1
|
Gu Y, Ding Y, Zhang X, Li Y, Shang Z. Secreted frizzled-related protein 3 alleviated cardiac remodeling induced by angiotensin II via inhibiting oxidative stress and apoptosis in mice. Eur J Pharmacol 2022; 934:175303. [PMID: 36174667 DOI: 10.1016/j.ejphar.2022.175303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 12/01/2022]
Abstract
Increased expression of secreted frizzled related protein 3 (SFRP3) is associated with adverse outcomes of heart failure. The purpose of this study was to investigate the effect of SFRP3 on cardiac remodeling and its mechanism. Cardiac remodeling was induced by angiotensin II (Ang II) infusion in the mice, and in the neonatal rat cardiomyocytes (NRCM) treated with Ang II. The expression decreased in the heart of mice, and NRCM and HL-1 cells with Ang II treatment. Ang II-induced hypertrophy and fibrosis of heart in mice were attenuated by upregulation of SFRP3, and were further deteriorated by downregulation of SFRP3. Ang II-induced hypertrophy of NRCM and HL-1 cells were improved by SFRP3 overexpression, and were further deteriorated by SFRP3 knockdown. The oxidative stress increased in the heart of Ang II-treated mice, and this enhancement was inhibited by overexpressing of SFPR3, and was worsened by downregulation of SFPR3. These outcomes suggested that upregulation of SFPR3 could improve cardiac remodeling via inhibition of oxidative stress.
Collapse
Affiliation(s)
- Yang Gu
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Ying Ding
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xin Zhang
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Yong Li
- Department of Cardiology, The Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhenglu Shang
- Department of Cardiology, Wuxi Huishan District People's Hospital, Wuxi, Jiangsu, China.
| |
Collapse
|
2
|
Du Y, Demillard LJ, Ren J. Catecholamine-induced cardiotoxicity: A critical element in the pathophysiology of stroke-induced heart injury. Life Sci 2021; 287:120106. [PMID: 34756930 DOI: 10.1016/j.lfs.2021.120106] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 01/20/2023]
Abstract
Cerebrovascular diseases such as ischemic stroke, brain hemorrhage, and subarachnoid hemorrhage provoke cardiac complications such as heart failure, neurogenic stress-related cardiomyopathy and Takotsubo cardiomyopathy. With regards to the pathophysiology of stroke-induced heart injury, several mechanisms have been postulated to contribute to this complex interaction between brain and heart, including damage from gut dysbiosis, immune and systematic inflammatory responses, microvesicle- and microRNA-mediated vascular injury and damage from a surge of catecholamines. All these cerebrovascular diseases may trigger pronounced catecholamine surges through diverse ways, including stimulation of hypothalamic-pituitary adrenal axis, dysregulation of autonomic system, and secretion of adrenocorticotropic hormone. Primary catecholamines involved in this pathophysiological response include norepinephrine (NE) and epinephrine. Both are important neurotransmitters that connect the nervous system with the heart, leading to cardiac damage via myocardial ischemia, calcium (Ca2+) overload, oxidative stress, and mitochondrial dysfunction. In this review, we will aim to summarize the molecular mechanisms behind catecholamine-induced cardiotoxicity including Ca2+ overload, oxidative stress, apoptosis, cardiac hypertrophy, interstitial fibrosis, and inflammation. In addition, we will focus on how synchronization among these pathways evokes cardiotoxicity.
Collapse
Affiliation(s)
- Yuxin Du
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Laurie J Demillard
- School of Pharmacy, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
3
|
Liu J, Zheng X, Zhang C, Zhang C, Bu P. Lcz696 Alleviates Myocardial Fibrosis After Myocardial Infarction Through the sFRP-1/Wnt/β-Catenin Signaling Pathway. Front Pharmacol 2021; 12:724147. [PMID: 34539406 PMCID: PMC8443774 DOI: 10.3389/fphar.2021.724147] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Lcz696 (ARNI, angiotensin receptor–neprilysin inhibitor; sacubitril/valsartan) shows an inhibitory effect on fibrosis after myocardial infarction (MI). However, the underlying signaling mechanisms are poorly understood. The Wnt/β-catenin signaling pathway is activated after MI and participates in the process of myocardial fibrosis. Here, we aimed to assess the efficacy of ARNI for alleviating myocardial fibrosis after MI and hypothesized that ARNI alleviates myocardial fibrosis by inhibiting the Wnt/β-catenin signaling pathway and overexpressing sFRP-1, an inhibitor of the Wnt/β-catenin signaling pathway. Methods: Mice randomized at 1 week post-MI were administered lcz696 (60 mg/kg, n = 21), valsartan (30 mg/kg, n = 19), or corn oil (n = 13) orally for 4 weeks, while the sham-operated group received vehicle (corn oil, n = 19). Cardiac function and extent of myocardial fibrosis were measured. Western blotting and quantitative real-time polymerase chain reaction were used to detect the expression of Wnt/β-catenin pathway-related proteins. Furthermore, primary myocardial fibroblasts were stimulated with angiotensin II (Ang II) and cultured with lcz696 and the sFRP-1 inhibitor way316606 to detect the expression of Wnt/β-catenin pathway proteins. Results: Both lcz696 and valsartan alleviated myocardial fibrosis and improved cardiac function, but lcz696 had superior efficiency compared to valsartan. Furthermore, β-catenin expression was inhibited and sFRP-1 was overexpressed after drug treatment, which could be significantly improved by lcz696 in mice. In addition, lcz696 inhibited β-catenin expression in AngII-stimulated myocardial fibroblasts, and β-catenin expression increased after the inhibition of sFRP-1. Conclusion: ARNI alleviated cardiac fibrosis and cardiac remodeling by inhibiting the Wnt/β-catenin signaling pathway. In addition, ARNI can lead to overexpression of sFRP-1, which is an inhibitor of the Wnt/β-catenin signaling pathway. These results indicate a new therapeutic target of ARNI to improve myocardial fibrosis and prevent myocardial remodeling.
Collapse
Affiliation(s)
- Jing Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Cardiology, Heze Municipal Hospital, Heze, China
| | - Xuehui Zheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chen Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunmei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peili Bu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
4
|
Han M, Chen XC, Sun MH, Gai MT, Yang YN, Gao XM, Ma X, Chen BD, Ma YT. Overexpression of IκBα in cardiomyocytes alleviates hydrogen peroxide-induced apoptosis and autophagy by inhibiting NF-κB activation. Lipids Health Dis 2020; 19:150. [PMID: 32580730 PMCID: PMC7315514 DOI: 10.1186/s12944-020-01327-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/16/2020] [Indexed: 12/27/2022] Open
Abstract
Background Inflammation and oxidative stress play predominant roles in the initiation and progression of ischaemia/reperfusion (I/R) injury, with nuclear factor kappa B (NF-κB) serving as a crucial mediator. Overexpression of the inhibitor of κB alpha (IκBα) gene is hypothesized to have protective effects against apoptosis and autophagy in cardiomyocytes subjected to hydrogen peroxide (H2O2) by inhibiting the NF-κB pathway. Methods The IκBαS32A, S36A gene was transfected via adeno-associated virus serotype 9 (AAV9) delivery into neonatal rat ventricular cardiomyocytes (NRVMs) prior to H2O2 treatment. NRVMs were divided into control, H2O2, GFP + H2O2, IκBα+H2O2, and pyrrolidine dithiocarbamate (PDTC) + H2O2 groups. Nuclear translocation of the NF-κB p65 subunit was evaluated by immunofluorescence and Western blotting. Cell viability was assessed by Cell Counting Kit-8 assay. Supernatant lactate dehydrogenase (LDH) and intracellular malondialdehyde (MDA) were measured to identify H2O2-stimulated cytotoxicity. Apoptosis was determined by Annexin V-PE/7-AAD staining, and the mitochondrial membrane potential (ΔΨm) was detected by JC-1 staining. Western blotting was used to detect apoptosis- and autophagy-related proteins. Results IκBα transfection significantly increased cell viability and ΔΨm but decreased the supernatant LDH and cellular MDA levels in cardiomyocytes exposed to H2O2. Meanwhile, IκBα overexpression decreased H2O2-induced apoptosis by upregulating the Bcl-2/Bax ratio and reduced autophagy by downregulating the expression of Beclin-1 and the LC3-II/LC3-I ratio. These effects partly accounted for the ability of IκBα to inhibit the NF-κB signalling pathway, as evidenced by decreases in p65 phosphorylation and nuclear translocation. Indeed, the effects of inactivation of NF-κB signalling with the specific inhibitor PDTC resembled the cardioprotective effects of IκBα during H2O2 stimulation. Conclusion IκBα overexpression can ameliorate H2O2-induced apoptosis, autophagy, oxidative injury, and ΔΨm loss through inhibition of the NF-κB signalling pathway. These findings suggest that IκBα transfection can result in successful resistance to oxidative stress-induced damage by inhibiting NF-κB activation, which may provide a potential therapeutic target for the prevention of myocardial I/R injury.
Collapse
Affiliation(s)
- Min Han
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China.,Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Xiao-Cui Chen
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Ming-Hui Sun
- Department of Nephrology, Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, PR China
| | - Min-Tao Gai
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Yi-Ning Yang
- Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Xiao-Ming Gao
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Xiang Ma
- Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Bang-Dang Chen
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China.
| | - Yi-Tong Ma
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China. .,Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China.
| |
Collapse
|
5
|
Sun MH, Chen XC, Han M, Yang YN, Gao XM, Ma X, Huang Y, Li XM, Gai MT, Liu F, Ma YT, Chen BD. Cardioprotective effects of constitutively active MEK1 against H 2O 2-induced apoptosis and autophagy in cardiomyocytes via the ERK1/2 signaling pathway. Biochem Biophys Res Commun 2019; 512:125-130. [PMID: 30876692 DOI: 10.1016/j.bbrc.2019.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/01/2019] [Indexed: 12/11/2022]
Abstract
Oxidative stress injury is one of the main mechanisms of ischemia-reperfusion (I/R) injury. The extracellular signal-regulated kinase (ERK1/2) pathway plays an important role in cardioprotective during acute myocardial infarction. In this study, we used constitutively active MEK1 gene (CaMEK) transfection strategy to investigate whether CaMEK provides a protective effect against apoptosis and autophagy induced by Hydrogen peroxide (H2O2) in neonatal rat cardiac ventricular cardiomyocytes (NCMs) and the underlying mechanisms. As a result, CaMEK attenuated H2O2-induced apoptosis and cytotoxicity in NCMs, evidenced by decreased apoptotic cells and the ratio of Bax/Bcl-2, increased the mitochondrial membrane potential (Δψm) and cell vitality and reduced the level of lactate dehydrogenase (LDH). Further studies revealed that CaMEK attenuated H2O2-induced autophagy, evidenced by the decreased LC3-Ⅱ/LC3-Ⅰratio and SQSTM1/p62 (p62) degradation. Furthermore, we demonstrated that CaMEK phosphorylated the ERK1/2 pathway-related proteins, ERK1/2, p70S6K and GSK3β, in NCMs with H2O2 stimulation. In contrast, these effects could be reversed by co-treatment with the ERK1/2 inhibitor, PD98059. These results suggest that CaMEK plays an important role in protecting cardiomyocytes against H2O2-induced injury and autophagy in NCMs via ERK1/2 pathway. Therefore, transfection of CaMEK may provide a hopeful therapeutic strategy for I/R.
Collapse
Affiliation(s)
- Ming-Hui Sun
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China; Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, PR China; Department of Nephrology, Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, PR China
| | - Xiao-Cui Chen
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, PR China; Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Min Han
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China; Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, PR China
| | - Yi-Ning Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Xiao-Ming Gao
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, PR China; Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, 830054, PR China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, 830000, PR China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, 830054, PR China; Baker Heart and Diabetes Institute, Department of Surgery, Central Clinical School, Monash University, Melbourne, Australia
| | - Xiang Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Ying Huang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Xiao-Mei Li
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Min-Tao Gai
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, PR China; Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Fen Liu
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, PR China; Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, 830054, PR China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, PR China.
| | - Bang-Dang Chen
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, PR China; Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, 830054, PR China.
| |
Collapse
|
6
|
Hu Y, Guo Z, Lu J, Wang P, Sun S, Zhang Y, Li J, Zheng Q, Guo K, Wang J, Jiang J, Liu P. sFRP1 has a biphasic effect on doxorubicin-induced cardiotoxicity in a cellular location-dependent manner in NRCMs and Rats. Arch Toxicol 2018; 93:533-546. [PMID: 30377735 DOI: 10.1007/s00204-018-2342-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/23/2018] [Indexed: 12/22/2022]
Abstract
Doxorubicin (Dox) is an effective anticancer drug, however, its clinical application is restricted by the life-threatening cardiotoxic effects. Secreted Frizzled-related protein 1 (sFRP1) has been reported to participate in both the cancer and cardiovascular diseases and was one of the differential expression genes in normal hearts compared with Dox-treated hearts. Thus, it is important to reveal the potential role of sFRP1 in Dox-induced cardiotoxicity. Here, we show that sFRP1 has a biphasic effect on Dox-induced cardiotoxicity in a location-dependent manner. The secretion of sFRP1 was significantly increased in Dox-treated neonatal rat cardiomyocytes (NRCMs) (1 µM) and SD rats (5 mg/kg/injection at day 1, 5, and 9, i.p.). Adding the anti-sFRP1 antibody (0.5 µg/ml) and inhibiting sFRP1 secretion by caffeine (5 mM) both relieved Dox-induced cardiotoxicity through activating Wnt/β-catenin signaling, whereas increasing the secretion of sFRP1 by heparin (100 µg/ml) had the opposite effect. The intracellular level of sFRP1 was significantly decreased after Dox treatment both in vitro and in vivo. Knockdown of sFRP1 by sgRNA aggravated Dox-induced cardiotoxicity, while moderate overexpression of sFRP1 by Ad-sFRP1 exhibited protective effect. Besides, poly(ADP-ribosyl) polymerase-1 (PARP1) was screened as an interacting partner of sFRP1 in NRCMs by mass spectrometry. Our results suggested that the intracellular sFRP1 protected NRCMs from Dox-induced cardiotoxicity by interacting with PARP1. Thus, our results provide a novel evidence that sFRP1 has a biphasic effect on Dox-induced cardiotoxicity. In addition, the oversecretion of sFRP1 might be used as a biomarker to indicate the occurrence of cardiotoxicity induced by Dox treatment.
Collapse
Affiliation(s)
- Yuehuai Hu
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Sun Yat-sen University, 132 East Waihuan Road, Guangzhou, 510006, People's Republic of China
| | - Zhen Guo
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Sun Yat-sen University, 132 East Waihuan Road, Guangzhou, 510006, People's Republic of China
| | - Jing Lu
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Sun Yat-sen University, 132 East Waihuan Road, Guangzhou, 510006, People's Republic of China.
| | - Panxia Wang
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Sun Yat-sen University, 132 East Waihuan Road, Guangzhou, 510006, People's Republic of China
| | - Shuya Sun
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Yiqiang Zhang
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Jingyan Li
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Sun Yat-sen University, 132 East Waihuan Road, Guangzhou, 510006, People's Republic of China
| | - Qiyao Zheng
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Sun Yat-sen University, 132 East Waihuan Road, Guangzhou, 510006, People's Republic of China
| | - Kaiteng Guo
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Sun Yat-sen University, 132 East Waihuan Road, Guangzhou, 510006, People's Republic of China
| | - Junjian Wang
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Sun Yat-sen University, 132 East Waihuan Road, Guangzhou, 510006, People's Republic of China
| | - Jianmin Jiang
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Sun Yat-sen University, 132 East Waihuan Road, Guangzhou, 510006, People's Republic of China.
| | - Peiqing Liu
- School of Pharmaceutical Sciences, Guangzhou Higher Education Mega Center, Sun Yat-sen University, 132 East Waihuan Road, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
7
|
Meyer IS, Leuschner F. The role of Wnt signaling in the healing myocardium: a focus on cell specificity. Basic Res Cardiol 2018; 113:44. [PMID: 30327885 DOI: 10.1007/s00395-018-0705-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/01/2018] [Accepted: 10/09/2018] [Indexed: 12/18/2022]
Abstract
Various cell types are involved in the healing process after myocardial infarction (MI). Besides cardiac resident cells (such as cardiomyocytes, fibroblasts and endothelial cells) already present at the lesion site, a massive influx of leukocytes (mainly monocytes and neutrophils) is observed within hours after the ischemic event. So far, little is known about modes of interaction of these cells. Wnt signaling is an evolutionary conserved signaling cassette known to play an important role in cell-cell communication. While the overall reactivation of Wnt signaling upon ischemic injury is well described, the precise expression pattern of Wnt proteins, however, is far from understood. We here describe known Wnt components that partake in MI healing and differentiate cell-specific aspects. The secretion of Wnt proteins and their antagonists in the context of cardiac inflammation after MI appear to be tightly regulated in a spatial-temporal manner. Overall, we aim to stress the importance of elucidating not only Wnt component-specific aspects, but also their sometimes contradicting effects in different target cells. A better understanding of Wnt signaling in MI healing may eventually lead to the development of successful therapeutic approaches in an often considered "un-druggable" pathway.
Collapse
Affiliation(s)
- Ingmar Sören Meyer
- Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Florian Leuschner
- Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
8
|
Zhang P, Hua L, Hou H, Du X, He Z, Liu M, Hu X, Yan N. Sphingomyelin synthase 2 promotes H2O2-induced endothelial dysfunction by activating the Wnt/β-catenin signaling pathway. Int J Mol Med 2018; 42:3344-3354. [PMID: 30272329 PMCID: PMC6202097 DOI: 10.3892/ijmm.2018.3888] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 09/17/2018] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis (AS) is the primary cause of various cardiovascular and cerebrovascular diseases and has high morbidity and mortality rates. Oxidative stress-induced endothelial cells (ECs) dysfunction is the pathological basis of AS. In addition, sphingomyelin (SM) and the Wnt/β-catenin signaling pathway are considered to be closely associated with AS; however, the specific mechanism is not clear. Therefore, the present study investigated whether SM may induce ECs dysfunction through the Wnt/β-catenin signaling pathway. Firstly, a sphingomyelin synthase 2 (SMS2) overexpression cell model was constructed. It was identified that the expression of SMS2 was increased when ECs were treated with H2O2. In addition, these results demonstrated that SMS2 overexpression promoted apoptosis and macrophage adhesion of H2O2-induced ECs, thereby increasing the expression of β-catenin. Furthermore, SMS activity was inhibited with Dy105, combined with simultaneous treatment with LiCl or H2O2. This additionally confirmed that Dy105 significantly inhibited SMS activity and decreased the level of ECs dysfunction and β-catenin content; however, LiCl served a key role in activating the Wnt/β-catenin signaling pathway to promote ECs dysfunction. Collectively, these results suggested that SMS2 overexpression may promote ECs dysfunction by activating the Wnt/β-catenin signaling pathway, while Dy105 may inhibit the evolution of oxidative stress-induced dysfunction.
Collapse
Affiliation(s)
- Panpan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lingyue Hua
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Huan Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xingyue Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhiqiang He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Menghan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaojuan Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Nianlong Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
9
|
Axin1 up-regulated 1 accelerates stress-induced cardiomyocytes apoptosis through activating Wnt/β-catenin signaling. Exp Cell Res 2017; 359:441-448. [DOI: 10.1016/j.yexcr.2017.08.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/13/2022]
|
10
|
Wang X, Peng Q, Jiang F, Xue L, Li J, Fan Z, Chen P, Chen G, Cai Y. Secreted frizzled-related protein 5 protects against oxidative stress-induced apoptosis in human aortic endothelial cells via downregulation of Bax. J Biochem Mol Toxicol 2017; 31. [PMID: 28834606 DOI: 10.1002/jbt.21978] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022]
Abstract
This study was undertaken to determine the role of secreted frizzled-related protein 5 (SFRP5) in endothelial oxidative injury. Human aortic endothelial cells (HAECs) were exposed to different oxidative stimuli and examined for SFRP5 expression. The effects of SFRP5 overexpression and knockdown on cell viability, apoptosis, and reactive oxygen species production were measured. HAECs treated with angiotensin (Ang) II (1 μM) or oxidized low-density lipoprotein (oxLDL) (150 μg/mL) showed a significant increase in SFRP5 expression. Overexpression of SFRP5 significantly attenuated the viability suppression and apoptosis induction by Ang II and oxLDL, whereas the knockdown of SFRP5 exerted opposite effects. Overexpression of SFRP5 prevented ROS formation and β-catenin activation and reduced Bax expression. Co-expression of Bax significantly reversed the anti-apoptotic effect of SFRP5 overexpression, whereas knockdown of Bax restrained Ang II- and oxLDL-induced apoptosis in HAECs. Taken together, SFRP5 confers protection against oxidative stress-induced apoptosis through inhibition of β-catenin activation and downregulation of Bax.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Endocrinology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Qing Peng
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Feng Jiang
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Li Xue
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jiafu Li
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Pan Chen
- Department of Endocrinology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Guo Chen
- Department of Endocrinology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yihua Cai
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
11
|
Xing Z, Ni Y, Zhao J, Ma X. Hydrogen Peroxide-Induced Secreted Frizzled-Related Protein 1 Gene Demethylation Contributes to Hydrogen Peroxide-Induced Apoptosis in Human U251 Glioma Cells. DNA Cell Biol 2017; 36:347-353. [PMID: 28398872 DOI: 10.1089/dna.2016.3594] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Zhiguo Xing
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Yaping Ni
- Department of General Medicine and Geriatrics, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Junjie Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xudong Ma
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
12
|
Gao YY, Zeng P, Jia CL, Zhang Y, Zhang T, Jia Q, Li YM. Two new phenols from Lysimachia patungensis. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2017; 19:28-34. [PMID: 27308956 DOI: 10.1080/10286020.2016.1183652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Two new phenols, methyl 3-(2-O-β-d-glucopyranosyl-3-hydroxy-5-methoxyphenyl) propionate (1) and myricetin-3,3',5'-tri-O-α-l-rhamnopyranoside (2), together with six known phenols compounds (3-8), were isolated from the whole plant of Lysimachia patungensis Hand.-Mazz. Their structures were elucidated on the basis of the interpretation of spectroscopic data, viz., ESI-MS, HR-TOF-MS, UV, IR, and NMR. All the known phenols were isolated from the genus Lysimachia for the first time. A preliminary bioassay revealed that compounds 3 and 7 exhibited significant protective effects against hydrogen peroxide-induced damage in human retinal endothelial cells (HRECs) with the concentration of 10 μM, respectively. Compound 1 showed moderate activity against the HRECs damage at 100 μM.
Collapse
Affiliation(s)
- Yuan-Yuan Gao
- a School of Pharmacy , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Peng Zeng
- a School of Pharmacy , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Cheng-Lin Jia
- b Yueyang Hospital and Clinical Research Institute of Integrative Medicine , Shanghai University of Traditional Chinese Medicine , Shanghai 200437 , China
| | - Yong Zhang
- c Drug Discovery and Design Centre & CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Teng Zhang
- b Yueyang Hospital and Clinical Research Institute of Integrative Medicine , Shanghai University of Traditional Chinese Medicine , Shanghai 200437 , China
| | - Qi Jia
- a School of Pharmacy , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Yi-Ming Li
- a School of Pharmacy , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| |
Collapse
|
13
|
Tao J, Abudoukelimu M, Ma YT, Yang YN, Li XM, Chen BD, Liu F, He CH, Li HY. Secreted frizzled related protein 1 protects H9C2 cells from hypoxia/re-oxygenation injury by blocking the Wnt signaling pathway. Lipids Health Dis 2016; 15:72. [PMID: 27048460 PMCID: PMC4822324 DOI: 10.1186/s12944-016-0240-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/30/2016] [Indexed: 11/17/2022] Open
Abstract
Background In animal models, secreted frizzled related protein 1 (Sfrp1) inhibition of the Wnt signaling pathway is beneficial because Sfrp1 reduces myocardial apoptosis and prevents heart failure. The mechanisms mediating the cellular survival effect of Sfrp1 has not been completely elucidated. The present study was designed to investigate the possible protective actions of Sfrp1 on cardiac muscle cells using an in vitro model of ischemia/reperfusion, and to evaluate the possible involvement of the Wnt signaling pathway. Methods We used a recombinant AAV9 vector to deliver the Sfrp1 gene into H9C2 rat cardiomyoblasts and adopted an in vitro model of ischemia/reperfusion. Cell vitality was measured by CKK-8 and the trypan blue exclusion assay. Western blot was used to evaluate the expression of Dvl-1, β-catenin, c-Myc, Bax, and Bcl-2. Flow cytometry analysis of cardiomyocyte apoptosis was performed. Results We confirmed that Sfrp1 significantly increased cell viability (assayed by trypan blue and CKK-8) and decreased apoptosis (assayed by flow cytometry analysis and the Bax/Bcl-2 ratio). These effects were partly attributable to the ability of Sfrp1 to down-regulate Wnt signaling pathway (assayed by Western blot to evaluate the expression of Dvl-1, β-catenin, and c-Myc). Indeed, reactivation of the Wnt signaling pathway activity with the specific activator, Licl, reduced Sfrp1-induced cardioprotection during hypoxia and reoxygenation. Conclusions The present study demonstrated that Sfrp1 directly protected H9C2 cells from hypoxia and reoxygenation-induced reperfusion injury and apoptosis through inhibition of the Wnt signaling pathway, and added new mechanistic insight regarding the cardioprotective role of Sfrp1 on ischemic damage. Electronic supplementary material The online version of this article (doi:10.1186/s12944-016-0240-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing Tao
- Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.,Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China
| | - Mayila Abudoukelimu
- Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.,Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China
| | - Yi-tong Ma
- Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China. .,Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China. .,Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.
| | - Yi-ning Yang
- Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China
| | - Xiao-mei Li
- Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China
| | - Bang-dang Chen
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China
| | - Fen Liu
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China
| | - Chun-hui He
- Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.,Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China
| | - Hua-yin Li
- Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China.,Xinjiang Medical University, Li Yu Shan South Road 137, Urumqi, 830001, People's Republic of China
| |
Collapse
|
14
|
Li S, Xu M, Niu Q, Xu S, Ding Y, Yan Y, Guo S, Li F. Efficacy of Procyanidins against In Vivo Cellular Oxidative Damage: A Systematic Review and Meta-Analysis. PLoS One 2015; 10:e0139455. [PMID: 26426531 PMCID: PMC4591260 DOI: 10.1371/journal.pone.0139455] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/14/2015] [Indexed: 01/18/2023] Open
Abstract
AIMS In this study, the efficacy of proanthocyanidins (PCs) against oxidative damage was systematically reviewed to facilitate their use in various applications. METHODS A meta-analysis was performed by two researchers. Each investigator independently searched electronic databases, including Cochrane, PubMed, Springer, Web of Science, China National Knowledge Infrastructure (CKNI), China Science and Technology Journal Database (CSTJ), and WanFang Data, and analyzed published data from 29 studies on the effects of PCs against oxidative damage. Oxidative stress indexes included superoxide dismutase (SOD), malondialdehyde (MDA), catalase (CAT), glutathione (GSH), glutathione peroxidase (GPx), and total antioxidative capacity (T-AOC). RESULTS Compared with the oxidative damage model group, PCs effectively improved the T-AOC, SOD, GSH, GPx, and CAT levels, and reduced the MDA levels; these differences were statistically significant (P < 0.05). In studies that used the gavage method, SOD (95% CI, 2.33-4.00) and GPx (95% CI, 2.10-4.05) were 3.16-fold and 3.08-fold higher in the PC group than in the control group, respectively. In studies that used the feeding method, SOD (95% CI, 0.32-1.74) and GPx (95% CI, -0.31 to 1.65) were 1.03-fold and 0.67-fold higher in the PC group than in the control group, respectively. Statistically significant differences in the effects of PCs (P < 0.00001) were observed between these two methods. MDA estimated from tissue samples (95% CI, -5.82 to -2.60) was 4.32-fold lower in the PC group than in the control group. In contrast, MDA estimated using serum samples (95% CI, -4.07 to -2.06) was 3.06-fold lower in the PC group than in the control group. The effect of PCs on MDA was significantly greater in tissue samples than in serum samples (P = 0.02). CONCLUSION PCs effectively antagonize oxidative damage and enhance antioxidant capacity. The antagonistic effect may be related to intervention time, intervention method, and the source from which the indexes are estimated.
Collapse
Affiliation(s)
- Shugang Li
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Xinjiang, China
| | - Mengchuan Xu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Xinjiang, China
| | - Qiang Niu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Xinjiang, China
| | - Shangzhi Xu
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Xinjiang, China
| | - Yusong Ding
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Xinjiang, China
| | - Yizhong Yan
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Xinjiang, China
| | - Shuxia Guo
- Department of Public Health and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Xinjiang, China
| | - Feng Li
- Department of Pathology and Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| |
Collapse
|