51
|
Curdione Ameliorated Doxorubicin-Induced Cardiotoxicity Through Suppressing Oxidative Stress and Activating Nrf2/HO-1 Pathway. J Cardiovasc Pharmacol 2019; 74:118-127. [DOI: 10.1097/fjc.0000000000000692] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
52
|
Liu F, Zhang H, Li Y, Lu X. Nobiletin suppresses oxidative stress and apoptosis in H9c2 cardiomyocytes following hypoxia/reoxygenation injury. Eur J Pharmacol 2019; 854:48-53. [DOI: 10.1016/j.ejphar.2019.03.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/19/2019] [Accepted: 03/28/2019] [Indexed: 01/10/2023]
|
53
|
Cui C, Li Z, Wu D. The long non-coding RNA H19 induces hypoxia/reoxygenation injury by up-regulating autophagy in the hepatoma carcinoma cells. Biol Res 2019; 52:32. [PMID: 31196153 PMCID: PMC6567522 DOI: 10.1186/s40659-019-0239-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/25/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Long non-coding RNA H19 (H19) plays an important role by regulating protein expression in different tissues and organs of the body. However, whether H19 induces hypoxia/reoxygenation (h/R) injury via increase of autophagy in the hepatoma carcinoma cells is unknown. RESULTS H19 was expressed in the hepatoma carcinoma cells (Hep G2 and HCCLM3 cells) and its expression was most in 8 h/24R. The knockdown of H19 and 3-MA (an autophagy inhibitor) protected against h/R-induced apoptosis, cell damage, the expression of cleaved caspase-3 and cleaved caspase-9, the release of cytochrome c (Cyt c). The knockdown of H19 and 3-MA also decreased the autophagic vesicles (AVs) and the expression of Beclin-1 and the ration of LC3-II/LC3-I, and increased cell viability, the expression of Bcl-2 and P62 and the phosphorylation of PI3K, Akt and mTOR. In addition, chloroquine (CQ, an inhibitor of autophagy flux) markedly decreased formation of autophagy flux (the ration of LC3-II/LC3-I). The results of the knockdown of H19 group were similar to those of the 3-MA (or CQ) group. Rapamycin (a mTOR inhibitor, an autophagy activator) further down-regulated h/R-induced decrease of the phosphorylated PI3K, Akt and mTOR. The knockdown of H19 cancelled the effect of rapamycin. The overexpression of H19 further expanded h/R-induced increase of the ration of LC3-II/LC3-I and decrease of the phosphorylated PI3K, Akt and mTOR. CONCLUSIONS Our results suggest that the long non-coding RNA H19 induces h/R injury by up-regulation of autophagy via activation of PI3K-Akt-mTOR pathway in the hepatoma carcinoma cells.
Collapse
Affiliation(s)
- Chao Cui
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Baojian Road, Harbin, 150086, China
| | - Zhiyu Li
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Baojian Road, Harbin, 150086, China
| | - Dequan Wu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Baojian Road, Harbin, 150086, China.
| |
Collapse
|
54
|
Yasuda J, Okada M, Yamawaki H. Protective effect of T3 peptide, an active fragment of tumstatin, against ischemia/reperfusion injury in rat heart. J Pharmacol Sci 2019; 139:193-200. [PMID: 30827890 DOI: 10.1016/j.jphs.2019.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/11/2019] [Accepted: 01/17/2019] [Indexed: 12/28/2022] Open
Abstract
Ischemia/reperfusion (I/R)-induced oxidative stress is a serious clinical problem in the reperfusion therapy for ischemic diseases. Tumstatin is an endogenous bioactive peptide cleaved from type IV collagen α3 chain. We previously reported that T3 peptide, an active subfragment of tumstatin, exerts cytoprotective effects on H2O2-induced apoptosis through the inhibition of intracellular reactive oxygen species (ROS) production in H9c2 cardiomyoblasts. In this study, we investigated whether T3 peptide has cardioprotective effects against I/R injury by using in vitro and ex vivo experimental models. H9c2 cardiomyoblasts were stimulated with oxygen and glucose deprivation (OGD) for 12 h followed by reoxygenation for 1-8 h (OGD/R; in vitro model). The cells were treated with T3 peptide (30-1000 ng/ml) during OGD. Ten minutes after the pre-perfusion of T3 peptide (300 ng/ml), Langendorff perfused rat hearts were exposed to ischemia for 30 min followed by reperfusion for 1 h (ex vivo model). T3 peptide inhibited OGD/R-induced apoptosis through the inhibition of mitochondrial ROS production and dysfunction in H9c2 cardiomyoblasts. T3 peptide also prevented I/R-induced cardiac dysfunction, arrhythmia and myocardial infarction in the perfused rat heart. In conclusion, we for the first time demonstrated that T3 peptide exerts cardioprotective effects against I/R injury.
Collapse
Affiliation(s)
- Jumpei Yasuda
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 Bancho 35-1, Towada City, Aomori, 034-8628, Japan
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 Bancho 35-1, Towada City, Aomori, 034-8628, Japan.
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Higashi 23 Bancho 35-1, Towada City, Aomori, 034-8628, Japan
| |
Collapse
|
55
|
The lncRNA H19/miR-675 axis regulates myocardial ischemic and reperfusion injury by targeting PPARα. Mol Immunol 2019; 105:46-54. [DOI: 10.1016/j.molimm.2018.11.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 10/24/2018] [Accepted: 11/14/2018] [Indexed: 12/21/2022]
|
56
|
Cowley PM, Wang G, Swigart PM, Raghunathan A, Reddy N, Dulam P, Lovett DH, Simpson PC, Baker AJ. Reversal of right ventricular failure by chronic α 1A-subtype adrenergic agonist therapy. Am J Physiol Heart Circ Physiol 2019; 316:H224-H232. [PMID: 30412439 PMCID: PMC6859419 DOI: 10.1152/ajpheart.00507.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/01/2018] [Accepted: 11/06/2018] [Indexed: 12/31/2022]
Abstract
Right ventricular (RV) failure (RVF) is a serious disease with no effective treatment available. We recently reported a disease prevention study showing that chronic stimulation of α1A-adrenergic receptors (α1A-ARs), started at the time of RV injury, prevented the development of RVF. The present study used a clinically relevant disease reversal design to test if chronic α1A-AR stimulation, started after RVF was established, could reverse RVF. RVF was induced surgically by pulmonary artery constriction in mice. Two weeks after pulmonary artery constriction, in vivo RV fractional shortening as assessed by MRI was reduced by half relative to sham-operated controls (25 ± 2%, n = 27, vs. 52 ± 2%, n = 13, P < 10-11). Subsequent chronic treatment with the α1A-AR agonist A61603 for a further 2 wk resulted in a substantial recovery of RV fractional shortening (to 41 ± 2%, n = 17, P < 10-7 by a paired t-test) along with recovery of voluntary exercise capacity. Mechanistically, chronic A61603 treatment resulted in increased activation of the prosurvival kinase ERK, increased abundance of the antiapoptosis factor Bcl-2, and decreased myocyte necrosis evidenced by a decreased serum level of cardiac troponin. Moreover, A61603 treatment caused increased abundance of the antioxidant glutathione peroxidase-1, decreased level of reactive oxygen species, and decreased oxidative modification (carbonylation) of myofilament proteins. Consistent with these effects, A61603 treatment resulted in increased force development by cardiac myofilaments, which might have contributed to increased RV function. These findings suggest that the α1A-AR is a therapeutic target to reverse established RVF. NEW & NOTEWORTHY Currently, there are no effective therapies for right ventricular (RV) failure (RVF). This project evaluated a novel therapy for RVF. In a mouse model of RVF, chronic stimulation of α1A-adrenergic receptors with the agonist A61603 resulted in recovery of in vivo RV function, improved exercise capacity, reduced oxidative stress-related carbonylation of contractile proteins, and increased myofilament force generation. These results suggest that the α1A-adrenergic receptor is a therapeutic target to treat RVF.
Collapse
Affiliation(s)
- Patrick M Cowley
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Guanying Wang
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Philip M Swigart
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Anaha Raghunathan
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Nikitha Reddy
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Pranavi Dulam
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - David H Lovett
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Paul C Simpson
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| | - Anthony J Baker
- Veterans Affairs Medical Center, San Francisco, California, and Department of Medicine, University of California , San Francisco, California
| |
Collapse
|
57
|
Roles of apoptosis and inflammation in a rat model of acute lung injury induced right ventricular dysfunction. Biomed Pharmacother 2018; 108:1105-1114. [PMID: 30372811 DOI: 10.1016/j.biopha.2018.09.115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 12/17/2022] Open
Abstract
AIM The effect of intratracheal lipopolysaccharide (LPS) instillation on right ventricular dysfunction in rats was studied with the aim of exploring underlying mechanisms. MAIN METHODS A single dose of LPS (10 mg/kg) or an equal volume of saline was instilled intratracheally and lung injury evaluated using histopathologic scoring and wet/dry (W/D) weight ratio at 6 or 12 h post-administration. Besides, serum atrial natriuretic peptide (ANP) was detected using an enzyme-linked immunosorbent assay (ELISA) and right ventricle β-myosin heavy chain (β-MHC) presence was examined using reverse transcription polymerase chain reaction (RT-PCR). Echocardiography examined pulmonary artery acceleration time (PAAT), right ventricular free wall thickness (RVFWT), tricuspid annular plane systolic excursion (TAPSE), and right ventricular end-diastolic diameter (RVEDD). In addition, right ventricular TUNEL staining and Western blots of Bax and Bcl-2 were performed. Right ventricle and left ventricle caspases-3, -8, and -9 activity were examined using fluorometric assay. Finally, right ventricle myeloperoxidase (MPO) neutrophil staining, and right ventricle and plasma cytokines TNF-α, IL-1β, IL-6 detection was performed. KEY FINDINGS Histopathologic lung injury and increased W/D weight ratio was seen at 6 h after LPS intratracheal instillation, along with increased ANP, but not β-MHC. Pulmonary hypertension was indicated by decreased PAAT at 6 h post-exposure. Right ventricular systolic dysfunction and dilation were observed at 12 h post-exposure, as indicated by a significant decrease of TAPSE and increase of RVEDD. Of note, the procedure led to an increased right ventricle TUNEL positive cardiomyocytes, an increased Bax/Bcl-2 ratio, and increased right and left ventricle caspases-3, -8, and -9 activity as early as 6 h post-exposure, which was paralleled by increased right ventricle MPO staining and increased expression of right ventricle and serum cytokines TNF-α, IL-1β, and IL-6. SIGNIFICANCE As well as acute lung injury, a single dose of LPS intratracheally instilled can induce pulmonary hypertension at 6 h post-exposure, with obvious right ventricular systolic dysfunction and right ventricular dilation present at 12 h post-exposure, possibly via cardiomyocytes apoptosis and inflammation.
Collapse
|
58
|
A P, P SR, M PR, K G R. Apoptosis in angiotensin II-stimulated hypertrophic cardiac cells -modulation by phenolics rich extract of Boerhavia diffusa L. Biomed Pharmacother 2018; 108:1097-1104. [PMID: 30372810 DOI: 10.1016/j.biopha.2018.09.114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/19/2018] [Accepted: 09/19/2018] [Indexed: 02/04/2023] Open
Abstract
Herein, we investigated the effects of B. diffusa (BDE), a well-known cardiotonic edible medicinal plant against apoptosis in Angiotensin II (Ang II)-stimulated hypertrophic cardiac cells (H9c2). The cells were analyzed for viability, markers of hypertrophy, apoptosis, and the expression of various proteins related to apoptosis. Ang II (100 nM for 48 h)-exposed H9c2 cells treated with BDE (75 μg/ml) showed a significant reduction in apoptosis (58.60%↓) compared to Ang II-alone treated cells. BDE treatment significantly reduced the up-regulation of Bax and cytosolic cytochrome-C caused by Ang II as well as reduced the degree of Ang II- induced down-regulation of Bcl-2. A reduction in caspase-3 activity (33.77%↓) and down-regulation of TNF-α was also observed in BDE treated cells stimulated with Ang II. Furthermore, the up-regulation of phospho-p38 MAPK was attenuated by BDE treatment. Bioactive components in the extract were identified as boeravinone B, quercetin, kaempferol, and caffeic acid as evident from high-performance liquid chromatography (HPLC). Overall, our study shows that B. diffusa is effective in attenuating apoptosis in cardiac cells, which is a major contributor to sudden cardiac death in addition to its nutraceutical properties.
Collapse
Affiliation(s)
- Prathapan A
- Biochemistry & Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, Kerala, India
| | - Salin Raj P
- Biochemistry & Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, Kerala, India
| | - Priya Rani M
- Biochemistry & Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, Kerala, India
| | - Raghu K G
- Biochemistry & Molecular Mechanism Laboratory, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, Kerala, India.
| |
Collapse
|
59
|
Liu S, Wu N, Miao J, Huang Z, Li X, Jia P, Guo Y, Jia D. Protective effect of morin on myocardial ischemia‑reperfusion injury in rats. Int J Mol Med 2018; 42:1379-1390. [PMID: 29956744 PMCID: PMC6089753 DOI: 10.3892/ijmm.2018.3743] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022] Open
Abstract
Morin, a natural flavonol, exhibits antioxidative, anti-inflammatory and anti-apoptotic effects in various pathological and physiological processes. However, whether morin exerts a protective effect on myocardial ischemia-reperfusion injury (MIRI) is unknown. The present study aimed to determine the effect of morin on MIRI in cultured cardiomyocytes and isolated rat hearts, and to additionally explore the underlying mechanism. The effect of morin on the viability, lactate dehydrogenase (LDH) activity and apoptosis of H9c2 cardiomyocytes subjected to hypoxia/reoxygenation, and cardiac function and infarct size of rat hearts following ischemia/reperfusion in an animal model were measured. Furthermore, the mitochondrial permeability transition pore (MPTP) opening, mitochondrial membrane potential (ΔΨm), and the change in the expression levels of B-cell lymphoma 2 (Bcl2)-associated X protein (Bax), Bcl-2 and mitochondrial apoptosis-associated proteins following MPTP opening were also detected. The results indicated that morin treatment significantly increased cell viability, decreased LDH activity and cell apoptosis, improved the recovery of cardiac function and decreased the myocardial infarct size. Furthermore, morin treatment markedly inhibited MPTP opening, prevented the decrease of ΔΨm, and decreased the expression of cytochrome c, apoptotic protease activating factor-1, caspase-9, caspase-3 and the Bax/Bcl-2 ratio. However, these beneficial effects were reversed by treatment with atractyloside, an MPTP opener. The present study demonstrated that morin may prevent MIRI by inhibiting MPTP opening and revealed the possible mechanism of the cardioprotection of morin and its acting target. It also provided an important theoretical basis for the research on drug interventions for MIRI in clinical applications.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Nan Wu
- The Central Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jiaxin Miao
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zijun Huang
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xuying Li
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Pengyu Jia
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuxuan Guo
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Dalin Jia
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
60
|
Liu GS, Gardner G, Adly G, Jiang M, Cai WF, Lam CK, Alogaili F, Robbins N, Rubinstein J, Kranias EG. A novel human S10F-Hsp20 mutation induces lethal peripartum cardiomyopathy. J Cell Mol Med 2018; 22:3911-3919. [PMID: 29761889 PMCID: PMC6050507 DOI: 10.1111/jcmm.13665] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/30/2018] [Indexed: 01/20/2023] Open
Abstract
Heat shock protein 20 (Hsp20) has been shown to be a critical regulator of cardiomyocyte survival upon cardiac stress. In this study, we investigated the functional significance of a novel human Hsp20 mutation (S10F) in peripartum cardiomyopathy. Previous findings showed that cardiac-specific overexpression of this mutant were associated with reduced autophagy, left ventricular dysfunction and early death in male mice. However, this study indicates that females have normal function with no alterations in autophagy but died within a week after 1-4 pregnancies. Further examination of mutant females revealed left ventricular chamber dilation and hypertrophic remodelling. Echocardiography demonstrated increases in left ventricular end-systolic volume and left ventricular end-diastolic volume, while ejection fraction and fractional shortening were depressed following pregnancy. Subsequent studies revealed that cardiomyocyte apoptosis was elevated in mutant female hearts after the third delivery, associated with decreases in the levels of Bcl-2/Bax and Akt phosphorylation. These results indicate that the human S10F mutant is associated with dysregulation of cell survival signalling, accelerated heart failure and early death post-partum.
Collapse
Affiliation(s)
- Guan-Sheng Liu
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - George Gardner
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - George Adly
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Min Jiang
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wen-Feng Cai
- Department of Pathology & Lab Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Chi Keung Lam
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fawzi Alogaili
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nathan Robbins
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jack Rubinstein
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Evangelia G Kranias
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Molecular Biology Division, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
61
|
Gu M, Wang J, Wang Y, Xu Y, Zhang Y, Wu W, Liao S. MiR-147b inhibits cell viability and promotes apoptosis of rat H9c2 cardiomyocytes via down-regulating KLF13 expression. Acta Biochim Biophys Sin (Shanghai) 2018; 50:288-297. [PMID: 29377979 DOI: 10.1093/abbs/gmx144] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Indexed: 11/12/2022] Open
Abstract
Recently, microRNAs (miRNAs) have been shown to involve in the process of heart failure. This study aims to investigate the functional role of miR-147b in rat H9c2 cardiomyocytes and explore the underlying molecular mechanisms. Cell viability of H9c2 cells was detected by MTT assay. Cell apoptosis was detected by flow cytometry. Expression of miR-147b and KLF13 mRNA was detected by quantitative real-time PCR. The relationship between miR-147b and KLF13 was verified by dual-luciferase reporter assay. Protein levels were detected by western blot analysis. It was found that H2O2 inhibited cell viability and promoted cell apoptosis of H9c2 cells in a concentration-dependent manner. MiR-147b overexpression suppressed cell viability and increased apoptosis in H9c2 cells, while knock-down of miR-147b increased cell viability and reduced apoptosis in H2O2-treated H9c2 cells. Luciferase reporter assay and in vitro functional assay showed that KLF13 was a downstream target of miR-147b, and KLF13 knock-down suppressed cell viability and induced apoptosis in H9c2 cells. Enforced expression of KLF13 restored the effects of miR-147b overexpression on cell viability and apoptosis in H9c2 cells. MiR-147b modulated the expression levels of apoptosis-related proteins, and the effects of miR-147b overexpression on apoptosis-related proteins levels were prevented by enforced expression of KLF13 in H9c2 cells. The in vivo experiments showed that miR-147b was up-regulated, and KLF13 was down-regulated in the myocardial tissues from rats with chronic heart failure. Collectively, miR-147b inhibits viability and promotes cell apoptosis by targeting KLF13 in H9c2 cells, which may be associated with the pathogenesis of heart failure.
Collapse
Affiliation(s)
- Mingxia Gu
- Department of Cardiology, Nanjing Central Hospital, Nanjing 210018, China
| | - Jing Wang
- Department of Cardiology, Nanjing Central Hospital, Nanjing 210018, China
| | - Yi Wang
- Department of Cardiology, Nanjing Central Hospital, Nanjing 210018, China
| | - Yanjuan Xu
- Department of Cardiology, Nanjing Central Hospital, Nanjing 210018, China
| | - Yingqiang Zhang
- Department of Cardiology, Nanjing Central Hospital, Nanjing 210018, China
| | - Weiqing Wu
- Department of Physical Examination, The Second Clinical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Shuping Liao
- Department of Physical Examination, The Second Clinical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China
| |
Collapse
|
62
|
Li Y, Lou C, Wang W. STIM1 deficiency protects the liver from ischemia/reperfusion injury in mice. Biochem Biophys Res Commun 2018; 496:422-428. [PMID: 29305862 DOI: 10.1016/j.bbrc.2018.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023]
Abstract
Hepatic ischemia reperfusion (I/R) injury is unavoidable in various clinical conditions. Despite considerable investigation, the underlying molecular mechanism revealing liver I/R injury remains elusive. Stromal interaction molecule 1 (STIM1) plays essential role in regulating the induction of cellular responses to a number of stress conditions, including temperature changes, elevated ROS, and hypoxia. Here, to explore if STIM1 is involved in hepatic injury, wild type (WT) and STIM1-knockout (STIM1-/-) mice were subjected to I/R. Our results indicated that the WT mice with hepatic I/R injury showed higher STIM1 expressions from gene and protein levels in liver tissue samples. Similar results were observed in hypoxia-exposed cells in vitro. Significantly, STIM1-/- attenuated hepatic injury compared to the WT mice after I/R, as evidenced by the improved pathological alterations in liver sections. WT mice subjected to liver I/R showed higher serum alanine aminotransferase (ALT) and aminotransferase (AST) levels, as well as pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-1β, which were significantly reduced by STIM1-/-. In addition, STIM1-/- also decreased the liver mRNA levels of pro-inflammatory cytokines in mice after I/R injury. Furthermore, significantly decreased oxidative stress was found in STIM1-/- mice after I/R injury compared to the WT group of mice, evidenced by the enhanced superoxide dismutase (SOD) activity and the reduced malondialdehyde (MDA) and reactive oxygen species (ROS) levels in liver tissue samples. Moreover, STIM1-/- mice with hepatic I/R injury displayed the down-regulated nuclear factor of activated T cell (NFAT1), Orai1 and cleaved Caspase-3 levels in liver, contributing to apoptosis suppression. The results above were confirmed in hypoxia-treated cells lacking of STIM1 expression. Together, the findings suggested that STIM1-deletion protects the liver from I/R injury in mice through inhibiting inflammation, oxidative stress and apoptosis. STIM1 could be considered as a potential therapeutic target to ameliorate I/R injury.
Collapse
Affiliation(s)
- Yanyang Li
- Department of Pediatrics, Huaihe Hospital, Henan University, Kaifeng 475000, China.
| | - Chunyan Lou
- Department of Pediatrics, Huaihe Hospital, Henan University, Kaifeng 475000, China
| | - Weiying Wang
- Department of Pediatrics, Huaihe Hospital, Henan University, Kaifeng 475000, China
| |
Collapse
|
63
|
Zhang W, Shao M, He X, Wang B, Li Y, Guo X. Overexpression of microRNA-146 protects against oxygen-glucose deprivation/recovery-induced cardiomyocyte apoptosis by inhibiting the NF-κB/TNF-α signaling pathway. Mol Med Rep 2017; 17:1913-1918. [PMID: 29257202 DOI: 10.3892/mmr.2017.8073] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 08/04/2017] [Indexed: 11/06/2022] Open
Abstract
MicroRNA (miR) has been reported to be associated with ischemia and reperfusion (I/R) and cell apoptosis. Suppression of cell apoptosis may reduce the irreversible damage induced by reperfusion. The aims of the current study were to explore the cytoprotective effects of miR-146 against oxygen-glucose deprivation/recovery (OGD/R)-induced injury in H9c2 rat myocardial cells, as well as the underlying mechanisms. Following stimulation with OGD/R, the cells were transfected with miR-146 mimics or negative controls. The levels of miR-146 were analyzed by reverse transcription-quantitative polymerase chain reaction. Thereafter, cell viability and cell apoptosis were analyzed by MTT assay and terminal deoxynucleotidyl-transferase-mediated dUTP nick-end labeling assay, respectively. In addition, the levels of tumor necrosis factor (TNF)-α were determined by ELISA and the levels of B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax), Bcl-2 and phosphorylated (p)-nuclear factor (NF)-κB were measured by western blotting. The results demonstrated that overexpression of miR-146 significantly increased cell viability and decreased apoptosis (P<0.05). It was observed that overexpression of miR-146 statistically reduced the levels of Bax, TNF-α and p-NF-κB but markedly upregulated the levels of Bcl-2 (P<0.05). These results indicate that overexpression of miR-146 may protect against OGD/R-induced cardiomyocyte apoptosis. Overexpression of miR-146 may alleviate the irreversible injury associated with reperfusion and the effects may be achieved by inhibiting the NF-κB/TNF-α signaling pathway.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Mengmeng Shao
- Department of Rehabilitation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaojie He
- Department of Ophthalmology, The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Benji Wang
- Department of Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuechun Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xianyang Guo
- Department of Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
64
|
Conversion of Bim-BH3 from Activator to Inhibitor of Bak through Structure-Based Design. Mol Cell 2017; 68:659-672.e9. [DOI: 10.1016/j.molcel.2017.11.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/15/2017] [Accepted: 10/31/2017] [Indexed: 12/26/2022]
|
65
|
Hu X, Zhang K, Chen Z, Jiang H, Xu W. The HMGB1‑IL‑17A axis contributes to hypoxia/reoxygenation injury via regulation of cardiomyocyte apoptosis and autophagy. Mol Med Rep 2017; 17:336-341. [PMID: 29115425 DOI: 10.3892/mmr.2017.7839] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 03/17/2017] [Indexed: 11/06/2022] Open
Abstract
Both the high‑mobility group box 1 protein (HMGB1) and interleukin (IL)‑17A serve important roles in myocardial ischemia and reperfusion injury. The purpose of the present study was to evaluate whether HMGB1 could induce IL‑17A secretion and lead to cardiomyocyte hypoxia/reoxygenation (H/R) injury. Neonatal rat cardiomyocytes were treated with HMGB1‑neutralizing antibody, IL‑17A‑neutralizing antibody, recombinant HMGB1 (rHMGB1) and recombinant IL‑17A (rIL‑17A), respectively. Cell viabilities, lactate dehydrogenase and creatine kinase levels were measured. Apoptotic cells were assessed by flow cytometry. The expression of HMGB1, IL‑17A, microtubule‑associated proteins 1A/1B light chain 3B (LC3), Beclin‑1, B‑cell lymphoma (Bcl)‑2 and Bcl‑2‑associated X protein were assessed by western blot analysis. The results demonstrated that HMGB1 significantly increased the expression of IL‑17A. HMGB1 or IL‑17A antibody significantly ameliorated H/R‑induced cell injury and improved the cell viability. In contrast, rHMGB1 or rIL‑17A aggravated cell injury and inhibited the cell viability. Furthermore, cardiomyocytes were treated with HMGB1 or IL‑17A antibody significantly increased Bcl‑2 protein expression and had fewer apoptotic cells, whereas rHMGB1 or rIL‑17A‑treated cardiomyocytes markedly decreased Bcl‑2 protein expression and had more apoptotic cells. Moreover, HMGB1 or IL‑17A antibodies significantly inhibited H/R induced autophagy dysfunction (as determined by the inhibition of Beclin‑1 expression, a lower ratio of LC3‑II to LC3‑I), whereas rHMGB1 or rIL‑17A may promote cardiomyocyte autophagy. Together, these results suggested that the HMGB1‑IL‑17A axis contributes to H/R injury via regulation of cardiomyocyte apoptosis and autophagy.
Collapse
Affiliation(s)
- Xiaorong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Kai Zhang
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, Hubei 435000, P.R. China
| | - Zhiqiang Chen
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, Hubei 435000, P.R. China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Weipan Xu
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, Hubei 435000, P.R. China
| |
Collapse
|
66
|
Li X, Geng J, Chen Y, Chen F, Liu C, Xu Q, Zhao J, Hu J, Xie J, Xu B. Exposure to particulate matter induces cardiomyocytes apoptosis after myocardial infarction through NFκB activation. Biochem Biophys Res Commun 2017; 488:224-231. [DOI: 10.1016/j.bbrc.2017.05.047] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 05/08/2017] [Indexed: 02/02/2023]
|
67
|
Jing R, Zhou Z, Kuang F, Huang L, Li C. microRNA-99a Reduces Lipopolysaccharide-Induced Oxidative Injury by Activating Notch Pathway in H9c2 Cells. Int Heart J 2017; 58:422-427. [PMID: 28484120 DOI: 10.1536/ihj.16-261] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
microRNA-99a (miR-99a) is recently recognized as a key regulator in various cancers and cardiovascular diseases. In the present study, we sought to investigate the effects of miR-99a in rat cardiomyocyte H9c2 cells against oxidative injury induced by lipopolysaccharide (LPS).MTT assay, reactive oxygen species (ROS) assay, flow cytometry and lactate dehydrogenase (LDH) assay were respectively used to explore viability, ROS levels, apoptosis, and cell death in H9c2 cells. Quantitative PCR (qRT-PCR) was performed to confirm the expression of miR-99a. Western blot was performed to determine the expression of Notch pathway factors.LPS could significantly suppress viability and increase cell death, apoptosis, and ROS level (P < 0.05). However, miR-99a could significantly increase the viability and decrease apoptosis and ROS level of H9c2 cells (P < 0.05). Overexpression of miR-99a could activate a Notch pathway and regulate the expression of B-cell CLL/lymphoma 2 (BCL2) and cleaved caspase 3.Our study found that overexpression of miR-99a could attenuate LPS-induced oxidative injury in H9c2 cells, possibly via a Notch pathway. These findings suggest that miR-99a may be a key factor in cardiomyocyte oxidative injury and could be a new therapeutic strategy for cardiovascular diseases.
Collapse
Affiliation(s)
- Ran Jing
- Department of Cardiovascular, Xiangya Hospital Central South University
| | - Zhengming Zhou
- Department of Radiology, Xiangya Hospital Central South University
| | - Feng Kuang
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University
| | - Lei Huang
- Department of Cardiac Surgery, Peking University Shenzhen Hospital
| | - Chuanchang Li
- Department of Geriatrics, Xiangya Hospital Central South University
| |
Collapse
|
68
|
Abstract
Apoptosis plays an important role in the myocardial loss after acute myocardial infarction and participates in the process of subsequent left ventricular remodeling and development of symptomatic heart failure. Finding a sensitive apoptotic marker that would help in prognostic stratification of patients after acute myocardial infarction and offer new therapeutic strategies is thus of a great importance. Several studies suggest that tumor necrosis factor-related apoptosis inducing ligand (TRAIL) represents a very promising marker of prognosis in patients with acute myocardial infarction. This review article provides an overview of current knowledge on the role of apoptosis in ischemic heart disease and highlights potentially beneficial apoptotic markers in clinical practice.
Collapse
Affiliation(s)
- Elena Teringova
- Cardiocenter, Department of Cardiology, 3rd Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Srobarova 50, 100 34, Prague 10, Czech Republic
| | - Petr Tousek
- Cardiocenter, Department of Cardiology, 3rd Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Srobarova 50, 100 34, Prague 10, Czech Republic.
| |
Collapse
|
69
|
Yang J, Han J, Li Y, Dong B. Esculetin inhibits the apoptosis in H9c2 cardiomyocytes via the MAPK signaling pathway following hypoxia/reoxygenation injury. Pharmacotherapy 2017. [DOI: 10.1016/j.biopha.2017.01.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
70
|
AKT2 Blocks Nucleus Translocation of Apoptosis-Inducing Factor (AIF) and Endonuclease G (EndoG) While Promoting Caspase Activation during Cardiac Ischemia. Int J Mol Sci 2017; 18:ijms18030565. [PMID: 28272306 PMCID: PMC5372581 DOI: 10.3390/ijms18030565] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 02/13/2017] [Accepted: 02/23/2017] [Indexed: 02/04/2023] Open
Abstract
The AKT (protein kinase B, PKB) family has been shown to participate in diverse cellular processes, including apoptosis. Previous studies demonstrated that protein kinase B2 (AKT2−/−) mice heart was sensitized to apoptosis in response to ischemic injury. However, little is known about the mechanism and apoptotic signaling pathway. Here, we show that AKT2 inhibition does not affect the development of cardiomyocytes but increases cell death during cardiomyocyte ischemia. Caspase-dependent apoptosis of both the extrinsic and intrinsic pathway was inactivated in cardiomyocytes with AKT2 inhibition during ischemia, while significant mitochondrial disruption was observed as well as intracytosolic translocation of cytochrome C (Cyto C) together with apoptosis-inducing factor (AIF) and endonuclease G (EndoG), both of which are proven to conduct DNA degradation in a range of cell death stimuli. Therefore, mitochondria-dependent cell death was investigated and the results suggested that AIF and EndoG nucleus translocation causes cardiomyocyte DNA degradation during ischemia when AKT2 is blocked. These data are the first to show a previous unrecognized function and mechanism of AKT2 in regulating cardiomyocyte survival during ischemia by inducing a unique mitochondrial-dependent DNA degradation pathway when it is inhibited.
Collapse
|
71
|
|
72
|
Liu N, Shi YF, Diao HY, Li YX, Cui Y, Song XJ, Tian X, Li TY, Liu B. MicroRNA-135a Regulates Apoptosis Induced by Hydrogen Peroxide in Rat Cardiomyoblast Cells. Int J Biol Sci 2017; 13:13-21. [PMID: 28123342 PMCID: PMC5264257 DOI: 10.7150/ijbs.16769] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress and apoptosis are the most important pathologic features of ischemic heart disease. Recent research has indicated that microRNAs (miRs) play an essential role in apoptosis. However, whether miRs might regulate B cell lymphoma-2 (Bcl-2) protein in apoptosis during ischemic heart disease is still unclear. The aim of this study, therefore, was to confirm the regulation of microRNA-135a (miR-135a) in oxidative stress injuries induced by hydrogen peroxide (H2O2) in rat cardiomyoblast cells H9c2. To this end, we analyzed the effects of H2O2 treatment on miR-135a expression in rat cardiomyocytes. Furthermore, we upregulated and inhibited miR-135a using mimics and inhibitors, respectively, and examined the effects on cell viability and apoptosis-related proteins. We observed that miR-135a was markedly up-regulated under H2O2 treatment in rat cardiomyoblast cells. Overexpression of miR-135a blocked the Bcl-2 protein and enhanced the apoptosis induced by H2O2, and miR-135a inhibition restored Bcl-2 protein expression. Interestingly, miR-135a inhibition did not attenuate H2O2-induced apoptosis with Bcl-2 knockdown. The results of the present study indicate that miR-135a regulates H2O2-induced apoptosis in H9c2 cells via targeting Bcl-2, and that miR-135a may be a novel therapeutic target for ischemic heart disease.
Collapse
Affiliation(s)
- Ning Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Yong-Feng Shi
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Hong-Ying Diao
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Yang-Xue Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Yan Cui
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Xian-Jing Song
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Xin Tian
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Tian-Yi Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| |
Collapse
|
73
|
Protective Effects of Tirofiban on Myocardial Ischemia–Reperfusion Injury in Rabbits. Am J Ther 2016; 23:e1427-e1435. [DOI: 10.1097/mjt.0000000000000280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
74
|
Schubert C, Raparelli V, Westphal C, Dworatzek E, Petrov G, Kararigas G, Regitz-Zagrosek V. Reduction of apoptosis and preservation of mitochondrial integrity under ischemia/reperfusion injury is mediated by estrogen receptor β. Biol Sex Differ 2016; 7:53. [PMID: 27688871 PMCID: PMC5035458 DOI: 10.1186/s13293-016-0104-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/09/2016] [Indexed: 12/30/2022] Open
Abstract
Background Estrogen improves cardiac recovery after ischemia/reperfusion (I/R) by yet incompletely understood mechanisms. Mitochondria play a crucial role in I/R injury through cytochrome c-dependent apoptosis activation. We tested the hypothesis that 17β-estradiol (E2) as well as a specific ERβ agonist improve cardiac recovery through estrogen receptor (ER)β-mediated mechanisms by reducing mitochondria-induced apoptosis and preserving mitochondrial integrity. Methods We randomized ovariectomized C57BL/6N mice 24h before I/R to pre-treatment with E2 or a specific ERβ agonist (ERβA). Isolated hearts were perfused for 20min prior to 30min global ischemia followed by 40min reperfusion. Results Compared with controls, ERβA and E2 treated groups showed a significant improvement in cardiac recovery, i.e. an increase in left ventricular developed pressure, dP/dtmax and dP/dtmin. ERβA and E2 pre-treatment led to a significant reduction in apoptosis with decreased cytochrome c release from the mitochondria and increased mitochondrial levels of anti-apoptotic Bcl2 and ACAA2. Protein levels of mitochondrial translocase inner membrane (TIM23) and mitochondrial complex I of respiratory chain were increased by ERβA and E2 pre-treatment. Furthermore, we found a significant increase of myosin light chain 2 (MLC2) phosphorylation together with ERK1/2 activation in E2, but not in ERβA treated groups. Conclusions Activation of ERβ is essential for the improvement of cardiac recovery after I/R through the inhibition of apoptosis and preservation of mitochondrial integrity and can be a achieved by a specific ERβ agonist. Furthermore, E2 modulates MLC2 activation after I/R independent of ERβ. Electronic supplementary material The online version of this article (doi:10.1186/s13293-016-0104-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carola Schubert
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charité-Universitaetsmedizin, Hessische Str. 3-4, 10115 Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Valeria Raparelli
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charité-Universitaetsmedizin, Hessische Str. 3-4, 10115 Berlin, Germany.,Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Elke Dworatzek
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charité-Universitaetsmedizin, Hessische Str. 3-4, 10115 Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - George Petrov
- Klinik für Kardiovaskuläre Chirurgie, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Georgios Kararigas
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charité-Universitaetsmedizin, Hessische Str. 3-4, 10115 Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Vera Regitz-Zagrosek
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charité-Universitaetsmedizin, Hessische Str. 3-4, 10115 Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| |
Collapse
|
75
|
Ramnath D, Powell EE, Scholz GM, Sweet MJ. The toll-like receptor 3 pathway in homeostasis, responses to injury and wound repair. Semin Cell Dev Biol 2016; 61:22-30. [PMID: 27552920 DOI: 10.1016/j.semcdb.2016.08.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 12/11/2022]
Abstract
In addition to their established roles in host defence, Toll-like Receptors (TLRs) have emerging roles in control of homeostasis, injury and wound repair. The dsRNA-sensing receptor, TLR3, has been particularly implicated in such processes in several different tissues including the skin, intestine and liver, as well as in the control of reparative mechanisms in the brain, heart and kidneys, following ischemia reperfusion injury. In this review, we provide an overview of TLR3 signalling and functions in inflammation, tissue damage and repair processes, as well as therapeutic opportunities that may arise in the future from knowledge of such pathways.
Collapse
Affiliation(s)
- Divya Ramnath
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Qld 4072, Australia; IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Qld 4072, Australia; Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Elizabeth E Powell
- Centre for Liver Disease Research, School of Medicine, The University of Queensland, Translational Research Institute, Brisbane, Qld 4102, Australia; Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Qld 4102, Australia
| | - Glen M Scholz
- Melbourne Dental School and Oral Health Cooperative Research Centre, The University of Melbourne, Parkville 3010, Australia; Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville 3010, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Qld 4072, Australia; IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Qld 4072, Australia; Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, Qld 4072, Australia.
| |
Collapse
|
76
|
Eupatilin inhibits the apoptosis in H9c2 cardiomyocytes via the Akt/GSK-3β pathway following hypoxia/reoxygenation injury. Biomed Pharmacother 2016; 82:373-8. [DOI: 10.1016/j.biopha.2016.05.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 05/17/2016] [Accepted: 05/17/2016] [Indexed: 11/20/2022] Open
|
77
|
Murphy E, Ardehali H, Balaban RS, DiLisa F, Dorn GW, Kitsis RN, Otsu K, Ping P, Rizzuto R, Sack MN, Wallace D, Youle RJ. Mitochondrial Function, Biology, and Role in Disease: A Scientific Statement From the American Heart Association. Circ Res 2016; 118:1960-91. [PMID: 27126807 PMCID: PMC6398603 DOI: 10.1161/res.0000000000000104] [Citation(s) in RCA: 303] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cardiovascular disease is a major leading cause of morbidity and mortality in the United States and elsewhere. Alterations in mitochondrial function are increasingly being recognized as a contributing factor in myocardial infarction and in patients presenting with cardiomyopathy. Recent understanding of the complex interaction of the mitochondria in regulating metabolism and cell death can provide novel insight and therapeutic targets. The purpose of this statement is to better define the potential role of mitochondria in the genesis of cardiovascular disease such as ischemia and heart failure. To accomplish this, we will define the key mitochondrial processes that play a role in cardiovascular disease that are potential targets for novel therapeutic interventions. This is an exciting time in mitochondrial research. The past decade has provided novel insight into the role of mitochondria function and their importance in complex diseases. This statement will define the key roles that mitochondria play in cardiovascular physiology and disease and provide insight into how mitochondrial defects can contribute to cardiovascular disease; it will also discuss potential biomarkers of mitochondrial disease and suggest potential novel therapeutic approaches.
Collapse
|
78
|
Li X, Zeng Z, Li Q, Xu Q, Xie J, Hao H, Luo G, Liao W, Bin J, Huang X, Liao Y. Inhibition of microRNA-497 ameliorates anoxia/reoxygenation injury in cardiomyocytes by suppressing cell apoptosis and enhancing autophagy. Oncotarget 2016; 6:18829-44. [PMID: 26299920 PMCID: PMC4643066 DOI: 10.18632/oncotarget.4774] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/20/2015] [Indexed: 12/21/2022] Open
Abstract
MiR-497 is predicted to target anti-apoptosis gene Bcl2 and autophagy gene microtubule-associated protein 1 light chain 3 B (LC3B), but the functional consequence of miR-497 in response to anoxia/reoxygenation (AR) or ischemia/reperfusion (IR) remains unknown. This study was designed to investigate the influences of miR-497 on myocardial AR or IR injury. We noted that miR-497 was enriched in cardiac tissues, while its expression was dynamically changed in murine hearts subjected to myocardial infarction and in neonatal rat cardiomyocytes (NRCs) subjected to AR. Forced expression of miR-497 (miR-497 mimic) induced apoptosis in NRCs as determined by Hoechst staining and TUNEL assay. In response to AR, silencing of miR-497 using a miR-497 sponge significantly reduced cell apoptosis and enhanced autophagic flux. Furthermore, the infarct size induced by IR in adenovirus (Ad)-miR-497 sponge infected mice was significantly smaller than in mice receiving Ad-vector or vehicle treatment, while Ad-miR-497 increased infarct size. The expression of Bcl-2 and LC3B-II in NRCs or in murine heart was significantly decreased by miR-497 mimic and enhanced by miR-497 sponge. These findings demonstrate that inhibition of miR-497 holds promise for limiting myocardial IR injury.
Collapse
Affiliation(s)
- Xixian Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhi Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qingman Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qiulin Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiahe Xie
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Huixin Hao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guangjin Luo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jianping Bin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaobo Huang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yulin Liao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
79
|
Luz A, Santos M, Magalhães R, Oliveira JC, Pacheco A, Silveira J, Cabral S, Torres S, Leite-Moreira AF, Carvalho H. Soluble TNF-related apoptosis induced ligand (sTRAIL) is augmented by Post-Conditioning and correlates to infarct size and left ventricle dysfunction in STEMI patients: a substudy from a randomized clinical trial. Heart Vessels 2016; 32:117-125. [DOI: 10.1007/s00380-016-0851-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/20/2016] [Indexed: 01/15/2023]
|
80
|
Li L, Li M, Li Y, Sun W, Wang Y, Bai S, Li H, Wu B, Yang G, Wang R, Wu L, Li H, Xu C. Exogenous H2S contributes to recovery of ischemic post-conditioning-induced cardioprotection by decrease of ROS level via down-regulation of NF-κB and JAK2-STAT3 pathways in the aging cardiomyocytes. Cell Biosci 2016; 6:26. [PMID: 27096074 PMCID: PMC4836181 DOI: 10.1186/s13578-016-0090-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/04/2016] [Indexed: 11/10/2022] Open
Abstract
Background Hydrogen sulfide (H2S), a third member of gasotransmitter family along with nitric oxide and carbon monoxide, generated from mainly catalyzed by cystathionine-lyase, possesses important functions in the cardiovascular system. Ischemic post-conditioning (PC) strongly protects against the hypoxia/reoxygenation (H/R)-induced injury and apoptosis of cardiomyocytes. However, PC protection is ineffective in the aging cardiomyocytes. Whether H2S restores PC-induced cardioprotection by decrease of reactive oxygen species (ROS) level in the aging cardiomyocytes is unknown. Methods The aging cardiomyocytes were induced by treatment of primary cultures of neonatal cardiomyocytes using d-galactose and were exposed to H/R and PC protocols. Cell viability was observed by CCK-8 kit. Apoptosis was detected by Hoechst 33342 staining and flow cytometry. ROS level was analyzed using spectrofluorimeter. Related protein expressions were detected through Western blot. Results Treatment of NaHS (a H2S donor) protected against H/R-induced apoptosis, cell damage, the expression of cleaved caspase-3 and cleaved caspase-9, the release of cytochrome c (Cyt c). The supplementation of NaHS also decreased the activity of LDH and CK, MDA contents, ROS levels and the phosphorylation of IκBα, NF-κB, JNK2 and STAT3, and increased cell viability, the expression of Bcl-2, the activity of SOD, CAT and GSH-PX. PC alone did not provide cardioprotection in H/R-treated aging cardiomyocytes, which was significantly restored by the addition of NaHS. The beneficial role of NaHS was similar to the supply of N-acetyl-cysteine (NAC, an inhibitor of ROS), Ammonium pyrrolidinedithiocarbamate (PDTC, an inhibitor of NF-κB) and AG 490 (an inhibitor of JNK2), respectively, during PC. Conclusion Our results suggest that exogenous H2S contributes to recovery of PC-induced cardioprotection by decrease of ROS level via down-regulation of NF-κB and JAK2/STAT3 pathways in the aging cardiomyocytes.
Collapse
Affiliation(s)
- Lina Li
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China.,The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Meixiu Li
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China.,The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Youyou Li
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China.,The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Weiming Sun
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China.,The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Yuehong Wang
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China.,The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Shuzhi Bai
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China.,The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Hongxia Li
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China.,The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Bo Wu
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China.,The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Guangdong Yang
- The Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON Canada
| | - Rui Wang
- The Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON Canada
| | - Lingyun Wu
- The Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON Canada
| | - Hongzhu Li
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China.,The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| | - Changing Xu
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China.,The Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, China
| |
Collapse
|
81
|
Jayachandran KS, Vasanthi AHR, Gurusamy N. Steroidal Saponin Diosgenin from Dioscorea bulbifera Protects Cardiac Cells from Hypoxia-reoxygenation Injury Through Modulation of Pro-survival and Pro-death Molecules. Pharmacogn Mag 2016; 12:S14-20. [PMID: 27041852 PMCID: PMC4791993 DOI: 10.4103/0973-1296.176114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Diosgenin, a steroidal saponin from plants, exhibits many biological potentials. Herein, the cardioprotective role of diosgenin is studied. Materials and Methods: The effect of diosgenin, isolated from Dioscorea bulbifera, was studied on hypoxia-reoxygenation (HR) in H9c2 cardiomyoblast cells. The amount of diosgenin in the plant extract was analyzed by high-performance thin layer chromatography using a solvent system comprising of chloroform:methanol:acetic acid:formic acid (13:4.5:1.5:1). Cardioprotection was checked by (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Further, the release of lactate dehydrogenase, an enzyme released during cell death was checked. The proteins responsible for cell death (Bax) and cell survival (Bcl-2, hemeoxygenase-1 and Akt) were analyzed using Western blot to check the cardioprotective role of diosgenin. Conclusion: Supplementation of diosgenin mitigates HR injury, thereby exhibiting cardioprotective potential. SUMMARY The cardioprotective effect of Diosgenin was evidenced from the improved cell survival after hypoxia-reoxygenation injury demonstrated through MTT cell survival assay. The release of lactate dehydrogenase, an enzyme released during cell death was decreased by Diosgenin. Diosgenin upregulated the pro-survival molecules like B-cell lymphoma 2 (Bcl-2), heme oxygenase-1 and the phosphorylation of ATK (at serine 473); and at the same time pro-.death molecules like Bax was downregulated. Thus, Diosgenin as a plant based steroidal saponin is confirmed to mitigate ischemic reperfusion injury.
Collapse
Affiliation(s)
| | | | - Narasimman Gurusamy
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Kingdom of Saudi Arabia
| |
Collapse
|
82
|
Kong F, Wang H, Guo J, Peng M, Ji H, Yang H, Liu B, Wang J, Zhang X, Li S. Hsp70 suppresses apoptosis of BRL cells by regulating the expression of Bcl-2, cytochrome C, and caspase 8/3. In Vitro Cell Dev Biol Anim 2016; 52:568-75. [DOI: 10.1007/s11626-016-0005-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/20/2016] [Indexed: 01/07/2023]
|
83
|
Cho GW, Altamirano F, Hill JA. Chronic heart failure: Ca(2+), catabolism, and catastrophic cell death. Biochim Biophys Acta Mol Basis Dis 2016; 1862:763-777. [PMID: 26775029 DOI: 10.1016/j.bbadis.2016.01.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 12/28/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022]
Abstract
Robust successes have been achieved in recent years in conquering the acutely lethal manifestations of heart disease. Many patients who previously would have died now survive to enjoy happy and productive lives. Nevertheless, the devastating impact of heart disease continues unabated, as the spectrum of disease has evolved with new manifestations. In light of this ever-evolving challenge, insights that culminate in novel therapeutic targets are urgently needed. Here, we review fundamental mechanisms of heart failure, both with reduced (HFrEF) and preserved (HFpEF) ejection fraction. We discuss pathways that regulate cardiomyocyte remodeling and turnover, focusing on Ca(2+) signaling, autophagy, and apoptosis. In particular, we highlight recent insights pointing to novel connections among these events. We also explore mechanisms whereby potential therapeutic approaches targeting these processes may improve morbidity and mortality in the devastating syndrome of heart failure.
Collapse
Affiliation(s)
- Geoffrey W Cho
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Francisco Altamirano
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
84
|
Shi M, Liu B, Liu G, Wang P, Yang M, Li Y, Zhou J. Low intensity-pulsed ultrasound induced apoptosis of human hepatocellular carcinoma cells in vitro. ULTRASONICS 2016; 64:43-53. [PMID: 26231998 DOI: 10.1016/j.ultras.2015.07.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/18/2015] [Accepted: 07/21/2015] [Indexed: 05/15/2023]
Abstract
The present study was conducted to determine whether low intensity-pulsed ultrasound (LIPUS) could induce apoptosis of human hepatocellular carcinoma cells, SMMC-7721, and to define the mechanism of ultrasound-induced apoptosis, in vitro. MTT assay was used to measure cell proliferation. Apoptosis was investigated by multiple methods such as flow cytometry, DNA fragmentation, Ca(2+) mobilizations, pro- and anti-apoptotic protein expression, and light as well as ultramicroscopic morphology. The results provide evidence that LIPUS induced a dose-dependent effect on cell viability and apoptosis of SMMC-7721 cells. Specifically, exposure of cells to >0.5 W/cm(2) intensity significantly increased cell apoptosis, caused shifts in cell cycle phase, and induced structural changes. Ultrasound significantly increased intracellular Ca(2+) concentrations and modulated expression of caspase-3, Bcl-2 and Bax. The findings suggest that this novel technology can be used to induce SMMC-7721 apoptosis via the Ca(2+)/mitochondrial pathway and could potentially be of clinical use for the treatment of hepatocellular carcinoma (SMMC-7721 cell line) and other cancers.
Collapse
Affiliation(s)
- Mingfang Shi
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bangzhong Liu
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Guanghua Liu
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ping Wang
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mingzhen Yang
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yun Li
- Department of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
85
|
Badalzadeh R, Mokhtari B, Yavari R. Contribution of apoptosis in myocardial reperfusion injury and loss of cardioprotection in diabetes mellitus. J Physiol Sci 2015; 65:201-15. [PMID: 25726180 PMCID: PMC10717803 DOI: 10.1007/s12576-015-0365-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 02/11/2015] [Indexed: 12/21/2022]
Abstract
Ischemic heart disease is one of the major causes of death worldwide. Ischemia is a condition in which blood flow of the myocardium declines, leading to cardiomyocyte death. However, reperfusion of ischemic regions decreases the rate of mortality, but it can also cause later complications. In a clinical setting, ischemic heart disease is always coincident with other co-morbidities such as diabetes. The risk of heart disease increases 2-3 times in diabetic patients. Apoptosis is considered to be one of the main pathophysiological mechanisms of myocardial ischemia-reperfusion injury. Diabetes can disrupt the anti-apoptotic intracellular signaling cascades involved in myocardial protection. Therefore, targeting these changes may be an effective cardioprotective approach in the diabetic myocardium against ischemia-reperfusion injury. In this article, we review the interaction of diabetes with the pathophysiology of myocardial ischemia-reperfusion injury, focusing on the contribution of apoptosis in this context, and then discuss the alterations of pro-apoptotic or anti-apoptotic pathways probably responsible for the loss of cardioprotection in diabetes.
Collapse
Affiliation(s)
- Reza Badalzadeh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Mokhtari
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raana Yavari
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
86
|
Wang H, Li J, Chi H, Zhang F, Zhu X, Cai J, Yang X. MicroRNA-181c targets Bcl-2 and regulates mitochondrial morphology in myocardial cells. J Cell Mol Med 2015; 19:2084-97. [PMID: 25898913 PMCID: PMC4568913 DOI: 10.1111/jcmm.12563] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/20/2015] [Indexed: 12/21/2022] Open
Abstract
Apoptosis is an important mechanism for the development of heart failure. Mitochondria are central to the execution of apoptosis in the intrinsic pathway. The main regulator of mitochondrial pathway of apoptosis is Bcl-2 family which includes pro- and anti-apoptotic proteins. MicroRNAs are small noncoding RNA molecules that regulate gene expression by inhibiting mRNA translation and/or inducing mRNA degradation. It has been proposed that microRNAs play critical roles in the cardiovascular physiology and pathogenesis of cardiovascular diseases. Our previous study has found that microRNA-181c, a miRNA expressed in the myocardial cells, plays an important role in the development of heart failure. With bioinformatics analysis, we predicted that miR-181c could target the 3' untranslated region of Bcl-2, one of the anti-apoptotic members of the Bcl-2 family. Thus, we have suggested that miR-181c was involved in regulation of Bcl-2. In this study, we investigated this hypothesis using the Dual-Luciferase Reporter Assay System. Cultured myocardial cells were transfected with the mimic or inhibitor of miR-181c. We found that the level of miR-181c was inversely correlated with the Bcl-2 protein level and that transfection of myocardial cells with the mimic or inhibitor of miR-181c resulted in significant changes in the levels of caspases, Bcl-2 and cytochrome C in these cells. The increased level of Bcl-2 caused by the decrease in miR-181c protected mitochondrial morphology from the tumour necrosis factor alpha-induced apoptosis.
Collapse
Affiliation(s)
- Hongjiang Wang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jing Li
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hongjie Chi
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Fan Zhang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiaoming Zhu
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jun Cai
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xinchun Yang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
87
|
Li H, Wang Y, Wei C, Bai S, Zhao Y, Li H, Wu B, Wang R, Wu L, Xu C. Mediation of exogenous hydrogen sulfide in recovery of ischemic post-conditioning-induced cardioprotection via down-regulating oxidative stress and up-regulating PI3K/Akt/GSK-3β pathway in isolated aging rat hearts. Cell Biosci 2015; 5:11. [PMID: 25789157 PMCID: PMC4364662 DOI: 10.1186/s13578-015-0003-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/02/2015] [Indexed: 11/16/2022] Open
Abstract
The physiological and pathological roles of hydrogen sulfide (H2S) in the regulation of cardiovascular functions have been recognized. Cystathionine gamma-lyase (CSE) is a major H2S-producing enzyme in cardiovascular system. Ischemic post-conditioning (PC) provides cadioprotection in young hearts but lost in the aging hearts. The involvement of H2S in the recovery of PC-induced cardioprotection in the aging hearts is unclear. In the present study, we demonstrated that ischemia/reperfusion (I/R) decreased H2S production rate and CSE expression, aggravated cardiomyocytes damage, apoptosis and myocardial infarct size, reduced cardiac function, increased the levels of Bcl-2, caspase-3 and caspase-9 mRNA, enhanced oxidative stress in isolated young and aging rat hearts. I/R also increased the release of cytochrome c and down-regulated the phosphorylation of PI3K, Akt and GSK-3β in the aging rat hearts. We further found that PC increased H2S production rate and CSE expressions, and protected young hearts from I/R-induced cardiomyocytes damage, all of which were disappeared in the aging hearts. Supply of NaHS not only increased PC-induced cardioprotection in the young hearts, but also lightened I/R induced-myocardial damage and significantly recovered the cardioprotective role of PC against I/R induced myocardial damage in the aging hearts. LY294002 (a PI3K inhibitor) abolished but N-acetyl-cysteine (NAC, an inhibitor of reactive oxygen species, ROS) further enhanced the protective role of H2S against I/R induced myocardial damage in the aging hearts. In conclusion, these results demonstrate that exogenous H2S recovers PC-induced cardioprotection via inhibition of oxidative stress and up-regulation of PI3K-Akt-GSK-3β pathway in the aging rat hearts. These findings suggested that H2S might be a novel target for the treatment of aging cardiovascular diseases.
Collapse
Affiliation(s)
- Hongzhu Li
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China
| | - Yuehong Wang
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China
| | - Can Wei
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China
| | - Shuzhi Bai
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China
| | - Yajun Zhao
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China
| | - Hongxia Li
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China
| | - Bo Wu
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China
| | - Rui Wang
- Department of Biology, Lakehead University, Thunder Bay, ON P7B5E1 Canada
| | - Lingyun Wu
- Department of Health Science, Lakehead University, Thunder Bay, ON P7B5E1 Canada
| | - Changqing Xu
- Department of Pathophysiology, Harbin Medical University, Baojian Road, Harbin, 150081 China
| |
Collapse
|
88
|
Chen Q, Xu H, Xu A, Ross T, Bowler E, Hu Y, Lesnefsky EJ. Inhibition of Bcl-2 sensitizes mitochondrial permeability transition pore (MPTP) opening in ischemia-damaged mitochondria. PLoS One 2015; 10:e0118834. [PMID: 25756500 PMCID: PMC4354902 DOI: 10.1371/journal.pone.0118834] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 01/15/2015] [Indexed: 11/21/2022] Open
Abstract
Background Mitochondria are critical to cardiac injury during reperfusion as a result of damage sustained during ischemia, including the loss of bcl-2. We asked if bcl-2 depletion not only leads to selective permeation of the outer mitochondrial membrane (MOMP) favoring cytochrome c release and programmed cell death, but also favors opening of the mitochondrial permeability transition pore (MPTP). An increase in MPTP susceptibility would support a role for bcl-2 depletion mediated cell death in the calcium overload setting of early reperfusion via MPTP as well as later in reperfusion via MOMP as myocardial calcium content normalizes. Methods Calcium retention capacity (CRC) was used to reflect the sensitivity of the MPTP opening in isolated cardiac mitochondria. To study the relationship between bcl-2 inhibition and MPTP opening, mitochondria were incubated with a bcl-2 inhibitor (HA14-1) and CRC measured. The contribution of preserved bcl-2 content to MPTP opening following ischemia-reperfusion was explored using transgenic bcl-2 overexpressed mice. Results CRC was decreased in mitochondria following reperfusion compared to ischemia alone, indicating that reperfusion further sensitizes to MPTP opening. Incubation of ischemia-damaged mitochondria with increasing HA14-1concentrations increased calcium-stimulated MPTP opening, supporting that functional inhibition of bcl-2 during simulated reperfusion favors MPTP opening. Moreover, HA14-1 sensitivity was increased by ischemia compared to non-ischemic controls. Overexpression of bcl-2 attenuated MPTP opening in following ischemia-reperfusion. HA14-1 inhibition also increased the permeability of the outer membrane in the absence of exogenous calcium, indicating that bcl-2 inhibition favors MOMP when calcium is low. Conclusions The depletion and functional inhibition of bcl-2 contributes to cardiac injury by increasing susceptibility to MPTP opening in high calcium environments and MOMP in the absence of calcium overload. Thus, ischemia-damaged mitochondria with decreased bcl-2 content are susceptible to MPTP opening in early reperfusion and MOMP later in reperfusion when cytosolic calcium has normalized.
Collapse
Affiliation(s)
- Qun Chen
- Department of Medicine, Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Haishan Xu
- Department of Medicine, Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Aijun Xu
- Department of Medicine, Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Thomas Ross
- Department of Medicine, Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Elizabeth Bowler
- Department of Medicine, Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- University of the West of England, Bristol, United Kingdom
| | - Ying Hu
- Department of Medicine, Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Edward J. Lesnefsky
- Department of Medicine, Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Medicine, Pauley Heart Center, Division of Biochemistry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- McGuire Department of Veterans Affairs Medical Center, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
89
|
Euler G. Good and bad sides of TGFβ-signaling in myocardial infarction. Front Physiol 2015; 6:66. [PMID: 25788886 PMCID: PMC4349055 DOI: 10.3389/fphys.2015.00066] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/07/2015] [Indexed: 12/21/2022] Open
Abstract
Myocardial infarction is a prevailing cause of death in industrial countries. In spite of the good opportunities we have nowadays in interventional cardiology to reopen the clotted coronary arteries for reperfusion of ischemic areas, post-infarct remodeling emerges and contributes to unfavorable structural conversion processes in the myocardium, finally resulting in heart failure. The growth factor TGFβ is upregulated during these processes. In this review, an overview on the functional role of TGFβ signaling in the process of cardiac remodeling is given, as it can influence apoptosis, fibrosis and hypertrophy thereby predominantly aggravating ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Gerhild Euler
- Institute of Physiology, Justus-Liebig-University Giessen, Germany
| |
Collapse
|
90
|
|
91
|
Zirpoli H, Abdillahi M, Quadri N, Ananthakrishnan R, Wang L, Rosario R, Zhu Z, Deckelbaum RJ, Ramasamy R. Acute administration of n-3 rich triglyceride emulsions provides cardioprotection in murine models after ischemia-reperfusion. PLoS One 2015; 10:e0116274. [PMID: 25559887 PMCID: PMC4283969 DOI: 10.1371/journal.pone.0116274] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 12/08/2014] [Indexed: 11/19/2022] Open
Abstract
Dietary n-3 fatty acids (FAs) may reduce cardiovascular disease risk. We questioned whether acute administration of n-3 rich triglyceride (TG) emulsions could preserve cardiac function and decrease injury after ischemia/reperfusion (I/R) insult. We used two different experimental models: in vivo, C57BL/6 mice were exposed to acute occlusion of the left anterior descending coronary artery (LAD), and ex-vivo, C57BL/6 murine hearts were perfused using Langendorff technique (LT). In the LAD model, mice treated with n-3 TG emulsion (1.5g/kg body weight), immediately after ischemia and 1h later during reperfusion, significantly reduced infarct size and maintained cardiac function (p<0.05). In the LT model, administration of n-3 TG emulsion (300mgTG/100ml) during reperfusion significantly improved functional recovery (p<0.05). In both models, lactate dehydrogenase (LDH) levels, as a marker of injury, were significantly reduced by n-3 TG emulsion. To investigate the mechanisms by which n-3 FAs protects hearts from I/R injury, we investigated changes in key pathways linked to cardioprotection. In the ex-vivo model, we showed that n-3 FAs increased phosphorylation of AKT and GSK3β proteins (p<0.05). Acute n-3 TG emulsion treatment also increased Bcl-2 protein level and reduced an autophagy marker, Beclin-1 (p<0.05). Additionally, cardioprotection by n-3 TG emulsion was linked to changes in PPARγ protein expression (p<0.05). Rosiglitazone and p-AKT inhibitor counteracted the positive effect of n-3 TG; GSK3β inhibitor plus n-3 TG significantly inhibited LDH release. We conclude that acute n-3 TG injection during reperfusion provides cardioprotection. This may prove to be a novel acute adjunctive reperfusion therapy after treating patients with myocardial infarction.
Collapse
Affiliation(s)
- Hylde Zirpoli
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Mariane Abdillahi
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Nosirudeen Quadri
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Radha Ananthakrishnan
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Lingjie Wang
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Rosa Rosario
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Zhengbin Zhu
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
| | - Richard J. Deckelbaum
- Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- * E-mail: (R. Ramasamy); (RJD)
| | - Ravichandran Ramasamy
- Department of Medicine, New York University School of Medicine, New York, New York, United States of America
- * E-mail: (R. Ramasamy); (RJD)
| |
Collapse
|
92
|
Wang Z, Wang Y, Ye J, Lu X, Cheng Y, Xiang L, Chen L, Feng W, Shi H, Yu X, Lin L, Zhang H, Xiao J, Li X. bFGF attenuates endoplasmic reticulum stress and mitochondrial injury on myocardial ischaemia/reperfusion via activation of PI3K/Akt/ERK1/2 pathway. J Cell Mol Med 2014; 19:595-607. [PMID: 25533999 PMCID: PMC4369816 DOI: 10.1111/jcmm.12346] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 05/14/2014] [Indexed: 12/11/2022] Open
Abstract
Extensive research focused on finding effective strategies to prevent or improve recovery from myocardial ischaemia/reperfusion (I/R) injury. Basic fibroblast growth factor (bFGF) has been shown to have therapeutic potential in some heart disorders, including ischaemic injury. In this study, we demonstrate that bFGF administration can inhibit the endoplasmic reticulum (ER) stress and mitochondrial dysfunction induced in the heart in a mouse model of I/R injury. In vitro, bFGF exerts a protective effect by inhibiting the ER stress response and mitochondrial dysfunction proteins that are induced by tert-Butyl hydroperoxide (TBHP) treatment. Both of these in vivo and in vitro effects are related to the activation of two downstream signalling pathways, PI3K/Akt and ERK1/2. Inhibition of these PI3K/Akt and ERK1/2 pathways by specific inhibitors, LY294002 and PD98059, partially reduces the protective effect of bFGF. Taken together, our results indicate that the cardioprotective role of bFGF involves the suppression of ER stress and mitochondrial dysfunction in ischaemic oxidative damage models and oxidative stress-induced H9C2 cell injury; furthermore, these effects underlie the activation of the PI3K/Akt and ERK1/2 signalling pathways.
Collapse
Affiliation(s)
- Zhouguang Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Jilin University, Changchun, China; School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Shin SY, Kim T, Lee HS, Kang JH, Lee JY, Cho KH, Kim DH. The switching role of β-adrenergic receptor signalling in cell survival or death decision of cardiomyocytes. Nat Commun 2014; 5:5777. [PMID: 25517116 PMCID: PMC4284638 DOI: 10.1038/ncomms6777] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 11/06/2014] [Indexed: 01/21/2023] Open
Abstract
How cell fate (survival or death) is determined and whether such determination depends on the strength of stimulation has remained unclear. In this study, we discover that the cell fate of cardiomyocytes switches from survival to death with the increase of β-adrenergic receptor (β-AR) stimulation. Mathematical simulations combined with biochemical experimentation of β-AR signalling pathways show that the gradual increment of isoproterenol (a non-selective β1/β2-AR agonist) induces the switching response of Bcl-2 expression from the initial increase followed by a decrease below its basal level. The ERK1/2 and ICER-mediated feed-forward loop is the hidden design principle underlying such cell fate switching characteristics. Moreover, we find that β1-blocker treatment increases the survival effect of β-AR stimuli through the regulation of Bcl-2 expression leading to the resistance to cell death, providing new insight into the mechanism of therapeutic effects. Our systems analysis further suggests a novel potential therapeutic strategy for heart disease. The contribution of signal strength on cell fate decisions is often not reflected in signalling networks. By combining mathematical simulation and biochemical experiments in cultured adult cardiomyocytes, Shin et al. show that the concentration of a β-adrenergic receptor agonist affects the expression of Bcl-2, influencing the balance between cell survival and death.
Collapse
Affiliation(s)
- Sung-Young Shin
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Taeyong Kim
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, Korea
| | - Ho-Sung Lee
- 1] Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea [2] Graduate School of Medical Science and Engineering, KAIST, Daejeon 305-701, Korea
| | - Jun Hyuk Kang
- 1] Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea [2] Graduate School of Medical Science and Engineering, KAIST, Daejeon 305-701, Korea
| | - Ji Young Lee
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, Korea
| | - Kwang-Hyun Cho
- 1] Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea [2] Graduate School of Medical Science and Engineering, KAIST, Daejeon 305-701, Korea
| | - Do Han Kim
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, Korea
| |
Collapse
|
94
|
GUO XINHONG, CAO WENJIANG, YAO JIAMING, YUAN YONG, HONG YE, WANG XINCHUN, XING JIANGUO. Cardioprotective effects of tilianin in rat myocardial ischemia-reperfusion injury. Mol Med Rep 2014; 11:2227-33. [DOI: 10.3892/mmr.2014.2954] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 07/04/2014] [Indexed: 11/05/2022] Open
|
95
|
Senyo SE, Lee RT, Kühn B. Cardiac regeneration based on mechanisms of cardiomyocyte proliferation and differentiation. Stem Cell Res 2014; 13:532-41. [PMID: 25306390 PMCID: PMC4435693 DOI: 10.1016/j.scr.2014.09.003] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 09/10/2014] [Accepted: 09/16/2014] [Indexed: 12/23/2022] Open
Abstract
Cardiomyocyte proliferation and progenitor differentiation are endogenous mechanisms of myocardial development. Cardiomyocytes continue to proliferate in mammals for part of post-natal development. In adult mammals under homeostatic conditions, cardiomyocytes proliferate at an extremely low rate. Because the mechanisms of cardiomyocyte generation provide potential targets for stimulating myocardial regeneration, a deep understanding is required for developing such strategies. We will discuss approaches for examining cardiomyocyte regeneration, review the specific advantages, challenges, and controversies, and recommend approaches for interpretation of results. We will also draw parallels between developmental and regenerative principles of these mechanisms and how they could be targeted for treating heart failure.
Collapse
Affiliation(s)
- Samuel E Senyo
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Brigham Regenerative Medicine Center, Cambridge, MA 02139, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Richard T Lee
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Brigham Regenerative Medicine Center, Cambridge, MA 02139, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Bernhard Kühn
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
96
|
Parvin AA, Pranap RA, Shalini U, Devendran A, Baker JE, Dhanasekaran A. Erythropoietin protects cardiomyocytes from cell death during hypoxia/reperfusion injury through activation of survival signaling pathways. PLoS One 2014; 9:e107453. [PMID: 25237819 PMCID: PMC4169563 DOI: 10.1371/journal.pone.0107453] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 05/29/2014] [Indexed: 12/17/2022] Open
Abstract
Hypoxia/Reoxygenation (H/R) cardiac injury is of great importance in understanding Myocardial Infarctions, which affect a major part of the working population causing debilitating side effects and often-premature mortality. H/R injury primarily consists of apoptotic and necrotic death of cardiomyocytes due to a compromise in the integrity of the mitochondrial membrane. Major factors associated in the deregulation of the membrane include fluctuating reactive oxygen species (ROS), deregulation of mitochondrial permeability transport pore (MPTP), uncontrolled calcium (Ca2+) fluxes, and abnormal caspase-3 activity. Erythropoietin (EPO) is strongly inferred to be cardioprotective and acts by inhibiting the above-mentioned processes. Surprisingly, the underlying mechanism of EPO's action and H/R injury is yet to be fully investigated and elucidated. This study examined whether EPO maintains Ca2+ homeostasis and the mitochondrial membrane potential (ΔΨm) in cardiomyocytes when subjected to H/R injury and further explored the underlying mechanisms involved. H9C2 cells were exposed to different concentrations of EPO post-H/R, and 20 U/ml EPO was found to significantly increase cell viability by inhibiting the intracellular production of ROS and caspase-3 activity. The protective effect of EPO was abolished when H/R-induced H9C2 cells were treated with Wortmannin, an inhibitor of Akt, suggesting the mechanism of action through the activation Akt, a major survival pathway.
Collapse
Affiliation(s)
- Asiya A Parvin
- Centre for Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | - Raj A Pranap
- Centre for Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | - U Shalini
- Centre for Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | - Ajay Devendran
- Centre for Biotechnology, Anna University, Chennai, Tamil Nadu, India
| | - John E Baker
- Department of Surgery, Division of Cardiothoracic Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | | |
Collapse
|
97
|
Agarwal B, Stowe DF, Dash RK, Bosnjak ZJ, Camara AKS. Mitochondrial targets for volatile anesthetics against cardiac ischemia-reperfusion injury. Front Physiol 2014; 5:341. [PMID: 25278902 PMCID: PMC4165278 DOI: 10.3389/fphys.2014.00341] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/20/2014] [Indexed: 12/15/2022] Open
Abstract
Mitochondria are critical modulators of cell function and are increasingly recognized as proximal sensors and effectors that ultimately determine the balance between cell survival and cell death. Volatile anesthetics (VA) are long known for their cardioprotective effects, as demonstrated by improved mitochondrial and cellular functions, and by reduced necrotic and apoptotic cell death during cardiac ischemia and reperfusion (IR) injury. The molecular mechanisms by which VA impart cardioprotection are still poorly understood. Because of the emerging role of mitochondria as therapeutic targets in diseases, including ischemic heart disease, it is important to know if VA-induced cytoprotective mechanisms are mediated at the mitochondrial level. In recent years, considerable evidence points to direct effects of VA on mitochondrial channel/transporter protein functions and electron transport chain (ETC) complexes as potential targets in mediating cardioprotection. This review furnishes an integrated overview of targets that VA impart on mitochondrial channels/transporters and ETC proteins that could provide a basis for cation regulation and homeostasis, mitochondrial bioenergetics, and reactive oxygen species (ROS) emission in redox signaling for cardiac cell protection during IR injury.
Collapse
Affiliation(s)
- Bhawana Agarwal
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - David F. Stowe
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
- Department of Physiology, Medical College of WisconsinMilwaukee, WI, USA
- Cardiovascular Research Center, Medical College of WisconsinMilwaukee, WI, USA
- Zablocki VA Medical CenterMilwaukee, WI, USA
- Department of Biomedical Engineering, Marquette UniversityMilwaukee, WI, USA
| | - Ranjan K. Dash
- Department of Physiology, Medical College of WisconsinMilwaukee, WI, USA
- Department of Biomedical Engineering, Marquette UniversityMilwaukee, WI, USA
- Biotechnology and Bioengineering Center, Medical College of WisconsinMilwaukee, WI, USA
| | - Zeljko J. Bosnjak
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
- Department of Physiology, Medical College of WisconsinMilwaukee, WI, USA
- Cardiovascular Research Center, Medical College of WisconsinMilwaukee, WI, USA
| | - Amadou K. S. Camara
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
- Cardiovascular Research Center, Medical College of WisconsinMilwaukee, WI, USA
| |
Collapse
|
98
|
Yan P, Chen SQ, Li ZP, Zhang J, Xue JK, Wang WT, Huang WJ, Cheng JY, Li HP. Effect of exogenous phosphocreatine on cardiomycytic apoptosis and expression of Bcl-2 and Bax after cardiopulmonary resuscitation in rats. World J Emerg Med 2014; 2:291-5. [PMID: 25215026 DOI: 10.5847/wjem.j.1920-8642.2011.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 10/10/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Ischemia-reperfusion injury in the myocardium after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) is an important pathologic basis of post-cardiac arrest of syndrome (PCAS), and apoptosis is one of the major mechanisms in myocardial ischemia-reperfusion injury. To lessen myocardial ischemia-reperfusion injury after cardiac arrest and CPR, it is important to reduce energy consumption and to increase energy supply in the myocardium. This study aimed to observe changes of cell apoptosis and expression of Bcl-2 and Bax protein on the myocardium after CPR in rats, and the protective effects of different doses of exogenous phosphocreatine (creatine phosphate, CP) on them. METHODS A total of 32 male adult Sprague-Dawley rats were randomly divided into 4 groups: control group (group A), CPR group (group B), low-dose CP group (group C, CP 0.5 g/kg at beginning of CPR and 1.0 g/kg at 2 hours after CPR) and high-dose CP group (group D, CP 1.0 g/kg at beginning of CPR and 2.0 g/kg at 2 hours after CPR). Cardiac arrest was induced by asphyxiation and CPR started at 7 minutes after asphyxiation in groups B, C and D. Myocardium samples were taken at 24 hours after CPR. Cardiomycytic apoptosis was detected by the TdT-mediated dUTP-biotin nick end labeling (TUNEL) method. The expression of Bcl-2 and Bax protein was measured by immunohistochemistry. RESULTS Cardiomyocytic apoptosis index (AI) and expression of Bcl-2 and Bax protein increased more significantly in groups B, C and D than in group A (P<0.01), but Bcl-2/Bax ratio significantly decreased (P<0.01). Cardiomyocytic AI and expression of Bcl-2 and Bax protein decreased more significantly in groups C and D than in group B (P<0.01), but Bcl-2/Bax ratio increased more significantly (P<0.01). Cardiomyocytic AI and expression of Bcl-2 and Bax protein decreased more significantly in group D than in group C (P<0.05), but Bcl-2/Bax ratio increased more significantly (P<0.05). CONCLUSION Exogenous phosphocreatine, especially at a large dose, could inhibit cardiomyocytic apoptosis and alleviate myocardial injury after CPR in rats.
Collapse
Affiliation(s)
- Ping Yan
- Department of Emergency Medicine, First and Second Affiliated Hospitals of Wenzhou Medical College, Wenzhou 325000, China
| | - Shou-Quan Chen
- Department of Emergency Medicine, First and Second Affiliated Hospitals of Wenzhou Medical College, Wenzhou 325000, China
| | - Zhang-Ping Li
- Department of Emergency Medicine, First and Second Affiliated Hospitals of Wenzhou Medical College, Wenzhou 325000, China
| | - Jie Zhang
- Department of Emergency Medicine, First and Second Affiliated Hospitals of Wenzhou Medical College, Wenzhou 325000, China
| | - Ji-Ke Xue
- Department of Emergency Medicine, First and Second Affiliated Hospitals of Wenzhou Medical College, Wenzhou 325000, China
| | - Wan-Tie Wang
- Department of Emergency Medicine, First and Second Affiliated Hospitals of Wenzhou Medical College, Wenzhou 325000, China
| | - Wei-Jia Huang
- Department of Emergency Medicine, First and Second Affiliated Hospitals of Wenzhou Medical College, Wenzhou 325000, China
| | - Jun-Yan Cheng
- Department of Emergency Medicine, First and Second Affiliated Hospitals of Wenzhou Medical College, Wenzhou 325000, China
| | - Hui-Ping Li
- Department of Emergency Medicine, First and Second Affiliated Hospitals of Wenzhou Medical College, Wenzhou 325000, China
| |
Collapse
|
99
|
Zang X, Zheng F, Hong HJ, Jiang Y, Song Y, Xia Y. Neutrophil gelatinase-associated lipocalin protects renal tubular epithelial cells in hypoxia–reperfusion by reducing apoptosis. Int Urol Nephrol 2014; 46:1673-9. [DOI: 10.1007/s11255-014-0749-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 05/16/2014] [Indexed: 10/25/2022]
|
100
|
Abstract
Accumulation of protein aggregates is a hallmark of several neurodegenerative disorders as well as for a number of protein conformation-based diseases, including those affecting muscle, liver and heart. Desminopathy or desmin-related myopathy (DRM) is a skeletal myopathy characterized by bilateral muscle weakness, but is often accompanied by cardiomyopathy as well. DRM can be caused by mutations in desmin, alphaB crystallin, myotilin, Z-band alternatively spliced PDZ-containing protein (ZASP), filamin C (FLNC) or Bcl-2-associated athanogene-3 (BAG3). The common pathological pattern in DRM is accumulation of misfolded proteins, however, clinical manifestations can differ significantly.
Collapse
|