1
|
Xu X, Liu Y, Lan M, Liu F, Xia H, Zeng J. Suppression of SRC protein kinase activity alleviates the severity of aganglionosis by impairing CAV1/FLNA expression. Sci Prog 2025; 108:368504251336287. [PMID: 40296549 DOI: 10.1177/00368504251336287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
ObjectiveHirschsprung's disease (HSCR) is a rare congenital disorder attributed to the defects of enteric neural crest cells. We aim to identify characteristic phosphorylation proteins and preliminarily explore underlying related action mechanisms in HSCR.MethodsColon samples from HSCR patients underwent proteomic and phosphoproteomic sequencing to identify differentially expressed phosphoproteins (DEPPs) and proteins (DEPs). Interaction network construction and analysis of correlations with upstream phosphorylating kinases were employed to pinpoint core proteins. HSCR rat models were established through enema administration of Benzalkonium chloride and evaluated by measuring colon cross-sectional area, colon weight, AchE, and PGP9.5 levels. Histopathological damage was assessed via hematoxylin and eosin staining. Protein expression was analyzed using western blotting. Furthermore, the impact of SRC kinase in HSCR was investigated utilizing an SRC-specific inhibitor in HSCR rat models.ResultsA total of 5725 DEPPs were identified, with SRC kinase emerging as a key regulatory protein. In the HSCR rat model, SRC expression was elevated along with increased pCAV1 and FLNA levels. Notably, inhibition of SRC protein kinase activity by 1-(tert-butyl)-3-(4-chlorophenyl)-1H-pyrazolo[3,4-d] pyrimidin-4-amine (PP2) led to reduced colon cross-sectional area and weight, an increase in the number of colonic ganglion cells, heightened AchE levels, enhanced PGP9.5 expression, and slight enlargement of the crypt, thereby alleviating HSCR symptoms in rats. Additionally, SRC kinase inhibition following PP2 treatment decreased the expression of pCAV1 and FLNA.ConclusionsInhibition of SRC kinase activity may potentially reduce CAV1/FLNA expression, ultimately alleviating the severity of HSCR in rats.
Collapse
Affiliation(s)
- Xiaogang Xu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yanqing Liu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Menglong Lan
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Fei Liu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jixiao Zeng
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| |
Collapse
|
2
|
Zhang S, Niu Q, Zong W, Song Q, Tian S, Wang J, Liu J, Zhang H, Wang Z, Li B. Endotype-driven Co-module mechanisms of danhong injection in the Co-treatment of cardiovascular and cerebrovascular diseases: A modular-based drug and disease integrated analysis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118287. [PMID: 38705429 DOI: 10.1016/j.jep.2024.118287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiovascular and cerebrovascular diseases are the leading causes of death worldwide and interact closely with each other. Danhong Injection (DHI) is a widely used preparation for the co-treatment of brain and heart diseases (CTBH). However, the underlying molecular endotype mechanisms of DHI in the CTBH remain unclear. AIM OF THIS STUDY To elucidate the underlying endotype mechanisms of DHI in the CTBH. MATERIALS AND METHODS In this study, we proposed a modular-based disease and drug-integrated analysis (MDDIA) strategy for elucidating the systematic CTBH mechanisms of DHI using high-throughput transcriptome-wide sequencing datasets of DHI in the treatment of patients with stable angina pectoris (SAP) and cerebral infarction (CI). First, we identified drug-targeted modules of DHI and disease modules of SAP and CI based on the gene co-expression networks of DHI therapy and the protein-protein interaction networks of diseases. Moreover, module proximity-based topological analyses were applied to screen CTBH co-module pairs and driver genes of DHI. At the same time, the representative driver genes were validated via in vitro experiments on hypoxia/reoxygenation-related cardiomyocytes and neuronal cell lines of H9C2 and HT22. RESULTS Seven drug-targeted modules of DHI and three disease modules of SAP and CI were identified by co-expression networks. Five modes of modular relationships between the drug and disease modules were distinguished by module proximity-based topological analyses. Moreover, 13 targeted module pairs and 17 driver genes associated with DHI in the CTBH were also screened. Finally, the representative driver genes AKT1, EDN1, and RHO were validated by in vitro experiments. CONCLUSIONS This study, based on clinical sequencing data and modular topological analyses, integrated diseases and drug targets. The CTBH mechanism of DHI may involve the altered expression of certain driver genes (SRC, STAT3, EDN1, CYP1A1, RHO, RELA) through various enriched pathways, including the Wnt signaling pathway.
Collapse
Affiliation(s)
- Siqi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qikai Niu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wenjing Zong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qi Song
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Siwei Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jingai Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huamin Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
3
|
Sahadevan R, Binoy A, Shajan I, Sadhukhan S. Mitochondria-targeting EGCG derivatives protect H9c2 cardiomyocytes from H 2O 2-induced apoptosis: design, synthesis and biological evaluation. RSC Adv 2023; 13:29477-29488. [PMID: 37818277 PMCID: PMC10561634 DOI: 10.1039/d3ra04527g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/02/2023] [Indexed: 10/12/2023] Open
Abstract
Pathologies related to cardiovascular diseases mostly emerge as a result of oxidative stress buildup in cardiomyocytes. The heavy load of mitochondrial oxidative phosphorylation in cardiac tissues corresponds to a surge in oxidative stress leading to mitochondrial dysfunction and cellular apoptosis. Thus, scavenging the reactive oxygen species (ROS) linked to mitochondria can significantly improve cardio-protection. Epigallocatechin-3-gallate (EGCG), the major polyphenol found in green tea has been extensively studied for its profound health-beneficial activities. Herein, we designed and synthesized a series of mitochondrial-targeting EGCG derivatives, namely MitoEGCGn (n = 4, 6, 8) by incorporating triphenylphosphonium ion onto it using different linkers. MitoEGCGn were found to be non-toxic to H9c2 rat cardiomyocyte cells even at higher doses in comparison to its parent molecule EGCG. Interestingly, MitoEGCG4 and MitoEGCG6 protected the H9c2 cardiomyocyte cells from the oxidative damage induced by H2O2 whereas EGCG was found to be toxic and ineffective in protecting the cells from H2O2 damage. MitoEGCG4 and MitoEGCG6 also protected the cells from the H2O2-induced disruption of mitochondrial membrane potential as well as activation of apoptosis as revealed by pro-caspase 3 expression profile, DNA fragmentation assay, and AO/EtBr staining. Taken together, our study shows that the mitochondria targeting EGCG derivatives were able to effectively combat the H2O2-induced oxidative stress in H9c2 cardiomyocytes. They eventually augmented the mitochondrial health of cardiomyocytes by maintaining the mitochondrial function and attenuating apoptosis. Overall, MitoEGCG4 and MitoEGCG6 could provision a cardioprotective role to H9c2 cardiomyocytes at the time of oxidative insults related to mitochondrial dysfunction-associated injuries.
Collapse
Affiliation(s)
- Revathy Sahadevan
- Department of Chemistry, Indian Institute of Technology Palakkad Kerala 678 623 India
| | - Anupama Binoy
- Department of Chemistry, Indian Institute of Technology Palakkad Kerala 678 623 India
| | - Irene Shajan
- Department of Chemistry, Indian Institute of Technology Palakkad Kerala 678 623 India
| | - Sushabhan Sadhukhan
- Department of Chemistry, Indian Institute of Technology Palakkad Kerala 678 623 India
- Physical & Chemical Biology Laboratory, Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad Kerala 678 623 India
| |
Collapse
|
4
|
Zhang X, Xu M, Cai S, Chen B, Lin H, Liu Z. Effects of astaxanthin on microRNA expression in a rat cardiomyocyte anoxia-reoxygenation model. Front Pharmacol 2023; 14:1103971. [PMID: 36817156 PMCID: PMC9936191 DOI: 10.3389/fphar.2023.1103971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction: The protective effects of astaxanthin against myocardial ischemia-reperfusion injuries are well documented, although the mechanisms are not defined. Methods: The anoxia-reoxygenation injury model was established after astaxanthin treated H9c2 cells for 24 h. Cell viability, lactate dehydrogenase, oxidative stress level and western blot were tested. Secondly, measured the effects of astaxanthin pretreatment on microRNA expression in a rat myocardial cell anoxia-reoxygenation injury model. Results: After anoxia-reoxygenation injury, in a dose dependent manner, astaxanthin increased cell viability, superoxide dismutase and glutathione peroxidase activity, decreased lactate dehydrogenase and malondialdehyde levels, downregulated protein expression of caspase-3, caspase-8, nuclear factor erythroid-2-related factor 2 and heme oxygenase-1, and upregulated the Bcl-2/Bax ratio. High-throughput sequencing and qPCR showed that microRNAs rno-miR-125b-5p and rno-let-7c-1-3p were differentially expressed (|log2| ≥ 0.585, q < 0.1) between the normal, anoxia-reoxygenation, and astaxanthin (1.25 μM) groups. Kyoto Encyclopedia of Genes and Genomes and GO Gene ontology pathway enrichment analyses showed that TNF signaling, axon guidance, NF-κB signaling pathway, and other pathways displayed differentially expressed microRNA target genes associated with myocardial injuries. Discussion: These results suggested that thetarget genes of rno-miR-125b-5p were enriched in inflammation and apoptosis-related signaling pathways. Also, the results imply that simultaneous targeting of these related signaling pathways could significantly prevent myocardial anoxia-reoxygenation injury in the presence of astaxanthin.
Collapse
Affiliation(s)
- Xinxin Zhang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China,Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, National Research and Development Center for Marine Fish Processing, Xiamen, China,Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, China
| | - Min Xu
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, National Research and Development Center for Marine Fish Processing, Xiamen, China,College of Ocean Food and Biological Engineering, Jimei University, Xiamen, China
| | - Shuilin Cai
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, National Research and Development Center for Marine Fish Processing, Xiamen, China
| | - Bei Chen
- Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, National Research and Development Center for Marine Fish Processing, Xiamen, China
| | - Hetong Lin
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China,Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, China
| | - Zhiyu Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China,Key Laboratory of Cultivation and High-Value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, National Research and Development Center for Marine Fish Processing, Xiamen, China,*Correspondence: Zhiyu Liu,
| |
Collapse
|
5
|
Wang L, Sheng G, Cui J, Yao Y, Bai X, Chen F, Yu W. Electroacupuncture attenuates ischemic injury after stroke and promotes angiogenesis via activation of EPO mediated Src and VEGF signaling pathways. PLoS One 2022; 17:e0274620. [PMID: 36108080 PMCID: PMC9477374 DOI: 10.1371/journal.pone.0274620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Although electroacupuncture (EA) has been shown to be effective in the treatment of stroke, its mechanisms of action remain undefined. This study explored the therapeutic effects of EA in rats with cerebral ischemia-reperfusion injury (CIRI) and evaluated its possible mechanisms in promoting angiogenesis. To evaluate the effect of EA, we used 2, 3, 5-Triphenyl-2H-Tetrazolium Chloride (TTC) staining and behavior score to calculate the cerebral infarct volume and neurological deficit score after CIRI. Western blot (WB) analysis was employed to evaluate the expression of cluster of differentiation 34 (CD34), erythropoietin (EPO), vascular endothelial growth factor (VEGF) and phospho-Src (p-Src) in the brain of the rats with CIRI. On the other hand, we established an oxygen-glucose deprivation/reoxygenation (OGD/R) injury model using brain microvascular endothelial cells (BMECs), and analyzed cell viability and expression of VEGF or p-Src using cell counting kit-8 (CCK-8) and WB, respectively. Our data showed that EA at the GV26 acupoint could significantly promote the expression of CD34, EPO, VEGF and p-Src in CIRI rats. Our CCK-8 results demonstrated that intervention with recombinant EPO and VEGF proteins remarkably improved the viability of BMECs after OGD/R, while a Src inhibitor, PP1, reversed this phenotype. The WB results showed that the recombinant EPO protein increased the expression of VEGF and p-Src, which was significantly inhibited by PP1. Taken together, our findings showed that EA at the GV26 acupoint can significantly attenuate ischemic injury after stroke and promote angiogenesis via activation of EPO-mediated Src and VEGF signaling pathways. Besides, the upregulation of VEGF may also be associated with the activation of Src by EPO.
Collapse
Affiliation(s)
- Lifen Wang
- Shaanxi Academy of Traditional Chinese Medicine, Shaanxi Provincial Hospital of Chinese Medicine, Xi’an, China
| | - Gang Sheng
- Shaanxi Academy of Traditional Chinese Medicine, Shaanxi Provincial Hospital of Chinese Medicine, Xi’an, China
| | - Jinjun Cui
- Department of Neurology, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, China
| | - Yanling Yao
- Shaanxi Academy of Traditional Chinese Medicine, Shaanxi Provincial Hospital of Chinese Medicine, Xi’an, China
| | - Xue Bai
- College of Acupuncture-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Xian yang, China
| | - Fan Chen
- College of Acupuncture-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Xian yang, China
| | - Wei Yu
- Department of Physiology, Xi’an Medical University, Xi’an, China
| |
Collapse
|
6
|
Tsai YT, Ruan JW, Chang CS, Ko ML, Chou HC, Lin CC, Lin CM, Huang CT, Wei YS, Liao EC, Chen HY, Lin LH, Lin MW, Kao CY, Chan HL. Proteomic and microbial assessments on the effect of Antrodia cinnamomea in C57BL/6 mice. Arch Biochem Biophys 2021; 713:109058. [PMID: 34627749 DOI: 10.1016/j.abb.2021.109058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 01/01/2023]
Abstract
Antrodia cinnamomea (AC) is a nutraceutical fungus and studies have suggested that AC has the potential to prevent or alleviate diseases. However, little is known about the AC-induced phenotypes on the intestine-liver axis and gut microbial alterations. Here, we performed two-dimensional difference gel electrophoresis (2D-DIGE) and MALDI-Biotyper to elaborate the AC-induced phenotypes on the intestine-liver axis and gut microbial distribution of C57BL/6 mice. The experimental outcomes showed that the hepatic density may increase by elevating hepatic redox regulation, lipid degradation and glycolysis-related proteins and alleviating cholesterol biosynthesis and transport-related proteins in C57BL/6 mice with AC treatment. Moreover, AC facilitates intestinal glycolysis, TCA cycle, redox and cytoskeleton regulation-related proteins, but also reduces intestinal vesicle transport-related proteins in C57BL/6 mice. However, the body weight, GTT, daily food/water intake, and fecal/urine weight were unaffected by AC supplementation in C57BL/6 mice. Notably, the C57BL/6-AC mice had a higher gut microbial abundance of Alistipes shahii (AS) than C57BL/6-Ctrl mice. In summary, the AC treatment affects intestinal permeability by regulating redox and cytoskeleton-related proteins and elevates the gut microbial abundance of AS in C57BL/6 mice that might be associated with increasing hepatic density and metabolism-related proteins of the liver in C57BL/6 mice. Our study provides an insight into the mechanisms of AC-induced phenotypes and a comprehensive assessment of AC's nutraceutical effect in C57BL/6 mice.
Collapse
Affiliation(s)
- Yi-Ting Tsai
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Jhen-Wei Ruan
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan.
| | - Cherng-Shyang Chang
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan.
| | - Mei-Lan Ko
- Department of Ophthalmology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, 30059, Taiwan.
| | - Hsiu-Chuan Chou
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Chi-Chien Lin
- Department of Life Sciences, Institute of Biomedical Science, National Chung Hsing University, Taichung, 402, Taiwan.
| | - Chiao-Mei Lin
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan.
| | - Chih-Ting Huang
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan.
| | - Yu-Shan Wei
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - En-Chi Liao
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Hsin-Yi Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Li-Hsun Lin
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Meng-Wei Lin
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Cheng-Yuan Kao
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, 35053, Taiwan.
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology and Department of Medical Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
7
|
Kim KW, Kim SW, Lim S, Yoo KJ, Hwang KC, Lee S. Neutralization of hexokinase 2-targeting miRNA attenuates the oxidative stress-induced cardiomyocyte apoptosis. Clin Hemorheol Microcirc 2021; 78:57-68. [PMID: 33523042 DOI: 10.3233/ch-200924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hexokinase 2 (HK2) is a metabolic sensor that couples glycolysis and oxidative phosphorylation of mitochondria by binding to the outer mitochondrial membrane (OMM), and it also has been implicated in induction of apoptotic process by regulating the integrity of OMM. When HK2 detaches from the mitochondria, it triggers permeability increase of the OMM and subsequently facilitates the cytosolic release of cytochrome c, a major apoptosis-inducing factor. According to previous studies, a harsh microenvironment created by ischemic heart disease such as low tissue oxygen and nutrients, and increased reactive oxygen species (ROS) can cause cardiomyocyte apoptosis. Under these conditions, the expression of HK2 in heart significantly decrease and such down-regulation of HK2 was correlated to the increased apoptosis of cardiomyocytes. Therefore, prevention of HK2 down-regulation may salvage cardiomyocytes from apoptosis. MicroRNAs are short, non-coding RNAs that either inhibit transcription of target mRNAs or degrade the targeted mRNAs via complementary binding to the 3'UTR (untranslated region) of the targeted mRNAs. Since miRNAs are known to be involved in virtually every biological processes, it is reasonable to assume that the expression of HK2 is also regulated by miRNAs. Currently, to my best knowledge, there is no previous study examined the miRNA-mediated regulation of HK2 in cardiomyocytes. Thus, in the present study, miRNA-mediated modulation of HK2 during ROS (H2O2)-induced cardiomyocyte apoptosis was investigated. First, the expression of HK2 in cardiomyocytes exposed to H2O2 was evaluated. H2O2 (500 μM) induced cardiomyocyte apoptosis and it also decreased the mitochondrial expression of HK2. Based on miRNA-target prediction databases and empirical data, miR-181a was identified as a HK2-targeting miRNA. To further examine the effect of negative regulation of the selected HK2-targeting miRNA on cardiomyocyte apoptosis, anti-miR-181a, which neutralizes endogenous miR-181a, was utilized. Delivery of anti-miR-181a significantly abrogated the H2O2-induced suppression of HK2 expression and subsequent disruption of mitochondrial membrane potential, improving the survival of cardiomyocytes exposed to H2O2. These findings suggest that miR-181a-mediated down-regulation of HK2 contributes to the apoptosis of cardiomyocytes exposed to ROS. Neutralizing miR-181a can be a viable and effective means to prevent cardiomyocyte from apoptosis in ischemic heart disease.
Collapse
Affiliation(s)
- Kwan Wook Kim
- Department of Medicine, The Graduate School, Yonsei University, Seoul, South Korea.,Department of Thoracic and Cardiovascular Surgery, CHA Bundang Medical Center, CHA University, Pangyo, South Korea
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, South Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, South Korea
| | - Kyung-Jong Yoo
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, South Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, South Korea
| |
Collapse
|
8
|
Varela R, Rauschert I, Romanelli G, Alberro A, Benech JC. Hyperglycemia and hyperlipidemia can induce morphophysiological changes in rat cardiac cell line. Biochem Biophys Rep 2021; 26:100983. [PMID: 33912691 PMCID: PMC8063753 DOI: 10.1016/j.bbrep.2021.100983] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/04/2021] [Accepted: 03/08/2021] [Indexed: 01/04/2023] Open
Abstract
H9c2 cardiac cells were incubated under the control condition and at different hyperglycemic and hyperlipidemic media, and the following parameters were determined and quantified: a) cell death, b) type of cell death, and c) changes in cell length, width and height. Of all the proven media, the one that showed the greatest differences compared to the control was the medium glucose (G) 33 mM + 500 μM palmitic acid. This condition was called the hyperglycemic and hyperlipidemic condition (HHC). Incubation of H9c2 cells in HHC promoted 5.2 times greater total cell death when compared to the control. Of the total death ofthe HHC cells, 38.6% was late apoptotic and 8.3% early apoptotic. HHC also changes cell morphology. The reordering of the actin cytoskeleton and cell stiffness was also studied in control and HHC cells. The actin cytoskeleton was quantified and the number and distance of actin bundles were not the same in the control as under HHC. Young's modulus images show a map of cell stiffness. Cells incubated in HHC with the reordered actin cytoskeleton were stiffer than those incubated in control. The region of greatest stiffness was the peripheral zone of HHC cells (where the number of actin bundles was higher and the distance between them smaller). Our results suggest a correlation between the reordering of the actin cytoskeleton and cell stiffness. Thus, our study showed that HHC can promote morphophysiological changes in rat cardiac cells confirming that gluco-and lipotoxicity may play a central role in the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Rocío Varela
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay
| | - Inés Rauschert
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay.,Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay
| | - Gerardo Romanelli
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay
| | - Andrés Alberro
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay
| | - Juan C Benech
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay.,Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Av. Italia, 3318, CP, 11600, Montevideo, Uruguay
| |
Collapse
|
9
|
A systematic review of post-translational modifications in the mitochondrial permeability transition pore complex associated with cardiac diseases. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165992. [PMID: 33091565 DOI: 10.1016/j.bbadis.2020.165992] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/08/2020] [Indexed: 12/28/2022]
Abstract
The mitochondrial permeability transition pore (mPTP) opening is involved in the pathophysiology of multiple cardiac diseases, such as ischemia/reperfusion injury and heart failure. A growing number of evidence provided by proteomic screening techniques has demonstrated the role of post-translational modifications (PTMs) in several key components of the pore in response to changes in the extra/intracellular environment and bioenergetic demand. This could lead to a fine, complex regulatory mechanism that, under pathological conditions, can shift the state of mitochondrial functions and, thus, the cell's fate. Understanding the complex relationship between these PTMs is still under investigation and can provide new, promising therapeutic targets and treatment approaches. This review, using a systematic review of the literature, presents the current knowledge on PTMs of the mPTP and their role in health and cardiac disease.
Collapse
|
10
|
Wang X, Tang S, Qin F, Liu Y, Liang Z, Cai H, Mo L, Xiao D, Guo S, Ouyang Y, Sun B, Lu C, Li X. Proteomics and phosphoproteomics study of LCMT1 overexpression and oxidative stress: overexpression of LCMT1 arrests H 2O 2-induced lose of cells viability. Redox Rep 2020; 24:1-9. [PMID: 30898057 PMCID: PMC6748586 DOI: 10.1080/13510002.2019.1595332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objectives: Protein phosphatase 2A (PP2A), a major serine/threonine
phosphatase, is also known to be a target of ROS. The methylation of PP2A can be
catalyzed by leucine carboxyl methyltransferase-1 (LCMT1), which regulates PP2A
activity and substrate specificity. Methods: In the previous study, we have showed that LCMT1-dependent
PP2Ac methylation arrests H2O2-induced cell oxidative
stress damage. To explore the possible protective mechanism, we performed
iTRAQ-based comparative quantitative proteomics and phosphoproteomics studies of
H2O2-treated vector control and LCMT1-overexpressing
cells. Results: A total of 4480 non-redundant proteins and 3801 unique
phosphopeptides were identified by this means. By comparing the
H2O2-regulated proteins in LCMT1-overexpressing and
vector control cells, we found that these differences were mainly related to
protein phosphorylation, gene expression, protein maturation, the cytoskeleton
and cell division. Further investigation of LCMT1 overexpression-specific
regulated proteins under H2O2 treatment supported the idea
that LCMT1 overexpression induced ageneral dephosphorylation of proteins and
indicated increased expression of non-erythrocytic hemoglobin, inactivation of
MAPK3 and regulation of proteins related to Rho signal transduction, which were
known to be linked to the regulation of the cytoskeleton. Discussion: These data provide proteomics and phosphoproteomics
insights into the association of LCMT1-dependent PP2Ac methylation and oxidative
stress and indirectly indicate that the methylation of PP2A plays an important
role against oxidative stress.
Collapse
Affiliation(s)
- Xinhang Wang
- a School of Preclinical Medicine , Guangxi Medical University , Nanning , People's Republic of China
| | - Shen Tang
- a School of Preclinical Medicine , Guangxi Medical University , Nanning , People's Republic of China
| | - Fu Qin
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Yuyang Liu
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Ziwei Liang
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Haiqing Cai
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Laiming Mo
- a School of Preclinical Medicine , Guangxi Medical University , Nanning , People's Republic of China
| | - Deqiang Xiao
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China
| | - Songcao Guo
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China
| | - Yiqiang Ouyang
- d Laboratory Animal Centre , Guangxi Medical University , Nanning , People's Republic of China
| | - Bin Sun
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Cailing Lu
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| | - Xiyi Li
- b School of Public Health, Guangxi Medical University , Nanning , People's Republic of China.,c Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases , Guangxi Medical University , Nanning , People's Republic of China
| |
Collapse
|
11
|
Maekawa S, Takada S, Nambu H, Furihata T, Kakutani N, Setoyama D, Ueyanagi Y, Kang D, Sabe H, Kinugawa S. Linoleic acid improves assembly of the CII subunit and CIII2/CIV complex of the mitochondrial oxidative phosphorylation system in heart failure. Cell Commun Signal 2019; 17:128. [PMID: 31619261 PMCID: PMC6796462 DOI: 10.1186/s12964-019-0445-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 09/24/2019] [Indexed: 12/20/2022] Open
Abstract
Background Linoleic acid is the major fatty acid moiety of cardiolipin, which is central to the assembly of components involved in mitochondrial oxidative phosphorylation (OXPHOS). Although linoleic acid is an essential nutrient, its excess intake is harmful to health. On the other hand, linoleic acid has been shown to prevent the reduction in cardiolipin content and to improve mitochondrial function in aged rats with spontaneous hypertensive heart failure (HF). In this study, we found that lower dietary intake of linoleic acid in HF patients statistically correlates with greater severity of HF, and we investigated the mechanisms therein involved. Methods HF patients, who were classified as New York Heart Association (NYHA) functional class I (n = 45), II (n = 93), and III (n = 15), were analyzed regarding their dietary intakes of different fatty acids during the one month prior to the study. Then, using a mouse model of HF, we confirmed reduced cardiolipin levels in their cardiac myocytes, and then analyzed the mechanisms by which dietary supplementation of linoleic acid improves cardiac malfunction of mitochondria. Results The dietary intake of linoleic acid was significantly lower in NYHA III patients, as compared to NYHA II patients. In HF model mice, both CI-based and CII-based OXPHOS activities were affected together with reduced cardiolipin levels. Silencing of CRLS1, which encodes cardiolipin synthetase, in cultured cardiomyocytes phenocopied these events. Feeding HF mice with linoleic acid improved both CI-based and CII-based respiration as well as left ventricular function, together with an increase in cardiolipin levels. However, although assembly of the respirasome (i.e., CI/CIII2/CIV complex), as well as assembly of CII subunits and the CIII2/CIV complex statistically correlated with cardiolipin levels in cultured cardiomyocytes, respirasome assembly was not notably restored by dietary linoleic acid in HF mice. Therefore, although linoleic acid may significantly improve both CI-based and CII-based respiration of cardiomyocytes, respirasomes impaired by HF were not easily repaired by the dietary intake of linoleic acid. Conclusions Dietary supplement of linoleic acid is beneficial for improving cardiac malfunction in HF, but is unable to completely cure HF.
Collapse
Affiliation(s)
- Satoshi Maekawa
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Shingo Takada
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan. .,Department of Molecular Biology, Hokkaido University Graduate School of Medicine, Sapporo, Japan. .,Faculty of Lifelong Sport, Department of Sports Education, Hokusho University, Ebetsu, Japan.
| | - Hideo Nambu
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Takaaki Furihata
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Naoya Kakutani
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan.,Research Fellow of the Japan Society for the Promotion of Science, Tokyo, Japan
| | - Daiki Setoyama
- Clinical Laboratories, Kyushu University Hospital, Fukuoka, Japan
| | - Yasushi Ueyanagi
- Clinical Laboratories, Kyushu University Hospital, Fukuoka, Japan.,Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dongchon Kang
- Clinical Laboratories, Kyushu University Hospital, Fukuoka, Japan.,Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hisataka Sabe
- Department of Molecular Biology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| |
Collapse
|
12
|
Guan W, Liu Y, Liu Y, Wang Q, Ye HL, Cheng YG, Kuang HX, Jiang XC, Yang BY. Proteomics Research on the Protective Effect of Mangiferin on H9C2 Cell Injury Induced by H 2O 2. Molecules 2019; 24:molecules24101911. [PMID: 31109015 PMCID: PMC6572523 DOI: 10.3390/molecules24101911] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/06/2019] [Accepted: 05/15/2019] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is one of the leading causes of morbidity and mortality worldwide. Mangiferin is a natural glucosylxanthone with antioxidant and anti-inflammatory properties, which has been confirmed to protect cardiac cells from myocardial infarction and myocardial ischemia reperfusion injury (MIRI); however, the underlying mechanism is still unclear. As oxidative stress is a major pathogenesis of MIRI, an H9C2 cell injury induced by hydrogen peroxide (H2O2) was established to simulate MIRI in vitro. Herein, the protective effect of mangiferin against MIRI was evaluated and the isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics was applied to explore the underlying molecular mechanism. In this research, mangiferin markedly ameliorated the oxidative imbalance by increasing the antioxidative capacity of the H9C2 cell. Moreover, proteomics analysis revealed that mangiferin pretreatment brought twenty differently-expressed proteins back to normal, most of which were related to glucose and fatty acid metabolism. Glycolysis, citrate cycle, and fatty acid degradation pathways were highlighted by Kyoto Encyclopedia of Gene and Genomes (KEGG) analysis. Western blot validation of six cardiac metabolism-related proteins were consistent with the proteomics analysis. Taken together, mangiferin protected the cardiomyocytes from MIRI by enhancing the antioxidant capacity and increasing the activities of glycolysis, citrate cycle, and fatty acid degradation pathways.
Collapse
Affiliation(s)
- Wei Guan
- Key Laboratory of Chinese Materia Medica, Ministry of Education of Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Yan Liu
- Key Laboratory of Chinese Materia Medica, Ministry of Education of Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Yuan Liu
- Key Laboratory of Chinese Materia Medica, Ministry of Education of Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Qi Wang
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150036, China.
| | - Hong-Liang Ye
- Key Laboratory of Chinese Materia Medica, Ministry of Education of Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Yan-Gang Cheng
- Key Laboratory of Chinese Materia Medica, Ministry of Education of Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Hai-Xue Kuang
- Key Laboratory of Chinese Materia Medica, Ministry of Education of Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Xi-Cheng Jiang
- Key Laboratory of Chinese Materia Medica, Ministry of Education of Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Bing-You Yang
- Key Laboratory of Chinese Materia Medica, Ministry of Education of Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
13
|
Zhang Y, Gao J, Sun W, Wen X, Xi Y, Wang Y, Wei C, Xu C, Li H. H 2S restores the cardioprotective effects of ischemic post-conditioning by upregulating HB-EGF/EGFR signaling. Aging (Albany NY) 2019; 11:1745-1758. [PMID: 30912763 PMCID: PMC6461169 DOI: 10.18632/aging.101866] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/07/2019] [Indexed: 04/08/2023]
Abstract
Hydrogen sulfide (H2S) reduces ischemia/reperfusion (I/R) injury and apoptosis and restores the cardioprotective effects of ischemic post-conditioning (PC) in aged cardiomyocytes by inhibiting oxidative stress and endoplasmic reticulum stress and increasing autophagy. However, the mechanism is unclear. In the present study, we observed a loss of PC-mediated cardioprotection of aged cardiomyocytes. NaHS (a H2S donor) exerted significant protective effects against H/R-induced cell damage, apoptosis, production of cleaved caspase-3 and caspase-9, and release of cytochrome c. NaHS also reversed the H/R-induced reduction in cell viability and increased HB-EGF expression, cellular HB-EGF content, and EGFR phosphorylation. Additionally, NaHS increased expression of Bcl-2, c-myc, c-fos and c-jun, and the phosphorylation of ERK1/2, PI3K, Akt and GSK-3β. PC alone did not provide protection to H/R-treated aged cardiomyocytes, but it was significantly restored by supplementation of NaHS. The beneficial effects of NaHS during PC were inhibited by EGFR knockdown, AG1478 (EGFR inhibitor), PD98059 (ERK1/2 inhibitor) or LY294002 (PI3K inhibitor). These results suggest that exogenous H2S restores PC-mediated cardioprotection by up-regulating HB-EGF/EGFR signaling, which activates the ERK1/2-c-myc (and fos and c-jun) and PI3K-Akt- GSK-3β pathways in the aged cardiomyocytes.
Collapse
Affiliation(s)
- Yuanzhou Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
- Equal contribution
| | - Jun Gao
- Department of Osteology, the First Hospital of Harbin, Harbin, China
- Equal contribution
| | - Weiming Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Xin Wen
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Yuxin Xi
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Yuehong Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Can Wei
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Changqing Xu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Hongzhu Li
- Department of Pathophysiology, Harbin Medical University, Harbin, China
- The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| |
Collapse
|
14
|
Zhang W, Hou X, Huang M, Zeng X, He X, Liao Y. TDCPP protects cardiomyocytes from H2O2-induced injuries via activating PI3K/Akt/GSK3β signaling pathway. Mol Cell Biochem 2018; 453:53-64. [DOI: 10.1007/s11010-018-3431-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/16/2018] [Indexed: 10/28/2022]
|
15
|
Proteomic analysis of honokiol-induced cytotoxicity in thyroid cancer cells. Life Sci 2018; 207:184-204. [PMID: 29883720 DOI: 10.1016/j.lfs.2018.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 05/24/2018] [Accepted: 06/02/2018] [Indexed: 12/16/2022]
Abstract
AIMS Honokiol is a natural product extracted from herbal plants such as the Magnolia species which have been shown to exhibit anti-tumor and anti-metastatic properties. However, the effects of honokiol on thyroid cancers are largely unknown. MATERIALS AND METHODS To determine whether honokiol might be useful for the treatment of thyroid cancer and to elucidate the mechanism of toxicity of honokiol, we analyzed the impact of honokiol treatment on differential protein expression in human thyroid cancer cell line ARO using lysine-labeling two-dimensional difference gel electrophoresis (2D-DIGE) combined with mass spectrometry (MS). KEY FINDINGS This study revealed 178 proteins that showed a significant change in expression levels and also revealed that honokiol-induced cytotoxicity in thyroid cancer cells involves dysregulation of cytoskeleton, protein folding, transcription control and glycolysis. SIGNIFICANCE Our work shows that combined proteomic strategy provides a rapid method to study the molecular mechanisms of honokiol-induced cytotoxicity in thyroid cancer cells. The identified targets may be useful for further evaluation as potential targets in thyroid cancer therapy.
Collapse
|
16
|
Hwang IC, Kim JY, Kim JH, Lee JE, Seo JY, Lee JW, Park J, Yang HM, Kim SH, Cho HJ, Kim HS. Therapeutic Potential of a Novel Necrosis Inhibitor, 7-Amino-Indole, in Myocardial Ischemia-Reperfusion Injury. Hypertension 2018; 71:1143-1155. [PMID: 29661840 PMCID: PMC5959205 DOI: 10.1161/hypertensionaha.117.09405] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 04/04/2017] [Accepted: 03/08/2018] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Opening of mitochondrial permeability transition pore and Ca2+ overload are main contributors to myocardial ischemia–reperfusion injury, which paradoxically causes a wide variety of myocardial damage. We investigated the protective role of a novel necrosis inhibitor (NecroX-7; NecX) against myocardial ischemia–reperfusion injury using in vitro and in vivo models. H9C2 rat cardiomyoblasts and neonatal cardiomyocytes were exposed to hypoxia–reoxygenation stress after pre-treatment with NecX, vitamin C, a combination of vitamin C and E, N-acetylcysteine, an apoptosis inhibitor (Z-VAD-fmk), or cyclosporine A. The main mechanism of cell death after hypoxia–reoxygenation stress was not apoptosis but necrosis, which was prevented by NecX. Protective effect of NecX was based on its potent reactive oxygen species scavenging activity, especially on mitochondrial reactive oxygen species. NecX preserved mitochondrial membrane potential through prevention of Ca2+ influx and inhibition of mitochondrial permeability transition pore opening, which was more potent than that by cyclosporine A. Using Sprague-Dawley rats exposed to myocardial ischemia for 45 minutes followed by reperfusion, we compared therapeutic efficacies of NecX with cyclosporine A, vitamin C, a combination of vitamin C and E, and 5% dextrose, each administered 5 minutes before reperfusion. NecX markedly inhibited myocardial necrosis and reduced fibrotic area to a greater extent than did cyclosporine A and other treated groups. In addition, NecX preserved systolic function and prevented pathological dilatory remodeling of left ventricle. The novel necrosis inhibitor has a significant protective effect against myocardial ischemia–reperfusion injury through inhibition of mitochondrial permeability transition pore opening, indicating that it is a promising candidate for cardioprotective adjunctive measure on top of reperfusion therapy.
Collapse
Affiliation(s)
- In-Chang Hwang
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.).,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Ju-Young Kim
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.).,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Ji-Hyun Kim
- National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.)
| | - Joo-Eun Lee
- National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Ji-Yun Seo
- National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Jae-Won Lee
- National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.)
| | - Jonghanne Park
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.).,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.)
| | - Han-Mo Yang
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.).,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Soon-Ha Kim
- R&D Campus, LG Chem/Ltd., Daejeon, Republic of Korea (S.-H.K.)
| | - Hyun-Jai Cho
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.).,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,Strategic Center of Cell and Bio Therapy for Heart, Diabetes, and Cancer, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-Y.K., J.-E.L., J.-W.L., H.-M.Y., H.-J.C., H.-S.K.)
| | - Hyo-Soo Kim
- From the Cardiovascular Center and Department of Internal Medicine, Seoul National University Hospital, Republic of Korea (I.-C.H., J.-H.K., J.P., H.-M.Y., H.-J.C., H.-S.K.) .,National Leading Laboratory for Cardiovascular Stem Cell, Seoul National University College of Medicine, Republic of Korea (I.-C.H., J.-Y.K., J.-H.K., J.-E.L., J.-Y.S., J.-W.L., J.P., H.-M.Y., H.-J.C., H.-S.K.).,and Department of Molecular Medicine and Biopharmaceutical Science, Seoul National University, Republic of Korea (H.-S.K.).,R&D Campus, LG Chem/Ltd., Daejeon, Republic of Korea (S.-H.K.)
| |
Collapse
|
17
|
Zhang YS, Tang LJ, Tu H, Wang SJ, Liu B, Zhang XJ, Li NS, Luo XJ, Peng J. Fasudil ameliorates the ischemia/reperfusion oxidative injury in rat hearts through suppression of myosin regulatory light chain/NADPH oxidase 2 pathway. Eur J Pharmacol 2018; 822:1-12. [PMID: 29337194 DOI: 10.1016/j.ejphar.2018.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 12/17/2017] [Accepted: 01/10/2018] [Indexed: 11/18/2022]
Abstract
Fasudil is a potent Rho-kinase (ROCK) inhibitor and can relax smooth muscle or cardiac muscle contraction through decreasing the phosphorylation level of myosin regulatory light chain (p-MLC20 or p-MLC2v), while p-MLC2v can function as a transcription factor to promote the NADPH oxidase 2 (NOX2) expression in rat hearts subjected to ischemia/reperfusion (I/R). This study aims to explore whether fasudil can protect the rat hearts against I/R oxidative injury through suppressing NOX2 expression via reduction of p-MLC2v level. The SD rat hearts were subjected to 1h-ischemia plus 3h-reperfusion, which showed myocardial injuries (myocardial fiber loss and disarray, increase of creatine kinase release and myocardial infarction/apoptosis), increase in ROCK activity and nuclear p-MLC2v level concomitant with up-regulation of NOX2 and H2O2 production; these phenomena were attenuated by fasudil in a dose-dependent manner. Next, we verified the cardioprotective effect of fasudil and the underlying mechanisms in hypoxia-reoxygenation (H/R) -treated H9c2 cells. Consistent with the results in vivo, the H/R-treated H9c2 cells showed cellular injury (increase in apoptotic ratio), elevation in ROCK activity and nuclear p-MLC2v level, accompanied by up-regulation of NOX2 and H2O2 production; these effects were blocked in the presence of fasudil in a dose-dependent way. Based on these observations, we conclude that beneficial effect of fasudil against myocardial I/R or H/R oxidative injury is related to the suppression of NOX2 expression through decrease of the p-MLC2v level. Our findings also highlight that intervention of MLC2v phosphorylation by drugs may provide a novel strategy to protect heart from I/R oxidative injury.
Collapse
Affiliation(s)
- Yi-Shuai Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Li-Jing Tang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Hua Tu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Shi-Jing Wang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Bin Liu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xiao-Jie Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Nian-Sheng Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| |
Collapse
|
18
|
Huang J, Liu Z, Xu P, Zhang Z, Yin D, Liu J, He H, He M. Capsaicin prevents mitochondrial damage, protects cardiomyocytes subjected to anoxia/reoxygenation injury mediated by 14-3-3η/Bcl-2. Eur J Pharmacol 2018; 819:43-50. [DOI: 10.1016/j.ejphar.2017.11.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 11/07/2017] [Accepted: 11/16/2017] [Indexed: 10/18/2022]
|
19
|
Kankeu C, Clarke K, Van Haver D, Gevaert K, Impens F, Dittrich A, Roderick HL, Passante E, Huber HJ. Quantitative proteomics and systems analysis of cultured H9C2 cardiomyoblasts during differentiation over time supports a 'function follows form' model of differentiation. Mol Omics 2018; 14:181-196. [PMID: 29770421 DOI: 10.1039/c8mo00036k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The rat cardiomyoblast cell line H9C2 has emerged as a valuable tool for studying cardiac development, mechanisms of disease and toxicology. We present here a rigorous proteomic analysis that monitored the changes in protein expression during differentiation of H9C2 cells into cardiomyocyte-like cells over time. Quantitative mass spectrometry followed by gene ontology (GO) enrichment analysis revealed that early changes in H9C2 differentiation are related to protein pathways of cardiac muscle morphogenesis and sphingolipid synthesis. These changes in the proteome were followed later in the differentiation time-course by alterations in the expression of proteins involved in cation transport and beta-oxidation. Studying the temporal profile of the H9C2 proteome during differentiation in further detail revealed eight clusters of co-regulated proteins that can be associated with early, late, continuous and transient up- and downregulation. Subsequent reactome pathway analysis based on these eight clusters further corroborated and detailed the results of the GO analysis. Specifically, this analysis confirmed that proteins related to pathways in muscle contraction are upregulated early and transiently, and proteins relevant to extracellular matrix organization are downregulated early. In contrast, upregulation of proteins related to cardiac metabolism occurs at later time points. Finally, independent validation of the proteomics results by immunoblotting confirmed hereto unknown regulators of cardiac structure and ionic metabolism. Our results are consistent with a 'function follows form' model of differentiation, whereby early and transient alterations of structural proteins enable subsequent changes that are relevant to the characteristic physiology of cardiomyocytes.
Collapse
Affiliation(s)
- Cynthia Kankeu
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Ledee D, Kang MA, Kajimoto M, Purvine S, Brewer H, Pasa-Tolic L, Portman MA. Quantitative cardiac phosphoproteomics profiling during ischemia-reperfusion in an immature swine model. Am J Physiol Heart Circ Physiol 2017; 313:H125-H137. [PMID: 28455290 PMCID: PMC5538860 DOI: 10.1152/ajpheart.00842.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 01/26/2023]
Abstract
Ischemia-reperfusion (I/R) results in altered metabolic and molecular responses, and phosphorylation is one of the most noted regulatory mechanisms mediating signaling mechanisms during physiological stresses. To expand our knowledge of the potential phosphoproteomic changes in the myocardium during I/R, we used Isobaric Tags for Relative and Absolute Quantitation-based analyses in left ventricular samples obtained from porcine hearts under control or I/R conditions. The data are available via ProteomeXchange with identifier PXD006066. We identified 1,896 phosphopeptides within left ventricular control and I/R porcine samples. Significant differential phosphorylation between control and I/R groups was discovered in 111 phosphopeptides from 86 proteins. Analysis of the phosphopeptides using Motif-x identified five motifs: (..R..S..), (..SP..), (..S.S..), (..S…S..), and (..S.T..). Semiquantitative immunoblots confirmed site location and directional changes in phosphorylation for phospholamban and pyruvate dehydrogenase E1, two proteins known to be altered by I/R and identified by this study. Novel phosphorylation sites associated with I/R were also identified. Functional characterization of the phosphopeptides identified by our methodology could expand our understanding of the signaling mechanisms involved during I/R damage in the heart as well as identify new areas to target therapeutic strategies.NEW & NOTEWORTHY We used Isobaric Tags for Relative and Absolute Quantitation technology to investigate the phosphoproteomic changes that occur in cardiac tissue under ischemia-reperfusion conditions. The results of this study provide an extensive catalog of phosphoproteins, both predicted and novel, associated with ischemia-reperfusion, thereby identifying new pathways for investigation.
Collapse
Affiliation(s)
- Dolena Ledee
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, Washington
- Division of Cardiology, Department of Pediatrics, University of Washington, Seattle, Washington
| | - Min A Kang
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, Washington
| | - Masaki Kajimoto
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, Washington
| | - Samuel Purvine
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington; and
| | - Heather Brewer
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington; and
| | - Ljiljana Pasa-Tolic
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington; and
| | - Michael A Portman
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, Washington;
- Division of Cardiology, Department of Pediatrics, University of Washington, Seattle, Washington
| |
Collapse
|
21
|
Li F, Zhang Y, Zeng D, Xia Y, Fan X, Tan Y, Kou J, Yu B. The Combination of Three Components Derived from Sheng MaiSan Protects Myocardial Ischemic Diseases and Inhibits Oxidative Stress via Modulating MAPKs and JAK2-STAT3 Signaling Pathways Based on Bioinformatics Approach. Front Pharmacol 2017; 8:21. [PMID: 28197101 PMCID: PMC5282471 DOI: 10.3389/fphar.2017.00021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/11/2017] [Indexed: 01/25/2023] Open
Abstract
GRS is a drug combination of three components including ginsenoside Rb1, ruscogenin and schisandrin. It derived from the well-known TCM formula Sheng MaiSan, a widely used traditional Chinese medicine for the treatment of cardiovascular diseases in clinic. The present study illuminates its underlying mechanisms against myocardial ischemic diseases based on the combined methods of bioinformatic prediction and experimental verification. A protein database was established through constructing the drug-protein network. And the target-pathway interaction network clustered the potential signaling pathways and targets of GRS in treatment of myocardial ischemic diseases. Several target proteins, such as NFKB1, STAT3 and MAPK14, were identified as the candidate key proteins, and MAPKs and JAK-STAT signaling pathway were suggested as the most related pathways, which were in accordance with the gene ontology analysis. Then, the predictive results were further validated and we found that GRS treatment alleviated hypoxia/reoxygenation (H/R)-induced cardiomyocytes injury via suppression of MDA levels and ROS generation, and potential mechanisms might related to the suppression of activation of MAPKs and JAK2-STAT3 signaling pathways. Conclusively, our results offer the evidence that GRS attenuates myocardial ischemia injury via regulating oxidative stress and MAPKs and JAK2-STAT3 signaling pathways, which supplied some new insights for its prevention and treatment of myocardial ischemia diseases.
Collapse
Affiliation(s)
- Fang Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of Traditional Chinese Medicine, China Pharmaceutical University Nanjing, China
| | - Yu Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of Traditional Chinese Medicine, China Pharmaceutical University Nanjing, China
| | - Donglin Zeng
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of Traditional Chinese Medicine, China Pharmaceutical University Nanjing, China
| | - Yu Xia
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of Traditional Chinese Medicine, China Pharmaceutical University Nanjing, China
| | - Xiaoxue Fan
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of Traditional Chinese Medicine, China Pharmaceutical University Nanjing, China
| | - Yisha Tan
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of Traditional Chinese Medicine, China Pharmaceutical University Nanjing, China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of Traditional Chinese Medicine, China Pharmaceutical University Nanjing, China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of Traditional Chinese Medicine, China Pharmaceutical University Nanjing, China
| |
Collapse
|
22
|
Yang CS, Wei YS, Tsai HL, Cheong IS, Chang SJ, Chou HC, Lee YR, Chan HL. Proteomic analysis of prognostic plasma biomarkers in peripheral arterial occlusive disease. MOLECULAR BIOSYSTEMS 2017; 13:1297-1303. [DOI: 10.1039/c7mb00229g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A comprehensive patient-based plasma proteomic approach for the identification of potential plasma biomarkers for the screening and detection of good/poor prognosis of peripheral arterial occlusive disease (PAOD).
Collapse
Affiliation(s)
- Cheng-San Yang
- Division of Plastic Surgery
- Department of Surgery
- Ditmanson Medical Foundation Chia-Yi Christian Hospital
- Chiayi
- Taiwan
| | - Yu-Shan Wei
- Institute of Bioinformatics and Structural Biology and Department of Medical Science
- National Tsing Hua University
- Hsinchu
- Taiwan
| | - Han-Lin Tsai
- Division of Cardiology
- Department of Internal Medicine
- Ditmanson Medical Foundation Chia-Yi Christian Hospital
- Chiayi
- Taiwan
| | - Ian-Seng Cheong
- Divisions of Urology
- Department of Surgery
- Ditmanson Medical Foundation Chia-Yi Christian Hospital
- Chiayi
- Taiwan
| | - Shing-Jyh Chang
- Gynecologic Oncology Section
- Department of Obstetrics and Gynecology
- Hsinchu Mackay Memorial Hospital
- Hsinchu
- Taiwan
| | - Hsiu-Chuan Chou
- Department of Biomedical Engineering and Environmental Sciences
- National Tsing Hua University
- Hsinchu
- Taiwan
| | - Ying-Ray Lee
- Department of Medical Research
- Ditmanson Medical Foundation Chia-Yi Christian Hospital
- Chiayi
- Taiwan
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology and Department of Medical Science
- National Tsing Hua University
- Hsinchu
- Taiwan
| |
Collapse
|
23
|
Abstract
Ischemic disorders, such as myocardial infarction, stroke, and peripheral vascular disease, are the most common causes of debilitating disease and death in westernized cultures. The extent of tissue injury relates directly to the extent of blood flow reduction and to the length of the ischemic period, which influence the levels to which cellular ATP and intracellular pH are reduced. By impairing ATPase-dependent ion transport, ischemia causes intracellular and mitochondrial calcium levels to increase (calcium overload). Cell volume regulatory mechanisms are also disrupted by the lack of ATP, which can induce lysis of organelle and plasma membranes. Reperfusion, although required to salvage oxygen-starved tissues, produces paradoxical tissue responses that fuel the production of reactive oxygen species (oxygen paradox), sequestration of proinflammatory immunocytes in ischemic tissues, endoplasmic reticulum stress, and development of postischemic capillary no-reflow, which amplify tissue injury. These pathologic events culminate in opening of mitochondrial permeability transition pores as a common end-effector of ischemia/reperfusion (I/R)-induced cell lysis and death. Emerging concepts include the influence of the intestinal microbiome, fetal programming, epigenetic changes, and microparticles in the pathogenesis of I/R. The overall goal of this review is to describe these and other mechanisms that contribute to I/R injury. Because so many different deleterious events participate in I/R, it is clear that therapeutic approaches will be effective only when multiple pathologic processes are targeted. In addition, the translational significance of I/R research will be enhanced by much wider use of animal models that incorporate the complicating effects of risk factors for cardiovascular disease. © 2017 American Physiological Society. Compr Physiol 7:113-170, 2017.
Collapse
Affiliation(s)
- Theodore Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Christopher P. Baines
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Ronald J. Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
24
|
Lindoso RS, Sandim V, Collino F, Carvalho AB, Dias J, da Costa MR, Zingali RB, Vieyra A. Proteomics of cell-cell interactions in health and disease. Proteomics 2015; 16:328-44. [PMID: 26552723 DOI: 10.1002/pmic.201500341] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 09/29/2015] [Accepted: 11/02/2015] [Indexed: 12/11/2022]
Abstract
The mechanisms of cell-cell communications are now under intense study by proteomic approaches. Proteomics has unraveled changes in protein profiling as the result of cell interactions mediated by ligand/receptor, hormones, soluble factors, and the content of extracellular vesicles. Besides being a brief overview of the main and profitable methodologies now available (evaluating theory behind the methods, their usefulness, and pitfalls), this review focuses on-from a proteome perspective-some signaling pathways and post-translational modifications (PTMs), which are essential for understanding ischemic lesions and their recovery in two vital organs in mammals, the heart, and the kidney. Knowledge of misdirection of the proteome during tissue recovery, such as represented by the convergence between fibrosis and cancer, emerges as an important tool in prognosis. Proteomics of cell-cell interaction is also especially useful for understanding how stem cells interact in injured tissues, anticipating clues for rational therapeutic interventions. In the effervescent field of induced pluripotency and cell reprogramming, proteomic studies have shown what proteins from specialized cells contribute to the recovery of infarcted tissues. Overall, we conclude that proteomics is at the forefront in helping us to understand the mechanisms that underpin prevalent pathological processes.
Collapse
Affiliation(s)
- Rafael S Lindoso
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, RJ, Brazil
| | - Vanessa Sandim
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, RJ, Brazil.,Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Federica Collino
- Department of Medical Sciences and Molecular Biotechnology Center, University of Turin, Turin, Italy.,Translational Center of Regenerative Medicine, University of Turin/Fresenius Medical Care, Turin, Italy
| | - Adriana B Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, RJ, Brazil
| | - Juliana Dias
- National Institute of Cancer, Rio de Janeiro, RJ, Brazil
| | - Milene R da Costa
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Russolina B Zingali
- National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, RJ, Brazil.,Leopoldo de Meis Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Proteomic Network of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Adalberto Vieyra
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,National Institute of Science and Technology for Structural Biology and Bioimaging, Rio de Janeiro, RJ, Brazil.,Translational Biomedicine Graduate Program, Grand Rio University, Duque de Caxias, RJ, Brazil
| |
Collapse
|
25
|
Qian L, Shi J, Zhang C, Lu J, Lu X, Wu K, Yang C, Yan D, Zhang C, You Q, Liu X. Downregulation of RACK1 is associated with cardiomyocyte apoptosis after myocardial ischemia/reperfusion injury in adult rats. In Vitro Cell Dev Biol Anim 2015; 52:305-313. [PMID: 26659395 DOI: 10.1007/s11626-015-9981-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 11/09/2015] [Indexed: 12/12/2022]
Abstract
The receptor for activated C kinase 1 (RACK1) is a multifaceted scaffolding protein that mediates the shuttling of activated protein kinase C (PKC) to cellular membranes. In addition, RACK1 could decrease cell apoptosis in a variety of disease models. However, the function of RACK1 in cardiomyocyte apoptosis after myocardial ischemia/reperfusion (I/R) is unknown. In this study, male Sprague-Dawley rats were anesthetized and subjected to myocardial I/R insult consisting of 30 min left anterior descending coronary artery (LAD) occlusion followed by reperfusion for 1, 2, 4, 6, 8, 12, and 24 h. The expression of RACK1 was decreased after myocardial I/R and was associated with cardiomyocyte apoptosis. To further verify the relationship between RACK1 and cardiomyocyte apoptosis, H9c2 cardiomyocytes were cultured under hypoxia for 6 h, then maintained in the regular incubator to reoxygenation. After H9c2 cells were transfected with Flag-RACK1 to overexpress RACK1, RACK1 expression was upregulated in hypoxia/reoxygenation (H/R) 4 h group accompanied with the decrease of cleaved caspase-3 and the increase of Bcl-2 expression. Terminal transferase-mediated biotin dUTP nick end labeling (TUNEL) assay showed that RACK1 overexpression inhibited H9c2 cell apoptosis induced by H/R treatment. Our data suggested that RACK1 might suppress cardiomyocyte apoptosis after I/R, providing a novel molecular target for the therapy of ischemia heart disease.
Collapse
Affiliation(s)
- Long Qian
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Jiahai Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Chi Zhang
- Department of Vasculocardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Jiawei Lu
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Xiaoning Lu
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Kunpeng Wu
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Chen Yang
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Daliang Yan
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Chao Zhang
- Department of Vasculocardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Qingsheng You
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, People's Republic of China. .,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.
| | - Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China. .,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.
| |
Collapse
|
26
|
Lenčo J, Lenčová-Popelová O, Link M, Jirkovská A, Tambor V, Potůčková E, Stulík J, Šimůnek T, Štěrba M. Proteomic investigation of embryonic rat heart-derived H9c2 cell line sheds new light on the molecular phenotype of the popular cell model. Exp Cell Res 2015; 339:174-86. [DOI: 10.1016/j.yexcr.2015.10.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 10/20/2015] [Accepted: 10/23/2015] [Indexed: 01/15/2023]
|
27
|
Zhao Y, Yang C, Organ C, Li Z, Bhushan S, Otsuka H, Pacheco A, Kang J, Aguilar HC, Lefer DJ, Xian M. Design, Synthesis, and Cardioprotective Effects of N-Mercapto-Based Hydrogen Sulfide Donors. J Med Chem 2015; 58:7501-11. [PMID: 26317692 PMCID: PMC4766970 DOI: 10.1021/acs.jmedchem.5b01033] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Hydrogen sulfide (H2S) is a signaling molecule which plays regulatory roles in many physiological and/or pathological processes. Therefore, regulation of H2S levels could have great potential therapeutic value. In this work, we report the design, synthesis, and evaluation of a class of N-mercapto (N-SH)-based H2S donors. Thirty-three donors were synthesized and tested. Our results indicated that controllable H2S release from these donors could be achieved upon structural modifications. Selected donors (NSHD-1, NSHD-2, and NSHD-6) were tested in cellular models of oxidative damage and showed significant cytoprotective effects. Moreover, NSHD-1 and NSHD-2 were also found to exhibit potent protective effects in a murine model of myocardial ischemia reperfusion (MI/R) injury.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Chemistry, Washington State University , Pullman, Washington 99164, United States
| | - Chuntao Yang
- Department of Physiology, Guangzhou Medical University , Guangzhou 511436, China
| | - Chelsea Organ
- Cardiovascular Center of Excellence, Louisiana State University Health Science Center , New Orleans, Louisiana 70112, United States
| | - Zhen Li
- Cardiovascular Center of Excellence, Louisiana State University Health Science Center , New Orleans, Louisiana 70112, United States
| | - Shashi Bhushan
- Cardiovascular Center of Excellence, Louisiana State University Health Science Center , New Orleans, Louisiana 70112, United States
| | - Hiro Otsuka
- Cardiovascular Center of Excellence, Louisiana State University Health Science Center , New Orleans, Louisiana 70112, United States
| | - Armando Pacheco
- Department of Chemistry, Washington State University , Pullman, Washington 99164, United States
| | - Jianming Kang
- Department of Chemistry, Washington State University , Pullman, Washington 99164, United States
| | - Hector C Aguilar
- Paul G. Allen School for Global Animal Health, Washington State University , Pullman, Washington 99164, United States
| | - David J Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Science Center , New Orleans, Louisiana 70112, United States
| | - Ming Xian
- Department of Chemistry, Washington State University , Pullman, Washington 99164, United States
| |
Collapse
|
28
|
Huang HJ, Lin CC, Chou HC, Chen YW, Lin ST, Lin YC, Lin DY, Lyu KW, Chan HL. Proteomic analysis of rhein-induced cyt: ER stress mediates cell death in breast cancer cells. MOLECULAR BIOSYSTEMS 2015; 10:3086-100. [PMID: 25259860 DOI: 10.1039/c4mb00451e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Rhein is a natural product purified from herbal plants such as Rheum palmatum, which has been shown to have anti-angiogenesis and anti-tumor metastasis properties. However, the biological effects of rhein on the behavior of breast cancers are not completely elucidated. To evaluate whether rhein might be useful in the treatment of breast cancer and its cytotoxic mechanism, we analyzed the impact of rhein treatment on differential protein expression as well as redox regulation in a non-invasive breast cancer cell line, MCF-7, and an invasive breast cancer cell line, MDA-MB-231, using lysine- and cysteine-labeling two-dimensional difference gel electrophoresis (2D-DIGE) combined with MALDI-TOF/TOF mass spectrometry. This proteomic study revealed that 73 proteins were significantly changed in protein expression; while 9 proteins were significantly altered in thiol reactivity in both MCF-7 and MDA-MB-231 cells. The results also demonstrated that rhein-induced cytotoxicity in breast cancer cells mostly involves dysregulation of cytoskeleton regulation, protein folding, the glycolysis pathway and transcription control. A further study also indicated that rhein promotes misfolding of cellular proteins as well as unbalancing of the cellular redox status leading to ER-stress. Our work shows that the current proteomic strategy offers a high-through-put platform to study the molecular mechanisms of rhein-induced cytotoxicity in breast cancer cells. The identified differentially expressed proteins might be further evaluated as potential targets in breast cancer therapy.
Collapse
Affiliation(s)
- Hui-Ju Huang
- Institute of Bioinformatics and Structural Biology & Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kuo CY, Chiu YC, Lee AYL, Hwang TL. Mitochondrial Lon protease controls ROS-dependent apoptosis in cardiomyocyte under hypoxia. Mitochondrion 2015; 23:7-16. [PMID: 25922169 DOI: 10.1016/j.mito.2015.04.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 04/14/2015] [Accepted: 04/21/2015] [Indexed: 11/30/2022]
Abstract
Apoptosis of cardiomyocytes, under ischemic conditions, has been identified as an essential process in the progression of heart failure. Under hypoxic conditions, mitochondria can become a threat to the cell because of their capacity to generate reactive oxygen species (ROS). As ROS appear to have a critical role in heart failure, there has been considerable interest in identifying the candidate proteins involved and in developing strategies to reduce oxidative stress. Lon protease (Lon) is a multifunctional protein that mediates protein quality control and stress response in mitochondria. However, comprehensive and detailed studies, on the role of Lon in hypoxia-induced cardiomyocyte apoptosis, have yet to be carried out. In the present study, we demonstrated that hypoxia induced ROS-dependent cardiomyocyte apoptosis. Lon was upregulated in hypoxia-induced cardiomyocytes. Lon downregulation attenuated hypoxia-induced cardiomyocyte apoptosis through a reduction of ROS level. Moreover, overexpression of Lon stimulated ROS production and induced apoptosis under normoxic conditions in cardiomyocytes. Our results identify Lon as a novel regulator of cardiomyocyte fate and offers exciting new insights into the therapeutic potential of hypoxia-induced cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Chan-Yen Kuo
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, and Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan 33302, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Yi-Chieh Chiu
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Alan Yueh-Luen Lee
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, and Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan 33302, Taiwan; Department of Cosmetic Science and Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan; Immunology Consortium, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan.
| |
Collapse
|
30
|
Li M, Tang YQ, Du RH, Shi FH, Hussein HK, Dai DZ, Dai Y. CPUY11018, an azimilide derivative, ameliorates isoproterenol-induced cardiac insufficiency through relieving dysfunctional mitochondria and endoplasmic reticulum. J Pharm Pharmacol 2015; 67:1029-41. [DOI: 10.1111/jphp.12401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 01/18/2015] [Indexed: 12/11/2022]
Abstract
Abstract
Objectives
Deterioration of cardiac performance under stress may be partly mediated by dysfunctional mitochondria and endoplasmic reticulum (ER) that is likely related to an activation of NADPH oxidase (NOX) and an increase in pro-inflammatory factors. We investigated if a new compound CPUY11018 (CPUY) derived from Azimilide could ameliorate the stress impaired cardiac performance.
Methods
Forty-eight male Sprague Dawley rats were randomly divided into six groups and were injected with isoproterenol (ISO, 1 ml/kg, s.c.) for 10 days. Cardiac myocytes and fibroblasts from neonate rats were incubated with ISO. CPUY was employed and compared with apocynin (APO) – an inhibitor of NOX.
Key findings
In ISO-treated group, the compromised haemodynamics and cardiac remodelling were significant with dysfunctional mitochondria indicated by decreased MnSOD and mitochondrial membrane potential, and an enhanced reactive oxygen species genesis. Downregulation of FKBP12.6, CASQ2 and SERCA2a was also remarkable in vivo and in vitro implying an abnormal ER. Upregulated Nox4, p22phox and p47phox were significant, associated with upregulation of Src, IκBβ and NFκB, and downregulation of pAMPK/AMPK and Cx40 in vivo and in vitro. These abnormalities were relieved by CPUY and APO.
Conclusions
CPUY is potential in managing cardiac insufficiency through normalizing mitochondria and ER in the affected heart.
Collapse
Affiliation(s)
- Meng Li
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Yi-Qun Tang
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Rong-Hui Du
- Department of Pharmacology, The Medical School, University of Nanjing, Nanjing, China
| | - Fang-Hong Shi
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Humed Khan Hussein
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
- Pharmacology Department, Faculty of Pharmacy, University of Aden, Aden, Yemen
| | - De-Zai Dai
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Yin Dai
- Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
31
|
Wang SH, Lee WC, Chou HC. Retinal proteins associated with redox regulation and protein folding play central roles in response to high glucose conditions. Electrophoresis 2015; 36:902-9. [DOI: 10.1002/elps.201400591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/11/2014] [Accepted: 12/17/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Ssu-Han Wang
- Department of Applied Science; National Hsinchu University of Education; Hsinchu Taiwan
| | - Wen-Chi Lee
- Department of Applied Science; National Hsinchu University of Education; Hsinchu Taiwan
| | - Hsiu-Chuan Chou
- Department of Applied Science; National Hsinchu University of Education; Hsinchu Taiwan
| |
Collapse
|
32
|
Feng CL, Chou HC. Hyperglycemia initiates N-cadherin rearrangement and diabetic monocytes promote inflammatory responses in human microvascular endothelial cells. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.bgm.2014.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Hsieh SR, Cheng WC, Su YM, Chiu CH, Liou YM. Molecular targets for anti-oxidative protection of green tea polyphenols against myocardial ischemic injury. Biomedicine (Taipei) 2014; 4:23. [PMID: 25520936 PMCID: PMC4264984 DOI: 10.7603/s40681-014-0023-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/14/2014] [Indexed: 12/20/2022] Open
Abstract
Ischemic heart disease is the leading cause of death worldwide. An improved understanding of the mechanisms involved in myocardial injury would allow intervention downstream in the pathway where certain drugs including natural products could be efficiently applied to target the end effectors of the cell death pathway. Green tea polyphenols (GTPs) have potent anti-oxidative capabilities, which may account for their beneficial effects in preventing oxidative stress associated with ischemia injury. Although studies have provided convincing evidence to support the protective effects of GTPs in cardiovascular system, the potential end effectors that mediate cardiac protection are only beginning to be addressed. Proteomics analyses widely used to identify the protein targets for many cardiovascular diseases have advanced the discovery of the signaling mechanism for GTPs-mediated cardio-protection. This review focuses on putative triggers, mediators, and end effectors for the GTPs-mediated cardio-protection signaling pathways engaged in myocardial ischemia crisis, allowing a promising natural product to be used for ameliorating oxidative stress associated with ischemic heart diseases.
Collapse
Affiliation(s)
- Shih-Rong Hsieh
- Department of Cardiovascular Surgery, Taichung Veterans General Hospital, 407 Taichung, Taiwan
| | - Wei-Chen Cheng
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, 300 Hsinchu, Taiwan
| | - Yi-Min Su
- Department of Life Sciences, National Chung-Hsing University, 402 No. 250, Kuokang Road, Taichung, Taiwan
| | - Chun-Hwei Chiu
- Department of Life Sciences, National Chung-Hsing University, 402 No. 250, Kuokang Road, Taichung, Taiwan
| | - Ying-Ming Liou
- Department of Life Sciences, National Chung-Hsing University, 402 No. 250, Kuokang Road, Taichung, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, 402 Taichung, Taiwan
| |
Collapse
|
34
|
Mitra A, Basak T, Ahmad S, Datta K, Datta R, Sengupta S, Sarkar S. Comparative Proteome Profiling during Cardiac Hypertrophy and Myocardial Infarction Reveals Altered Glucose Oxidation by Differential Activation of Pyruvate Dehydrogenase E1 Component Subunit β. J Mol Biol 2014; 427:2104-20. [PMID: 25451023 DOI: 10.1016/j.jmb.2014.10.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 10/09/2014] [Accepted: 10/29/2014] [Indexed: 12/19/2022]
Abstract
Cardiac hypertrophy and myocardial infarction (MI) are two etiologically different disease forms with varied pathological characteristics. However, the precise molecular mechanisms and specific causal proteins associated with these diseases are obscure to date. In this study, a comparative cardiac proteome profiling was performed in Wistar rat models for diseased and control (sham) groups using two-dimensional difference gel electrophoresis followed by matrix-assisted laser desorption/ionization tandem time-of-flight mass spectrometry. Proteins were identified using Protein Pilot™ software (version 4.0) and were subjected to stringent statistical analysis. Alteration of key proteins was validated by Western blot analysis. The differentially expressed protein sets identified in this study were associated with different functional groups, involving various metabolic pathways, stress responses, cytoskeletal organization, apoptotic signaling and other miscellaneous functions. It was further deciphered that altered energy metabolism during hypertrophy in comparison to MI may be predominantly attributed to induced glucose oxidation level, via reduced phosphorylation of pyruvate dehydrogenase E1 component subunit β (PDHE1-B) protein during hypertrophy. This study reports for the first time the global changes in rat cardiac proteome during two etiologically different cardiac diseases and identifies key signaling regulators modulating ontogeny of these two diseases culminating in heart failure. This study also pointed toward differential activation of PDHE1-B that accounts for upregulation of glucose oxidation during hypertrophy. Downstream analysis of altered proteome and the associated modulators would enhance our present knowledge regarding altered pathophysiology of these two etiologically different cardiac disease forms.
Collapse
Affiliation(s)
- Arkadeep Mitra
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700 019, India
| | - Trayambak Basak
- Genomics and Molecular Medicine Unit, CSIR Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110 020, India
| | - Shadab Ahmad
- Genomics and Molecular Medicine Unit, CSIR Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110 020, India
| | - Kaberi Datta
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700 019, India
| | - Ritwik Datta
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700 019, India
| | - Shantanu Sengupta
- Genomics and Molecular Medicine Unit, CSIR Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110 020, India
| | - Sagartirtha Sarkar
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700 019, India.
| |
Collapse
|
35
|
Yang YC, Wang XD, Huang K, Wang L, Jiang ZL, Qi YX. Temporal phosphoproteomics to investigate the mechanotransduction of vascular smooth muscle cells in response to cyclic stretch. J Biomech 2014; 47:3622-9. [DOI: 10.1016/j.jbiomech.2014.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 09/27/2014] [Accepted: 10/05/2014] [Indexed: 12/28/2022]
|
36
|
Keilhoff G, Lucas B, Pinkernelle J, Steiner M, Fansa H. Effects of cerebrolysin on motor-neuron-like NSC-34 cells. Exp Cell Res 2014; 327:234-55. [PMID: 24997385 DOI: 10.1016/j.yexcr.2014.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/12/2014] [Accepted: 06/26/2014] [Indexed: 01/01/2023]
Abstract
Although the peripheral nervous system is capable of regeneration, this capability is limited. As a potential means of augmenting nerve regeneration, the effects of cerebrolysin (CL)--a proteolytic peptide fraction--were tested in vitro on the motor-neuron-like NSC-34 cell line and organotypic spinal cord cultures. Therefore, NSC-34 cells were subjected to mechanical stress by changing media and metabolic stress by oxygen glucose deprivation. Afterwards, cell survival/proliferation using MTT and BrdU-labeling (FACS) and neurite sprouting using ImageJ analysis were evaluated. Calpain-1, Src and α-spectrin protein expression were analyzed by Western blot. In organotypic cultures, the effect of CL on motor neuron survival and neurite sprouting was tested by immunohistochemistry. CL had a temporary anti-proliferative but initially neuroprotective effect on OGD-stressed NSC-34 cells. High-dosed or repeatedly applied CL was deleterious for cell survival. CL amplified neurite reconstruction to limited extent, affected calpain-1 protein expression and influenced calpain-mediated spectrin cleavage as a function of Src expression. In organotypic spinal cord slice cultures, CL was not able to support motor neuron survival/neurite sprouting. Moreover, it hampered astroglia and microglia activities. The data suggest that CL may have only isolated positive effects on injured spinal motor neurons. High-dosed or accumulated CL seemed to have adverse effects in treatment of spinal cord injury. Further experiments are required to optimize the conditions for a safe clinical administration of CL in spinal cord injuries.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Benjamin Lucas
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Josephine Pinkernelle
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Michael Steiner
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Hisham Fansa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, Klinikum Bielefeld, Teutoburger Str. 50, D-33604 Bielefeld, Germany
| |
Collapse
|
37
|
Chen WC, Hsieh SR, Chiu CH, Hsu BD, Liou YM. Molecular identification for epigallocatechin-3-gallate-mediated antioxidant intervention on the H2O2-induced oxidative stress in H9c2 rat cardiomyoblasts. J Biomed Sci 2014; 21:56. [PMID: 24913014 PMCID: PMC4070642 DOI: 10.1186/1423-0127-21-56] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 06/03/2014] [Indexed: 11/10/2022] Open
Abstract
Background Epigallocatechin-3-gallate (EGCG) has been documented for its beneficial effects protecting oxidative stress to cardiac cells. Previously, we have shown the EGCG-mediated cardiac protection by attenuating reactive oxygen species and cytosolic Ca2+ in cardiac cells during oxidative stress and myocardial ischemia. Here, we aimed to seek a deeper elucidation of the molecular anti-oxidative capabilities of EGCG in an H2O2-induced oxidative stress model of myocardial ischemia injury using H9c2 rat cardiomyoblasts. Results Proteomics analysis was used to determine the differential expression of proteins in H9c2 cells cultured in the conditions of control, 400 μM H2O2 exposure for 30 min with and/or without 10 to 20 μM EGCG pre-treatment. In this model, eight proteins associated with energy metabolism, mitochondrial electron transfer, redox regulation, signal transduction, and RNA binding were identified to take part in EGCG-ameliorating H2O2-induced injury in H9c2 cells. H2O2 exposure increased oxidative stress evidenced by increases in reactive oxygen species and cytosolic Ca2+ overload, increases in glycolytic protein, α-enolase, decreases in antioxidant protein, peroxiredoxin-4, as well as decreases in mitochondrial proteins, including aldehyde dehydrogenase-2, ornithine aminotransferase, and succinate dehydrogenase ubiquinone flavoprotein subunit. All of these effects were reversed by EGCG pre-treatment. In addition, EGCG attenuated the H2O2-induced increases of Type II inositol 3, 4-bisphosphate 4-phosphatase and relieved its subsequent inhibition of the downstream signalling for Akt and glycogen synthase kinase-3β (GSK-3β)/cyclin D1 in H9c2 cells. Pre-treatment with EGCG or GSK-3β inhibitor (SB 216763) significantly improved the H2O2-induced suppression on cell viability, phosphorylation of pAkt (S473) and pGSK-3β (S9), and level of cyclin D1 in cells. Conclusions Collectively, these findings suggest that EGCG blunts the H2O2-induced oxidative effect on the Akt activity through the modulation of PIP3 synthesis leading to the subsequent inactivation of GSK-3β mediated cardiac cell injury.
Collapse
Affiliation(s)
| | | | | | - Ban-Dar Hsu
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | | |
Collapse
|
38
|
Lin LH, Chang SJ, Hu RY, Lin MW, Lin ST, Huang SH, Lyu PC, Chou HC, Lai ZY, Chuang YJ, Chan HL. Biomarker discovery for neuroendocrine cervical cancer. Electrophoresis 2014; 35:2039-45. [DOI: 10.1002/elps.201400014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 03/20/2014] [Accepted: 04/02/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Li-Hsun Lin
- Department of Medical Sciences; Institute of Bioinformatics and Structural Biology; National Tsing Hua University; Hsinchu Taiwan
| | - Shing-Jyh Chang
- Gynecologic Oncology Section, Department of Obstetrics and Gynecology; Hsinchu Mackay Memorial Hospital; Hsinchu Taiwan
| | - Ren-Yu Hu
- Department of Medical Sciences; Institute of Bioinformatics and Structural Biology; National Tsing Hua University; Hsinchu Taiwan
| | - Meng-Wei Lin
- Department of Applied Science; National Hsinchu University of Education; Hsinchu Taiwan
| | - Szu-Ting Lin
- Department of Medical Sciences; Institute of Bioinformatics and Structural Biology; National Tsing Hua University; Hsinchu Taiwan
| | - Shun-Hong Huang
- Department of Medical Sciences; Institute of Bioinformatics and Structural Biology; National Tsing Hua University; Hsinchu Taiwan
| | - Ping-Chiang Lyu
- Department of Medical Sciences; Institute of Bioinformatics and Structural Biology; National Tsing Hua University; Hsinchu Taiwan
| | - Hsiu-Chuan Chou
- Department of Applied Science; National Hsinchu University of Education; Hsinchu Taiwan
| | - Zih-Yin Lai
- Department of Medical Sciences; Institute of Bioinformatics and Structural Biology; National Tsing Hua University; Hsinchu Taiwan
| | - Yung-Jen Chuang
- Department of Medical Sciences; Institute of Bioinformatics and Structural Biology; National Tsing Hua University; Hsinchu Taiwan
| | - Hong-Lin Chan
- Department of Medical Sciences; Institute of Bioinformatics and Structural Biology; National Tsing Hua University; Hsinchu Taiwan
| |
Collapse
|
39
|
Maiwulanjiang M, Chen J, Xin G, Gong AGW, Miernisha A, Du CYQ, Lau KM, Lee PSC, Chen J, Dong TTX, Aisa HA, Tsim KWK. The volatile oil of Nardostachyos Radix et Rhizoma inhibits the oxidative stress-induced cell injury via reactive oxygen species scavenging and Akt activation in H9c2 cardiomyocyte. JOURNAL OF ETHNOPHARMACOLOGY 2014; 153:491-498. [PMID: 24632018 DOI: 10.1016/j.jep.2014.03.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/03/2014] [Accepted: 03/04/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nardostachyos Radix et Rhizoma (NRR; the root and rhizome of Nardostachys jatamansi DC.) is a well-known medicinal herb widely used in Chinese, Uyghur and Ayurvedic medicines for the treatment of cardiovascular disorders. The oxidative stress-induced cardiomyocyte loss is the major pathogenesis of heart disorders. Here, the total volatile oil of NRR was isolated, and its function in preventing the cell death of cardiomyocyte was demonstrated. MATERIALS AND METHODS The cyto-protective effect of volatile oil of NRR against tBHP-induced H9c2 cardiomyocyte injury was measured by MTT assay. A promoter-report construct (pARE-Luc) containing four repeats of antioxidant response element (ARE) was applied to study the transcriptional activation of ARE. The amounts of phase ΙΙ antioxidant enzymes were analyzed by quantitative real-time polymer chain reaction (qPCR) upon the volatile oil treatment at 30 μg/mL for 24 h. The activation of Akt pathway was analyzed by western blot. RESULTS In cultured H9c2 cardiomyocytes, application of NRR volatile oil exhibited strong potency in preventing tBHP-induced cell death and accumulation of intracellular reactive oxygen species (ROS) in a concentration-dependent manner. In addition, the application of NRR volatile oil in cultures stimulated the gene expressions of self-defense antioxidant enzymes, which was mediated by the transcriptional activation of antioxidant response element (ARE). The induced genes were glutathione S-transferase, NAD(P)H quinone oxidoreductase, glutamate-cysteine ligase catalytic and modulatory subunits. In addition, the volatile oil of NRR activated the phosphorylation of Akt in cultured H9c2 cells. The treatment of LY294002, an Akt inhibitor, significantly inhibited the volatile oil-mediated ARE transcriptional activity, as well as the cell protective effect of NRR oil. CONCLUSION These results demonstrated that NRR volatile oil prevented the oxidative stress-induced cell death in H9c2 cells by (i) reducing intracellular ROS production, (ii) inducing antioxidant enzymes and (iii) activating Akt phosphorylation.
Collapse
Affiliation(s)
- Maitinuer Maiwulanjiang
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | - Jianping Chen
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | - Guizhong Xin
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | - Amy G W Gong
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | - Abudureyimu Miernisha
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | - Crystal Y Q Du
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | - Kei M Lau
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | - Pinky S C Lee
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | - Jihang Chen
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | - Tina T X Dong
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China
| | - Haji A Aisa
- Xinjiang Key Laboratory of Plant Resources and Natural Products Chemistry, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, China
| | - Karl W K Tsim
- Division of Life Science and Centre for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong, China.
| |
Collapse
|
40
|
Huang HH, Shao ZH, Li CQ, Vanden Hoek TL, Li J. Baicalein Protects Cardiomyocytes Against Mitochondrial Oxidant Injury Associated with JNK Inhibition and Mitochondrial Akt Activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 42:79-94. [DOI: 10.1142/s0192415x14500050] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Baicalein, a flavonoid derived from Scutellaria baicalensis Georgi, possesses cardioprotection against oxidant injury by scavenging reactive oxygen species (ROS). Few studies investigate whether baicalein protection is mediated by attenuating mitochondrial ROS and modulating the prosurvival and proapoptotic signaling. Primary cultured chick cardiomyocytes were used to study the role of baicalein in mitochondrial superoxide [Formula: see text] generation and signaling of Akt and JNK. Cells were exposed to H 2 O 2 for 2 h and baicalein was given 2 h prior to and during 2 h of H 2 O 2 exposure. Cell viability was assessed by propidium iodide and DNA fragmentation. H 2 O 2 (500 μM) significantly induced 45.3 ± 6.2% of cell death compared to the control (p < 0.001) and resulted in DNA laddering. Baicalein (10, 25 or 50 μM) dose-dependently reduced the cell death to 38.7 ± 5.6% (p = 0.226); 31.2 ± 3.9% (p < 0.01); 30.3 ± 5.3% (p < 0.01), respectively. It also attenuated DNA laddering. Further, baicalein decreased intracellular ROS and mitochondrial [Formula: see text] generation that was confirmed by superoxide dismutase PEG-SOD and mitochondria electron transport chain complex III inhibitor stigmatellin. In addition, baicalein increased Akt phosphorylation and decreased JNK phosphorylation in H 2 O 2-exposed cells. Moreover, baicalein augmented mitochondrial phosphorylation of Akt Thr308 and GSK3β Ser9, and prevented mitochondrial cytochrome c release assessed by cellular fractionation. Our results suggest that baicalein cardioprotection may involve an attenuation of mitochondrial [Formula: see text] and an increase in mitochondrial phosphorylation of Akt and GSK3β while decreasing JNK activation.
Collapse
Affiliation(s)
- Hsien-Hao Huang
- Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, USA
- Department of Emergency Medicine, Taipei Veterans General Hospital and Emergency Medicine, College of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Zuo-Hui Shao
- Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, USA
| | - Chang-Qing Li
- Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, USA
| | - Terry L. Vanden Hoek
- Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, USA
| | - Jing Li
- Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, USA
| |
Collapse
|
41
|
Chou HC, Chan HL. Effect of glutathione reductase knockdown in response to UVB-induced oxidative stress in human lung adenocarcinoma. Proteome Sci 2014; 12:2. [PMID: 24405781 PMCID: PMC3905656 DOI: 10.1186/1477-5956-12-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 12/03/2013] [Indexed: 01/19/2023] Open
Abstract
Background Glutathione reductase (GR) plays a critical role in the maintenance of physiological redox status in cells. However, the comprehensive investigations of GR-modulated oxidative stress have not been reported. Methods In the present study, we cultured a human lung adenocarcinoma line CL1-0 and its GR-knockdown derivative CL1-0ΔGR to evaluate their differential responses to UVB-irradiation. Results We identified 18 proteins that showed significant changes under UVB-irradiation in CL1-0ΔGR cells rather than in CL1-0 cells. Several proteins involving protein folding, metabolism, protein biosynthesis and redox regulation showed significant changes in expression. Conclusions In summary, the current study used a comprehensive lung adenocarcinoma-based proteomic approach for the identification of GR-modulated protein expression in response to UVB-irradiation. To our knowledge, this is the first global proteomic analysis to investigate the role of GR under UVB-irradiation in mammalian cell model.
Collapse
Affiliation(s)
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology & Department of Medical Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
42
|
Chou HC, Chan HL. 5-Methoxytryptophan-dependent protection of cardiomyocytes from heart ischemia reperfusion injury. Arch Biochem Biophys 2013; 543:15-22. [PMID: 24384558 DOI: 10.1016/j.abb.2013.12.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/01/2013] [Accepted: 12/17/2013] [Indexed: 10/25/2022]
Abstract
5-Methoxytryptophan (5-MTP), a catabolic product of tryptophan, can block Cox-2 overexpression in cancer cells as well as suppress cancer cell growth, migration and invasion. The aim of this study was to in vitro examine whether 5-MTP is able to reduce reactive oxygen species (ROS)-induced heart ischemia reperfusion injury and activate the cardiomyocyte's damage surveillance systems. Accordingly, rattus cardiomyocytes were treated with H2O2 as a heart ischemia reperfusion model prior to incubation with/without 5-MTP and proteomic analysis was performed to investigate the physiologic protection of 5-MTP in H2O2-induced ischemia reperfusion in cardiomyocyte. Our data demonstrated that 5-MTP treatment does protect cardiomyocyte in the ROS-induced ischemia reperfusion model. 5-MTP has also been shown to significantly facilitate cell migration and wound healing via cytoskeletal regulations. Additionally, two-dimensional differential gel electrophoresis (2D-DIGE) combined matrix assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF/TOF MS) analysis showed that 5-MTP might modulate growth-associated proteins, cytoskeleton regulation, redox regulation and protein folding to stimulate wound healing as well as prevent these ischemia reperfusion-damaged cardiomyocytes from cell death through maintaining cellular redox-balance and reducing ER-stress. To our knowledge, we report for the first time the cell repair mechanism of 5-MTP against ischemia reperfusion-damage in cardiomyocytes based on cell biology and proteomic analysis.
Collapse
Affiliation(s)
- Hsiu-Chuan Chou
- Department of Applied Science, National Hsinchu University of Education, Hsinchu, Taiwan
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology & Department of Medical Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
43
|
Hsieh SR, Hsu CS, Lu CH, Chen WC, Chiu CH, Liou YM. Epigallocatechin-3-gallate-mediated cardioprotection by Akt/GSK-3β/caveolin signalling in H9c2 rat cardiomyoblasts. J Biomed Sci 2013; 20:86. [PMID: 24251870 PMCID: PMC3871020 DOI: 10.1186/1423-0127-20-86] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 11/18/2013] [Indexed: 12/30/2022] Open
Abstract
Background Epigallocatechin-3-gallate (EGCg) with its potent anti-oxidative capabilities is known for its beneficial effects ameliorating oxidative injury to cardiac cells. Although studies have provided convincing evidence to support the cardioprotective effects of EGCg, it remains unclear whether EGCg affect trans-membrane signalling in cardiac cells. Here, we have demonstrated the potential mechanism for cardioprotection of EGCg against H2O2-induced oxidative stress in H9c2 cardiomyoblasts. Results Exposing H9c2 cells to H2O2 suppressed cell viability and altered the expression of adherens and gap junction proteins with increased levels of intracellular reactive oxygen species and cytosolic Ca2+. These detrimental effects were attenuated by pre-treating cells with EGCg for 30 min. EGCg also attenuated H2O2-mediated cell cycle arrest at the G1-S phase through the glycogen synthase kinase-3β (GSK-3β)/β-catenin/cyclin D1 signalling pathway. To determine how EGCg targets H9c2 cells, enhanced green fluorescence protein (EGFP) was ectopically expressed in these cells. EGFP-emission fluorescence spectroscopy revealed that EGCg induced dose-dependent fluorescence changes in EGFP expressing cells, suggesting that EGCg signalling events might trigger proximity changes of EGFP expressed in these cells. Proteomics studies showed that EGFP formed complexes with the 67 kD laminin receptor, caveolin-1 and -3, β-actin, myosin 9, vimentin in EGFP expressing cells. Using in vitro oxidative stress and in vivo myocardial ischemia models, we also demonstrated the involvement of caveolin in EGCg-mediated cardioprotection. In addition, EGCg-mediated caveolin-1 activation was found to be modulated by Akt/GSK-3β signalling in H2O2-induced H9c2 cell injury. Conclusions Our data suggest that caveolin serves as a membrane raft that may help mediate cardioprotective EGCg transmembrane signalling.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying-Ming Liou
- Department of Life Sciences, National Chung-Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
44
|
Chen JY, Chou HC, Chen YH, Chan HL. High glucose-induced proteome alterations in hepatocytes and its possible relevance to diabetic liver disease. J Nutr Biochem 2013; 24:1889-910. [DOI: 10.1016/j.jnutbio.2013.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/16/2013] [Accepted: 05/24/2013] [Indexed: 12/11/2022]
|
45
|
Lee TR, Lee HY, Huang SH, Chan HT, Lyu PC, Chan HL. Comparative proteomics analysis of normal and memory-deficient Drosophila melanogaster heads. Zool Stud 2013. [DOI: 10.1186/1810-522x-52-10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Abstract
Background
Learning and memory are extremely complex and dynamic processes. Proteins that participate in memory formation are strictly regulated by various pathways and may require protein synthesis and/or post-translational modifications. To examine the formation of memory, Drosophila was genetically engineered with the mutated memory-related gene, Amn
X8
, which induces normal learning and memory behavior within the first 30 min of training. However, the process through which learning occurred could not be retained after the 30 min of training, indicating that these mutants possessed deficits in middle-term memory. A proteomics platform based on two-dimensional differential gel electrophoresis and matrix-assisted laser desorption/ionization time of flight mass spectrometry was employed to examine the head proteome alterations between the wild-type 2u strain and the memory-deficient mutant Amn
X8
strain.
Results
The results indicated that 30 differentially expressed head proteins that mainly function in metabolic pathways and cell structure/cytoskeleton proteins were involved in memory formation. A bioinformatics analysis demonstrated that mitochondrial proteins had critical roles in modulating this process.
Conclusions
This is the first study of a comparative head proteomics analysis of a memory mutant strain and a normal control fruit fly strain. The fundamental proteomics analysis provides potential candidates for further elucidation of the biological mechanism of the memory formation process in Drosophila.
Collapse
|
46
|
Li JM, Chou HC, Wang SH, Wu CL, Chen YW, Lin ST, Chen YH, Chan HL. Hyaluronic acid-dependent protection against UVB-damaged human corneal cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2013; 54:429-449. [PMID: 23813585 DOI: 10.1002/em.21794] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 05/20/2013] [Accepted: 05/21/2013] [Indexed: 06/02/2023]
Abstract
Within ultraviolet radiation, ultraviolet B (UVB) is the most energetic and damaging to humans. At the protein level, UVB irradiation downregulates the expression of antioxidant enzymes leading to the accumulation of reactive oxygen species (ROS). Due to lacking of a global analysis of UVB-modulated corneal proteome, we investigate in vitro the mechanism of UVB-induced corneal damage to determine whether hyaluronic acid (HA) is able to reduce UVB irradiation-induced injury in human corneal epithelial cells. Accordingly, human corneal epithelial cell lines (HCE-2) were irradiated with UVB, followed by incubation with low molecular weight HA (LMW-HA, 100 kDa) or high molecular weight HA (HMW-HA, 1,000 kDa) to investigate the physiologic protection of HMW-HA in UVB-induced corneal injury, and to perform a global proteomic analysis. The data demonstrated that HA treatment protects corneal epithelial cells in the UVB-induced wound model, and that the molecular weight of HA is a crucial factor. Only HMW-HA significantly reduces the UVB-induced cytotoxic effects in corneal cells and increases cell migration and wound-healing ability. In addition, proteomic analysis showed that HMW-HA might modulate cytoskeleton regulation, signal transduction, biosynthesis, redox regulation, and protein folding to stimulate wound healing and to prevent these UVB-damaged cells from cell death. Further studies evidenced membrane-associated progesterone receptor component 1 (mPR) and malate dehydrogenase (MDH2) play essential roles in protecting corneal cells from UVB irradiation. This study reports on UVB-modulated cellular proteins that might play an important role in UVB-induced corneal cell injury and show HMW-HA to be a potential substance for protecting corneal cells from UVB-induced injury.
Collapse
Affiliation(s)
- Ji-Min Li
- Institute of Bioinformatics and Structural Biology & Department of Medical Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Cardioprotective Effects of Quercetin in Cardiomyocyte under Ischemia/Reperfusion Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:364519. [PMID: 23573126 PMCID: PMC3612448 DOI: 10.1155/2013/364519] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 11/22/2012] [Accepted: 02/07/2013] [Indexed: 11/30/2022]
Abstract
Quercetin, a polyphenolic compound existing in many vegetables, fruits, has antiinflammatory, antiproliferation, and antioxidant effect on mammalian cells. Quercetin was evaluated for protecting cardiomyocytes from ischemia/reperfusion injury, but its protective mechanism remains unclear in the current study. The cardioprotective effects of quercetin are achieved by reducing the activity of Src kinase, signal transducer and activator of transcription 3 (STAT3), caspase 9, Bax, intracellular reactive oxygen species production, and inflammatory factor and inducible MnSOD expression. Fluorescence two-dimensional differential gel electrophoresis (2D-DIGE) and matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) can reveal the differentially expressed proteins of H9C2 cells treated with H2O2 or quercetin. Although 17 identified proteins were altered in H2O2-induced cells, these proteins such as alpha-soluble NSF attachment protein (α-SNAP), Ena/VASP-like protein (Evl), and isopentenyl-diphosphate delta-isomerase 1 (Idi-1) were reverted by pretreatment with quercetin, which correlates with kinase activation, DNA repair, lipid, and protein metabolism. Quercetin dephosphorylates Src kinase in H2O2-induced H9C2 cells and likely blocks the H2O2-induced inflammatory response through STAT3 kinase modulation. This probably contributes to prevent ischemia/reperfusion injury in cardiomyocytes.
Collapse
|
48
|
Liddy KA, White MY, Cordwell SJ. Functional decorations: post-translational modifications and heart disease delineated by targeted proteomics. Genome Med 2013; 5:20. [PMID: 23445784 PMCID: PMC3706772 DOI: 10.1186/gm424] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The more than 300 currently identified post-translational modifications (PTMs) provides great scope for subtle or dramatic alteration of protein structure and function. Furthermore, the rapid and transient nature of many PTMs allows efficient signal transmission in response to internal and environmental stimuli. PTMs are predominantly added by enzymes, and the enzymes responsible (such as kinases) are thus attractive targets for therapeutic interventions. Modifications can be grouped according to their stability or transience (reversible versus irreversible): irreversible types (such as irreversible redox modifications or protein deamidation) are often associated with aging or tissue injury, whereas transient modifications are associated with signal propagation and regulation. This is particularly important in the setting of heart disease, which comprises a diverse range of acute (such as ischemia/reperfusion), chronic (such as heart failure, dilated cardiomyopathy) and genetic (such as hypertrophic cardiomyopathy) disease states, all of which have been associated with protein PTM. Recently the interplay between diverse PTMs has been suggested to also influence cellular function, with cooperation or competition for sites of modification possible. Here we discuss the utility of proteomics for examining PTMs in the context of the molecular mechanisms of heart disease.
Collapse
Affiliation(s)
- Kiersten A Liddy
- School of Molecular Bioscience, The University of Sydney, 2006 Sydney, Australia
| | - Melanie Y White
- School of Molecular Bioscience, The University of Sydney, 2006 Sydney, Australia ; Discipline of Pathology, School of Medical Sciences, The University of Sydney, 2006 Sydney, Australia
| | - Stuart J Cordwell
- School of Molecular Bioscience, The University of Sydney, 2006 Sydney, Australia ; Discipline of Pathology, School of Medical Sciences, The University of Sydney, 2006 Sydney, Australia
| |
Collapse
|
49
|
Franco C, Soares R, Pires E, Koci K, Almeida AM, Santos R, Coelho AV. Understanding regeneration through proteomics. Proteomics 2013; 13:686-709. [DOI: 10.1002/pmic.201200397] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/31/2012] [Accepted: 11/06/2012] [Indexed: 12/29/2022]
Affiliation(s)
- Catarina Franco
- Instituto de Tecnologia Química e Biológica; Universidade Nova de Lisboa; Oeiras Portugal
| | - Renata Soares
- Instituto de Tecnologia Química e Biológica; Universidade Nova de Lisboa; Oeiras Portugal
| | - Elisabete Pires
- Instituto de Tecnologia Química e Biológica; Universidade Nova de Lisboa; Oeiras Portugal
| | - Kamila Koci
- Instituto de Tecnologia Química e Biológica; Universidade Nova de Lisboa; Oeiras Portugal
| | - André M. Almeida
- Instituto de Tecnologia Química e Biológica; Universidade Nova de Lisboa; Oeiras Portugal
- Instituto de Investigação Científica Tropical; Lisboa Portugal
| | - Romana Santos
- Unidade de Investigação em Ciências Orais e Biomédicas, Faculdade de Medicina Dentária; Universidade de Lisboa; Portugal
| | - Ana Varela Coelho
- Instituto de Tecnologia Química e Biológica; Universidade Nova de Lisboa; Oeiras Portugal
| |
Collapse
|
50
|
Bouétard A, Besnard AL, Vassaux D, Lagadic L, Coutellec MA. Impact of the redox-cycling herbicide diquat on transcript expression and antioxidant enzymatic activities of the freshwater snail Lymnaea stagnalis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2013; 126:256-265. [PMID: 23237706 DOI: 10.1016/j.aquatox.2012.11.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 11/06/2012] [Accepted: 11/17/2012] [Indexed: 06/01/2023]
Abstract
The presence of pesticides in the environment results in potential unwanted effects on non-target species. Freshwater organisms inhabiting water bodies adjacent to agricultural areas, such as ditches, ponds and marshes, are good models to test such effects as various pesticides may reach these habitats through several ways, including aerial drift, run-off, and drainage. Diquat is a non-selective herbicide used for crop protection or for weed control in such water bodies. In this study, we investigated the effects of diquat on a widely spread aquatic invertebrate, the holarctic freshwater snail Lymnaea stagnalis. Due to the known redox-cycling properties of diquat, we studied transcript expression and enzymatic activities relative to oxidative and general stress in the haemolymph and gonado-digestive complex (GDC). As diquat is not persistent, snails were exposed for short times (5, 24, and 48 h) to ecologically relevant concentrations (22.2, 44.4, and 222.2 μg l(-1)) of diquat dibromide. RT-qPCR was used to quantify the transcription of genes encoding catalase (cat), a cytosolic superoxide dismutase (Cu/Zn-sod), a selenium-dependent glutathione peroxidase (gpx), a glutathione reductase (gred), the retinoid X receptor (rxr), two heat shock proteins (hsp40 and hsp70), cortactin (cor) and the two ribosomal genes r18S and r28s. Enzymatic activities of SOD, Gpx, Gred and glutathione S-transferase (GST) were investigated in the GDC using spectrophoto/fluorometric methods. Opposite trends were obtained in the haemolymph depending on the herbicide concentration. At the lowest concentration, effects were mainly observed after 24 h of exposure, with over-transcription of cor, hsp40, rxr, and sod, whereas higher concentrations down-regulated the expression of most of the studied transcripts, especially after 48 h of exposure. In the GDC, earlier responses were observed and the fold-change magnitude was generally much higher: transcription of all target genes increased significantly (or non-significantly for cat) after 5 h of exposure, and went back to control levels afterwards, suggesting the onset of an early response to oxidative stress associated to the unbalance of reactive oxygen species (ROS) in hepatocytes. Although increases obtained for Gred and SOD activities were globally consistent with their respective transcript expressions, up-regulation of transcription was not always correlated with increase of enzymatic activity, indicating that diquat might affect steps downstream of transcription. However, constitutive levels of enzymatic activities were at least maintained. In conclusion, diquat was shown to affect expression of the whole set of studied transcripts, reflecting their suitability as markers of early response to oxidative stress in L. stagnalis.
Collapse
Affiliation(s)
- Anthony Bouétard
- INRA, UMR INRA-Agrocampus Ouest ESE, Equipe Ecotoxicologie et Qualité des Milieux Aquatiques, Rennes, France.
| | | | | | | | | |
Collapse
|