1
|
Kuang H, Li D, Chen Y, Zou H, Li F, Gong Z, Long Y, Zhou H, Du H, Yin Y. Valine acts as an early biomarker and exacerbates pathological cardiac hypertrophy by impairing mitochondrial quality control. Atherosclerosis 2025; 405:119216. [PMID: 40318256 DOI: 10.1016/j.atherosclerosis.2025.119216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVE Pathological cardiac hypertrophy is an independent risk factor for heart failure (HF). Early identification and timely treatment are crucial for significantly delaying the progression of HF. METHODS Targeted amino acid metabolomics and RNA sequencing (RNA-seq) were combined to explore the underlying mechanism. In vitro, H9c2 cells were stimulated with angiotensin II (Ang II) or were incubated with extra valine after Ang II stimulation. The branched chain alpha-ketoate dehydrogenase kinase (Bckdk) inhibitor 3,6-dichlorobenzo[b]thiophene-2-carboxylic acid (BT2) and rapamycin were utilized to confirm the role of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway in this process. RESULTS A significant accumulation of valine was detected within hypertrophic hearts from spontaneously hypertensive rats (SHR). When branched chain amino acid (BCAA) degradation was increased by BT2, the most pronounced decrease was observed in the valine level (Δ = 0.185 μmol/g, p < 0.001), and cardiac hypertrophy was ameliorated. The role of imbalanced mitochondrial quality control (MQC), including the suppression of mitophagy and excessive mitochondrial fission, was revealed in myocardial hypertrophy. In vitro, high concentrations of valine exacerbated cardiomyocyte hypertrophy stimulated by Any II, resulting in the accumulation of impaired mitochondria and respiratory chain dysfunction. BT2, rapamycin, and mitochondrial division inhibitor 1 (Mdivi-1) all ameliorated MQC imbalance, mitochondrial damage and oxidative stress in hypertensive models with high valine concentration. CONCLUSION Valine exacerbated pathological cardiac hypertrophy by causing a MQC imbalance, probably as an early biomarker for cardiac hypertrophy under chronic hypertension.
Collapse
Affiliation(s)
- Hongyu Kuang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, China; Chongqing Key Laboratory of Cardiac Electrophysiology, Cardiovascular Neuromodulation Research and Treatment Center, China; Department of Cardiology, University-town Hospital of Chongqing Medical University, China
| | - Dan Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, China; Chongqing Key Laboratory of Cardiac Electrophysiology, Cardiovascular Neuromodulation Research and Treatment Center, China
| | - Yunlin Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, China; Chongqing Key Laboratory of Cardiac Electrophysiology, Cardiovascular Neuromodulation Research and Treatment Center, China
| | - Hongmi Zou
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, China
| | - Fang Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, China; Chongqing Key Laboratory of Cardiac Electrophysiology, Cardiovascular Neuromodulation Research and Treatment Center, China
| | - Zhiyan Gong
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, China
| | - Yuxiang Long
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, China; Chongqing Key Laboratory of Cardiac Electrophysiology, Cardiovascular Neuromodulation Research and Treatment Center, China
| | - Hao Zhou
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, China; Chongqing Key Laboratory of Cardiac Electrophysiology, Cardiovascular Neuromodulation Research and Treatment Center, China
| | - Huaan Du
- Department of Cardiology, University-town Hospital of Chongqing Medical University, China.
| | - Yuehui Yin
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, China; Chongqing Key Laboratory of Cardiac Electrophysiology, Cardiovascular Neuromodulation Research and Treatment Center, China.
| |
Collapse
|
2
|
Jia H, Song Y, Hua Y, Li K, Li S, Wang Y. Molecular Mechanism of Aerobic Exercise Ameliorating Myocardial Mitochondrial Injury in Mice with Heart Failure. Int J Mol Sci 2025; 26:2136. [PMID: 40076760 PMCID: PMC11901053 DOI: 10.3390/ijms26052136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 03/14/2025] Open
Abstract
To explore the molecular mechanism of aerobic exercise to improve heart failure and to provide a theoretical basis and experimental reference for the treatment of heart failure. Nine-week-old male mice were used to establish a left ventricular pressure overload-induced heart failure model by transverse aortic constriction (TAC). The mice were randomly divided into four groups: a sham group (SHAM), heart failure group (HF), heart failure + SKQ1 group (HS) and heart failure + aerobic exercise group (HE). The mice in the HE group were subjected to moderate-intensity aerobic exercise interventions. The mitochondrion-targeting antioxidant (SKQ1) contains the lipophilic cation TPP, which targets scavenging mitochondrial ROS. The HS group was subjected to SKQ1 (100 nmol/kg/d) interventions, which were initiated 1 week after the surgery, and the interventions lasted 8 weeks. Cardiac function was assessed by ultrasound, cardiomyocyte size by H&E and WGA staining, myocardial fibrosis by Masson's staining, and myocardial tissue oxidative stress and apoptosis by DHE and TUNEL fluorescence staining, respectively. Western blotting was used to detect the expression of mitochondrial quality control, inflammation, and apoptosis-related proteins. In the cellular level, an in vitro cellular model was established by isolating primary cardiomyocytes from neonatal mice (2-3 days) and intervening with Ang II (1 μM) to mimic heart failure. Oxidative stress and mitochondrial membrane potential were determined in the cardiomyocytes of each group by DHE and JC-1 staining, respectively. Myocardial fibrosis was increased significantly and cardiac function was reduced significantly in the heart failure mice. Aerobic exercise and SKQ1 intervention improved cardiac function and reduced myocardial hypertrophy and myocardial fibrosis in the heart failure mice significantly. Meanwhile, aerobic exercise and SKQ1 intervention reduced the number of DHE-positive particles (p < 0.01) and inhibited myocardial oxidative stress in the heart failure mice significantly. Aerobic exercise also reduced DRP1, Parkin, and BNIP3 protein expression (p < 0.05, p < 0.01), and increased OPA1 and PINK1 protein expression (p < 0.05, p < 0.01) significantly. Moreover, aerobic exercise and SKQ1 intervention decreased the number of TUNEL-positive particles and the expression of inflammation- and apoptosis-related proteins NLRP3, TXNIP, Caspase-1, IL-1β, BAX, BAK, and p53 significantly (p < 0.05, p < 0.01). In addition, the AMPK agonist AICAR and the mitochondria-targeted ROS scavenger (SKQ1) ameliorated AngII-induced mitochondrial fragmentation and decreased mitochondrial membrane potential in cardiomyocytes significantly. It was shown that inhibition of mitochondrial ROS by aerobic exercise, which in turn inhibits mitochondrial damage, improves mitochondrial quality control, and reduces myocardial inflammatory and apoptosis, may be an important molecular mechanism by which aerobic exercise exerts endogenous antioxidant protective effects to improve cardiac function.
Collapse
Affiliation(s)
| | | | | | | | | | - Youhua Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shanxi Normal University, Xi’an 710119, China; (H.J.); (Y.S.); (Y.H.); (K.L.); (S.L.)
| |
Collapse
|
3
|
Zhang H, Zhang Y, Zhang J, Jia D. Exercise Alleviates Cardiovascular Diseases by Improving Mitochondrial Homeostasis. J Am Heart Assoc 2024; 13:e036555. [PMID: 39291488 DOI: 10.1161/jaha.124.036555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Engaging in regular exercise and physical activity contributes to delaying the onset of cardiovascular diseases (CVDs). However, the physiological mechanisms underlying the benefits of regular exercise or physical activity in CVDs remain unclear. The disruption of mitochondrial homeostasis is implicated in the pathological process of CVDs. Exercise training effectively delays the onset and progression of CVDs by significantly ameliorating the disruption of mitochondrial homeostasis. This includes improving mitochondrial biogenesis, increasing mitochondrial fusion, decreasing mitochondrial fission, promoting mitophagy, and mitigating mitochondrial morphology and function. This review provides a comprehensive overview of the benefits of physical exercise in the context of CVDs, establishing a connection between the disruption of mitochondrial homeostasis and the onset of these conditions. Through a detailed examination of the underlying molecular mechanisms within mitochondria, the study illuminates how exercise can provide innovative perspectives for future therapies for CVDs.
Collapse
Affiliation(s)
- Huijie Zhang
- School of Exercise and health Shanghai University of Sport Shanghai China
| | - Yuxuan Zhang
- School of Exercise and health Shanghai University of Sport Shanghai China
| | - Jiaqiao Zhang
- School of Exercise and health Shanghai University of Sport Shanghai China
| | - Dandan Jia
- School of Exercise and health Shanghai University of Sport Shanghai China
| |
Collapse
|
4
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
5
|
Eden M, Leye M, Hahn J, Heilein E, Luzarowski M, Völschow B, Tannert C, Sossalla S, Lucena-Porcel C, Frank D, Frey N. Mst4, a novel cardiac STRIPAK complex-associated kinase, regulates cardiomyocyte growth and survival and is upregulated in human cardiomyopathy. J Biol Chem 2024; 300:107255. [PMID: 38579991 PMCID: PMC11087964 DOI: 10.1016/j.jbc.2024.107255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/07/2024] Open
Abstract
Myocardial failure is associated with adverse remodeling, including loss of cardiomyocytes, hypertrophy, and alterations in cell-cell contacts. Striatin-interacting phosphatase and kinase (STRIPAK) complexes and their mammalian STE20-like kinase 4 (Mst4) have been linked to development of different diseases. The role and targets of Mst4 in cardiomyocytes have not been investigated yet. Multitissue immunoblot experiments show highly enriched Mst4 expression in rodent hearts. Analyses of human biopsy samples from patients suffering from dilated cardiomyopathy revealed that Mst4 is upregulated (5- to 8-fold p < 0.001) compared with nonfailing controls. Increased abundance of Mst4 could also be detected in mouse models of cardiomyopathy. We confirmed that Mst4 interacts with STRIPAK components in neonatal rat ventricular cardiomyocytes, indicating that STRIPAK is present in the heart. Immunofluorescence stainings and molecular interaction studies revealed that Mst4 is localized to the intercalated disc and interacts with several intercalated disc proteins. Overexpression of Mst4 in cardiomyocytes results in hypertrophy compared with controls. In adult rat cardiomyocytes, Mst4 overexpression increases cellular and sarcomeric fractional shortening (p < 0.05), indicating enhanced contractility. Overexpression of Mst4 also inhibits apoptosis shown by reduction of cleaved caspase3 (-69%, p < 0.0001), caspase7 (-80%, p < 0.0001), and cleaved Parp1 (-27%, p < 0.001). To elucidate potential Mst4 targets, we performed phosphoproteomics analyses in neonatal rat cardiomyocytes after Mst4 overexpression and inhibition. The results revealed target candidates of Mst4 at the intercalated disc. We identified Mst4 as a novel cardiac kinase that is upregulated in cardiomyopathy-regulating cardiomyocyte growth and survival.
Collapse
Affiliation(s)
- Matthias Eden
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Marius Leye
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Justus Hahn
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Emanuel Heilein
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany
| | - Marcin Luzarowski
- Core Facility for Mass Spectrometry and Proteomics, Center for Molecular Biology at Heidelberg University (ZMBH), Heidelberg, Germany
| | - Bill Völschow
- German Centre for Cardiovascular Research, Kiel, Lübeck, Hamburg, Germany; Department of Cardiology, University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Christin Tannert
- German Centre for Cardiovascular Research, Kiel, Lübeck, Hamburg, Germany; Department of Internal Medicine III (Cardiology and Angiology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Samuel Sossalla
- Department of Cardiology, University of Giessen, Giessen and Kerckhoff Heart and Lung Centre, Giessen, Germany
| | - Carlota Lucena-Porcel
- Tissue Bank of the National Center of Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Derk Frank
- German Centre for Cardiovascular Research, Kiel, Lübeck, Hamburg, Germany; Department of Internal Medicine III (Cardiology and Angiology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Norbert Frey
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, Mannheim/Heidelberg, Germany.
| |
Collapse
|
6
|
Mota GAF, de Souza SLB, Vileigas DF, da Silva VL, Sant'Ana PG, Costa LCDS, Padovani CR, Zanatti Bazan SG, Buzalaf MAR, Santos LDD, Okoshi MP, Gatto M, Cicogna AC. Myocardial proteome changes in aortic stenosis rats subjected to long-term aerobic exercise. J Cell Physiol 2024; 239:e31199. [PMID: 38291668 DOI: 10.1002/jcp.31199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 02/01/2024]
Abstract
The effects of exercise training (ET) on the heart of aortic stenosis (AS) rats are controversial and the mechanisms involved in alterations induced by ET have been poorly clarified. In this study, we analyzed the myocardial proteome to identify proteins modulated by moderate-intensity aerobic ET in rats with chronic supravalvular AS. Wistar rats were divided into four groups: sedentary control (C-Sed), exercised control (C-Ex), sedentary aortic stenosis (AS-Sed), and exercised AS (AS-Ex). ET consisted of five treadmill running sessions per week for 16 weeks. Statistical analysis was performed by ANOVA or Kruskal-Wallis and Goodman tests. Results were discussed at a significance level of 5%. At the end of the experiment, AS-Ex rats had higher functional capacity, lower blood lactate concentration, and better cardiac structural and left ventricular (LV) functional parameters than the AS-Sed. Myocardial proteome analysis showed that AS-Sed had higher relative protein abundance related to the glycolytic pathway, oxidative stress, and inflammation, and lower relative protein abundance related to beta-oxidation than C-Sed. AS-Ex had higher abundance of one protein related to mitochondrial biogenesis and lower relative protein abundance associated with oxidative stress and inflammation than AS-Sed. Proteomic data were validated for proteins related to lipid and glycolytic metabolism. Chronic pressure overload changes the abundance of myocardial proteins that are mainly involved in lipid and glycolytic energy metabolism in rats. Moderate-intensity aerobic training attenuates changes in proteins related to oxidative stress and inflammation and increases the COX4I1 protein, related to mitochondrial biogenesis. Protein changes are combined with improved functional capacity, cardiac remodeling, and LV function in AS rats.
Collapse
Affiliation(s)
- Gustavo Augusto Ferreira Mota
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | - Sérgio Luiz Borges de Souza
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | | | - Vitor Loureiro da Silva
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | - Paula Grippa Sant'Ana
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | - Licia Carla da Silva Costa
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Carlos Roberto Padovani
- Department of Biostatistics, Institute of Bioscience, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Silméia Garcia Zanatti Bazan
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | | | | | - Marina Politi Okoshi
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | - Mariana Gatto
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | - Antonio Carlos Cicogna
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Wang X, Yu Q, Liao X, Fan M, Liu X, Liu Q, Wang M, Wu X, Huang CK, Tan R, Yuan J. Mitochondrial Dysfunction in Arrhythmia and Cardiac Hypertrophy. Rev Cardiovasc Med 2023; 24:364. [PMID: 39077079 PMCID: PMC11272842 DOI: 10.31083/j.rcm2412364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 07/31/2024] Open
Abstract
Arrhythmia and cardiac hypertrophy are two very common cardiovascular diseases that can lead to heart failure and even sudden death, thus presenting a serious threat to human life and health. According to global statistics, nearly one million people per year die from arrhythmia, cardiac hypertrophy and other associated cardiovascular diseases. Hence, there is an urgent need to find new treatment targets and to develop new intervention measures. Recently, mitochondrial dysfunction has been examined in relation to heart disease with a view to lowering the incidence of arrhythmia and cardiac hypertrophy. The heart is the body's largest energy consuming organ, turning over about 20 kg of adenosine triphosphate (ATP) per day in the mitochondria. Mitochondrial oxidative phosphorylation (OXPHOS) produces up to 90% of the ATP needed by cardiac muscle cells for contraction and relaxation. Dysfunction of heart mitochondria can therefore induce arrhythmia, cardiac hypertrophy and other cardiovascular diseases. Mitochondrial DNA (mtDNA) mutations cause disorders in OXPHOS and defects in the synthesis of muscle contraction proteins. These lead to insufficient production of secondary ATP, increased metabolic requirements for ATP by the myocardium, and the accumulation of reactive oxygen species (ROS). The resulting damage to myocardial cells eventually induces arrhythmia and cardiac hypertrophy. Mitochondrial damage decreases the efficiency of energy production, which further increases the production of ROS. The accumulation of ROS causes mitochondrial damage and eventually leads to a vicious cycle of mitochondrial damage and low efficiency of mitochondrial energy production. In this review, the mechanism underlying the development of arrhythmia and cardiac hypertrophy is described in relation to mitochondrial energy supply, oxidative stress, mtDNA mutation and Mitochondrial dynamics. Targeted therapy for arrhythmia and cardiac hypertrophy induced by mitochondrial dysfunction is also discussed in terms of its potential clinical value. These strategies should improve our understanding of mitochondrial biology and the pathogenesis of arrhythmia and cardiac hypertrophy. They may also identify novel strategies for targeting mitochondria in the treatment of these diseases.
Collapse
Affiliation(s)
- Xiaomei Wang
- College of Basic Medical, Jining Medical University, 272067 Jining,
Shandong, China
| | - Qianxue Yu
- College of Basic Medical, Jining Medical University, 272067 Jining,
Shandong, China
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
| | - Xuemei Liao
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
- College of Second Clinical Medicine, Jining Medical University, 272067
Jining, Shandong, China
| | - Mengying Fan
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
- College of Second Clinical Medicine, Jining Medical University, 272067
Jining, Shandong, China
| | - Xibin Liu
- College of Basic Medical, Jining Medical University, 272067 Jining,
Shandong, China
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
| | - Qian Liu
- College of Basic Medical, Jining Medical University, 272067 Jining,
Shandong, China
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
| | - Manru Wang
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
- College of Second Clinical Medicine, Jining Medical University, 272067
Jining, Shandong, China
| | - Xinyu Wu
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
- College of Second Clinical Medicine, Jining Medical University, 272067
Jining, Shandong, China
| | - Chun-Kai Huang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University,
School of Medicine, 200025 Shanghai, China
| | - Rubin Tan
- College of Basic Medical, Xuzhou Medical University, 221004 Xuzhou,
Jiangsu, China
| | - Jinxiang Yuan
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
- Lin He's Academician Workstation of New Medicine and Clinical Translation,
Jining Medical University, 272067 Jining, Shandong, China
| |
Collapse
|
8
|
Wang Y, Dai X, Li H, Jiang H, Zhou J, Zhang S, Guo J, Shen L, Yang H, Lin J, Yan H. The role of mitochondrial dynamics in disease. MedComm (Beijing) 2023; 4:e462. [PMID: 38156294 PMCID: PMC10753647 DOI: 10.1002/mco2.462] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/14/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023] Open
Abstract
Mitochondria are multifaceted and dynamic organelles regulating various important cellular processes from signal transduction to determining cell fate. As dynamic properties of mitochondria, fusion and fission accompanied with mitophagy, undergo constant changes in number and morphology to sustain mitochondrial homeostasis in response to cell context changes. Thus, the dysregulation of mitochondrial dynamics and mitophagy is unsurprisingly related with various diseases, but the unclear underlying mechanism hinders their clinical application. In this review, we summarize the recent developments in the molecular mechanism of mitochondrial dynamics and mitophagy, particularly the different roles of key components in mitochondrial dynamics in different context. We also summarize the roles of mitochondrial dynamics and target treatment in diseases related to the cardiovascular system, nervous system, respiratory system, and tumor cell metabolism demanding high-energy. In these diseases, it is common that excessive mitochondrial fission is dominant and accompanied by impaired fusion and mitophagy. But there have been many conflicting findings about them recently, which are specifically highlighted in this view. We look forward that these findings will help broaden our understanding of the roles of the mitochondrial dynamics in diseases and will be beneficial to the discovery of novel selective therapeutic targets.
Collapse
Affiliation(s)
- Yujuan Wang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Xinyan Dai
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Hui Li
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Huiling Jiang
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Junfu Zhou
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Shiying Zhang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Jiacheng Guo
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Lidu Shen
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Huantao Yang
- Immunotherapy LaboratoryQinghai Tibet Plateau Research InstituteSouthwest Minzu UniversityChengduSichuanChina
| | - Jie Lin
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| | - Hengxiu Yan
- Immunotherapy LaboratoryCollege of PharmacologySouthwest Minzu UniversityChengduSichuanChina
| |
Collapse
|
9
|
Reitz CJ, Kuzmanov U, Gramolini AO. Multi-omic analyses and network biology in cardiovascular disease. Proteomics 2023; 23:e2200289. [PMID: 37691071 DOI: 10.1002/pmic.202200289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/11/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023]
Abstract
Heart disease remains a leading cause of death in North America and worldwide. Despite advances in therapies, the chronic nature of cardiovascular diseases ultimately results in frequent hospitalizations and steady rates of mortality. Systems biology approaches have provided a new frontier toward unraveling the underlying mechanisms of cell, tissue, and organ dysfunction in disease. Mapping the complex networks of molecular functions across the genome, transcriptome, proteome, and metabolome has enormous potential to advance our understanding of cardiovascular disease, discover new disease biomarkers, and develop novel therapies. Computational workflows to interpret these data-intensive analyses as well as integration between different levels of interrogation remain important challenges in the advancement and application of systems biology-based analyses in cardiovascular research. This review will focus on summarizing the recent developments in network biology-level profiling in the heart, with particular emphasis on modeling of human heart failure. We will provide new perspectives on integration between different levels of large "omics" datasets, including integration of gene regulatory networks, protein-protein interactions, signaling networks, and metabolic networks in the heart.
Collapse
Affiliation(s)
- Cristine J Reitz
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
| | - Uros Kuzmanov
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
| | - Anthony O Gramolini
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Chang YW, Chen YC, Chen CC. Identification of Novel Targeting Sites of Calcineurin and CaMKII in Human Ca V3.2 T-Type Calcium Channel. Biomedicines 2023; 11:2891. [PMID: 38001892 PMCID: PMC10669385 DOI: 10.3390/biomedicines11112891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
The Cav3.2 T-type calcium channel is implicated in various pathological conditions, including cardiac hypertrophy, epilepsy, autism, and chronic pain. Phosphorylation of Cav3.2 by multiple kinases plays a pivotal role in regulating its calcium channel function. The calcium/calmodulin-dependent serine/threonine phosphatase, calcineurin, interacts physically with Cav3.2 and modulates its activity. However, it remains unclear whether calcineurin dephosphorylates Cav3.2, the specific spatial regions on Cav3.2 involved, and the extent of the quantitative impact. In this study, we elucidated the serine/threonine residues on Cav3.2 targeted by calcineurin using quantitative mass spectrometry. We identified six serine residues in the N-terminus, II-III loop, and C-terminus of Cav3.2 that were dephosphorylated by calcineurin. Notably, a higher level of dephosphorylation was observed in the Cav3.2 C-terminus, where calcineurin binds to this channel. Additionally, a previously known CaMKII-phosphorylated site, S1198, was found to be dephosphorylated by calcineurin. Furthermore, we also discovered that a novel CaMKII-phosphorylated site, S2137, underwent dephosphorylation by calcineurin. In CAD cells, a mouse central nervous system cell line, membrane depolarization led to an increase in the phosphorylation of endogenous Cav3.2 at S2137. Mutation of S2137 affected the calcium channel function of Cav3.2. Our findings advance the understanding of Cav3.2 regulation not only through kinase phosphorylation but also via calcineurin phosphatase dephosphorylation.
Collapse
Affiliation(s)
- Yu-Wang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan;
| | | | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan;
| |
Collapse
|
11
|
The Drp1-Mediated Mitochondrial Fission Protein Interactome as an Emerging Core Player in Mitochondrial Dynamics and Cardiovascular Disease Therapy. Int J Mol Sci 2023; 24:ijms24065785. [PMID: 36982862 PMCID: PMC10057413 DOI: 10.3390/ijms24065785] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
Mitochondria, the membrane-bound cell organelles that supply most of the energy needed for cell function, are highly regulated, dynamic organelles bearing the ability to alter both form and functionality rapidly to maintain normal physiological events and challenge stress to the cell. This amazingly vibrant movement and distribution of mitochondria within cells is controlled by the highly coordinated interplay between mitochondrial dynamic processes and fission and fusion events, as well as mitochondrial quality-control processes, mainly mitochondrial autophagy (also known as mitophagy). Fusion connects and unites neighboring depolarized mitochondria to derive a healthy and distinct mitochondrion. In contrast, fission segregates damaged mitochondria from intact and healthy counterparts and is followed by selective clearance of the damaged mitochondria via mitochondrial specific autophagy, i.e., mitophagy. Hence, the mitochondrial processes encompass all coordinated events of fusion, fission, mitophagy, and biogenesis for sustaining mitochondrial homeostasis. Accumulated evidence strongly suggests that mitochondrial impairment has already emerged as a core player in the pathogenesis, progression, and development of various human diseases, including cardiovascular ailments, the leading causes of death globally, which take an estimated 17.9 million lives each year. The crucial factor governing the fission process is the recruitment of dynamin-related protein 1 (Drp1), a GTPase that regulates mitochondrial fission, from the cytosol to the outer mitochondrial membrane in a guanosine triphosphate (GTP)-dependent manner, where it is oligomerized and self-assembles into spiral structures. In this review, we first aim to describe the structural elements, functionality, and regulatory mechanisms of the key mitochondrial fission protein, Drp1, and other mitochondrial fission adaptor proteins, including mitochondrial fission 1 (Fis1), mitochondrial fission factor (Mff), mitochondrial dynamics 49 (Mid49), and mitochondrial dynamics 51 (Mid51). The core area of the review focuses on the recent advances in understanding the role of the Drp1-mediated mitochondrial fission adaptor protein interactome to unravel the missing links of mitochondrial fission events. Lastly, we discuss the promising mitochondria-targeted therapeutic approaches that involve fission, as well as current evidence on Drp1-mediated fission protein interactions and their critical roles in the pathogeneses of cardiovascular diseases (CVDs).
Collapse
|
12
|
Liu X, Guo C, Zhang Q. Novel insights into the involvement of mitochondrial fission/fusion in heart failure: From molecular mechanisms to targeted therapies. Cell Stress Chaperones 2023; 28:133-144. [PMID: 36652120 PMCID: PMC10050249 DOI: 10.1007/s12192-023-01321-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/19/2023] Open
Abstract
Mitochondria are dynamic organelles that alter their morphology through fission (fragmentation) and fusion (elongation). These morphological changes correlate highly with mitochondrial functional adaptations to stressors, such as hypoxia, pressure overload, and inflammation, and are important in the setting of heart failure. Pathological mitochondrial remodeling, characterized by increased fission and reduced fusion, is associated with impaired mitochondrial respiration, increased mitochondrial oxidative stress, abnormal cytoplasmic calcium handling, and increased cardiomyocyte apoptosis. Considering the impact of the mitochondrial morphology on mitochondrial behavior and cardiomyocyte performance, altered mitochondrial dynamics could be expected to induce or exacerbate the pathogenesis and progression of heart failure. However, whether alterations in mitochondrial fission and fusion accelerate or retard the progression of heart failure has been the subject of intense debate. In this review, we first describe the physiological processes and regulatory mechanisms of mitochondrial fission and fusion. Then, we extensively discuss the pathological contributions of mitochondrial fission and fusion to heart failure. Lastly, we examine potential therapeutic approaches targeting mitochondrial fission/fusion to treat patients with heart failure.
Collapse
Affiliation(s)
- Xinxin Liu
- Department of First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Chenchen Guo
- Neck, Shoulder, Waist and Leg Pain Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiming Zhang
- Department of First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China.
| |
Collapse
|
13
|
Zhu T, Hu Q, Yuan Y, Yao H, Zhang J, Qi J. Mitochondrial dynamics in vascular remodeling and target-organ damage. Front Cardiovasc Med 2023; 10:1067732. [PMID: 36860274 PMCID: PMC9970102 DOI: 10.3389/fcvm.2023.1067732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
Vascular remodeling is the pathological basis for the development of many cardiovascular diseases. The mechanisms underlying endothelial cell dysfunction, smooth muscle cell phenotypic switching, fibroblast activation, and inflammatory macrophage differentiation during vascular remodeling remain elusive. Mitochondria are highly dynamic organelles. Recent studies showed that mitochondrial fusion and fission play crucial roles in vascular remodeling and that the delicate balance of fusion-fission may be more important than individual processes. In addition, vascular remodeling may also lead to target-organ damage by interfering with the blood supply to major body organs such as the heart, brain, and kidney. The protective effect of mitochondrial dynamics modulators on target-organs has been demonstrated in numerous studies, but whether they can be used for the treatment of related cardiovascular diseases needs to be verified in future clinical studies. Herein, we summarize recent advances regarding mitochondrial dynamics in multiple cells involved in vascular remodeling and associated target-organ damage.
Collapse
Affiliation(s)
- Tong Zhu
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingxun Hu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University, School of Medicine, Shanghai University, Shanghai, China,Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
| | - Yanggang Yuan
- Department of Nephrology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Huijuan Yao
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Zhang
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Jian Zhang,
| | - Jia Qi
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Jia Qi,
| |
Collapse
|
14
|
Quiles JM, Gustafsson ÅB. The role of mitochondrial fission in cardiovascular health and disease. Nat Rev Cardiol 2022; 19:723-736. [PMID: 35523864 PMCID: PMC10584015 DOI: 10.1038/s41569-022-00703-y] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/02/2022] [Indexed: 02/07/2023]
Abstract
Mitochondria are organelles involved in the regulation of various important cellular processes, ranging from ATP generation to immune activation. A healthy mitochondrial network is essential for cardiovascular function and adaptation to pathological stressors. Mitochondria undergo fission or fusion in response to various environmental cues, and these dynamic changes are vital for mitochondrial function and health. In particular, mitochondrial fission is closely coordinated with the cell cycle and is linked to changes in mitochondrial respiration and membrane permeability. Another key function of fission is the segregation of damaged mitochondrial components for degradation by mitochondrial autophagy (mitophagy). Mitochondrial fission is induced by the large GTPase dynamin-related protein 1 (DRP1) and is subject to sophisticated regulation. Activation requires various post-translational modifications of DRP1, actin polymerization and the involvement of other organelles such as the endoplasmic reticulum, Golgi apparatus and lysosomes. A decrease in mitochondrial fusion can also shift the balance towards mitochondrial fission. Although mitochondrial fission is necessary for cellular homeostasis, this process is often aberrantly activated in cardiovascular disease. Indeed, strong evidence exists that abnormal mitochondrial fission directly contributes to disease development. In this Review, we compare the physiological and pathophysiological roles of mitochondrial fission and discuss the therapeutic potential of preventing excessive mitochondrial fission in the heart and vasculature.
Collapse
Affiliation(s)
- Justin M Quiles
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
15
|
Zhao X, Xu H, Li Y, Liu Y, Li X, Zhou W, Wang J, Guo C, Sun Z, Li Y. Silica nanoparticles perturbed mitochondrial dynamics and induced myocardial apoptosis via PKA-DRP1-mitochondrial fission signaling. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 842:156854. [PMID: 35750168 DOI: 10.1016/j.scitotenv.2022.156854] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 05/20/2023]
Abstract
Silica nanoparticles (SiNPs) are among the most abundantly produced nanosized particles in the global market, and their potential toxicity has aroused a great concern. Increasing epidemiological investigations and experimental evidence revealed the threaten of SiNPs exposure to cardiovascular system. The myocardial toxicity caused by SiNPs was gradually demonstrated, nevertheless, the underlying mechanisms remain unclear. In view of mitochondria serving as the centrality in the prominent of cardiovascular disease, we investigated the role of mitochondria and related mechanisms in SiNPs-induced adverse effects on cardiomyocytes. As a result, SiNPs were found in cytoplasm, accompanied with morphological alterations in mitochondria, such as cristae fracture or disappearance, vacuolation. The induction of mitochondrial dysfunction by SiNPs was confirmed, as indicated by the excessive reactive oxygen species (ROS) formation, and blockage of cellular respiratory and ATP production. Concomitantly, SiNPs activated mitochondria-mediated apoptotic signaling in view of the up-regulated BAX, increased Caspase-9 cleavage and declined Bcl-2, ultimately resulting in myocardial apoptosis. It was noteworthy that SiNPs disturbed mitochondrial dynamics toward fission phenotype, which was supported by the dysregulated fission/fusion regulators. Especially, DRP1 and its phosphorylated level at s616 (p-DRP1s616) were up-regulated, whilst its phosphorylated level at s637 (p-DRP1s637) and PKA phosphorylation were down-regulated in SiNPs-treated cardiomyocytes in a dose-dependent manner. More importantly, the mechanistic investigations revealed PKA-DRP1-mediated mitochondrial fission was responsible for SiNPs-induced cardiomyocyte apoptosis through the mitochondria-mediated apoptotic way. This study firstly demonstrated the disturbance of mitochondrial dynamics played a crucial role in cardiomyocyte apoptosis caused by SiNPs, attributing to PKA-DRP1-mitochondrial fission signaling.
Collapse
Affiliation(s)
- Xinying Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Hailin Xu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yan Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Yufan Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Xueyan Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ji Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Caixia Guo
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yanbo Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
16
|
Protective roles of MITOL against myocardial senescence and ischemic injury partly via Drp1 regulation. iScience 2022; 25:104582. [PMID: 35789860 PMCID: PMC9249672 DOI: 10.1016/j.isci.2022.104582] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/30/2021] [Accepted: 06/07/2022] [Indexed: 11/21/2022] Open
Abstract
Abnormal mitochondrial fragmentation by dynamin-related protein1 (Drp1) is associated with the progression of aging-associated heart diseases, including heart failure and myocardial infarction (MI). Here, we report a protective role of outer mitochondrial membrane (OMM)-localized E3 ubiquitin ligase MITOL/MARCH5 against cardiac senescence and MI, partly through Drp1 clearance by OMM-associated degradation (OMMAD). Persistent Drp1 accumulation in cardiomyocyte-specific MITOL conditional-knockout mice induced mitochondrial fragmentation and dysfunction, including reduced ATP production and increased ROS generation, ultimately leading to myocardial senescence and chronic heart failure. Furthermore, ischemic stress-induced acute downregulation of MITOL, which permitted mitochondrial accumulation of Drp1, resulted in mitochondrial fragmentation. Adeno-associated virus-mediated delivery of the MITOL gene to cardiomyocytes ameliorated cardiac dysfunction induced by MI. Our findings suggest that OMMAD activation by MITOL can be a therapeutic target for aging-associated heart diseases, including heart failure and MI. MITOL is essential for maintaining cardiac function partly via Drp1 clearance MITOL deficiency causes cardiac aging partly via Drp1 accumulation Ischemic stress induces a rapid downregulation of MITOL MITOL expression attenuates cardiac dysfunction in acute myocardial infarction
Collapse
|
17
|
Yin X, Li Z, Lyu C, Wang Y, Ding S, Ma C, Wang J, Cui S, Wang J, Guo D, Xu R. Induced Effect of Zinc oxide nanoparticles on human acute myeloid leukemia cell apoptosis by regulating mitochondrial division. IUBMB Life 2022; 74:519-531. [PMID: 35383422 DOI: 10.1002/iub.2615] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/28/2022] [Accepted: 03/31/2022] [Indexed: 11/11/2022]
Abstract
Zinc oxide nanoparticles (ZnO NPs) have exhibited excellent anti-tumor, the present study aimed to elucidate the underlying mechanism of ZnO NPs induced apoptosis in acute myeloid leukemia (AML) cells by regulating mitochondrial division. THP-1 cells, an AML cell line, were first incubated with different concentrations ZnO NPs for 24 h. Next, the expression of Drp-1, Bcl-2, Bax mRNA and protein was detected, and the effects of ZnO NPs on the levels of reactive oxygen species (ROS), mitochondrial membrane potential (Δψm), apoptosis and ATP generation in THP-1 cells were measured. Moreover, the effect of Drp-1 inhibitor Mdivi-1 and ZnO NPs on THP-1 cells was also detected. The results showed that the THP-1 cells survival rate decreased with the increment of ZnO NPs concentration and incubation time in a dose- and time-dependent manner. ZnO NPs can reduce the cell Δψm and ATP levels, induce the ROS production, and increase the levels of mitochondrial division and apoptosis. In contrast, the apoptotic level was significantly reduced after intervention of Drp-1 inhibitor, suggesting that ZnO NPs can induce the apoptosis of THP-1 cells by regulating mitochondrial division. Overall, ZnO NPs may provide a new basis and idea in treating human acute myeloid leukemia in clinical practice. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xuewei Yin
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zonghong Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chunyi Lyu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Wang
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Shumin Ding
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Chenchen Ma
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Jingyi Wang
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Siyuan Cui
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Jinxin Wang
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruirong Xu
- Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| |
Collapse
|
18
|
Chen BR, Wei TW, Tang CP, Sun JT, Shan TK, Fan Y, Yang TT, Li YF, Ma Y, Wang SB, Wang ZM, Wang H, Shi JZ, Liu L, Chen JW, Zhou LH, Du C, Sun R, Wang QM, Wang LS. MNK2-eIF4E axis promotes cardiac repair in the infarcted mouse heart by activating cyclin D1. J Mol Cell Cardiol 2022; 166:91-106. [DOI: 10.1016/j.yjmcc.2022.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 02/15/2022] [Accepted: 02/23/2022] [Indexed: 10/19/2022]
|
19
|
Ala M, Ala M. Metformin for Cardiovascular Protection, Inflammatory Bowel Disease, Osteoporosis, Periodontitis, Polycystic Ovarian Syndrome, Neurodegeneration, Cancer, Inflammation and Senescence: What Is Next? ACS Pharmacol Transl Sci 2021; 4:1747-1770. [PMID: 34927008 DOI: 10.1021/acsptsci.1c00167] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Indexed: 12/15/2022]
Abstract
Diabetes is accompanied by several complications. Higher prevalence of cancers, cardiovascular diseases, chronic kidney disease (CKD), obesity, osteoporosis, and neurodegenerative diseases has been reported among patients with diabetes. Metformin is the oldest oral antidiabetic drug and can improve coexisting complications of diabetes. Clinical trials and observational studies uncovered that metformin can remarkably prevent or alleviate cardiovascular diseases, obesity, polycystic ovarian syndrome (PCOS), osteoporosis, cancer, periodontitis, neuronal damage and neurodegenerative diseases, inflammation, inflammatory bowel disease (IBD), tuberculosis, and COVID-19. In addition, metformin has been proposed as an antiaging agent. Numerous mechanisms were shown to be involved in the protective effects of metformin. Metformin activates the LKB1/AMPK pathway to interact with several intracellular signaling pathways and molecular mechanisms. The drug modifies the biologic function of NF-κB, PI3K/AKT/mTOR, SIRT1/PGC-1α, NLRP3, ERK, P38 MAPK, Wnt/β-catenin, Nrf2, JNK, and other major molecules in the intracellular signaling network. It also regulates the expression of noncoding RNAs. Thereby, metformin can regulate metabolism, growth, proliferation, inflammation, tumorigenesis, and senescence. Additionally, metformin modulates immune response, autophagy, mitophagy, endoplasmic reticulum (ER) stress, and apoptosis and exerts epigenetic effects. Furthermore, metformin protects against oxidative stress and genomic instability, preserves telomere length, and prevents stem cell exhaustion. In this review, the protective effects of metformin on each disease will be discussed using the results of recent meta-analyses, clinical trials, and observational studies. Thereafter, it will be meticulously explained how metformin reprograms intracellular signaling pathways and alters molecular and cellular interactions to modify the clinical presentations of several diseases.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), 1416753955 Tehran, Iran
| | - Mahan Ala
- School of Dentistry, Golestan University of Medical Sciences (GUMS), 4814565589 Golestan, Iran
| |
Collapse
|
20
|
Lohanadan K, Molt S, Dierck F, van der Ven PFM, Frey N, Höhfeld J, Fürst DO. Isoform-specific functions of synaptopodin-2 variants in cytoskeleton stabilization and autophagy regulation in muscle under mechanical stress. Exp Cell Res 2021; 408:112865. [PMID: 34637763 DOI: 10.1016/j.yexcr.2021.112865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 10/01/2021] [Accepted: 10/03/2021] [Indexed: 11/17/2022]
Abstract
Protein homeostasis (proteostasis) in multicellular organisms depends on the maintenance of force-bearing and force-generating cellular structures. Within myofibrillar Z-discs of striated muscle, isoforms of synaptopodin-2 (SYNPO2/myopodin) act as adapter proteins that are engaged in proteostasis of the actin-crosslinking protein filamin C (FLNc) under mechanical stress. SYNPO2 directly binds F-actin, FLNc and α-actinin and thus contributes to the architectural features of the actin cytoskeleton. By its association with autophagy mediating proteins, i.e. BAG3 and VPS18, SYNPO2 is also engaged in protein quality control and helps to target mechanical unfolded and damaged FLNc for degradation. Here we show that deficiency of all SYNPO2-isoforms in myotubes leads to decreased myofibrillar stability and deregulated autophagy under mechanical stress. In addition, isoform-specific proteostasis functions were revealed. The PDZ-domain containing variant SYNPO2b and the shorter, PDZ-less isoform SYNPO2e both localize to Z-discs. Yet, SYNPO2e is less stably associated with the Z-disc than SYNPO2b, and is dynamically transferred into FLNc-containing myofibrillar lesions under mechanical stress. SYNPO2e also recruits BAG3 into these lesions via interaction with the WW domain of BAG3. Our data provide evidence for a role of myofibrillar lesions as a transient quality control compartment essential to prevent and repair contraction-induced myofibril damage in muscle and indicate an important coordinating activity for SYNPO2 therein.
Collapse
Affiliation(s)
- Keerthika Lohanadan
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Sibylle Molt
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Franziska Dierck
- Department of Internal Medicine III, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Norbert Frey
- Department of Internal Medicine III, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany; German Centre for Cardiovascular Research, Partner Site Heidelberg, 69120 Heidelberg, Germany
| | - Jörg Höhfeld
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany.
| |
Collapse
|
21
|
Chang YW, Song ZH, Chen CC. FAK regulates cardiomyocyte mitochondrial fission and function through Drp1. FEBS J 2021; 289:1897-1910. [PMID: 34739186 DOI: 10.1111/febs.16263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 09/19/2021] [Accepted: 11/03/2021] [Indexed: 11/29/2022]
Abstract
Loss of the mitochondrial fission enzyme dynamin-related protein 1 (Drp1) in cardiomyocytes results in energy shortage and heart failure. We aim to understand the intracellular signal pathway and extracellular factors regulating Drp1 phosphorylation and mitochondrial morphology and function in cardiomyocytes. We found cyclic mechanical stretching induced mitochondrial fission through Drp1 and focal adhesion kinase (FAK) in neonatal rat ventricular myocytes (NRVMs). FAK regulated phosphorylation of Drp1 and mitochondrial Drp1 levels. Extracellular fibronectin activated Drp1 and caused mitochondrial fission through FAK and extracellular signal-regulated kinase 1/2 (ERK1/2). Fibronectin increased NRVMs oxygen consumption rate and ATP content via FAK-ERK1/2-Drp1. Inhibition of the FAK-ERK1/2-Drp1 pathway caused cellular energy shortage. In addition, the FAK-ERK1/2-Drp1 pathway was rapidly activated by adrenergic agonists and contributed to agonists-stimulated NRVMs respiration. Interestingly, fibronectin limited the adrenergic agonists-induced NRVMs respiration by restricting phosphorylation of Drp1. Our results suggest that extracellular fibronectin and adrenergic stimulations use the FAK-ERK1/2-Drp1 pathway to regulate mitochondrial morphology and function in cardiomyocytes.
Collapse
Affiliation(s)
- Yu-Wang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taiwan
| | - Zong-Han Song
- Institute of Biomedical Sciences, Academia Sinica, Taiwan
| | | |
Collapse
|
22
|
Exploration of the lactation function of protein phosphorylation sites in goat mammary tissues by phosphoproteome analysis. BMC Genomics 2021; 22:703. [PMID: 34583635 PMCID: PMC8479986 DOI: 10.1186/s12864-021-07993-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/08/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Protein phosphorylation plays an important role in lactation. Differentially modified phosphorylation sites and phosphorylated proteins between peak lactation (PL, 90 days postpartum) and late lactation (LL, 280 days postpartum) were investigated using an integrated approach, namely, liquid chromatography with tandem mass spectrometry (LC-MS/MS) and tandem mass tag (TMT) labeling, to determine the molecular changes in the mammary tissues during the different stages of goat lactation. RESULTS A total of 1,938 (1,111 upregulated, 827 downregulated) differentially modified phosphorylation sites of 1,172 proteins were identified (P values < 0.05 and fold change of phosphorylation ratios > 1.5). Multiple phosphorylation sites of FASN, ACACA, mTOR, PRKAA, IRS1, RPS6KB, EIF4EBP1, JUN, and TSC2 were different in PL compared with LL. In addition, the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that the calcium signaling pathway, oxytocin signaling pathway and MAPK signaling pathway were enriched. The western blot results showed that the phosphorylation levels of ACACA (Ser80), EIF4EBP1 (Thr46) and IRS1 (Ser312) increased and JUN (Ser63) decreased in PL compared with LL. These results were consistent with the phosphoproteome results. CONCLUSIONS In this study, we identified for the first time the differentially modified phosphorylation sites in goat mammary tissues between PL and LL. These results indicate that the multiple differentially modified phosphorylation sites of FASN, ACACA, mTOR, PRKAA, IRS1, RPS6KB, EIF4EBP1, TSC2, and JUN and proteins involved in the calcium signaling pathway, oxytocin signaling pathway, and MAPK signaling pathway are worthy of further exploration.
Collapse
|
23
|
Abstract
Cardiomyopathy affects approximately 1 in 500 adults and is the leading cause of death. Familial cases are common, and mutations in many genes are involved in cardiomyopathy, especially those in genes encoding cytoskeletal, sarcomere, and nuclear envelope proteins. Filamin C is an actin-binding protein encoded by filamin C (FLNC) gene and participates in sarcomere stability maintenance. FLNC was first demonstrated to be a causal gene of myofibrillar myopathy; recently, it has been found that FLNC mutation plays a critical role in the pathogenesis of cardiomyopathy. In this review, we summarized the physiological roles of filamin C in cardiomyocytes and the genetic evidence for links between FLNC mutations and cardiomyopathies. Truncated FLNC is enriched in dilated cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy. Non-truncated FLNC is enriched in hypertrophic cardiomyopathy and restrictive cardiomyopathy. Two major pathomechanisms in FLNC-related cardiomyopathy have been described: protein aggregation resulting from non-truncating mutations and haploinsufficiency triggered by filamin C truncation. Therefore, it is important to understand the cellular biology and molecular regulation of FLNC to design new therapies to treat patients with FLNC-related cardiomyopathy.
Collapse
|
24
|
Aung LHH, Chen X, Cueva Jumbo JC, Li Z, Wang SY, Zhao C, Liu Z, Wang Y, Li P. Cardiomyocyte mitochondrial dynamic-related lncRNA 1 (CMDL-1) may serve as a potential therapeutic target in doxorubicin cardiotoxicity. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:638-651. [PMID: 34589283 PMCID: PMC8463323 DOI: 10.1016/j.omtn.2021.08.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/13/2021] [Indexed: 11/19/2022]
Abstract
Doxorubicin (DOX)-induced cardiotoxicity has been one of the major limitations for its clinical use. Although extensive studies have been conducted to decipher the molecular mechanisms underlying DOX cardiotoxicity, no effective preventive or therapeutic measures have yet been identified. Microarray analysis showed that multiple long non-coding RNAs (lncRNAs) are differentially expressed between control- and DOX-treated cardiomyocytes. Functional enrichment analysis indicated that the differentially expressed genes are annotated to cardiac hypertrophic pathways. Among differentially expressed lncRNAs, cardiomyocyte mitochondrial dynamic-related lncRNA 1 (CMDL-1) is the most significantly downregulated lncRNA in cardiomyocytes after DOX exposure. The protein-RNA interaction analysis showed that CMDL-1 may target dynamin-related protein 1 (Drp1). Mechanistic analysis shows that lentiviral overexpression of CMDL-1 prevents DOX-induced mitochondrial fission and apoptosis in cardiomyocytes. However, overexpression of CMDL-1 cannot effectively reduce mitochondrial fission when Drp1 is minimally expressed by small interfering RNA Drp1 (siDrp1). Overexpression of CMDL-1 promotes the association between CMDL-1 and Drp1, as well as with phosphorylated (p-)Drp1, as evidenced by RNA immunoprecipitation analysis. These data indicate the role of CMDL-1 in posttranslational modification of a target protein via regulating its phosphorylation. Collectively, our data indicate that CMDL-1 may play an anti-apoptotic role in DOX cardiotoxicity by regulating Drp1 S637 phosphorylation. Thus, CMDL-1 may serve as a potential therapeutic target in DOX cardiotoxicity.
Collapse
Affiliation(s)
- Lynn Htet Htet Aung
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266000, Shandong, China
- Center for Bioinformatics, Institute of Translational Medicine, College of Medicine, Qingdao University, Qingdao 266000, Shandong, China
| | - Xiatian Chen
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266000, Shandong, China
| | - Juan Carlos Cueva Jumbo
- School of Preclinical Medicine, Nanobody Research Center, Guangxi Medical University, Nanning 530021, China
| | - Zhe Li
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266000, Shandong, China
| | - Shao-ying Wang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266000, Shandong, China
| | - Cheng Zhao
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266000, Shandong, China
| | - Ziqian Liu
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266000, Shandong, China
| | - Yin Wang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266000, Shandong, China
| | - Peifeng Li
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266000, Shandong, China
- Center for Bioinformatics, Institute of Translational Medicine, College of Medicine, Qingdao University, Qingdao 266000, Shandong, China
| |
Collapse
|
25
|
Aung LHH, Jumbo JCC, Wang Y, Li P. Therapeutic potential and recent advances on targeting mitochondrial dynamics in cardiac hypertrophy: A concise review. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:416-443. [PMID: 34484866 PMCID: PMC8405900 DOI: 10.1016/j.omtn.2021.06.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pathological cardiac hypertrophy begins as an adaptive response to increased workload; however, sustained hemodynamic stress will lead it to maladaptation and eventually cardiac failure. Mitochondria, being the powerhouse of the cells, can regulate cardiac hypertrophy in both adaptive and maladaptive phases; they are dynamic organelles that can adjust their number, size, and shape through a process called mitochondrial dynamics. Recently, several studies indicate that promoting mitochondrial fusion along with preventing mitochondrial fission could improve cardiac function during cardiac hypertrophy and avert its progression toward heart failure. However, some studies also indicate that either hyperfusion or hypo-fission could induce apoptosis and cardiac dysfunction. In this review, we summarize the recent knowledge regarding the effects of mitochondrial dynamics on the development and progression of cardiac hypertrophy with particular emphasis on the regulatory role of mitochondrial dynamics proteins through the genetic, epigenetic, and post-translational mechanisms, followed by discussing the novel therapeutic strategies targeting mitochondrial dynamic pathways.
Collapse
Affiliation(s)
- Lynn Htet Htet Aung
- Center for Molecular Genetics, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.,Center for Bioinformatics, Institute for Translational Medicine, School of Basic Science, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Juan Carlos Cueva Jumbo
- School of Preclinical Medicine, Nanobody Research Center, Guangxi Medical University, Nanning 530021, China
| | - Yin Wang
- Center for Molecular Genetics, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Center for Molecular Genetics, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.,Center for Bioinformatics, Institute for Translational Medicine, School of Basic Science, College of Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
26
|
Kwon HK, Choi H, Park SG, Park WJ, Kim, DH, Park ZY. Integrated Quantitative Phosphoproteomics and Cell-based Functional Screening Reveals Specific Pathological Cardiac Hypertrophy-related Phosphorylation Sites. Mol Cells 2021; 44:500-516. [PMID: 34158421 PMCID: PMC8334354 DOI: 10.14348/molcells.2021.4002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 01/07/2019] [Indexed: 12/29/2022] Open
Abstract
Cardiac hypertrophic signaling cascades resulting in heart failure diseases are mediated by protein phosphorylation. Recent developments in mass spectrometry-based phosphoproteomics have led to the identification of thousands of differentially phosphorylated proteins and their phosphorylation sites. However, functional studies of these differentially phosphorylated proteins have not been conducted in a large-scale or high-throughput manner due to a lack of methods capable of revealing the functional relevance of each phosphorylation site. In this study, an integrated approach combining quantitative phosphoproteomics and cell-based functional screening using phosphorylation competition peptides was developed. A pathological cardiac hypertrophy model, junctate-1 transgenic mice and control mice, were analyzed using label-free quantitative phosphoproteomics to identify differentially phosphorylated proteins and sites. A cell-based functional assay system measuring hypertrophic cell growth of neonatal rat ventricle cardiomyocytes (NRVMs) following phenylephrine treatment was applied, and changes in phosphorylation of individual differentially phosphorylated sites were induced by incorporation of phosphorylation competition peptides conjugated with cell-penetrating peptides. Cell-based functional screening against 18 selected phosphorylation sites identified three phosphorylation sites (Ser-98, Ser-179 of Ldb3, and Ser-1146 of palladin) displaying near-complete inhibition of cardiac hypertrophic growth of NRVMs. Changes in phosphorylation levels of Ser-98 and Ser-179 in Ldb3 were further confirmed in NRVMs and other pathological/physiological hypertrophy models, including transverse aortic constriction and swimming models, using site-specific phospho-antibodies. Our integrated approach can be used to identify functionally important phosphorylation sites among differentially phosphorylated sites, and unlike conventional approaches, it is easily applicable for large-scale and/or high-throughput analyses.
Collapse
Affiliation(s)
- Hye Kyeong Kwon
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Hyunwoo Choi
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Sung-Gyoo Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Woo Jin Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Do Han Kim,
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Zee-Yong Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| |
Collapse
|
27
|
Drp1-dependent mitochondrial fission in cardiovascular disease. Acta Pharmacol Sin 2021; 42:655-664. [PMID: 32913266 DOI: 10.1038/s41401-020-00518-y] [Citation(s) in RCA: 207] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/19/2020] [Indexed: 12/21/2022]
Abstract
Mitochondria are highly dynamic organelles undergoing cycles of fusion and fission to modulate their morphology, distribution, and function, which are referred as 'mitochondrial dynamics'. Dynamin-related protein 1 (Drp1) is known as the major pro-fission protein whose activity is tightly regulated to clear the damaged mitochondria via mitophagy, ensuring a strict control over the intricate process of cellular and organ dynamics in heart. Various posttranslational modifications (PTMs) of Drp1 have been identified including phosphorylation, SUMOylation, palmitoylation, ubiquitination, S-nitrosylation, and O-GlcNAcylation, which implicate a role in the regulation of mitochondrial dynamics. An intact mitochondrial homeostasis is critical for heart to fuel contractile function and cardiomyocyte metabolism, while defects in mitochondrial dynamics constitute an essential part of the pathophysiology underlying various cardiovascular diseases (CVDs). In this review, we summarize current knowledge on the critical role of Drp1 in the pathogenesis of CVDs including endothelial dysfunction, smooth muscle remodeling, cardiac hypertrophy, pulmonary arterial hypertension, myocardial ischemia-reperfusion, and myocardial infarction. We also highlight how the targeting of Drp1 could potentially contribute to CVDs treatments.
Collapse
|
28
|
Jiang SJ, Wang W. Research progress on the role of CaMKII in heart disease. Am J Transl Res 2020; 12:7625-7639. [PMID: 33437349 PMCID: PMC7791482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/21/2020] [Indexed: 06/12/2023]
Abstract
In the heart, Ca2+ participates in electrical activity and myocardial contraction, which is closely related to the generation of action potential and excitation contraction coupling (ECC) and plays an important role in various signal cascades and regulates different physiological processes. In the Ca2+ related physiological activities, CaMKII is a key downstream regulator, involving autophosphorylation and post-translational modification, and plays an important role in the excitation contraction coupling and relaxation events of cardiomyocytes. This paper reviews the relationship between CaMKII and various substances in the pathological process of myocardial apoptosis and necrosis, myocardial hypertrophy and arrhythmia, and what roles it plays in the development of disease in complex networks. This paper also introduces the drugs targeting at CaMKII to treat heart disease.
Collapse
Affiliation(s)
- Shi-Jun Jiang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| | - Wei Wang
- Department of Cardiology, Affiliated Taihe Hospital of Hubei University of MedicineShiyan 442000, Hubei, China
| |
Collapse
|
29
|
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
|
30
|
Williams AL, Khadka VS, Anagaran MCT, Lee K, Avelar A, Deng Y, Shohet RV. miR-125 family regulates XIRP1 and FIH in response to myocardial infarction. Physiol Genomics 2020; 52:358-368. [PMID: 32716698 DOI: 10.1152/physiolgenomics.00041.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are powerful regulators of protein expression. Many play important roles in cardiac development and disease. While several miRNAs and targets have been well characterized, the abundance of miRNAs and the numerous potential targets for each suggest that the vast majority of these interactions have yet to be described. The goal of this study was to characterize miRNA expression in the mouse heart after coronary artery ligation (LIG) and identify novel mRNA targets altered during the initial response to ischemic stress. We performed small RNA sequencing (RNA-Seq) of ischemic heart tissue 1 day and 3 days after ligation and identified 182 differentially expressed miRNAs. We then selected relevant mRNA targets from all potential targets by correlating miRNA and mRNA expression from a corresponding RNA-Seq data set. From this analysis we chose to focus, as proof of principle, on two miRNAs from the miR-125 family, miR-125a and miR-351, and two of their potential mRNA targets, Xin actin-binding repeat-containing protein 1 (XIRP1) and factor inhibiting hypoxia-inducible factor (FIH). We found miR-125a to be less abundant and XIRP1 more abundant after ligation. In contrast, the related murine miRNA miR-351 was substantially upregulated in response to ischemic injury, and FIH expression correspondingly decreased. Luciferase reporter assays confirmed direct interactions between these miRNAs and targets. In summary, we utilized a correlative analysis strategy combining miRNA and mRNA expression data to identify functional miRNA-mRNA relationships in the heart after ligation. These findings provide insight into the response to ischemic injury and suggest future therapeutic targets.
Collapse
Affiliation(s)
- Allison Lesher Williams
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Vedbar S Khadka
- Bioinformatics Core, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Ma C T Anagaran
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Katie Lee
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Abigail Avelar
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Youping Deng
- Bioinformatics Core, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Ralph V Shohet
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| |
Collapse
|
31
|
Mapping signalling perturbations in myocardial fibrosis via the integrative phosphoproteomic profiling of tissue from diverse sources. Nat Biomed Eng 2020; 4:889-900. [PMID: 32661320 DOI: 10.1038/s41551-020-0585-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/14/2020] [Indexed: 12/13/2022]
Abstract
Study of the molecular basis of myocardial fibrosis is hampered by limited access to tissues from human patients and by confounding variables associated with sample accessibility, collection, processing and storage. Here, we report an integrative strategy based on mass spectrometry for the phosphoproteomic profiling of normal and fibrotic cardiac tissue obtained from surgical explants from patients with hypertrophic cardiomyopathy, from a transaortic-constriction mouse model of cardiac hypertrophy and fibrosis, and from a heart-on-a-chip model of cardiac fibrosis. We used the integrative approach to map the relative abundance of thousands of proteins, phosphoproteins and phosphorylation sites specific to each tissue source, to identify key signalling pathways driving fibrosis and to screen for anti-fibrotic compounds targeting glycogen synthase kinase 3, which has a consistent role as a key mediator of fibrosis in all three types of tissue specimen. The integrative disease-modelling strategy may reveal new insights into mechanisms of cardiac disease and serve as a test bed for drug screening.
Collapse
|
32
|
Mitochondrial dynamic modulation exerts cardiometabolic protection in obese insulin-resistant rats. Clin Sci (Lond) 2020; 133:2431-2447. [PMID: 31808509 DOI: 10.1042/cs20190960] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/15/2019] [Accepted: 12/06/2019] [Indexed: 12/26/2022]
Abstract
Obese insulin resistance impairs cardiac mitochondrial dynamics by increasing mitochondrial fission and decreasing mitochondrial fusion, leading to mitochondrial damage, myocardial cell death and cardiac dysfunction. Therefore, inhibiting fission and promoting fusion could provide cardioprotection in this pre-diabetic condition. We investigated the combined effects of the mitochondrial fission inhibitor (Mdivi1) and fusion promoter (M1) on cardiac function in obese insulin-resistant rats. We hypothesized that Mdivi1 and M1 protect heart against obese insulin-resistant condition, but also there will be greater improvement using Mdivi1 and M1 as a combined treatment. Wistar rats (n=56, male) were randomly assigned to a high-fat diet (HFD) and normal diet (ND) fed groups. After feeding with either ND or HFD for 12 weeks, rats in each dietary group were divided into groups to receive either the vehicle, Mdivi1 (1.2 mg/kg, i.p.), M1 (2 mg/kg, i.p.) or combined treatment for 14 days. The cardiac function, cardiac mitochondrial function, metabolic and biochemical parameters were monitored before and after the treatment. HFD rats developed obese insulin resistance which led to impaired dynamics balance and function of mitochondria, increased cardiac cell apoptosis and dysfunction. Although Mdivi1, M1 and combined treatment exerted similar cardiometabolic benefits in HFD rats, the combined therapy showed a greater reduction in mitochondrial reactive oxygen species (ROS). Mitochondrial fission inhibitor and fusion promoter exerted similar levels of cardioprotection in a pre-diabetic condition.
Collapse
|
33
|
Fan Y, Cheng Y, Li Y, Chen B, Wang Z, Wei T, Zhang H, Guo Y, Wang Q, Wei Y, Chen F, Sha J, Guo X, Wang L. Phosphoproteomic Analysis of Neonatal Regenerative Myocardium Revealed Important Roles of Checkpoint Kinase 1 via Activating Mammalian Target of Rapamycin C1/Ribosomal Protein S6 Kinase b-1 Pathway. Circulation 2020; 141:1554-1569. [PMID: 32098494 DOI: 10.1161/circulationaha.119.040747] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND In mammals, regenerative therapy after myocardial infarction is hampered by the limited regenerative capacity of adult heart, whereas a transient regenerative capacity is maintained in the neonatal heart. Systemic phosphorylation signaling analysis on ischemic neonatal myocardium might be helpful to identify key pathways involved in heart regeneration. Our aim was to define the kinase-substrate network in ischemic neonatal myocardium and to identify key pathways involved in heart regeneration after ischemic insult. METHODS Quantitative phosphoproteomics profiling was performed on infarct border zone of neonatal myocardium, and kinase-substrate network analysis revealed 11 kinases with enriched substrates and upregulated phosphorylation levels, including checkpoint kinase 1 (CHK1) kinase. The effect of CHK1 on cardiac regeneration was tested on Institute of Cancer Research CD1 neonatal and adult mice that underwent apical resection or myocardial infarction. RESULTS In vitro, CHK1 overexpression promoted whereas CHK1 knockdown blunted cardiomyocyte proliferation. In vivo, inhibition of CHK1 hindered myocardial regeneration on resection border zone in neonatal mice. In adult myocardial infarction mice, CHK1 overexpression on infarct border zone upregulated mammalian target of rapamycin C1/ribosomal protein S6 kinase b-1 pathway, promoted cardiomyocyte proliferation, and improved cardiac function. Inhibiting mammalian target of rapamycin activity by rapamycin blunted the neonatal cardiomyocyte proliferation induced by CHK1 overexpression in vitro. CONCLUSIONS Our study indicates that phosphoproteome of neonatal regenerative myocardium could help identify important signaling pathways involved in myocardial regeneration. CHK1 is found to be a key signaling responsible for neonatal regeneration. Myocardial overexpression of CHK1 could improve cardiac regeneration in adult hearts by activating the mammalian target of rapamycin C1/ribosomal protein S6 kinase b-1 pathway. Thus, CHK1 might serve as a potential novel target in myocardial repair after myocardial infarction.
Collapse
Affiliation(s)
- Yi Fan
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University (Y.F., Y.L., B.C., Z.W., T.W., Q.W., L.W.), Nanjing Medical University, China
| | - Yiwei Cheng
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University (Y.F., Y.L., B.C., Z.W., T.W., Q.W., L.W.), Nanjing Medical University, China.,State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology (Y.C., H.Z., Y.G., J.S., X.G.), Nanjing Medical University, China
| | | | - Bingrui Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University (Y.F., Y.L., B.C., Z.W., T.W., Q.W., L.W.), Nanjing Medical University, China
| | - Zimu Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University (Y.F., Y.L., B.C., Z.W., T.W., Q.W., L.W.), Nanjing Medical University, China
| | - Tianwen Wei
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University (Y.F., Y.L., B.C., Z.W., T.W., Q.W., L.W.), Nanjing Medical University, China
| | - Hao Zhang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology (Y.C., H.Z., Y.G., J.S., X.G.), Nanjing Medical University, China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology (Y.C., H.Z., Y.G., J.S., X.G.), Nanjing Medical University, China
| | - Qiming Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University (Y.F., Y.L., B.C., Z.W., T.W., Q.W., L.W.), Nanjing Medical University, China
| | - Yongyue Wei
- Department of Biostatistics, School of Public Health, China International Cooperation Center for Environment and Human Health (Y.W., F.C.), Nanjing Medical University, China
| | - Feng Chen
- Department of Biostatistics, School of Public Health, China International Cooperation Center for Environment and Human Health (Y.W., F.C.), Nanjing Medical University, China
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology (Y.C., H.Z., Y.G., J.S., X.G.), Nanjing Medical University, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology (Y.C., H.Z., Y.G., J.S., X.G.), Nanjing Medical University, China
| | - Liansheng Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University (Y.F., Y.L., B.C., Z.W., T.W., Q.W., L.W.), Nanjing Medical University, China
| |
Collapse
|
34
|
Catanzaro MP, Weiner A, Kaminaris A, Li C, Cai F, Zhao F, Kobayashi S, Kobayashi T, Huang Y, Sesaki H, Liang Q. Doxorubicin-induced cardiomyocyte death is mediated by unchecked mitochondrial fission and mitophagy. FASEB J 2019; 33:11096-11108. [PMID: 31291545 PMCID: PMC6766652 DOI: 10.1096/fj.201802663r] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 06/17/2019] [Indexed: 02/06/2023]
Abstract
Doxorubicin (Dox) is a widely used antineoplastic agent that can cause heart failure. Dox cardiotoxicity is closely associated with mitochondrial damage. Mitochondrial fission and mitophagy are quality control mechanisms that normally help maintain a pool of healthy mitochondria. However, unchecked mitochondrial fission and mitophagy may compromise the viability of cardiomyocytes, predisposing them to cell death. Here, we tested this possibility by using Dox-treated H9c2 cardiac myoblast cells expressing either the mitochondria-targeted fluorescent protein MitoDsRed or the novel dual-fluorescent mitophagy reporter mt-Rosella. Dox induced mitochondrial fragmentation as shown by reduced form factor, aspect ratio, and mean mitochondrial size. This effect was abolished by short interference RNA-mediated knockdown of dynamin-related protein 1 (DRP1), a major regulator of fission. Importantly, DRP1 knockdown decreased cell death as indicated by the reduced number of propidium iodide-positive cells and the cleavage of caspase-3 and poly (ADP-ribose) polymerase. Moreover, DRP1-deficient mice were protected from Dox-induced cardiac damage, strongly supporting a role for DRP1-dependent mitochondrial fragmentation in Dox cardiotoxicity. In addition, Dox accelerated mitophagy flux, which was attenuated by DRP1 knockdown, as assessed by the mitophagy reporter mt-Rosella, suggesting the necessity of mitochondrial fragmentation in Dox-induced mitophagy. Knockdown of parkin, a positive regulator of mitophagy, dramatically diminished Dox-induced cell death, whereas overexpression of parkin had the opposite effect. Together, these results suggested that Dox cardiotoxicity was mediated, at least in part, by the increased mitochondrial fragmentation and accelerated mitochondrial degradation by the lysosome. Strategies that limit mitochondrial fission and mitophagy in the physiologic range may help reduce Dox cardiotoxicity.-Catanzaro, M. P., Weiner, A., Kaminaris, A., Li, C., Cai, F., Zhao, F., Kobayashi, S., Kobayashi, T., Huang, Y., Sesaki, H., Liang, Q. Doxorubicin-induced cardiomyocyte death is mediated by unchecked mitochondrial fission and mitophagy.
Collapse
Affiliation(s)
- Michael P. Catanzaro
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Ashley Weiner
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Amanda Kaminaris
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Cairong Li
- Clinical Medical College, Hubei University of Science and Technology, Xianning, China
| | - Fei Cai
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
| | - Fengyi Zhao
- Department of Endocrinology, The First Affiliated Hospital, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Satoru Kobayashi
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Tamayo Kobayashi
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Yuan Huang
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Qiangrong Liang
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| |
Collapse
|
35
|
Fert-Bober J, Murray CI, Parker SJ, Van Eyk JE. Precision Profiling of the Cardiovascular Post-Translationally Modified Proteome: Where There Is a Will, There Is a Way. Circ Res 2019; 122:1221-1237. [PMID: 29700069 DOI: 10.1161/circresaha.118.310966] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There is an exponential increase in biological complexity as initial gene transcripts are spliced, translated into amino acid sequence, and post-translationally modified. Each protein can exist as multiple chemical or sequence-specific proteoforms, and each has the potential to be a critical mediator of a physiological or pathophysiological signaling cascade. Here, we provide an overview of how different proteoforms come about in biological systems and how they are most commonly measured using mass spectrometry-based proteomics and bioinformatics. Our goal is to present this information at a level accessible to every scientist interested in mass spectrometry and its application to proteome profiling. We will specifically discuss recent data linking various protein post-translational modifications to cardiovascular disease and conclude with a discussion for enablement and democratization of proteomics across the cardiovascular and scientific community. The aim is to inform and inspire the readership to explore a larger breadth of proteoform, particularity post-translational modifications, related to their particular areas of expertise in cardiovascular physiology.
Collapse
Affiliation(s)
- Justyna Fert-Bober
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Christopher I Murray
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Sarah J Parker
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA.
| | - Jennifer E Van Eyk
- From the Advanced Clinical BioSystems Research Institute, Smidt Heart Institute, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
36
|
Marrocco V, Bogomolovas J, Ehler E, Dos Remedios CG, Yu J, Gao C, Lange S. PKC and PKN in heart disease. J Mol Cell Cardiol 2019; 128:212-226. [PMID: 30742812 PMCID: PMC6408329 DOI: 10.1016/j.yjmcc.2019.01.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 12/22/2022]
Abstract
The protein kinase C (PKC) and closely related protein kinase N (PKN) families of serine/threonine protein kinases play crucial cellular roles. Both kinases belong to the AGC subfamily of protein kinases that also include the cAMP dependent protein kinase (PKA), protein kinase B (PKB/AKT), protein kinase G (PKG) and the ribosomal protein S6 kinase (S6K). Involvement of PKC family members in heart disease has been well documented over the years, as their activity and levels are mis-regulated in several pathological heart conditions, such as ischemia, diabetic cardiomyopathy, as well as hypertrophic or dilated cardiomyopathy. This review focuses on the regulation of PKCs and PKNs in different pathological heart conditions and on the influences that PKC/PKN activation has on several physiological processes. In addition, we discuss mechanisms by which PKCs and the closely related PKNs are activated and turned-off in hearts, how they regulate cardiac specific downstream targets and pathways, and how their inhibition by small molecules is explored as new therapeutic target to treat cardiomyopathies and heart failure.
Collapse
Affiliation(s)
- Valeria Marrocco
- Division of Cardiology, School of Medicine, University of California-San Diego, La Jolla, USA
| | - Julius Bogomolovas
- Division of Cardiology, School of Medicine, University of California-San Diego, La Jolla, USA; Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, School of Cardiovascular Medicine and Sciences, British Heart Foundation Research Excellence Centre, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | | | - Jiayu Yu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Gao
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, University of California-Los Angeles, Los Angeles, USA.
| | - Stephan Lange
- Division of Cardiology, School of Medicine, University of California-San Diego, La Jolla, USA; University of Gothenburg, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden.
| |
Collapse
|
37
|
Nishimura A, Shimauchi T, Tanaka T, Shimoda K, Toyama T, Kitajima N, Ishikawa T, Shindo N, Numaga-Tomita T, Yasuda S, Sato Y, Kuwahara K, Kumagai Y, Akaike T, Ide T, Ojida A, Mori Y, Nishida M. Hypoxia-induced interaction of filamin with Drp1 causes mitochondrial hyperfission-associated myocardial senescence. Sci Signal 2018; 11:11/556/eaat5185. [PMID: 30425165 DOI: 10.1126/scisignal.aat5185] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Defective mitochondrial dynamics through aberrant interactions between mitochondria and actin cytoskeleton is increasingly recognized as a key determinant of cardiac fragility after myocardial infarction (MI). Dynamin-related protein 1 (Drp1), a mitochondrial fission-accelerating factor, is activated locally at the fission site through interactions with actin. Here, we report that the actin-binding protein filamin A acted as a guanine nucleotide exchange factor for Drp1 and mediated mitochondrial fission-associated myocardial senescence in mice after MI. In peri-infarct regions characterized by mitochondrial hyperfission and associated with myocardial senescence, filamin A colocalized with Drp1 around mitochondria. Hypoxic stress induced the interaction of filamin A with the GTPase domain of Drp1 and increased Drp1 activity in an actin-binding-dependent manner in rat cardiomyocytes. Expression of the A1545T filamin mutant, which potentiates actin aggregation, promoted mitochondrial hyperfission under normoxia. Furthermore, pharmacological perturbation of the Drp1-filamin A interaction by cilnidipine suppressed mitochondrial hyperfission-associated myocardial senescence and heart failure after MI. Together, these data demonstrate that Drp1 association with filamin and the actin cytoskeleton contributes to cardiac fragility after MI and suggests a potential repurposing of cilnidipine, as well as provides a starting point for innovative Drp1 inhibitor development.
Collapse
Affiliation(s)
- Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan.,Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tsukasa Shimauchi
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Tomohiro Tanaka
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan
| | - Kakeru Shimoda
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan
| | - Takashi Toyama
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.,Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Naoyuki Kitajima
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tatsuya Ishikawa
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.,EA Pharma Co. Inc., Tokyo 104-0042, Japan
| | - Naoya Shindo
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takuro Numaga-Tomita
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan
| | - Satoshi Yasuda
- National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Yoji Sato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.,National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | | | - Yoshito Kumagai
- Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Takaaki Akaike
- Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Tomomi Ide
- Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Akio Ojida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasuo Mori
- Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Motohiro Nishida
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan. .,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan.,Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
38
|
Hasan P, Saotome M, Ikoma T, Iguchi K, Kawasaki H, Iwashita T, Hayashi H, Maekawa Y. Mitochondrial fission protein, dynamin-related protein 1, contributes to the promotion of hypertensive cardiac hypertrophy and fibrosis in Dahl-salt sensitive rats. J Mol Cell Cardiol 2018; 121:103-106. [PMID: 29981304 DOI: 10.1016/j.yjmcc.2018.07.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/27/2018] [Accepted: 07/03/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Hypertension promotes cardiac hypertrophy which finally leads to cardiac dysfunction. Although aberrant mitochondrial dynamics is known to be a relevant contributor of pathogenesis in heart disease, little is known about the relationship between mitochondrial dynamics and cardiac hypertrophy. We investigated the pathophysiological roles of Dynamin-related protein1 (Drp1, a mitochondrial fission protein) on the hypertensive cardiac hypertrophy. METHODS & RESULTS Dahl salt-sensitive rats were fed with a low-salt (0.3% NaCl) or a high-salt (8% NaCl) chow to promote hypertension with and without administration of mdivi1 (an inhibitor of Drp1: 1 mg/kg/every alternative day), and then the hypertensive cardiac hypertrophy was assessed. High-salt fed rats exhibited left ventricular hypertrophy (LVH), myocytes hypertrophy, and cardiac fibrosis, and mdivi-1 suppressed them without alteration of the blood pressure. Mdivi1 also reduced ROS production by hypertension, which subsequently suppressed the Ca2+-activated protein phosphatase calcineurin and Ca2+/calmodulin-dependent kinase II (CaMKII). CONCLUSIONS Our results suggest that Drp1 contributes to the pathogenesis of hypertensive cardiac hypertrophy via ROS production and the Drp1 suppression may be effective to prevent the hypertensive cardiac hypertrophy.
Collapse
Affiliation(s)
- Prottoy Hasan
- Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masao Saotome
- Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Takenori Ikoma
- Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Keisuke Iguchi
- Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideya Kawasaki
- Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Toshihide Iwashita
- Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideharu Hayashi
- Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuichiro Maekawa
- Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
39
|
Zhao Y, Ponnusamy M, Liu C, Tian J, Dong Y, Gao J, Wang C, Zhang Y, Zhang L, Wang K, Li P. MiR-485-5p modulates mitochondrial fission through targeting mitochondrial anchored protein ligase in cardiac hypertrophy. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2871-2881. [DOI: 10.1016/j.bbadis.2017.07.034] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 07/11/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022]
|
40
|
Sivakumar A, Subbiah R, Balakrishnan R, Rajendhran J. Cardiac mitochondrial dynamics: miR-mediated regulation during cardiac injury. J Mol Cell Cardiol 2017; 110:26-34. [PMID: 28705612 DOI: 10.1016/j.yjmcc.2017.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 07/06/2017] [Accepted: 07/07/2017] [Indexed: 12/20/2022]
Abstract
Mitochondrial integrity is indispensable for cardiac health. With the advent of modern imaging technologies, mitochondrial motility and dynamics within the cell are extensively studied. Terminally differentiated and well-structured cardiomyocytes depict little mitochondrial division and fusion, questioning the contribution of mitochondrial fusion proteins (Mitofusin 1/2 and Optic Atrophy 1 protein) and fission factors (Dynamin-like protein 1 and mitochondrial fission 1 protein) in cardiomyocyte homeostasis. Emerging evidences suggest that alterations in mitochondrial morphology from globular, elongated network to punctate fragmented disconnected structures are a pathological response to ensuing cardiac stress and cardiomyocyte cell death, bringing forth the following question, "what maintains this balance between fusion and fission?" The answer hinges upon the classical "junk" DNA: microRNAs, the endogenous non-coding RNAs. Because of their essential role in numerous signaling pathways, microRNAs are considered to play major roles in the pathogenesis of various diseases. Mitochondria are not exempted from microRNA-mediated regulation. This review defines the importance of mitochondrial structural integrity and the microRNA-mitochondrial dynamics tandem, an imminent dimension of the cardiac homeostasis network. Elucidating their coordinated interaction could spur RNA-based therapeutics for resuscitating functional mitochondrial population during cardiovascular disorders.
Collapse
Affiliation(s)
- Anusha Sivakumar
- Cardiac Hypertrophy Laboratory, Department of Molecular Biology, School of Biological Sciences, Madurai Kamaraj University, Madurai 625 021, Tamilnadu, India
| | - Ramasamy Subbiah
- Cardiac Hypertrophy Laboratory, Department of Molecular Biology, School of Biological Sciences, Madurai Kamaraj University, Madurai 625 021, Tamilnadu, India.
| | - Rekha Balakrishnan
- Cardiac Hypertrophy Laboratory, Department of Molecular Biology, School of Biological Sciences, Madurai Kamaraj University, Madurai 625 021, Tamilnadu, India
| | - Jeyaprakash Rajendhran
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai 625 021, Tamil Nadu, India
| |
Collapse
|
41
|
Yu Q, Shi X, Feng Y, Kent KC, Li L. Improving data quality and preserving HCD-generated reporter ions with EThcD for isobaric tag-based quantitative proteomics and proteome-wide PTM studies. Anal Chim Acta 2017; 968:40-49. [PMID: 28395773 PMCID: PMC5509462 DOI: 10.1016/j.aca.2017.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 02/03/2017] [Accepted: 03/02/2017] [Indexed: 11/22/2022]
Abstract
Mass spectrometry (MS)-based isobaric labeling has undergone rapid development in recent years due to its capability for high throughput quantitation. Apart from its originally designed use with collision-induced dissociation (CID) and higher-energy collisional dissociation (HCD), isobaric tagging technique could also work with electron-transfer dissociation (ETD), which provides complementarity to CID and is preferred in sequencing peptides with post-translational modifications (PTMs). However, ETD suffers from long reaction time, reduced duty cycle and bias against peptides with lower charge states. In addition, common fragmentation mechanism in ETD results in altered reporter ion production, decreased multiplexing capability, and even loss of quantitation capability for some of the isobaric tags, including custom-designed dimethyl leucine (DiLeu) tags. Here, we demonstrate a novel electron-transfer/higher-energy collision dissociation (EThcD) approach that preserves original reporter ion channels, mitigates bias against lower charge states, improves sensitivity, and significantly improves data quality for quantitative proteomics and proteome-wide PTM studies. Systematic optimization was performed to achieve a balance between data quality and sensitivity. We provide direct comparison of EThcD with ETD and HCD for DiLeu- and TMT-labeled HEK cell lysate and IMAC enriched phosphopeptides. Results demonstrate improved data quality and phosphorylation localization accuracy while preserving sufficient reporter ion production. Biological studies were performed to investigate phosphorylation changes in a mouse vascular smooth muscle cell line treated with four different conditions. Overall, EThcD exhibits superior performance compared to conventional ETD and offers distinct advantages compared to HCD in isobaric labeling based quantitative proteomics and quantitative PTM studies.
Collapse
Affiliation(s)
- Qing Yu
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Xudong Shi
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
| | - Yu Feng
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - K Craig Kent
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53706, USA.
| |
Collapse
|
42
|
Singh S, Sharma S. Dynamin-related protein-1 as potential therapeutic target in various diseases. Inflammopharmacology 2017; 25:383-392. [PMID: 28409390 DOI: 10.1007/s10787-017-0347-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/31/2017] [Indexed: 12/19/2022]
Abstract
Mitochondria can interchange morphology due to their dynamic nature. It can exist in either fragmented disconnected arrangement or elongated interconnected mitochondrial networks due to fission and fusion, respectively. The recent studies have revealed the remarkable and unexpected insights into the physiological impact and molecular regulation of mitochondrial morphology. The balance between fission and fusion governs the faith of the cell. The active targeting of DRP 1 to the outer mitochondrial membrane (OMM) is done by non-GTPase receptor proteins such as mitochondrial fission factor, mitochondrial fission protein 1 and mitochondrial elongation factor 1. The active targeting of DRP 1 to OMM leads to the fission of mitochondria. However, the imbalance of DRP 1-dependent mitochondrial fission and modulation of equilibrium of fission and fusion has been documented to be involved in several cardiovascular and neurodegenerative disorders. In this review, we are focusing on the active participation of DRP 1 in various diseases and also the factors responsible for the activation of DRP 1 for its action.
Collapse
Affiliation(s)
- Surinder Singh
- Cardiovascular Division, Department of Pharmacology, I.S.F. College of Pharmacy, Moga, 142001, Punjab, India
| | - Saurabh Sharma
- Cardiovascular Division, Department of Pharmacology, I.S.F. College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
43
|
Ong SB, Kalkhoran SB, Hernández-Reséndiz S, Samangouei P, Ong SG, Hausenloy DJ. Mitochondrial-Shaping Proteins in Cardiac Health and Disease - the Long and the Short of It! Cardiovasc Drugs Ther 2017; 31:87-107. [PMID: 28190190 PMCID: PMC5346600 DOI: 10.1007/s10557-016-6710-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondrial health is critically dependent on the ability of mitochondria to undergo changes in mitochondrial morphology, a process which is regulated by mitochondrial shaping proteins. Mitochondria undergo fission to generate fragmented discrete organelles, a process which is mediated by the mitochondrial fission proteins (Drp1, hFIS1, Mff and MiD49/51), and is required for cell division, and to remove damaged mitochondria by mitophagy. Mitochondria undergo fusion to form elongated interconnected networks, a process which is orchestrated by the mitochondrial fusion proteins (Mfn1, Mfn2 and OPA1), and which enables the replenishment of damaged mitochondrial DNA. In the adult heart, mitochondria are relatively static, are constrained in their movement, and are characteristically arranged into 3 distinct subpopulations based on their locality and function (subsarcolemmal, myofibrillar, and perinuclear). Although the mitochondria are arranged differently, emerging data supports a role for the mitochondrial shaping proteins in cardiac health and disease. Interestingly, in the adult heart, it appears that the pleiotropic effects of the mitochondrial fusion proteins, Mfn2 (endoplasmic reticulum-tethering, mitophagy) and OPA1 (cristae remodeling, regulation of apoptosis, and energy production) may play more important roles than their pro-fusion effects. In this review article, we provide an overview of the mitochondrial fusion and fission proteins in the adult heart, and highlight their roles as novel therapeutic targets for treating cardiac disease.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Siavash Beikoghli Kalkhoran
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Parisa Samangouei
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Derek John Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore. .,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore. .,The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK. .,The National Institute of Health Research, University College London Hospitals Biomedical Research Centre, London, UK.
| |
Collapse
|
44
|
Wang S, Zhang F, Zhao G, Cheng Y, Wu T, Wu B, Zhang YE. Mitochondrial PKC-ε deficiency promotes I/R-mediated myocardial injury via GSK3β-dependent mitochondrial permeability transition pore opening. J Cell Mol Med 2017; 21:2009-2021. [PMID: 28266127 PMCID: PMC5571523 DOI: 10.1111/jcmm.13121] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 01/05/2017] [Indexed: 11/29/2022] Open
Abstract
Mitochondrial fission is critically involved in cardiomyocyte apoptosis, which has been considered as one of the leading causes of ischaemia/reperfusion (I/R)‐induced myocardial injury. In our previous works, we demonstrate that aldehyde dehydrogenase‐2 (ALDH2) deficiency aggravates cardiomyocyte apoptosis and cardiac dysfunction. The aim of this study was to elucidate whether ALDH2 deficiency promotes mitochondrial injury and cardiomyocyte death in response to I/R stress and the underlying mechanism. I/R injury was induced by aortic cross‐clamping for 45 min. followed by unclamping for 24 hrs in ALDH2 knockout (ALDH2−/−) and wild‐type (WT) mice. Then myocardial infarct size, cell apoptosis and cardiac function were examined. The protein kinase C (PKC) isoform expressions and their mitochondrial translocation, the activity of dynamin‐related protein 1 (Drp1), caspase9 and caspase3 were determined by Western blot. The effects of N‐acetylcysteine (NAC) or PKC‐δ shRNA treatment on glycogen synthase kinase‐3β (GSK‐3β) activity and mitochondrial permeability transition pore (mPTP) opening were also detected. The results showed that ALDH2−/− mice exhibited increased myocardial infarct size and cardiomyocyte apoptosis, enhanced levels of cleaved caspase9, caspase3 and phosphorylated Drp1. Mitochondrial PKC‐ε translocation was lower in ALDH2−/− mice than in WT mice, and PKC‐δ was the opposite. Further data showed that mitochondrial PKC isoform ratio was regulated by cellular reactive oxygen species (ROS) level, which could be reversed by NAC pre‐treatment under I/R injury. In addition, PKC‐ε inhibition caused activation of caspase9, caspase3 and Drp1Ser616 in response to I/R stress. Importantly, expression of phosphorylated GSK‐3β (inactive form) was lower in ALDH2−/− mice than in WT mice, and both were increased by NAC pre‐treatment. I/R‐induced mitochondrial translocation of GSK‐3β was inhibited by PKC‐δ shRNA or NAC pre‐treatment. In addition, mitochondrial membrane potential (∆Ψm) was reduced in ALDH2−/− mice after I/R, which was partly reversed by the GSK‐3β inhibitor (SB216763) or PKC‐δ shRNA. Collectively, our data provide the evidence that abnormal PKC‐ε/PKC‐δ ratio promotes the activation of Drp1 signalling, caspase cascades and GSK‐3β‐dependent mPTP opening, which results in mitochondrial injury‐triggered cardiomyocyte apoptosis and myocardial dysfuction in ALDH2−/− mice following I/R stress.
Collapse
Affiliation(s)
- Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Feng Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gang Zhao
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Cheng
- Heart Centre of Zhengzhou Ninth People's Hospital, Zhengzhou, Henan, China
| | - Ting Wu
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Bing Wu
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - You-En Zhang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
45
|
Reimann L, Wiese H, Leber Y, Schwäble AN, Fricke AL, Rohland A, Knapp B, Peikert CD, Drepper F, van der Ven PFM, Radziwill G, Fürst DO, Warscheid B. Myofibrillar Z-discs Are a Protein Phosphorylation Hot Spot with Protein Kinase C (PKCα) Modulating Protein Dynamics. Mol Cell Proteomics 2016; 16:346-367. [PMID: 28028127 DOI: 10.1074/mcp.m116.065425] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Indexed: 11/06/2022] Open
Abstract
The Z-disc is a protein-rich structure critically important for the development and integrity of myofibrils, which are the contractile organelles of cross-striated muscle cells. We here used mouse C2C12 myoblast, which were differentiated into myotubes, followed by electrical pulse stimulation (EPS) to generate contracting myotubes comprising mature Z-discs. Using a quantitative proteomics approach, we found significant changes in the relative abundance of 387 proteins in myoblasts versus differentiated myotubes, reflecting the drastic phenotypic conversion of these cells during myogenesis. Interestingly, EPS of differentiated myotubes to induce Z-disc assembly and maturation resulted in increased levels of proteins involved in ATP synthesis, presumably to fulfill the higher energy demand of contracting myotubes. Because an important role of the Z-disc for signal integration and transduction was recently suggested, its precise phosphorylation landscape further warranted in-depth analysis. We therefore established, by global phosphoproteomics of EPS-treated contracting myotubes, a comprehensive site-resolved protein phosphorylation map of the Z-disc and found that it is a phosphorylation hotspot in skeletal myocytes, underscoring its functions in signaling and disease-related processes. In an illustrative fashion, we analyzed the actin-binding multiadaptor protein filamin C (FLNc), which is essential for Z-disc assembly and maintenance, and found that PKCα phosphorylation at distinct serine residues in its hinge 2 region prevents its cleavage at an adjacent tyrosine residue by calpain 1. Fluorescence recovery after photobleaching experiments indicated that this phosphorylation modulates FLNc dynamics. Moreover, FLNc lacking the cleaved Ig-like domain 24 exhibited remarkably fast kinetics and exceedingly high mobility. Our data set provides research community resource for further identification of kinase-mediated changes in myofibrillar protein interactions, kinetics, and mobility that will greatly advance our understanding of Z-disc dynamics and signaling.
Collapse
Affiliation(s)
- Lena Reimann
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Heike Wiese
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Yvonne Leber
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Anja N Schwäble
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anna L Fricke
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anne Rohland
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Bettina Knapp
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Christian D Peikert
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Friedel Drepper
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Peter F M van der Ven
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Gerald Radziwill
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,§BIOSS Centre for Biological Signalling Studies, University of Freiburg
| | - Dieter O Fürst
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Bettina Warscheid
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; .,§BIOSS Centre for Biological Signalling Studies, University of Freiburg
| |
Collapse
|
46
|
Kebir S, Orfanos Z, Schuld J, Linhart M, Lamberz C, van der Ven PFM, Schrickel J, Kirfel G, Fürst DO, Meyer R. Sarcomeric lesions and remodeling proximal to intercalated disks in overload-induced cardiac hypertrophy. Exp Cell Res 2016; 348:95-105. [PMID: 27639425 DOI: 10.1016/j.yexcr.2016.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 09/09/2016] [Accepted: 09/13/2016] [Indexed: 10/21/2022]
Abstract
Pressure overload induces cardiac remodeling involving both the contractile machinery and intercalated disks (IDs). Filamin C (FlnC) and Xin actin-binding repeat-containing proteins (XIRPs) are multi-adapters localizing in IDs of higher vertebrates. Knockout of the gene encoding Xin (Xirp1) in mice leads to a mild cardiac phenotype with ID mislocalization. In order to amplify this phenotype, we performed transverse aortic constriction (TAC) on control and Xirp1-deficient mice. TAC induced similar left ventricular hypertrophy in both genotypes, suggesting that the lack of Xin does not lead to higher susceptibility to cardiac overload. However, in both genotypes, FlnC appeared in "streaming" localizations across multiple sarcomeres proximal to the IDs, suggesting a remodeling response. Furthermore, FlnC-positive areas of remodeling, reminiscent of sarcomeric lesions previously described for skeletal muscles (but so far unreported in the heart), were also observed. These adaptations reflect a similarly strong effect of the pressure induced by TAC in both genotypes. However, 2 weeks post-operation TAC-treated knockout hearts had reduced levels of connexin43 and slightly increased incidents of ventricular tachycardia compared to their wild-type (WT) counterparts. Our findings highlight the FlnC-positive sarcomeric lesions and ID-proximal streaming as general remodeling responses in cardiac overload-induced hypertrophy.
Collapse
Affiliation(s)
- Sied Kebir
- Institute of Physiology II, University Hospital Bonn, Nussallee 11, 53115 Bonn, Germany.
| | - Zacharias Orfanos
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Julia Schuld
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Markus Linhart
- Department of Medicine-Cardiology, University of Bonn Medical Center, Sigmund-Freud-Straße 25, 53127 Bonn, Germany.
| | - Christian Lamberz
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Peter F M van der Ven
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Jan Schrickel
- Department of Medicine-Cardiology, University of Bonn Medical Center, Sigmund-Freud-Straße 25, 53127 Bonn, Germany.
| | - Gregor Kirfel
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Dieter O Fürst
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| | - Rainer Meyer
- Institute of Physiology II, University Hospital Bonn, Nussallee 11, 53115 Bonn, Germany.
| |
Collapse
|
47
|
Leber Y, Ruparelia AA, Kirfel G, van der Ven PFM, Hoffmann B, Merkel R, Bryson-Richardson RJ, Fürst DO. Filamin C is a highly dynamic protein associated with fast repair of myofibrillar microdamage. Hum Mol Genet 2016; 25:2776-2788. [PMID: 27206985 DOI: 10.1093/hmg/ddw135] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 11/12/2022] Open
Abstract
Filamin c (FLNc) is a large dimeric actin-binding protein located at premyofibrils, myofibrillar Z-discs and myofibrillar attachment sites of striated muscle cells, where it is involved in mechanical stabilization, mechanosensation and intracellular signaling. Mutations in the gene encoding FLNc give rise to skeletal muscle diseases and cardiomyopathies. Here, we demonstrate by fluorescence recovery after photobleaching that a large fraction of FLNc is highly mobile in cultured neonatal mouse cardiomyocytes and in cardiac and skeletal muscles of live transgenic zebrafish embryos. Analysis of cardiomyocytes from Xirp1 and Xirp2 deficient animals indicates that both Xin actin-binding repeat-containing proteins stabilize FLNc selectively in premyofibrils. Using a novel assay to analyze myofibrillar microdamage and subsequent repair in cultured contracting cardiomyocytes by live cell imaging, we demonstrate that repair of damaged myofibrils is achieved within only 4 h, even in the absence of de novo protein synthesis. FLNc is immediately recruited to these sarcomeric lesions together with its binding partner aciculin and precedes detectable assembly of filamentous actin and recruitment of other myofibrillar proteins. These data disclose an unprecedented degree of flexibility of the almost crystalline contractile machinery and imply FLNc as a dynamic signaling hub, rather than a primarily structural protein. Our myofibrillar damage/repair model illustrates how (cardio)myocytes are kept functional in their mechanically and metabolically strained environment. Our results help to better understand the pathomechanisms and pathophysiology of early stages of FLNc-related myofibrillar myopathy and skeletal and cardiac diseases preceding pathological protein aggregation.
Collapse
Affiliation(s)
- Yvonne Leber
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Avnika A Ruparelia
- School of Biological Sciences, Monash University, Melbourne, Victoria 3800, Australia
| | - Gregor Kirfel
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| | - Bernd Hoffmann
- Department of Biomechanics (ICS-7), Institute of Complex Systems, Forschungszentrum Jülich, D52428 Jülich, Germany and
| | - Rudolf Merkel
- Department of Biomechanics (ICS-7), Institute of Complex Systems, Forschungszentrum Jülich, D52428 Jülich, Germany and.,Department of Biomechanics, Institute for Physical and Theoretical Chemistry, University of Bonn, D53115 Bonn, Germany
| | | | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, D53121 Bonn, Germany
| |
Collapse
|
48
|
Mitochondrial fission/fusion and cardiomyopathy. Curr Opin Genet Dev 2016; 38:38-44. [PMID: 27061490 DOI: 10.1016/j.gde.2016.03.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/23/2016] [Accepted: 03/04/2016] [Indexed: 01/08/2023]
Abstract
Mitochondria are highly abundant in and essential to the beat-to-beat contractile performance of hearts. However, relatively few cardiac diseases have been attributed to primary mitochondrial dysfunction. The paucity of evidence for 'primary mitochondrial cardiac diseases' may be because such an entity does not exist. Alternately, the consequences of mitochondrial dysfunction on hearts may be so severe that long-term viability is severely impaired and affected individuals are therefore not included in standard genetic screens of adult heart disease subjects. Here, I review accumulating experimental evidence that impairing mitochondrial fission or fusion causes cardiomyopathy in otherwise normal mice, and consider how these data could motivate screening of perinatal cardiomyopathy subjects for damaging mutations of mitochondrial fission and fusion factors.
Collapse
|
49
|
Abstract
Mitochondrial dynamics, fission and fusion, were first identified in yeast with investigation in heart cells beginning only in the last 5 to 7 years. In the ensuing time, it has become evident that these processes are not only required for healthy mitochondria, but also, that derangement of these processes contributes to disease. The fission and fusion proteins have a number of functions beyond the mitochondrial dynamics. Many of these functions are related to their membrane activities, such as apoptosis. However, other functions involve other areas of the mitochondria, such as OPA1's role in maintaining cristae structure and preventing cytochrome c leak, and its essential (at least a 10 kDa fragment of OPA1) role in mtDNA replication. In heart disease, changes in expression of these important proteins can have detrimental effects on mitochondrial and cellular function.
Collapse
Affiliation(s)
- A A Knowlton
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| | - T T Liu
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| |
Collapse
|
50
|
Ong SB, Kalkhoran SB, Cabrera-Fuentes HA, Hausenloy DJ. Mitochondrial fusion and fission proteins as novel therapeutic targets for treating cardiovascular disease. Eur J Pharmacol 2015; 763:104-14. [PMID: 25987420 PMCID: PMC4784719 DOI: 10.1016/j.ejphar.2015.04.056] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 03/28/2015] [Accepted: 04/09/2015] [Indexed: 12/11/2022]
Abstract
The past decade has witnessed a number of exciting developments in the field of mitochondrial dynamics - a phenomenon in which changes in mitochondrial shape and movement impact on cellular physiology and pathology. By undergoing fusion and fission, mitochondria are able to change their morphology between elongated interconnected networks and discrete fragmented structures, respectively. The cardiac mitochondria, in particular, have garnered much interest due to their unique spatial arrangement in the adult cardiomyocyte, and the multiple roles they play in cell death and survival. In this article, we review the role of the mitochondrial fusion and fission proteins as novel therapeutic targets for treating cardiovascular disease.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Department of Clinical Sciences, Faculty of Biosciences and Medical Engineering, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | | | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; Institute of Biochemistry, Medical School, Justus-Liebig University, Giessen, Germany; Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, UK; The National Institute of Health Research University College London Hospitals Biomedical Research Centre, UK.
| |
Collapse
|