101
|
Gao K, Li Y, Su Y, Lin Z, Yang X, Xu M, Huang Y, Chen S, Xie Y, Li Z. High uric acid promotes mitophagy through the ROS/CaMKIIδ/Parkin pathway in cardiomyocytes in vitro and in vivo. Am J Transl Res 2021; 13:8754-8765. [PMID: 34539992 PMCID: PMC8430116 DOI: pmid/34539992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 06/13/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Increasing evidence has suggested that high uric acid (HUA) is closely related to cardiovascular disease (CVD). Mitophagy abnormalities have been reported to participate in multiple pathogenic processes of CVD. However, the potential molecular mechanisms remain unclear. Herein, we investigated the effect of HUA-induced mitophagy and its potential molecular mechanism in cardiomyocytes. METHODS We established a model of cardiomyocytes induced by HUA in vitro and in vivo. Mitochondrial membrane potential (MMP), reactive oxygen species (ROS) production and adenosine triphosphate (ATP) content were measured. The mitophagy-related protein expression of LC3B-II, Parkin, Ca2+/calmodulin-dependent protein kinase II δ (CaMKIIδ) and P62 was measured by Western blot. Based on the colocalization of lysosomes and mitochondria, a confocal microscope was used to detect mitophagy. Additionally, we established a mitophagy inhibitor group (3-MA) and CaMKIIδ inhibitor group (KN-93) to verify the pathway. RESULTS In the HUA stimulation model, ROS production was increased, and mitochondrial injury indexes (MMP and ATP contents) were decreased. Moreover, these indicators were reversed by 3-MA and KN-93. Under HUA stimulation, the expression of LC3B-II, Parkin, CaMKIIδ and P62 increased significantly. Furthermore, these protein levels were reduced by 3-MA and KN-93. CONCLUSION HUA can promote cardiomyocyte mitophagy activation through the ROS/CaMKIIδ/parkin pathway axis. This study may provide a new target and theoretical basis for the prevention and treatment of HUA-related metabolic heart disease in the future.
Collapse
Affiliation(s)
- Kai Gao
- Emergency Department, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong Province, China
| | - Yanbing Li
- Department of Cardiology, Beijing Youan Hospital, Capital Medical UniversityBeijing, China
| | - Yiwan Su
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong Province, China
| | - Zhishan Lin
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong Province, China
| | - Xiangbin Yang
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong Province, China
| | - Meiling Xu
- Emergency Department, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong Province, China
| | - Yanting Huang
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong Province, China
| | - Shuqin Chen
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong Province, China
| | - Yang Xie
- Emergency Department, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong Province, China
| | - Zhi Li
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical CollegeShantou, Guangdong Province, China
| |
Collapse
|
102
|
Telomerase therapy attenuates cardiotoxic effects of doxorubicin. Mol Ther 2021; 29:1395-1410. [PMID: 33388418 PMCID: PMC8058493 DOI: 10.1016/j.ymthe.2020.12.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/17/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
Doxorubicin is one of the most potent chemotherapeutic agents. However, its clinical use is restricted due to the severe risk of cardiotoxicity, partially attributed to elevated production of reactive oxygen species (ROS). Telomerase canonically maintains telomeres during cell division but is silenced in adult hearts. In non-dividing cells such as cardiomyocytes, telomerase confers pro-survival traits, likely owing to the detoxification of ROS. Therefore, we hypothesized that pharmacological overexpression of telomerase may be used as a therapeutic strategy for the prevention of doxorubicin-induced cardiotoxicity. We used adeno-associated virus (AAV)-mediated gene therapy for long-term expression of telomerase in in vitro and in vivo models of doxorubicin-induced cardiotoxicity. Overexpression of telomerase protected the heart from doxorubicin-mediated apoptosis and rescued cardiac function, which was accompanied by preserved cardiomyocyte size. At the mechanistic level, we observed altered mitochondrial morphology and dynamics in response to telomerase expression. Complementary in vitro experiments confirmed the anti-apoptotic effects of telomerase overexpression in human induced pluripotent stem cell-derived cardiomyocytes after doxorubicin treatment. Strikingly, elevated levels of telomerase translocated to the mitochondria upon doxorubicin treatment, which helped to maintain mitochondrial function. Thus, telomerase gene therapy could be a novel preventive strategy for cardiotoxicity by chemotherapy agents such as the anthracyclines.
Collapse
|
103
|
Chang X, Zhang W, Zhao Z, Ma C, Zhang T, Meng Q, Yan P, Zhang L, Zhao Y. Regulation of Mitochondrial Quality Control by Natural Drugs in the Treatment of Cardiovascular Diseases: Potential and Advantages. Front Cell Dev Biol 2020; 8:616139. [PMID: 33425924 PMCID: PMC7793684 DOI: 10.3389/fcell.2020.616139] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/18/2020] [Indexed: 12/13/2022] Open
Abstract
Mitochondria are double-membraned cellular organelles that provide the required energy and metabolic intermediates to cardiomyocytes. Mitochondrial respiratory chain defects, structure abnormalities, and DNA mutations can affect the normal function of cardiomyocytes, causing an imbalance in intracellular calcium ion homeostasis, production of reactive oxygen species, and apoptosis. Mitochondrial quality control (MQC) is an important process that maintains mitochondrial homeostasis in cardiomyocytes and involves multi-level regulatory mechanisms, such as mitophagy, mitochondrial fission and fusion, mitochondrial energy metabolism, mitochondrial antioxidant system, and mitochondrial respiratory chain. Furthermore, MQC plays a role in the pathological mechanisms of various cardiovascular diseases (CVDs). In recent years, the regulatory effects of natural plants, drugs, and active ingredients on MQC in the context of CVDs have received significant attention. Effective active ingredients in natural drugs can influence the production of energy-supplying substances in the mitochondria, interfere with the expression of genes associated with mitochondrial energy requirements, and regulate various mechanisms of MQC modulation. Thus, these ingredients have therapeutic effects against CVDs. This review provides useful information about novel treatment options for CVDs and development of novel drugs targeting MQC.
Collapse
Affiliation(s)
- Xing Chang
- China Academy of Chinese Medical Sciences, Beijing, China.,Guang'anmen Hospital of Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Wenjin Zhang
- China Academy of Chinese Medical Sciences, Beijing, China.,College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Zhenyu Zhao
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Chunxia Ma
- Shandong Analysis and Test Center, Qilu University of Technology, Jinan, China
| | - Tian Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingyan Meng
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peizheng Yan
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Zhang
- Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yuping Zhao
- China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
104
|
Kobayashi S, Zhao F, Zhang Z, Kobayashi T, Huang Y, Shi B, Wu W, Liang Q. Mitochondrial Fission and Mitophagy Coordinately Restrict High Glucose Toxicity in Cardiomyocytes. Front Physiol 2020; 11:604069. [PMID: 33362579 PMCID: PMC7758327 DOI: 10.3389/fphys.2020.604069] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/16/2020] [Indexed: 11/15/2022] Open
Abstract
Hyperglycemia-induced mitochondrial dysfunction plays a key role in the pathogenesis of diabetic cardiomyopathy. Injured mitochondrial segments are separated by mitochondrial fission and eliminated by autophagic sequestration and subsequent degradation in the lysosome, a process termed mitophagy. However, it remains poorly understood how high glucose affects the activities of, and the relationship between, mitochondrial fission and mitophagy in cardiomyocytes. In this study, we determined the functional roles of mitochondrial fission and mitophagy in hyperglycemia-induced cardiomyocyte injury. High glucose (30 mM, HG) reduced mitochondrial connectivity and particle size and increased mitochondrial number in neonatal rat ventricular cardiomyocytes, suggesting an enhanced mitochondrial fragmentation. SiRNA knockdown of the pro-fission factor dynamin-related protein 1 (DRP1) restored mitochondrial size but did not affect HG toxicity, and Mdivi-1, a DRP1 inhibitor, even increased HG-induced cardiomyocyte injury, as shown by superoxide production, mitochondrial membrane potential and cell death. However, DRP1 overexpression triggered mitochondrial fragmentation and mitigated HG-induced cardiomyocyte injury, suggesting that the increased mitochondrial fission is beneficial, rather than detrimental, to cardiomyocytes cultured under HG conditions. This is in contrast to the prevailing hypothesis that mitochondrial fragmentation mediates or contributes to HG cardiotoxicity. Meanwhile, HG reduced mitophagy flux as determined by the difference in the levels of mitochondria-associated LC3-II or the numbers of mitophagy foci indicated by the novel dual fluorescent reporter mt-Rosella in the absence and presence of the lysosomal inhibitors. The ability of HG to induce mitochondrial fragmentation and inhibit mitophagy was reproduced in adult mouse cardiomyocytes. Overexpression of Parkin, a positive regulator of mitophagy, or treatment with CCCP, a mitochondrial uncoupler, induced mitophagy and attenuated HG-induced cardiomyocyte death, while Parkin knockdown had opposite effects, suggesting an essential role of mitophagy in cardiomyocyte survival under HG conditions. Strikingly, Parkin overexpression increased mitochondrial fragmentation, while DRP1 overexpression accelerated mitophagy flux, demonstrating a reciprocal activation loop that controls mitochondrial fission and mitophagy. Thus, strategies that promote the mutual positive interaction between mitochondrial fission and mitophagy while simultaneously maintain their levels within the physiological range would be expected to improve mitochondrial health, alleviating hyperglycemic cardiotoxicity.
Collapse
Affiliation(s)
- Satoru Kobayashi
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Fengyi Zhao
- Department of Endocrinology, The First affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ziying Zhang
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tamayo Kobayashi
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Yuan Huang
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Bingyin Shi
- Department of Endocrinology, The First affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weihua Wu
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiangrong Liang
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY, United States
| |
Collapse
|
105
|
Chai Q, Miao J, Liu M, Zhang Z, Meng Z, Wu W. Knockdown of SGLT1 prevents the apoptosis of cardiomyocytes induced by glucose fluctuation via relieving oxidative stress and mitochondrial dysfunction. Biochem Cell Biol 2020; 99:356-363. [PMID: 33259229 DOI: 10.1139/bcb-2020-0491] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fluctuations in the concentration of glucose in the blood is more detrimental than a constantly high level of glucose with respect to the development of cardiovascular complications associated with diabetes mellitus (DM). Sodium glucose cotransporter 2 (SGLT2) inhibitors have been developed as antidiabetic drugs with cardiovascular benefits; however, whether inhibition of SGLT1 protects the diabetic heart remains to be determined. This study investigated the role of SGLT1 in rat H9c2 cardiomyocytes subjected to fluctuating levels of glucose and the underlying mechanisms. The results indicated that knockdown of SGLT1 restored cell proliferation and suppressed the cytotoxicity associated with fluctuating glucose levels. Oxidative stress was induced in H9c2 cells subjected to fluctuating glucose levels, but these changes were effectively reversed by knockdown of SGLT1, as manifested by reductions in the level of intracellular reactive oxygen species and increased antioxidant activity. Further study demonstrated that knockdown of SGLT1 attenuated the mitochondrial dysfunction in H9c2 cells exposed to fluctuating glucose levels, by restoring mitochondrial membrane potential and promoting mitochondrial fusion. In addition, knockdown of SGLT1 downregulated the expression of Bax, upregulated the expression of Bcl-2, and reduced the activation of caspase-3 in H9c2 cells subjected to fluctuating levels of glucose. Collectively, our results show that knockdown of SGLT1 ameliorates the apoptosis of cardiomyocyte caused by fluctuating glucose levels via regulating oxidative stress and combatting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Qian Chai
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Jiajing Miao
- Department of Endocrinology, Affiliated Hospital of Jilin Medical College, Jilin 132013, P.R. China
| | - Meili Liu
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Ziying Zhang
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Ziang Meng
- Department of Urology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Weihua Wu
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| |
Collapse
|
106
|
Ishikawa K, Yamamoto S, Hattori S, Nishimura N, Matsumoto H, Miyakawa T, Nakada K. Neuronal degeneration and cognitive impairment can be prevented via the normalization of mitochondrial dynamics. Pharmacol Res 2020; 163:105246. [PMID: 33086082 DOI: 10.1016/j.phrs.2020.105246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 01/27/2023]
Abstract
Neuronal cells possess a certain degree of plasticity to recover from cell damage. When the stress levels are higher than their plasticity capabilities, neuronal degeneration is triggered. However, the factors correlated to the plasticity capabilities need to be investigated. In this study, we generated a novel mouse model that able to express in an inducible manner a dominant-negative form of MFN2, a mitochondrial fusion factor. We then compared the phenotype of the mice continuously expressing the mutated MFN2 with that of the mice only transiently expressing it. Remarkably, the phenotypes of the group transiently expressing mutant MFN2 could be divided into 3 types: equivalent to what was observed in the continuous expression group, intermediate between the continuous expression group and the control group, and equivalent to the control group. In particular, in the continuous expression group, we observed remarkable hyperactivity and marked cognitive impairments, which were not seen, or were very mild in the transient expression group. These results indicate that abnormal mitochondrial dynamics lead to stress, triggering neuron degeneration; therefore, the neurodegeneration progression can be prevented via the normalization of the mitochondrial dynamics. Since the availability of mouse models suitable for the reproduction of both neurodegeneration and recovery at least partially is very limited, our mouse model can be a useful tool to investigate neuronal plasticity mechanisms and neurodegeneration.
Collapse
Affiliation(s)
- Kaori Ishikawa
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8572, Japan; Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8572, Japan.
| | - Satoshi Yamamoto
- Integrated Technology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd. 26-1, 2-chome, Muraoka-Higashi, Fujisawa, Kanagawa, 251-8555, Japan
| | - Satoko Hattori
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Naoya Nishimura
- Integrated Technology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd. 26-1, 2-chome, Muraoka-Higashi, Fujisawa, Kanagawa, 251-8555, Japan
| | - Hirokazu Matsumoto
- Integrated Technology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd. 26-1, 2-chome, Muraoka-Higashi, Fujisawa, Kanagawa, 251-8555, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, 1-98, Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Kazuto Nakada
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8572, Japan; Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| |
Collapse
|
107
|
Sangweni NF, Moremane M, Riedel S, van Vuuren D, Huisamen B, Mabasa L, Barry R, Johnson R. The Prophylactic Effect of Pinocembrin Against Doxorubicin-Induced Cardiotoxicity in an In Vitro H9c2 Cell Model. Front Pharmacol 2020; 11:1172. [PMID: 32903793 PMCID: PMC7438920 DOI: 10.3389/fphar.2020.01172] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 07/17/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The clinical use of Doxorubicin (Dox) is significantly limited by its dose-dependent cardiotoxic side effect. Accumulative evidence suggests that the use of flavonoids, such as the antioxidative Pinocembrin (Pin), could be effective in the prevention of Dox-induced cardiotoxicity. Accordingly, we investigated the ability of pinocembrin (Pin) to attenuate Dox-induced cardiotoxicity in an in vitro H9c2 cardiomyoblast model. METHODOLOGY The cardioprotective potential of Pin was established in H9c2 cells. Here, cells were treated with Dox (2μM), Dox (2μM) + Pin (1μM), and Dox (2μM) + Dexrazoxane (20μM) for 6 days. Thereafter, the safe co-administration of Pin with Dox, in a cancer environment, was investigated in MCF-7 breast cancer cells subjected to the same experimental conditions. Untreated cells served as the control. Subsequently, Pin's ability to attenuate Dox-mediated oxidative stress, impaired mitochondrial bioenergetics and potential, as well as aggravated apoptosis was quantified using biochemical assays. RESULTS The results demonstrated that co-treatment with Pin mitigates Dox-induced oxidative stress by alleviating the antioxidant enzyme activity of the H9c2 cells. Pin further reduced the rate of apoptosis and necrosis inferred by Dox by improving mitochondrial bioenergetics. Interestingly, Pin did not decrease the efficacy of Dox but, rather increased the rate of apoptosis and necrosis in Dox-treated MCF-7 cells. CONCLUSION The findings presented in this study showed, for the first time, that Pin attenuates Dox-induced cardiotoxicity without reducing its chemotherapeutic effect. We propose that additional studies, using in vivo models, should be conducted to further investigate Pin as a suitable candidate in the prevention of the cardiovascular dysfunction inferred by Dox administration.
Collapse
Affiliation(s)
- Nonhlakanipho F. Sangweni
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Malebogo Moremane
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Sylvia Riedel
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Derick van Vuuren
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Barbara Huisamen
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Lawrence Mabasa
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, South Africa
| | - Reenen Barry
- Research and Development Department, Biopharm, Hamilton, New Zealand
| | - Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
108
|
Zhou H, He L, Xu G, Chen L. Mitophagy in cardiovascular disease. Clin Chim Acta 2020; 507:210-218. [DOI: 10.1016/j.cca.2020.04.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 02/08/2023]
|
109
|
Kalkhoran SB, Hernandez-Resendiz S, Ong SG, Ramachandra CJ, Hausenloy DJ. Mitochondrial shaping proteins as novel treatment targets for cardiomyopathies. CONDITIONING MEDICINE 2020; 3:216-226. [PMID: 33134886 PMCID: PMC7595308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Heart failure (HF) is one of the leading causes of death and disability worldwide. The prevalence of HF continues to rise, and its outcomes are worsened by risk factors such as age, diabetes, obesity, hypertension, and ischemic heart disease. Hence, there is an unmet need to identify novel treatment targets that can prevent the development and progression of HF in order to improve patient outcomes. In this regard, cardiac mitochondria play an essential role in generating the ATP required to maintain normal cardiac contractile function. Mitochondrial dysfunction is known to contribute to the pathogenesis of a number of cardiomyopathies including those secondary to diabetes, pressure-overload left ventricular hypertrophy (LVH), and doxorubicin cardiotoxicity. Mitochondria continually change their shape by undergoing fusion and fission, and an imbalance in mitochondrial fusion and fission have been shown to impact on mitochondrial function, and contribute to the pathogenesis of these cardiomyopathies. In this review article, we focus on the role of mitochondrial shaping proteins as contributors to the development of three cardiomyopathies, and highlight their therapeutic potential as novel treatment targets for preventing the onset and progression of HF.
Collapse
Affiliation(s)
- Siavash Beikoghli Kalkhoran
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, UK
- Cardiovascular and Metabolic Disorder Programme, Duke-NUS Medical School, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore
| | - Sauri Hernandez-Resendiz
- Cardiovascular and Metabolic Disorder Programme, Duke-NUS Medical School, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Nuevo Leon, Mexico
| | - Sang-Ging Ong
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Chrishan J.A. Ramachandra
- Cardiovascular and Metabolic Disorder Programme, Duke-NUS Medical School, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore
| | - Derek J. Hausenloy
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, UK
- Cardiovascular and Metabolic Disorder Programme, Duke-NUS Medical School, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore
- Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan
| |
Collapse
|
110
|
Selective protection of human cardiomyocytes from anthracycline cardiotoxicity by small molecule inhibitors of MAP4K4. Sci Rep 2020; 10:12060. [PMID: 32694738 PMCID: PMC7374628 DOI: 10.1038/s41598-020-68907-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 06/25/2020] [Indexed: 12/11/2022] Open
Abstract
Given the poor track record to date of animal models for creating cardioprotective drugs, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have been proposed as a therapeutically relevant human platform to guide target validation and cardiac drug development. Mitogen-Activated Protein Kinase Kinase Kinase Kinase-4 (MAP4K4) is an “upstream” member of the MAPK superfamily that is implicated in human cardiac muscle cell death from oxidative stress, based on gene silencing and pharmacological inhibition in hPSC-CMs. A further role for MAP4K4 was proposed in heart muscle cell death triggered by cardiotoxic anti-cancer drugs, given its reported activation in failing human hearts with doxorubicin (DOX) cardiomyopathy, and its activation acutely by DOX in cultured cardiomyocytes. Here, we report successful protection from DOX in two independent hPSC-CM lines, using two potent, highly selective MAP4K4 inhibitors. The MAP4K4 inhibitors enhanced viability and reduced apoptosis at otherwise lethal concentrations of DOX, and preserved cardiomyocyte function, as measured by spontaneous calcium transients, at sub-maximal ones. Notably, in contrast, no intereference was seen in tumor cell killing, caspase activation, or mitochondrial membrane dissipation by DOX, in human cancer cell lines. Thus, MAP4K4 is a plausible, tractable, selective therapeutic target in DOX-induced human heart muscle cell death.
Collapse
|
111
|
Li L, Li J, Wang Q, Zhao X, Yang D, Niu L, Yang Y, Zheng X, Hu L, Li Y. Shenmai Injection Protects Against Doxorubicin-Induced Cardiotoxicity via Maintaining Mitochondrial Homeostasis. Front Pharmacol 2020; 11:815. [PMID: 32581790 PMCID: PMC7289952 DOI: 10.3389/fphar.2020.00815] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
Shenmai injection (SMI), as a patented traditional Chinese medicine, is extracted from Panax ginseng and Ophiopogon japonicus. It commonly used in the treatment of cardiovascular disease and in the control of cardiac toxicity induced by doxorubicin (DOX) treatment. However, its anti-cardiotoxicity mechanism remains unknown. The purpose of this study was to investigate the underlying mitochondrial protective mechanisms of SMI on DOX-induced myocardial injury. The cardioprotective effect of SMI against DOX-induced myocardial damage was evaluated in C57BL/6 mice and H9c2 cardiomyocytes. In vivo, myocardial injury, apoptosis and phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB/Akt)/glycogen synthase kinase 3 beta (GSK-3β) signaling pathway related proteins were measured. In vitro, apoptosis, mitochondrial superoxide, mitochondrial membrane potential, mitochondrial morphology, levels of mitochondrial fission/fusion associated proteins, mitochondrial respiratory function, and AMP-activated protein kinase (AMPK) activity were assessed. To further elucidate the regulating effects of SMI on AMPK and PI3K/Akt/GSK-3β signaling pathway, compound C and LY294002 were utilized. In vivo, SMI decreased mortality rate, levels of creatine kinase, and creatine kinase-MB. SMI significantly prevented DOX-induced cardiac dysfunction and apoptosis, decreased levels of Bax/Bcl-2 and cleaved-Caspase3, increased levels of PI3K, p-Akt, and p-GSK-3β. In vitro, SMI rescued DOX-injured H9c2 cardiomyocytes from apoptosis, excessive mitochondrial reactive oxygen species production and descending mitochondrial membrane potential, which were markedly suppressed by LY294002. SMI increased ratio of L-OPA1 to S-OPA1, levels of AMPK phosphorylation, and DRP1 phosphorylation (Ser637) in order to prevent DOX-induced excessive mitochondrial fission and insufficient mitochondrial fusion. In conclusion, SMI prevents DOX-induced cardiotoxicity, inhibits mitochondrial oxidative stress and mitochondrial fragmentation through activation of AMPK and PI3K/Akt/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Lin Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinghao Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qilong Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin Zhao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dongli Yang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lu Niu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanze Yang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xianxian Zheng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Limin Hu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhong Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
112
|
Xiao D, Chang W, Ding W, Wang Y, Fa H, Wang J. Enhanced mitophagy mediated by the YAP/Parkin pathway protects against DOX-induced cardiotoxicity. Toxicol Lett 2020; 330:96-107. [PMID: 32434049 DOI: 10.1016/j.toxlet.2020.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
The clinical usage of Doxorubicin (DOX) is limited due to its cardiotoxicity. Although the precise mechanism remains unclear, there is an increasing body of evidence that has demonstrated that mitophagy is responsible for DOX-induced cardiotoxicity. In the present study, Parkin, a key protein for mitophagy initiation, was revealed to be downregulated in mouse hearts and in H9c2 cells upon DOX treatment. Enforced expression of Parkin led to mitophagy activation and attenuated cell apoptosis in H9c2 cells. Parkin transgenic mice inhibited DOX-induced cardiotoxicity. Furthermore, Yes-associatd protein, as a transcription co-activator, regulated the gene expression of Parkin, and in turn Parkin overexpression protected against cell apoptosis induced by DOX treatment. Taken together, enhanced mitophagy mediated by YAP/Parkin pathway protects against DOX-induced cardiotoxicity in mouse heart. These studies revealed the complex pathological process of DOX-induced cardiotoxicity and provided novel insight into potential chemotherapy targets.
Collapse
Affiliation(s)
- Dandan Xiao
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China; Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China.
| | - Wenguang Chang
- Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China.
| | - Wei Ding
- Affiliated Hospital, Qingdao University, Qingdao, 266003, China.
| | - Yu Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Hongge Fa
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China; Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
113
|
Wang Q, Liu B, Wang Y, Bai B, Yu T, Chu XM. The biomarkers of key miRNAs and target genes associated with acute myocardial infarction. PeerJ 2020; 8:e9129. [PMID: 32440375 PMCID: PMC7229769 DOI: 10.7717/peerj.9129] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Background Acute myocardial infarction (AMI) is considered one of the most prominent causes of death from cardiovascular disease worldwide. Knowledge of the molecular mechanisms underlying AMI remains limited. Accurate biomarkers are needed to predict the risk of AMI and would be beneficial for managing the incidence rate. The gold standard for the diagnosis of AMI, the cardiac troponin T (cTnT) assay, requires serial testing, and the timing of measurement with respect to symptoms affects the results. As attractive candidate diagnostic biomarkers in AMI, circulating microRNAs (miRNAs) are easily detectable, generally stable and tissue specific. Methods The Gene Expression Omnibus (GEO) database was used to compare miRNA expression between AMI and control samples, and the interactions between miRNAs and mRNAs were analysed for expression and function. Furthermore, a protein-protein interaction (PPI) network was constructed. The miRNAs identified in the bioinformatic analysis were verified by RT-qPCR in an H9C2 cell line. The miRNAs in plasma samples from patients with AMI (n = 11) and healthy controls (n = 11) were used to construct receiver operating characteristic (ROC) curves to evaluate the clinical prognostic value of the identified miRNAs. Results We identified eight novel miRNAs as potential candidate diagnostic biomarkers for patients with AMI. In addition, the predicted target genes provide insight into the molecular mechanisms underlying AMI.
Collapse
Affiliation(s)
- Qi Wang
- Department of Cardiology, The Affiliated hospital of Qingdao University, Qingdao, China
| | - Bingyan Liu
- School of Basic Medicine, Qingdao University, Qingdao, China.,Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Yuanyong Wang
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Baochen Bai
- Department of Cardiology, The Affiliated hospital of Qingdao University, Qingdao, China
| | - Tao Yu
- Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Xian-Ming Chu
- Department of Cardiology, The Affiliated hospital of Qingdao University, Qingdao, China.,Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
114
|
Liang X, Wang S, Wang L, Ceylan AF, Ren J, Zhang Y. Mitophagy inhibitor liensinine suppresses doxorubicin-induced cardiotoxicity through inhibition of Drp1-mediated maladaptive mitochondrial fission. Pharmacol Res 2020; 157:104846. [PMID: 32339784 DOI: 10.1016/j.phrs.2020.104846] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/17/2020] [Accepted: 04/16/2020] [Indexed: 01/04/2023]
Abstract
Doxorubicin (DOX) is one of the most effective antineoplastic drugs. However, its clinical application has been greatly limited due to the development of cardiotoxicity with DOX utilization. A number of theories have been postulated for DOX-induced cardiotoxicity with a pivotal contribution from unchecked (excess) mitophagy and mitochondrial fission. Liensinine (LIEN), a newly identified mitophagy inhibitor, strengthens the antineoplastic efficacy of DOX although its action on hearts remains elusive. This study was designed to examine the effect of LIEN on DOX-induced cardiotoxicity and the underlying mechanisms involved with a focus on mitochondrial dynamics. Our data revealed that LIEN alleviated DOX-induced cardiac dysfunction and apoptosis through inhibition of dynamin-related protein 1 (Drp1)-mediated excess (unchecked) mitochondrial fission. LIEN treatment decreased Drp1 phosphorylation at Ser616 site, inhibited mitochondrial fragmentation, mitophagy (assessed by TOM20 and TIM23), oxidative stress, cytochrome C leakage, cardiomyocyte apoptosis, as well as improved mitochondrial function and cardiomyocyte contractile function in DOX-induced cardiac injury. In DOX-challenged neonatal mouse ventricular myocytes (NMVMs), LIEN-suppressed Drp1 phosphorylation, mitochondrial fragmentation, and apoptosis were blunted by Rab7 overexpression, the effect of which was reversed by the ERK inhibitor U0126. Moreover, activation of ERK or Drp1 abolished the protective effects of LIEN on cardiomyocyte mechanical anomalies. These data shed some lights towards understanding the role of LIEN as a new protective agent against DOX-associated cardiotoxicity without compromising its anti-tumor effects.
Collapse
Affiliation(s)
- Xinyue Liang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Shuyi Wang
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA
| | - Lifeng Wang
- Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA; Department of Physiology, Basic Medicine College, Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Asli F Ceylan
- Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA.
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| |
Collapse
|
115
|
The role of Drp1 in mitophagy and cell death in the heart. J Mol Cell Cardiol 2020; 142:138-145. [PMID: 32302592 DOI: 10.1016/j.yjmcc.2020.04.015] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/25/2020] [Accepted: 04/11/2020] [Indexed: 12/20/2022]
Abstract
Maintenance of mitochondrial function and integrity is critical for normal cell survival, particularly in non-dividing cells with a high-energy demand such as cardiomyocytes. Well-coordinated quality control mechanisms in cardiomyocytes, involving mitochondrial biogenesis, mitochondrial dynamics-fission and fusion, and mitophagy, act to protect against mitochondrial dysfunction. Mitochondrial fission, which requires dynamin-related protein 1 (Drp1), is essential for segregation of damaged mitochondria for degradation. Alterations in this process have been linked to cardiomyocyte apoptosis and cardiomyopathy. In this review, we discuss the role of Drp1 in mitophagy and apoptosis in the context of cardiac pathology, including myocardial ischemia and heart failure.
Collapse
|
116
|
Tang Z, Luo C, Jun Y, Yao M, Zhang M, He K, Jin L, Ma J, Chen S, Sun S, Tao M, Ding L, Sun X, Chen X, Zhang L, Gao Y, Wang QL. Nanovector Assembled from Natural Egg Yolk Lipids for Tumor-Targeted Delivery of Therapeutics. ACS APPLIED MATERIALS & INTERFACES 2020; 12:7984-7994. [PMID: 31971362 DOI: 10.1021/acsami.9b22293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Nanomedicine uses nanotechnology-based strategies for precision tumor therapy, including passive and ligand-mediated active tumor targeting by nanocarriers. However, the possible biotoxicity of chemosynthetic nanovectors limits their clinical applications. A novel natural egg yolk lipid nanovector (EYLN) was developed for effective loading and delivery of therapeutic agents. Lipids were extracted from egg yolks and reassembled into nanosized particles. EYLNs' stability, cellular uptake, toxicity, and delivery capacity for therapeutic agents were evaluated in vitro. The systemic toxicity and biodistribution of EYLNs were analyzed in normal mice, and the therapeutic effects of doxorubicin (Dox)-loaded EYLNs were evaluated in mouse breast cancer and hepatoma models. EYLNs had a particle size of ∼40 nm and a surface ζ-potential of -45 mV and were effectively internalized by tumor cells, without showing toxicity and side effects in vitro and in vivo. Importantly, their excellent permeability and retention effect significantly enhanced the distribution of EYLNs at tumor sites, and EYLN-Dox effectively inhibited the tumor growth in both mouse models. Targeted modification with folic acid further promoted vector-mediated drug distribution in tumors. This study demonstrates that lipids with specific proportions in the egg yolk can be used to construct natural drug vectors, providing a new strategy for nano-oncology research.
Collapse
Affiliation(s)
- Zhuang Tang
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Chao Luo
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Yali Jun
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Mengchu Yao
- Department of Clinical Oncology, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Mengyan Zhang
- Department of Clinical Oncology, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Kang He
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Luhao Jin
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Jianshe Ma
- School of Basic Medicine , Wenzhou Medical University , Wenzhou 325035 , China
| | - Song Chen
- Institute of Medicinal Biotechnology , Jiangsu College of Nursing , Huai'an 223300 , China
| | - SuAn Sun
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Mingyue Tao
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Lianshu Ding
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Xiaoyang Sun
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Xiaofei Chen
- Department of Clinical Oncology, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Li Zhang
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Yong Gao
- Department of Clinical Oncology, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| | - Qi-Long Wang
- Department of Central Laboratory, The Affiliated Huaian No.1 People's Hospital , Nanjing Medical University , Huai'an 223300 , China
| |
Collapse
|
117
|
Xu H, Yu W, Sun S, Li C, Zhang Y, Ren J. Luteolin Attenuates Doxorubicin-Induced Cardiotoxicity Through Promoting Mitochondrial Autophagy. Front Physiol 2020; 11:113. [PMID: 32116805 PMCID: PMC7033739 DOI: 10.3389/fphys.2020.00113] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/30/2020] [Indexed: 02/02/2023] Open
Abstract
Doxorubicin is a valuable antineoplastic drug although its clinical use is greatly hindered by its severe cardiotoxicity with dismal target therapy available. Luteolin is a natural product extracted from vegetables and fruits with a wide range of biological efficacies including anti-oxidative, anti-tumorigenic, and anti-inflammatory properties. This study was designed to examine the possible effect of luteolin on doxorubicin-induced cardiotoxicity, if any, and the mechanism(s) involved with a focus on mitochondrial autophagy. Luteolin application (10 μM) in adult mouse cardiomyocytes overtly improved doxorubicin-induced cardiomyocyte contractile dysfunction including elevated peak shortening amplitude and maximal velocity of shortening/relengthening along with unchanged duration of shortening and relengthening. Luteolin alleviated doxorubicin-induced cardiotoxicity including apoptosis, accumulation of reactive oxygen species (ROS) and loss of mitochondrial membrane potential. Furthermore, luteolin attenuated doxorubicin-induced cardiotoxicity through promoting mitochondrial autophagy in association with facilitating phosphorylation of Drp1 at Ser616, and upregulating TFEB expression. In addition, luteolin treatment partially attenuated low dose doxorubicin-induced elongation of mitochondria. Treatment of Mdivi-1, a Drp1 GTPase inhibitor, negated the protective effect of luteolin on levels of TFEB, LAMP1, and LC3B, as well as loss of mitochondrial membrane potential and cardiomyocyte contractile dysfunction in the face of doxorubicin challenge. Taken together, these findings provide novel insights for the therapeutic efficacy of luteolin against doxorubicin-induced cardiotoxicity possibly through improved mitochondrial autophagy.
Collapse
Affiliation(s)
- Haixia Xu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Wenjun Yu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Shiqun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| |
Collapse
|
118
|
Potential targets for intervention against doxorubicin-induced cardiotoxicity based on genetic studies: a systematic review of the literature. J Mol Cell Cardiol 2020; 138:88-98. [DOI: 10.1016/j.yjmcc.2019.11.150] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/24/2019] [Accepted: 11/03/2019] [Indexed: 12/12/2022]
|
119
|
Zou X, Shi Y, Zhu R, Han J, Han S. Organelle-Redirected Chameleon Sensor-Enabled Live Cell Imaging of Mitochondrial DNA. Anal Chem 2019; 91:15899-15907. [PMID: 31743646 DOI: 10.1021/acs.analchem.9b04364] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mitochondrial DNA (mtDNA) plays important roles in diverse physiological processes and myriad diseases. We herein report mtDNA imaging with a chameleon sensor containing a cationic rhodamine B (RB) entity for mitochondria targeting and a fluorogenic SYBR Green-I (SG) entity for DNA sensing. SG-RB selectively binds to mtDNA and gives green SG fluorescence in mitochondria of living cells but gives red RB fluorescence upon delivery of mitochondria into lysosomes in mitophagy. With the dual-color imaging, mtDNA aggregation and elevated mitophagy were identified in HeLa cells stressed with anticancer doxorubicin. These results suggest the utility of organelle-redirected DNA sensors for live cell imaging of mtDNA involved in myriad pathological disorders.
Collapse
Affiliation(s)
- Xiaoxue Zou
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory for Physical Chemistry of Solid Surfaces, the Key Laboratory for Chemical Biology of Fujian Province, the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network , Xiamen University , Xiamen 361005 , China
| | - Yilong Shi
- State key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences , Xiamen University , Xiamen 361005 , China
| | - Rui Zhu
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory for Physical Chemistry of Solid Surfaces, the Key Laboratory for Chemical Biology of Fujian Province, the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network , Xiamen University , Xiamen 361005 , China
| | - Jiahuai Han
- State key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences , Xiamen University , Xiamen 361005 , China
| | - Shoufa Han
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory for Physical Chemistry of Solid Surfaces, the Key Laboratory for Chemical Biology of Fujian Province, the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Innovation Center for Cell Signaling Network , Xiamen University , Xiamen 361005 , China.,State key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences , Xiamen University , Xiamen 361005 , China
| |
Collapse
|
120
|
Zhen J, Yu H, Ji H, Cai L, Leng J, Keller BB. Neonatal murine engineered cardiac tissue toxicology model: Impact of dexrazoxane on doxorubicin induced injury. Life Sci 2019; 239:117070. [PMID: 31751580 DOI: 10.1016/j.lfs.2019.117070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/28/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023]
Abstract
Doxorubicin (DOX) induced cardiotoxicity is a life-threatening side effect of chemotherapy and decreased cardiac function can present years after treatment. Despite the investigation of a broad range of pharmacologic interventions, to date the only drug shown to reduce DOX-related cardiotoxicity in preclinical studies and limited clinical trials is the iron chelating agent, dexrazoxane (DRZ), although the mechanisms responsible for DRZ mediated protection from DOX related cardiotoxicity remain unclear. Engineered cardiac tissues (ECTs) can be used for tissue repair strategies and as in vitro surrogate models to test cardiac toxicities and preventative countermeasures. Neonatal murine ECTs display cardiotoxicity in response to the environmental toxin, cadmium, and reduced cadmium toxicity with Zinc co-treatment, in part via the induction of the anti-oxidant Metallothionein (MT). We adapted our in vitro ECT model to determine the feasibility of using the ECT approach to investigate DOX-related cardiac injury and DRZ prevention. We found: (1) DOX induced dose and time dependent cell death in ECTs; (2) Zinc did not show protection from DOX cardiotoxicity; (3) MT overexpression induced by Zinc, low dose Cd pretreatment, or MT-overexpression (MT-TG) did not reduce ECT DOX cardiotoxicity; (4) DRZ reduced ECT DOX induced cell death; and (5) The mechanism of DRZ ECT protection from DOX cardiotoxicity was topoisomerase 2B (TOP2B) inhibition rather than reduced reactive oxygen species. Our data support the feasibility of ECTs as an in vitro platform technology for the investigation of drug induced cardiotoxicities including the role of TOP2B in DOX toxicity and DRZ mediated DOX toxicity prevention.
Collapse
Affiliation(s)
- Juan Zhen
- The First Hospital of Jilin University, Changchun 130021, China; The Pediatric Research Institute, Department of Pediatrics, the University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Haitao Yu
- The First Hospital of Jilin University, Changchun 130021, China; The Pediatric Research Institute, Department of Pediatrics, the University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Honglei Ji
- The First Hospital of Jilin University, Changchun 130021, China
| | - Lu Cai
- The Pediatric Research Institute, Department of Pediatrics, the University of Louisville School of Medicine, Louisville, KY 40292, USA; Department of Radiation Oncology, the University of Louisville School of Medicine, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Jiyan Leng
- The First Hospital of Jilin University, Changchun 130021, China.
| | - Bradley B Keller
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA; Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
121
|
The role of toll-like receptors in myocardial toxicity induced by doxorubicin. Immunol Lett 2019; 217:56-64. [PMID: 31707054 DOI: 10.1016/j.imlet.2019.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/21/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023]
Abstract
Doxorubicin is an effective antitumor drug commonly used in the treatment of a wide variety of cancers. However, doxorubicin may cause cardiac toxicity, which can cause congestive heart failure in severe cases, and this seriously limits its clinical application. It is believed that doxorubicin promotes the formation of reactive oxygen species, inducing oxidative stress, and at the same time, reduces the content of antioxidant substances in cardiac tissues, causing adverse effects. Toll-like receptors (TLRs) are biomolecules expressed on the surfaces of macrophages, dendritic cells, and epithelial cells that can recognize various types of pathogen-related or damage-related molecular patterns. In recent years, a large number of studies have confirmed that TLRs play important roles in the cardiac toxicity induced by doxorubicin. This review aimed to explore the role of TLRs in the cardiac toxicity induced by doxorubicin and provide possible solutions.
Collapse
|
122
|
Mitophagy, Mitochondrial Dynamics, and Homeostasis in Cardiovascular Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9825061. [PMID: 31781358 PMCID: PMC6875274 DOI: 10.1155/2019/9825061] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022]
Abstract
Biological aging is an inevitable and independent risk factor for a wide array of chronic diseases including cardiovascular and metabolic diseases. Ample evidence has established a pivotal role for interrupted mitochondrial homeostasis in the onset and development of aging-related cardiovascular anomalies. A number of culprit factors have been suggested in aging-associated mitochondrial anomalies including oxidative stress, lipid toxicity, telomere shortening, metabolic disturbance, and DNA damage, with recent findings revealing a likely role for compromised mitochondrial dynamics and mitochondrial quality control machinery such as autophagy. Mitochondria undergo consistent fusion and fission, which are crucial for mitochondrial homeostasis and energy adaptation. Autophagy, in particular, mitochondria-selective autophagy, namely, mitophagy, refers to a highly conservative cellular process to degrade and clear long-lived or damaged cellular organelles including mitochondria, the function of which gradually deteriorates with increased age. Mitochondrial homeostasis could be achieved through a cascade of independent but closely related processes including fusion, fission, mitophagy, and mitochondrial biogenesis. With improved health care and increased human longevity, the ever-rising aging society has imposed a high cardiovascular disease prevalence. It is thus imperative to understand the role of mitochondrial homeostasis in the regulation of lifespan and healthspan. Targeting mitochondrial homeostasis should offer promising novel therapeutic strategies against aging-related complications, particularly cardiovascular diseases.
Collapse
|
123
|
Zhang YJ, Huang H, Liu Y, Kong B, Wang G. MD-1 Deficiency Accelerates Myocardial Inflammation and Apoptosis in Doxorubicin-Induced Cardiotoxicity by Activating the TLR4/MAPKs/Nuclear Factor kappa B (NF-κB) Signaling Pathway. Med Sci Monit 2019; 25:7898-7907. [PMID: 31636246 PMCID: PMC6820359 DOI: 10.12659/msm.919861] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/02/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Myocardial apoptosis and inflammation play important roles in doxorubicin (DOX)-caused cardiotoxicity. Our prior studies have characterized the effects of myeloid differentiation protein 1(MD-1) in pathological cardiac remodeling and myocardial ischemia/reperfusion (I/R) injury, but its participations and potential molecular mechanisms in DOX-caused cardiotoxicity remain unknown. MATERIAL AND METHODS In the present study, MD-1 knockout mice were generated, and a single intraperitoneal injection of DOX (15 mg/kg) was performed to elicit DOX-induced cardiotoxicity. Cardiac function, histological change, mitochondrial structure, myocardial death, apoptosis, inflammation, and molecular alterations were measured systemically. RESULTS The results showed that the protein and mRNA levels of MD-1 were dramatically downregulated in DOX-treated cardiomyocytes. DOX insult markedly accelerated cardiac dysfunction and injury, followed by enhancements of apoptosis and inflammation, all of which were further aggravated in MD-1 knockout mice. Mechanistically, the TLR4/MAPKs/NF-kappaB pathways, which were over-activated in MD-1-deficient mice, were significantly increased in DOX-damaged cardiomyocytes. Moreover, the abolishment of TLR4 or NF-kappaB via a specific inhibitor exerted protective effects against the adverse effects of MD-1 loss on DOX-caused cardiotoxicity. CONCLUSIONS Collectively, these findings suggest that MD-1 is a novel target for the treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ying-Jun Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
- Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, P.R. China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
- Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, P.R. China
| | - Yu Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
- Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, P.R. China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
- Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, P.R. China
| | - Guangji Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
- Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, P.R. China
| |
Collapse
|