, and gingival tissue (GT) was collected to evaluate gene expression and extracellular matrix components (ECM). Immunohistochemical (IHC) analyses were used to localize RAS in GT. The production of C-reactive protein (CRP), IL-1β, CXCL2, and CCL8 was evaluated by enzyme-linked immunosorbent assay (ELISA). Renin was found to exacerbate the inflammation and periodontal bone loss at 14 days after PD, and Alisk inhibited this process in GT of N and D. PD increased CRP, CXCL2, CCL8, and IL-1β production in both animals. Alisk could inhibit CRP, CXCL2, and CCL8 primarily in D animals. However, only CCL8 was decreased in N animals after Alisk pretreatment. PD enhanced expression and production of AGT, ACE, AT1R, and AT2R in both N and D. AT1R expression was higher in D with PD, and AT2R expression was higher in N with PD. ACE2 and receptor Mas (MasR) expression and production was elevated in the control group of both animals. PD inhibited ACE2 in N but not in D. MasR expression was unaffected in both N and D with PD. Alisk reduced expression and production of all RAS components in GT of both animals, except for ACE2 in N. RAS staining was observed in all layers of epithelium, basal cell layer, and lamina propria and was higher in N with PD. Col1a1, Col1a2, Col3a1, and fibronectin (Fn1) were increased in both animals with PD. Alisk inhibited Col1a1 and Fn in both animals, Col1a2 was decreased only in D, while levels of Col3a1 remained unchanged in all animal groups. In conclusion, these data demonstrated the presence and functional role of local RAS in GT, exacerbating the inflammatory response, periodontal bone loss, and wound healing processes in both N and D animal groups. In addition, Alisk was able to significantly reduce gingival inflammation, excessive wound healing processes, and periodontal bone loss.
Collapse
Affiliation(s)
- Sandra Helena Penha Oliveira
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil.,Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - Victor Gustavo Balera Brito
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil.,Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - Sabrina Cruz Tfaile Frasnelli
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - Bianca da Silva Ribeiro
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - Milena Nunes Ferreira
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - Dayane Priscilla Queiroz
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil.,Programa Multicêntrico de Pós-graduação em Ciências Fisiológicas, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - Carluci Taís Beltan
- Department of Basic Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - Vanessa Soares Lara
- Department of Stomatology, Bauru School of Dentistry, University of São Paulo (USP), Bauru, Brazil
| | - Carlos Ferreira Santos
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo (USP), Bauru, Brazil
| |
Collapse
|
54
|
Development of calcific aortic valve disease: Do we know enough for new clinical trials? J Mol Cell Cardiol 2019; 132:189-209. [PMID: 31136747 DOI: 10.1016/j.yjmcc.2019.05.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/11/2019] [Accepted: 05/19/2019] [Indexed: 12/19/2022]
Abstract
Calcific aortic valve disease (CAVD), previously thought to represent a passive degeneration of the valvular extracellular matrix (VECM), is now regarded as an intricate multistage disorder with sequential yet intertangled and interacting underlying processes. Endothelial dysfunction and injury, initiated by disturbed blood flow and metabolic disorders, lead to the deposition of low-density lipoprotein cholesterol in the VECM further provoking macrophage infiltration, oxidative stress, and release of pro-inflammatory cytokines. Such changes in the valvular homeostasis induce differentiation of normally quiescent valvular interstitial cells (VICs) into synthetically active myofibroblasts producing excessive quantities of the VECM and proteins responsible for its remodeling. As a result of constantly ongoing degradation and re-deposition, VECM becomes disorganised and rigid, additionally potentiating myofibroblastic differentiation of VICs and worsening adaptation of the valve to the blood flow. Moreover, disrupted and excessively vascularised VECM is susceptible to the dystrophic calcification caused by calcium and phosphate precipitating on damaged collagen fibers and concurrently accompanied by osteogenic differentiation of VICs. Being combined, passive calcification and biomineralisation synergistically induce ossification of the aortic valve ultimately resulting in its mechanical incompetence requiring surgical replacement. Unfortunately, multiple attempts have failed to find an efficient conservative treatment of CAVD; however, therapeutic regimens and clinical settings have also been far from the optimal. In this review, we focused on interactions and transitions between aforementioned mechanisms demarcating ascending stages of CAVD, suggesting a predisposing condition (bicuspid aortic valve) and drug combination (lipid-lowering drugs combined with angiotensin II antagonists and cytokine inhibitors) for the further testing in both preclinical and clinical trials.
Collapse
|
55
|
Ait-Aissa K, Heisner JS, Norwood Toro LE, Bruemmer D, Doyon G, Harmann L, Geurts A, Camara AKS, Beyer AM. Telomerase Deficiency Predisposes to Heart Failure and Ischemia-Reperfusion Injury. Front Cardiovasc Med 2019; 6:31. [PMID: 31001540 PMCID: PMC6454001 DOI: 10.3389/fcvm.2019.00031] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/07/2019] [Indexed: 12/13/2022] Open
Abstract
Introduction: Elevated levels of mitochondrial reactive oxygen species (ROS) contribute to the development of numerous cardiovascular diseases. TERT, the catalytic subunit of telomerase, has been shown to translocate to mitochondria to suppress ROS while promoting ATP production. Acute overexpression of TERT increases survival and decreases infarct size in a mouse model of myocardial infarct, while decreased telomerase activity predisposes to mitochondrial defects and heart failure. In the present study, we examined the role of TERT on cardiac structure and function under basal conditions and conditions of acute or prolonged stress in a novel rat model of TERT deficiency. Methods: Cardiac structure and function were evaluated via transthoracic echocardiogram. Langendorff preparations were used to test the effects of acute global ischemia reperfusion injury on cardiac function and infarction. Coronary flow and left ventricular pressure were measured during and after ischemia/reperfusion (I/R). Mitochondrial DNA integrity was measured by PCR and mitochondrial respiration was assessed in isolated mitochondria using an Oxygraph. Angiotensin II infusion was used as an established model of systemic stress. Results: No structural changes (echocardiogram) or coronary flow/left ventricle pressure (isolated hearts) were observed in TERT-/- rats at baseline; however, after I/R, coronary flow was significantly reduced in TERT-/- compared to wild type (WT) rats, while diastolic Left Ventricle Pressure was significantly elevated (n = 6 in each group; p < 0.05) in the TERT-/-. Interestingly, infarct size was less in TERT-/- rats compared to WT rats, while mitochondrial respiratory control index decreased and mitochondrial DNA lesions increased in TERT-/- compared to WT. Angiotensin II treatment did not alter cardiac structure or function; however, it augmented the infarct size significantly more in TERT-/- compared to the WT. Conclusion: Absence of TERT activity increases susceptibility to stress like cardiac injury. These results suggest a critical role of telomerase in chronic heart disease.
Collapse
Affiliation(s)
- Karima Ait-Aissa
- Cardiovascular Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - James S. Heisner
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Laura E. Norwood Toro
- Cardiovascular Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Dennis Bruemmer
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Genevieve Doyon
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Leanne Harmann
- Cardiovascular Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Aron Geurts
- Cardiovascular Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Amadou K. S. Camara
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Andreas M. Beyer
- Cardiovascular Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
56
|
Jönsson S, Becirovic-Agic M, Isackson H, Tveitarås MK, Skogstrand T, Narfström F, Karlsen TV, Lidén Å, Leh S, Ericsson M, Nilsson SK, Reed RK, Hultström M. Angiotensin II and salt-induced decompensation in Balb/CJ mice is aggravated by fluid retention related to low oxidative stress. Am J Physiol Renal Physiol 2019; 316:F914-F933. [PMID: 30785350 DOI: 10.1152/ajprenal.00483.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Balb/CJ mice are more sensitive to treatment with angiotensin II (ANG II) and high-salt diet compared with C57BL/6J mice. Together with higher mortality, they develop edema, signs of heart failure, and acute kidney injury. The aim of the present study was to identify differences in renal gene regulation that may affect kidney function and fluid balance, which could contribute to decompensation in Balb/CJ mice after ANG II + salt treatment. Male Balb/CJ and C57BL/6J mice were divided into the following five different treatment groups: control, ANG II, salt, ANG II + salt, and ANG II + salt + N-acetylcysteine. Gene expression microarrays were used to explore differential gene expression after treatment and between the strains. Published data from the Mouse Genome Database were used to identify the associated genomic differences. The glomerular filtration rate (GFR) was measured using inulin clearance, and fluid balance was measured using metabolic cages. Gene ontology enrichment analysis of gene expression microarrays identified glutathione transferase (antioxidant system) as highly enriched among differentially expressed genes. Balb/CJ mice had similar GFR compared with C57BL/6J mice but excreted less Na+ and water, although net fluid and electrolyte balance did not differ, suggesting that Balb/CJ mice may be inherently more prone to decompensation. Interestingly, C57BL/6J mice had higher urinary oxidative stress despite their relative protection from decompensation. In addition, treatment with the antioxidant N-acetylcysteine decreased oxidative stress in C57BL/6J mice, reduced urine excretion, and increased mortality. Balb/CJ mice are more sensitive than C57BL/6J to ANG II + salt, in part mediated by lower oxidative stress, which favors fluid and Na+ retention.
Collapse
Affiliation(s)
- Sofia Jönsson
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Mediha Becirovic-Agic
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Henrik Isackson
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Cardiology, Uppsala University , Uppsala , Sweden
| | | | | | - Fredrik Narfström
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Tine V Karlsen
- Department of Biomedicine, University of Bergen , Norway
| | - Åsa Lidén
- Department of Biomedicine, University of Bergen , Norway
| | - Sabine Leh
- Department of Pathology, Haukeland University Hospital , Bergen , Norway.,Department of Clinical Medicine, University of Bergen , Norway
| | | | - Stefan K Nilsson
- Department of Medical Biosciences, Umeå University , Umeå , Sweden
| | - Rolf K Reed
- Department of Biomedicine, University of Bergen , Norway.,Centre for Cancer Biomarkers (CCBIO), University of Bergen , Norway
| | - Michael Hultström
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Department of Biomedicine, University of Bergen , Norway.,Anesthesia and Intensive Care, Department of Surgical Sciences, Uppsala University , Uppsala , Sweden
| |
Collapse
|
57
|
Inhibitory Effects of Roseoside and Icariside E4 Isolated from a Natural Product Mixture (No-ap) on the Expression of Angiotensin II Receptor 1 and Oxidative Stress in Angiotensin II-Stimulated H9C2 Cells. Molecules 2019; 24:molecules24030414. [PMID: 30678135 PMCID: PMC6384670 DOI: 10.3390/molecules24030414] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/12/2019] [Accepted: 01/22/2019] [Indexed: 12/13/2022] Open
Abstract
Hypertension is a major risk factor for the development of cardiovascular diseases. This study aimed to elucidate whether the natural product mixture No-ap (NA) containing Pine densiflora, Annona muricate, and Monordica charantia, or its single components have inhibitory effects on hypertension-related molecules in Angiotensin II (Ang II)-stimulated H9C2 cells. Individual functional components were isolated and purified from NA using various columns and solvents, and then their structures were analyzed using ESI–MS, 1H-NMR, and 13H-NMR spectra. H9C2 cells were stimulated with 300 nM Ang II for 7 h. NA, telmisartan, ginsenoside, roseoside (Roseo), icariside E4 (IE4), or a combination of two components (Roseo and IE4) were administered to the cells 1 h before Ang II stimulation. The expression and activity of hypertension-related molecules or oxidative molecules were determined using RT-PCR, western blot, and ELISA. Ang II stimulation increased the expression of Ang II receptor 1 (AT1), tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), tumor growth factor-β (TGF-β) mRNA, and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity and the levels of hydrogen peroxide (H2O2) and superoxide anion (•O2−) and reduced anti-oxidant enzyme activity. NA significantly improved the expression or activities of all hypertension-related molecules altered in Ang II-stimulated cells. Roseo or IE4 pretreatment either decreased or increased the expression or activities of all hypertension-related molecules similar to NA, but to a lesser extent. The pretreatment with a combination of Roseo and IE4 (1:1) either decreased or increased the expression of all hypertension-related molecules, compared to each single component, revealing a synergistic action of the two compounds. Thus, the combination of single components could exert promising anti-hypertensive effects similar to NA, which should be examined in future animal and clinical studies.
Collapse
|
58
|
Manuneedhi Cholan P, Cartland SP, Dang L, Rayner BS, Patel S, Thomas SR, Kavurma MM. TRAIL protects against endothelial dysfunction in vivo and inhibits angiotensin-II-induced oxidative stress in vascular endothelial cells in vitro. Free Radic Biol Med 2018; 126:341-349. [PMID: 30165101 DOI: 10.1016/j.freeradbiomed.2018.08.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/16/2018] [Accepted: 08/24/2018] [Indexed: 12/22/2022]
Abstract
The vascular endothelium is critical for maintenance of cardiovascular homeostasis. Endothelial dysfunction is a key event of atherosclerosis, with oxidative stress mediated by reactive oxygen species (ROS) playing a major role. Tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is increasingly recognised to play a protective role in atherosclerosis, however the molecular mechanisms by which it exerts its beneficial effects are unclear. Here we examined if TRAIL could attenuate vascular oxidative stress and improve endothelial cell (EC) function. In coronary artery disease patients, plasma TRAIL levels were significantly reduced compared to healthy individuals, and negatively correlated with the levels of circulating 8-iso Prostaglandin F2α, a marker of in vivo oxidative stress. In vivo, high-fat fed, atherosclerotic Trail-/-Apoe-/- mice exhibited a significant impairment in endothelial-dependent vasorelaxation, which correlated with increased vascular ROS and 4-hydroxynonenal compared to Apoe-/- mice. Endothelial permeability measured by Evan's blue dye extravasation was increased in several organs of Trail-/- mice compared to wild-type mice, which correlated with a decrease in VE-cadherin expression. In vitro in ECs, angiotensin II (AngII)-induced ROS generation involving the mitochondria, NADPH oxidase-4 (NOX-4) and eNOS, was inhibited by pre-treatment with TRAIL. Furthermore, AngII-augmented VCAM-1 expression and monocyte adhesion to ECs was inhibited by TRAIL. Finally, AngII reduced VE-cadherin expression and redistributed this protein, all of which was brought back to baseline by TRAIL pre-treatment. These findings demonstrate for the first time that TRAIL protects against several forms of endothelial dysfunction involving its ability to control EC ROS generation. Understanding the role TRAIL plays in normal physiology and disease, may lead to potential new therapies to improve endothelial function and atherosclerosis.
Collapse
Affiliation(s)
- Pradeep Manuneedhi Cholan
- Heart Research Institute, Sydney 2042, Australia; Sydney Medical School, The University of Sydney, Sydney 2006, Australia
| | - Siân P Cartland
- Heart Research Institute, Sydney 2042, Australia; Sydney Medical School, The University of Sydney, Sydney 2006, Australia
| | - Lei Dang
- School of Medical Sciences, University of New South Wales, Sydney 2052, Australia
| | - Benjamin S Rayner
- Heart Research Institute, Sydney 2042, Australia; Sydney Medical School, The University of Sydney, Sydney 2006, Australia
| | - Sanjay Patel
- Sydney Medical School, The University of Sydney, Sydney 2006, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Shane R Thomas
- School of Medical Sciences, University of New South Wales, Sydney 2052, Australia
| | - Mary M Kavurma
- Heart Research Institute, Sydney 2042, Australia; Sydney Medical School, The University of Sydney, Sydney 2006, Australia.
| |
Collapse
|
59
|
Wang X, Yang C, Liu X, Yang P. Ghrelin Alleviates Angiotensin II-Induced H9c2 Apoptosis: Impact of the miR-208 Family. Med Sci Monit 2018; 24:6707-6716. [PMID: 30244257 PMCID: PMC6178878 DOI: 10.12659/msm.908096] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Ghrelin is a novel peptide with abundant cardioprotective effects. The miR-208 family, consisting of cardiac-specifically expressed microRNAs, are not only involved in hypertrophy and fibrosis, but are also closely related with myocyte apoptosis. This study explored the role of the miR-208 family in the protective effect of ghrelin on angiotensin II (Ang II)-induced apoptosis. MATERIAL AND METHODS H9c2 cells were exposed to Ang II with or without ghrelin. Cell viability was detected by MTT assay and the percentage of apoptotic cells was confirmed by flow cytometry. miRNAs expression levels were measured by qRT-PCR. Then, cells transfected with miR-208 negative control, mimics, and inhibitors were treated with Ang II and ghrelin, followed by flow cytometry. PCR array was performed to explore the pathways affected by miR-208. RESULTS The miR-208 level was reduced in Ang II-treated H9c2 cells, accompanied with increased cell apoptosis, which were both reversed by ghrelin administration. Flow cytometry revealed that miR-208 inhibitors clearly upregulated the apoptotic percentage, whereas miR-208 mimics showed the opposite effects in the Ang II group. miR-208a further alleviated apoptosis when treated with ghrelin. miR-208 mainly affected caspase, inflammatory-related genes, and several signaling pathways. CONCLUSIONS We provide new evidence that the miR-208 family is regulated by Ang II and ghrelin. Overexpression of miR-208 family alleviated Ang II-induced cell apoptosis and miR-208a assisted in the protective effect of ghrelin. Several apoptosis pathways affected by miR-208 family were found. These findings suggest the pathogenesis of cardiomyocyte apoptosis and the protective mechanism of ghrelin.
Collapse
Affiliation(s)
- Xiaotong Wang
- Department of Cardiololgy, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China (mainland)
| | - Chunyan Yang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China (mainland)
| | - Xueyan Liu
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China (mainland)
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
60
|
Wang X, Yang C, Liu X, Yang P. The impact of microRNA-122 and its target gene Sestrin-2 on the protective effect of ghrelin in angiotensin II-induced cardiomyocyte apoptosis. RSC Adv 2018; 8:10107-10114. [PMID: 35540851 PMCID: PMC9078835 DOI: 10.1039/c7ra13028g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/06/2018] [Indexed: 12/15/2022] Open
Abstract
Ghrelin with n-octanoylated serine 3 residue is a peptide hormone with well-known cardioprotective properties. MicroRNA-122 is associated with the pathogenesis of many cardiovascular diseases, including apoptosis and was found highly increased in our previous rat model of post-myocardial infarction heart failure. In this study, we aimed to identify the target gene of microRNA-122 and to evaluate their impacts on the protective effect of acylated ghrelin in angiotensin II-induced apoptosis. The results showed that microRNA-122 was upregulated in the angiotensin II administration group accompanied by increased cell apoptosis, which were both reversed by ghrelin. Furthermore, microRNA-122 mimics upregulated numerous pro-apoptotic genes and increased apoptosis. The luciferase activity assay revealed Sestrin-2 as a direct target of microRNA-122. The expression of Sestrin-2 was downregulated by angiotensin II and upregulated by co-treatment with ghrelin. Inhibition of microRNA-122 and overexpression of Sestrin-2 alleviated apoptosis which was further reduced upon administered of ghrelin. Together, these results indicated that Sestrin-2 expression is inhibited by microRNA-122 and that this inhibition is involved in the protective effect of ghrelin and angiotensin II-induced apoptosis. We also found that microRNA-122 influenced several apoptosis pathways including the caspase cascade reaction and death receptor-mediated pathways. Collectively, our data reveal that microRNA-122 and its target gene Sestrin-2, under the regulation of angiotensin II and ghrelin, are important players in cardiomyocyte apoptosis. We therefore believe that microRNA-122 and Sestrin-2 can be developed as potential therapeutic targets against apoptosis in cardiovascular diseases. Inhibition of microRNA-122 and overexpression of Sestrin-2 alleviated angiotensin II-induced cardiomyocyte apoptosis and enhanced the protective effect of ghrelin.![]()
Collapse
Affiliation(s)
- Xiaotong Wang
- The Department of Cardiology, China-Japan Union Hospital, Jilin University Changchun 130011 Jilin China
| | - Chunyan Yang
- The Department of Cardiology, China-Japan Union Hospital, Jilin University Changchun 130011 Jilin China
| | - Xueyan Liu
- The Department of Cardiology, China-Japan Union Hospital, Jilin University Changchun 130011 Jilin China
| | - Ping Yang
- The Department of Cardiology, China-Japan Union Hospital, Jilin University Changchun 130011 Jilin China
| |
Collapse
|
61
|
Zhou H, Huang J, Zhu L, Cao Y. Erythropoietin alleviates post-resuscitation myocardial dysfunction in rats potentially through increasing the expression of angiotensin II receptor type 2 in myocardial tissues. Mol Med Rep 2018; 17:5184-5192. [PMID: 29393490 PMCID: PMC5865983 DOI: 10.3892/mmr.2018.8473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023] Open
Abstract
Activation of renin-angiotensin system (RAS) is one of the pathological mechanisms associated with myocardial ischemia-reperfusion injury following resuscitation. The present study aimed to determine whether erythropoietin (EPO) improves post‑resuscitation myocardial dysfunction and how it affects the renin‑angiotensin system. Sprague‑Dawley rats were randomly divided into sham, vehicle, epinephrine (EP), EPO and EP + EPO groups. Excluding the sham group, all groups underwent cardiopulmonary resuscitation (CPR) 4 min after asphyxia‑induced cardiac arrest (CA). EP and/or EPO was administrated by intravenous injection when CPR began. The results demonstrated that the vehicle group exhibited lower mean arterial pressure, left ventricular systolic pressure, maximal ascending rate of left ventricular pressure during left ventricular isovolumic contraction and maximal descending rate of left ventricular pressure during left ventricular isovolumic relaxation (+LVdP/dt max and ‑LVdP/dt max, respectively), and higher left ventricular end‑diastolic pressure, compared with the sham group following return of spontaneous circulation (ROSC). Few significant differences were observed concerning the myocardial function between the vehicle and EP groups; however, compared with the vehicle group, EPO reversed myocardial function indices following ROSC, excluding‑LVdP/dt max. Serum renin and angiotensin (Ang) II levels were measured by ELISA. The serum levels of renin and Ang II were significantly increased in the vehicle group compared with the sham group, which was also observed for the myocardial expression of renin and Ang II receptor type 1 (AT1R), as determined by reverse transcription‑quantitative polymerase chain reaction and western blotting. EPO alone did not significantly reduce the high serum levels of renin and Ang II post-resuscitation, but changed the protein levels of renin and AT1R expression in myocardial tissues. However, EPO enhanced the myocardial expression of Ang II receptor type 2 (AT2R) following ROSC. In conclusion, the present study confirmed that CA resuscitation activated the renin‑Ang II‑AT1R signaling pathway, which may contribute to myocardial dysfunction in rats. The present study confirmed that EPO treatment is beneficial for protecting cardiac function post‑resuscitation, and the roles of EPO in alleviating post‑resuscitation myocardial dysfunction may potentially be associated with enhanced myocardial expression of AT2R.
Collapse
Affiliation(s)
- Hourong Zhou
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jia Huang
- Emergency Department, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Li Zhu
- Institute of Anesthesia, Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Yu Cao
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
62
|
Liu W, Gong W, He M, Liu Y, Yang Y, Wang M, Wu M, Guo S, Yu Y, Wang X, Sun F, Li Y, Zhou L, Qin S, Zhang Z. Spironolactone Protects against Diabetic Cardiomyopathy in Streptozotocin-Induced Diabetic Rats. J Diabetes Res 2018; 2018:9232065. [PMID: 30406151 PMCID: PMC6204188 DOI: 10.1155/2018/9232065] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/16/2018] [Accepted: 08/02/2018] [Indexed: 12/16/2022] Open
Abstract
Spironolactone (SPR) has been shown to protect diabetic cardiomyopathy (DCM), but the specific mechanisms are not fully understood. Here, we determined the cardioprotective role of SPR in diabetic mice and further explored the potential mechanisms in both in vivo and in vitro models. Streptozotocin- (STZ-) induced diabetic rats were used as the in vivo model. After the onset of diabetes, rats were treated with either SPR (STZ + SPR) or saline (STZ + NS) for 12 weeks; nondiabetic rats were used as controls (NDCs). In vitro, H9C2 cells were exposed to aldosterone, with or without SPR. Cardiac structure was investigated with transmission electron microscopy and pathological examination; immunohistochemistry was performed to detect nitrotyrosine, collagen-1, TGF-β1, TNF-α, and F4/80 expression; and gene expression of markers for oxidative stress, inflammation, fibrosis, and energy metabolism was detected. Our results suggested that SPR attenuated mitochondrial morphological abnormalities and sarcoplasmic reticulum enlargement in diabetic rats. Compared to the STZ + NS group, cardiac oxidative stress, fibrosis, inflammation, and mitochondrial dysfunction were improved by SPR treatment. Our study showed that SPR had cardioprotective effects in diabetic rats by ameliorating mitochondrial dysfunction and reducing fibrosis, oxidative stress, and inflammation. This study, for the first time, indicates that SPR might be a potential treatment for DCM.
Collapse
Affiliation(s)
- Wenjuan Liu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Wei Gong
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Min He
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Yemei Liu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Department of Endocrinology, The Second People's Hospital, 4 Duchun Road, Wuhu, Anhui 241001, China
| | - Yeping Yang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Meng Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Meng Wu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, 1055 Sanxiang Rd, Suzhou, Jiangsu 215000, China
| | - Shizhe Guo
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Yifei Yu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Xuanchun Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Fei Sun
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Yiming Li
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Linuo Zhou
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| | - Shengmei Qin
- Department of Cardiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Zhaoyun Zhang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, 12 Wulumuqi Road, Shanghai 200040, China
| |
Collapse
|
63
|
Ramadan A, Singh KK, Quan A, Plant PJ, Al-Omran M, Teoh H, Verma S. Loss of vascular smooth muscle cell autophagy exacerbates angiotensin II-associated aortic remodeling. J Vasc Surg 2017; 68:859-871. [PMID: 29273297 DOI: 10.1016/j.jvs.2017.08.086] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 08/25/2017] [Indexed: 11/18/2022]
Abstract
OBJECTIVE The pathophysiologic processes of abdominal aortic aneurysms (AAAs) and atherosclerosis often intersect. Given that anomalies in vascular smooth muscle cell (SMC) autophagy have been noted in models of atherosclerosis, we sought to evaluate the potential role that SMC autophagy may play in the initiation and progression of AAAs. METHODS Studies were conducted in ATG7flx/flxSM22α-Cretg/+ (SMC ATG7 knockout [SMC-ATG7-KO]) and ATG7WT/WT; SM22α-Cretg/+ (SMC ATG7 wild-type [SMC-ATG7-WT]) littermates that were continuously infused with angiotensin II (Ang II; 1.5 mg/kg/d) for up to 12 weeks. Mortality, morbidity, hemodynamics, and aortic remodeling were documented. RESULTS During the 12-week observation window, all of the Ang II-treated SMC-ATG-WT mice (n = 6) survived, whereas 10 of the 19 Ang II-treated SMC-ATG-KO mice had died by week 7 (log-rank test, P < .001). Mean arterial pressure (128.07 ± 3.4 mm Hg for Ang II-treated SMC-ATG-KO vs 138.5 ± 5.87 mm Hg for Ang II-treated SMC-ATG-WT mice) and diastolic arterial pressure (109.7 ± 2.55 mm Hg for Ang II-treated SMC-ATG7-KO vs 119.4 ± 2.12 mm Hg for Ang II-treated SMC-ATG7-WT mice) were significantly different between the two groups (P < .01). Cardiac rupture, myocardial infarct, end-organ damage, pleural effusion, and venous distention were noted in Ang II-treated SMC-ATG7-KO but not in Ang II-treated SMC-ATG7-WT mice. Although the suprarenal aortic diameters of the Ang II-treated SMC-ATG7-KO group demonstrated a trending increase (at week 4, 1.26 ± 0.06 mm [n = 14] for Ang II-treated SMC-ATG-KO mice vs 1.09 ± 0.02 mm [n = 5] for Ang II-treated SMC-ATG-WT mice; P < .05), only 2 of the 19 developed abdominal aortic dissections. CONCLUSIONS Mice with SMC ATG7 deficiency that are chronically infused with Ang II do not tend to develop dissecting AAA but do exhibit adverse aortic remodeling and appreciable cardiac failure-associated mortality.
Collapse
Affiliation(s)
- Azza Ramadan
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Krishna K Singh
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Division of Vascular Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Quan
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Pamela J Plant
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Mohammed Al-Omran
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Division of Vascular Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Surgery, King Saud University, Riyadh, Kingdom of Saudi Arabia; King Saud University-Li Ka Shing Collaborative Research Program, Riyadh, Kingdom of Saudi Arabia
| | - Hwee Teoh
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Division of Endocrinology and Metabolism, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Subodh Verma
- Division of Cardiac Surgery, St. Michael's Hospital, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
64
|
Agostini S, Lionetti V. New insights into the non-hemostatic role of von Willebrand factor in endothelial protection. Can J Physiol Pharmacol 2017; 95:1183-1189. [PMID: 28715643 DOI: 10.1139/cjpp-2017-0126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During exposure to ischemia-reperfusion (I/R) insult, angiotensin II (AngII)-induced endothelin-1 (ET-1) upregulation in endothelial cells progressively impairs nitric oxide (NO) bioavailability while increasing levels of superoxide anion (O2-) and leading to the onset of endothelial dysfunction. Moreover, the overexpression of ET-1 increases the endothelial and circulating levels of von Willebrand factor (vWF), a glycoprotein with a crucial role in arterial thrombus formation. Nowadays, the non-hemostatic role of endothelial vWF is emerging, although we do not yet know whether its increased expression is cause or consequence of endothelial dysfunction. Notably, the vWF blockade or depletion leads to endothelial protection in cultured cells, animal models of vascular injury, and patients as well. Despite the recent efforts to develop an effective pharmacological strategy, the onset of endothelial dysfunction is still difficult to prevent and remains closely related to adverse clinical outcome. Unraveling the non-hemostatic role of endothelial vWF in the onset of endothelial dysfunction could provide new avenues for protection against vascular injury mediated by AngII.
Collapse
Affiliation(s)
- Silvia Agostini
- a Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Vincenzo Lionetti
- a Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,b UOS Anesthesiology, Fondazione Toscana "G. Monasterio", Pisa, Italy
| |
Collapse
|
65
|
Kim J, Kim J, Kook H, Park WJ. PICOT alleviates myocardial ischemia-reperfusion injury by reducing intracellular levels of reactive oxygen species. Biochem Biophys Res Commun 2017; 485:807-813. [PMID: 28257842 DOI: 10.1016/j.bbrc.2017.02.136] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 12/31/2022]
Abstract
Excessive generation of reactive oxygen species (ROS) is one of the main causes of myocardial ischemia-reperfusion (I/R) injury. In this study, we investigated the role of protein kinase C-interacting cousin of thioredoxin (PICOT; Grx3) during myocardial I/R using PICOT transgenic (TG) and knockdown (KD) mice. Infarction and apoptosis were attenuated in PICOT TG mice but exacerbated in PICOT KD mice upon I/R. In parallel, I/R-induced generation of ROS was attenuated in PICOT TG mice but exacerbated in PICOT KD mice. Angiotensin II (AngII)-mediated increases in ROS and free iron levels were also attenuated in cardiomyocytes isolated from PICOT TG mice but exacerbated in cardiomyocytes from PICOT KD mice. Accordingly, H2O2-mediated cell death was attenuated in cardiomyocytes isolated from PICOT TG mice but exacerbated in cardiomyocytes from PICOT KD mice. Taken together, these data show that PICOT alleviates myocardial I/R injury by regulating intracellular ROS and free iron levels. We suggest that PICOT presents a novel therapeutic strategy for myocardial I/R injury.
Collapse
Affiliation(s)
- Jihwa Kim
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Jooyeon Kim
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology and Medical Research Center for Gene Regulation, Chonnam National University Medical School, 160 Baekseo-ro, Dong-ku, Gwangju 61469, Republic of Korea
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea.
| |
Collapse
|
66
|
Zhou L, Ma B, Han X. The role of autophagy in angiotensin II-induced pathological cardiac hypertrophy. J Mol Endocrinol 2016; 57:R143-R152. [PMID: 27620875 DOI: 10.1530/jme-16-0086] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 09/12/2016] [Indexed: 12/18/2022]
Abstract
Pathological cardiac hypertrophy is associated with nearly all forms of heart failure. It develops in response to disorders such as coronary artery disease, hypertension and myocardial infarction. Angiotensin II (Ang II) has direct effects on the myocardium and promotes hypertension. Chronic elevation of Ang II can lead to pathological cardiac hypertrophy and cardiac failure. Autophagy is an important process in the pathogenesis of cardiovascular diseases. Under physiological conditions, autophagy is an essential homeostatic mechanism to maintain the global cardiac structure function by ridding damaged cells or unwanted macromolecules and organelles. Dysregulation of autophagy may play an important role in Ang II-induced cardiac hypertrophy although conflicting reports on the effects of Ang II on autophagy and cardiac hypertrophy exist. Some studies showed that autophagy activation attenuated Ang II-induced cardiac dysfunction. Others suggested that inhibition of the Ang II induced autophagy should be protective. The discrepancies may be due to different model systems and different signaling pathway involved. Ang II-induced cardiac hypertrophy may be alleviated through regulation of autophagy. This review focuses on Ang II to highlight the molecular targets and pathways identified in the prevention and treatment of Ang II-induced pathological cardiac hypertrophy by regulating autophagy.
Collapse
Affiliation(s)
- Lichun Zhou
- Department of PharmacologySchool of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province, China
| | - Baohua Ma
- Pharmaceutical Preparation SectionCentral Hospital of Qingdao, Qingdao, Shandong Province, China
| | - Xiuzhen Han
- Department of PharmacologySchool of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
67
|
Haque ZK, Wang DZ. How cardiomyocytes sense pathophysiological stresses for cardiac remodeling. Cell Mol Life Sci 2016; 74:983-1000. [PMID: 27714411 DOI: 10.1007/s00018-016-2373-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/01/2016] [Accepted: 09/19/2016] [Indexed: 12/14/2022]
Abstract
In the past decades, the cardiovascular community has laid out the fundamental signaling cascades that become awry in the cardiomyocyte during the process of pathologic cardiac remodeling. These pathways are initiated at the cell membrane and work their way to the nucleus to mediate gene expression. Complexity is multiplied as the cardiomyocyte is subjected to cross talk with other cells as well as a barrage of extracellular stimuli and mechanical stresses. In this review, we summarize the signaling cascades that play key roles in cardiac function and then we proceed to describe emerging concepts of how the cardiomyocyte senses the mechanical and environmental stimuli to transition to the deleterious genetic program that defines pathologic cardiac remodeling. As a highlighting example of these processes, we illustrate the transition from a compensated hypertrophied myocardium to a decompensated failing myocardium, which is clinically manifested as decompensated heart failure.
Collapse
Affiliation(s)
- Zaffar K Haque
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 1260 John F. Enders Research Bldg, 320 Longwood Ave, Boston, MA, 02115, USA.
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 1260 John F. Enders Research Bldg, 320 Longwood Ave, Boston, MA, 02115, USA
| |
Collapse
|
68
|
Ramalingam L, Menikdiwela K, LeMieux M, Dufour JM, Kaur G, Kalupahana N, Moustaid-Moussa N. The renin angiotensin system, oxidative stress and mitochondrial function in obesity and insulin resistance. Biochim Biophys Acta Mol Basis Dis 2016; 1863:1106-1114. [PMID: 27497523 DOI: 10.1016/j.bbadis.2016.07.019] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 07/25/2016] [Indexed: 12/13/2022]
Abstract
Obesity is a complex disease characterized by excessive expansion of adipose tissue and is an important risk factor for chronic diseases such as cardiovascular disorders, hypertension and type 2 diabetes. Moreover, obesity is a major contributor to inflammation and oxidative stress, all of which are key underlying causes for diabetes and insulin resistance. Specifically, adipose tissue secretes bioactives molecules such as inflammatory hormone angiotensin II, generated in the Renin Angiotensin System (RAS) from its precursor angiotensinogen. Accumulated evidence suggests that RAS may serve as a strong link between obesity and insulin resistance. Dysregulation of RAS also occurs in several other tissues including those involved in regulation of glucose and whole body homeostasis as well as insulin sensitivity such as muscle, liver and pancreas and heart. Here we review the scientific evidence for these interactions and potential roles for oxidative stress, inflammation and mitochondrial dysfunction in these target tissues which may mediate effects of RAS in metabolic diseases. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.
Collapse
Affiliation(s)
- Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States
| | - Kalhara Menikdiwela
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States
| | - Monique LeMieux
- Department of Nutrition and Food Sciences, Texas Women's University, Denton, TX, United States
| | - Jannette M Dufour
- Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States; Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Gurvinder Kaur
- Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States; Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Nishan Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States; Department of Physiology, Faculty of Medicine, University of Peradeniya, Sri Lanka
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States; Obesity Research Cluster, Texas Tech University, Lubbock, TX, United States.
| |
Collapse
|
69
|
Sakr HF, Abbas AM, Elsamanoudy AZ. Effect of valsartan on cardiac senescence and apoptosis in a rat model of cardiotoxicity. Can J Physiol Pharmacol 2016; 94:588-98. [DOI: 10.1139/cjpp-2015-0461] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The clinical application of doxorubicin is limited by its cardiotoxicity. The present study investigated the effect of valsartan on doxorubicin-induced cardiotoxicity in rats. Rats were divided into 6 groups: control, control + valsartan (10 mg/kg, for 14 days, orally), doxorubicin-treated (2.5 mg/kg, 3 times/week for 2 weeks, intraperitoneally), valsartan then doxorubicin, valsartan + doxorubicin, and doxorubicin then valsartan. ECG, isolated heart, lipid peroxidation (thiobaribituric acid reactive substances (TBARS)), total antioxidant capacity (TAC), and Bax, Bcl-2, and senescence marker protein 30 (SMP30) gene expression were measured in cardiac tissue. Blood samples were collected to measure lactate dehydrogenase (LDH) and creatine kinase MB (CK-MB). Doxorubicin significantly increased LDH, CK-MB, TBARS, heart rate (HR), Bax gene expression, and −dP/dtmax and decreased TAC, Bcl-2 and SMP30 gene expression, left ventricular developed pressure (LVDP), and +dP/dtmax. Also, doxorubicin lengthened ST, QT, and QTc intervals. Concurrent or post- but not pre-treatment of doxorubicin-treated rats with valsartan reduced LDH, CK-MB, TBARS, HR, Bax gene expression, −dP/dtmax, and ST, QT, and QTc intervals and increased TAC, Bcl-2 and SMP30 gene expression, LVDP, and +dP/dtmax. Therefore, we conclude that concurrent or post- but not pre-treatment of doxorubicin-induced rats with valsartan attenuated doxorubicin-induced cardiotoxicity through inhibiting oxidative stress, apoptosis, and senescence.
Collapse
Affiliation(s)
- Hussein F. Sakr
- Medical Physiology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amr M. Abbas
- Medical Physiology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ayman Z. Elsamanoudy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
70
|
Chuang KJ, Lee KY, Pan CH, Lai CH, Lin LY, Ho SC, Ho KF, Chuang HC. Effects of zinc oxide nanoparticles on human coronary artery endothelial cells. Food Chem Toxicol 2016; 93:138-44. [PMID: 27185063 DOI: 10.1016/j.fct.2016.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 12/11/2022]
Abstract
Inhalation of zinc oxide (ZnO) metal fumes is known to cause metal fume fever and to have systemic effects; however, the effects of ZnO nanoparticles (ZnONPs) on the cardiovascular system remain unclear. The objective of this study was to investigate the cardiovascular toxicity of ZnONPs. Human coronary artery endothelial cells (HCAECs) were exposed to ZnONPs of different sizes to investigate the cell viability, 8-hydroxy-2'-deoxyguanosine (8-OHdG), interleukin (IL)-6, nitric oxide (NO), and regulation of cardiovascular disease-related genes. Exposure of HCAECs to ZnONPs resulted in decreased cell viability and increased levels of 8-OHdG, IL-6, and NO. Downregulation of cardiovascular-associated genes was observed in response to ZnONPs in HCAECs determined by qPCR, suggesting that the calcium signaling pathway, neuroactive ligand-receptor interaction, hypertrophic cardiomyopathy, dilated cardiomyopathy, and renin-angiotensin system are important affected pathways in response to ZnONPs. Furthermore, we observed a significant response of AGTR1 to ZnONP exposure in HCAECs. Our results suggest that ZnONPs cause toxicity to HCAECs, which could be associated with cardiovascular dysfunction.
Collapse
Affiliation(s)
- Kai-Jen Chuang
- School of Public Health, College of Public Health and Nutrition, Taipei Medical University, Taipei, Taiwan; Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Chih-Hong Pan
- Institute of Labor, Occupational Safety and Health, Ministry of Labor, New Taipei City, Taiwan; School of Public Health, National Defense Medical Center, Taipei, Taiwan.
| | - Ching-Huang Lai
- School of Public Health, National Defense Medical Center, Taipei, Taiwan.
| | - Lian-Yu Lin
- Department of Internal Medicine, Division of Cardiology, National Taiwan University Hospital, Taipei, Taiwan.
| | - Shu-Chuan Ho
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Kin-Fai Ho
- Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Shenzhen Municipal Key Laboratory for Health Risk Analysis, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Hsiao-Chi Chuang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
71
|
Peng K, Tian X, Qian Y, Skibba M, Zou C, Liu Z, Wang J, Xu Z, Li X, Liang G. Novel EGFR inhibitors attenuate cardiac hypertrophy induced by angiotensin II. J Cell Mol Med 2016; 20:482-94. [PMID: 26762600 PMCID: PMC4759478 DOI: 10.1111/jcmm.12763] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 11/26/2015] [Indexed: 12/28/2022] Open
Abstract
Cardiac hypertrophy is an important risk factor for heart failure. Epidermal growth factor receptor (EGFR) has been found to play a role in the pathogenesis of various cardiovascular diseases. The aim of this current study was to examine the role of EGFR in angiotensin II (Ang II)-induced cardiac hypertrophy and identify the underlying molecular mechanisms. In this study, we observed that both Ang II and EGF could increase the phospohorylation of EGFR and protein kinase B (AKT)/extracellular signal-regulated kinase (ERK), and then induce cell hypertrophy in H9c2 cells. Both pharmacological inhibitors and genetic silencing significantly reduced Ang II-induced EGFR signalling pathway activation, hypertrophic marker overexpression, and cell hypertrophy. In addition, our results showed that Ang II-induced EGFR activation is mediated by c-Src phosphorylation. In vivo, Ang II treatment significantly led to cardiac remodelling including cardiac hypertrophy, disorganization and fibrosis, accompanied by the activation of EGFR signalling pathway in the heart tissues, while all these molecular and pathological alterations were attenuated by the oral administration with EGFR inhibitors. In conclusion, the c-Src-dependent EGFR activation may play an important role in Ang II-induced cardiac hypertrophy, and inhibition of EGFR by specific molecules may be an effective strategy for the treatment of Ang II-associated cardiac diseases.
Collapse
Affiliation(s)
- Kesong Peng
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinqiao Tian
- Department of Ultrasonography, The 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuanyuan Qian
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Melissa Skibba
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunpeng Zou
- Department of Ultrasonography, The 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiguo Liu
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingying Wang
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zheng Xu
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaokun Li
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
72
|
Hamilton DJ, Zhang A, Li S, Cao TN, Smith JA, Vedula I, Cordero-Reyes AM, Youker KA, Torre-Amione G, Gupte AA. Combination of angiotensin II and l-NG-nitroarginine methyl ester exacerbates mitochondrial dysfunction and oxidative stress to cause heart failure. Am J Physiol Heart Circ Physiol 2016; 310:H667-80. [PMID: 26747502 DOI: 10.1152/ajpheart.00746.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/22/2015] [Indexed: 11/22/2022]
Abstract
Mitochondrial dysfunction has been implicated as a cause of energy deprivation in heart failure (HF). Herein, we tested individual and combined effects of two pathogenic factors of nonischemic HF, inhibition of nitric oxide synthesis [with l-N(G)-nitroarginine methyl ester (l-NAME)] and hypertension [with angiotensin II (AngII)], on myocardial mitochondrial function, oxidative stress, and metabolic gene expression. l-NAME and AngII were administered individually and in combination to mice for 5 wk. Although all treatments increased blood pressure and reduced cardiac contractile function, the l-NAME + AngII group was associated with the most severe HF, as characterized by edema, hypertrophy, oxidative stress, increased expression of Nppa and Nppb, and decreased expression of Atp2a2 and Camk2b. l-NAME + AngII-treated mice exhibited robust deterioration of cardiac mitochondrial function, as observed by reduced respiratory control ratios in subsarcolemmal mitochondria and reduced state 3 levels in interfibrillar mitochondria for complex I but not for complex II substrates. Cardiac myofibrils showed reduced ADP-supported and oligomycin-inhibited oxygen consumption. Mitochondrial functional impairment was accompanied by reduced mitochondrial DNA content and activities of pyruvate dehydrogenase and complex I but increased H2O2 production and tissue protein carbonyls in hearts from AngII and l-NAME + AngII groups. Microarray analyses revealed the majority of the gene changes attributed to the l-NAME + AngII group. Pathway analyses indicated significant changes in metabolic pathways, such as oxidative phosphorylation, mitochondrial function, cardiac hypertrophy, and fatty acid metabolism in l-NAME + AngII hearts. We conclude that l-NAME + AngII is associated with impaired mitochondrial respiratory function and increased oxidative stress compared with either l-NAME or AngII alone, resulting in nonischemic HF.
Collapse
Affiliation(s)
- Dale J Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas; Houston Methodist Department of Medicine, Weill Cornell Medical College, Houston, Texas
| | - Aijun Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas
| | - Shumin Li
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas
| | - Tram N Cao
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas
| | - Jessie A Smith
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas
| | - Indira Vedula
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas
| | - Andrea M Cordero-Reyes
- Houston Methodist Department of Cardiology, Weill Cornell Medical College, Houston, Texas
| | - Keith A Youker
- Houston Methodist Department of Cardiology, Weill Cornell Medical College, Houston, Texas
| | - Guillermo Torre-Amione
- Houston Methodist Department of Cardiology, Weill Cornell Medical College, Houston, Texas; Catedra de Cardiologia y Medicina Vascular, Tecnologico de Monterrey, Nuevo Leon, Mexico
| | - Anisha A Gupte
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas;
| |
Collapse
|
73
|
Zeinali N, Hashemi M, Mirmohammadsadeghi M, Mirmohammadsadeghi H, Eskandari N, Sabzghabaee AM. Association of Angiotensin-Converting Enzyme Genotype, Insertion/Deletion Polymorphism and Saphenous Vein Graft Atherosclerosis in Iranian Patients. Braz J Cardiovasc Surg 2016; 30:557-61. [PMID: 26735603 PMCID: PMC4690661 DOI: 10.5935/1678-9741.20150069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 09/23/2015] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE The aim of this study was to evaluate possible interactions among
Angiotensin-I converting enzyme genotype, insertion/deletion polymorphism
and atherosclerosis of vein grafts in Iranian patients, and characterize
their clinical and demographic profile. METHODS In this cross-sectional study, patients who underwent coronary artery bypass
graft surgery more than five years ago, were included for angiographic
analysis. Atherosclerosis was determined by quantitative angiography and
adjusted Gensini score. The gene angiotensin converting enzyme I/D
polymorphism was detected by polymerase chain reaction. RESULTS A total of 102 patients participated in this study. Eighty-four patients
were male. The frequency distribution of DD, ID and II polymorphism were
23.6%, 62.7% and 13.7% respectively. There were no differences among
genotypic groups in age, sex, number of risk factors, number of vein grafts
and months since bypass surgery. According to adjusted Gensini score
[0.18±0.12 (II) vs. 0.11±0.09 (ID) and
0.1±0.09 (DD) P=0.021] the II genotype was
associated with severity of vein graft atherosclerosis. CONCLUSION Although there are conflicting results about gene angiotensin converting
enzyme I/D polymorphism and the degree of venous bypass graft degeneration,
this study suggests an association between ACE genotype II and
atherosclerosis of saphenous vein grafts, however, large samples considering
clinical, demographic and ethnic profile are necessary to confirm these
results.
Collapse
Affiliation(s)
- Neda Zeinali
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Hashemi
- Department of Cardiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Mirmohammadsadeghi
- Department of Cardiothoracic Surgery, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Mirmohammadsadeghi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Mohammad Sabzghabaee
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
74
|
Abstract
Heart failure is a significant burden to the global healthcare system and represents an underserved market for new pharmacologic strategies, especially therapies which can address root cause myocyte dysfunction. Modern drugs, surgeries, and state-of-the-art interventions are costly and do not improve survival outcome measures. Gene therapy is an attractive strategy, whereby selected gene targets and their associated regulatory mechanisms can be permanently managed therapeutically in a single treatment. This in theory could be sustainable for the patient's life. Despite the promise, however, gene therapy has numerous challenges that must be addressed together as a treatment plan comprising these key elements: myocyte physiologic target validation, gene target manipulation strategy, vector selection for the correct level of manipulation, and carefully utilizing an efficient delivery route that can be implemented in the clinic to efficiently transfer the therapy within safety limits. This chapter summarizes the key developments in cardiac gene therapy from the perspective of understanding each of these components of the treatment plan. The latest pharmacologic gene targets, gene therapy vectors, delivery routes, and strategies are reviewed.
Collapse
Affiliation(s)
- Anthony S Fargnoli
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Center, New York, NY, USA.
| | - Michael G Katz
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Center, New York, NY, USA
| | - Charles R Bridges
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Center, New York, NY, USA
| | - Roger J Hajjar
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Center, New York, NY, USA
| |
Collapse
|
75
|
Woitek F, Zentilin L, Hoffman NE, Powers JC, Ottiger I, Parikh S, Kulczycki AM, Hurst M, Ring N, Wang T, Shaikh F, Gross P, Singh H, Kolpakov MA, Linke A, Houser SR, Rizzo V, Sabri A, Madesh M, Giacca M, Recchia FA. Intracoronary Cytoprotective Gene Therapy: A Study of VEGF-B167 in a Pre-Clinical Animal Model of Dilated Cardiomyopathy. J Am Coll Cardiol 2015; 66:139-53. [PMID: 26160630 DOI: 10.1016/j.jacc.2015.04.071] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 04/24/2015] [Accepted: 04/28/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF)-B activates cytoprotective/antiapoptotic and minimally angiogenic mechanisms via VEGF receptors. Therefore, VEGF-B might be an ideal candidate for the treatment of dilated cardiomyopathy, which displays modest microvascular rarefaction and increased rate of apoptosis. OBJECTIVES This study evaluated VEGF-B gene therapy in a canine model of tachypacing-induced dilated cardiomyopathy. METHODS Chronically instrumented dogs underwent cardiac tachypacing for 28 days. Adeno-associated virus serotype 9 viral vectors carrying VEGF-B167 genes were infused intracoronarily at the beginning of the pacing protocol or during compensated heart failure. Moreover, we tested a novel VEGF-B167 transgene controlled by the atrial natriuretic factor promoter. RESULTS Compared with control subjects, VEGF-B167 markedly preserved diastolic and contractile function and attenuated ventricular chamber remodeling, halting the progression from compensated to decompensated heart failure. Atrial natriuretic factor-VEGF-B167 expression was low in normally functioning hearts and stimulated by cardiac pacing; it thus functioned as an ideal therapeutic transgene, active only under pathological conditions. CONCLUSIONS Our results, obtained with a standard technique of interventional cardiology in a clinically relevant animal model, support VEGF-B167 gene transfer as an affordable and effective new therapy for nonischemic heart failure.
Collapse
Affiliation(s)
- Felix Woitek
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; University of Leipzig-Heart Center, Department of Cardiology/Internal Medicine, Leipzig, Germany
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Nicholas E Hoffman
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Jeffery C Powers
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Isabel Ottiger
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; University of Leipzig-Heart Center, Department of Cardiology/Internal Medicine, Leipzig, Germany
| | - Suraj Parikh
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Anna M Kulczycki
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Marykathryn Hurst
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Nadja Ring
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Tao Wang
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Farah Shaikh
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Polina Gross
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Harinder Singh
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Mikhail A Kolpakov
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Axel Linke
- University of Leipzig-Heart Center, Department of Cardiology/Internal Medicine, Leipzig, Germany
| | - Steven R Houser
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Victor Rizzo
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Abdelkarim Sabri
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Muniswamy Madesh
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Fabio A Recchia
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania; Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.
| |
Collapse
|
76
|
Lu Y, Li S, Wu H, Bian Z, Xu J, Gu C, Chen X, Yang D. Beneficial effects of astragaloside IV against angiotensin II-induced mitochondrial dysfunction in rat vascular smooth muscle cells. Int J Mol Med 2015; 36:1223-32. [PMID: 26398547 PMCID: PMC4601744 DOI: 10.3892/ijmm.2015.2345] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023] Open
Abstract
Angiotensin II (Ang II)-induced mitochondrial dysfunction is a prominent characteristic of the majority of cardiovascular diseases. Astragaloside IV (As-IV), the major active ingredient of Astragalus membranaceus (Fisch.) Bge. (a traditional Chinese herbal medicine), possesses antioxidant properties. The present study was carried out to examine whether As-IV can reverse Ang II-induced mitochondrial dysfunction in vascular smooth muscle cells (VSMCs) and to elucidate the underlying molecular mechanisms. Cultured rat aortic VSMCs treated with Ang II (1 µM) for 24 h exhibited mitochondrial dysfunction, including a decrease in mitochondrial oxygen consumption rates (OCRs), adenosine triphosphate (ATP) production and mitochondrial DNA (mtDNA) levels, as well as the disruption of mitochondrial structural integrity. Following treatment with Ang II, As-IV (50 µg/ml) was added to the culture medium followed by incubation for a further 24 h. The administration of As-IV significantly increased the mitochondrial OCRs, ATP production and the mtDNA levels, and reversed the mitochondrial morphological changes which occurred in the VSMCs. Treatment with As-IV also reversed the Ang II-induced increase in the production of reactive oxygen species (ROS), the increase in NADPH oxidase and xanthine oxidase activity, as well as the decrease in mitochondrial membrane potential (ΔΨm) and manganese superoxide dismutase (Mn-SOD) activity. Furthermore, treatment with As-IV led to an increase in the mRNA expression of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) and mitochondrial transcription factor A (Tfam), and in the protein expression of PGC-1α, parkin and dynamin 1-like protein 1 (Drp1) in the VSMCs. These results indicate that As-IV exerts beneficial effects on Ang II-induced mitochondrial dysfunction in rat VSMCs and that these effects are mediated through the inhibition of ROS overproduction, as well as the promotion of mitochondrial autophagy and mitochondrial biogenesis. These data demonstrate the antioxidant properties of As-IV.
Collapse
Affiliation(s)
- Yao Lu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Su Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hengfang Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zhiping Bian
- Research Institute of Cardiovascular Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jindan Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chunrong Gu
- Research Institute of Cardiovascular Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiangjian Chen
- Research Institute of Cardiovascular Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Di Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
77
|
Papinska AM, Mordwinkin NM, Meeks CJ, Jadhav SS, Rodgers KE. Angiotensin-(1-7) administration benefits cardiac, renal and progenitor cell function in db/db mice. Br J Pharmacol 2015; 172:4443-4453. [PMID: 26075703 PMCID: PMC4562506 DOI: 10.1111/bph.13225] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 05/05/2015] [Accepted: 06/07/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Diabetic patients are at an increased risk of cardiovascular disease, in part due to inflammation and oxidative stress. These two pathological mechanisms also affect other organs and cells including the kidneys and progenitor cells. Angiotensin-(1-7) [Ang-(1-7)] has previously been shown to counterbalance pathological effects of angiotensin II, including inflammation and oxidative stress. The aim of this study was to investigate the effects of short-term (2 weeks) Ang-(1-7) treatment on cardiovascular and renal function in a mouse model of type 2 diabetes (db/db). EXPERIMENTAL APPROACH Eight- to nine-week-old db/db mice were administered either vehicle, Ang-(1-7) alone, or Ang-(1-7) combined with an inhibitor (losartan, PD123319, A-779, L-NAME or icatibant) daily for 14 days. KEY RESULTS An improvement in physiological heart function was observed in Ang-(1-7)-treated mice. Ang-(1-7) also reduced cardiomyocyte hypertrophy, fibrosis and inflammatory cell infiltration of the heart tissue and increased blood vessel number. These changes were blocked by antagonists of the MAS1, AT2 and bradykinin receptors and inhibition of NO formation. Treatment with Ang-(1-7) reduced glomerular damage and oxidative stress in kidney tissue. Bone marrow and circulating endothelial progenitors, as well as bone marrow mesenchymal stem cells, were increased in mice treated with Ang-(1-7). CONCLUSIONS AND IMPLICATIONS Short-term Ang-(1-7) treatment of young db/db mice improved heart function and reduced kidney damage. Treatment also improved bone marrow and circulating levels of endothelial and mesenchymal stem cells. All of this may contribute to improved cardiovascular and renal function.
Collapse
Affiliation(s)
- A M Papinska
- School of Pharmacy, University of Southern CaliforniaLos Angeles, CA, USA
| | - N M Mordwinkin
- School of Pharmacy, University of Southern CaliforniaLos Angeles, CA, USA
| | - C J Meeks
- School of Pharmacy, University of Southern CaliforniaLos Angeles, CA, USA
| | - S S Jadhav
- School of Pharmacy, University of Southern CaliforniaLos Angeles, CA, USA
| | - K E Rodgers
- School of Pharmacy, University of Southern CaliforniaLos Angeles, CA, USA
| |
Collapse
|
78
|
Pesce P, Cecchetto L, Brocco S, Bolognesi M, Sodhi K, Abraham NG, Sacerdoti D. Characterization of a murine model of cardiorenal syndrome type 1 by high-resolution Doppler sonography. J Ultrasound 2015; 18:229-35. [PMID: 26261465 PMCID: PMC4529411 DOI: 10.1007/s40477-014-0129-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/14/2014] [Indexed: 12/25/2022] Open
Abstract
ABSTRACT Cardiorenal syndrome type 1 (CRS-1) is the acute kidney disfunction caused by an acute worsening of cardiac function. CRS-1 is the consequence of renal vasoconstriction secondary to renin-angiotensin system (RAS) activation. No animal models of CRS-1 are described in literature. PURPOSE To characterize a murine model of CRS-1 by using a high-resolution ultrasound echo-color Doppler system (VEVO2100). MATERIALS Post-ischemic heart failure was induced by coronary artery ligation (LAD) in seven CD1 mice. Fifteen and thirty days after surgery, mice underwent cardiac and renal echo-color Doppler. Serum creatinine and plasma renin activity were measured after killing. Animals were compared to seven CD1 control mice. RESULTS Heart failure with left ventricle dilatation (end diastolic area, p < 0.05 vs. controls) and significantly reduced ejection fraction (EF; p < 0.01 vs. controls) was evident 15 days after LAD. We measured a significant renal vasoconstriction in infarcted mice characterized by increased renal pulsatility index (PI; p < 0.05 vs. controls) associated to increased creatinine and renin levels (p < 0.05 vs. controls). CONCLUSIONS The mice model of LAD is a good model of CRS-1 evaluable by Doppler sonography and characterized by renal vasoconstriction due to the activation of the renin-angiotensin system secondary to heart failure.
Collapse
Affiliation(s)
- P. Pesce
- />Department of Medicine (DIMED), University of Padova, Padua, Italy
| | - L. Cecchetto
- />Department of Medicine (DIMED), University of Padova, Padua, Italy
| | - S. Brocco
- />Department of Medicine (DIMED), University of Padova, Padua, Italy
| | - M. Bolognesi
- />Department of Medicine (DIMED), University of Padova, Padua, Italy
| | - K. Sodhi
- />Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755 USA
| | - N. G. Abraham
- />Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755 USA
| | - D. Sacerdoti
- />Department of Medicine (DIMED), University of Padova, Padua, Italy
| |
Collapse
|
79
|
Zhang W, Chen L, Zhang L, Xiao M, Ding J, Goltzman D, Miao D. Administration of exogenous 1,25(OH)2D3 normalizes overactivation of the central renin-angiotensin system in 1α(OH)ase knockout mice. Neurosci Lett 2015; 588:184-9. [PMID: 25576706 DOI: 10.1016/j.neulet.2015.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/31/2014] [Accepted: 01/05/2015] [Indexed: 12/21/2022]
Abstract
Previously, we reported that active vitamin D deficiency in mice causes secondary hypertension and cardiac dysfunction, but the underlying mechanism remains largely unknown. To clarify whether exogenous active vitamin D rescues hypertension by normalizing the altered central renin-angiotensin system (RAS) via an antioxidative stress mechanism, 1-alpha-hydroxylase [1α(OH)ase] knockout mice [1α(OH)ase(-/-)] and their wild-type littermates were fed a normal diet alone or with 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], or a high-calcium, high-phosphorus "rescue" diet with or without antioxidant N-acetyl-l-cysteine (NAC) supplementation for 4 weeks. Compared with their wild-type littermates, 1α(OH)ase(-/-)mice had high mean arterial pressure, increased levels of renin, angiotensin II (Ang II), and Ang II type 1 receptor, and increased malondialdehyde levels, but decreased anti-peroxiredoxin I and IV proteins and the antioxidative genes glutathione reductase (Gsr) and glutathione peroxidase 4 (Gpx4) in the brain samples. Except Ang II type 1 receptor, these pathophysiological changes were rescued by exogenous 1,25(OH)2D3 or NAC plus rescue diet, but not by rescue diet alone. We conclude that 1,25(OH)2D3 normalizes the altered central RAS in 1α(OH)ase(-/-)mice, at least partially, through a central antioxidative mechanism.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China; Department of Human Anatomy, Kangda College, Lianyungang, PR China
| | - Lulu Chen
- Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Luqing Zhang
- Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China.
| | - Ming Xiao
- Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Jiong Ding
- Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - David Goltzman
- Calcium Research Laboratory, McGill University Health Center and Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Dengshun Miao
- Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| |
Collapse
|
80
|
Monu SR, Pesce P, Sodhi K, Boldrin M, Puri N, Fedorova L, Sacerdoti D, Peterson SJ, Abraham NG, Kappas A. HO-1 induction improves the type-1 cardiorenal syndrome in mice with impaired angiotensin II-induced lymphocyte activation. Hypertension 2013; 62:310-6. [PMID: 23753410 DOI: 10.1161/hypertensionaha.111.00495] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type-1 cardiorenal syndrome, characterized by acute kidney dysfunction secondary to cardiac failure and renal arteriolar vasoconstriction, is mediated by the renin-angiotensin-aldosterone axis and sympathetic nervous system activation. Previous reports indicate that angiotensin II modulates immune function and causes recruitment and activation of T-lymphocytes. The goal of this study was to evaluate the effects of postischemic heart failure on renal morphology and circulation and the beneficial effects of heme oxygenase-1 (HO-1) induction in T-lymphocyte-suppressed severe combined immune deficiency (SCID) mice. Mice were divided into 4 groups: sham, myocardial infarction (MI), MI treated with an HO-1 inducer, cobalt protoporphyrin, and with or without stannous mesoporphyrin, an inhibitor of HO activity. Heart and kidney function were studied 30 days after surgery. Fractional area change was reduced 30 days after surgery in both the C57 and SCID MI-groups as compared with their respective controls (P<0.01). Renal Pulsatility Index and renal injury were increased in C57 and SCID MI-groups compared with the sham group. HO-1 induction improved renal vasoconstriction as well as ameliorated renal injury in both the SCID and C57 MI-groups (P<0.01). However, improvement was more evident in SCID mice. In addition, our results showed that plasma creatinine, angiotensin II, and renin were significantly increased in the C57 and SCID MI-groups as compared with their respective controls. HO-1 induction decreased these parameters in both MI groups. Stannous mesoporphyrin reversed the beneficial effect of cobalt protoporphyrin in both mouse strains. The study demonstrates that T-lymphocyte suppression facilitated the HO-1-dependent improvement in the attenuation of type-1 cardiorenal syndrome.
Collapse
Affiliation(s)
- Sumit R Monu
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701-3655, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Rosenbaugh EG, Savalia KK, Manickam DS, Zimmerman MC. Antioxidant-based therapies for angiotensin II-associated cardiovascular diseases. Am J Physiol Regul Integr Comp Physiol 2013; 304:R917-28. [PMID: 23552499 DOI: 10.1152/ajpregu.00395.2012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases, including hypertension and heart failure, are associated with activation of the renin-angiotensin system (RAS) and increased circulating and tissue levels of ANG II, a primary effector peptide of the RAS. Through its actions on various cell types and organ systems, ANG II contributes to the pathogenesis of cardiovascular diseases by inducing cardiac and vascular hypertrophy, vasoconstriction, sodium and water reabsorption in kidneys, sympathoexcitation, and activation of the immune system. Cardiovascular research over the past 15-20 years has clearly implicated an important role for elevated levels of reactive oxygen species (ROS) in mediating these pathophysiological actions of ANG II. As such, the use of antioxidants, to reduce the elevated levels of ROS, as potential therapies for various ANG II-associated cardiovascular diseases has been intensely investigated. Although some antioxidant-based therapies have shown therapeutic impact in animal models of cardiovascular disease and in human patients, others have failed. In this review, we discuss the benefits and limitations of recent strategies, including gene therapy, dietary sources, low-molecular-weight free radical scavengers, polyethylene glycol conjugation, and nanomedicine-based technologies, which are designed to deliver antioxidants for the improved treatment of cardiovascular diseases. Although much work has been completed, additional research focusing on developing specific antioxidant molecules or proteins and identifying the ideal in vivo delivery system for such antioxidants is necessary before the use of antioxidant-based therapies for cardiovascular diseases become a clinical reality.
Collapse
Affiliation(s)
- Erin G Rosenbaugh
- Department of Cellular and Integrative Physiology, Nebraska Center for Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | |
Collapse
|