51
|
Li J, Xie J, Wang YZ, Gan YR, Wei L, Ding GW, Ding YH, Xie DX. Overexpression of lncRNA Dancr inhibits apoptosis and enhances autophagy to protect cardiomyocytes from endoplasmic reticulum stress injury via sponging microRNA-6324. Mol Med Rep 2020; 23:116. [PMID: 33300079 PMCID: PMC7723073 DOI: 10.3892/mmr.2020.11755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/19/2020] [Indexed: 12/17/2022] Open
Abstract
Endoplasmic reticulum stress (ERS) contributes to the pathogenesis of myocardial ischemia/reperfusion injury and myocardial infarction (MI). Long non-coding RNAs (lncRNAs) serve an important role in cardiovascular diseases, and lncRNA discrimination antagonizing non-protein coding RNA (Dancr) alleviates cardiomyocyte damage. microRNA (miR)-6324 was upregulated in MI model rats and was predicted to bind to Dancr. The present study aimed to investigate the role of Dancr in ERS-induced cardiomyocytes and the potential underlying mechanisms. Tunicamycin (Tm) was used to induce ERS. Cell viability, apoptosis and levels of associated proteins, ERS and autophagy in Dancr-overexpression H9C2 cells and miR-6234 mimic-transfected H9C2 cells were assessed using Cell Counting Kit-8, TUNEL staining and western blot assay, respectively. The results suggested that Dancr expression levels and cell viability were downregulated by Tm in a concentration-dependent manner compared with the control group. Tm induced apoptosis, ERS and autophagy, as indicated by an increased ratio of apoptotic cells, increased expression levels of Bax, cleaved (c)-caspase-3/9, glucose-regulated protein 78 kDa (GRP78), phosphorylated (p)-inositol-requiring enzyme-1α (IRE1α), spliced X-box-binding protein 1 (Xbp1s), IRE1α, activating transcription factor (ATF)6, ATF4, Beclin 1 and microtubule associated protein 1 light chain 3α (LC3)II/I, and decreased expression levels of Bcl-2, unspliced Xbp1 (Xbp1u) and p62 in the Tm group compared with the control group. Moreover, the results indicated that compared with the Tm + overexpression (Oe)-negative control (NC) group, the Tm + Oe-Dancr group displayed decreased apoptosis, but enhanced ERS and autophagy to restore cellular homeostasis. Compared with the Tm + Oe-NC group, the Tm + Oe-Dancr group decreased the ratio of apoptotic cells, decreased expression levels of Bax, c-caspase-3/9 and Xbp1u, and increased expression levels of Bcl-2, p-IRE1α, Xbp1s, Beclin 1 and LC3II/I. Dancr overexpression also significantly downregulated miR-6324 expression compared with Oe-NC. The dual-luciferase reporter assay further indicated an interaction between Dancr and miR-6324. In addition, miR-6324 mimic partially reversed the effects of Dancr overexpression on Tm-induced apoptosis, ERS and autophagy. In conclusion, lncRNA Dancr overexpression protected cardiomyocytes against ERS injury via sponging miR-6324, thus inhibiting apoptosis, enhancing autophagy and restoring ER homeostasis.
Collapse
Affiliation(s)
- Jiong Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jing Xie
- Department of Ultrasonic Diagnosis, The First People's Hospital of Lanzhou, Lanzhou, Gansu 730050, P.R. China
| | - Yan-Zhen Wang
- Gansu Cardiovascular Institute, Lanzhou, Gansu 730050, P.R. China
| | - Yi-Rong Gan
- Gansu Cardiovascular Institute, Lanzhou, Gansu 730050, P.R. China
| | - Ling Wei
- Outpatient Department, The First People's Hospital of Lanzhou, Lanzhou, Gansu 730050, P.R. China
| | - Guan-Waner Ding
- Medical Department, Shijiazhuang People's Medical College, Shijiazhuang, Hebei 050599, P.R. China
| | - Yan-Hong Ding
- Anesthesiology Department, The First People's Hospital of Lanzhou, Lanzhou, Gansu 730050, P.R. China
| | - Ding-Xiong Xie
- Gansu Cardiovascular Institute, Lanzhou, Gansu 730050, P.R. China
| |
Collapse
|
52
|
Haas Bueno R, Recamonde-Mendoza M. Meta-analysis of Transcriptomic Data Reveals Pathophysiological Modules Involved with Atrial Fibrillation. Mol Diagn Ther 2020; 24:737-751. [PMID: 33095430 DOI: 10.1007/s40291-020-00497-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is a complex disease and affects millions of people around the world. The biological mechanisms that are involved with AF are complex and still need to be fully elucidated. Therefore, we performed a meta-analysis of transcriptome data related to AF to explore these mechanisms aiming at more sensitive and reliable results. METHODS Ten public transcriptomic datasets were downloaded, analyzed for quality control, and individually pre-processed. Differential expression analysis was carried out for each dataset, and the results were meta-analytically aggregated using the rth ordered p value method. We analyzed the final list of differentially expressed genes through network analysis, namely topological and modularity analysis, and functional enrichment analysis. RESULTS The meta-analysis of transcriptomes resulted in 1197 differentially expressed genes, whose protein-protein interaction network presented 39 hubs-bottlenecks and four main identified functional modules. These modules were enriched for 39, 20, 64, and 10 biological pathways involved with the pathophysiology of AF, especially with the disease's structural and electrical remodeling processes. The stress of the endoplasmic reticulum, protein catabolism, oxidative stress, and inflammation are some of the enriched processes. Among hub-bottlenecks genes, which are highly connected and probably have a key role in regulating these processes, HSPA5, ANK2, CTNNB1, and MAPK1 were identified. CONCLUSION Our approach based on transcriptome meta-analysis revealed a set of key genes that demonstrated consistent overall changes in expression patterns associated with AF despite data heterogeneity related, among others, to type of tissue. Further experimental investigation of our findings may shed light on the pathophysiology of the disease and contribute to the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Rodrigo Haas Bueno
- Experimental and Molecular Cardiovascular Laboratory, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil
- Bioinformatics Core, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil
| | - Mariana Recamonde-Mendoza
- Experimental and Molecular Cardiovascular Laboratory, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil.
- Bioinformatics Core, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil.
- Institute of Informatics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
53
|
Kaur N, Raja R, Ruiz-Velasco A, Liu W. Cellular Protein Quality Control in Diabetic Cardiomyopathy: From Bench to Bedside. Front Cardiovasc Med 2020; 7:585309. [PMID: 33195472 PMCID: PMC7593653 DOI: 10.3389/fcvm.2020.585309] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022] Open
Abstract
Heart failure is a serious comorbidity and the most common cause of mortality in diabetes patients. Diabetic cardiomyopathy (DCM) features impaired cellular structure and function, culminating in heart failure; however, there is a dearth of specific clinical therapy for treating DCM. Protein homeostasis is pivotal for the maintenance of cellular viability under physiological and pathological conditions, particularly in the irreplaceable cardiomyocytes; therefore, it is tightly regulated by a protein quality control (PQC) system. Three evolutionarily conserved molecular processes, the unfolded protein response (UPR), the ubiquitin-proteasome system (UPS), and autophagy, enhance protein turnover and preserve protein homeostasis by suppressing protein translation, degrading misfolded or unfolded proteins in cytosol or organelles, disposing of damaged and toxic proteins, recycling essential amino acids, and eliminating insoluble protein aggregates. In response to increased cellular protein demand under pathological insults, including the diabetic condition, a coordinated PQC system retains cardiac protein homeostasis and heart performance, on the contrary, inappropriate PQC function exaggerates cardiac proteotoxicity with subsequent heart dysfunction. Further investigation of the PQC mechanisms in diabetes propels a more comprehensive understanding of the molecular pathogenesis of DCM and opens new prospective treatment strategies for heart disease and heart failure in diabetes patients. In this review, the function and regulation of cardiac PQC machinery in diabetes mellitus, and the therapeutic potential for the diabetic heart are discussed.
Collapse
Affiliation(s)
- Namrita Kaur
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Rida Raja
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Andrea Ruiz-Velasco
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Wei Liu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
54
|
Liu Y, Zhao Y, Tang R, Jiang X, Wang Y, Gu T. Effect of TFAM on ATP content in tachypacing primary cultured cardiomyocytes and atrial fibrillation patients. Mol Med Rep 2020; 22:5105-5112. [PMID: 33174030 PMCID: PMC7646973 DOI: 10.3892/mmr.2020.11593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/31/2020] [Indexed: 12/16/2022] Open
Abstract
Atrial fibrillation (AF) is one of the most common types of arrhythmia worldwide; although a number of theories have been proposed to explain the mechanisms of AF, the treatment of AF is far from satisfactory. Energy metabolism is associated with the development of AF. Mitochondrial transcription factor A (TFAM) serves a role in the maintenance and transcription of mitochondrial DNA. The present study aimed to investigate the association between TFAM and AF and the effect of TFAM on ATP content in cardiomyocytes. Left atrial appendage tissues were collected from 20 patients with normal sinus rhythm (SR) and 20 patients with AF, and the expression levels of TFAM in SR and AF tissues were evaluated. In addition, a tachypacing model of primary cultured cardiomyocytes was constructed to assess ATP content, cell viability and expression levels of TFAM, mitochondrially encoded (MT)-NADH dehydrogenase 1 (ND1), MT-cytochrome c oxidase 1 (CO1), NADH ubiquinone oxidoreductase core subunit 1 (NDUFS1) and cytochrome c oxidase subunit 6C (COX6C). Finally, the effects of overexpression and inhibition of TFAM on ATP content, cell viability and the expression levels of MT-ND1 and MT-CO1 were investigated. The expression levels of TFAM were decreased in AF tissues compared with SR tissues (P<0.05). The ATP content, cell viability and expression levels of TFAM, MT-ND1 and MT-CO1 were decreased in tachypacing cardiomyocytes compared with non-pacing cardiomyocytes (P<0.05), whereas the expression levels of NDUFS1 and COX6C were not changed (P>0.05). Overexpression of TFAM increased ATP content, cell viability and expression levels of MT-ND1 and MT-CO1 (P<0.05). The inhibition of TFAM decreased ATP content, cell viability and expression levels of MT-ND1 and MT-CO1 (P<0.05). In summary, the results of the present study demonstrated that the expression levels of TFAM were decreased in AF tissues and tachypacing cardiomyocytes and that the restoration of TFAM increased the ATP content by upregulating the expression levels of MT-ND1 and MT-CO1 in tachypacing cardiomyocytes. Thus, TFAM may be a novel beneficial target for treatment of patients with AF.
Collapse
Affiliation(s)
- Yueheng Liu
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ye Zhao
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Rui Tang
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xuan Jiang
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuchao Wang
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Tianxiang Gu
- Department of Cardiac Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
55
|
Impaired Mitophagy: A New Potential Mechanism of Human Chronic Atrial Fibrillation. Cardiol Res Pract 2020; 2020:6757350. [PMID: 33062320 PMCID: PMC7547333 DOI: 10.1155/2020/6757350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/17/2020] [Accepted: 09/02/2020] [Indexed: 12/16/2022] Open
Abstract
Mitophagy is an autophagic response and plays essential roles in survival, development, and homeostasis of cells. It has been reported that mitophagic dysfunction is involved in several cardiovascular diseases. However, the effect of mitophagy on atrial fibrillation (AF) is still unknown. Therefore, we investigated the exact role of mitophagy in human chronic AF. Western blot was used to detect the protein abundance. The mitochondrial morphology and structure were observed by transmission electron microscopy. Immunofluorescent stainings were performed to analyze colocalization of mitochondria with autophagosomes or lysosomes. Totally, 43 patients with valvular heart disease undergoing cardiac surgery were selected, including 21 patients with chronic AF. Comparing with the sinus rhythm (SR) group, we found the size and number of mitochondria in atrial myocytes of patients with AF increased significantly. In addition, expression of LC3B II and LC3B II/LC3B I ratio was significantly decreased in the AF group. Moreover, the expression of p62 was markedly elevated in the AF group compared with that in the SR group. The results of immunofluorescence staining and western blot showed an enhanced expression of Cox IV in the AF group. Dual immunofluorescent stainings revealed that mitophagy defect in atrial myocytes of patients with AF resulted from dysfunction in the process of delivery of mitochondria into autophagosomes. For the first time, impaired mitophagy, during the phagocytosis of mitochondria, is associated with human chronic AF. Mitophagy could be a potential therapeutic target for AF.
Collapse
|
56
|
Wu X, Liu Z, Yu XY, Xu S, Luo J. Autophagy and cardiac diseases: Therapeutic potential of natural products. Med Res Rev 2020; 41:314-341. [PMID: 32969064 DOI: 10.1002/med.21733] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 08/28/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022]
Abstract
The global incidence of cardiac diseases is expected to increase in the coming years, imposing a substantial socioeconomic burden on healthcare systems. Autophagy is a tightly regulated lysosomal degradation mechanism important for cell survival, homeostasis, and function. Accumulating pieces of evidence have indicated a major role of autophagy in the regulation of cardiac homeostasis and function. It is well established that dysregulation of autophagy in cardiomyocytes is involved in cardiac hypertrophy, myocardial infarction, diabetic cardiomyopathy, and heart failure. In this sense, autophagy seems to be an attractive therapeutic target for cardiac diseases. Recently, multiple natural products/phytochemicals, such as resveratrol, berberine, and curcumin have been shown to regulate cardiomyocyte autophagy via different pathways. The autophagy-modifying capacity of these compounds should be taken into consideration for designing novel therapeutic agents. This review focuses on the role of autophagy in various cardiac diseases and the pharmacological basis and therapeutic potential of reported natural products in cardiac diseases by modifying autophagic processes.
Collapse
Affiliation(s)
- Xiaoqian Wu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zumei Liu
- Department of Central Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Suowen Xu
- Department of Endocrinology and Metabolism, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Jiandong Luo
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
57
|
van Marion DMS, Lanters EAH, Ramos KS, Li J, Wiersma M, Baks-te Bulte L, J. Q. M. Muskens A, Boersma E, de Groot NMS, Brundel BJJM. Evaluating Serum Heat Shock Protein Levels as Novel Biomarkers for Atrial Fibrillation. Cells 2020; 9:E2105. [PMID: 32947824 PMCID: PMC7564530 DOI: 10.3390/cells9092105] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Staging of atrial fibrillation (AF) is essential to understanding disease progression and the accompanied increase in therapy failure. Blood-based heat shock protein (HSP) levels may enable staging of AF and the identification of patients with higher risk for AF recurrence after treatment. Objective: This study evaluates the relationship between serum HSP levels, presence of AF, AF stage and AF recurrence following electrocardioversion (ECV) or pulmonary vein isolation (PVI). Methods: To determine HSP27, HSP70, cardiovascular (cv)HSP and HSP60 levels, serum samples were collected from control patients without AF and patients with paroxysmal atrial fibrillation (PAF), persistent (PeAF) and longstanding persistent (LSPeAF) AF, presenting for ECV or PVI, prior to intervention and at 3-, 6- and 12-months post-PVI. Results: The study population (n = 297) consisted of 98 control and 199 AF patients admitted for ECV (n = 98) or PVI (n = 101). HSP27, HSP70, cvHSP and HSP60 serum levels did not differ between patients without or with PAF, PeAF or LSPeAF. Additionally, baseline HSP levels did not correlate with AF recurrence after ECV or PVI. However, in AF patients with AF recurrence, HSP27 levels were significantly elevated post-PVI relative to baseline, compared to patients without recurrence. Conclusions: No association was observed between baseline HSP levels and the presence of AF, AF stage or AF recurrence. However, HSP27 levels were increased in serum samples of patients with AF recurrence within one year after PVI, suggesting that HSP27 levels may predict recurrence of AF after ablative therapy.
Collapse
Affiliation(s)
- Denise M. S. van Marion
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
| | - Eva A. H. Lanters
- Department of Cardiology, Erasmus MC, 3000CA Rotterdam, The Netherlands; (E.A.H.L.); (A.J.Q.M.M.); (E.B.); (N.M.S.d.G.)
| | - Kennedy S. Ramos
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
- Department of Cardiology, Erasmus MC, 3000CA Rotterdam, The Netherlands; (E.A.H.L.); (A.J.Q.M.M.); (E.B.); (N.M.S.d.G.)
| | - Jin Li
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
| | - Marit Wiersma
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
- Netherlands Heart Institute, 3511EP Utrecht, The Netherlands
| | - Luciënne Baks-te Bulte
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
| | - Agnes J. Q. M. Muskens
- Department of Cardiology, Erasmus MC, 3000CA Rotterdam, The Netherlands; (E.A.H.L.); (A.J.Q.M.M.); (E.B.); (N.M.S.d.G.)
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, 3000CA Rotterdam, The Netherlands; (E.A.H.L.); (A.J.Q.M.M.); (E.B.); (N.M.S.d.G.)
| | - Natasja M. S. de Groot
- Department of Cardiology, Erasmus MC, 3000CA Rotterdam, The Netherlands; (E.A.H.L.); (A.J.Q.M.M.); (E.B.); (N.M.S.d.G.)
| | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije University, 1081HV Amsterdam, The Netherlands; (D.M.S.v.M.); (K.S.R.); (J.L.); (M.W.); (L.B.-t.B.)
| |
Collapse
|
58
|
Wang Q, Zhao Y, Dong X, Li C, Zhou L, Zou C, Li X, Zhou N, Liu J, Sun Y, Wang J. The Occurrence of Valvular Atrial Fibrillation: Involvement of NGF/TrKA Signaling Pathway. J INVEST SURG 2020; 34:1379-1386. [PMID: 32781864 DOI: 10.1080/08941939.2020.1798570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Nerve growth factor (NGF) and tropomyosin kinase receptors A (TrKA) exert a crucial effect on the regulation of autonomic nervous system which contributes to the progress of atrial fibrillation (AF). Valvular heart disease (VHD) patients are more easily to induce the AF. We investigated whether NGF/TrKA could impact the occurrence of AF in VHD patients. MATERIALS AND METHODS Atrial tissues were resected from 30 VHD patients with chronic AF (n = 15, AF >6 months) or sinus rhythm (SR, n = 15). The expression of NGF, TrKA, protein kinase B (PKB/Akt), beta-isoforms of glycogen synthase kinase-3 (GSK3β), Serine473 phosphorylation of Akt (p-Ser473 Akt), Serine9 phosphorylation of GSK-3β (p-Ser9 GSK3β) in right atrial tissues and peripheral blood lymphocyte were quantified by Western blot. The localization of those genes expression was measured by immunohistochemistry. Double sandwich enzyme-linked immunosorbent assay was used to observe the trace changes of NGF-β in peripheral plasma. RESULTS Our results revealed that the NGF expression was markedly elevated in the tissue of right atrial appendage and peripheral blood lymphocytes from AF patients compared with the SR patients. But, the expression of TrKA, GSK3β, p-Akt and p-GSK3β were decreased. There was no difference about the expression of Akt from the AF patients and the SR patients. The NGF-β level in peripheral blood plasma of patients with AF and SR was not statistical difference. CONCLUSION Thus, we thought that NGF/TrKA signaling pathway may be involved in the AF in the patients with VHD, inactivation of GSK3β could increase the incidence of AF, but not relevant to phosphorylation.
Collapse
Affiliation(s)
- Qianli Wang
- Department of Geriatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China.,Cardiovascular Surgery Intensive Care Unit, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, PR China
| | - Yong Zhao
- Department of Geriatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China.,Department of Geriatric Cardiology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, PR China
| | - Xin Dong
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Cong Li
- Intensive Care Unit, Shouguang People's Hospital, Shouguang, PR China
| | - Lin Zhou
- Department of Geriatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China.,Department of Geriatric Cardiology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, PR China
| | - Chengwei Zou
- Department of Geriatric Cardiology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, PR China.,Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, PR China
| | - Xiaodong Li
- Department of Geriatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China.,Department of Geriatric Cardiology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, PR China
| | - Nannan Zhou
- Department of Geriatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China.,Department of Geriatric Cardiology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, PR China
| | - Junni Liu
- Department of Geriatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China.,Department of Geriatric Cardiology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, PR China
| | - Yuanyuan Sun
- Department of Geriatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China.,Department of Geriatric Cardiology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, PR China
| | - Jianchun Wang
- Department of Geriatric Cardiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, PR China.,Department of Geriatric Cardiology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, PR China
| |
Collapse
|
59
|
Lu F, Lu B, Zhang L, Wen J, Wang M, Zhang S, Li Q, Shu F, Sun Y, Liu N, Peng S, Zhao Y, Dong S, Zhao D, Lu F, Zhang W. Hydrogen sulphide ameliorating skeletal muscle atrophy in db/db mice via Muscle RING finger 1 S-sulfhydration. J Cell Mol Med 2020; 24:9362-9377. [PMID: 32633463 PMCID: PMC7417732 DOI: 10.1111/jcmm.15587] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 12/25/2022] Open
Abstract
Muscle atrophy occurs in many pathological states, including cancer, diabetes and sepsis, whose results primarily from accelerated protein degradation and activation of the ubiquitin‐proteasome pathway. Expression of Muscle RING finger 1 (MuRF1), an E3 ubiquitin ligase, was increased to induce the loss of muscle mass in diabetic condition. However, hydrogen sulphide (H2S) plays a crucial role in the variety of physiological functions, including antihypertension, antiproliferation and antioxidant. In this study, db/db mice and C2C12 myoblasts treated by high glucose and palmitate and oleate were chose as animal and cellular models. We explored how exogenous H2S attenuated the degradation of skeletal muscle via the modification of MuRF1 S‐sulfhydration in db/db mice. Our results show cystathionine‐r‐lyase expression, and H2S level in skeletal muscle of db/db mice was reduced. Simultaneously, exogenous H2S could alleviate ROS production and reverse expression of ER stress protein markers. Exogenous H2S could decrease the ubiquitination level of MYOM1 and MYH4 in db/db mice. In addition, exogenous H2S reduced the interaction between MuRF1 with MYOM1 and MYH4 via MuRF1 S‐sulfhydration. Based on these results, we establish that H2S prevented the degradation of skeletal muscle via MuRF1 S‐sulfhydration at the site of Cys44 in db/db mice.
Collapse
Affiliation(s)
- Fangping Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Baoling Lu
- Department of Infectious, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Linxue Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - JingChen Wen
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Mengyi Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiwu Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Qianzhu Li
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Feng Shu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yu Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shuo Peng
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yajun Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| |
Collapse
|
60
|
Abstract
Atrial fibrillation (AF), the most common progressive and age-related cardiac arrhythmia, affects millions of people worldwide. AF is associated with common risk factors, including hypertension, diabetes mellitus, and obesity, and serious complications such as stroke and heart failure. Notably, AF is progressive in nature, and because current treatment options are mainly symptomatic, they have only a moderate effect on prevention of arrhythmia progression. Hereto, there is an urgent unmet need to develop mechanistic treatments directed at root causes of AF. Recent research findings indicate a key role for inflammasomes and derailed proteostasis as root causes of AF. Here, we elaborate on the molecular mechanisms of these 2 emerging key pathways driving the pathogenesis of AF. First the role of NLRP3 (NACHT, LRR, and PYD domains-containing protein 3) inflammasome on AF pathogenesis and cardiomyocyte remodeling is discussed. Then we highlight pathways of proteostasis derailment, including exhaustion of cardioprotective heat shock proteins, disruption of cytoskeletal proteins via histone deacetylases, and the recently discovered DNA damage-induced nicotinamide adenine dinucleotide+ depletion to underlie AF. Moreover, potential interactions between the inflammasomes and proteostasis pathways are discussed and possible therapeutic targets within these pathways indicated.
Collapse
Affiliation(s)
- Na Li
- From the Department of Medicine (Cardiovascular Research) (N.L.), Baylor College of Medicine, Houston, TX.,Department of Molecular Physiology and Biophysics (N.L.), Baylor College of Medicine, Houston, TX.,Cardiovascular Research Institute (N.L.), Baylor College of Medicine, Houston, TX
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, the Netherlands (B.J.J.M.B.)
| |
Collapse
|
61
|
Cell-Free Circulating Mitochondrial DNA: A Potential Blood-Based Marker for Atrial Fibrillation. Cells 2020; 9:cells9051159. [PMID: 32397106 PMCID: PMC7290331 DOI: 10.3390/cells9051159] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 04/30/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023] Open
Abstract
Atrial fibrillation (AF), the most common, progressive tachyarrhythmia is associated with serious complications, such as stroke and heart failure. Early recognition of AF, essential to prevent disease progression and therapy failure, is hampered by the lack of accurate diagnostic serum biomarkers to identify the AF stage. As we previously showed mitochondrial dysfunction to drive experimental and human AF, we evaluated whether cell-free circulating mitochondrial DNA (cfc-mtDNA) represents a potential serum marker. Therefore, the levels of two mtDNA genes, COX3 and ND1, were measured in 84 control patients (C), 59 patients undergoing cardiac surgery without a history of AF (SR), 100 paroxysmal (PAF), 116 persistent (PeAF), and 20 longstanding-persistent (LS-PeAF) AF patients undergoing either cardiac surgery or AF treatment (electrical cardioversion or pulmonary vein isolation). Cfc-mtDNA levels were significantly increased in PAF patients undergoing AF treatment, especially in males and patients with AF recurrence after AF treatment. In PeAF and LS-PeAF, cfc-mtDNA levels gradually decreased. Importantly, cfc-mtDNA in serum may originate from cardiomyocytes, as in vitro tachypaced cardiomyocytes release mtDNA in the medium. The findings suggest that cfc-mtDNA is associated with AF stage, especially in males, and with patients at risk for AF recurrence after treatment.
Collapse
|
62
|
Correlation between increased atrial expression of genes related to fatty acid metabolism and autophagy in patients with chronic atrial fibrillation. PLoS One 2020; 15:e0224713. [PMID: 32315296 PMCID: PMC7173849 DOI: 10.1371/journal.pone.0224713] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 04/03/2020] [Indexed: 01/07/2023] Open
Abstract
Atrial metabolic disturbance contributes to the onset and development of atrial fibrillation (AF). Autophagy plays a role in maintaining the cellular energy balance. We examined whether atrial gene expressions related to fatty acid metabolism and autophagy are altered in chronic AF and whether they are related to each other. Right atrial tissue was obtained during heart surgery from 51 patients with sinus rhythm (SR, n = 38) or chronic AF (n = 13). Preoperative fasting serum free-fatty-acid levels were significantly higher in the AF patients. The atrial gene expression of fatty acid binding protein 3 (FABP3), which is involved in the cells' fatty acid uptake and intracellular fatty acid transport, was significantly increased in AF patients compared to SR patients; in the SR patients it was positively correlated with the right atrial diameter and intra-atrial electromechanical delay (EMD), parameters of structural and electrical atrial remodeling that were evaluated by an echocardiography. In contrast, the two groups' atrial contents of diacylglycerol (DAG), a toxic fatty acid metabolite, were comparable. Importantly, the atrial gene expression of microtubule-associated protein light chain 3 (LC3) was significantly increased in AF patients, and autophagy-related genes including LC3 were positively correlated with the atrial expression of FABP3. In conclusion, in chronic AF patients, the atrial expression of FABP3 was upregulated in association with autophagy-related genes without altered atrial DAG content. Our findings may support the hypothesis that dysregulated cardiac fatty acid metabolism contributes to the progression of AF and induction of autophagy has a cardioprotective effect against cardiac lipotoxicity in chronic AF.
Collapse
|
63
|
Starreveld R, Knops P, Ramos KS, Roos-Serote MC, Bogers AJJC, Brundel BJJM, de Groot NMS. Atrial fibrillation fingerprinting; spotting bio-electrical markers to early recognize atrial fibrillation by the use of a bottom-up approach (AFFIP): Rationale and design. Clin Cardiol 2020; 43:546-552. [PMID: 32304106 PMCID: PMC7298972 DOI: 10.1002/clc.23370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 12/23/2022] Open
Abstract
Background The exact pathophysiology of atrial fibrillation (AF) remains incompletely understood and treatment of AF is associated with high recurrence rates. Persistence of AF is rooted in the presence of electropathology, defined as complex electrical conduction disorders caused by structural damage of atrial tissue. The atrial fibrillation fingerprinting (AFFIP) study aims to characterize electropathology, enabling development of a novel diagnostic instrument to predict AF onset and early progression. Hypotheses History of AF, development of post‐operative AF, age, gender, underlying heart disease, and other clinical characteristics impact the degree of electropathology. Methods This study is a prospective observational study with a planned duration of 48 months. Three study groups are defined: (1) patients with (longstanding) persistent AF, (2) patients with paroxysmal AF, and (3) patients without a history of AF, all undergoing open‐chest cardiac surgery. Intra‐operative high‐resolution epicardial mapping is performed to identify the patient‐specific electrical profile, whereas the patient‐specific biological profile is assessed by evaluating proteostasis markers in blood samples and atrial appendage tissue samples. Post‐operative continuous rhythm monitoring is performed for detection of early post‐operative AF. Late post‐operative AF (during 5‐year follow‐up) is documented by either electrocardiogram or 24‐hour Holter registration. Results The required sample size for this study is estimated at 447 patients. Up till now, 105 patients were included, of whom 36 have a history of AF. Conclusion The AFFIP study will elucidate whether electrophysiological and structural characteristics represent a novel diagnostic tool, the AF fingerprint, to predict onset and early progression of AF in cardiac surgery patients.
Collapse
Affiliation(s)
- Roeliene Starreveld
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Paul Knops
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Kennedy S Ramos
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, UK
| | | | - Ad J J C Bogers
- Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, UK
| | | |
Collapse
|
64
|
The Role of Proteostasis in the Regulation of Cardiac Intercellular Communication. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:279-302. [DOI: 10.1007/978-3-030-38266-7_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
65
|
van Gorp PRR, Trines SA, Pijnappels DA, de Vries AAF. Multicellular In vitro Models of Cardiac Arrhythmias: Focus on Atrial Fibrillation. Front Cardiovasc Med 2020; 7:43. [PMID: 32296716 PMCID: PMC7138102 DOI: 10.3389/fcvm.2020.00043] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia in clinical practice with a large socioeconomic impact due to its associated morbidity, mortality, reduction in quality of life and health care costs. Currently, antiarrhythmic drug therapy is the first line of treatment for most symptomatic AF patients, despite its limited efficacy, the risk of inducing potentially life-threating ventricular tachyarrhythmias as well as other side effects. Alternative, in-hospital treatment modalities consisting of electrical cardioversion and invasive catheter ablation improve patients' symptoms, but often have to be repeated and are still associated with serious complications and only suitable for specific subgroups of AF patients. The development and progression of AF generally results from the interplay of multiple disease pathways and is accompanied by structural and functional (e.g., electrical) tissue remodeling. Rational development of novel treatment modalities for AF, with its many different etiologies, requires a comprehensive insight into the complex pathophysiological mechanisms. Monolayers of atrial cells represent a simplified surrogate of atrial tissue well-suited to investigate atrial arrhythmia mechanisms, since they can easily be used in a standardized, systematic and controllable manner to study the role of specific pathways and processes in the genesis, perpetuation and termination of atrial arrhythmias. In this review, we provide an overview of the currently available two- and three-dimensional multicellular in vitro systems for investigating the initiation, maintenance and termination of atrial arrhythmias and AF. This encompasses cultures of primary (animal-derived) atrial cardiomyocytes (CMs), pluripotent stem cell-derived atrial-like CMs and (conditionally) immortalized atrial CMs. The strengths and weaknesses of each of these model systems for studying atrial arrhythmias will be discussed as well as their implications for future studies.
Collapse
Affiliation(s)
| | | | | | - Antoine A. F. de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
66
|
Role of HDACs in cardiac electropathology: Therapeutic implications for atrial fibrillation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118459. [DOI: 10.1016/j.bbamcr.2019.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 12/21/2022]
|
67
|
Liu C, Zhong G, Zhou Y, Yang Y, Tan Y, Li Y, Gao X, Sun W, Li J, Jin X, Cao D, Yuan X, Liu Z, Liang S, Li Y, Du R, Zhao Y, Xue J, Zhao D, Song J, Ling S, Li Y. Alteration of calcium signalling in cardiomyocyte induced by simulated microgravity and hypergravity. Cell Prolif 2020; 53:e12783. [PMID: 32101357 PMCID: PMC7106961 DOI: 10.1111/cpr.12783] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/21/2020] [Accepted: 02/02/2020] [Indexed: 12/14/2022] Open
Abstract
Objectives Cardiac Ca2+ signalling plays an essential role in regulating excitation‐contraction coupling and cardiac remodelling. However, the response of cardiomyocytes to simulated microgravity and hypergravity and the effects on Ca2+ signalling remain unknown. Here, we elucidate the mechanisms underlying the proliferation and remodelling of HL‐1 cardiomyocytes subjected to rotation‐simulated microgravity and 4G hypergravity. Materials and Methods The cardiomyocyte cell line HL‐1 was used in this study. A clinostat and centrifuge were used to study the effects of microgravity and hypergravity, respectively, on cells. Calcium signalling was detected with laser scanning confocal microscopy. Protein and mRNA levels were detected by Western blotting and real‐time PCR, respectively. Wheat germ agglutinin (WGA) staining was used to analyse cell size. Results Our data showed that spontaneous calcium oscillations and cytosolic calcium concentration are both increased in HL‐1 cells after simulated microgravity and 4G hypergravity. Increased cytosolic calcium leads to activation of calmodulin‐dependent protein kinase II/histone deacetylase 4 (CaMKII/HDAC4) signalling and upregulation of the foetal genes ANP and BNP, indicating cardiac remodelling. WGA staining indicated that cell size was decreased following rotation‐simulated microgravity and increased following 4G hypergravity. Moreover, HL‐1 cell proliferation was increased significantly under hypergravity but not rotation‐simulated microgravity. Conclusions Our study demonstrates for the first time that Ca2+/CaMKII/HDAC4 signalling plays a pivotal role in myocardial remodelling under rotation‐simulated microgravity and hypergravity.
Collapse
Affiliation(s)
- Caizhi Liu
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Guohui Zhong
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | | | | | - Yingjun Tan
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yuheng Li
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xingcheng Gao
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Weijia Sun
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Jianwei Li
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoyan Jin
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Dengchao Cao
- State Key Laboratory of Agrobiotechnology, College of Life Sciences, China Agricultural University, Beijing, China
| | - Xinxin Yuan
- State Key Laboratory of Agrobiotechnology, College of Life Sciences, China Agricultural University, Beijing, China
| | - Zizhong Liu
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Shuai Liang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Youyou Li
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Ruikai Du
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yinlong Zhao
- Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Jianqi Xue
- Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Dingsheng Zhao
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Jinping Song
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Shukuan Ling
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yingxian Li
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
68
|
SIRT1 Protects the Heart from ER Stress-Induced Injury by Promoting eEF2K/eEF2-Dependent Autophagy. Cells 2020; 9:cells9020426. [PMID: 32059483 PMCID: PMC7072417 DOI: 10.3390/cells9020426] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/11/2022] Open
Abstract
Many recent studies have demonstrated the involvement of endoplasmic reticulum (ER) stress in the development of cardiac diseases and have suggested that modulation of ER stress response could be cardioprotective. Previously, we demonstrated that the deacetylase Sirtuin 1 (SIRT1) attenuates ER stress response and promotes cardiomyocyte survival. Here, we investigated whether and how autophagy plays a role in SIRT1-afforded cardioprotection against ER stress. The results revealed that protective autophagy was initiated before cell death in response to tunicamycin (TN)-induced ER stress in cardiac cells. SIRT1 inhibition decreased ER stress-induced autophagy, whereas its activation enhanced autophagy. In response to TN- or isoproterenol-induced ER stress, mice deficient for SIRT1 exhibited suppressed autophagy along with exacerbated cardiac dysfunction. At the molecular level, we found that in response to ER stress (i) the extinction of eEF2 or its kinase eEF2K not only reduced autophagy but further activated cell death, (ii) inhibition of SIRT1 inhibited the phosphorylation of eEF2, (iii) eIF2α co-immunoprecipitated with eEF2K, and (iv) knockdown of eIF2α reduced the phosphorylation of eEF2. Our results indicate that in response to ER stress, SIRT1 activation promotes cardiomyocyte survival by enhancing autophagy at least through activation of the eEF2K/eEF2 pathway.
Collapse
|
69
|
Tang X, Li PH, Chen HZ. Cardiomyocyte Senescence and Cellular Communications Within Myocardial Microenvironments. Front Endocrinol (Lausanne) 2020; 11:280. [PMID: 32508749 PMCID: PMC7253644 DOI: 10.3389/fendo.2020.00280] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/15/2020] [Indexed: 01/10/2023] Open
Abstract
Cardiovascular diseases have become the leading cause of human death. Aging is an independent risk factor for cardiovascular diseases. Cardiac aging is associated with maladaptation of cellular metabolism, dysfunction (or senescence) of cardiomyocytes, a decrease in angiogenesis, and an increase in tissue scarring (fibrosis). These events eventually lead to cardiac remodeling and failure. Senescent cardiomyocytes show the hallmarks of DNA damage, endoplasmic reticulum stress, mitochondria dysfunction, contractile dysfunction, hypertrophic growth, and senescence-associated secreting phenotype (SASP). Metabolism within cardiomyocytes is essential not only to fuel the pump function of the heart but also to maintain the functional homeostasis and participate in the senescence of cardiomyocytes. The senescence of cardiomyocyte is also regulated by the non-myocytes (endothelial cells, fibroblasts, and immune cells) in the local microenvironment. On the other hand, the senescent cardiomyocytes alter their phenotypes and subsequently affect the non-myocytes in the local microenvironment and contribute to cardiac aging and pathological remodeling. In this review, we first summarized the hallmarks of the senescence of cardiomyocytes. Then, we discussed the metabolic switch within senescent cardiomyocytes and provided a discussion of the cellular communications between dysfunctional cardiomyocytes and non-myocytes in the local microenvironment. We also addressed the functions of metabolic regulators within non-myocytes in modulating myocardial microenvironment. Finally, we pointed out some interesting and important questions that are needed to be addressed by further studies.
Collapse
Affiliation(s)
- Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xiaoqiang Tang ;
| | - Pei-Heng Li
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Hou-Zao Chen ;
| |
Collapse
|
70
|
Li J, Zhang D, Brundel BJJM, Wiersma M. Imbalance of ER and Mitochondria Interactions: Prelude to Cardiac Ageing and Disease? Cells 2019; 8:cells8121617. [PMID: 31842269 PMCID: PMC6952992 DOI: 10.3390/cells8121617] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022] Open
Abstract
Cardiac disease is still the leading cause of morbidity and mortality worldwide, despite some exciting and innovative improvements in clinical management. In particular, atrial fibrillation (AF) and heart failure show a steep increase in incidence and healthcare costs due to the ageing population. Although research revealed novel insights in pathways driving cardiac disease, the exact underlying mechanisms have not been uncovered so far. Emerging evidence indicates that derailed proteostasis (i.e., the homeostasis of protein expression, function and clearance) is a central component driving cardiac disease. Within proteostasis derailment, key roles for endoplasmic reticulum (ER) and mitochondrial stress have been uncovered. Here, we describe the concept of ER and mitochondrial stress and the role of interactions between the ER and mitochondria, discuss how imbalance in the interactions fuels cardiac ageing and cardiac disease (including AF), and finally assess the potential of drugs directed at conserving the interaction as an innovative therapeutic target to improve cardiac function.
Collapse
Affiliation(s)
- Jin Li
- Correspondence: (J.L.); (M.W.)
| | | | | | | |
Collapse
|
71
|
Mitochondrial Dysfunction Underlies Cardiomyocyte Remodeling in Experimental and Clinical Atrial Fibrillation. Cells 2019; 8:cells8101202. [PMID: 31590355 PMCID: PMC6829298 DOI: 10.3390/cells8101202] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/21/2022] Open
Abstract
Atrial fibrillation (AF), the most common progressive tachyarrhythmia, results in structural remodeling which impairs electrical activation of the atria, rendering them increasingly permissive to the arrhythmia. Previously, we reported on endoplasmic reticulum stress and NAD+ depletion in AF, suggesting a role for mitochondrial dysfunction in AF progression. Here, we examined mitochondrial function in experimental model systems for AF (tachypaced HL-1 atrial cardiomyocytes and Drosophila melanogaster) and validated findings in clinical AF. Tachypacing of HL-1 cardiomyocytes progressively induces mitochondrial dysfunction, evidenced by impairment of mitochondrial Ca2+-handling, upregulation of mitochondrial stress chaperones and a decrease in the mitochondrial membrane potential, respiration and ATP production. Atrial biopsies from AF patients display mitochondrial dysfunction, evidenced by aberrant ATP levels, upregulation of a mitochondrial stress chaperone and fragmentation of the mitochondrial network. The pathophysiological role of mitochondrial dysfunction is substantiated by the attenuation of AF remodeling by preventing an increased mitochondrial Ca2+-influx through partial blocking or downregulation of the mitochondrial calcium uniporter, and by SS31, a compound that improves bioenergetics in mitochondria. Together, these results show that conservation of the mitochondrial function protects against tachypacing-induced cardiomyocyte remodeling and identify this organelle as a potential novel therapeutic target.
Collapse
|
72
|
Liu Y, Bai F, Liu N, Ouyang F, Liu Q. The Warburg effect: A new insight into atrial fibrillation. Clin Chim Acta 2019; 499:4-12. [PMID: 31473195 DOI: 10.1016/j.cca.2019.08.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/23/2019] [Accepted: 08/28/2019] [Indexed: 12/28/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia. Atrial remodeling, including electrical/structural/autonomic remodeling, plays a vital role in AF pathogenesis. All of these have been shown to contribute continuously to the self-perpetuating nature of AF. The Warburg effect was found to play important roles in tumor and non-tumor disease. Recently, lots of studies documented altered atrial metabolism in AF, but the specific mechanism and the impact of these changes upon AF initiation/progression remain unclear. In this article, we review the metabolic consideration in AF comprehensively and observe the footprints of the Warburg effect. We also summarize the signaling pathway involved in the Warburg effect during AF-HIF-1α and AMPK, and discuss their potential roles in AF maintenance and progression. In conclusion, we give the innovative idea that the Warburg effect exists in AF and promotes the progression of AF. Targeting it may provide new therapies for AF treatment.
Collapse
Affiliation(s)
- Yaozhong Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Hunan Province, China
| | - Fan Bai
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Hunan Province, China
| | - Na Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Hunan Province, China
| | - Feifan Ouyang
- Department of Cardiology, Asklepios-Klinik St Georg, Hamburg, Germany
| | - Qiming Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Hunan Province, China.
| |
Collapse
|
73
|
Heat shock protein inducer GGA*-59 reverses contractile and structural remodeling via restoration of the microtubule network in experimental Atrial Fibrillation. J Mol Cell Cardiol 2019; 134:86-97. [PMID: 31302117 DOI: 10.1016/j.yjmcc.2019.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Atrial Fibrillation (AF) is the most common progressive tachyarrhythmia. AF progression is driven by abnormalities in electrical impulse formation and contractile function due to structural remodeling of cardiac tissue. Previous reports indicate that structural remodeling is rooted in derailment of protein homeostasis (proteostasis). Heat shock proteins (HSPs) play a critical role in facilitating proteostasis. Hence, the HSP-inducing compound geranylgeranylacetone (GGA) and its derivatives protect against proteostasis derailment in experimental models for AF. Whether these compounds also accelerate reversibility from structural remodeling in tachypaced cardiomyocytes is unknown. OBJECTIVE To investigate whether the potent HSP inducer GGA*-59 restores structural remodeling and contractile dysfunction in tachypaced cardiomyocytes and explore the underlying mechanisms. MATERIALS AND RESULTS HL-1 cardiomyocytes post-treated with GGA*-59 or recombinant HSPB1 (rcHSPB1) revealed increased levels of HSPB1 expression and accelerated recovery from tachypacing (TP)-induced calcium transient (CaT) loss compared to non-treated cardiomyocytes. In addition, protein levels of the microtubule protein (acetylated) α-tubulin, and contractile proteins cardiac troponin I (cTnI) and troponin T (cTnT) were reduced after TP and significantly recovered by GGA*-59 or rcHSPB1 post-treatment. The mRNA levels of α-tubulin encoding genes, but not cardiac troponin genes, were reduced upon TP and during recovery, but significantly enhanced by GGA*-59 and rcHSPB1 post-treatment. In addition, TP increased calpain activity, which remained increased during recovery and GGA*-59 post-treatment. However, HDAC6 activity, which deacetylates α-tubulin resulting in microtubule disruption, was significantly increased after TP and during recovery, but normalized to control levels by GGA*-59 or rcHSPB1 post-treatment in HL-1 cardiomyocytes. CONCLUSIONS Our results imply that the HSP inducer GGA*-59 and recombinant HSPB1 accelerate recovery from TP-induced structural remodeling and contractile dysfunction in HL-1 cardiomyocytes. GGA*-59 increases HSPB1 levels, represses HDAC6 activity and restores contractile protein and microtubule levels after TP, indicating that HSP-induction is an interesting target to accelerate recovery from AF-induced remodeling.
Collapse
|
74
|
Castañeda D, Gabani M, Choi SK, Nguyen QM, Chen C, Mapara A, Kassan A, Gonzalez AA, Ait-Aissa K, Kassan M. Targeting Autophagy in Obesity-Associated Heart Disease. Obesity (Silver Spring) 2019; 27:1050-1058. [PMID: 30938942 DOI: 10.1002/oby.22455] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/30/2019] [Indexed: 01/18/2023]
Abstract
Over the past three decades, the increasing rates of obesity have led to an alarming obesity epidemic worldwide. Obesity is associated with an increased risk of cardiovascular diseases; thus, it is essential to define the molecular mechanisms by which obesity affects heart function. Individuals with obesity and overweight have shown changes in cardiac structure and function, leading to cardiomyopathy, hypertrophy, atrial fibrillation, and arrhythmia. Autophagy is a highly conserved recycling mechanism that delivers proteins and damaged organelles to lysosomes for degradation. In the hearts of patients and mouse models with obesity, this process is impaired. Furthermore, it has been shown that autophagy flux restoration in obesity models improves cardiac function. Therefore, autophagy may play an important role in mitigating the adverse effects of obesity on the heart. Throughout this review, we will discuss the benefits of autophagy on the heart in obesity and how regulating autophagy might be a therapeutic tool to reduce the risk of obesity-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Diana Castañeda
- Department of Biological Sciences, California State University, Los Angeles, California, USA
| | - Mohanad Gabani
- Cardiovascular Division, Department of Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Soo-Kyoung Choi
- Department of Physiology, College of Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Quynh My Nguyen
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California, USA
| | - Cheng Chen
- Department of Emergency and Critical Care, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China, Shanghai
| | - Ayesha Mapara
- Department of Biology, Northeastern Illinois University, Chicago, Illinois, USA
| | - Adam Kassan
- School of Pharmacy, West Coast University, Los Angeles, California, USA
| | - Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Karima Ait-Aissa
- Cardiovascular Division, Department of Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Modar Kassan
- Cardiovascular Division, Department of Medicine, Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
75
|
|
76
|
Belmadani S, Matrougui K. Broken heart: A matter of the endoplasmic reticulum stress bad management? World J Cardiol 2019; 11:159-170. [PMID: 31367278 PMCID: PMC6658386 DOI: 10.4330/wjc.v11.i6.159] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/29/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases are the number one cause of morbidity and mortality in the United States and worldwide. The induction of the endoplasmic reticulum (ER) stress, a result of a disruption in the ER homeostasis, was found to be highly associated with cardiovascular diseases such as hypertension, diabetes, ischemic heart diseases and heart failure. This review will discuss the latest literature on the different aspects of the involvement of the ER stress in cardiovascular complications and the potential of targeting the ER stress pathways as a new therapeutic approach for cardiovascular complications.
Collapse
Affiliation(s)
- Souad Belmadani
- Department of Physiological Science, Eastern Virginia Medical School, Norfolk, VA 23501, United States
| | - Khalid Matrougui
- Department of Physiological Science, Eastern Virginia Medical School, Norfolk, VA 23501, United States
| |
Collapse
|
77
|
Sun X, Zhao D, Lu F, Peng S, Yu M, Liu N, Sun Y, Du H, Wang B, Chen J, Dong S, Lu F, Zhang W. Hydrogen sulfide regulates muscle RING finger-1 protein S-sulfhydration at Cys 44 to prevent cardiac structural damage in diabetic cardiomyopathy. Br J Pharmacol 2019; 177:836-856. [PMID: 30734268 DOI: 10.1111/bph.14601] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 12/04/2018] [Accepted: 12/19/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Hydrogen sulfide (H2 S) plays important roles as a gasotransmitter in pathologies. Increased expression of the E3 ubiquitin ligase, muscle RING finger-1 (MuRF1), may be involved in diabetic cardiomyopathy. Here we have investigated whether and how exogenous H2 S alleviates cardiac muscle degradation through modifications of MuRF1 S-sulfhydration in db/db mice. EXPERIMENTAL APPROACH Neonatal rat cardiomyocytes were treated with high glucose (40 mM), oleate (100 μM), palmitate (400 μM), and NaHS (100 μM) for 72 hr. MuRF1 was silenced with siRNA technology and mutation at Cys44 . Endoplasmic reticulum stress markers, MuRF1 expression, and ubiquitination level were measured. db/db mice were injected with NaHS (39 μmol·kg-1 ) for 20 weeks. Echocardiography, cardiac ultrastructure, cystathionine-γ-lyase, cardiac structure proteins expression, and S-sulfhydration production were measured. KEY RESULTS H2 S levels and cystathionine-γ-lyase protein expression in myocardium were decreased in db/db mice. Exogenous H2 S reversed endoplasmic reticulum stress, including impairment of the function of cardiomyocytes and structural damage in db/db mice. Exogenous H2 S could suppress the levels of myosin heavy chain 6 and myosin light chain 2 ubiquitination in cardiac tissues of db/db mice, and MuRF1 was modified by S-sulfhydration, following treatment with exogenous H2 S, to reduce the interaction between MuRF1 and myosin heavy chain 6 and myosin light chain 2. CONCLUSIONS AND IMPLICATIONS Our findings suggest that H2 S regulates MuRF1 S-sulfhydration at Cys44 to prevent myocardial degradation in the cardiac tissues of db/db mice. LINKED ARTICLES This article is part of a themed section on Hydrogen Sulfide in Biology & Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.4/issuetoc.
Collapse
Affiliation(s)
- Xiaojiao Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fangping Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shuo Peng
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Miao Yu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yu Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Haining Du
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Bingzhu Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jian Chen
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China.,Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University Ministry of Education, Harbin, China
| |
Collapse
|
78
|
Huang S, Lin Y, Liang Z, Wu Z, Chen Y, Chen C. Erythropoietin helix B surface peptide modulates miR-21/Atg12 axis to alleviates cardiomyocyte hypoxia-reoxygenation injury. Am J Transl Res 2019; 11:2422-2430. [PMID: 31105848 PMCID: PMC6511769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 03/25/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND The erythropoietin helix B surface peptide (HBSP) has been shown to have neuroprotective and repair-damaging myocardium effects similar to erythropoietin (EPO). However, the protective mechanism of HBSP on cardiomyocyte hypoxia-reoxygenation (H/R) injury is not clear. METHODS H9C2 cells were pretreated with HBSP and subjected to hypoxia/reoxygenation (H/R), changes in cell function, autophagy and apoptosis were assessed, respectively. Cells were transfected with miR-21 mimic and miR-NC, and the relative expression of miR-21 and Atg12 were detected by qRT-PCR. The target role of miR-21 and Atg12 was evaluated by dual-luciferase reporter. After transfected with si-Atg12 and si-NC, western blot was used to assess autophagy and apoptosis proteins, flow cytometry assay was used to detect apoptosis rate. RESULTS We found the expression of miR-21 was significantly down-regulated, accompanied by remarkably activated of autophagy and apoptosis in H9C2 cells during H/R injury. Pleasantly, HBSP pretreatment has a similar effect as transfection of miR-21 mimic, which is to evidently inhibit autophagy and apoptosis by up-regulating miR-21 expression. Moreover, Bioinformatics analysis and luciferase reporter assay revealed that Atg12 was directly bond to miR-21. To further understand whether Atg12 is involved in the process of miR-21 regulating autophagy, si-Atg12 and si-NC were transfected into H9C2 cell, the results showed that knockdown of Atg12 enhances the inhibition autophagy and apoptosis effect of HBSP. CONCLUSION These results demonstrate that HBSP inhibits myocardial H/R injury induced by autophagy over-activation and apoptosis via miR-21/Atg12 axis.
Collapse
Affiliation(s)
- Song Huang
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical CollegeShantou 515041, Guangdong, China
| | - Yongluan Lin
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical CollegeShantou 515041, Guangdong, China
| | - Zhanbo Liang
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical CollegeShantou 515041, Guangdong, China
| | - Zhuomin Wu
- Department of Pharmacy, The First Affiliated Hospital of Shantou University Medical CollegeShantou 515041, Guangdong, China
| | - Yequn Chen
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical CollegeShantou 515041, Guangdong, China
| | - Chang Chen
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical CollegeShantou 515041, Guangdong, China
| |
Collapse
|
79
|
Zhang D, Hu X, Li J, Liu J, Baks-Te Bulte L, Wiersma M, Malik NUA, van Marion DMS, Tolouee M, Hoogstra-Berends F, Lanters EAH, van Roon AM, de Vries AAF, Pijnappels DA, de Groot NMS, Henning RH, Brundel BJJM. DNA damage-induced PARP1 activation confers cardiomyocyte dysfunction through NAD + depletion in experimental atrial fibrillation. Nat Commun 2019; 10:1307. [PMID: 30898999 PMCID: PMC6428932 DOI: 10.1038/s41467-019-09014-2] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/28/2019] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common clinical tachyarrhythmia with a strong tendency to progress in time. AF progression is driven by derailment of protein homeostasis, which ultimately causes contractile dysfunction of the atria. Here we report that tachypacing-induced functional loss of atrial cardiomyocytes is precipitated by excessive poly(ADP)-ribose polymerase 1 (PARP1) activation in response to oxidative DNA damage. PARP1-mediated synthesis of ADP-ribose chains in turn depletes nicotinamide adenine dinucleotide (NAD+), induces further DNA damage and contractile dysfunction. Accordingly, NAD+ replenishment or PARP1 depletion precludes functional loss. Moreover, inhibition of PARP1 protects against tachypacing-induced NAD+ depletion, oxidative stress, DNA damage and contractile dysfunction in atrial cardiomyocytes and Drosophila. Consistently, cardiomyocytes of persistent AF patients show significant DNA damage, which correlates with PARP1 activity. The findings uncover a mechanism by which tachypacing impairs cardiomyocyte function and implicates PARP1 as a possible therapeutic target that may preserve cardiomyocyte function in clinical AF.
Collapse
Affiliation(s)
- Deli Zhang
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HZ, Amsterdam, The Netherlands.
| | - Xu Hu
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HZ, Amsterdam, The Netherlands
| | - Jin Li
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HZ, Amsterdam, The Netherlands
| | - Jia Liu
- Department of Cardiology, Laboratory of Experimental Cardiology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Luciënne Baks-Te Bulte
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HZ, Amsterdam, The Netherlands
| | - Marit Wiersma
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HZ, Amsterdam, The Netherlands
| | - Noor-Ul-Ann Malik
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HZ, Amsterdam, The Netherlands
| | - Denise M S van Marion
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HZ, Amsterdam, The Netherlands
| | - Marziyeh Tolouee
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Femke Hoogstra-Berends
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Eva A H Lanters
- Department of Cardiology, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Arie M van Roon
- Department of Internal Medicine, Division of Vascular Medicine, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands
| | - Antoine A F de Vries
- Department of Cardiology, Laboratory of Experimental Cardiology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Daniël A Pijnappels
- Department of Cardiology, Laboratory of Experimental Cardiology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Natasja M S de Groot
- Department of Cardiology, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HZ, Amsterdam, The Netherlands.
| |
Collapse
|
80
|
van Marion DM, Hu X, Zhang D, Hoogstra-Berends F, Seerden JPG, Loen L, Heeres A, Steen H, Henning RH, Brundel BJ. Screening of novel HSP-inducing compounds to conserve cardiomyocyte function in experimental atrial fibrillation. Drug Des Devel Ther 2019; 13:345-364. [PMID: 30705583 PMCID: PMC6342224 DOI: 10.2147/dddt.s176924] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background The heat shock protein (HSP) inducer, geranylgeranylacetone (GGA), was previously found to protect against atrial fibrillation (AF) remodeling in experimental model systems. Clinical application of GGA in AF is limited, due to low systemic concentrations owing to the hydrophobic character of GGA. Objectives To identify novel HSP-inducing compounds, with improved physicochemical properties, that prevent contractile dysfunction in experimental model systems for AF. Methods Eighty-one GGA-derivatives were synthesized and explored for their HSP-inducing properties by assessment of HSP expression in HL-1 cardiomyocytes pretreated with or without a mild heat shock (HS), followed by incubation with 10 µM GGA or GGA-derivative. Subsequently, the most potent HSP-inducers were tested for preservation of calcium transient (CaT) amplitudes or heart wall contraction in pretreated tachypaced HL-1 cardiomyocytes (with or without HSPB1 siRNA) and Drosophilas, respectively. Finally, CaT recovery in tachypaced HL-1 cardiomyocytes posttreated with GGA or protective GGA-derivatives was determined. Results Thirty GGA-derivatives significantly induced HSPA1A expression after HS, and seven showed exceeding HSPA1A expression compared to GGA. GGA and nine GGA-derivatives protected significantly from tachypacing (TP)-induced CaT loss, which was abrogated by HSPB1 suppression. GGA and four potent GGA-derivatives protected against heart wall dysfunction after TP compared to non-paced control Drosophilas. Of these compounds, GGA and three GGA-derivatives induced a significant restoration from CaT loss after TP of HL-1 cardiomyocytes. Conclusion We identified novel GGA-derivatives with improved physicochemical properties compared to GGA. GGA-derivatives, particularly GGA*-59, boost HSP expression resulting in prevention and restoration from TP-induced remodeling, substantiating their role as novel therapeutics in clinical AF.
Collapse
Affiliation(s)
- Denise Ms van Marion
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands, ;
| | - Xu Hu
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands, ;
| | - Deli Zhang
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands, ;
| | - Femke Hoogstra-Berends
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Andre Heeres
- Syncom BV, Groningen, The Netherlands.,Hanze University of Applied Sciences, Groningen, The Netherlands
| | | | - Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - Bianca Jjm Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands, ;
| |
Collapse
|
81
|
Li J, Zhang D, Wiersma M, Brundel BJJM. Role of Autophagy in Proteostasis: Friend and Foe in Cardiac Diseases. Cells 2018; 7:cells7120279. [PMID: 30572675 PMCID: PMC6316637 DOI: 10.3390/cells7120279] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/13/2018] [Accepted: 12/18/2018] [Indexed: 12/11/2022] Open
Abstract
Due to ageing of the population, the incidence of cardiovascular diseases will increase in the coming years, constituting a substantial burden on health care systems. In particular, atrial fibrillation (AF) is approaching epidemic proportions. It has been identified that the derailment of proteostasis, which is characterized by the loss of homeostasis in protein biosynthesis, folding, trafficking, and clearance by protein degradation systems such as autophagy, underlies the development of common cardiac diseases. Among various safeguards within the proteostasis system, autophagy is a vital cellular process that modulates clearance of misfolded and proteotoxic proteins from cardiomyocytes. On the other hand, excessive autophagy may result in derailment of proteostasis and therefore cardiac dysfunction. Here, we review the interplay between autophagy and proteostasis in the healthy heart, discuss the imbalance between autophagy and proteostasis during cardiac diseases, including AF, and finally explore new druggable targets which may limit cardiac disease initiation and progression.
Collapse
Affiliation(s)
- Jin Li
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands.
| | - Deli Zhang
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands.
| | - Marit Wiersma
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands.
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
82
|
Pabon MA, Manocha K, Cheung JW, Lo JC. Linking Arrhythmias and Adipocytes: Insights, Mechanisms, and Future Directions. Front Physiol 2018; 9:1752. [PMID: 30568603 PMCID: PMC6290087 DOI: 10.3389/fphys.2018.01752] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/20/2018] [Indexed: 01/14/2023] Open
Abstract
Obesity and atrial fibrillation have risen to epidemic levels worldwide and may continue to grow over the next decades. Emerging evidence suggests that obesity promotes atrial and ventricular arrhythmias. This has led to trials employing various strategies with the ultimate goal of decreasing the atrial arrhythmic burden in obese patients. The effectiveness of these interventions remains to be determined. Obesity is defined by the expansion of adipose mass, making adipocytes a prime candidate to mediate the pro-arrhythmogenic effects of obesity. The molecular mechanisms linking obesity and adipocytes to increased arrhythmogenicity in both the atria and ventricles remain poorly understood. In this focused review, we highlight areas of potential molecular interplay between adipocytes and cardiomyocytes. The effects of adipocytes may be direct, local or remote. Direct effect refers to adipocyte or fatty infiltration of the atrial and ventricular myocardium itself, possibly causing increased dispersion of normal myocardial electrical signals and fibrotic substrate of adipocytes that promote reentry or adipocytes serving as a direct source of aberrant signals. Local effects may originate from nearby adipose depots, specifically epicardial adipose tissue (EAT) and pericardial adipose tissue, which may play a role in the secretion of adipokines and chemokines that can incite inflammation given the direct contact and disrupt the conduction system. Adipocytes can also have a remote effect on the myocardium arising from their systemic secretion of adipokines, cytokines and metabolites. These factors may lead to mitochondrial dysfunction, oxidative stress, autophagy, mitophagy, autonomic dysfunction, and cardiomyocyte death to ultimately produce a pro-arrhythmogenic state. By better understanding the molecular mechanisms connecting dysfunctional adipocytes and arrhythmias, novel therapies may be developed to sever the link between obesity and arrhythmias.
Collapse
Affiliation(s)
- Maria A Pabon
- Joan and Sanford I. Weill, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Kevin Manocha
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Jim W Cheung
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - James C Lo
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States.,Metabolic Health Center, Weill Cornell Medicine, New York, NY, United States.,Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
83
|
Converse role of class I and class IIa HDACs in the progression of atrial fibrillation. J Mol Cell Cardiol 2018; 125:39-49. [PMID: 30321539 DOI: 10.1016/j.yjmcc.2018.09.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 12/25/2022]
Abstract
Atrial fibrillation (AF), the most common persistent clinical tachyarrhythmia, is associated with altered gene transcription which underlies cardiomyocyte dysfunction, AF susceptibility and progression. Recent research showed class I and class IIa histone deacetylases (HDACs) to regulate pathological and fetal gene expression, and thereby induce hypertrophy and cardiac contractile dysfunction. Whether class I and class IIa HDACs are involved in AF promotion is unknown. We aim to elucidate the role of class I and class IIa HDACs in tachypacing-induced contractile dysfunction in experimental model systems for AF and clinical AF. METHODS AND RESULTS: Class I and IIa HDACs were overexpressed in HL-1 cardiomyocytes followed by calcium transient (CaT) measurements. Overexpression of class I HDACs, HDAC1 or HDAC3, significantly reduced CaT amplitude in control normal-paced (1 Hz) cardiomyocytes, which was further reduced by tachypacing (5 Hz) in HDAC3 overexpressing cardiomyocytes. HDAC3 inhibition by shRNA or by the specific inhibitor, RGFP966, prevented contractile dysfunction in both tachypaced HL-1 cardiomyocytes and Drosophila prepupae. Conversely, overexpression of class IIa HDACs (HDAC4, HDAC5, HDAC7 or HDAC9) did not affect CaT in controls, with HDAC5 and HDAC7 overexpression even protecting against tachypacing-induced CaT loss. Notably, the protective effect of HDAC5 and HDAC7 was abolished in cardiomyocytes overexpressing a dominant negative HDAC5 or HDAC7 mutant, bearing a mutation in the binding domain for myosin enhancer factor 2 (MEF2). Furthermore, tachypacing induced phosphorylation of HDAC5 and promoted its translocation from the nucleus to cytoplasm, leading to up-regulation of MEF2-related fetal gene expression (β-MHC, BNP). In accord, boosting nuclear localization of HDAC5 by MC1568 or Go6983 attenuated CaT loss in tachypaced HL-1 cardiomyocytes and preserved contractile function in Drosophila prepupae. Findings were expanded to clinical AF. Here, patients with AF showed a significant increase in expression levels and activity of HDAC3, phosphorylated HDAC5 and fetal genes (β-MHC, BNP) in atrial tissue compared to controls in sinus rhythm. CONCLUSION: Class I and class IIa HDACs display converse roles in AF progression. Whereas overexpression of Class I HDAC3 induces cardiomyocyte dysfunction, class IIa HDAC5 overexpression reveals protective properties. Accordingly, HDAC3 inhibitors and HDAC5 nuclear boosters show protection from tachypacing-induced changes and therefore may represent interesting therapeutic options in clinical AF.
Collapse
|
84
|
Yuan Y, Zhao J, Gong Y, Wang D, Wang X, Yun F, Liu Z, Zhang S, Li W, Zhao X, Sun L, Sheng L, Pan Z, Li Y. Autophagy exacerbates electrical remodeling in atrial fibrillation by ubiquitin-dependent degradation of L-type calcium channel. Cell Death Dis 2018; 9:873. [PMID: 30158642 PMCID: PMC6115437 DOI: 10.1038/s41419-018-0860-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/16/2018] [Accepted: 06/18/2018] [Indexed: 12/20/2022]
Abstract
Autophagy, a bidirectional degradative process extensively occurring in eukaryotes, has been revealed as a potential therapeutic target for several cardiovascular diseases. However, its role in atrial fibrillation (AF) remains largely unknown. This study aimed to determine the role of autophagy in atrial electrical remodeling under AF condition. Here, we reported that autophagic flux was markedly activated in atria of persistent AF patients and rabbit model of atrial rapid pacing (RAP). We also observed that the key autophagy-related gene7 (ATG7) significantly upregulated in AF patients as well as tachypacing rabbits. Moreover, lentivirus-mediated ATG7 knockdown and overexpression in rabbits were employed to clarify the effects of autophagy on atrial electrophysiology via intracardiac operation and patch-clamp experiments. Lentivirus-mediated ATG7 knockdown or autophagy inhibitor chloroquine (CQ) restored the shortened atrial effective refractory period (AERP) and alleviated the AF vulnerability caused by tachypacing in rabbits. Conversely, ATG7 overexpression significantly promoted the incidence and persistence of AF and decreased L-type calcium channel (Cav1.2 α-subunits), along with abbreviated action potential duration (APD) and diminished L-type calcium current (ICa,L). Furthermore, the co-localization and interaction of Cav1.2 with LC3B-positive autophagosomes enhanced when autophagy was activated in atrial myocytes. Tachypacing-induced autophagic degradation of Cav1.2 required ubiquitin signal through the recruitment of ubiquitin-binding proteins RFP2 and p62, which guided Cav1.2 to autophagosomes. These findings suggest that autophagy induces atrial electrical remodeling via ubiquitin-dependent selective degradation of Cav1.2 and provide a novel and promising strategy for preventing AF development.
Collapse
Affiliation(s)
- Yue Yuan
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Jing Zhao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China.,Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, 150001, Harbin, China
| | - Yongtai Gong
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Dingyu Wang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Xiaoyu Wang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Fengxiang Yun
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Zhaorui Liu
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Song Zhang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Wenpeng Li
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Xinbo Zhao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Li Sun
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Li Sheng
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China
| | - Zhenwei Pan
- Department of Pharmacology, Harbin Medical University, 150081, Harbin, China
| | - Yue Li
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, 150001, Harbin, China. .,Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, 150001, Harbin, China. .,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, 150081, Harbin, China.
| |
Collapse
|
85
|
Laredo M, Waldmann V, Khairy P, Nattel S. Age as a Critical Determinant of Atrial Fibrillation: A Two-sided Relationship. Can J Cardiol 2018; 34:1396-1406. [PMID: 30404745 DOI: 10.1016/j.cjca.2018.08.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/23/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022] Open
Abstract
The incidence of atrial fibrillation (AF), the most common sustained arrhythmia and a major public health burden, increases exponentially with age. However, mechanisms underlying this long-recognized association remain incompletely understood. Experimental and human studies have demonstrated the involvement of aging in several arrhythmogenic processes, including atrial electrical and structural remodelling, disturbed calcium homeostasis, and enhanced atrial ectopic activity/increased vulnerability to re-entry induction. Given this wide range of putative mechanisms, the task of delineating the specific effects of aging responsible for AF promotion is not simple, as aging is itself associated with increasing prevalence of a host of AF-predisposing conditions, including heart failure, coronary artery disease, and hypertension. Although we usually think of old age promoting AF, there is also evidence that young age may actually have a protective effect against AF occurrence. For example, the low AF incidence among populations of young patients with significant structural congenital heart disease and substantial atrial enlargement/remodelling suggests that younger age might protect against fibrillation in the diseased atrium; efforts at understating how younger age may prevent AF might be helpful in elucidating missing mechanistic links between AF and age. The goal of this paper is to review the epidemiologic and pathophysiologic evidence regarding mechanisms underlying age-related AF. Although the therapeutic options for AF have recently improved, major gaps still remain and a better understanding of the special relationship between age and AF may be important for the identification of new targets for therapeutic innovation.
Collapse
Affiliation(s)
- Mikael Laredo
- Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada; AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Institut de Cardiologie, Paris, France
| | - Victor Waldmann
- Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada
| | - Paul Khairy
- Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada.
| | - Stanley Nattel
- Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Québec, Canada; Institute of Pharmacology, University Duisburg-Essen, Essen, Germany; LIRYC Center, Bordeaux, France.
| |
Collapse
|
86
|
Effect of Endoplasmic Reticulum Stress and Autophagy in the Regulation of Post-infarct Cardiac Repair. Arch Med Res 2018; 49:576-582. [PMID: 30017234 DOI: 10.1016/j.arcmed.2018.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 07/02/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Acute myocardial infarction (AMI) is reported to be accompanied by endoplasmic reticulum (ER) stress and autophagy induction. Nevertheless, the roles of ER stress and autophagy in post-infarct reparative fibrosis remain to be elucidated. AIM To investigate the effects of ER stress and autophagy on the regulation of post-infarct reparative fibrosis. METHODS The expression of GRP78 and LC3 in cardiac fibroblasts in human heart tissues obtained from patients with or without AMI was assessed by immunofluorescence. In vitro, human cardiac fibroblasts (HCFs) were stimulated by various agents, the expression of GRP78, LC3 and fibronectin in these was evaluated by immunoblot and/or immunofluorescence. RESULTS After AMI, HCFs expressed significantly higher levels of GRP78 and LC3. ER stress inducer, tunicamycin (200 ng/mL) significantly increased the level of autophagy and reduced expression of fibronectin in HCFs, both of which were reversed by 4 Phenylbutyric acid. Under the condition of ER stress, the expression of fibronectin in HCFs was regulated by different levels of autophagy. LC3 co-localized with fibronectin when stimulated HCFs with tunicamycin. CONCLUSION AMI induces ER stress in cardiac fibroblasts, down-regulating fibronectin via enhanced autophagy. These findings suggest that ER stress and autophagy may be a therapeutic target to improve prognosis of patients with AMI.
Collapse
|
87
|
Meijering RAM, Wiersma M, Zhang D, Lanters EAH, Hoogstra-Berends F, Scholma J, Diks S, Qi X, de Groot NMS, Nattel S, Henning RH, Brundel BJJM. Application of kinomic array analysis to screen for altered kinases in atrial fibrillation remodeling. Heart Rhythm 2018; 15:1708-1716. [PMID: 29902583 DOI: 10.1016/j.hrthm.2018.06.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Indexed: 12/01/2022]
Abstract
BACKGROUND Dysregulation of protein kinase-mediated signaling is an early event in many diseases, including the most common clinical cardiac arrhythmia, atrial fibrillation (AF). Kinomic profiling represents a promising technique to identify candidate kinases. OBJECTIVE In this study we used kinomic profiling to identify kinases altered in AF remodeling using atrial tissue from a canine model of AF (atrial tachypacing). METHODS Left atrial tissue obtained in a previous canine study was used for kinomic array (containing 1024 kinase pseudosubstrates) analysis. Three groups of dogs were included: nonpaced controls and atrial tachypaced dogs, which were contrasted with geranylgeranylacetone-treated dogs with AF, which are protected from AF promotion, to enhance specificity of detection of putative kinases. RESULTS While tachypacing changed activity of 50 kinases, 40 of these were prevented by geranylgeranylacetone and involved in differentiation and proliferation (SRC), contraction, metabolism, immunity, development, cell cycle (CDK4), and survival (Akt). Inhibitors of Akt (MK2206) and CDK4 (PD0332991) and overexpression of a dominant-negative CDK4 phosphorylation mutant protected against tachypacing-induced contractile dysfunction in HL-1 cardiomyocytes. Moreover, patients with AF show down- and upregulation of SRC and Akt phosphorylation, respectively, similar to findings of the kinome array. CONCLUSION Contrasting kinomic array analyses of controls and treated subjects offer a versatile tool to identify kinases altered in atrial remodeling owing to tachypacing, which include Akt, CDK4, and SRC. Ultimately, pharmacological targeting of altered kinases may offer novel therapeutic possibilities to treat clinical AF.
Collapse
Affiliation(s)
- Roelien A M Meijering
- Department of Clinical Pharmacy and Pharmacology, Groningen University Institute for Drug Exploration (GUIDE), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marit Wiersma
- Department of Clinical Pharmacy and Pharmacology, Groningen University Institute for Drug Exploration (GUIDE), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Deli Zhang
- Department of Clinical Pharmacy and Pharmacology, Groningen University Institute for Drug Exploration (GUIDE), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Eva A H Lanters
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Femke Hoogstra-Berends
- Department of Clinical Pharmacy and Pharmacology, Groningen University Institute for Drug Exploration (GUIDE), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jetse Scholma
- Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - Sander Diks
- Department of Pediatric Oncology, Beatrix Children's hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - XiaoYan Qi
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | | | - Stanley Nattel
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Robert H Henning
- Department of Clinical Pharmacy and Pharmacology, Groningen University Institute for Drug Exploration (GUIDE), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bianca J J M Brundel
- Department of Clinical Pharmacy and Pharmacology, Groningen University Institute for Drug Exploration (GUIDE), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
88
|
Ducheix S, Magré J, Cariou B, Prieur X. Chronic O-GlcNAcylation and Diabetic Cardiomyopathy: The Bitterness of Glucose. Front Endocrinol (Lausanne) 2018; 9:642. [PMID: 30420836 PMCID: PMC6215811 DOI: 10.3389/fendo.2018.00642] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/09/2018] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes (T2D) is a major risk factor for heart failure. Diabetic cardiomyopathy (DC) is characterized by diastolic dysfunction and left ventricular hypertrophy. Epidemiological data suggest that hyperglycaemia contributes to the development of DC. Several cellular pathways have been implicated in the deleterious effects of high glucose concentrations in the heart: oxidative stress, accumulation of advanced glycation end products (AGE), and chronic hexosamine biosynthetic pathway (HBP) activation. In the present review, we focus on the effect of chronic activation of the HBP on diabetic heart function. The HBP supplies N-acetylglucosamine moiety (O-GlcNAc) that is O-linked by O-GlcNAc transferase (OGT) to proteins on serine or threonine residues. This post-translational protein modification modulates the activity of the targeted proteins. In the heart, acute activation of the HBP in response to ischaemia-reperfusion injury appears to be protective. Conversely, chronic activation of the HBP in the diabetic heart affects Ca2+ handling, contractile properties, and mitochondrial function and promotes stress signaling, such as left ventricular hypertrophy and endoplasmic reticulum stress. Many studies have shown that O-GlcNAc impairs the function of key protein targets involved in these pathways, such as phospholamban, calmodulin kinase II, troponin I, and FOXO1. The data show that excessive O-GlcNAcylation is a major trigger of the glucotoxic events that affect heart function under chronic hyperglycaemia. Supporting this finding, pharmacological or genetic inhibition of the HBP in the diabetic heart improves heart function. In addition, the SGLT2 inhibitor dapagliflozin, a glucose lowering agent, has recently been shown to lower cardiac HBP in a lipodystophic T2D mice model and to concomitantly improve the diastolic dysfunction of these mice. Therefore, targeting cardiac-excessive O-GlcNAcylation or specific target proteins represents a potential therapeutic option to treat glucotoxicity in the diabetic heart.
Collapse
Affiliation(s)
- Simon Ducheix
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Jocelyne Magré
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Bertrand Cariou
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Xavier Prieur
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
- *Correspondence: Xavier Prieur
| |
Collapse
|
89
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia despite substantial efforts to understand the pathophysiology of the condition and develop improved treatments. Identifying the underlying causative mechanisms of AF in individual patients is difficult and the efficacy of current therapies is suboptimal. Consequently, the incidence of AF is steadily rising and there is a pressing need for novel therapies. Research has revealed that defects in specific molecular pathways underlie AF pathogenesis, resulting in electrical conduction disorders that drive AF. The severity of this so-called electropathology correlates with the stage of AF disease progression and determines the response to AF treatment. Therefore, unravelling the molecular mechanisms underlying electropathology is expected to fuel the development of innovative personalized diagnostic tools and mechanism-based therapies. Moreover, the co-creation of AF studies with patients to implement novel diagnostic tools and therapies is a prerequisite for successful personalized AF management. Currently, various treatment modalities targeting AF-related electropathology, including lifestyle changes, pharmaceutical and nutraceutical therapy, substrate-based ablative therapy, and neuromodulation, are available to maintain sinus rhythm and might offer a novel holistic strategy to treat AF.
Collapse
Affiliation(s)
- Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam University Medical Centers, VU Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,
| | - Xun Ai
- Department of Physiology and Cell Biology, College of Medicine/Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | | | - Myrthe F. Kuipers
- AFIPonline.org, Atrial Fibrillation Innovation Platform, Amsterdam, Netherlands
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|