1
|
Fan L, Sun W, Lyu Y, Ju F, Sun W, Chen J, Ma H, Yang S, Zhou X, Wu N, Yi W, Chen E, Villaseñor R, Baubec T, Yan J. Chrom-seq identifies RNAs at chromatin marks. SCIENCE ADVANCES 2024; 10:eadn1397. [PMID: 39083617 PMCID: PMC11290522 DOI: 10.1126/sciadv.adn1397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024]
Abstract
Chromatin marks are associated with transcriptional regulatory activities. However, very few lncRNAs have been characterized with the role in regulating epigenetic marks, largely due to the technical difficulty in identifying chromatin-associating RNA. Current methods are largely limited by the availability of ChIP-grade antibody and the crosslinking, which generates high noise. Here, we developed a method termed Chrom-seq to efficiently capture RNAs associated with various chromatin marks in living cells. Chrom-seq jointly applies highly specific chromatin mark reader with APEX2, which catalyzes the oxidation of biotin-aniline to label the adjacent RNAs for isolation by streptavidin-coated beads. Using the readers of mCBX7/dPC, mCBX1, and mTAF3, we detected RNA species significantly associated with H3K27me3, H3K9me3, and H3K4me3, respectively. We demonstrated that Chrom-seq outperformed other equivalent methods in terms of sensitivity, efficiency, and cost of practice. It provides an antibody-free approach to systematically map RNAs at chromatin marks with potential regulatory roles in epigenetic events.
Collapse
Affiliation(s)
- Ligang Fan
- Ministry of Education Key Laboratory of Resource Biology and Biotechnology in Western China; Shaanxi Provincial Key Laboratory of Biotechnology; School of Medicine, Northwest University, Xi’an, China
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences; The Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, China
| | | | - Yitong Lyu
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences; The Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, China
| | - Furong Ju
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences; The Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, China
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong SAR, China
| | - Wenju Sun
- Ministry of Education Key Laboratory of Resource Biology and Biotechnology in Western China; Shaanxi Provincial Key Laboratory of Biotechnology; School of Medicine, Northwest University, Xi’an, China
| | - Jie Chen
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences; The Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, China
| | - Haiqian Ma
- Ministry of Education Key Laboratory of Resource Biology and Biotechnology in Western China; Shaanxi Provincial Key Laboratory of Biotechnology; School of Medicine, Northwest University, Xi’an, China
| | - Shifei Yang
- Ministry of Education Key Laboratory of Resource Biology and Biotechnology in Western China; Shaanxi Provincial Key Laboratory of Biotechnology; School of Medicine, Northwest University, Xi’an, China
| | - Xiaomin Zhou
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences; The Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, China
| | - Nan Wu
- Ministry of Education Key Laboratory of Resource Biology and Biotechnology in Western China; Shaanxi Provincial Key Laboratory of Biotechnology; School of Medicine, Northwest University, Xi’an, China
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences; The Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, China
| | - Wenkai Yi
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences; The Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, China
| | - Erfei Chen
- Ministry of Education Key Laboratory of Resource Biology and Biotechnology in Western China; Shaanxi Provincial Key Laboratory of Biotechnology; School of Medicine, Northwest University, Xi’an, China
| | - Rodrigo Villaseñor
- Division of Molecular Biology, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tuncay Baubec
- Institute of Biodynamics and Biocomplexity, Department of Biology, Science Faculty, Utrecht University, Utrecht, the Netherlands
| | - Jian Yan
- Ministry of Education Key Laboratory of Resource Biology and Biotechnology in Western China; Shaanxi Provincial Key Laboratory of Biotechnology; School of Medicine, Northwest University, Xi’an, China
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences; The Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, China
- Department of Precision Diagnostic and Therapeutic Technology, The City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, China
| |
Collapse
|
2
|
Sun Y, Hao M, Wu H, Zhang C, Wei D, Li S, Song Z, Tao Y. Unveiling the role of CaMKII in retinal degeneration: from biological mechanism to therapeutic strategies. Cell Biosci 2024; 14:59. [PMID: 38725013 PMCID: PMC11084033 DOI: 10.1186/s13578-024-01236-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a family of broad substrate specificity serine (Ser)/threonine (Thr) protein kinases that play a crucial role in the Ca2+-dependent signaling pathways. Its significance as an intracellular Ca2+ sensor has garnered abundant research interest in the domain of neurodegeneration. Accumulating evidences suggest that CaMKII is implicated in the pathology of degenerative retinopathies such as diabetic retinopathy (DR), age-related macular degeneration (AMD), retinitis pigmentosa (RP) and glaucoma optic neuropathy. CaMKII can induce the aberrant proliferation of retinal blood vessels, influence the synaptic signaling, and exert dual effects on the survival of retinal ganglion cells and pigment epithelial cells. Researchers have put forth multiple therapeutic agents, encompassing small molecules, peptides, and nucleotides that possess the capability to modulate CaMKII activity. Due to its broad range isoforms and splice variants therapeutic strategies seek to inhibit specifically the CaMKII are confronted with considerable challenges. Therefore, it becomes crucial to discern the detrimental and advantageous aspects of CaMKII, thereby facilitating the development of efficacious treatment. In this review, we summarize recent research findings on the cellular and molecular biology of CaMKII, with special emphasis on its metabolic and regulatory mechanisms. We delve into the involvement of CaMKII in the retinal signal transduction pathways and discuss the correlation between CaMKII and calcium overload. Furthermore, we elaborate the therapeutic trials targeting CaMKII, and introduce recent developments in the zone of CaMKII inhibitors. These findings would enrich our knowledge of CaMKII, and shed light on the development of a therapeutic target for degenerative retinopathy.
Collapse
Affiliation(s)
- Yuxin Sun
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Mengyu Hao
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Hao Wu
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Chengzhi Zhang
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Dong Wei
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Siyu Li
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Zongming Song
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| | - Ye Tao
- Department of Ophthalmology, Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
3
|
Le LTT, Nhu CXT. The Role of Long Non-Coding RNAs in Cardiovascular Diseases. Int J Mol Sci 2023; 24:13805. [PMID: 37762106 PMCID: PMC10531487 DOI: 10.3390/ijms241813805] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 09/29/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are non-coding RNA molecules longer than 200 nucleotides that regulate gene expression at the transcriptional, post-transcriptional, and translational levels. Abnormal expression of lncRNAs has been identified in many human diseases. Future improvements in diagnostic, prognostic, and therapeutic techniques will be facilitated by a deeper understanding of disease etiology. Cardiovascular diseases (CVDs) are the main cause of death globally. Cardiac development involves lncRNAs, and their abnormalities are linked to many CVDs. This review examines the relationship and function of lncRNA in a variety of CVDs, including atherosclerosis, myocardial infarction, myocardial hypertrophy, and heart failure. Therein, the potential utilization of lncRNAs in clinical diagnostic, prognostic, and therapeutic applications will also be discussed.
Collapse
Affiliation(s)
- Linh T. T. Le
- Biotechnology Department, Ho Chi Minh City Open University, Ho Chi Minh City 70000, Vietnam;
| | | |
Collapse
|
4
|
Zhao H, Tan Z, Zhou J, Wu Y, Hu Q, Ling Q, Ling J, Liu M, Ma J, Zhang D, Wang Y, Zhang J, Yu P, Jiang Y, Liu X. The regulation of circRNA and lncRNAprotein binding in cardiovascular diseases: Emerging therapeutic targets. Biomed Pharmacother 2023; 165:115067. [PMID: 37392655 DOI: 10.1016/j.biopha.2023.115067] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/18/2023] [Accepted: 06/23/2023] [Indexed: 07/03/2023] Open
Abstract
Noncoding ribonucleic acids (ncRNAs) are a class of ribonucleic acids (RNAs) that carry cellular information and perform essential functions. This class encompasses various RNAs, such as small nuclear ribonucleic acids (snRNA), small interfering ribonucleic acids (siRNA) and many other kinds of RNA. Of these, circular ribonucleic acids (circRNAs) and long noncoding ribonucleic acids (lncRNAs) are two types of ncRNAs that regulate crucial physiological and pathological processes, including binding, in several organs through interactions with other RNAs or proteins. Recent studies indicate that these RNAs interact with various proteins, including protein 53, nuclear factor-kappa B, vascular endothelial growth factor, and fused in sarcoma/translocated in liposarcoma, to regulate both the histological and electrophysiological aspects of cardiac development as well as cardiovascular pathogenesis, ultimately leading to a variety of genetic heart diseases, coronary heart disease, myocardial infarction, rheumatic heart disease and cardiomyopathies. This paper presents a thorough review of recent studies on circRNA and lncRNAprotein binding within cardiac and vascular cells. It offers insight into the molecular mechanisms involved and emphasizes potential implications for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Huilei Zhao
- Department of Anesthesiology, The Third Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Ziqi Tan
- Department of Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jin Zhou
- Department of Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yifan Wu
- Department of Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qingwen Hu
- Department of Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qing Ling
- Department of Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jitao Ling
- Department of Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Menglu Liu
- Department of Cardiology, Seventh People's Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Yue Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Peng Yu
- Department of Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Yuan Jiang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangzhou, China.
| | - Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangzhou, China.
| |
Collapse
|
5
|
Azman AA, Siok-Fong C, Rajab NF, Md Zin RR, Ahmad Daud NN, Mohamad Hanif EA. The potential roles of lncRNA TINCR in triple negative breast cancer. Mol Biol Rep 2023; 50:7909-7917. [PMID: 37442895 DOI: 10.1007/s11033-023-08661-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 07/03/2023] [Indexed: 07/15/2023]
Abstract
Triple negative breast cancer (TNBC) is the most aggressive intrinsic breast cancer subtype characterized by the lack of estrogen receptor (ER), progesterone receptor (PR), and low levels of human epidermal growth factor receptor 2 (HER2). The complex nature of TNBC has resulted in little therapeutic progress for the past several decades. The standard of care remains the FEC cocktail (5-fluorouracil (5-FU), epirubicin and cyclophosphamide). However, early relapse and metastasis in TNBC patients persists in causing dismal clinical outcomes. Due to complex heterogeneity features of TNBC, identifying the biomarker associated to the chemoresistance remains a challenge. The emergence of the long non-coding RNA (lncRNA) as a potential signature may have proven to be a new deterrent to diagnostic and treatment options. Previous studies unveiled the associations of lncRNA in the development of TNBCs whereby the aggressiveness and response to therapies may be associated by the abrogation of the molecular mechanism lncRNA. Terminal differentiation induced ncRNA (TINCR) is a lncRNA which have been linked with many cancers including TNBC. The expression and behavior of TINCR may exert unfavorable outcome in TNBCs. Nevertheless, the underlying molecular mechanism of TINCR in driving chemoresistance in TNBC is not well understood. This review will highlight the potential molecular mechanisms of TINCR in TNBC chemoresistance and how it can serve as a future potential prognostic and therapeutic target for a better treatment intervention.
Collapse
Affiliation(s)
- Afreena Afiqah Azman
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Jalan Ya'acob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Chin Siok-Fong
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Jalan Ya'acob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Nor Fadilah Rajab
- Centre for Healthy Aging & Wellness, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Reena Rahayu Md Zin
- Faculty of Medicine (Pathology Department), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nurul Nadiah Ahmad Daud
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Jalan Ya'acob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Ezanee Azlina Mohamad Hanif
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Jalan Ya'acob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
6
|
Han X, Li C, Ji Q, Zhang L, Xie X, Shang H, Ye H. SLC26A4-AS1 Aggravates AngII-induced Cardiac Hypertrophy by Enhancing SLC26A4 Expression. Arq Bras Cardiol 2023; 120:e20210933. [PMID: 37098982 PMCID: PMC10263427 DOI: 10.36660/abc.20210933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2022] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND It has been reported that solute carrier family 26 members 4 antisense RNA 1 (SLC26A4-AS1) is highly related to cardiac hypertrophy. OBJECTIVE This research aims to investigate the role and specific mechanism of SLC26A4-AS1 in cardiac hypertrophy, providing a novel marker for cardiac hypertrophy treatment. METHODS Angiotensin II (AngII) was infused into neonatal mouse ventricular cardiomyocytes (NMVCs) to induce cardiac hypertrophy. Gene expression was detected by quantitative real-time PCR (RT-qPCR). Protein levels were evaluated via western blot. Functional assays analyzed the role of SLC26A4-AS1. The mechanism of SLC26A4-AS1 was assessed by RNA-binding protein immunoprecipitation (RIP), RNA pull-down, and luciferase reporter assays. The P value <0.05 was identified as statistical significance. Student's t-test evaluated the two-group comparison. The difference between different groups was analyzed by one-way analysis of variance (ANOVA). RESULTS SLC26A4-AS1 is upregulated in AngII-treated NMVCs and promotes AngII-induced cardiac hypertrophy. SLC26A4-AS1 regulates its nearby gene solute carrier family 26 members 4 (SLC26A4) via functioning as a competing endogenous RNA (ceRNA) to modulate the microRNA (miR)-301a-3p and miR-301b-3p in NMVCs. SLC26A4-AS1 promotes AngII-induced cardiac hypertrophy via upregulating SLC26A4 or sponging miR-301a-3p/miR-301b-3p. CONCLUSION SLC26A4-AS1 aggravates AngII-induced cardiac hypertrophy via sponging miR-301a-3p or miR-301b-3p to enhance SLC26A4 expression.
Collapse
Affiliation(s)
- Xiaoliang Han
- Departamento de CardiologiaInstituto de Controle de Tuberculose de AnhuiHefeiAnhuiChinaDepartamento de Cardiologia, Anhui Provincial Chest Hospital, (Instituto de Controle de Tuberculose de Anhui), Hefei, Anhui – China
| | - Chao Li
- Departamento de CardiologiaHospital HefeiMedical University of AnhuiHefeiAnhuiChinaDepartamento de Cardiologia, the Second People’s Hospital of Hefei (Hospital Hefei afiliado à Medical University of Anhui), Hefei, Anhui – China
| | - Qinjiong Ji
- Departamento de CardiologiaInstituto de Controle de Tuberculose de AnhuiHefeiAnhuiChinaDepartamento de Cardiologia, Anhui Provincial Chest Hospital, (Instituto de Controle de Tuberculose de Anhui), Hefei, Anhui – China
| | - Ling Zhang
- Departamento de CardiologiaInstituto de Controle de Tuberculose de AnhuiHefeiAnhuiChinaDepartamento de Cardiologia, Anhui Provincial Chest Hospital, (Instituto de Controle de Tuberculose de Anhui), Hefei, Anhui – China
| | - Xiaofei Xie
- Departamento de CardiologiaInstituto de Controle de Tuberculose de AnhuiHefeiAnhuiChinaDepartamento de Cardiologia, Anhui Provincial Chest Hospital, (Instituto de Controle de Tuberculose de Anhui), Hefei, Anhui – China
| | - Huijuan Shang
- Departamento de CardiologiaInstituto de Controle de Tuberculose de AnhuiHefeiAnhuiChinaDepartamento de Cardiologia, Anhui Provincial Chest Hospital, (Instituto de Controle de Tuberculose de Anhui), Hefei, Anhui – China
| | - Hong Ye
- Departamento de CardiologiaInstituto de Controle de Tuberculose de AnhuiHefeiAnhuiChinaDepartamento de Cardiologia, Anhui Provincial Chest Hospital, (Instituto de Controle de Tuberculose de Anhui), Hefei, Anhui – China
| |
Collapse
|
7
|
Emami Meybodi SM, Soleimani N, Yari A, Javadifar A, Tollabi M, Karimi B, Emami Meybodi M, Seyedhossaini S, Brouki Milan P, Dehghani Firoozabadi A. Circulatory long noncoding RNAs (circulatory-LNC-RNAs) as novel biomarkers and therapeutic targets in cardiovascular diseases: Implications for cardiovascular diseases complications. Int J Biol Macromol 2023; 225:1049-1071. [PMID: 36414082 DOI: 10.1016/j.ijbiomac.2022.11.167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Abstract
Cardiovascular diseases (CVDs) are a group of disorders with major global health consequences. The prevalence of CVDs continues to grow due to population-aging and lifestyle modifications. Non-coding RNAs (ncRNAs) as key regulators of cell signaling pathways have gained attention in the occurrence and development of CVDs. Exosomal-lncRNAs (exos-lncRNAs) are emerging biomarkers due to their high sensitivity and specificity, stability, accuracy and accessibility in the biological fluids. Recently, circulatory and exos-based-lncRNAs are emerging and novel bio-tools in various pathogenic conditions. It is worth mentioning that dysregulation of these molecules has been found in different types of CVDs. In this regard, we aimed to discuss the knowledge gaps and suggest research priorities regarding circulatory and exos-lncRNAs as novel bio-tools and therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Seyed Mahdi Emami Meybodi
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Nafiseh Soleimani
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Abolfazl Yari
- Cellular and Molecular Research Center, Birjand University of Medical Mciences, Birjand, Iran.
| | - Amin Javadifar
- Immunology Research Center, Inflammation and Inflammatory Disease Division, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Tollabi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Bahareh Karimi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Mahmoud Emami Meybodi
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Seyedmostafa Seyedhossaini
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Peiman Brouki Milan
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Ali Dehghani Firoozabadi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Garcia-Padilla C, Lozano-Velasco E, Garcia-Lopez V, Aranega A, Franco D, Garcia-Martinez V, Lopez-Sanchez C. Comparative Analysis of Non-Coding RNA Transcriptomics in Heart Failure. Biomedicines 2022; 10:3076. [PMID: 36551832 PMCID: PMC9775550 DOI: 10.3390/biomedicines10123076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Heart failure constitutes a clinical complex syndrome with different symptomatic characteristics depending on age, sex, race and ethnicity, among others, which has become a major public health issue with an increasing prevalence. One of the most interesting tools seeking to improve prevention, diagnosis, treatment and prognosis of this pathology has focused on finding new molecular biomarkers since heart failure relies on deficient cardiac homeostasis, which is regulated by a strict gene expression. Therefore, currently, analyses of non-coding RNA transcriptomics have been oriented towards human samples. The present review develops a comparative study emphasizing the relevance of microRNAs, long non-coding RNAs and circular RNAs as potential biomarkers in heart failure. Significantly, further studies in this field of research are fundamental to supporting their widespread clinical use. In this sense, the various methodologies used by the authors should be standardized, including larger cohorts, homogeneity of the samples and uniformity of the bioinformatic pipelines used to reach stratification and statistical significance of the results. These basic adjustments could provide promising steps to designing novel strategies for clinical management of patients with heart failure.
Collapse
Affiliation(s)
- Carlos Garcia-Padilla
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
| | - Estefanía Lozano-Velasco
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Medina Foundation, 18016 Granada, Spain
| | - Virginio Garcia-Lopez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Amelia Aranega
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Medina Foundation, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Medina Foundation, 18016 Granada, Spain
| | - Virginio Garcia-Martinez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Carmen Lopez-Sanchez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| |
Collapse
|
9
|
Khan FB, Uddin S, Elderdery AY, Goh KW, Ming LC, Ardianto C, Palakot AR, Anwar I, Khan M, Owais M, Huang CY, Daddam JR, Khan MA, Shoaib S, Khursheed M, Reshadat S, Khayat Kashani HR, Mirza S, Khaleel AA, Ayoub MA. Illuminating the Molecular Intricacies of Exosomes and ncRNAs in Cardiovascular Diseases: Prospective Therapeutic and Biomarker Potential. Cells 2022; 11:cells11223664. [PMID: 36429092 PMCID: PMC9688392 DOI: 10.3390/cells11223664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/19/2022] Open
Abstract
Cardiovascular diseases (CVDs) are one of the leading causes of death worldwide. Accumulating evidences have highlighted the importance of exosomes and non-coding RNAs (ncRNAs) in cardiac physiology and pathology. It is in general consensus that exosomes and ncRNAs play a crucial role in the maintenance of normal cellular function; and interestingly it is envisaged that their potential as prospective therapeutic candidates and biomarkers are increasing rapidly. Considering all these aspects, this review provides a comprehensive overview of the recent understanding of exosomes and ncRNAs in CVDs. We provide a great deal of discussion regarding their role in the cardiovascular system, together with providing a glimpse of ideas regarding strategies exploited to harness their potential as a therapeutic intervention and prospective biomarker against CVDs. Thus, it could be envisaged that a thorough understanding of the intricacies related to exosomes and ncRNA would seemingly allow their full exploration and may lead clinical settings to become a reality in near future.
Collapse
Affiliation(s)
- Farheen Badrealam Khan
- Department of Biology, College of Science, The United Arab Emirates University, Al Ain 15551, United Arab Emirates
- Correspondence: (F.B.K.); (M.A.A.); (C.A.)
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Khang Wen Goh
- Faculty of Data Sciences and Information Technology, INTI International University, Nilai 78100, Malaysia
| | - Long Chiau Ming
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
- Correspondence: (F.B.K.); (M.A.A.); (C.A.)
| | - Abdul Rasheed Palakot
- Department of Biology, College of Science, The United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Irfa Anwar
- Department of Biology, College of Science, The United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Mohsina Khan
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mohammad Owais
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Chih-Yang Huang
- Department of Biotechnology, Asia University, Taichung 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Centre of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Jayasimha Rayalu Daddam
- Department of Ruminant Science, Institute of Animal Sciences, Agriculture Research Organization, Volcani Center, Rishon Lezion 7505101, Israel
| | - Meraj Alam Khan
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children & DigiBiomics Inc, Toronto, ON M51X8, Canada
| | - Shoaib Shoaib
- Department Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Md Khursheed
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
| | - Sara Reshadat
- Department of Internal Medicine, Semnan University of Medical Sciences, Semnan 3513119111, Iran
| | | | - Sameer Mirza
- Department of Chemistry, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Abbas A. Khaleel
- Department of Chemistry, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Mohammed Akli Ayoub
- Department of Biology, College of Science, The United Arab Emirates University, Al Ain 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates
- Department of Biology, College of Arts and Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
- Correspondence: (F.B.K.); (M.A.A.); (C.A.)
| |
Collapse
|
10
|
Cai Y, Li Y. LncRNA Gm43843 Promotes Cardiac Hypertrophy via miR-153-3p/Cacna1c Axis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2160804. [PMID: 36262165 PMCID: PMC9576395 DOI: 10.1155/2022/2160804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been reported to engage in many human diseases, including cardiac hypertrophy. Cardiac hypertrophy was mainly caused by excessive pressure load, which can eventually lead to a decline in myocardial contractility. Gm43843, a novel lncRNA, has not been well explored in cardiac hypertrophy so far. Herein, we are going to search the function and the underlying molecular mechanism of Gm43843 in cardiac hypertrophy. Gm43843 levels were measured via qRT-PCR in mouse myocardial cells when they are treated with angiogenin II (Ang II) or transfected with different plasmids. Western blot assay was implemented to detect the cardiac hypertrophy-related protein markers, while the cell was analyzed via immunofluorescence (IF) assay to evaluate the hypertrophy. Meanwhile, the binding of Gm43843 and the putative targets was examined based on mechanistic assay results. We found that Gm43843 expression was increased with the elevated concentration of Ang II. Inhibited Gm43843 was detected to reduce the hypertrophy of mouse myocardial cells. Meanwhile, Gm43843/miR-153-3p/Cacna1c axis was found to modulate cardiac hypertrophy. In short, Gm43843 promotes cardiac hypertrophy via miR-153-3p/Cacna1c axis.
Collapse
Affiliation(s)
- Yuhua Cai
- Department of Cardiology, Jingzhou First Municipal Hospital, Jingzhou 434000, Hubei Province, China
| | - Yunpeng Li
- Department of Cardiovasology, Dongfeng Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| |
Collapse
|
11
|
Ji H, Qu J, Peng W, Yang L. Downregulation of lncRNA MALAT1 Inhibits Angiotensin II-induced Hypertrophic Effects of Cardiomyocytes by Regulating SIRT4 via miR-93-5p. Int Heart J 2022; 63:602-611. [PMID: 35650160 DOI: 10.1536/ihj.21-332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cardiac hypertrophy is a leading risk for heart failure and sudden death. Long non-coding RNAs (lncRNAs) have been implicated in a variety of human diseases, including cardiac hypertrophy. We aimed to investigate the potential role and functional mechanism of lncRNA metastasis-associated in lung adenocarcinoma transcript 1 (MALAT1) in cardiac hypertrophy. C57BL/6 mice underwent transverse aortic constriction (TAC) to induce cardiac hypertrophy in vivo. The expression of MALAT1, miR-93-5p, and sirtuin 4 (SIRT4) mRNA was detected using a quantitative real-time polymerase chain reaction. The protein levels of cardiac hypertrophy-related markers, including atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP) and β-myosin heavy chain (β-MHC), and SIRT4 were measured via western blotting. The putative interaction between miR-93-5p and MALAT1 or SIRT4 was verified using a dual-luciferase reporter assay, RNA immunoprecipitation assay, or pull-down assay. Consequently, the expression of MALAT1 and SIRT4 was increased in TAC-treated mouse heart and angiotensin II (Ang-II)-induced cardiomyocytes, whereas the expression of miR-93-5p was decreased. Ang-II promoted the expression of ANP, BNP, and β-MHC and the surface area of cardiomyocytes, whereas MALAT1 downregulation impaired their expression and cell area. MiR-93-5p was a target of MALAT1, and its inhibition reversed the effects of MALAT1 downregulation. More importantly, MALAT1 modulated SIRT4 expression by degrading miR-93-5p. The expression of ANP, BNP, and β-MHC suppressed by miR-93-5p restoration was recovered by SIRT4 promotion. Overall, MALAT1 knockdown ameliorated cardiac hypertrophy partly by regulating the miR-93-5p/SIRT4 network, indicating that MALAT1 was a substantial indicator of cardiac hypertrophy.
Collapse
Affiliation(s)
- Huanchun Ji
- Department of Cardiology, Dalian Second People's Hospital
| | - Jingxian Qu
- Department of Cardiology, Dalian Second People's Hospital
| | - Wei Peng
- Department of Cardiology, Dalian Second People's Hospital
| | - Long Yang
- Department of Cardiology, Dalian Second People's Hospital
| |
Collapse
|
12
|
Non-Coding RNAs in the Therapeutic Landscape of Pathological Cardiac Hypertrophy. Cells 2022; 11:cells11111805. [PMID: 35681500 PMCID: PMC9180404 DOI: 10.3390/cells11111805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases are a major health problem, and long-term survival for people diagnosed with heart failure is, still, unrealistic. Pathological cardiac hypertrophy largely contributes to morbidity and mortality, as effective therapeutic approaches are lacking. Non-coding RNAs (ncRNAs) arise as active regulators of the signaling pathways and mechanisms that govern this pathology, and their therapeutic potential has received great attention in the last decades. Preclinical studies in large animal models have been successful in ameliorating cardiac hypertrophy, and an antisense drug for the treatment of heart failure has, already, entered clinical trials. In this review, we provide an overview of the molecular mechanisms underlying cardiac hypertrophy, the involvement of ncRNAs, and the current therapeutic landscape of oligonucleotides targeting these regulators. Strategies to improve the delivery of such therapeutics and overcome the actual challenges are, also, defined and discussed. With the fast advance in the improvement of oligonucleotide drug delivery, the inclusion of ncRNAs-targeting therapies for cardiac hypertrophy seems, increasingly, a closer reality.
Collapse
|
13
|
Cuesta-Llavona E, Lorca R, Rolle V, Alonso B, Iglesias S, Rodríguez-Reguero J, Duarte-Herrera ID, Pérez-Oliveira S, Junco-Vicente A, Lago CG, Coto E, Gómez J. Association of the Genetic Variation in the Long Non-Coding RNA FENDRR with the Risk of Developing Hypertrophic Cardiomyopathy. Life (Basel) 2022; 12:life12060818. [PMID: 35743849 PMCID: PMC9225451 DOI: 10.3390/life12060818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/20/2022] [Accepted: 05/28/2022] [Indexed: 11/16/2022] Open
Abstract
Background: In around 40−60% of Hypertrophic Cardiomyopathy (HCM) cases pathogenic variants are not identified. Our aim was to evaluate the possible association of lncRNAs with the risk of developing HCM. Methods: We sequenced 10 lncRNAs coding genes that have been associated with cardiovascular disease in a discovery cohort (238 HCM patients and 212 controls) by NGS, and genotyped rs74035787 G>A and rs1424019 A>G polymorphism in a validation cohort (962 HCM patients and 923 controls). Finally, we sequenced the FENDRR promoter by Sanger sequencing. Results: We observed by NGS that FENDRR rs39527, rs39529 and rs40384 polymorphisms were significantly associated with HCM in our cohort (p = 0.0284; OR: 0.24, 95%CI: 0.07−0.86). NGS results were confirmed by genotyping rs74035787 polymorphism (p = 0.001; OR:0.38, 95%CI: 0.21−0.66). Moreover, it is also associated when stratification by sex (p = 0.003; OR:0.20, 95%CI: 0.06−0.53), and age (≥50 years old p = 0.001, OR:0.33, 95%CI: 0.16−0.63) Moreover, the risk of HCM in the carriers of the GG genotype of the rs1424019 polymorphism was significantly higher than that of the AA/AG genotypes carriers in the elderly subjects (p = 0.045, OR:1.24, 95%CI: 1.01−1.53). On the other hand, we observed significant differences in the rs74035787 A/rs1424019 G haplotype frequency (p = 0.0035; OR: 0.20, 95%CI: 0.07−0.59). Conclusions: Our study suggested a significant association between FENDRR gene variants and HCM.
Collapse
Affiliation(s)
- Elías Cuesta-Llavona
- Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain; (E.C.-L.); (R.L.); (B.A.); (S.I.); (J.R.-R.); (I.D.D.-H.); (S.P.-O.); (A.J.-V.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (V.R.); (C.G.L.)
- Redes de Investigación Cooperativa Orientadas a Resultadosen Salud (RICORs), 28029 Madrid, Spain
| | - Rebeca Lorca
- Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain; (E.C.-L.); (R.L.); (B.A.); (S.I.); (J.R.-R.); (I.D.D.-H.); (S.P.-O.); (A.J.-V.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (V.R.); (C.G.L.)
- Redes de Investigación Cooperativa Orientadas a Resultadosen Salud (RICORs), 28029 Madrid, Spain
- Unidad de Cardiopatías Familiares del HUCA, 33011 Oviedo, Spain
| | - Valeria Rolle
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (V.R.); (C.G.L.)
| | - Belén Alonso
- Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain; (E.C.-L.); (R.L.); (B.A.); (S.I.); (J.R.-R.); (I.D.D.-H.); (S.P.-O.); (A.J.-V.)
| | - Sara Iglesias
- Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain; (E.C.-L.); (R.L.); (B.A.); (S.I.); (J.R.-R.); (I.D.D.-H.); (S.P.-O.); (A.J.-V.)
| | - Julian Rodríguez-Reguero
- Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain; (E.C.-L.); (R.L.); (B.A.); (S.I.); (J.R.-R.); (I.D.D.-H.); (S.P.-O.); (A.J.-V.)
- Unidad de Cardiopatías Familiares del HUCA, 33011 Oviedo, Spain
| | - Israel David Duarte-Herrera
- Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain; (E.C.-L.); (R.L.); (B.A.); (S.I.); (J.R.-R.); (I.D.D.-H.); (S.P.-O.); (A.J.-V.)
| | - Sergio Pérez-Oliveira
- Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain; (E.C.-L.); (R.L.); (B.A.); (S.I.); (J.R.-R.); (I.D.D.-H.); (S.P.-O.); (A.J.-V.)
| | - Alejandro Junco-Vicente
- Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain; (E.C.-L.); (R.L.); (B.A.); (S.I.); (J.R.-R.); (I.D.D.-H.); (S.P.-O.); (A.J.-V.)
| | - Claudia García Lago
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (V.R.); (C.G.L.)
| | - Eliecer Coto
- Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain; (E.C.-L.); (R.L.); (B.A.); (S.I.); (J.R.-R.); (I.D.D.-H.); (S.P.-O.); (A.J.-V.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (V.R.); (C.G.L.)
- Redes de Investigación Cooperativa Orientadas a Resultadosen Salud (RICORs), 28029 Madrid, Spain
- Unidad de Cardiopatías Familiares del HUCA, 33011 Oviedo, Spain
- Medicicine Department, Universidad de Oviedo, 33003 Oviedo, Spain
- Correspondence: (E.C.); (J.G.); Tel.: +34-985-10-80-00 (ext. 37484) (J.G.)
| | - Juan Gómez
- Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain; (E.C.-L.); (R.L.); (B.A.); (S.I.); (J.R.-R.); (I.D.D.-H.); (S.P.-O.); (A.J.-V.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (V.R.); (C.G.L.)
- Redes de Investigación Cooperativa Orientadas a Resultadosen Salud (RICORs), 28029 Madrid, Spain
- Unidad de Cardiopatías Familiares del HUCA, 33011 Oviedo, Spain
- CIBER-Enfermedades Respiratorias, 28029 Madrid, Spain
- Correspondence: (E.C.); (J.G.); Tel.: +34-985-10-80-00 (ext. 37484) (J.G.)
| |
Collapse
|
14
|
Zhu C, Wang M, Yu X, Shui X, Tang L, Chen Z, Xiong Z. lncRNA NBR2 attenuates angiotensin II-induced myocardial hypertrophy through repressing ER stress via activating LKB1/AMPK/Sirt1 pathway. Bioengineered 2022; 13:13667-13679. [PMID: 35703318 PMCID: PMC9275985 DOI: 10.1080/21655979.2022.2062527] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Myocardial hypertrophy leads to heart failure (HF), and emerging researchers have illustrated that long noncoding RNAs (lncRNAs) modulate myocardial hypertrophy. Here, we explored the role and mechanism of a novel lncRNA, NBR2, in modulating angiotensin II (Ang II)-induced myocardial hypertrophy. First, we examined plasma NBR2 levels in 25 patients with HF and myocardial hypertrophy and ten healthy donors and analyzed the correlation between NBR2 profiles and patients’ clinical indicators. In addition, the overexpression experiment of NBR2 was carried out to probe the influence of NBR2 on myocardial hypertrophy. lncRNA NBR2 was down-regulated in plasma of patients with HF and myocardial hypertrophy (vs. healthy controls), and its level was negatively correlated with cardiac function (represented by left ventricular end-diastolic diameter and left ventricular ejection fraction) and degree of myocardial hypertrophy. Besides, Ang II treatment intensified the hypertrophy of human myocardial cell lines (HCM and AC16) and curbed the NBR2 expression. Overexpressing lncRNA NBR2 alleviated Angiotension II–induced myocardial hypertrophy and declined the profiles of hypertrophic markers. Moreover, up-regulating lncRNA NBR2 weakened Ang II-mediated endoplasmic reticulum (ER) stress and activated the LKB1/AMPK/Sirt1 pathway. Interfering with the LKB1/AMPK/Sirt1 axis abated the lncRNA NBR2-mediated inhibitory effect on myocardial hypertrophy and ER stress. This study confirmed that lncRNA NBR2 dampened myocardial hypertrophy and ER stress by modulating the LKB1/AMPK/Sirt1 pathway. Our study provides the first evidence that lncRNA NBR2 is positively associated with myocardial hypertrophy.
Collapse
Affiliation(s)
- Cansheng Zhu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Min Wang
- Department of Cardiovascular Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xianguan Yu
- Department of Cardiovascular Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xing Shui
- Department of Cardiovascular Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Leile Tang
- Department of Cardiovascular Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zefeng Chen
- Department of Cardiovascular Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhaojun Xiong
- Department of Cardiovascular Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Mangraviti N, De Windt LJ. Long Non-Coding RNAs in Cardiac Hypertrophy. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:836418. [PMID: 39086960 PMCID: PMC11285587 DOI: 10.3389/fmmed.2022.836418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/08/2022] [Indexed: 08/02/2024]
Abstract
Heart disease represents one of the main challenges in modern medicine with insufficient treatment options. Whole genome sequencing allowed for the discovery of several classes of non-coding RNA (ncRNA) and widened our understanding of disease regulatory circuits. The intrinsic ability of long ncRNAs (lncRNAs) and circular RNAs (circRNAs) to regulate gene expression by a plethora of mechanisms make them candidates for conceptually new treatment options. However, important questions remain to be addressed before we can fully exploit the therapeutic potential of these molecules. Increasing our knowledge of their mechanisms of action and refining the approaches for modulating lncRNAs expression are just a few of the challenges we face. The accurate identification of novel lncRNAs is hampered by their relatively poor cross-species sequence conservation and their low and context-dependent expression pattern. Nevertheless, progress has been made in their annotation in recent years, while a few experimental studies have confirmed the value of lncRNAs as new mechanisms in the development of cardiac hypertrophy and other cardiovascular diseases. Here, we explore cardiac lncRNA biology and the evidence that this class of molecules has therapeutic benefit to treat cardiac hypertrophy.
Collapse
Affiliation(s)
| | - Leon J. De Windt
- Department of Molecular Genetics, Faculty of Science and Engineering, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
16
|
Tu S, Wang XY, Zeng LX, Shen ZJ, Zhang ZH. LncRNA TINCR improves cardiac hypertrophy by regulating the miR-211-3p-VEGFB-SDF-1α-CXCR4 pathway. J Transl Med 2022; 102:253-262. [PMID: 34732848 DOI: 10.1038/s41374-021-00678-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/12/2021] [Accepted: 09/07/2021] [Indexed: 11/08/2022] Open
Abstract
Cardiac hypertrophy is a common cardiovascular disease that is found worldwide and is characterized by heart enlargement, eventually resulting in heart failure. Exploring the regulatory mechanism of cardiac hypertrophy is beneficial for understanding its pathogenesis and treatment. In our study, we have showed TINCR was downregulated and miR-211-3p was upregulated in TAC- or Ang II-induced models of cardiac hypertrophy. Dual luciferase and RIP assays revealed that TINCR served as a competitive endogenous RNA (ceRNA) for miR-211-3p. Then, we observed that knockdown of miR-211-3p alleviated TAC- or Ang II-induced cardiac hypertrophy both in vivo and in vitro. Mechanistically, we demonstrated that miR-211-3p directly targeted VEGFB and thus regulated the expression of SDF-1α and CXCR4. Rescue assays further confirmed that TINCR suppressed the progression of cardiac hypertrophy by competitively binding to miR-211-3p, thereby enhancing the expression of VEGFB and activating the VEGFB-SDF-1α- CXCR4 signal. Furthermore, overexpression of TINCR suppressed TAC-induced cardiac hypertrophy in vivo by targeting miR-211-3p-VEGFB-SDF-1α- CXCR4 signalling. In conclusion, our research suggests that LncRNA TINCR improves cardiac hypertrophy by targeting miR-211-3p, thus relieving its suppressive effects on the VEGFB-SDF-1α-CXCR4 signalling axis. TINCR and miR-211-3p might act as therapeutic targets for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Shan Tu
- Department of Cardiology, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Xiao-Yan Wang
- Department of Cardiology, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Li-Xiong Zeng
- Department of Cardiology, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Zhi-Jie Shen
- Department of Cardiology, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Zhi-Hui Zhang
- Department of Cardiology, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, China.
| |
Collapse
|
17
|
Shahzadi SK, Naidoo N, Alsheikh-Ali A, Rizzo M, Rizvi AA, Santos RD, Banerjee Y. Reconnoitering the Role of Long-Noncoding RNAs in Hypertrophic Cardiomyopathy: A Descriptive Review. Int J Mol Sci 2021; 22:ijms22179378. [PMID: 34502285 PMCID: PMC8430576 DOI: 10.3390/ijms22179378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/05/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common form of hereditary cardiomyopathy. It is characterized by an unexplained non-dilated hypertrophy of the left ventricle with a conserved or elevated ejection fraction. It is a genetically heterogeneous disease largely caused by variants of genes encoding for cardiac sarcomere proteins, including MYH7, MYBPC3, ACTC1, TPM1, MYL2, MYL3, TNNI3, and TNNT23. Preclinical evidence indicates that the enhanced calcium sensitivity of the myofilaments plays a key role in the pathophysiology of HCM. Notably, this is not always a direct consequence of sarcomeric variations but may also result from secondary mutation-driven alterations. Long non-coding RNAs (lncRNAs) are a large class of transcripts ≥200 nucleotides in length that do not encode proteins. Compared to coding mRNAs, most lncRNAs are not as well-annotated and their functions are greatly unexplored. Nevertheless, increasing evidence shows that lncRNAs are involved in a variety of biological processes and diseases including HCM. Accumulating evidence has indicated that lncRNAs are dysregulated in HCM, and closely related to sarcomere construction, calcium channeling and homeostasis of mitochondria. In this review, we have summarized the known regulatory and functional roles of lncRNAs in HCM.
Collapse
Affiliation(s)
- Syeda K. Shahzadi
- Department of Basic Medical Sciences, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (S.K.S.); (A.A.-A.)
| | - Nerissa Naidoo
- Department of Basic Medical Sciences, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (S.K.S.); (A.A.-A.)
- Correspondence: (N.N.); (Y.B.); Tel.: +971-4383-8728 (N.N.); +971-4383-8710 (Y.B.)
| | - Alawi Alsheikh-Ali
- Department of Basic Medical Sciences, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (S.K.S.); (A.A.-A.)
- Dubai Health Authority, Dubai 66566, United Arab Emirates
| | - Manfredi Rizzo
- Department of Health Promotion Sciences, Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy;
| | - Ali A. Rizvi
- Division of Endocrinology, Metabolism, and Lipids, School of Medicine, Emory University, Atlanta, GA 30322, USA;
| | - Raul D. Santos
- The Heart Institute, Faculty of Medicine, University of São Paulo, São Paulo 01000, Brazil;
| | - Yajnavalka Banerjee
- Department of Basic Medical Sciences, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates; (S.K.S.); (A.A.-A.)
- Centre of Medical Education, School of Medicine, University of Dundee, Dundee DD1 4HN, UK
- Correspondence: (N.N.); (Y.B.); Tel.: +971-4383-8728 (N.N.); +971-4383-8710 (Y.B.)
| |
Collapse
|
18
|
Li D, Yang Y, Wang S, He X, Liu M, Bai B, Tian C, Sun R, Yu T, Chu X. Role of acetylation in doxorubicin-induced cardiotoxicity. Redox Biol 2021; 46:102089. [PMID: 34364220 PMCID: PMC8350499 DOI: 10.1016/j.redox.2021.102089] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/23/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
As a potent chemotherapeutic agent, doxorubicin (DOX) is widely used for the treatment of a variety of cancers However, its clinical utility is limited by dose-dependent cardiotoxicity, and pathogenesis has traditionally been attributed to the formation of reactive oxygen species (ROS). Accordingly, the prevention of DOX-induced cardiotoxicity is an indispensable goal to optimize therapeutic regimens and reduce morbidity. Acetylation is an emerging and important epigenetic modification regulated by histone deacetylases (HDACs) and histone acetyltransferases (HATs). Despite extensive studies of the molecular basis and biological functions of acetylation, the application of acetylation as a therapeutic target for cardiotoxicity is in the initial stage, and further studies are required to clarify the complex acetylation network and improve the clinical management of cardiotoxicity. In this review, we summarize the pivotal functions of HDACs and HATs in DOX-induced oxidative stress, the underlying mechanisms, the contributions of noncoding RNAs (ncRNAs) and exercise-mediated deacetylases to cardiotoxicity. Furthermore, we describe research progress related to several important SIRT activators and HDAC inhibitors with potential clinical value for chemotherapy and cardiotoxicity. Collectively, a comprehensive understanding of specific roles and recent developments of acetylation in doxorubicin-induced cardiotoxicity will provide a basis for improved treatment outcomes in cancer and cardiovascular diseases.
Collapse
Affiliation(s)
- Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, Qingdao, 266071, China
| | - Shizhong Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xiangqin He
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Meixin Liu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Baochen Bai
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chao Tian
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ruicong Sun
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Basic Medicine School, Qingdao University, 38 Deng Zhou Road, Qingdao, 266021, China.
| | - Xianming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China; Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266071, China.
| |
Collapse
|
19
|
Abstract
Cardiac hypertrophy (CH) is generally considered adaptive responses that may occur after myocardial infarction, pressure overload, volume overload, inflammatory heart muscle disease, or idiopathic dilated cardiomyopathy, whereas long-term stimulation eventually leads to heart failure (HF). However, the current molecular mechanisms involved in CH are unclear. Recently, increasing evidences reveal that long non-coding RNAs (lncRNAs) play vital roles in CH. Different lncRNAs can promote or inhibit the pathological process of CH by different mechanisms, while the regulation of lncRNAs expression can improve CH. Thus, CH-related lncRNAs may become a novel field of research on CH.
Collapse
Affiliation(s)
- Jinghui Sun
- Cardiovascular Disease Research Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Haidian District, Beijing, 100091, China
| | - Chenglong Wang
- Cardiovascular Disease Research Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Haidian District, Beijing, 100091, China.
| |
Collapse
|
20
|
Rey F, Urrata V, Gilardini L, Bertoli S, Calcaterra V, Zuccotti GV, Cancello R, Carelli S. Role of long non-coding RNAs in adipogenesis: State of the art and implications in obesity and obesity-associated diseases. Obes Rev 2021; 22:e13203. [PMID: 33443301 PMCID: PMC8244036 DOI: 10.1111/obr.13203] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 12/14/2022]
Abstract
Obesity is an evolutionary, chronic, and relapsing disease that consists of a pathological accumulation of adipose tissue able to increase morbidity for high blood pressure, type 2 diabetes, metabolic syndrome, and obstructive sleep apnea in adults, children, and adolescents. Despite intense research over the last 20 years, obesity remains today a disease with a complex and multifactorial etiology. Recently, long non-coding RNAs (lncRNAs) are emerging as interesting new regulators as different lncRNAs have been found to play a role in early and late phases of adipogenesis and to be implicated in obesity-associated complications onset. In this review, we discuss the most recent advances on the role of lncRNAs in adipocyte biology and in obesity-associated complications. Indeed, more and more researchers are focusing on investigating the underlying roles that these molecular modulators could play. Even if a significant number of evidence is correlation-based, with lncRNAs being differentially expressed in a specific disease, recent works are now focused on deeply analyzing how lncRNAs can effectively modulate the disease pathogenesis onset and progression. LncRNAs possibly represent new molecular markers useful in the future for both the early diagnosis and a prompt clinical management of patients with obesity.
Collapse
Affiliation(s)
- Federica Rey
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Milan, Italy.,Pediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Valentina Urrata
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Milan, Italy.,Pediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Luisa Gilardini
- Obesity Unit-Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Simona Bertoli
- Obesity Unit-Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy.,International Center for the Assessment of Nutritional Status (ICANS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Valeria Calcaterra
- Pediatrics and Adolescentology Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy.,Department of Pediatrics, Children's Hospital "V. Buzzi", Milan, Italy
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Milan, Italy.,Pediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy.,Department of Pediatrics, Children's Hospital "V. Buzzi", Milan, Italy
| | - Raffaella Cancello
- Obesity Unit-Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Milan, Italy.,Pediatric Clinical Research Center Fondazione "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| |
Collapse
|
21
|
Qin J, Guo N, Tong J, Wang Z. Function of histone methylation and acetylation modifiers in cardiac hypertrophy. J Mol Cell Cardiol 2021; 159:120-129. [PMID: 34175302 DOI: 10.1016/j.yjmcc.2021.06.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 06/14/2021] [Accepted: 06/19/2021] [Indexed: 12/15/2022]
Abstract
Cardiac hypertrophy is an adaptive response of the heart to increased workload induced by various physiological or pathological stimuli. It is a common pathological process in multiple cardiovascular diseases, and it ultimately leads to heart failure. The development of cardiac hypertrophy is accompanied by gene expression reprogramming, a process that is largely dependent on epigenetic regulation. Histone modifications such as methylation and acetylation are dynamically regulated under cardiac stress. These consequently contribute to the pathogenesis of cardiac hypertrophy via compensatory or maladaptive transcriptome reprogramming. Histone methylation and acetylation modifiers play crucial roles in epigenetic remodeling during the pathogenesis of cardiac hypertrophy. Regulation of histone methylation and acetylation modifiers serves as a bridge between signal transduction and downstream gene reprogramming. Exploring the role of histone modifiers in cardiac hypertrophy provides novel therapeutic strategies to treat cardiac hypertrophy and heart failure. In this review, we summarize the recent advancements in functional histone methylation and acetylation modifiers in cardiac hypertrophy, with an emphasis on the underlying mechanisms and the therapeutic potential.
Collapse
Affiliation(s)
- Jian Qin
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ningning Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingjing Tong
- School of Life Sciences, Central China Normal University, Wuhan, China
| | - Zhihua Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, China; State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
22
|
Bu S, Singh KK. Epigenetic Regulation of Autophagy in Cardiovascular Pathobiology. Int J Mol Sci 2021; 22:ijms22126544. [PMID: 34207151 PMCID: PMC8235464 DOI: 10.3390/ijms22126544] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the number one cause of debilitation and mortality worldwide, with a need for cost-effective therapeutics. Autophagy is a highly conserved catabolic recycling pathway triggered by various intra- or extracellular stimuli to play an essential role in development and pathologies, including CVDs. Accordingly, there is great interest in identifying mechanisms that govern autophagic regulation. Autophagic regulation is very complex and multifactorial that includes epigenetic pathways, such as histone modifications to regulate autophagy-related gene expression, decapping-associated mRNA degradation, microRNAs, and long non-coding RNAs; pathways are also known to play roles in CVDs. Molecular understanding of epigenetic-based pathways involved in autophagy and CVDs not only will enhance the understanding of CVDs, but may also provide novel therapeutic targets and biomarkers for CVDs.
Collapse
Affiliation(s)
| | - Krishna K. Singh
- Correspondence: ; Tel.: +1-519-661-2111 (ext. 80542) (Office) or (ext. 85683) (Lab)
| |
Collapse
|
23
|
Wei S, Ma W, Zhang B, Li W. NLRP3 Inflammasome: A Promising Therapeutic Target for Drug-Induced Toxicity. Front Cell Dev Biol 2021; 9:634607. [PMID: 33912556 PMCID: PMC8072389 DOI: 10.3389/fcell.2021.634607] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/18/2021] [Indexed: 12/13/2022] Open
Abstract
Drug-induced toxicity, which impairs human organ function, is a serious problem during drug development that hinders the clinical use of many marketed drugs, and the underlying mechanisms are complicated. As a sensor of infections and external stimuli, nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome plays a key role in the pathological process of various diseases. In this review, we specifically focused on the role of NLRP3 inflammasome in drug-induced diverse organ toxicities, especially the hepatotoxicity, nephrotoxicity, and cardiotoxicity. NLRP3 inflammasome is involved in the initiation and deterioration of drug-induced toxicity through multiple signaling pathways. Therapeutic strategies via inhibiting NLRP3 inflammasome for drug-induced toxicity have made significant progress, especially in the protective effects of the phytochemicals. Growing evidence collected in this review indicates that NLRP3 is a promising therapeutic target for drug-induced toxicity.
Collapse
Affiliation(s)
- Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Wanjun Ma
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
24
|
Kay M, Soltani BM. LncRNAs in Cardiomyocyte Maturation: New Window for Cardiac Regenerative Medicine. Noncoding RNA 2021; 7:ncrna7010020. [PMID: 33802186 PMCID: PMC8005985 DOI: 10.3390/ncrna7010020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiomyocyte (CM) maturation, which is characterized by structural, functional, and metabolic specializations, is the last phase of CM development that prepares the cells for efficient and forceful contraction throughout life. Over the past decades, CM maturation has gained increased attention due to the fact that pluripotent stem cell-derived CMs are structurally, transcriptionally, and functionally immature and embryonic-like, which causes a defect in cell replacement therapy. The current challenge is to discover and understand the molecular mechanisms, which control the CM maturation process. Currently, emerging shreds of evidence emphasize the role of long noncoding RNAs (lncRNAs) in regulating different aspects of CM maturation, including myofibril maturation, electrophysiology, and Ca2+ handling maturation, metabolic maturation and proliferation to hypertrophy transition. Here, we describe the structural and functional characteristics of mature CMs. Furthermore, this review highlights the lncRNAs as crucial regulators of different aspects in CM maturation, which have the potential to be used for mature CM production. With the current advances in oligonucleotide delivery; lncRNAs may serve as putative therapeutic targets to produce highly mature CMs for research and regenerative medicine.
Collapse
|
25
|
Long Non-Coding RNAs (lncRNAs) in Cardiovascular Disease Complication of Type 2 Diabetes. Diagnostics (Basel) 2021; 11:diagnostics11010145. [PMID: 33478141 PMCID: PMC7835902 DOI: 10.3390/diagnostics11010145] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
The discovery of non-coding RNAs (ncRNAs) has opened a new paradigm to use ncRNAs as biomarkers to detect disease progression. Long non-coding RNAs (lncRNA) have garnered the most attention due to their specific cell-origin and their existence in biological fluids. Type 2 diabetes patients will develop cardiovascular disease (CVD) complications, and CVD remains the top risk factor for mortality. Understanding the lncRNA roles in T2D and CVD conditions will allow the future use of lncRNAs to detect CVD complications before the symptoms appear. This review aimed to discuss the roles of lncRNAs in T2D and CVD conditions and their diagnostic potential as molecular biomarkers for CVD complications in T2D.
Collapse
|
26
|
Jiang Y, Sun-Waterhouse D, Chen Y, Li F, Li D. Epigenetic mechanisms underlying the benefits of flavonoids in cardiovascular health and diseases: are long non-coding RNAs rising stars? Crit Rev Food Sci Nutr 2021; 62:3855-3872. [PMID: 33427492 DOI: 10.1080/10408398.2020.1870926] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cardiovascular diseases (CVDs) rank as the first leading cause of death globally. High dietary polyphenol (especially flavonoids) intake has strongly been associated with low incidence of the primary outcome, overall mortality, blood pressure, inflammatory biomarkers, onset of new-onset type 2 diabetes mellitus (T2DM), and obesity. Phytogenic flavonoids affect the physiological and pathological processes of CVDs by modulating various biochemical signaling pathways. Non-coding RNAs (ncRNAs) have attracted increasing attention as fundamental regulator of gene expression involved in CVDs. Among the different ncRNA subgroups, long ncRNAs (lncRNAs) have recently emerged as regulatory eukaryotic transcripts and therapeutic targets with important and diverse functions in health and diseases. lncRNAs may be associated with the initiation, development and progression of CVDs by modulating acute and chronic inflammation, adipogenesis and lipid metabolism, and cellular physiology. This review summarizes this research on the modulatory effects of lncRNAs and their roles in mediating cellular processes. The mechanisms of action of flavonoids underlying their therapeutic effects on CVDs are also discussed. Based on our review, flavonoids might facilitate a significant epigenetic modification as part (if not full) of their tissue-/cell-related biological effects. This finding may be attributed to their interaction with cellular signaling pathways involved in chronic diseases. Certain lncRNAs might be the target of specific flavonoids, and some critical signaling processes involved in the intervention of CVDs might mediate the therapeutic roles of flavonoids.
Collapse
Affiliation(s)
- Yang Jiang
- College of Food Science and Engineering, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Shandong Agricultural University, Taian, PR China
| | | | - Yilun Chen
- College of Food Science and Engineering, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Shandong Agricultural University, Taian, PR China
| | - Feng Li
- College of Food Science and Engineering, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Shandong Agricultural University, Taian, PR China
| | - Dapeng Li
- College of Food Science and Engineering, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Shandong Agricultural University, Taian, PR China
| |
Collapse
|
27
|
Li H, Shi H, Zhang F, Xue H, Wang L, Tian J, Xu J, Han Q. LncRNA Tincr regulates PKCɛ expression in a miR-31-5p-dependent manner in cardiomyocyte hypertrophy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:2495-2506. [PMID: 32157348 DOI: 10.1007/s00210-020-01847-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 02/28/2020] [Indexed: 12/16/2022]
Abstract
Cardiomyocyte hypertrophy is a fatal factor in heart disease resulting in heart failure and even mortality. Although many studies have been focusing on the pathogenesis of cardiomyocyte hypertrophy, the exact molecular mechanisms are still unexclusive. In this study, we first found that the expression level of lncRNA Tincr was significantly decreased in the myocardial tissues of TAC mouse models of cardiomyocyte hypertrophy, and this result was further confirmed in H9C2 cells, a widely used rat myoblast cell lines. More intriguingly, we demonstrated that the aberration of Tincr is essential to the pathogenesis of cardiomyocyte hypertrophy, indicated by the re-induction of Tincr improving the heart functions of hypertrophic mice. In mechanism, we identified miR-31-5p as a direct target of Tincr using a widely used online bioinformatics tool StarBase, and this result was further experimentally validated using dual-luciferase reporter assay and real-time PCR. Also, we identified PRKCE as a direct target of miR-31-5p, and loss function of miR-31-5p significantly blocks the positive regulatory effect of Tincr on PRKCE expression in H9C2 cells. The knockdown of Tincr resulted in increased cardiomyocyte size, and, however, inhibition of miR-31-5p or overexpression of PRKCE significantly reversed the increased cardiomyocyte size. Taken together, our study showed that a novel Tincr-miR-31-5p axis targeting PRKCE was involved in cardiomyocyte hypertrophy, indicating that it may provide potential therapy in cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Hao Li
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Hongtao Shi
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Fan Zhang
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Honghong Xue
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Lei Wang
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- The Affiliated Cardiovascular Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Jing Tian
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Jianrong Xu
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Qinghua Han
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China.
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China.
| |
Collapse
|
28
|
Wang M, Liu S, Wang H, Tang R, Chen Z. Morphine post-conditioning-induced up-regulation of lncRNA TINCR protects cardiomyocytes from ischemia-reperfusion injury via inhibiting degradation and ubiquitination of FGF1. QJM 2020; 113:859-869. [PMID: 32176291 DOI: 10.1093/qjmed/hcaa088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/24/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Our previous study has demonstrated that morphine post-conditioning (MpostC) protects cardiomyocytes from ischemia/reperfusion (I/R) injury partly through activating protein kinase-epsilon (PKCε) signaling pathway and subsequently inhibiting mitochondrial permeability transition pore (mPTP) opening. AIM In this study, we aim to investigate the relationship between long non-coding RNA TINCR and PKCε in cardiomyocytes under MpostC-treated I/R injury. DESIGN The myocardial I/R rat model was established by the ligation of lower anterior descending coronary artery for 45 min followed by the reperfusion for 1 h, and MpostC was performed before the reperfusion. METHOD H/R and MpostC were performed in the rat cardiomyocyte cell line (H9C2), and the Cytochrome-c release in cytosol and mPTP opening were determined. Cell viability was detected by using Cell Counting Kit-8, and cell apoptosis was determined by using flow cytometry or TUNEL assay. RESULTS The results indicated that MpostC restored the expression of TINCR in I/R rat myocardial tissues. In cardiomyocytes, the therapeutic effect of MpostC, including reduced mPTP opening, reduced Cytochrome-c expression, increased cell viability and reduced cell apoptosis, was dramatically negated by interfering TINCR. The expression of fibroblast growth factor 1 (FGF1), a protein that activates PKCε signaling pathway, was positively correlated with TINCR. The RNA immunoprecipitation and RNA pull-down assay further confirmed the binding between FGF1 and TINCR. Furthermore, TINCR was demonstrated to inhibit the degradation and ubiquitination of FGF1 in cardiomyocytes using the cycloheximide experiment and the ubiquitination assay. The TINCR/FGF1/PKCε axis was revealed to mediate the protective effect of MpostC against hypoxia/reoxygenation injury both in vitro and in vivo. CONCLUSION In conclusion, our findings demonstrated that MpostC-induced up-regulation of TINCR protects cardiomyocytes from I/R injury via inhibiting degradation and ubiquitination of FGF1, and subsequently activating PKCε signaling pathway, which provides a novel insight in the mechanism of TINCR and PKCε during MpostC treatment of I/R injury.
Collapse
Affiliation(s)
- M Wang
- Department of Anesthesiology, Qingdao Women and Children's Hospital, Shandong University, Qingdao, Shandong 266034, China
- Department of Anesthesiology, Weifang Medical University, Weifang, Shandong 261053, China
| | - S Liu
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - H Wang
- Department of Anesthesiology, Qingdao Women and Children's Hospital, Shandong University, Qingdao, Shandong 266034, China
| | - R Tang
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Z Chen
- Department of Anesthesiology, Qingdao Women and Children's Hospital, Shandong University, Qingdao, Shandong 266034, China
- Department of Anesthesiology, Qingdao Binhai University Affiliated Hospital, Qingdao, Shandong 266404, China
| |
Collapse
|
29
|
Zhang H, Liu B, Shi X, Sun X. Long noncoding RNAs: Potential therapeutic targets in cardiocerebrovascular diseases. Pharmacol Ther 2020; 221:107744. [PMID: 33181193 DOI: 10.1016/j.pharmthera.2020.107744] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
Cardiocerebrovascular disease is a collective term for cardiovascular and cerebrovascular diseases. Because of the complex mechanisms involved in cardiocerebrovascular diseases, limited effective treatments have been developed. With advancements in precision medicine, studies have focused on long noncoding RNAs (lncRNAs) in cerebrovascular diseases. LncRNAs, which are over 200 nucleotides long, regulate gene expression at epigenetic, transcriptional, and post-transcriptional levels. Moreover, lncRNAs play pivotal roles in the progression of cardiocerebrovascular diseases. For example, recent studies suggested that abnormal expression of lncRNAs are closely related to the occurrence and progression of these diseases. LncRNAs regulate gene expression by specifically binding to mRNA to modulate disease progression, serving as biomarkers for the diagnosis and prognosis of cardiocerebrovascular diseases. In this review, we discuss the roles, mechanisms, and clinical value of lncRNAs in cardiocerebrovascular diseases, providing a new perspective for the diagnosis and treatment of the diseases.
Collapse
Affiliation(s)
- Hao Zhang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Bo Liu
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| |
Collapse
|
30
|
Chen Y, Zhang Z, Zhu D, Zhao W, Li F. Knockdown of KCNQ1OT1 attenuates cardiac hypertrophy through modulation of the miR-2054/AKT3 axis. J Thorac Dis 2020; 12:4771-4780. [PMID: 33145050 PMCID: PMC7578479 DOI: 10.21037/jtd-20-203] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Background Persistent cardiac hypertrophy threatens health worldwide. Long non-coding RNAs (lncRNAs) attracted lots of attention in cardiac diseases such as cardiac hypertrophy. In this study, we aimed to study the function of KCNQ1OT1 in cardiac hypertrophy. Methods We first used qRT-PCR to detect the expression of KCNQ1OT1 in Ang-II-induced cardiomyocytes and mouse cardiac hypertrophy models. The function of KCNQ1OT1 was investigated by a loss-of-function test. Analysis of the luciferase reporter gene and RNA pulldown confirmed the interaction between KCNQ1OT1 and miR-2054. The target gene of miR-2054 was predicted by bioinformatics analysis and confirmed by luciferase reporter gene detection. Rescue experiments were performed to evaluate the role of miR-2054/AKT3 in the function of KCNQ1OT1. Results Our results suggested that KCNQ1OT1 was up-regulated in Ang-II-induced cardiomyocytes and transverse aortic constriction (TAC) mice. Knocking down of KCNQ1OT1 can reduce cell size and downregulate the expression of ANF, BNP and α-MHC in response to Ang-II. KCNQ1OT1 has been shown to compete competitively with miR-2054 and has a negative correlation with its expression. The combination of miR-2054 can reverse the effect of the KCNQ1OT1 combination in Ang-II-induced cardiomyocytes. In addition, AKT3 is a target of miR-2054 and mediates its effect on Ang-II-induced cardiomyocytes. Conclusions Knockdown of KCNQ1OT1 could attenuate cardiac hypertrophy through modulation of the miR-2054/AKT3 axis.
Collapse
Affiliation(s)
- Yiwei Chen
- Department of Cardiology, Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhifang Zhang
- Department of Cardiology, Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Diqi Zhu
- Department of Cardiology, Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenchuo Zhao
- Department of Cardiology, Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fen Li
- Department of Cardiology, Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Abstract
Gene expression is needed for the maintenance of heart function under normal conditions and in response to stress. Each cell type of the heart has a specific program controlling transcription. Different types of stress induce modifications of these programs and, if prolonged, can lead to altered cardiac phenotype and, eventually, to heart failure. The transcriptional status of a gene is regulated by the epigenome, a complex network of DNA and histone modifications. Until a few years ago, our understanding of the role of the epigenome in heart disease was limited to that played by histone deacetylation. But over the last decade, the consequences for the maintenance of homeostasis in the heart and for the development of cardiac hypertrophy of a number of other modifications, including DNA methylation and hydroxymethylation, histone methylation and acetylation, and changes in chromatin architecture, have become better understood. Indeed, it is now clear that many levels of regulation contribute to defining the epigenetic landscape required for correct cardiomyocyte function, and that their perturbation is responsible for cardiac hypertrophy and fibrosis. Here, we review these aspects and draw a picture of what epigenetic modification may imply at the therapeutic level for heart failure.
Collapse
Affiliation(s)
- Roberto Papait
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy; Humanitas Clinical Research Center-IRCCS, Rozzano, Italy; Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Italy; and National Research Council of Italy, Institute of Genetics and Biomedical Research, Milan Unit, Rozzano, Italy
| | - Simone Serio
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy; Humanitas Clinical Research Center-IRCCS, Rozzano, Italy; Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Italy; and National Research Council of Italy, Institute of Genetics and Biomedical Research, Milan Unit, Rozzano, Italy
| | - Gianluigi Condorelli
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy; Humanitas Clinical Research Center-IRCCS, Rozzano, Italy; Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Italy; and National Research Council of Italy, Institute of Genetics and Biomedical Research, Milan Unit, Rozzano, Italy
| |
Collapse
|
32
|
Cheng Z, Liu L, Li Q. lncRNA ZEB2-AS1 stimulates cardiac hypertrophy by downregulating PTEN. Exp Ther Med 2020; 20:92. [PMID: 32973941 DOI: 10.3892/etm.2020.9220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiac hypertrophy (CH) is closely related to a range of cardiovascular diseases, including heart failure and sudden cardiac death. The present study aimed to elucidate the role of long non-coding RNA (lncRNA) ZEB2 antisense RNA 1 (ZEB2-AS1) in regulating the hypertrophic process of cardiomyocytes and the potential underlying mechanism. An in vivo CH mouse model was established by performing transverse aortic constriction procedures. An in vitro CH model was established in primary cardiomyocytes isolated from mice by phenylephrine (PE) treatment. The relative protein levels of BNP, ANP and PTEN in cells with different groups (CH group and control group) were determined by western blotting. Relative expression levels of ZEB2-AS1, natriuretic peptide A (ANP) and brain natriuretic peptide (BNP) were determined in both in vivo and in vitro CH models. The regulatory effects of ZEB2-AS1/phosphatase and tensin homolog (PTEN) on cell surface area, and the relative expression levels of ANP and BNP were explored. ZEB2-AS1, ANP and BNP expression levels were increased in both in vivo and in vitro CH models compared with the sham and negative control groups, respectively. ZEB2-AS1 knockdown decreased cell surface area, and downregulated ANP and BNP expression levels in PE-treated primary cardiomyocytes. Similarly, PTEN overexpression reduced cell surface area, and downregulated ANP and BNP expression levels in PE-treated primary cardiomyocytes. Moreover, PTEN reversed the regulatory effects of ZEB2-AS1 on hypertrophic cardiomyocytes. Therefore, the present study suggested that lncRNA ZEB2-AS1 may influence the progression of CH by downregulating PTEN.
Collapse
Affiliation(s)
- Zhi Cheng
- Cardiovascular Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210003, P.R. China
| | - Lingyun Liu
- Cardiovascular Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210003, P.R. China
| | - Qingguo Li
- Cardiovascular Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210003, P.R. China
| |
Collapse
|
33
|
Liu L, Zhang D, Li Y. LncRNAs in cardiac hypertrophy: From basic science to clinical application. J Cell Mol Med 2020; 24:11638-11645. [PMID: 32896990 PMCID: PMC7579708 DOI: 10.1111/jcmm.15819] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/29/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac hypertrophy is a typical pathological phenotype of cardiomyopathy and a result from pathological remodelling of cardiomyocytes in humans. At present, emerging evidence demonstrated the roles of long non‐coding RNAs (lncRNAs) in regulating the pathophysiological process of cardiac hypertrophy. Herein, we would like to review the recent researches on this issue and try to analysis the potential therapeutic targets on lncRNA sites. Studies have revealed both genetic mutations related hypertrophic cardiomyopathy and the compensative cardiac hypertrophy due to pressure overload, inflammation, endocrine issues and other external stimulations, share a common molecular mechanism of ventricular hypertrophy. The emerging evidence identified the abnormal expression of lncRNAs would leading to the impairment the function of sarcomere, intracellular calcium handling and mitochondrial metabolisms. Several researches proved the therapeutic role of lncRNAs in preventing or reversing cardiac hypertrophy. With the development of delivery system for small pieces of oligonucleotide, clinicians could design gene therapy approaches to terminate the process of cardiac hypertrophy to provide better prognosis.
Collapse
Affiliation(s)
- Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Junho CVC, Caio-Silva W, Trentin-Sonoda M, Carneiro-Ramos MS. An Overview of the Role of Calcium/Calmodulin-Dependent Protein Kinase in Cardiorenal Syndrome. Front Physiol 2020; 11:735. [PMID: 32760284 PMCID: PMC7372084 DOI: 10.3389/fphys.2020.00735] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
Calcium/calmodulin-dependent protein kinases (CaMKs) are key regulators of calcium signaling in health and disease. CaMKII is the most abundant isoform in the heart; although classically described as a regulator of excitation–contraction coupling, recent studies show that it can also mediate inflammation in cardiovascular diseases (CVDs). Among CVDs, cardiorenal syndrome (CRS) represents a pressing issue to be addressed, considering the growing incidence of kidney diseases worldwide. In this review, we aimed to discuss the role of CaMK as an inflammatory mediator in heart and kidney interaction by conducting an extensive literature review using the database PubMed. Here, we summarize the role and regulating mechanisms of CaMKII present in several quality studies, providing a better understanding for future investigations of CamKII in CVDs. Surprisingly, despite the obvious importance of CaMKII in the heart, very little is known about CaMKII in CRS. In conclusion, more studies are necessary to further understand the role of CaMKII in CRS.
Collapse
Affiliation(s)
| | - Wellington Caio-Silva
- Center of Natural and Human Sciences (CCNH), Universidade Federal do ABC, Santo André, Brazil
| | - Mayra Trentin-Sonoda
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | | |
Collapse
|
35
|
|
36
|
Abstract
Cardiovascular disease is an enormous socioeconomic burden worldwide and remains a leading cause of mortality and disability despite significant efforts to improve treatments and personalize healthcare. Heart failure is the main manifestation of cardiovascular disease and has reached epidemic proportions. Heart failure follows a loss of cardiac homeostasis, which relies on a tight regulation of gene expression. This regulation is under the control of multiple types of RNA molecules, some encoding proteins (the so-called messenger RNAs) and others lacking protein-coding potential, named noncoding RNAs. In this review article, we aim to revisit the notion of regulatory RNA, which has been thus far mainly confined to noncoding RNA. Regulatory RNA, which we propose to abbreviate as regRNA, can include both protein-coding RNAs and noncoding RNAs, as long as they contribute, directly or indirectly, to the regulation of gene expression. We will address the regulation and functional role of messenger RNAs, microRNAs, long noncoding RNAs, and circular RNAs (ie, regRNAs) in heart failure. We will debate the utility of regRNAs to diagnose, prognosticate, and treat heart failure, and we will provide directions for future work.
Collapse
Affiliation(s)
| | - Blanche Schroen
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands (B.S., E.L.R., S.H.)
| | - Gabriela M. Kuster
- Clinic of Cardiology and Department of Biomedicine, University Hospital Basel and University of Basel, Switzerland (G.M.K.)
| | - Emma L. Robinson
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands (B.S., E.L.R., S.H.)
| | - Kerrie Ford
- Imperial College London, United Kingdom (K.F., C.E.)
| | - Iain B. Squire
- Department of Cardiovascular Sciences, University of Leicester, and NIHR Biomedical Research Centre, Glenfield Hospital, United Kingdom (I.B.S.)
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands (B.S., E.L.R., S.H.)
| | | | | | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, Strassen, Luxembourg (C.P.d.C.G., Y.D.)
| | - On behalf of the EU-CardioRNA COST Action (CA17129)
- Cardiovascular Research Unit, Luxembourg Institute of Health, Strassen, Luxembourg (C.P.d.C.G., Y.D.)
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands (B.S., E.L.R., S.H.)
- Clinic of Cardiology and Department of Biomedicine, University Hospital Basel and University of Basel, Switzerland (G.M.K.)
- Imperial College London, United Kingdom (K.F., C.E.)
- Department of Cardiovascular Sciences, University of Leicester, and NIHR Biomedical Research Centre, Glenfield Hospital, United Kingdom (I.B.S.)
- IRCCS Policlinico San Donato, Milan, Italy (F.M.)
| |
Collapse
|
37
|
He J, Luo Y, Song J, Tan T, Zhu H. Non-coding RNAs and Pathological Cardiac Hypertrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:231-245. [PMID: 32285415 DOI: 10.1007/978-981-15-1671-9_13] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cardiovascular disease (CVD) is a common disease which poses a serious threat to human health and it is characterized by high prevalence, high disability and high mortality. Myocardial hypertrophy (MH) is a common pathological process of various cardiovascular diseases and is considered as an independent risk factor for increased cardiovascular morbidity and mortality. Therefore, it is particularly important to understand its pathological mechanism and treatment. In recent years, it has been found that many non-coding RNAs (ncRNAs) play key regulatory roles in humans' various pathophysiological processes. Abnormal expression of ncRNAs in different types of cardiac cells is associated with pathological cardiac hypertrophy. Understanding the relationship between various ncRNAs and intercellular communication through extracellular vesicles (EV) can identify the key ncRNAs which are the accurate targets of precise therapy in this network of action, it also can potentially be a marker for clinical disease diagnosis, which will reflect the progress of the disease earlier and more accurately. There are many factors that regulate the occurrence and development of cardiac hypertrophy, ncRNAs are only a part of them. There are also mutual promotion or inhibition between ncRNAs and other molecules. It will be helpful for us to comprehend the mechanism of cardiac hypertrophy better and provide a sufficient theoretical basis for clinical diagnosis and treatment by defining these relationships.
Collapse
Affiliation(s)
- Jianfeng He
- Children's Hospital Chongqing Medical University, Chongqing, People's Republic of China
| | - Yanhong Luo
- Children's Hospital Chongqing Medical University, Chongqing, People's Republic of China
| | - Junxia Song
- Children's Hospital Chongqing Medical University, Chongqing, People's Republic of China
| | - Tao Tan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Hua Zhu
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
38
|
Jiang W, Agrawal DK, Boosani CS. Non-coding RNAs as Epigenetic Gene Regulators in Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:133-148. [PMID: 32285409 DOI: 10.1007/978-981-15-1671-9_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epigenetic gene regulations can be considered as de-novo initiation of abnormal molecular signaling events whose regulation is otherwise required during normal or specific developmental stages of the organisms. Primarily, three different mechanisms have been identified to participate in epigenetic gene regulations which include, DNA methylation, non-coding RNA species (microRNAs [miRNA], and long non-coding RNAs [LNC-RNA]) and histone modifications. These de-novo epigenetic mechanisms have been associated with altered normal cellular functions which eventually facilitate normal cells to transition into an abnormal phenotype. Among the three modes of regulation, RNA species which are usually considered to be less stable, can be speculated to initiate instant alterations in gene expression compared to DNA methylation or histone modifications. However, LNC-RNAs appear to be more stable in the cells than the other RNA species. Moreover, there is increasing literature which clearly suggests that a single specific LNC-RNA can regulate multiple mechanisms and disease phenotypes. With specific focus on cardiovascular diseases, here we attempt to provide UpToDate information on the functional role of miRNAs and LNC-RNAs. Here we discuss the role of these epigenetic mediators in different components of cardiovascular disease which include physiopathological heart development, athersclerosis, retenosis, diabetic hearts, myocardial infarction, ischemia-reperfusion, heart valve disease, aortic aneurysm, osteogenesis, angiogenesis and hypoxia in the heart. While there is abundant literature support that shows the involvement of many LNC-RNAs and miRNAs in cardiovascular diseases, very few RNA species have been identified which regulate epigenetic mechanisms which is the current focus in this article. Understanding the role of these RNA species in regulating epigenetic mechanisms in different cell types causing cardiovascular disease, would advance the field and promote disease prevention approaches that are aimed to target epigenetic mechanisms.
Collapse
Affiliation(s)
- Wanlin Jiang
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Devendra K Agrawal
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Chandra Shekhar Boosani
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
39
|
Yang X, Tao L, Zhu J, Zhang S. Long Noncoding RNA FTX Reduces Hypertrophy of Neonatal Mouse Cardiac Myocytes and Regulates the PTEN/PI3K/Akt Signaling Pathway by Sponging MicroRNA-22. Med Sci Monit 2019; 25:9609-9617. [PMID: 31840653 PMCID: PMC6929539 DOI: 10.12659/msm.919654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cardiac myocyte hypertrophy results from clinical conditions that include hypertension and valvular heart disease, and can result in heart failure. This study aimed to investigate the expression and role of the long noncoding RNA FTX (lnc-FTX), an X-inactive-specific transcript (XIST) regulator transcribed from the X chromosome, in hypertrophy of neonatal mouse cardiac myocytes induced by angiotensin II (Ang II) in vitro. MATERIAL AND METHODS Cardiac myocytes were isolated from neonatal mice and cultured with and without Ang II. Immunofluorescence, with localization of an antibody to alpha-smooth muscle actin (alpha-SMA), was used to identify the neonatal mouse cardiac myocytes. Quantitative real-time polymerase chain reaction (qRT-PCR) measured gene expression levels. The cell counting kit-8 (CCK-8) assay was used to determine cell viability, and Western blot measured protein expression. StarBase v2.0 bioinformatics software was used for target gene prediction and was confirmed with the luciferase reporter assay. RESULTS The expression of lnc-FTX was reduced in mouse cardiac myocytes treated with Ang II. Overexpression of lnc-FTX reduced cell apoptosis, cardiomyocyte contractility, and the expression of c-Jun, A-type natriuretic peptide (ANP), and B-type natriuretic peptide (BNP) induced by Ang II. The target of lnc-FTX was micro-RNA 22 (miRNA-22). The mechanism of action of lnc-FTX in neonatal mouse cardiac myocytes was through suppression of the PI3K/Akt signaling pathway by promoting the release of PTEN by sponging miRNA-22. CONCLUSIONS The expression of lnc-FTX was associated with reduced hypertrophy of neonatal mouse cardiac myocytes and regulated the PTEN/PI3K/Akt signaling pathway by sponging miRNA-22.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China (mainland)
| | - Lichan Tao
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China (mainland)
| | - Jin Zhu
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China (mainland)
| | - Sheng Zhang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China (mainland)
| |
Collapse
|
40
|
Zhang Y, Du W, Yang B. Long non-coding RNAs as new regulators of cardiac electrophysiology and arrhythmias: Molecular mechanisms, therapeutic implications and challenges. Pharmacol Ther 2019; 203:107389. [DOI: 10.1016/j.pharmthera.2019.06.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
|
41
|
Luo X, He S, Hu Y, Liu J, Chen X. Sp1-induced LncRNA CTBP1-AS2 is a novel regulator in cardiomyocyte hypertrophy by interacting with FUS to stabilize TLR4. Cardiovasc Pathol 2019; 42:21-29. [PMID: 31220774 DOI: 10.1016/j.carpath.2019.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/19/2019] [Accepted: 04/30/2019] [Indexed: 02/05/2023] Open
Abstract
Cardiomyocyte hypertrophy is a heart response adapting to increasing cardiac load. Prolonged cardiomyocyte hypertrophy indicates a higher risk of heart failure or even cardiac death. Long noncoding RNAs have been largely reported to modulate human diseases. CTPB1-AS2 is a newly discovered lncRNA reported as an oncogene in papillary thyroid cancer, but its function in cardiomyocyte hypertrophy has never been probed. Toll-like receptor 4 (TLR4) is recognized to play important roles in cardiomyocyte hypertrophy. The present study aimed to investigate the role of CTBP1-AS2 in cardiomyocyte hypertrophy. First, we discovered the low expression of CTBP1-AS2 in normal heart tissues in GETx database. Then, we established cardiomyocyte hypertrophy models on mice and cardiomyocytes through transverse aortic constriction surgery and Ang II treatment. We revealed the up-regulation of CTBP1-AS2 and TLR4 in cardiomyocyte hypertrophy models. Also, we confirmed the positive correlation between CTBP1-AS2 and TLR4 expressions in cardiomyocyte hypertrophy tissues. Loss-of-function assays confirmed that inhibiting CTBP1-AS2 attenuated the Ang II-induced cardiomyocyte hypertrophy. Mechanism research showed that CTBP1-AS2 stabilized TLR4 mRNA by recruiting FUS. Rescue assays certified that CTBP1-AS2 regulated cardiomyocyte hypertrophy through TLR4. Finally, we found Sp1 as an upstream activator for CTBP1-AS2 expression. In conclusion, our study uncovered CTBP1-AS2 as a novel regulator of cardiomyocyte hypertrophy through regulating TLR4, providing a new potential treatment target for cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Xiaojia Luo
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Cardiovascular Medicine, Chengdu Second People's Hospital, Chengdu, Sichuan, 610017, China
| | - Sen He
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yongmei Hu
- Department of Cardiovascular Medicine, Chengdu Second People's Hospital, Chengdu, Sichuan, 610017, China
| | - Jianxiong Liu
- Department of Cardiovascular Medicine, Chengdu Second People's Hospital, Chengdu, Sichuan, 610017, China
| | - Xiaoping Chen
- Department of Cardiovascular Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
42
|
Meng L, Lin H, Zhang J, Lin N, Sun Z, Gao F, Luo H, Ni T, Luo W, Chi J, Guo H. Doxorubicin induces cardiomyocyte pyroptosis via the TINCR-mediated posttranscriptional stabilization of NLR family pyrin domain containing 3. J Mol Cell Cardiol 2019; 136:15-26. [PMID: 31445005 DOI: 10.1016/j.yjmcc.2019.08.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/17/2019] [Accepted: 08/19/2019] [Indexed: 02/09/2023]
Abstract
AIMS Doxorubicin (DOX), a widely used powerful chemotherapeutic component for cancer treatment, can give rise to severe cardiotoxicity that limits its clinical use. Pyroptosis is characterized by proinflammation and has been defined as a new type of programmed cell death in recent years. However, whether the DOX-induced cardiotoxicity is related to pyroptosis, and if so, which genes are involved in this process is largely unknown. In this study, we sought to identify the effect of DOX on cardiomyocyte pyroptosis and further reveal the underlying regulatory mechanism. METHODS AND RESULTS In vitro and in vivo experiments showed that DOX treatment induced cardiomyocyte pyroptosis as evidenced by increased cell death and upregulated expression levels of NLR family pyrin domain containing 3 (NLRP3), caspase-3, IL-1β, IL-18 and GMDSD-N. Inhibition of NLRP3 rescued the DOX-induced pyroptosis. qRT-PCR showed that TINCR lncRNA was upregulated by DOX treatment and knockdown of TINCR reversed the DOX-induced pyroptosis both in vitro and in vivo. Mechanistic investigations revealed that TINCR increased NLRP3 level via recruiting IGF2BP1 to enhance NLRP3 mRNA. And the effect of TINCR on cardiomyocyte pyroptosis was attenuated by the inhibition of NLRP3 or IGF2BP1. Finally, TINCR was not involved in DOX-induced pyroptosis in cancer cells. CONCLUSION TINCR mediates the DOX-induced cardiotoxicity and pyroptosis in an IGF2BP1-dependent manner. Therefore, TINCR may serve as a promising therapeutic target to overcome the cardiotoxicity of chemotherapy for cancer therapy.
Collapse
Affiliation(s)
- Liping Meng
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Jie Zhang
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China; The First Clinical Medical College, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Na Lin
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Zhenzhu Sun
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China; The First Clinical Medical College, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Feidan Gao
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Hangqi Luo
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Tingjuan Ni
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Wenqiang Luo
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Jufang Chi
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China.
| | - Hangyuan Guo
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China; The First Clinical Medical College, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| |
Collapse
|
43
|
Han X, Zhang J, Liu Y, Fan X, Ai S, Luo Y, Li X, Jin H, Luo S, Zheng H, Yue Y, Chang Z, Yang Z, Tang F, He A, Shen X. The lncRNA Hand2os1/ Uph locus orchestrates heart development through regulation of precise expression of Hand2. Development 2019; 146:146/13/dev176198. [PMID: 31273086 DOI: 10.1242/dev.176198] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/09/2019] [Indexed: 12/30/2022]
Abstract
Exploration and dissection of potential actions and effects of long noncoding RNA (lncRNA) in animals remain challenging. Here, using multiple knockout mouse models and single cell RNA sequencing, we demonstrate that the divergent lncRNA Hand2os1/Uph has a key complex modulatory effect on the expression of its neighboring gene HAND2 and subsequently on heart development and function. Short deletion of the Hand2os1 promoter in mouse diminishes Hand2os1 transcription to ∼8-32%, but fails to affect HAND2 expression and yields no discernable heart phenotypes. Interestingly, full-length deletion of Hand2os1 in mouse causes moderate yet prevalent upregulation of HAND2 in hundreds of cardiac cells, leading to profound biological consequences, including dysregulated cardiac gene programs, congenital heart defects and perinatal lethality. We propose that the Hand2os1 locus dampens HAND2 expression to restrain cardiomyocyte proliferation, thereby orchestrating a balanced development of cardiac cell lineages. This study highlights the regulatory complexity of the lncRNA Hand2os1 on HAND2 expression, emphasizing the need for complementary genetic and single cell approaches to delineate the function and primary molecular effects of an lncRNA in animals.
Collapse
Affiliation(s)
- Xue Han
- Tsinghua Center for Life Sciences, School of Medicine, and School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jiejie Zhang
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Yaxi Liu
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Xiaoying Fan
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing 100871, China
| | - Shanshan Ai
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Yingjie Luo
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Xin Li
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Hengwei Jin
- Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Sai Luo
- Tsinghua Center for Life Sciences, School of Medicine, and School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hui Zheng
- Tsinghua Center for Life Sciences, School of Medicine, and School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanzhu Yue
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Zai Chang
- Tsinghua Center for Life Sciences, School of Medicine, and School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhongzhou Yang
- Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics, College of Life Sciences, Peking University, Beijing 100871, China
| | - Aibin He
- Peking Center for Life Sciences, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Xiaohua Shen
- Tsinghua Center for Life Sciences, School of Medicine, and School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
44
|
Popa-Fotea NM, Micheu MM, Bataila V, Scafa-Udriste A, Dorobantu L, Scarlatescu AI, Zamfir D, Stoian M, Onciul S, Dorobantu M. Exploring the Continuum of Hypertrophic Cardiomyopathy-From DNA to Clinical Expression. ACTA ACUST UNITED AC 2019; 55:medicina55060299. [PMID: 31234582 PMCID: PMC6630598 DOI: 10.3390/medicina55060299] [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: 03/23/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 12/29/2022]
Abstract
The concepts underlying hypertrophic cardiomyopathy (HCM) pathogenesis have evolved greatly over the last 60 years since the pioneering work of the British pathologist Donald Teare, presenting the autopsy findings of “asymmetric hypertrophy of the heart in young adults”. Advances in human genome analysis and cardiac imaging techniques have enriched our understanding of the complex architecture of the malady and shaped the way we perceive the illness continuum. Presently, HCM is acknowledged as “a disease of the sarcomere”, where the relationship between genotype and phenotype is not straightforward but subject to various genetic and nongenetic influences. The focus of this review is to discuss key aspects related to molecular mechanisms and imaging aspects that have prompted genotype–phenotype correlations, which will hopefully empower patient-tailored health interventions.
Collapse
Affiliation(s)
- Nicoleta Monica Popa-Fotea
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Vlad Bataila
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Alexandru Scafa-Udriste
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
- Department 4-Cardiothoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania.
| | - Lucian Dorobantu
- Cardiomyopathy Center, Monza Hospital, Tony Bulandra Street 27, 021968 Bucharest, Romania.
| | - Alina Ioana Scarlatescu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Diana Zamfir
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Monica Stoian
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
| | - Sebastian Onciul
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
- Department 4-Cardiothoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania.
| | - Maria Dorobantu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Floreasca Street 8, 014461 Bucharest, Romania.
- Department 4-Cardiothoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania.
| |
Collapse
|
45
|
Zhang Q, Wang F, Wang F, Wu N. Long noncoding RNA MAGI1-IT1 regulates cardiac hypertrophy by modulating miR-302e/DKK1/Wnt/beta-catenin signaling pathway. J Cell Physiol 2019; 235:245-253. [PMID: 31222747 DOI: 10.1002/jcp.28964] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/24/2019] [Accepted: 05/28/2019] [Indexed: 01/05/2023]
Abstract
Cardiac hypertrophy (CH) is an adaptive cardiac response to overload whose decompensation eventually leads to heart failure or sudden death. Recently, accumulating studies have indicated the implication of long noncoding RNAs (lncRNAs) in CH progression. MAGI1-IT1 is a newly-identified lncRNA that is highly associated with CH, while its specific role in CH progression remains masked. In this study, we uncovered that MAGI1-IT1 was distinctly downregulated in angiotensin (Ang) II-induced hypertrophic H9c2 cells. Also, MAGI1-IT1 overexpression in Ang II-treated H9c2 cells strikingly abolished the enlarged surface area and the enhanced levels of hypertrophic markers such as ANP, BNP, and β-MHC. Mechanically, we found MAGI1-IT1 sponged miR-302e which was identified as a hypertrophy-facilitator here, and that miR-302e upregulation countervailed the inhibition of MAGI1-IT1 overexpression on hypertrophic cells. Moreover, it was confirmed that MAGI1-IT1 boosted DKK1 expression by absorbing miR-302e. Subsequently, we also illustrated that MAGI1-IT1 inactivated Wnt/beta-catenin signaling through a DKK1-dependent pathway. Finally, both the DKK1 inhibition and LiCI (Wnt activator) supplement abrogated the hypertrophy-suppressive impact of MAGI1-IT1 on Ang II-simulated hypertrophic H9c2 cells. Jointly, our findings disclosed that MAGI1-IT1 functioned as a negative regulator in CH through inactivating Wnt/beta-catenin pathway via targeting miR-302e/DKK1 axis, revealing a novel road for CH treatment.
Collapse
Affiliation(s)
- Qinghua Zhang
- Department of Cardiovascular Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fengshuang Wang
- Pharmacy Intravenous Admixture Services, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fenghua Wang
- Medical Services Section, the 1st Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Naishi Wu
- Department of Cardiovascular Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
46
|
Zou X, Wang J, Tang L, Wen Q. LncRNA TUG1 contributes to cardiac hypertrophy via regulating miR-29b-3p. In Vitro Cell Dev Biol Anim 2019; 55:482-490. [PMID: 31183682 DOI: 10.1007/s11626-019-00368-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/05/2019] [Indexed: 12/19/2022]
Abstract
Cardiac hypertrophy with maladjusted cardiac remodeling is the leading cause of heart failure. In the past decades, long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) have been proved to exert multiple functions in cellular biological behaviors; however, their role in cardiac hypertrophy remains largely unclear. Presently, we first obtained hypertrophic H9c2 cells by treating with angiotensin II (Ang II) and uncovered upregulation of lncRNA taurine upregulated gene 1 (TUG1) in such H9c2 cells. Then, we demonstrated that silencing TUG1 attenuated Ang II-induced cardiac hypertrophy. Besides, a strong interactivity of TUG1 with miR-29b-3p at the putative sites was validated, suggesting that TUG1 was an endogenous sponge of miR-29b-3p in H9c2 cells. Additionally, the expression of miR-29b-3p was strikingly reduced by TUG1 upregulation and also inhibited under Ang II treatment, whereas it was restored after silencing TUG1 in hypertrophic cells. Also, we proved miR-29b-3p as a negative regulator in cardiac hypertrophy. Finally, miR-29b-3p inhibition abolished the anti-hypertrophy effect of TUG1 depletion in Ang II-treated H9c2 cells. Collectively, our findings confirmed that TUG1 functioned as a positive modulator of cardiac hypertrophy via sponging miR-29b-3p, indicating that TUG1 might serve as a potential target for the treatment of cardiac hypertrophy and even heart failure.
Collapse
Affiliation(s)
- Xue Zou
- Department of Cardiology, Daping Hospital, The Third Military Medical University, No.10 Changjiangzhilu, Daping, Yuzhong District, Chongqing, 40042, People's Republic of China
| | - Jialiang Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, No.10 Changjiangzhilu, Daping, Yuzhong District, Chongqing, 40042, People's Republic of China
| | - Li Tang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, No.10 Changjiangzhilu, Daping, Yuzhong District, Chongqing, 40042, People's Republic of China
| | - Qian Wen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, No.10 Changjiangzhilu, Daping, Yuzhong District, Chongqing, 40042, People's Republic of China.
| |
Collapse
|
47
|
Deng Y, Chen D, Wang L, Gao F, Jin B, Lv H, Zhang G, Sun X, Liu L, Mo D, Ma N, Song L, Huo X, Yan T, Miao Z. Silencing of Long Noncoding RNA Nespas Aggravates Microglial Cell Death and Neuroinflammation in Ischemic Stroke. Stroke 2019; 50:1850-1858. [PMID: 31167620 PMCID: PMC6594728 DOI: 10.1161/strokeaha.118.023376] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Ischemic stroke is one of the leading causes of morbidity and mortality worldwide and a major cause of long-term disability. Recently, long noncoding RNAs have been revealed, which are tightly associated with several human diseases. However, the functions of long noncoding RNAs in ischemic stroke still remain largely unknown. In the current study, for the first time, we investigated the role of long noncoding RNA Nespas in ischemic stroke. Methods- We used in vivo models of middle cerebral artery occlusion and in vitro models of oxygen-glucose deprivation to illustrate the effect of long noncoding RNA Nespas on ischemic stroke. Results- We found expression of Nespas was significantly increased in ischemic cerebral tissues and oxygen-glucose deprivation-treated BV2 cells in a time-dependent manner. Silencing of Nespas aggravated middle cerebral artery occlusion operation-induced IR injury and cell death. In addition, proinflammatory cytokine production and NF-κB (nuclear factor-κB) signaling activation were inhibited by Nespas overexpression. TAK1 (transforming growth factor-β-activated kinase 1) was found to directly interact with Nespas, and TAK1 activation was significantly suppressed by Nespas. At last, we found Nespas-inhibited TRIM8 (tripartite motif 8)-induced K63-linked polyubiquitination of TAK1. Conclusions- We showed that Nespas played anti-inflammatory and antiapoptotic roles in cultured microglial cells after oxygen-glucose deprivation stimulation and in mice after ischemic stroke by inhibiting TRIM8-related K63-linked polyubiquitination of TAK1.
Collapse
Affiliation(s)
- Yiming Deng
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,China National Clinical Research Center for Neurological Diseases (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.)
| | - Duanduan Chen
- From the School of Life Science (D.C., T.Y.), Beijing Institute of Technology, China.,Key Laboratory of Convergence Medical Engineering System and Healthcare Technology, Ministry of Industry and Information Technology (D.C., T.Y.), Beijing Institute of Technology, China
| | - Luyao Wang
- Intelligent Robotics Institute, School of Mechatronical Engineering (L.W.), Beijing Institute of Technology, China
| | - Feng Gao
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,China National Clinical Research Center for Neurological Diseases (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.)
| | - Bo Jin
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, China (B.J., H.L., G.Z.)
| | - Hong Lv
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, China (B.J., H.L., G.Z.)
| | - Guojun Zhang
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, China (B.J., H.L., G.Z.)
| | - Xuan Sun
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,China National Clinical Research Center for Neurological Diseases (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.)
| | - Lian Liu
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,China National Clinical Research Center for Neurological Diseases (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.)
| | - Dapeng Mo
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,China National Clinical Research Center for Neurological Diseases (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.)
| | - Ning Ma
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,China National Clinical Research Center for Neurological Diseases (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.)
| | - Ligang Song
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,China National Clinical Research Center for Neurological Diseases (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.)
| | - Xiaochuan Huo
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,China National Clinical Research Center for Neurological Diseases (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.)
| | - Tianyi Yan
- From the School of Life Science (D.C., T.Y.), Beijing Institute of Technology, China.,Key Laboratory of Convergence Medical Engineering System and Healthcare Technology, Ministry of Industry and Information Technology (D.C., T.Y.), Beijing Institute of Technology, China
| | - Zhongrong Miao
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,China National Clinical Research Center for Neurological Diseases (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.D., F.G., X.S., L.L., D.M., N.M., L.S., X.H., Z.M.)
| |
Collapse
|
48
|
Jing L, Li S, Wang J, Zhang G. Long non‐coding RNA small nucleolar RNA host gene 7 facilitates cardiac hypertrophy via stabilization of SDA1 domain containing 1 mRNA. J Cell Biochem 2019; 120:15089-15097. [PMID: 31026094 DOI: 10.1002/jcb.28770] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Ling Jing
- Department of Cardiology The First Affiliated Hospital of Harbin Medical University Harbin Heilongjiang China
| | - Shuang Li
- Department of Cardiology The First Affiliated Hospital of Harbin Medical University Harbin Heilongjiang China
| | - Jingyao Wang
- Cardiology Internal Medicine Major Graduate School of Harbin Medical University Harbin Heilongjiang China
| | - Guowei Zhang
- Department of Cardiovascular Surgery The First Affiliated Hospital of Harbin Medical University Harbin Heilongjiang China
| |
Collapse
|
49
|
Qin G, Song Y, Guo Y, Sun Y, Zeng W. LincRNA TINCR facilitates excessive proliferation and inflammation in post-burn skin fibroblasts by directly binding with SND1 protein and inducing SND1-mediated TGF-β1 expression. Biochem Biophys Res Commun 2019; 509:903-910. [DOI: 10.1016/j.bbrc.2019.01.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 01/04/2019] [Indexed: 10/27/2022]
|
50
|
Chen Y, Tan S, Liu M, Li J. LncRNA TINCR is downregulated in diabetic cardiomyopathy and relates to cardiomyocyte apoptosis. SCAND CARDIOVASC J 2019; 52:335-339. [PMID: 30453794 DOI: 10.1080/14017431.2018.1546896] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Yongheng Chen
- Department of Cardiovascular Internal Medicine, the First Affiliated Hospital of Changsha Medical University, Changsha City, P.R. China
| | - Shan Tan
- Department of Cardiovascular Internal Medicine, the First Affiliated Hospital of Changsha Medical University, Changsha City, P.R. China
| | - Mei Liu
- Department of Cardiovascular Internal Medicine, the First Affiliated Hospital of Changsha Medical University, Changsha City, P.R. China
| | - Jianming Li
- Department of Anatomy, Changsha Medical University, Changsha City, P.R. China
| |
Collapse
|