1
|
Xia X, Li Y, Xiao X, Zhang Z, Mao C, Li T, Wan M. Chemotactic Micro/Nanomotors for Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306191. [PMID: 37775935 DOI: 10.1002/smll.202306191] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/12/2023] [Indexed: 10/01/2023]
Abstract
In nature, many organisms respond chemotactically to external chemical stimuli in order to extract nutrients or avoid danger. Inspired by this natural chemotaxis, micro/nanomotors with chemotactic properties have been developed and applied to study a variety of disease models. This chemotactic strategy has shown promising results and has attracted the attention of an increasing number of researchers. This paper mainly reviews the construction methods of different types of chemotactic micro/nanomotors, the mechanism of chemotaxis, and the potential applications in biomedicine. First, based on the classification of materials, the construction methods and therapeutic effects of chemotactic micro/nanomotors based on natural cells and synthetic materials in cellular and animal experiments will be elaborated in detail. Second, the mechanism of chemotaxis of micro/nanomotors is elaborated in detail: chemical reaction induced chemotaxis and physical process driven chemotaxis. In particular, the main differences and significant advantages between chemotactic micro/nanomotors and magnetic, electrical and optical micro/nanomotors are described. The applications of chemotactic micro/nanomotors in the biomedical fields in recent years are then summarized, focusing on the mechanism of action and therapeutic effects in cancer and cardiovascular disease. Finally, the authors are looking forward to the future development of chemotactic micro/nanomotors in the biomedical fields.
Collapse
Affiliation(s)
- Xue Xia
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Yue Li
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Xiangyu Xiao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Ziqiang Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Ting Li
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
2
|
Li N, Huang C, Zhang J, Zhang J, Huang J, Li S, Xia X, Wu Z, Chen C, Tang S, Xiao X, Gong H, Dai Y, Mao C, Wan M. Chemotactic NO/H 2S Nanomotors Realizing Cardiac Targeting of G-CSF against Myocardial Ischemia-Reperfusion Injury. ACS NANO 2023. [PMID: 37327056 DOI: 10.1021/acsnano.3c02781] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Recombinant granulocyte colony-stimulating factor (G-CSF), with a direct repair effect on injured cardiomyocytes against myocardial infarction ischemia-reperfusion-injury (IRI), displays a poor effect owing to the limited cardiac targeting efficacy. There are almost no reports of nanomaterials that deliver G-CSF to the IRI site. Herein, we propose a way to protect G-CSF by constructing one layer of nitric oxide (NO)/hydrogen sulfide (H2S) nanomotors on its outside. NO/H2S nanomotors with specific chemotactic ability to high expression of reactive oxygen species (ROS)/induced nitric oxide synthase (iNOS) at the IRI site can deliver G-CSF to the IRI site efficiently. Meanwhile, superoxide dismutase is covalently bound to the outermost part, reducing ROS at the IRI site through a cascade effect with NO/H2S nanomotors. The synergistic effect between NO and H2S on the effective regulation of the IRI microenvironment can not only avoid toxicity caused by excessive concentration of a single gas but also reduce inflammation level and relieve calcium overload, so as to promote G-CSF to play a cardioprotective role.
Collapse
Affiliation(s)
- Nan Li
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chenxing Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200433, China
| | - Jie Zhang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junyue Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jia Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200433, China
| | - Shangshang Li
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xue Xia
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ziyu Wu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chenglong Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Shuwan Tang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xiangyu Xiao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Hui Gong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200433, China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200433, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
3
|
Zhu MX, Ma XF, Niu X, Fan GB, Li Y. Mitochondrial unfolded protein response in ischemia-reperfusion injury. Brain Res 2022; 1797:148116. [PMID: 36209898 DOI: 10.1016/j.brainres.2022.148116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/27/2022] [Accepted: 10/02/2022] [Indexed: 11/21/2022]
Abstract
Mitochondrial unfolded protein response (UPRmt) is a mitochondrial stress response that activates the transcriptional program of mitochondrial chaperone proteins and proteases to keep protein homeostasis in mitochondria. Ischemia-reperfusion injury results in multiple severe clinical issues linked to high morbidity and mortality in various disorders. The pathophysiology and pathogenesis of ischemia-reperfusion injury are complex and multifactorial. Emerging evidence showed the roles of UPRmt signaling in ischemia-reperfusion injury. Herein, we discuss the regulatory mechanisms underlying UPRmt signaling in C. elegans and mammals. Furthermore, we review the recent studies into the roles and mechanisms of UPRmt signaling in ischemia-reperfusion injury of the heart, brain, kidney, and liver. Further research of UPRmt signaling will potentially develop novel therapeutic strategies against ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ming-Xi Zhu
- Department of Anatomy, School of Basic Medicine and Life Science, Hainan Medical University, Hainan, China
| | - Xiao-Fei Ma
- Department of ICU, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xing Niu
- Department of Second Clinical College, Shengjing Hospital of China Medical University, Shenyang, China
| | - Gui-Bo Fan
- Department of Anesthesiology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Yan Li
- Department of Anesthesiology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
4
|
Yan H, Zhao H, Yi SW, Zhuang H, Wang DW, Jiang JG, Shen GF. Sphingosine-1-Phosphate Protects Against the Development of Cardiac Remodeling via Sphingosine Kinase 2 and the S1PR2/ERK Pathway. Curr Med Sci 2022; 42:702-710. [PMID: 35963947 DOI: 10.1007/s11596-022-2600-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 03/25/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Cardiac remodeling is a common pathological change in various cardiovascular diseases and can ultimately result in heart failure. Thus, there is an urgent need for more effective strategies to aid in cardiac protection. Our previous work found that sphingosine-1-phosphate (S1P) could ameliorate cardiac hypertrophy. In this study, we aimed to investigate whether S1P could prevent cardiac fibrosis and the associated mechanisms in cardiac remodeling. METHODS Eight-week-old male C57BL/6 mice were randomly divided into a sham, transverse aortic constriction (TAC) or a TAC+S1P treatment group. RESULTS We found that S1P treatment improved cardiac function in TAC mice and that the cardiac fibrosis ratio in the TAC+S1P group was significantly lower and was accompanied by a decrease in α-smooth muscle actin (α-SMA) and collagen type I (COL I) expression compared with the TAC group. We also found that one of the key S1P enzymes, sphingosine kinase 2 (SphK2), which was mainly distributed in cytoblasts, was downregulated in the cardiac remodeling case and recovered after S1P treatment in vivo and in vitro. In addition, our in vitro results showed that S1P treatment activated extracellular regulated protein kinases (ERK) phosphorylation mainly through the S1P receptor 2 (S1PR2) and spurred p-ERK transposition from the cytoplasm to cytoblast in H9c2 cells exposed to phenylephrine. CONCLUSION These findings suggest that SphK2 and the S1PR2/ERK pathway may participate in the anti-remodeling effect of S1P on the heart. This work therefore uncovers a novel potential therapy for the prevention of cardiac remodeling.
Collapse
Affiliation(s)
- Hui Yan
- Wuhan No. 4 Hospital, Wuhan Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hu Zhao
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shao-Wei Yi
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hang Zhuang
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dao-Wen Wang
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Gang Jiang
- Division of Cardiology, Department of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gui-Fen Shen
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
5
|
Park IH, Shen GY, Song YS, Jong Cho Y, Kim BS, Lee Y, Lim YH, Shin JH, Kim KS. Granulocyte colony-stimulating factor reduces the endoplasmic reticulum stress in a rat model of diabetic cardiomyopathy. Endocr J 2021; 68:1293-1301. [PMID: 34121048 DOI: 10.1507/endocrj.ej21-0016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Prolonged endoplasmic reticulum (ER) stress contributes to the apoptosis of cardiomyocytes, which leads to the development of diabetic cardiomyopathy. Previously, we reported that the granulocyte colony-stimulating factor (G-CSF) reduces the cardiomyocyte apoptosis in diabetic cardiomyopathy; however, the precise mechanisms associated with this process are not yet fully understood. Therefore, in this study, we investigated whether the mechanism of the anti-apoptotic effect of G-CSF was associated with ER stress in a rat model of diabetic cardiomyopathy. Diabetic cardiomyopathy was induced in rats using a high-fat diet combined with the administration of a low-dose of streptozotocin. Diabetic rats were treated with G-CSF or saline for 5 days. Cardiac function was evaluated using serial echocardiography before and 4 weeks after treatment. The rate of cardiomyocyte apoptosis and the expression levels of proteins related to ER stress, including glucose-regulated protein 78 (GRP78), caspase-9, and caspase-12 were analyzed in the cardiac tissue. G-CSF treatment significantly reduced cardiomyocyte apoptosis in the diabetic myocardium and downregulated the expression levels of these proteins in diabetic rats treated with low-dose streptozotocin when compared to that in rats treated with saline. In addition, G-CSF treatment significantly downregulated the expression levels of proteins related to ER stress, such as GRP78, inositol-requiring enzyme-1α (IRE-1α), and C/EBP homologous protein (CHOP) in H9c2 cells under high glucose (HG) conditions. Moreover, G-CSF treatment significantly improved the diastolic dysfunction in serial echocardiography assessments. In conclusion, the anti-apoptotic effect of G-CSF may be associated with the downregulation of ER stress.
Collapse
Affiliation(s)
- In-Hwa Park
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Guang-Yin Shen
- Department of Cardiology, Jilin University, Jilin Central Hospital, Jilin, China
| | - Yi-Sun Song
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Young Jong Cho
- Laboratory Medicine, College of Medicine, Kangwon National University, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Byung Sik Kim
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Yonggu Lee
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Young-Hyo Lim
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Jeong-Hun Shin
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Soo Kim
- Department of Cardiology, St. Peter's Hospital, Seoul, Republic of Korea
| |
Collapse
|
6
|
Mohapatra S, Kundu AK, Mishra SR, Senapati S, Jyotiranjan T, Panda G. HSF1 and GM-CSF expression, its association with cardiac health, and assessment of organ function during heat stress in crossbred Jersey cattle. Res Vet Sci 2021; 139:200-210. [PMID: 34358923 DOI: 10.1016/j.rvsc.2021.07.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/16/2021] [Accepted: 07/14/2021] [Indexed: 11/25/2022]
Abstract
The study aimed to evaluate the differential expression of HSF1 and GM-CSF mRNA in PBMCs and correlate it with myocardial injury in crossbred Jersey heifers during heat stress. The study also assessed the effect of heat stress on cardiac electrical activity, vascular health, liver function and kidney function. The experiment was conducted in two phases: for heat stressed animals; HS in June (THI ranged from 80.0 to 89.8) and for control group i.e. not exposed to heat stress in January (THI ranged between 70.1 and 71.4). Results of the study revealed that the relative abundance of HSF1 and GM-CSF mRNA increased significantly (P < 0.05) in HS. Serum cardiac biomarkers such as CK-MB, AST and CRP were significantly elevated (P < 0.05) in HS. cTnI was detected 'positive' in nineteen out of twenty four cases in HS. Correlation of HSF1 and GM-CSF expression with concentration of LDH, CKMB, CRP and AST in HS was negative but non-significant (P > 0.05). Significant (P < 0.05) ECG findings in HS were increased heart rate, decreased RR interval, decreased PR interval, decreased QRS amplitude and decreased amplitude of P wave. Marked reduction (P < 0.05) in serum cholesterol and triglyceride levels was observed in HS. ALP, AST, bilirubin and urea levels in serum were significantly elevated (P < 0.05) in HS. In conclusion, cardiac enzymes in serum were significantly elevated in HS indicating myocardial injury. HSF1 and GM-CSF mRNA expression alone was inadequate in conferring cytoprotection to cardiac cells in HS. Cardiac electrical activity, vascular status, liver and kidney function were significantly altered in HS.
Collapse
Affiliation(s)
- Swagat Mohapatra
- Department of Veterinary Physiology, C.V.Sc. & A.H., O.U.A.T., Bhubaneswar, Odisha, India
| | - Akshaya Kumar Kundu
- Department of Veterinary Physiology, C.V.Sc. & A.H., O.U.A.T., Bhubaneswar, Odisha, India
| | - Smruti Ranjan Mishra
- Department of Veterinary Physiology, C.V.Sc. & A.H., O.U.A.T., Bhubaneswar, Odisha, India
| | | | - Tushar Jyotiranjan
- Department of Veterinary Physiology, C.V.Sc. & A.H., O.U.A.T., Bhubaneswar, Odisha, India
| | - Gopalaxmi Panda
- Department of Biochemistry, SCB Medical College, Cuttack, Odisha, India.
| |
Collapse
|
7
|
Shen Y, Zhang X, Li C, Wang X, Ye Y, Yuan J, Gong H, Zou Y, Ge J. Pressure overload promotes cystatin C secretion of cardiomyocytes to regulate the MAPK signaling pathway and mediate cardiac hypertrophy. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1514. [PMID: 33313259 PMCID: PMC7729345 DOI: 10.21037/atm-20-7041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/18/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND This study aimed to compare serum cystatin C (CysC) levels between hypertensive and non-hypertensive patients, and to explore the correlation between serum CysC and left ventricular hypertrophy (LVH). We also investigated the effects of pressure overload on cardiac expression and secretion of CysC, and explored the direct effect of CysC on the hypertrophy of primary cardiomyocytes. METHODS Serum CysC was compared in patients with hypertension (634 patients) and those without hypertension (411 patients), and the correlation between serum CysC levels and LVH was explored. A transverse aortic constriction (TAC) mouse model and a mechanical stretch model of primary cardiomyocytes and fibroblasts were developed to compare cardiac expression and secretion of CysC under pressure overload. After intervention with exogenous CysC, we compared the cross-sectional area of primary cardiomyocytes, cardiac hypertrophy-associated gene expression, and phosphorylation of the MAPK signaling pathway. RESULTS In chronic kidney disease (CKD) stage 1 patients, serum CysC was higher in hypertensive patients independent of renal function. Serum CysC elevation was an independent predictor of LVH after correction for endogenous creatinine clearance rate (eCCr), left ventricular ejection fraction (LVEF), and NT-proBNP. Cardiac levels of CysC in TAC mice were elevated. CST3 gene expression was upregulated, and both intracellular and culture supernatant CysC levels increased after mechanical stretch of primary cardiomyocytes. After intervention with exogenous CysC, the cross-sectional area of primary cardiomyocytes increased, as well as the gene expression of Nppa, Nppb, and Myh7, and the phosphorylation of ERK, p38, and TAK1. CONCLUSIONS Serum CysC levels were higher in hypertensive patients, and serum CysC elevation was an independent predictor of LVH after correction for eCCr. Pressure overload induced greater cardiomyocyte secretion of CysC. Exogenous CysC can enter cardiomyocytes, having a pro-hypertrophic effect on primary cardiomyocytes through regulation of the MAPK signaling pathways.
Collapse
Affiliation(s)
- Yi Shen
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyi Zhang
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenguang Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiang Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Ye
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Yuan
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Multiple Progressive Thermopreconditioning Improves Cardiac Ischemia/Reperfusion-induced Left Ventricular Contractile Dysfunction and Structural Abnormality in Rat. Transplantation 2020; 104:1869-1878. [PMID: 32058468 DOI: 10.1097/tp.0000000000003176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Triple progressive thermopreconditioning (3PTP) may induce high Hsp-70 expression to maintain cardiac function. We suggest that 3PTP may reduce myocardial ischemia/reperfusion (I/R) injury during organ transplantation through Bag3/Hsp-70 mediated defense mechanisms. METHODS Male Wistar rats were divided into sham control group and 72 h after 3PTP in a 42°C water bath (3PTP) group. Rats underwent 60 min of ischemia by occlusion of the left anterior descending coronary artery followed by 240 min reperfusion. Hemodynamic parameters, including the electrocardiogram, microcirculation, heart rate, left ventricular end-diastolic pressure, maximal rate of rise (+dp/dt), and fall (-dp/dt) in the left ventricular pressure for index of contraction and relaxation were determined. Myocardial infarct size was evaluated by the Evans blue-2,3,5-triphenyltetrazolium chloride method. 3PTP-induced protective mechanisms were determined by Western blot and immunohistochemistry. RESULTS Cardiac I/R depressed cardiac microcirculation, induced S-T segment elevation, and R-R and P-R interval elongation increased infarct size associated with erythrocyte extravasation, leukocytes and macrophage/monocyte infiltration, granulocyte colony-stimulating factor, poly(ADP-ribose) polymerase 1 stain, and transferase-mediated dUTP-biotin nick end labeling positive cells. However, 3PTP evoked significant cardioprotection against I/R injury, characterized by the increased +dp/dt value and the decreased elevated left ventricular end-diastolic pressure, erythrocyte extravasation, leukocyte and macrophage/monocyte infiltration, granulocyte colony-stimulating factor expression, poly(ADP-ribose) polymerase 1 expression, transferase-mediated dUTP-biotin nick end labeling positive cells, and fragmentation and infarct area. In addition, 3PTP increased Hsp-70 and Bag3 expression and decreased Bax/Bcl-2 ratio, but did not affect the Beclin-1 and LC3-II/LC3-I ratio in the heart with I/R injury. CONCLUSIONS 3PTP therapies may through Bag3 upregulation alleviate I/R injury-induced left ventricular structural deterioration and dysfunction.
Collapse
|
9
|
Liu D, Wu L, Wu Y, Wei X, Wang W, Zhang S, Yi M, Li J, Liu H, Ma X. Heat shock factor 1-mediated transcription activation of Omi/HtrA2 induces myocardial mitochondrial apoptosis in the aging heart. Aging (Albany NY) 2019; 11:8982-8997. [PMID: 31627188 PMCID: PMC6834417 DOI: 10.18632/aging.102361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 10/05/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Increased cardiac apoptosis is a hallmark of the elderly, which in turn increases the risk for developing cardiac disease. The overexpression of Omi/HtrA2 mRNA and protein contributes to apoptosis in the aged heart. Heat shock factor 1 (HSF1) is a transcription factor that binds to the promoter of Omi/HtrA2 in the aging myocardium. However, whether HSF1 participates in cardiomyocyte apoptosis via transcriptional regulation of Omi/HtrA2 remains unclear. The present study was designed to investigate whether HSF1 plays a role in Omi/HtrA2 transcriptional regulation and myocardial apoptosis. METHODS AND RESULTS Assessment of the hearts of mice of different ages was performed, which indicated a decrease in cardiac function reserve and an increase in mitochondrial apoptosis. Omi/HtrA2 overexpression in the elderly was negatively correlated with left ventricular function after exercise overload and positively correlated with myocardial Caspase-9 apoptosis. Chromatin immunoprecipitation (ChIP) of aging hearts and plasmid transfection/RNA interference of H9C2 cells revealed that enhancement of HSF1 expression promotes Omi/HtrA2 expression by inducing the promoter activity of Omi/HtrA2 while also increasing mitochondrial apoptosis by upregulating Omi/HtrA2 expression. CONCLUSIONS HSF1 acts as a transcriptional factor that induces Omi/HtrA2 expression and Caspase-9 apoptosis in aged cardiomyocytes, while also decreasing cardiac function reserve.
Collapse
Affiliation(s)
- Dan Liu
- Department of Physiology and Pathophysiology, Yan Jing Medical College, Capital Medical University, Beijing 101300, China
| | - Linguo Wu
- Department of Pathology, Beijing LuHe Hospital of Capital Medical University, Beijing 101100, China
| | - Ye Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Xin Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Ming Yi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jing Li
- Department of Cardiology, XuanWu Hospital Capital Medical University, Beijing 100053, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Metabolic Disturbance Related Cardiovascular Disease, Beijing 100069, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
10
|
Haybar H, Shahrabi S, Rezaeeyan H, Shirzad R, Saki N. Protective role of heat shock transcription factor 1 in heart failure: A diagnostic approach. J Cell Physiol 2018; 234:7764-7770. [DOI: 10.1002/jcp.27639] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/02/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology Faculty of Medicine, Semnan University of Medical Sciences Semnan Iran
| | - Hadi Rezaeeyan
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Reza Shirzad
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| |
Collapse
|
11
|
Haybar H, Shahrabi S, Deris Zayeri Z, Pezeshki S. Strategies to increase cardioprotection through cardioprotective chemokines in chemotherapy-induced cardiotoxicity. Int J Cardiol 2018; 269:276-282. [DOI: 10.1016/j.ijcard.2018.07.087] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/19/2018] [Accepted: 07/18/2018] [Indexed: 01/08/2023]
|
12
|
Shen J, Xie Y, Liu Z, Zhang S, Wang Y, Jia L, Wang Y, Cai Z, Ma H, Xiang M. Increased myocardial stiffness activates cardiac microvascular endothelial cell via VEGF paracrine signaling in cardiac hypertrophy. J Mol Cell Cardiol 2018; 122:140-151. [PMID: 30138627 DOI: 10.1016/j.yjmcc.2018.08.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 11/21/2022]
Abstract
When the heart is subjected to an increased workload, mechanical stretch together with neurohumoral stimuli activate the "fetal gene program" and induce cardiac hypertrophy to optimize output. Due to a lack of effective methods/models to quantify and modulate cardiac mechanical properties, the connection between these properties and the development of cardiac hypertrophy remains largely unexplored. Here, we utilized an atomic force microscope (AFM) to directly measure the elastic modulus of the hypertrophic myocardium induced by pressure overload. Additionally, we investigated the effects of extracellular elasticity on angiogenesis, which provides blood and nutrition to support cardiomyocyte hypertrophic growth in this process. In response to pressure overload, the myocardium rapidly developed hypertrophy and correspondingly demonstrated a high elastic modulus property. This mechanical feature correlated with enhanced angiogenesis. Mechanistically, we found that a high elastic modulus promoted cultured cardiomyocytes to synthesize and paracrine vascular endothelial growth factor (VEGF) to activate cardiac microvascular endothelial cells. Further analysis showed that the increased elastic modulus enhanced the interaction between Talin1 and integrin β1 to activate the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/hypoxia-inducible factor 1α (Hif-1α) pathway, which contributed to VEGF production. Thus, our study revealed a critical role of the elastic modulus in regulating angiogenesis during the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Jian Shen
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Yao Xie
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Zhenjie Liu
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Shuning Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200000, China
| | - Yaping Wang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Liangliang Jia
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Yidong Wang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Zhejun Cai
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Hong Ma
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China.
| | - Meixiang Xiang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China.
| |
Collapse
|
13
|
Mechanical stresses induce paracrine β-2 microglobulin from cardiomyocytes to activate cardiac fibroblasts through epidermal growth factor receptor. Clin Sci (Lond) 2018; 132:1855-1874. [PMID: 30072448 DOI: 10.1042/cs20180486] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 11/17/2022]
Abstract
By employing a proteomic analysis on supernatant of mechanically stretched cardiomyocytes, we found that stretch induced a significantly high level of β-2 microglobulin (β2M), a non-glycosylated protein, which is related to inflammatory diseases but rarely known in cardiovascular diseases. The present data showed that serum β2M level was increased in patients with hypertension and further increased in patients with chronic heart failure (HF) as compared with control group, and the high level of serum β2M level correlated to cardiac dysfunction in these patients. In pressure overload mice model by transverse aortic constriction (TAC), β2M levels in serum and heart tissue increased progressively in a time-dependent manner. Exogenous β2M showed pro-fibrotic effects in cultured cardiac fibroblasts but few effects in cardiomyocytes. Adeno-associated virus 9 (AAV9)-mediated knockdown of β2M significantly reduced cardiac β2M level and inhibited myocardial fibrosis and cardiac dysfunction but not cardiac hypertrophy at 4 weeks after TAC. In vitro, mechanical stretch induced the rapid secretion of β2M mainly from cardiomyocytes by activation of extracellular-regulated protein kinase (ERK). Conditional medium (CM) from mechanically stretched cardiomyocytes activated cultured cardiac fibroblasts, and the effect was partly abolished by CM from β2M-knockdown cardiomyocytes. In vivo, knockdown of β2M inhibited the increase in phosphorylation of epidermal growth factor receptor (EGFR) induced by TAC. In cultured cardiac fibroblasts, inhibition of EGFR significantly attenuated the β2M-induced the activation of EGFR and pro-fibrotic responses. The present study suggests that β2M is a paracrine pro-fibrotic mediator and associated with cardiac dysfunction in response to pressure overload.
Collapse
|
14
|
Yuan L, Qiu L, Ye Y, Wu J, Wang S, Wang X, Zhou N, Zou Y. Heat-shock transcription factor 1 is critically involved in the ischaemia-induced cardiac hypertrophy via JAK2/STAT3 pathway. J Cell Mol Med 2018; 22:4292-4303. [PMID: 29992755 PMCID: PMC6111827 DOI: 10.1111/jcmm.13713] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/25/2018] [Indexed: 01/20/2023] Open
Abstract
Cardiac hypertrophy after myocardial infarction (MI) is an independent risk factor for heart failure. Regression of cardiac hypertrophy has emerged as a promising strategy in the treatment of MI patients. Here, we have been suggested that heat-shock transcription factor 1 (HSF1) is a novel repressor of ischaemia-induced cardiac hypertrophy. Ligation of left anterior descending coronary was used to produce MI in HSF1-deficient heterozygote (KO), HSF1 transgenic (TG) mice and their wild-type (WT) littermates, respectively. Neonatal rat cardiomyocytes (NRCMs) were treated by hypoxia to mimic MI in vitro. The HSF1 phosphorylation was significantly reduced in the infarct border zone of mouse left ventricles (LVs) 1 week after MI and in the hypoxia-treated NRCMs. HSF1 KO mice showed more significant maladaptive cardiac hypertrophy and deteriorated cardiac dysfunction 1 week after MI compared to WT MI mice. Deficiency of HSF1 by siRNA transfection notably increased the hypoxia-induced myocardial hypertrophy in NRCMs. Mechanistically, Janus kinase 2 (JAK2) and its effector, signal transducer and activator of transcription 3 (STAT3) were found to be significantly increased in the LV infarct border zone of WT mice after MI as well as the NRCMs treated by hypoxia. These alterations were more significant in HSF1 KO mice and NRCMs transfected with HSF1 SiRNA. Inversely, HSF1 TG mice showed significantly ameliorated cardiac hypertrophy and heart failure 1 week after LAD ligation compared to their WT littermates. Our data collectively demonstrated that HSF1 is critically involved in the pathological cardiac hypertrophy after MI via modulating JAK2/STAT3 signalling and may constitute a potential therapeutic target for MI patients.
Collapse
Affiliation(s)
- Lingyan Yuan
- Department of kinesiology, Institute of physical education, Shanghai Normal University, Shanghai, China
| | - Lin Qiu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Ye
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biological Science, Fudan University, Shanghai, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biological Science, Fudan University, Shanghai, China
| | - Shuchun Wang
- Department of Computer Tomography and Magnetic Imaging, Yidu Central Hospital, Weifang Medical College, Weifang, China
| | - Xingxu Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biological Science, Fudan University, Shanghai, China
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biological Science, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Lee PH. Adjusted analysis. J Mol Cell Cardiol 2018; 120:28-30. [PMID: 29777690 DOI: 10.1016/j.yjmcc.2018.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/15/2018] [Indexed: 10/16/2022]
Affiliation(s)
- Paul H Lee
- The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
16
|
Chen Z, Li Y, Jiang G, Yang C, Wang Y, Wang X, Fang B, Zhang G, Sun Y, Qian J, Gong H, Zou Y. Knockdown of LRP6 activates Drp1 to inhibit survival of cardiomyocytes during glucose deprivation. Biomed Pharmacother 2018; 103:1408-1414. [PMID: 29864925 DOI: 10.1016/j.biopha.2018.04.134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/15/2018] [Accepted: 04/17/2018] [Indexed: 10/17/2022] Open
Abstract
Lipoprotein receptor-related protein 6 (LRP6) binds to Wnt ligands to transduce signal by stabilization of β-catenin, which has been involved in the regulation of embryonic development and metabolism et al. Here, we observed LRP6 decreased in human hearts with dilated cardiomyopathy (DCM), and it also decreased in cultured cardiomyocytes under glucose- deprivation (GD). Knockdown of LRP6 greatly inhibited cell viability in cardiomyocytes under GD, but it didn't induce the effect in cardiomyocytes at baseline. Overexpression of LRP6 increased the cell viability in GD-cardiomyocytes. To explore potential molecular mechanisms, we detected the phosphorylation of dynamin-related protein 1(Drp1) and active β-catenin in cardiomyocytes under GD. Knockdown of LRP6 enhanced p-Drp1(S616) level while it didn't alter the p-Drp1(S637) and active β-catenin level in GD-cardiomyocytes. Drp1 inhibitor significantly suppressed the increase in p-Drp1 at S616 and improved the cell viability in GD-cardiomyocytes with knockdown of LRP6. Further analysis showed that knockdown of LRP6 also increased the phosphorylation of mammalian target of rapamycin (mTOR), and Drp1 inhibitor greatly inhibited the increase in p-mTOR level in GD-cardiomyocytes. The present study indicated that knockdown of LRP6 inhibited the cell viability by activation of Drp1 in GD-cardiomyocytes, and the phosphorylation of mTOR may be involved in the process. It suggests that LRP6 can prevent cardiomyocytes from death in nutrition-deprived condition.
Collapse
Affiliation(s)
- Zhidan Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yang Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Guoliang Jiang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Chunjie Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Ying Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiang Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Bo Fang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Guoping Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yongxin Sun
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Juying Qian
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Wang S, Wu J, You J, Shi H, Xue X, Huang J, Xu L, Jiang G, Yuan L, Gong X, Luo H, Ge J, Cui Z, Zou Y. HSF1 deficiency accelerates the transition from pressure overload-induced cardiac hypertrophy to heart failure through endothelial miR-195a-3p-mediated impairment of cardiac angiogenesis. J Mol Cell Cardiol 2018; 118:193-207. [PMID: 29626503 DOI: 10.1016/j.yjmcc.2018.03.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 03/10/2018] [Accepted: 03/27/2018] [Indexed: 01/30/2023]
|
18
|
Ma L, Ma X, Kong F, Guo J, Shi H, Zhu J, Zou Y, Ge J. Mammalian target of rapamycin inhibition attenuates myocardial ischaemia-reperfusion injury in hypertrophic heart. J Cell Mol Med 2018; 22:1708-1719. [PMID: 29314656 PMCID: PMC5824378 DOI: 10.1111/jcmm.13451] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/06/2017] [Indexed: 01/08/2023] Open
Abstract
Pathological cardiac hypertrophy aggravated myocardial infarction and is causally related to autophagy dysfunction and increased oxidative stress. Rapamycin is an inhibitor of serine/threonine kinase mammalian target of rapamycin (mTOR) involved in the regulation of autophagy as well as oxidative/nitrative stress. Here, we demonstrated that rapamycin ameliorates myocardial ischaemia reperfusion injury by rescuing the defective cytoprotective mechanisms in hypertrophic heart. Our results showed that chronic rapamycin treatment markedly reduced the phosphorylated mTOR and ribosomal protein S6 expression, but not Akt in both normal and aortic-banded mice. Moreover, chronic rapamycin treatment significantly mitigated TAC-induced autophagy dysfunction demonstrated by prompted Beclin-1 activation, elevated LC3-II/LC3-I ratio and increased autophagosome abundance. Most importantly, we found that MI/R-induced myocardial injury was markedly reduced by rapamycin treatment manifested by the inhibition of myocardial apoptosis, the reduction of myocardial infarct size and the improvement of cardiac function in hypertrophic heart. Mechanically, rapamycin reduced the MI/R-induced iNOS/gp91phox protein expression and decreased the generation of NO and superoxide, as well as the cytotoxic peroxynitrite. Moreover, rapamycin significantly mitigated MI/R-induced endoplasmic reticulum stress and mitochondrial impairment demonstrated by reduced Caspase-12 activity, inhibited CHOP activation, decreased cytoplasmic Cyto-C release and preserved intact mitochondria. In addition, inhibition of mTOR also enhanced the phosphorylated ERK and eNOS, and inactivated GSK3β, a pivotal downstream target of Akt and ERK signallings. Taken together, these results suggest that mTOR signalling protects against MI/R injury through autophagy induction and ERK-mediated antioxidative and anti-nitrative stress in mice with hypertrophic myocardium.
Collapse
Affiliation(s)
- Lei‐Lei Ma
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
- Department of Critical Care MedicineZhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical CollegeHangzhouChina
| | - Xin Ma
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
| | - Fei‐Juan Kong
- Department of Endocrinology and MetabolismShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Jun‐Jie Guo
- Department of CardiologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Hong‐Tao Shi
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
| | - Jian‐Bing Zhu
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
| | - Yun‐Zeng Zou
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
| | - Jun‐Bo Ge
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
| |
Collapse
|
19
|
Hypertrophied myocardium is vulnerable to ischemia/reperfusion injury and refractory to rapamycin-induced protection due to increased oxidative/nitrative stress. Clin Sci (Lond) 2018; 132:93-110. [PMID: 29175946 DOI: 10.1042/cs20171471] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 11/17/2022]
Abstract
Left ventricular hypertrophy (LVH) is causally related to increased morbidity and mortality following acute myocardial infarction (AMI) via still unknown mechanisms. Although rapamycin exerts cardioprotective effects against myocardial ischemia/reperfusion (MI/R) injury in normal animals, whether rapamycin-elicited cardioprotection is altered in the presence of LVH has yet to be determined. Pressure overload induced cardiac hypertrophied mice and sham-operated controls were exposed to AMI by coronary artery ligation, and treated with vehicle or rapamycin 10 min before reperfusion. Rapamycin produced marked cardioprotection in normal control mice, whereas pressure overload induced cardiac hypertrophied mice manifested enhanced myocardial injury, and was refractory to rapamycin-elicited cardioprotection evidenced by augmented infarct size, aggravated cardiomyocyte apoptosis, and worsening cardiac function. Rapamycin alleviated MI/R injury via ERK-dependent antioxidative pathways in normal mice, whereas cardiac hypertrophied mice manifested markedly exacerbated oxidative/nitrative stress after MI/R evidenced by the increased iNOS/gp91phox expression, superoxide production, total NO metabolites, and nitrotyrosine content. Moreover, scavenging superoxide or peroxynitrite by selective gp91phox assembly inhibitor gp91ds-tat or ONOO- scavenger EUK134 markedly ameliorated MI/R injury, as shown by reduced myocardial oxidative/nitrative stress, alleviated myocardial infarction, hindered cardiomyocyte apoptosis, and improved cardiac function in aortic-banded mice. However, no additional cardioprotective effects were achieved when we combined rapamycin and gp91ds-tat or EUK134 in ischemic/reperfused hearts with or without LVH. These results suggest that cardiac hypertrophy attenuated rapamycin-induced cardioprotection by increasing oxidative/nitrative stress and scavenging superoxide/peroxynitrite protects the hypertrophied heart from MI/R.
Collapse
|
20
|
Yan H, Yi S, Zhuang H, Wu L, Wang DW, Jiang J. Sphingosine-1-phosphate ameliorates the cardiac hypertrophic response through inhibiting the activity of histone deacetylase-2. Int J Mol Med 2017; 41:1704-1714. [PMID: 29286094 DOI: 10.3892/ijmm.2017.3325] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/29/2017] [Indexed: 11/06/2022] Open
Abstract
Inhibition of histone deacetylase-2 (HDAC2), which is a prohypertrophic factor in the heart, can functionally attenuate cardiac hypertrophy. The present study aimed to investigate whether sphingosine‑1‑phosphate (S1P), which has recently been reported to suppress HDAC2 activity, could ameliorate the cardiac hypertrophic response and improve cardiac function in mice with transverse aortic constriction (TAC), as well as to determine the underlying mechanisms. Briefly, 8‑week‑old male C57BL/6 mice were randomly divided into sham, TAC and TAC + S1P groups; the results indicated that S1P treatment attenuated TAC‑induced cardiac dysfunction. In addition, heart size and the expression levels of fetal cardiac genes were reduced in the TAC + S1P group compared with in the TAC group. Furthermore, in cultured H9c2 cells exposed to phenylephrine, S1P was revealed to decrease cardiomyocyte size and the exaggerated expression of fetal cardiac genes. The present study also demonstrated that S1P had no effect on HDAC2 expression, but it did suppress its activity and increase acetylation of histone H3 in vivo and in vitro. Krüppel‑like factor 4 (KLF4) is an antihypertrophic transcriptional regulator, which mediates HDAC inhibitor‑induced prevention of cardiac hypertrophy; in the present study, KLF4 was upregulated by S1P. Finally, the results indicated that S1P receptor 2 (S1PR2) may be involved in the antihypertrophic effects, whereas the suppressive effects of S1P on HDAC2 activity were independent of S1PR2. In conclusion, the present study demonstrated that S1P treatment may ameliorate the cardiac hypertrophic response, which may be partly mediated by the suppression of HDAC2 activity and the upregulation of KLF4; it was suggested that S1PR2 may also be involved. Therefore, S1P may be considered a potential therapy for the treatment of heart diseases caused by cardiac hypertrophy.
Collapse
Affiliation(s)
- Hui Yan
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shaowei Yi
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hang Zhuang
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Lujin Wu
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dao Wen Wang
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jiangang Jiang
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
21
|
Fan J, Qiu L, Shu H, Ma B, Hagenmueller M, Riffel JH, Meryer S, Zhang M, Hardt SE, Wang L, Wang DW, Qiu H, Zhou N. Recombinant frizzled1 protein attenuated cardiac hypertrophy after myocardial infarction via the canonical Wnt signaling pathway. Oncotarget 2017; 9:3069-3080. [PMID: 29423029 PMCID: PMC5790446 DOI: 10.18632/oncotarget.23149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 11/15/2017] [Indexed: 01/20/2023] Open
Abstract
Postinfarct cardiac hypertrophy is an independent risk factor for heart failure and sudden death. Regression of cardiac hypertrophy has emerged as a promising strategy in the treatment of myocardial infarction (MI). Here we hypothesized that frizzled1 (FZD1), a receptor of the canonical Wnt signaling pathway, is a novel mediator of ischemia-associated cardiac hypertrophy. MI was induced in mice by left anterior descending (LAD) coronary occlusion. One week after MI, the expression of FZD1 was found to be notably increased in the left ventricles (LVs) of the MI-mice compared to shams. Mouse recombinant FZD1 protein (RFP) was subcutaneously injected in the mice to provoke autoimmunization response. Anti-FZD1 antibody titer was significantly increased in the plasma of RFP-treated mice. RFP significantly mitigated the MI-induced cardiac hypertrophy and improved cardiac function in the MI mouse hearts. Moreover, increased heart and LV weights, myocardial size and the expression of β-myosin heavy chain in the MI-mice were also found to be attenuated by RFP. FZD1 was found to be significantly up-regulated in hypoxia-treated neonatal rat cardiomyocytes (NRCMs). Silencing FZD1 by siRNA transfection notably repressed the hypoxia-induced myocardial hypertrophy in NRCMs. Mechanistically, activation of canonical Wnt signaling induced by MI, e.g., β-catenin and glycogen synthase kinase-3β, was restrained in the LVs of the MI-mice treated by RFP, these inhibition on canonical Wnt signaling was further confirmed in hypoxic NRCMs transfected with FZD1 siRNA. In conclusion, immunization of RFP attenuated cardiac hypertrophy and improved cardiac function in the MI mice via blocking the canonical Wnt signaling pathway.
Collapse
Affiliation(s)
- Jingjing Fan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China.,Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Lin Qiu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ben Ma
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | | | - Johannes H Riffel
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Soeren Meryer
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Min Zhang
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Stefan E Hardt
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | - Lin Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hongyu Qiu
- Division of Physiology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
22
|
Chiu YH, Ku PM, Cheng YZ, Li Y, Cheng JT, Niu HS. Phosphorylation of signal transducer and activator of transcription 3 induced by hyperglycemia is different with that induced by lipopolysaccharide or erythropoietin via receptor‑coupled signaling in cardiac cells. Mol Med Rep 2017; 17:1311-1320. [PMID: 29115516 DOI: 10.3892/mmr.2017.7973] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/25/2017] [Indexed: 11/06/2022] Open
Abstract
The signal transducer and activator of transcription 3 (STAT3) is known to be involved in hypertrophy and fibrosis in cardiac dysfunction. The activation of STAT3 via the phosphorylation of STAT3 is required for the production of functional activity. It has been established that lipopolysaccharide (LPS)‑induced phosphorylation of STAT3 in cardiomyocytes primarily occurs through a direct receptor‑mediated action. This effect is demonstrated to be produced rapidly. STAT3 in cardiac fibrosis of diabetes is induced by high glucose through promotion of the STAT3‑associated signaling pathway. However, the time schedule for STAT3 activation between LPS and high glucose appears to be different. Therefore, the difference in STAT3 activation between LPS and hyperglycemia in cardiomyocytes requires elucidation. The present study investigated the phosphorylation of STAT3 induced by LPS and hyperglycemia in the rat cardiac cell line H9c2. Additionally, phosphorylation of STAT3 induced by erythropoietin (EPO) via receptor activation was compared. Then, the downstream signals for fibrosis, including the connective tissue growth factor (CTGF) and matrix metalloproteinase (MMP)‑9, were determined using western blotting, while the mRNA levels were quantified. LPS induced a rapid elevation of STAT3 phosphorylation in H9c2 cells within 30 min, similar to that produced by EPO. However, LPS or EPO failed to modify the mRNA level of STAT3, and/or the downstream signals for fibrosis. High glucose increased STAT3 phosphorylation to be stable after a long period of incubation. Glucose incubation for 24 h may augment the STAT3 expression in a dose‑dependent manner. Consequently, fibrosis‑associated signals, including CTGF and MMP‑9 protein, were raised in parallel. In the presence of tiron, an antioxidant, these changes by hyperglycemia were markedly reduced, demonstrating the mediation of oxidative stress. Therefore, LPS‑ or EPO‑induced STAT3 phosphorylation is different compared with that caused by high glucose in H9c2 cells. Sustained activation of STAT3 by hyperglycemia may promote the expression of fibrosis‑associated signals, including CTGF and MMP‑9, in H9c2 cells. Therefore, regarding the cardiac dysfunctions associated with diabetes and/or hyperglycemia, the identification of nuclear STAT3 may be more reliable compared with the assay of phosphorylated STAT3 in cardiac cells.
Collapse
Affiliation(s)
- Yu-Hsin Chiu
- Division of Infectious Diseases, Chi‑Mei Medical Center‑Liouying, Tainan 73601, Taiwan, R.O.C
| | - Po-Ming Ku
- Cardiovascular Center, Department of Internal Medicine, Chi‑Mei Medical Center‑Liouying, Tainan 73601, Taiwan, R.O.C
| | - Yung-Ze Cheng
- Department of Emergency Medicine, Chi‑Mei Medical Center, Tainan 71003, Taiwan, R.O.C
| | - Yingxiao Li
- Department of Medical Research, Chi‑Mei Medical Center, Tainan 71003, Taiwan, R.O.C
| | - Juei-Tang Cheng
- Department of Medical Research, Chi‑Mei Medical Center, Tainan 71003, Taiwan, R.O.C
| | - Ho-Shan Niu
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien 97005, Taiwan, R.O.C
| |
Collapse
|
23
|
He Z, Yang Y, Wen Z, Chen C, Xu X, Zhu Y, Wang Y, Wang DW. CYP2J2 metabolites, epoxyeicosatrienoic acids, attenuate Ang II-induced cardiac fibrotic response by targeting Gα 12/13. J Lipid Res 2017; 58:1338-1353. [PMID: 28554983 DOI: 10.1194/jlr.m074229] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/23/2017] [Indexed: 12/23/2022] Open
Abstract
The arachidonic acid-cytochrome P450 2J2-epoxyeicosatrienoic acid (AA-CYP2J2-EET) metabolic pathway has been identified to be protective in the cardiovascular system. This study explored the effects of the AA-CYP2J2-EET metabolic pathway on cardiac fibrosis from the perspective of cardiac fibroblasts and underlying mechanisms. In in vivo studies, 8-week-old male CYP2J2 transgenic mice (aMHC-CYP2J2-Tr) and littermates were infused with angiotensin II (Ang II) or saline for 2 weeks. Results showed that CYP2J2 overexpression increased EET production. Meanwhile, impairment of cardiac function and fibrotic response were attenuated by CYP2J2 overexpression. The effects of CYP2J2 were associated with reduced activation of the α subunits of G12 family G proteins (Gα12/13)/RhoA/Rho kinase (ROCK) cascade and elevation of the NO/cyclic guanosine monophosphate (cGMP) level in cardiac tissue. In in vitro studies, cardiac fibroblast activation, proliferation, migration, and collagen production induced by Ang II were associated with activation of the Gα12/13/RhoA/ROCK pathway, which was inhibited by exogenous 11,12-EET. Moreover, silencing of Gα12/13 or RhoA exerted similar effects as 11,12-EET. Furthermore, inhibitory effects of 11,12-EET on Gα12/13 were blocked by NO/cGMP pathway inhibitors. Our findings indicate that enhancement of the AA-CYP2J2-EET metabolic pathway by CYP2J2 overexpression attenuates Ang II-induced cardiac dysfunction and fibrosis by reducing the fibrotic response of cardiac fibroblasts by targeting the Gα12/13/RhoA/ROCK pathway via NO/cGMP signaling.
Collapse
Affiliation(s)
- Zuowen He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yong Yang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Xizhen Xu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yanfang Zhu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yan Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China.
| |
Collapse
|
24
|
Heat shock transcription factor 1 protects against pressure overload-induced cardiac fibrosis via Smad3. J Mol Med (Berl) 2017; 95:445-460. [PMID: 28091697 PMCID: PMC5357304 DOI: 10.1007/s00109-016-1504-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 12/17/2016] [Accepted: 12/30/2016] [Indexed: 12/31/2022]
Abstract
Abstract Fibrotic cardiac muscle exhibits high stiffness and low compliance which are major risk factors of heart failure. Although heat shock transcription factor 1 (HSF1) was identified as an intrinsic cardioprotective factor, the role that HSF1 plays in cardiac fibrosis remains unclear. Our study aims to investigate the role of HSF1 in pressure overload-induced cardiac fibrosis and the underlying mechanism. HSF1 phosphorylation was significantly downregulated in transverse aortic constriction (TAC)-treated mouse hearts and mechanically stretched cardiac fibroblasts (cFBs). HSF1 transgenic (TG) mice, HSF1 deficient heterozygote (KO) mice, and their wild-type littermates were subjected to sham or TAC surgery for 4 weeks. HSF1 overexpression significantly attenuated pressure overload-induced cardiac fibrosis and dysfunction. Conversely, HSF1 KO mice showed deteriorated fibrotic response and cardiac dysfunction upon TAC. Moreover, we uncovered that overexpression of HSF1 protected against fibrotic response of cFBs to pressure overload. Mechanistically, we observed that the phosphorylation and the nuclear distribution of the Smad family member 3 (Smad3) were significantly decreased in HSF1-overexpressing mouse hearts, while being greatly increased in HSF1 KO mouse hearts upon TAC, compared to the control hearts, respectively. Similar alteration of Smad3 phosphorylation and nuclear distribution were found in isolated mouse cardiac fibroblasts and mechanically stretched cFBs. Constitutively active Smad3 blocked the anti-fibrotic effect of HSF1 in cFBs. Furthermore, we found a direct binding of phosphorylated HSF1 and Smad3, which can be suppressed by mechanical stress. In conclusion, the present study demonstrated for the first time that HSF1 acts as a novel negative regulator of cardiac fibrosis by blocking Smad3 activation. Key messages HSF1 activity is decreased in fibrotic hearts. HSF1 overexpression attenuates pressure overload-induced cardiac fibrosis and dysfunction. Deficiency of HSF1 deteriorates fibrotic response and cardiac dysfunction upon TAC. HSF1 inhibits phosphorylation and nuclear distribution of Smad3 via direct binding to Smad3. Active Smad3 blocks the anti-fibrotic effect of HSF1.
Electronic supplementary material The online version of this article (doi:10.1007/s00109-016-1504-2) contains supplementary material, which is available to authorized users.
Collapse
|
25
|
Yang Y, Hu W, Di S, Ma Z, Fan C, Wang D, Jiang S, Li Y, Zhou Q, Li T, Luo E. Tackling myocardial ischemic injury: the signal transducer and activator of transcription 3 (STAT3) at a good site. Expert Opin Ther Targets 2016; 21:215-228. [PMID: 28001439 DOI: 10.1080/14728222.2017.1275566] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yang Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, Xi’an, China
| | - Yue Li
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| | - Qing Zhou
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| | - Erping Luo
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
26
|
Targeting the prodromal stage of spinocerebellar ataxia type 17 mice: G-CSF in the prevention of motor deficits via upregulating chaperone and autophagy levels. Brain Res 2016; 1639:132-48. [PMID: 26972528 DOI: 10.1016/j.brainres.2016.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/13/2016] [Accepted: 03/03/2016] [Indexed: 12/12/2022]
Abstract
Spinocerebellar ataxia type 17 (SCA17), an autosomal dominant cerebellar ataxia, is a devastating, incurable disease caused by the polyglutamine (polyQ) expansion of transcription factor TATA binding protein (TBP). The polyQ expansion causes misfolding and aggregation of the mutant TBP, further leading to cytotoxicity and cell death. The well-recognized prodromal phase in many forms of neurodegeneration suggests a prolonged period of partial neuronal dysfunction prior to cell loss that may be amenable to therapeutic intervention. The objective of this study was to assess the effects and molecular mechanisms of granulocyte-colony stimulating factor (G-CSF) therapy during the pre-symptomatic stage in SCA17 mice. Treatment with G-CSF at the pre-symptomatic stage improved the motor coordination of SCA17 mice and reduced the cell loss, insoluble mutant TBP protein, and vacuole formation in the Purkinje neurons of these mice. The neuroprotective effects of G-CSF may be produced by increases in Hsp70, Beclin-1, LC3-II and the p-ERK survival pathway. Upregulation of chaperone and autophagy levels further enhances the clearance of mutant protein aggregation, slowing the progression of pathology in SCA17 mice. Therefore, we showed that the early intervention of G-CSF has a neuroprotective effect, delaying the progression of SCA17 in mutant mice via increases in the levels of chaperone expression and autophagy.
Collapse
|
27
|
Bostancı MS, Bakacak M, İnanc F, Yaylalı A, Serin S, Attar R, Yildirim ÖK, Yildirim G. The protective effect of G-CSF on experimental ischemia/reperfusion injury in rat ovary. Arch Gynecol Obstet 2015; 293:789-95. [DOI: 10.1007/s00404-015-3878-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 08/31/2015] [Indexed: 10/23/2022]
|
28
|
Serban AI, Stanca L, Geicu OI, Dinischiotu A. AGEs-Induced IL-6 Synthesis Precedes RAGE Up-Regulation in HEK 293 Cells: An Alternative Inflammatory Mechanism? Int J Mol Sci 2015; 16:20100-17. [PMID: 26307981 PMCID: PMC4613191 DOI: 10.3390/ijms160920100] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/12/2015] [Accepted: 08/06/2015] [Indexed: 01/21/2023] Open
Abstract
Advanced glycation end products (AGEs) can activate the inflammatory pathways involved in diabetic nephropathy. Understanding these molecular pathways could contribute to therapeutic strategies for diabetes complications. We evaluated the modulation of inflammatory and oxidative markers, as well as the protective mechanisms employed by human embryonic kidney cells (HEK 293) upon exposure to 200 μg/mL bovine serum albumine (BSA) or AGEs–BSA for 12, 24 and 48 h. The mRNA and protein expression levels of AGEs receptor (RAGE) and heat shock proteins (HSPs) 27, 60 and 70, the activity of antioxidant enzymes and the expression levels of eight cytokines were analysed. Cell damage via oxidative mechanisms was evaluated by glutathione and malondialdehyde levels. The data revealed two different time scale responses. First, the up-regulation of interleukin-6 (IL-6), HSP 27 and high catalase activity were detected as early as 12 h after exposure to AGEs–BSA, while the second response, after 24 h, consisted of NF-κB p65, RAGE, HSP 70 and inflammatory cytokine up-regulation, glutathione depletion, malondialdehyde increase and the activation of antioxidant enzymes. IL-6 might be important in the early ignition of inflammatory responses, while the cellular redox imbalance, RAGE activation and NF-κB p65 increased expression further enhance inflammatory signals in HEK 293 cells.
Collapse
Affiliation(s)
- Andreea Iren Serban
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, district 5, Bucharest 050097, Romania.
| | - Loredana Stanca
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, district 5, Bucharest 050097, Romania.
| | - Ovidiu Ionut Geicu
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomical Sciences and Veterinary Medicine Bucharest, 105 Splaiul Independentei, district 5, Bucharest 050097, Romania.
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, district 5, Bucharest 050095, Romania.
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, district 5, Bucharest 050095, Romania.
| |
Collapse
|
29
|
Dai M, Wu L, He Z, Zhang S, Chen C, Xu X, Wang P, Gruzdev A, Zeldin DC, Wang DW. Epoxyeicosatrienoic acids regulate macrophage polarization and prevent LPS-induced cardiac dysfunction. J Cell Physiol 2015; 230:2108-19. [PMID: 25626689 DOI: 10.1002/jcp.24939] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 01/16/2015] [Indexed: 12/14/2022]
Abstract
Macrophages, owning tremendous phenotypic plasticity and diverse functions, were becoming the target cells in various inflammatory, metabolic and immune diseases. Cytochrome P450 epoxygenase 2J2 (CYP2J2) metabolizes arachidonic acid to form epoxyeicosatrienoic acids (EETs), which possess various beneficial effects on cardiovascular system. In the present study, we evaluated the effects of EETs treatment on macrophage polarization and recombinant adeno-associated virus (rAAV)-mediated CYP2J2 expression on lipopolysaccharide (LPS)-induced cardiac dysfunction, and sought to investigate the underlying mechanisms. In vitro studies showed that EETs (1µmol/L) significantly inhibited LPS-induced M1 macrophage polarization and diminished the proinflammatory cytokines at transcriptional and post-transcriptional level; meanwhile it preserved M2 macrophage related molecules expression and upregulated anti-inflammatory cytokine IL-10. Furthermore, EETs down-regulated NF-κB activation and up-regulated peroxisome proliferator-activated receptors (PPARα/γ) and heme oxygenase 1 (HO-1) expression, which play important roles in regulating M1 and M2 polarization. In addition, LPS treatment in mice induced cardiac dysfunction, heart tissue damage and infiltration of M1 macrophages, as well as the increase of inflammatory cytokines in serum and heart tissue, but rAAV-mediated CYP2J2 expression increased EETs generation in heart and significantly attenuated the LPS-induced harmful effects, which mechanisms were similar as the in vitro study. Taken together, the results indicate that CYP2J2/EETs regulates macrophage polarization by attenuating NF-κB signaling pathway via PPARα/γ and HO-1 activation and its potential use in treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Meiyan Dai
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lujin Wu
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zuowen He
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shasha Zhang
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xizhen Xu
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peihua Wang
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Artiom Gruzdev
- Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina
| | - Dao Wen Wang
- Departments of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Granato M, Chiozzi B, Filardi MR, Lotti LV, Di Renzo L, Faggioni A, Cirone M. Tyrosine kinase inhibitor tyrphostin AG490 triggers both apoptosis and autophagy by reducing HSF1 and Mcl-1 in PEL cells. Cancer Lett 2015; 366:191-7. [PMID: 26184999 DOI: 10.1016/j.canlet.2015.07.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/01/2015] [Accepted: 07/08/2015] [Indexed: 01/09/2023]
Abstract
PEL cells relay on the constitutive activation of STAT3 for their survival, thus its inhibition by AG490 leads to apoptotic cell death. In this study, we found that the cytotoxic activity of AG490 correlated with the reduction of HSP70 and its master regulator HSF1 that, based on knocking-down experiments, was found to play a pro-survival role in PEL cells. To counteract the pro-death effect mediated by HSF1/HSP70 down-regulation, AG490 induced a complete autophagy, whose inhibition potentiated its cytotoxic effect against PEL cells. AG490 as well as HSF1 siRNA reduced the expression of Mcl-1, a Bcl-2 family member that negatively regulates apoptosis and autophagy. These results suggest that STAT3 inhibition, by down-regulating the expression of HSF1/HSP70, reduces Mcl-1 and leads to both apoptosis and autophagy induction in PEL cells.
Collapse
Affiliation(s)
- Marisa Granato
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161 Rome, Italy
| | - Barbara Chiozzi
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161 Rome, Italy
| | - Maria Rosaria Filardi
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161 Rome, Italy
| | - Lavinia Vittoria Lotti
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161 Rome, Italy
| | - Livia Di Renzo
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161 Rome, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161 Rome, Italy.
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, viale Regina Elena 324, 00161 Rome, Italy.
| |
Collapse
|
31
|
Urotensin II Protects Cardiomyocytes from Apoptosis Induced by Oxidative Stress through the CSE/H2S Pathway. Int J Mol Sci 2015; 16:12482-98. [PMID: 26047336 PMCID: PMC4490456 DOI: 10.3390/ijms160612482] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 05/22/2015] [Accepted: 05/22/2015] [Indexed: 02/07/2023] Open
Abstract
Plasma urotensin II (UII) has been observed to be raised in patients with acute myocardial infarction; suggesting a possible cardiac protective role for this peptide. However, the molecular mechanism is unclear. Here, we treated cultured cardiomyocytes with H2O2 to induce oxidative stress; observed the effect of UII on H2O2-induced apoptosis and explored potential mechanisms. UII pretreatment significantly reduced the number of apoptotic cardiomyocytes induced by H2O2; and it partly abolished the increase of pro-apoptotic protein Bax and the decrease of anti-apoptotic protein Bcl-2 in cardiomyocytes induced by H2O2. SiRNA targeted to the urotensin II receptor (UT) greatly inhibited these effects. Further analysis revealed that UII increased the production of hydrogen sulfide (H2S) and the level of cystathionine-γ-lyase (CSE) by activating the ERK signaling in H2O2-treated-cardiomyocytes. Si-CSE or ERK inhibitor not only greatly inhibited the increase in CSE level or the phosphorylation of ERK induced by UII but also reversed anti-apoptosis of UII in H2O2-treated-cadiomyocytes. In conclusion, UII rapidly promoted the phosphorylation of ERK and upregulated CSE level and H2S production, which in turn activated ERK signaling to protect cardiomyocytes from apoptosis under oxidative stress. These results suggest that increased plasma UII level may protect cardiomyocytes at the early-phase of acute myocardial infarction in patients.
Collapse
|
32
|
Cai Z, Shen L, Ma H, Yang J, Yang D, Chen H, Wei J, Lu Q, Wang DW, Xiang M, Wang J. Involvement of Endoplasmic Reticulum Stress-Mediated C/EBP Homologous Protein Activation in Coxsackievirus B3-Induced Acute Viral Myocarditis. Circ Heart Fail 2015; 8:809-18. [PMID: 25985795 DOI: 10.1161/circheartfailure.114.001244] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/07/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND This study tested the hypothesis whether endoplasmic reticulum (ER) stress/C/EBP homologous protein (CHOP) signaling is linked with coxsackievirus B3 (CVB3)-induced acute viral myocarditis (AVMC) in vivo. METHODS AND RESULTS AVMC was induced by intraperitoneal injection of 1000 tissue culture infectious dose (TCID50) of CVB3 virus in mice. In AVMC mouse hearts (n=11), ER stress and CHOP were significantly activated, and were linked to the induction of proapoptotic signaling including reduction of Bcl-2, activation of Bax and caspase 3, compared with the controls (n=10), whereas these could be markedly blocked by ER stress inhibitor tauroursodeoxycholic acid administration (n=11). Moreover, chemical inhibition of ER stress significantly attenuated cardiomyocytes apoptosis, and prevented cardiac troponin I elevation, ameliorated cardiac dysfunction assessed by both hemodynamic and echocardiographic analysis, reduced viral replication, and increased survival rate after CVB3 inoculation. We further discovered that genetic ablation of CHOP (n=10) suppressed cardiac Bcl-2/Bax ratio reduction and caspase 3 activation, and prevented cardiomyotes apoptosis in vivo, compared with wild-type receiving CVB3 inoculation (n=10). Strikingly, CHOP deficiency exhibited dramatic protective effects on cardiac damage, cardiac dysfunction, viral replication, and promoted survival in CVB3-caused AVMC. CONCLUSIONS Our data imply the involvement of ER stress/CHOP signaling in CVB3-induced AVMC via proapoptotic pathways, and provide a novel strategy for AVMC treatment.
Collapse
Affiliation(s)
- Zhejun Cai
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.)
| | - Li Shen
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.)
| | - Hong Ma
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.)
| | - Jin Yang
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.)
| | - Du Yang
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.)
| | - Han Chen
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.)
| | - Jia Wei
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.)
| | - Qiulun Lu
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.)
| | - Dao Wen Wang
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.)
| | - Meixiang Xiang
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.).
| | - Jian'an Wang
- From the Key Laboratory of Cardiovascular Disease of Zhejiang Province and Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China (Z.C., L.S., H.M., D.Y., H.C., M.X., J. Wang); Department of Medicine, Blood Center of Zhejiang Province, Hangzhou, China (J.Y.); Transform Medical Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China (J.Y.); Department of Pediatric Surgery (J. Wei) and Institute of Hypertension and Department of Internal Medicine (D.W.W.), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.L.).
| |
Collapse
|
33
|
He Z, Zhang X, Chen C, Wen Z, Hoopes SL, Zeldin DC, Wang DW. Cardiomyocyte-specific expression of CYP2J2 prevents development of cardiac remodelling induced by angiotensin II. Cardiovasc Res 2015; 105:304-17. [PMID: 25618409 DOI: 10.1093/cvr/cvv018] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS Cardiac remodelling is one of the key pathological changes that occur with cardiovascular disease. Previous studies have demonstrated the beneficial effects of CYP2J2 expression on cardiac injury. In the present study, we investigated the effects of cardiomyocyte-specific CYP2J2 expression and EET treatment on angiotensin II-induced cardiac remodelling and sought to determine the underlying molecular mechanisms involved in this process. METHODS AND RESULTS Eight-week-old mice with cardiomyocyte-specific CYP2J2 expression (αMHC-CYP2J2-Tr) and wild-type (WT) control mice were treated with Ang-II. Ang-II treatment of WT mice induced changes in heart morphology, cardiac hypertrophy and dysfunction, as well as collagen accumulation; however, cardiomyocyte-specific expression of CYP2J2 attenuated these effects. The cardioprotective effects observed in α-MHC-CYP2J2-Tr mice were associated with peroxisome proliferator-activated receptor (PPAR)-γ activation, reduced oxidative stress, reduced NF-κB p65 nuclear translocation, and inhibition of TGF-β1/smad pathway. The effects seen with cardiomyocyte-specific expression of CYP2J2 were partially blocked by treatment with PPAR-γ antagonist GW9662. In in vitro studies, 11,12-EET(1 μmol/L) treatment attenuated cardiomyocyte hypertrophy and remodelling-related protein (collagen I, TGF-β1, TIMP1) expression by inhibiting the oxidative stress-mediated NF-κB pathway via PPAR-γ activation. Furthermore, conditioned media from neonatal cardiomyocytes treated with 11,12-EET inhibited activation of cardiac fibroblasts and TGF-β1/smad pathway. CONCLUSION Cardiomyocyte-specific expression of CYP2J2 or treatment with EETs protects against cardiac remodelling by attenuating oxidative stress-mediated NF-κBp65 nuclear translocation via PPAR-γ activation.
Collapse
Affiliation(s)
- Zuowen He
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan 430030, P. R. China
| | - Xu Zhang
- Department of Physiology, Tianjin Medical University, Tianjin, P. R. China
| | - Chen Chen
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan 430030, P. R. China
| | - Zheng Wen
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan 430030, P. R. China
| | - Samantha L Hoopes
- Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Dao Wen Wang
- Department of Internal Medicine and Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Avenue, Wuhan 430030, P. R. China
| |
Collapse
|
34
|
Bocchini CE, Kasembeli MM, Roh SH, Tweardy DJ. Contribution of chaperones to STAT pathway signaling. JAKSTAT 2014; 3:e970459. [PMID: 26413421 DOI: 10.4161/21623988.2014.970459] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 09/21/2014] [Accepted: 09/25/2014] [Indexed: 12/27/2022] Open
Abstract
Aberrant STAT signaling is associated with the development and progression of many cancers and immune related diseases. Recent findings demonstrate that proteostasis modulators under clinical investigation for cancer therapy have a significant impact on STAT signaling, which may be critical for mediating their anti-cancer effects. Chaperones are critical for protein folding, stability and function and, thus, play an essential role in the maintenance of proteostasis. In this review we discuss the role of chaperones in STAT and tyrosine kinase (TK) protein folding, modulation of STAT and TK activity, and degradation of TKs. We highlight the important role of chaperones in STAT signaling, and how this knowledge has provided a framework for the development of new therapeutic avenues of targeting STAT signaling related pathologies.
Collapse
Affiliation(s)
- Claire E Bocchini
- Section of Infectious Disease; Department of Pediatrics; Baylor College of Medicine ; Houston, TX USA
| | - Moses M Kasembeli
- Section of Infectious Disease; Department of Medicine; Baylor College of Medicine ; Houston, TX USA
| | - Soung-Hun Roh
- Department of Biochemistry & Molecular Biology; Baylor College of Medicine ; Houston, TX USA
| | - David J Tweardy
- Section of Infectious Disease; Department of Medicine; Baylor College of Medicine ; Houston, TX USA ; Department of Biochemistry & Molecular Biology; Baylor College of Medicine ; Houston, TX USA ; Department of Molecular & Cellular Biology; Baylor College of Medicine ; Houston, TX USA
| |
Collapse
|
35
|
He Q, Pu J, Yuan A, Lau WB, Gao E, Koch WJ, Ma XL, He B. Activation of liver-X-receptor α but not liver-X-receptor β protects against myocardial ischemia/reperfusion injury. Circ Heart Fail 2014; 7:1032-41. [PMID: 25277999 DOI: 10.1161/circheartfailure.114.001260] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Liver-X-receptors, LXRα (NR1H3) and LXRβ (NR1H2), encode 2 different but highly homologous isoforms of transcription factors belonging to the nuclear receptor superfamily. Whether LXRα and LXRβ subtypes have discrete roles in the regulation of cardiac physiology/pathology is unknown. We determine the role of each LXR subtype in myocardial ischemia/reperfusion (MI/R) injury. METHODS AND RESULTS Mice (wild type; those genetically depleted of LXRα, LXRβ, or both; and those overexpressing LXRα or LXRβ by in vivo intramyocardial adenoviral vector) were subjected to MI/R injury. Both LXRα and LXRβ were detected in wild-type mouse heart. LXRα, but not LXRβ, was significantly upregulated after MI/R. Dual activation of LXRα and LXRβ by natural and synthetic agonists reduced myocardial infarction and improved contractile function after MI/R. Mechanistically, LXR activation inhibited MI/R-induced oxidative stress and nitrative stress, attenuated endoplasmic reticulum stress and mitochondrial dysfunction, and reduced cardiomyocyte apoptosis in ischemic/reperfused myocardium. The aforementioned cardioprotective effects of LXR agonists were impaired in the setting of cardiac-specific gene silencing of LXRα, but not LXRβ subtype. Moreover, LXRα/β double-knockout and LXRα-knockout mice, but not LXRβ-knockout mice, increased MI/R injury, exacerbated MI/R-induced oxidative/nitrative stress, and aggravated endoplasmic reticulum stress and mitochondrial dysfunction. Furthermore, cardiac LXRα, not LXRβ, overexpression via adenoviral transfection suppressed MI/R injury. CONCLUSIONS Our study provides the first direct evidence that the LXRα, but not LXRβ, subtype is a novel endogenous cardiac protective receptor against MI/R injury. Drug development strategies specifically targeting LXRα may be beneficial in treating ischemic heart disease.
Collapse
Affiliation(s)
- Qing He
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.)
| | - Jun Pu
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.).
| | - Ancai Yuan
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.)
| | - Wayne Bond Lau
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.)
| | - Erhe Gao
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.)
| | - Walter J Koch
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.)
| | - Xin-Liang Ma
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.).
| | - Ben He
- From the Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Q.H., J.P., A.Y., B.H.); Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (W.B.L., X.-L.M.); and Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA (E.G., W.J.K.).
| |
Collapse
|
36
|
Zamperone A, Pietronave S, Colangelo D, Antonini S, Locatelli M, Travaglia F, Coïsson JD, Arlorio M, Prat M. Protective effects of clovamide against H2O2-induced stress in rat cardiomyoblasts H9c2 cell line. Food Funct 2014; 5:2542-51. [PMID: 25133994 DOI: 10.1039/c4fo00195h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cocoa contains phenolic compounds with known antioxidant and antiradical properties beneficial in different pathologies, including cardiovascular diseases. Herein, we have evaluated the protective effects of clovamide, a minor cocoa component, against oxidative stress induced in the rat cardiomyoblast cell line, also comparing it to its bio-isosteric form, rosmarinic acid, and to the main monomeric flavan-3-ol from low-molecular-weight polyphenol in cocoa, i.e. epicatechin. At nano-micro-molar concentrations, the three compounds inhibited the production of reactive oxygen species and apoptosis, evaluated under different aspects, namely, annexin V positivity, DNA fragmentation, caspase release and activation. These molecules can, thus, be considered for their bioactive beneficial activity in the context of cardiovascular pathologies and, particularly, in the protection towards oxidative stress that follows ischemic injury. Clovamide may, thus, be the primary compound for the development of innovative nutraceutical strategies towards cardiovascular diseases.
Collapse
Affiliation(s)
- Andrea Zamperone
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Fiaschi T, Magherini F, Gamberi T, Lucchese G, Faggian G, Modesti A, Modesti PA. Hyperglycemia and angiotensin II cooperate to enhance collagen I deposition by cardiac fibroblasts through a ROS-STAT3-dependent mechanism. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2603-10. [PMID: 25072659 DOI: 10.1016/j.bbamcr.2014.07.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 01/09/2023]
Abstract
Cardiac fibroblasts significantly contribute to diabetes-induced structural and functional changes in the myocardium. The objective of the present study was to determine the effects of high glucose (alone or supplemented with angiotensin II) in the activation of the JAK2/STAT3 pathway and its involvement in collagen I production by cardiac fibroblasts. We observed that the diabetic environment 1) enhanced tyrosine phosphorylation of JAK2 and STAT3; 2) induced nuclear localization of tyrosine phosphorylated STAT3 through a reactive oxygen species-mediated mechanism, with angiotensin II stimulation further enhancing STAT3 nuclear accumulation; and 3) stimulated collagen I production. The effects were inhibited by depletion of reactive oxygen species or silencing of STAT3 in high glucose alone or supplemented with exogenous angiotensin II. Combined, our data demonstrate that increased collagen I deposition in the setting of high glucose occurred through a reactive oxygen species- and STAT3-dependent mechanism. Our results reveal a novel role for STAT3 as a key signaling molecule of collagen I production in cardiac fibroblasts exposed to a diabetic environment.
Collapse
Affiliation(s)
- Tania Fiaschi
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Francesca Magherini
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Tania Gamberi
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Gianluca Lucchese
- Institute of Thoracic and Cardiovascular Surgery, University of Verona, Verona, Italy
| | - Giuseppe Faggian
- Institute of Thoracic and Cardiovascular Surgery, University of Verona, Verona, Italy
| | - Alessandra Modesti
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy.
| | - Pietro Amedeo Modesti
- Department of Clinical and Experimental Medicine, University of Florence, School of Medicine, Florence, Italy.
| |
Collapse
|
38
|
Meshkibaf S, William Gower M, Dekaban GA, Ouk Kim S. G-CSF preferentially supports the generation of gut-homing Gr-1high macrophages in M-CSF-treated bone marrow cells. J Leukoc Biol 2014; 96:549-561. [DOI: 10.1189/jlb.1a0314-172r] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
AbstractThe G-CSF is best known for its activity in the generation and activation of neutrophils. In addition, studies on G-CSF−/− or G-CSFR−/− mice and BMC cultures suggested a role of G-CSF in macrophage generation. However, our understanding on the role of G-CSF in macrophage development is limited. Here, using in vitro BMC models, we demonstrated that G-CSF promoted the generation of Gr-1high/F4/80+ macrophage-like cells in M-BMCs, likely through suppressing cell death and enhancing generation of Gr-1high/F4/80+ macrophage-like cells. These Gr-1high macrophage-like cells produced “M2-like” cytokines and surface markers in response to LPS and IL-4/IL-13, respectively. Adoptive transfer of EGFP-expressing (EGFP+) M-BMCs showed a dominant, gut-homing phenotype. The small intestinal lamina propria of G-CSFR−/− mice also harbored significantly reduced numbers of Gr-1high/F4/80+ macrophages compared with those of WT mice, but levels of Gr-1+/F4/80− neutrophil-like cells were similar between these mice. Collectively, these results suggest a novel function of G-CSF in the generation of gut-homing, M2-like macrophages.
Collapse
Affiliation(s)
- Shahab Meshkibaf
- Department of Microbiology and Immunology , London, Ontario, Canada
- Center for Human Immunology, and Schulich School of Medicine and Dentistry , London, Ontario, Canada
| | - Mark William Gower
- Department of Microbiology and Immunology , London, Ontario, Canada
- Center for Human Immunology, and Schulich School of Medicine and Dentistry , London, Ontario, Canada
| | - Gregory A Dekaban
- Department of Microbiology and Immunology , London, Ontario, Canada
- Department of Molecular Medicine, Robarts Research Institute, University of Western Ontario , London, Ontario, Canada
| | - Sung Ouk Kim
- Department of Microbiology and Immunology , London, Ontario, Canada
- Center for Human Immunology, and Schulich School of Medicine and Dentistry , London, Ontario, Canada
| |
Collapse
|
39
|
Xiao X, Moreno-Moral A, Rotival M, Bottolo L, Petretto E. Multi-tissue analysis of co-expression networks by higher-order generalized singular value decomposition identifies functionally coherent transcriptional modules. PLoS Genet 2014; 10:e1004006. [PMID: 24391511 PMCID: PMC3879165 DOI: 10.1371/journal.pgen.1004006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 10/22/2013] [Indexed: 12/27/2022] Open
Abstract
Recent high-throughput efforts such as ENCODE have generated a large body of genome-scale transcriptional data in multiple conditions (e.g., cell-types and disease states). Leveraging these data is especially important for network-based approaches to human disease, for instance to identify coherent transcriptional modules (subnetworks) that can inform functional disease mechanisms and pathological pathways. Yet, genome-scale network analysis across conditions is significantly hampered by the paucity of robust and computationally-efficient methods. Building on the Higher-Order Generalized Singular Value Decomposition, we introduce a new algorithmic approach for efficient, parameter-free and reproducible identification of network-modules simultaneously across multiple conditions. Our method can accommodate weighted (and unweighted) networks of any size and can similarly use co-expression or raw gene expression input data, without hinging upon the definition and stability of the correlation used to assess gene co-expression. In simulation studies, we demonstrated distinctive advantages of our method over existing methods, which was able to recover accurately both common and condition-specific network-modules without entailing ad-hoc input parameters as required by other approaches. We applied our method to genome-scale and multi-tissue transcriptomic datasets from rats (microarray-based) and humans (mRNA-sequencing-based) and identified several common and tissue-specific subnetworks with functional significance, which were not detected by other methods. In humans we recapitulated the crosstalk between cell-cycle progression and cell-extracellular matrix interactions processes in ventricular zones during neocortex expansion and further, we uncovered pathways related to development of later cognitive functions in the cortical plate of the developing brain which were previously unappreciated. Analyses of seven rat tissues identified a multi-tissue subnetwork of co-expressed heat shock protein (Hsp) and cardiomyopathy genes (Bag3, Cryab, Kras, Emd, Plec), which was significantly replicated using separate failing heart and liver gene expression datasets in humans, thus revealing a conserved functional role for Hsp genes in cardiovascular disease.
Collapse
Affiliation(s)
- Xiaolin Xiao
- Medical Research Council (MRC) Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Aida Moreno-Moral
- Medical Research Council (MRC) Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Maxime Rotival
- Medical Research Council (MRC) Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Leonardo Bottolo
- Department of Mathematics, Imperial College, London, United Kingdom
| | - Enrico Petretto
- Medical Research Council (MRC) Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Henderson B, Kaiser F. Do reciprocal interactions between cell stress proteins and cytokines create a new intra-/extra-cellular signalling nexus? Cell Stress Chaperones 2013; 18:685-701. [PMID: 23884786 PMCID: PMC3789882 DOI: 10.1007/s12192-013-0444-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/16/2013] [Accepted: 06/17/2013] [Indexed: 12/22/2022] Open
Abstract
Cytokine biology began in the 1950s, and by 1988, a large number of cytokines, with a myriad of biological actions, had been discovered. In 1988, the basis of the protein chaperoning function of the heat shock, or cell stress, proteins was identified, and it was assumed that this was their major activity. However, since this time, evidence has accumulated to show that cell stress proteins are secreted by cells and can stimulate cellular cytokine synthesis with the generation of pro- and/or anti-inflammatory cytokine networks. Cell stress can also control cytokine synthesis, and cytokines are able to induce, or even inhibit, the synthesis of selected cell stress proteins and may also promote their release. How cell stress proteins control the formation of cytokines is not understood and how cytokines control cell stress protein synthesis depends on the cellular compartment experiencing stress, with cytoplasmic heat shock factor 1 (HSF1) having a variety of actions on cytokine gene transcription. The endoplasmic reticulum unfolded protein response also exhibits a complex set of behaviours in terms of control of cytokine synthesis. In addition, individual intracellular cell stress proteins, such as Hsp27 and Hsp90, have major roles in controlling cellular responses to cytokines and in controlling cytokine synthesis in response to exogenous factors. While still confusing, the literature supports the hypothesis that cell stress proteins and cytokines may generate complex intra- and extra-cellular networks, which function in the control of cells to external and internal stressors and suggests the cell stress response as a key parameter in cytokine network generation and, as a consequence, in control of immunity.
Collapse
Affiliation(s)
- Brian Henderson
- />Department of Microbial Diseases, Eastman Dental Institute, University College London, London, UK
| | - Frank Kaiser
- />Department of Microbial Diseases, Eastman Dental Institute, University College London, London, UK
- />Division of Microbial Diseases, Eastman Dental Institute, University College London, 256 Gray’s Inn Road, London, WC1X 8LD UK
| |
Collapse
|
41
|
Ma B, Xiong X, Chen C, Li H, Xu X, Li X, Li R, Chen G, Dackor RT, Zeldin DC, Wang DW. Cardiac-specific overexpression of CYP2J2 attenuates diabetic cardiomyopathy in male streptozotocin-induced diabetic mice. Endocrinology 2013; 154:2843-56. [PMID: 23696562 PMCID: PMC3713213 DOI: 10.1210/en.2012-2166] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to biologically active cis-epoxyeicosatrienoic acids, which have potent vasodilatory, antiinflammatory, antiapoptotic, and antidiabetes properties. Here, we showed the effects of cardiac-specific overexpression of CYP epoxygenase 2J2 (CYP2J2) on diabetic cardiomyopathy and insulin resistance in high-fat (HF) diet fed, low-dose streptozotocin-treated mice. Diabetic cardiomyopathy was induced by HF and streptozotocin in cardiac-specific CYP2J2 transgenic mice. Physiological parameters and systemic metabolic parameters were monitored using ELISA kits. Intraperitoneal injection glucose tolerance test and hyperinsulinemic-euglycemic clamp study were implied to indicate insulin resistance. Cardiac function was assessed by echocardiography and Millar catheter system. Real-time PCR and Western blotting were used in signal pathway detection. αMHC-CYP2J2 transgenic mice showed significantly lower plasma glucose and insulin levels, improved glucose tolerance, and increased cardiac glucose uptake. Furthermore, αMHC-CYP2J2 transgenic mice were significantly protected from HF-streptozotocin-induced diabetic cardiomyopathy. Strikingly, CYP2J2 overexpression attenuated myocardial hypertrophy induced by diabetes. We conclude that cardiac-specific overexpression of CYP2J2 significantly protects against diabetic cardiomyopathy, which may be due to improved cardiac insulin resistance, glucose uptake, and reversal of cardiac hypertrophy. Relevant mechanisms may include up-regulation of peroxisome proliferator-activated receptor γ, activation of insulin receptor and AMP-activated protein kinase signaling pathways, and inhibition of nuclear factor of activated T cells c3 signal by enhanced atrial natriuretic peptide production. These results suggest that CYP2J2 epoxygenase metabolites likely play an important role in plasma glucose homeostasis, and enhancement of epoxyeicosatrienoic acids activation may serve as an effective therapeutic strategy to prevent diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Ben Ma
- The Institute of Hypertension and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People’s Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Cytokines are important mediators of host defense and immunity, and were first identified for their role in immunity to infections. It was then found that some of them are pathogenic mediators in inflammatory diseases and much of the emphasis is now on pro-inflammatory cytokines, also in consideration of the fact that TNF inhibitors became effective drugs in chronic inflammatory diseases. The recent studies on the tissue-protective activities of erythropoietin (EPO) led to the term "tissue-protective cytokine." We discuss here how tissue-protective actions might be common to other cytokines, particularly those of the 4-alpha helical structural superfamily.
Collapse
|
43
|
Fragoso MA, Patel AK, Nakamura REI, Yi H, Surapaneni K, Hackam AS. The Wnt/β-catenin pathway cross-talks with STAT3 signaling to regulate survival of retinal pigment epithelium cells. PLoS One 2012; 7:e46892. [PMID: 23056515 PMCID: PMC3464242 DOI: 10.1371/journal.pone.0046892] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 09/10/2012] [Indexed: 12/23/2022] Open
Abstract
Wnt/β-catenin signaling is an essential pathway that regulates numerous cellular processes, including cell survival. The molecular mechanisms contributing to pro-survival Wnt signaling are mostly unknown. Signal transducer and activator of transcription proteins (STATs) are a well-described family of transcription factors. STAT3 induces expression of anti-apoptotic genes in many tissues and is a downstream mediator of protective growth factors and cytokines. In this study, we investigated whether pro-survival Wnt signaling is mediated by STAT3. The Wnt3a ligand activated Wnt signaling in the retinal pigment epithelium ARPE-19 cell line and significantly increased the viability of cells exposed to oxidative stress. Furthermore, Wnt3a increased STAT3 activation and nuclear translocation, as measured by an antibody against phosphorylated STAT3. Reducing STAT3 levels with siRNA eliminated Wnt3a-dependent protection from oxidative stress. Together, these data demonstrate a previously unknown link between Wnt3a-mediated activation of STAT3 and cell survival, and indicate cross-talk between two important pro-survival signaling pathways.
Collapse
Affiliation(s)
- Miryam A. Fragoso
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Amit K. Patel
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Rei E. I. Nakamura
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Hyun Yi
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Krishna Surapaneni
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Abigail S. Hackam
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|