1
|
Hussen BM, Othman DI, Abdullah SR, Khudhur ZO, Samsami M, Taheri M. New insights of LncRNAs fingerprints in breast cancer progression: Tumorigenesis, drug resistance, and therapeutic opportunities. Int J Biol Macromol 2025; 287:138589. [PMID: 39662549 DOI: 10.1016/j.ijbiomac.2024.138589] [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: 11/11/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024]
Abstract
Breast cancer (BC) is one of the common female cancers and it is characterized by considerable problems regarding its development and therapy. Long non-coding RNAs (lncRNAs) have been identified as significant modulators in BC development, especially, in tumorigenicity and chemoresistance. We therefore endeavor to present an up-to-date understanding of lncRNAs and their impact on BC progression and treatment, concerning molecular processes, treatment options, and use as a therapeutic opportunity. LncRNAs are novel regulators of genes that cause therapeutic resistance and directly impact the functioning of both coding and non-coding genes in BC patients, but little is known about their mechanisms of actions. Thus, additional study is required to have a deeper understanding of their modes of action and possible roles in BC disease. This study aims to investigate the functions of lncRNAs in the development of BC, with particular attention to their role in tumorigenesis, drug resistance mechanisms, and therapeutic targets. This will help to identify novel therapeutic targets and improve the effectiveness of BC treatment.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq; Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Diyar Idris Othman
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Snur Rasool Abdullah
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Zhikal Omar Khudhur
- Department of Biology, Faculty of Education, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Majid Samsami
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany; Urology and Nephrology Research Center, Research Institute for Urology and Nephrology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Zhang Z, Zhu D, Shi P, Wu J, Li F, Chen Y. LncRNA XIST knockdown reduces myocardial damage in myocarditis by targeting the miR-140-3p/RIPK1 axis. Biotechnol Genet Eng Rev 2024; 40:1425-1437. [PMID: 36971142 DOI: 10.1080/02648725.2023.2194074] [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: 02/13/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023]
Abstract
Viral myocarditis (MC) is caused by Coxsackie virus B3 (CVB3)-induced cardiomyocyte apoptosis and inflammation, and changes in miRNA and lncRNA are linked to cardiac remodeling. The long non-coding RNA XIST (XIST) has been identified as a regulator in various pathological processes in heart diseases, but its role in CVB3-induced MC is not well understood. This research aimed to evaluate the impact that XIST has on CVB3-induced MC as well as the mechanism behind this effect. XIST expression in CVB3-exposed H9c2 cells (H9c2 cells) was evaluated by qRT-PCR. In CVB3-exposed H9c2 cells, reactive oxygen species production, inflammatory mediators, and apoptosis were experimentally observed. An investigation into and confirmation of the existence of an interaction involving XIST, miR-140-3p, and RIPK1 were carried out. The findings showed that CVB3 induced upregulation of XIST in H9c2 cells. However, XIST knockdown reduced oxidative stress, inflammation, and apoptosis of CVB3-exposed H9c2 cells. XIST was specifically bound to miR-140-3p, and there was mutual negative regulation between the two. Moreover, XIST downregulated RIPK1, which was mediated by miR-140-3p. The study suggests that downregulating XIST can alleviate inflammatory injury in CVB3-exposed H9c2 cells through the miR-140-3p/RIPK1 axis. These findings provide novel insights into the underlying mechanisms of MC.
Collapse
Affiliation(s)
- 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
| | - Ping Shi
- Department of Cardiology, Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jinjin Wu
- 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
| | - Yiwei Chen
- Department of Cardiology, Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Chen P, Yawar W, Farooqui AR, Ali S, Lathiya N, Ghous Z, Sultan R, Alhomrani M, Alghamdi SA, Almalki AA, Alghamdi AA, ALSuhaymi N, Razi Ul Islam Hashmi M, Hameed Y. Transcriptomics data integration and analysis to uncover hallmark genes in hypertrophic cardiomyopathy. Am J Transl Res 2024; 16:637-653. [PMID: 38463581 PMCID: PMC10918138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/24/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Hypertrophic cardiomyopathy (HCM) is a heterogeneous disease that mainly affects the myocardium. In the current study, we aim to explore HCM-related hub genes through the analysis of differentially expressed genes (DEGs) between HCM and normal sample groups. METHODS The GSE68316 and GSE36961 expression profiles were obtained from the Gene Expression Omnibus (GEO) database for the identification of DEGs, to explore hub genes, and to perform their expression analysis. Clinical HCM and control tissue samples were taken for expression and promoter methylation validation analysis via RNA-sequencing (RNA-seq) and targeted bisulfite sequencing (bisulfite-seq) analyses. Then, other different bioinformatics tools were employed to perform STRING, lncRNA-miRNA-mRNA regulatory networks, gene enrichment, and drug prediction analyses. RESULTS In total, the top 20 DEGs, including 10 up-regulated and 10 down-regulated, were obtained from GSE68316. Out of the 20 DEGs, we subsequently identified the 8 most important hub genes including 5 up-regulated genes (EPB42, UQCRH, CA1, PFDN5, and LSM5) and 3 down-regulated genes (RPS24, TNS1, and RPL26). Expression and promoter methylation dysregulation of these genes were further validated on clinical HCM samples paired with controls. Next, we further investigated hub genes' regulatory 6 miRNAs (has-mir-1-3p, has-mir-129-5p, has-mir-16-5p, has-mir-23b-3p, has-mir-27-3p, and has-mir-182-5p) and miRNAs regulatory 4 lncRNAs (NUTMB2-AS1, NEAT1, XIST, and GABPB1-AS1) in this study via the lncRNA-cricRNA-miRNA-mRNA regulatory network. Later on, gene enrichment analysis revealed that hub genes were enriched in various important pathways including Nitrogen metabolism, Ribosome, RNA degradation, Cardiac muscle contraction, and Coronavirus disease, etc. Finally, the drug prediction analysis highlighted different potential candidate drugs for altering the expression of hub genes in the treatment of HCM. CONCLUSION In summary, the identification of key hub genes and their enrichment analysis in the current study may shed light on the mechanisms behind the occurrence and development of HCM.
Collapse
Affiliation(s)
- Peng Chen
- Department of Cardiovascular Medicine, Taiyuan Central HospitalTaiyuan 030000, Shanxi, China
| | - Warda Yawar
- Department of Emergency, PPHISindh, Karachi 74800, Pakistan
| | | | - Saqib Ali
- Department of Computer Science, University of AgricultureFaisalabad 38040, Pakistan
| | - Nida Lathiya
- Department of Physiology, Jinnah Medical and Dental College, Sohail UniversityKarachi 74800, Pakistan
| | - Zeeshan Ghous
- Department of Cardiology, Punjab Institute of CardiologyLahore 54000, Pakistan
| | - Rizwana Sultan
- Department of Pathology, Faculty of Veterinary and Animal Sciences, Cholistan University of Veterinary and Animal SciencesBahawalpur, Pakistan
| | - Majid Alhomrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
- Research Centre for Health Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Saleh A Alghamdi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Abdulraheem Ali Almalki
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Ahmad A Alghamdi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Naif ALSuhaymi
- Department of Emergency Medical Services, Faculty of Health Sciences - AlQunfudah, Umm Al-Qura UniversityMekkah, Saudi Arabia
| | | | - Yasir Hameed
- Department of Biotechnology, Institute of Biochemistry Biotechnology and Bioinformatics, The Islamia University of BahawalpurBahawalpur 63100, Pakistan
| |
Collapse
|
4
|
Dysfunctional network of hub genes in hypertrophic cardiomyopathy patients. Am J Transl Res 2022; 14:8918-8933. [PMID: 36628247 PMCID: PMC9827312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/15/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Considering it is one of the major causes of sudden cardiac arrest, the proper management of hypertrophic cardiomyopathy (HCM) is essential. However, efficient treatment options for this disease are still lacking. The discovery of HCM-associated hub genes may help in diagnosis and offer a reliable tool for developing effective therapeutic strategies. METHODS We examined HCM-based gene expression datasets (GSE36961) from the Gene Expression Omnibus (GEO) database for the identification of differentially expressed genes (DEGs), PPI network development, module screening, and shortlisting of hub genes via GEOR2, STRING, and Cytoscape. Moreover, we also used another HCM-based gene expression dataset (GSE32453) for the expression validation of hub genes. Following this, we constructed the lncRNA-cricRNA-miRNA-mRNA regulatory network after retrieving information from the miRTarBase, miRDB, and MiRcode databases. Finally, we used DAVID to perform functional and pathway analysis of the hub genes. RESULTS From GSE36961, a total of the 262 most significant DEGs, including 162 down-regulated and 76 up-regulated, were identified between HCM patients and normal individuals. Among these DEGs, a total of 10 significantly down-regulated DEGs, including cluster of differentiation 14 (CD14), beta2 Integrin Gene (ITGB2), C1q subcomponent subunit B (C1QB), Cluster of Differentiation 163 (CD163), Hematopoietic Cell-Specific Lyn Substrate 1 (HCLS1), Arachidonate 5-Lipoxygenase Activating Protein (ALOX5AP), Pleckstrin (PLEK), Complement C1q C Chain (C1QC), Fc fragment Of IgE receptor Ig (FCER1G), and tyrosine kinase binding protein (TYROBP), were shortlisted as the hub genes. Pathway enrichment analysis showed that the identified hub genes were involved in the dysregulation of some diverse pathways in HCM patients. Such as, Pertussis, Complement and coagulation cascade, Legnionellosis, Asthma, Staphylococcus aureus infection, etc. Lastly, we also explored hub genes' regulatory 2 MicroRNAs (miRNAs, has-mir-7-5p and has-mir-27a-3p), one Long non-coding RNAs (lncRNA, OIP5-AS1-201), and one Circular RNA (cricRNA, CDR1as) via lncRNA-cricRNA-miRNA-mRNA regulatory network. CONCLUSION Our study revealed that ten hub genes (CD14, ITGB2, C1QB, CD163, HCLS1, ALOX5AP, PLEK, C1QC, FCER1G, and TYROBP) are involved in the development and progression of HCM. These genes can potentially be used as biomarkers and therapeutic targets for HCM patients.
Collapse
|
5
|
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
|
6
|
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
|
7
|
Chen JY, Xie ZX, Dai JZ, Han JY, Wang K, Lu LH, Jin JJ, Xue SJ. Reconstruction and analysis of potential biomarkers for hypertrophic cardiomyopathy based on a competing endogenous RNA network. BMC Cardiovasc Disord 2022; 22:422. [PMID: 36138345 PMCID: PMC9503253 DOI: 10.1186/s12872-022-02862-9] [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: 06/17/2022] [Accepted: 09/15/2022] [Indexed: 11/26/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a common heritable cardiomyopath. Although considerable effort has been made to understand the pathogenesis of HCM, the mechanism of how long noncoding RNA (lncRNA)-associated competing endogenous RNA (ceRNA) network result in HCM remains unknown. In this study, we acquired a total of 520 different expression profiles of lncRNAs (DElncRNAs) and 371 messenger RNAs (mRNA, DEGs) by microarray and 33 microRNAs (DEmiRNAs) by sequencing in plasma of patients with HCM and healthy controls. Then lncRNA–miRNA pairs were predicted using miRcode and starBase and crossed with DEmiRNAs. MiRNA–mRNA pairs were retrieved from miRanda and TargetScan and crossed with DEGs. Combined with these pairs, the ceRNA network with eight lncRNAs, three miRNAs, and 22 mRNAs was constructed. lncRNA RP11-66N24.4 and LINC00310 were among the top 10% nodes. The hub nodes were analyzed to reconstruct a subnetwork. Furthermore, quantitative real-time polymerase chain reaction results showed that LINC00310 was significantly decreased in patients with HCM. For LINC00310, GO analysis revealed that biological processes were enriched in cardiovascular system development, sprouting angiogenesis, circulatory system development, and pathway analysis in the cGMP-PKG signaling pathway. These results indicate that the novel lncRNA-related ceRNA network in HCM and LINC00310 may play a role in the mechanism of HCM pathogenesis, which could provide insight into the pathogenesis of HCM.
Collapse
Affiliation(s)
- Jin-Yan Chen
- Institute for Immunology, Fujian Academy of Medical Sciences, No. 7 Wusi Road, Fuzhou, 350001, China. .,Fujian Provincial Key Laboratory of Medical Analysis, Fuzhou, 350001, China.
| | - Zhang-Xin Xie
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Jia-Zhen Dai
- Department of Cardiology, Zhangzhou Affilated Hospital, Zhangzhou, China
| | - Jun-Yong Han
- Institute for Immunology, Fujian Academy of Medical Sciences, No. 7 Wusi Road, Fuzhou, 350001, China.,Fujian Provincial Key Laboratory of Medical Analysis, Fuzhou, 350001, China
| | - Kun Wang
- Institute for Immunology, Fujian Academy of Medical Sciences, No. 7 Wusi Road, Fuzhou, 350001, China.,Fujian Provincial Key Laboratory of Medical Analysis, Fuzhou, 350001, China
| | - Li-Hong Lu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.
| | - Jing-Jun Jin
- Institute for Immunology, Fujian Academy of Medical Sciences, No. 7 Wusi Road, Fuzhou, 350001, China.,Fujian Provincial Key Laboratory of Medical Analysis, Fuzhou, 350001, China
| | - Shi-Jie Xue
- Institute for Immunology, Fujian Academy of Medical Sciences, No. 7 Wusi Road, Fuzhou, 350001, China.,Fujian Provincial Key Laboratory of Medical Analysis, Fuzhou, 350001, China
| |
Collapse
|
8
|
Cao J, Yuan L. Identification of key genes for hypertrophic cardiomyopathy using integrated network analysis of differential lncRNA and gene expression. Front Cardiovasc Med 2022; 9:946229. [PMID: 35990977 PMCID: PMC9386162 DOI: 10.3389/fcvm.2022.946229] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Hypertrophic cardiomyopathy (HCM) is a complex heterogeneous heart disease. Recent reports found that long non-coding RNAs (lncRNAs) play an important role in the progression of cardiovascular diseases. The present study aimed to identify the novel lncRNAs and messenger RNAs (mRNAs) and determine the key pathways involved in HCM. Methods The lncRNA and mRNA sequencing datasets of GSE68316 and GSE130036 were downloaded from the Gene Expression Omnibus (GEO) database. An integrated co-expression network analysis was conducted to identify differentially expressed lncRNAs and differentially expressed mRNAs in patients with HCM. Then, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were explored to identify the biological functions and signaling pathways of the co-expression network. Protein–protein interaction (PPI) and hub gene networks were constructed by using Cytoscape software. Plasma samples of patients with HCM and the GSE89714 dataset were used to validate the bioinformatics results. Results A total of 1,426 differentially expressed long non-coding RNAs (lncRNAs) and 1,715 differentially expressed mRNAs were obtained from GSE68316, of which 965 lncRNAs and 896 mRNAs were upregulated and 461 lncRNAs and 819 mRNAs were downregulated. A total of 469 differentially expressed lncRNAs and 2,407 differentially expressed mRNAs were screened from GSE130036, of which 183 lncRNAs and 1,283 mRNAs were upregulated and 286 lncRNAs and 1,124 mRNAs were downregulated. A co-expression network was constructed and contained 30 differentially expressed lncRNAs and 63 differentially expressed mRNAs, which were primarily involved in ‘G-protein beta/gamma-subunit complex binding,' ‘polyubiquitin modification-dependent protein binding,' ‘Apelin signaling pathway,' and ‘Wnt signaling pathway.' The 10 hub genes in the upregulated network [G Protein Subunit Alpha I2 (GNAI2), G Protein Subunit Alpha I1 (GNAI1), G Protein Subunit Alpha I3 (GNAI3), G Protein Subunit Gamma 2 (GNG2), G Protein Subunit Beta 1 (GNB1), G Protein Subunit Gamma 13 (GNG13), G Protein Subunit Gamma Transducin 1 (GNGT1), G Protein Subunit Gamma 12 (GNG12), AKT Serine/Threonine Kinase 1 (AKT1) and GNAS Complex Locus (GNAS)] and the 10 hub genes in the downregulated network [Nucleotide-Binding Oligomerization Domain Containing Protein 2 (NOD2), Receptor-Interacting Serine/Threonine Kinase 2 (RIPK2), Nucleotide-Binding Oligomerization Domain Containing Protein 1 (NOD1), Mitochondrial Antiviral Signaling Protein (MAVS), Autophagy Related 16-Like 1 (ATG16L1), Interferon Induced With Helicase C Domain 1 (IFIH1), Autophagy Related 5 (ATG5), TANK-Binding Kinase 1 (TBK1), Caspase Recruitment Domain Family Member 9 (CARD9), and von Willebrand factor (VWF)] were screened using cytoHubba. The expression of LA16c-312E8.2 and RP5-1160K1.3 in the plasma of patients with HCM was elevated, and the expression of the MIR22 host gene (MIR22HG) was decreased, which was consistent with our analysis, while the expression of LINC00324 and Small Nucleolar RNA Host Gene 12 (SNHG12) was not significantly different between the two groups. Verification analyses performed on GSE89714 showed the upregulated mRNAs of Chloride Voltage-Gated Channel 7 (CLCN7), N-Acetylglucosamine-1-Phosphate Transferase Subunit Gamma (GNPTG), Unk Like Zinc Finger (UNKL), Adenosine Monophosphate Deaminase 2 (AMPD2), GNAI3, WD Repeat Domain 81 (WDR81), and Serpin Family F Member 1 (SERPINF1) and downregulated mRNAs of TATA-Box Binding Protein Associated Factor 12 (TAF12) co-expressed with five crucial lncRNAs. Moreover, GNAI2, GNAI3, GNG12, and vWF were upregulated and GNAS was downregulated in the top 10 hub genes of upregulated and downregulated PPI networks. Conclusion These findings from integrative biological analysis of lncRNA-mRNA co-expression networks explored the key genes and pathways and provide new insights into the understanding of the mechanism and discovering new therapeutic targets for HCM. Three differentially expressed pivotal lncRNAs (LA16c-312E8.2, RP5-1160K1.3, and MIR22HG) in the co-expression network may serve as biomarkers and intervention targets for the diagnosis and treatment of HCM.
Collapse
Affiliation(s)
- Jing Cao
- Department of Cardiovascular Medicine, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lei Yuan
- Department of Medical Affairs, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Lei Yuan
| |
Collapse
|
9
|
Wang T, Li J, Li H, Zhong X, Wang L, Zhao S, Liu X, Huang Z, Wang Y. Aerobic Exercise Inhibited P2X7 Purinergic Receptors to Improve Cardiac Remodeling in Mice With Type 2 Diabetes. Front Physiol 2022; 13:828020. [PMID: 35711309 PMCID: PMC9197582 DOI: 10.3389/fphys.2022.828020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Diabetic cardiomyopathy (DCM), the main complication of diabetes mellitus, presents as cardiac dysfunction by ventricular remodeling. In addition, the inhibition of P2X7 purinergic receptors (P2X7R) alleviates cardiac fibrosis and apoptosis in Type 1 diabetes. However, whether exercise training improves cardiac remodeling by regulating P2X7R remains unknown. Methods: Db/db mice spontaneously induced with type 2 diabetes and high-fat diet (HFD) and mice with streptozotocin (STZ)-induced type 2 diabetes mice were treated by 12-week treadmill training. Cardiac functions were observed by two-dimensional echocardiography. Hematoxylin-eosin staining, Sirius red staining and transmission electron microscopy were respectively used to detect cardiac morphology, fibrosis and mitochondria. In addition, real-time polymerase chain reaction and Western Blot were used to detect mRNA and protein levels. Results: Studying the hearts of db/db mice and STZ-induced mice, we found that collagen deposition and the number of disordered cells significantly increased compared with the control group. However, exercise markedly reversed these changes, and the same tendency was observed in the expression of MMP9, COL-I, and TGF-β, which indicated cardiac fibrotic and hypertrophic markers, including ANP and MyHC expression. In addition, the increased Caspase-3 level and the ratio of Bax/Bcl2 were reduced by exercise training, and similar results were observed in the TUNEL test. Notably, the expression of P2X7R was greatly upregulated in the hearts of db/db mice and HFD + STZ-induced DM mice and downregulated by aerobic exercise. Moreover, we indicated that P2X7R knock out significantly reduced the collagen deposition and disordered cells in the DM group. Furthermore, the apoptosis levels and TUNEL analysis were greatly inhibited by exercise or in the P2X7R-/- group in DM. We found significant differences between the P2X7R-/- + DM + EX group and DM + EX group in myocardial tissue apoptosis and fibrosis, in which the former is significantly milder. Moreover, compared with the P2X7R-/- + DM group, the P2X7R-/- + DM + EX group represented a lower level of cardiac fibrosis. The expression levels of TGF-β at the protein level and TGF-β and ANP at the genetic level were evidently decreased in the P2X7R-/- + DM + EX group. Conclusion: Aerobic exercise reversed cardiac remodeling in diabetic mice at least partly through inhibiting P2X7R expression in cardiomyocytes.
Collapse
Affiliation(s)
- Ting Wang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianmin Li
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hui Li
- Department of Ultrasound, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin Zhong
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Luya Wang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shujue Zhao
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuesheng Liu
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhouqing Huang
- The Key Laboratory of Cardiovascular Disease of Wenzhou, Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yonghua Wang
- Department of Physical Education, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
The lncRNA MIAT regulates CPT-1a mediated cardiac hypertrophy through m 6A RNA methylation reading protein Ythdf2. Cell Death Dis 2022; 8:167. [PMID: 35383152 PMCID: PMC8983679 DOI: 10.1038/s41420-022-00977-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023]
Abstract
Pathological cardiac hypertrophy is a key contributor in heart failure (HF). Long non-coding RNAs (lncRNAs) and N6-methyladenosine (m6A) modification play a vital role in cardiac hypertrophy respectively. Nevertheless, the interaction between lncRNA and m6A methylase in cardiac hypertrophy is scarcely reported. Here, we constructed a cardiac hypertrophy mouse model by transverse aortic constriction (TAC) surgery and H9c2 cell model by stimulating with AngII. We found that lncRNA MIAT mRNA level, and m6A RNA methylation reading protein Ythdf2 mRNA and protein levels, were significantly increased in the cardiac hypertrophy model both in vivo and vitro. MIAT or Ythdf2 overexpression aggravated cardiac hypertrophy, and vice versa. Through bioinformatics prediction, western blotting, FISH, RNA pull-down, and RIP, we found that MIAT bound to Ythdf2 and regulated its expression. Furthermore, we discovered that Ythdf2 function was a downstream of MIAT in cardiac hypertrophy. Finally, we found that MIAT was a necessary regulator of cardiac hypertrophy due to its regulation of the Ythdf2/PPARα/CPT-1a axis. This study indicated a new hypertrophic signaling pathway: MIAT/Ythdf2/PPARα/CPT-1a. The results provided a new understanding of the MIAT and m6A RNA methylation reading protein, Ythdf2, function and mechanism in cardiac hypertrophy and highlighted the potential therapeutic benefits in the heart.
Collapse
|
11
|
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
|
12
|
Noncoding RNAs in Cardiac Hypertrophy and Heart Failure. Cells 2022; 11:cells11050777. [PMID: 35269399 PMCID: PMC8908994 DOI: 10.3390/cells11050777] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023] Open
Abstract
Heart failure is a major global health concern. Noncoding RNAs (ncRNAs) are involved in physiological processes and in the pathogenesis of various diseases, including heart failure. ncRNAs have emerged as critical components of transcriptional regulatory pathways that govern cardiac development, stress response, signaling, and remodeling in cardiac pathology. Recently, studies of ncRNAs in cardiovascular disease have achieved significant development. Here, we discuss the roles of ncRNAs, including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs) that modulate the cardiac hypertrophy and heart failure.
Collapse
|
13
|
LncRNA ROR promotes NLRP3-mediated cardiomyocyte pyroptosis by upregulating FOXP1 via interactions with PTBP1. Cytokine 2022; 152:155812. [PMID: 35180562 DOI: 10.1016/j.cyto.2022.155812] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/12/2022] [Accepted: 01/21/2022] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The purpose of this design was to explore the specific role and related mechanism of long noncoding RNA (lncRNA) regulators of reprogramming (ROR) in viral myocarditis (VMC). METHODS AC16 cells were infected with coxsackievirus B3 (CVB3) to establish a VMC cell model in vitro. The release of interleukin (IL)-1β and IL-18 was evaluated by enzyme-linked immunosorbent assay (ELISA). Gene expression was calculated using quantitative real-time (qRT)-PCR. Cell pyroptosis was determined by flow cytometry and Western blot assays. Cell counting Kit-8 (CCK-8) detected cell viability. The molecular associations were verified by employing RNA immunoprecipitation (RIP), RNA pulldown and chromatin immunoprecipitation (ChIP) assays. RESULTS The lncRNA ROR was more highly expressed in CVB3 virus-infected AC16 cells than in controls. Knockdown of ROR markedly rescued cell viability and reduced the release of IL-1β and IL-18, cell pyroptosis and pyroptotic proteins such as NLRP3, ASC and cleaved caspase 1. Mechanistically, ROR destroyed the mRNA stability of Forkhead Box P Factor 1 (FOXP1) by binding polypyrimidine tract binding protein 1 (PTBP1). FOXP1 repressed the transcription of NLRP3 by directly interacting with its promoter. Importantly, coinhibition of FOXP1 impeded the protective role of ROR silencing in CVB3-infected AC16 cells. CONCLUSION In conclusion, these findings elucidated that ROR knockdown inhibited CVB3-induced cardiomyocyte inflammation and NLRP3-mediated pyroptosis by regulating the PTBP1/FOXP1 axis, implying that ROR might be a new inducer in CVB3-infected VMC.
Collapse
|
14
|
Sun Y, Xiao Z, Chen Y, Xu D, Chen S. Susceptibility Modules and Genes in Hypertrophic Cardiomyopathy by WGCNA and ceRNA Network Analysis. Front Cell Dev Biol 2022; 9:822465. [PMID: 35178407 PMCID: PMC8844202 DOI: 10.3389/fcell.2021.822465] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/28/2021] [Indexed: 02/05/2023] Open
Abstract
Background: We attempted to identify a regulatory competing endogenous RNA (ceRNA) network and a hub gene of Hypertrophic Cardiomyopathy (HCM). Methods: Microarray datasets of HCM tissue were obtained from NCBI Gene Expression Omnibus (GEO) database. The R package "limma" was used to identify differentially expressed genes. Online search databases were utilized to match the relation among differentially expressed long non-coding RNAs (lncRNAs), microRNAs (miRNAs) and mRNAs. Weighted correlation network analysis (WGCNA) was used to identify the correlations between key modules and HCM. STRING database was applied to construct PPI networks. Gene Set Enrichment Analysis (GSEA) was used to perform functional annotations and verified the hub genes. Results: A total of 269 DE-lncRNAs, 63 DE-miRNAs and 879 DE-mRNAs were identified in myocardial tissues from microarray datasets GSE130036, GSE36946 and GSE36961, respectively. According to online databases, we found 1 upregulated miRNA hsa-miR-184 that was targeted by 2 downregulated lncRNAs (SNHG9, AC010980.2), potentially targeted 2 downregulated mRNAs (LRRC8A, SLC7A5). 3 downregulated miRNAs (hsa-miR-17-5p, hsa-miR-876-3p, hsa-miR-139-5p) that were targeted by 9 upregulated lncRNAs, potentially targeted 21 upregulated mRNAs. Black and blue modules significantly related to HCM were identified by WGCNA. Hub gene IGFBP5 regulated by hsa-miR-17-5p, AC007389.5, AC104667.1, and AC002511.2 was identified. GSEA indicated that IGFBP5 might involve in the synthesis of myosin complex, participate in kinesin binding, motor activity and function via the regulation of actin cytoskeleton. Conclusion: The results provide a potential molecular regulatory mechanism for the diagnosis and treatment of HCM. IGFBP5 might play an important role in the progression of HCM.
Collapse
Affiliation(s)
- Yifan Sun
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zhongbo Xiao
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | | | | | | |
Collapse
|
15
|
Jia X, Huang J, Wu B, Yang M, Xu W. A Competitive Endogenous RNA Network Based on Differentially Expressed lncRNA in Lipopolysaccharide-Induced Acute Lung Injury in Mice. Front Genet 2021; 12:745715. [PMID: 34917127 PMCID: PMC8669720 DOI: 10.3389/fgene.2021.745715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/14/2021] [Indexed: 12/03/2022] Open
Abstract
Non-coding RNAs have remarkable roles in acute lung injury (ALI) initiation. Nevertheless, the significance of long non-coding RNAs (lncRNAs) in ALI is still unknown. Herein, we purposed to identify potential key genes in ALI and create a competitive endogenous RNA (ceRNA) modulatory network to uncover possible molecular mechanisms that affect lung injury. We generated a lipopolysaccharide-triggered ALI mouse model, whose lung tissue was subjected to RNA sequencing, and then we conducted bioinformatics analysis to select genes showing differential expression (DE) and to build a lncRNA-miRNA (microRNA)- mRNA (messenger RNA) modulatory network. Besides, GO along with KEGG assessments were conducted to identify major biological processes and pathways, respectively, involved in ALI. Then, RT-qPCR assay was employed to verify levels of major RNAs. A protein-protein interaction (PPI) network was created using the Search Tool for the Retrieval of Interacting Genes (STRING) database, and the hub genes were obtained with the Molecular Complex Detection plugin. Finally, a key ceRNA subnetwork was built from major genes and their docking sites. Overall, a total of 8,610 lncRNAs were identified in the normal and LPS groups. Based on the 308 DE lncRNAs [p-value < 0.05, |log2 (fold change) | > 1] and 3,357 DE mRNAs [p-value < 0.05, |log2 (fold change) | > 1], lncRNA-miRNA and miRNA-mRNA pairs were predicted using miRanda. The lncRNA-miRNA-mRNA network was created from 175 lncRNAs, 22 miRNAs, and 209 mRNAs in ALI. The RT-qPCR data keep in step with the RNA sequencing data. GO along with KEGG analyses illustrated that DE mRNAs in this network were mainly bound up with the inflammatory response, developmental process, cell differentiation, cell proliferation, apoptosis, and the NF-kappa B, PI3K-Akt, HIF-1, MAPK, Jak-STAT, and Notch signaling pathways. A PPI network on the basis of the 209 genes was established, and three hub genes (Nkx2-1, Tbx2, and Atf5) were obtained from the network. Additionally, a lncRNA-miRNA-hub gene subnetwork was built from 15 lncRNAs, 3 miRNAs, and 3 mRNAs. Herein, novel ideas are presented to expand our knowledge on the regulation mechanisms of lncRNA-related ceRNAs in the pathogenesis of ALI.
Collapse
Affiliation(s)
- Xianxian Jia
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jinhui Huang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bo Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Miao Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Correia CCM, Rodrigues LF, de Avila Pelozin BR, Oliveira EM, Fernandes T. Long Non-Coding RNAs in Cardiovascular Diseases: Potential Function as Biomarkers and Therapeutic Targets of Exercise Training. Noncoding RNA 2021; 7:65. [PMID: 34698215 PMCID: PMC8544698 DOI: 10.3390/ncrna7040065] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022] Open
Abstract
Despite advances in treatments and therapies, cardiovascular diseases (CVDs) remain one of the leading causes of death worldwide. The discovery that most of the human genome, although transcribed, does not encode proteins was crucial for focusing on the potential of long non-coding RNAs (lncRNAs) as essential regulators of cell function at the epigenetic, transcriptional, and post-transcriptional levels. This class of non-coding RNAs is related to the pathophysiology of the cardiovascular system. The different expression profiles of lncRNAs, in different contexts of CVDs, change a great potential in their use as a biomarker and targets of therapeutic intervention. Furthermore, regular physical exercise plays a protective role against CVDs; on the other hand, little is known about its underlying molecular mechanisms. In this review, we look at the accumulated knowledge on lncRNAs and their functions in the cardiovascular system, focusing on the cardiovascular pathology of arterial hypertension, coronary heart disease, acute myocardial infarction, and heart failure. We discuss the potential of these molecules as biomarkers for clinical use, their limitations, and how the manipulation of the expression profile of these transcripts through physical exercise can begin to be suggested as a strategy for the treatment of CVDs.
Collapse
Affiliation(s)
- Camila Caldas Martins Correia
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-030, Brazil;
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.d.A.P.); (E.M.O.)
| | - Luis Felipe Rodrigues
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.d.A.P.); (E.M.O.)
| | - Bruno Rocha de Avila Pelozin
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.d.A.P.); (E.M.O.)
| | - Edilamar Menezes Oliveira
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.d.A.P.); (E.M.O.)
| | - Tiago Fernandes
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.d.A.P.); (E.M.O.)
| |
Collapse
|
17
|
Tian C, Yang Y, Ke Y, Yang L, Zhong L, Wang Z, Huang H. Integrative Analyses of Genes Associated With Right Ventricular Cardiomyopathy Induced by Tricuspid Regurgitation. Front Genet 2021; 12:708275. [PMID: 34603374 PMCID: PMC8485137 DOI: 10.3389/fgene.2021.708275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022] Open
Abstract
Tricuspid regurgitation (TR) induces right ventricular cardiomyopathy, a common heart disease, and eventually leads to severe heart failure and serious clinical complications. Accumulating evidence shows that long non-coding RNAs (lncRNAs) are involved in the pathological process of a variety of cardiovascular diseases. However, the regulatory mechanisms and functional roles of RNA interactions in TR-induced right ventricular cardiomyopathy are still unclear. Accordingly, we performed integrative analyses of genes associated with right ventricular cardiomyopathy induced by TR to study the roles of lncRNAs in the pathogenesis of this disease. In this study, we used high-throughput sequencing data of tissue samples from nine clinical cases of right ventricular myocardial cardiomyopathy induced by TR and nine controls with normal right ventricular myocardium from the Genotype-Tissue Expression database. We identified differentially expressed lncRNAs and constructed a protein-protein interaction and lncRNA-messenger RNA (mRNA) co-expression network. Furthermore, we determined hub lncRNA-mRNA modules related to right ventricular myocardial disease induced by TR and constructed a competitive endogenous RNA network for TR-induced right ventricular myocardial disease by integrating the interaction of lncRNA-miRNA-mRNA. In addition, we analyzed the immune infiltration using integrated data and the correlation of each immune-related gene with key genes of the integrated expression matrix. The present study identified 648 differentially expressed mRNAs, 201 differentially expressed miRNAs, and 163 differentially expressed lncRNAs. Protein-protein interaction network analysis confirmed that ADRA1A, AVPR1B, OPN4, IL-1B, IL-1A, CXCL4, ADCY2, CXCL12, GNB4, CCL20, CXCL8, and CXCL1 were hub genes. CTD-2314B22.3, hsa-miR-653-5p, and KIF17ceRNA; SRGAP3-AS2, hsa-miR-539-5p, and SHANK1; CERS6-AS1, hsa-miR-497-5p, and OPN4; INTS6-AS1, hsa-miR-4262, and NEURL1B; TTN-AS1, hsa-miR-376b-3p, and TRPM5; and DLX6-AS1, hsa-miR-346, and BIRC7 axes were obtained by constructing the ceRNA networks. Through the immune infiltration analysis, we found that the proportion of CD4 and CD8 T cells was about 20%, and the proportion of fibroblasts and endothelial cells was high. Our findings provide some insights into the mechanisms of RNA interaction in TR-induced right ventricular cardiomyopathy and suggest that lncRNAs are a potential therapeutic target for treating right ventricular myocardial disease induced by TR.
Collapse
Affiliation(s)
- Chengnan Tian
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital, Guangzhou, China.,The First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| | - Yanchen Yang
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Yingjie Ke
- Nanhai Hospital of Guangdong Provincial People's Hospital, Foshan, China
| | - Liang Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Lishan Zhong
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Zhenzhong Wang
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Huanlei Huang
- Department of Cardiovascular Surgery, Guangdong Provincial People's Hospital, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
Han L, Yang L. Multidimensional Mechanistic Spectrum of Long Non-coding RNAs in Heart Development and Disease. Front Cardiovasc Med 2021; 8:728746. [PMID: 34604357 PMCID: PMC8483262 DOI: 10.3389/fcvm.2021.728746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022] Open
Abstract
With the large-scale genome-wide sequencing, long non-coding RNAs (lncRNAs) have been found to compose of a large portion of the human transcriptome. Recent studies demonstrated the multidimensional functions of lncRNAs in heart development and disease. The subcellular localization of lncRNA is considered as a key factor that determines lncRNA function. Cytosolic lncRNAs mainly regulate mRNA stability, mRNA translation, miRNA processing and function, whereas nuclear lncRNAs epigenetically regulate chromatin remodeling, structure, and gene transcription. In this review, we summarize the molecular mechanisms of cytosolic and nuclear lncRNAs in heart development and disease separately, and emphasize the recent progress to dictate the crosstalk of cytosolic and nuclear lncRNAs in orchestrating the same biological process. Given the low evolutionary conservation of most lncRNAs, deeper understanding of human lncRNA will uncover a new layer of human regulatory mechanism underlying heart development and disease, and benefit the future clinical treatment for human heart disease.
Collapse
Affiliation(s)
- Lei Han
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lei Yang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
19
|
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: 0.5] [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
|
20
|
Abstract
Cardiac hypertrophy, characterized by the enlargement of cardiomyocytes, is initially an adaptive response to physiological and pathological stimuli. Decompensated cardiac hypertrophy is related to fibrosis, inflammatory cytokine, maladaptive remodeling, and heart failure. Although pathological myocardial hypertrophy is the main cause of hypertrophy-related morbidity and mortality, our understanding of its mechanism is still poor. Long noncoding RNAs (lncRNAs) are noncoding RNAs that regulate various physiological and pathological processes through multiple molecular mechanisms. Recently, accumulating evidence has indicated that lncRNA-H19 is a potent regulator of the progression of cardiac hypertrophy. For the first time, this review summarizes the current studies about the role of lncRNA-H19 in cardiac hypertrophy, including its pathophysiological processes and underlying pathological mechanism, including calcium regulation, fibrosis, apoptosis, angiogenesis, inflammation, and methylation. The context within which lncRNA-H19 might be developed as a target for cardiac hypertrophy treatment is then discussed to gain better insight into the possible biological functions of lncRNA-H19 in cardiac hypertrophy.
Collapse
|
21
|
Zhang Z, Sang M, Liu S, Shao J, Cai Y. Differential expression of long non-coding RNA Regulator of reprogramming and its molecular mechanisms in polycystic ovary syndrome. J Ovarian Res 2021; 14:79. [PMID: 34148561 PMCID: PMC8215827 DOI: 10.1186/s13048-021-00829-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/21/2021] [Indexed: 12/13/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is a common endocrine disease in women of reproductive age. Multiple studies have shown that long non-coding RNAs (lncRNA) and microRNAs (miRNA) play a role in PCOS. This study aimed to explore the role and molecular mechanism of lncRNA -Regulator of reprogramming (lncROR) in PCOS. Results Expression level of lncROR in PCOS patients was up-regulated, while level of miR-206 was down-regulated in comparison with control group (P < 0.001). Logistics regression analysis showed that lncROR and miR-206 were independent predictors of PCOS. The ROC curve showed that lncROR had a high diagnostic value for PCOS with an AUC value of 0.893. Pearson correlation coefficient indicated that the expression level of miR-206 was negatively correlated with the level of lncROR. CCK-8 assay and apoptosis assay revealed that downregulation of lncROR up-regulated the expression of miR-206, thereby inhibiting cell proliferation and promoting cell apoptosis. However, silencing the expression of miR-206 reversed the above effects caused by down-regulation of lncROR expression. Luciferase reporter gene assay suggested that there was a target relationship between lncROR and miR-206. VEGF was proved to be the target gene of miR-206. Conclusions Highly expressed lncROR indirectly up-regulated the expression of VEGF by down-regulating the expression of miR-206, thereby promoting the proliferation of KGN cells and inhibiting apoptosis, and further promoting the development of PCOS.
Collapse
Affiliation(s)
- Zhihong Zhang
- Department of Obstetrics and Gynecology, General Hospital of Daqing Oilfield, Daqing, 163000, Heilongjiang, China
| | - Min Sang
- Gynecology Clinic, The First Hospital of Harbin, No.151 Diduan Street, Heilongjiang, 150010, Harbin, China.
| | - Siqin Liu
- Laboratory Department, General Hospital of Daqing Oilfield, Daqing, 163000, Heilongjiang, China
| | - Jing Shao
- Department of Obstetrics and Gynecology, General Hospital of Daqing Oilfield, Daqing, 163000, Heilongjiang, China
| | - Yunjiang Cai
- Department of Psychology, Harbin Medical Univercity (Daqing), Daqing, 163319, Heilongjiang, 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: 13] [Impact Index Per Article: 3.3] [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
|
Luo S, Zhang M, Wu H, Ding X, Li D, Dong X, Hu X, Su S, Shang W, Wu J, Xiao H, Yang W, Zhang Q, Zhang J, Lu Y, Pan Z. SAIL: a new conserved anti-fibrotic lncRNA in the heart. Basic Res Cardiol 2021; 116:15. [PMID: 33675440 DOI: 10.1007/s00395-021-00854-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/12/2021] [Indexed: 12/19/2022]
Abstract
Long non-coding RNAs (lncRNAs) account for a large proportion of genomic transcripts and are critical regulators in various cardiac diseases. Though lncRNAs have been reported to participate in the process of diverse cardiac diseases, the contribution of lncRNAs in cardiac fibrosis remains to be fully elucidated. Here, we identified a novel anti-fibrotic lncRNA, SAIL (scaffold attachment factor B interacting lncRNA). SAIL was reduced in cardiac fibrotic tissue and activated cardiac fibroblasts. Gain- and loss-of-function studies showed that knockdown of SAIL promoted proliferation and collagen production of cardiac fibroblasts with or without TGF-β1 (transforming growth factor beta1) treatment, while overexpression of SAIL did the opposite. In mouse cardiac fibrosis induced by myocardial infarction, knockdown of SAIL exacerbated, whereas overexpression of SAIL alleviated cardiac fibrosis. Mechanically, SAIL inhibited the fibrotic process by directly binding with SAFB via 23 conserved nucleotide sequences, which in turn blocked the access of SAFB to RNA pol II (RNA polymerase II) and reduced the transcription of fibrosis-related genes. Intriguingly, the human conserved fragment of SAIL (hSAIL) significantly suppressed the proliferation and collagen production of human cardiac fibroblasts. Our findings demonstrate that SAIL regulates cardiac fibrosis by regulating SAFB-mediated transcription of fibrotic related genes. Both SAIL and SAFB hold the potential to become novel therapeutic targets for cardiac fibrosis.
Collapse
Affiliation(s)
- Shenjian Luo
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Mingyu Zhang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Hao Wu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Xin Ding
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Danyang Li
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Xue Dong
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Xiaoxi Hu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Shuang Su
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Wendi Shang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Jiaxu Wu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Hongwen Xiao
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Wanqi Yang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Qi Zhang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Jifan Zhang
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Yanjie Lu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Zhenwei Pan
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, 150081, Heilongjiang, People's Republic of China.
| |
Collapse
|
24
|
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: 3.3] [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
|
25
|
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: 3.0] [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
|
26
|
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: 0.8] [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
|
27
|
He Y, Feng Z, Lu J, Wang R, Huang C, Zhou Y. Exploring biomarkers and therapeutic targets for pressure overload induced heart failure based on microarray data. Cardiovasc Diagn Ther 2020; 10:1226-1237. [PMID: 33224746 DOI: 10.21037/cdt-20-465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Heart failure (HF) is an end stage heart condition with poor prognosis which brings about tremendous social medical cost. Along decades, mechanism and treatments of HF have been under restless research. Methods In the present study, we first established pressure overload induced HF model using transaortic arch constriction (TAC) method in mice. The global expression profiles of long noncoding RNA (lncRNA), microRNA (miRNA) and messenger RNA (mRNA) were obtained by microarray probes, which were further confirmed by quantitative PCR (qPCR). Bioinformatics analysis was performed using multiple methods including volcano plotting, heatmapping and hierarchical clustering, Gene Ontology (GO) and pathway enrichment analysis, and competing endogenous RNA (ceRNA) regulatory network construction. Results Totally, 1,139 differentially expressed mRNAs (DEmRNAs), 3,830 lncRNAs (DElncRNAs) and 13 miRNAs (DEmiRNAs) were identified in HF group compared to control group, which could distinctly differentiate HF from normal control and were potential candidate biomarkers for HF. GO and pathway enrichment analysis revealed that multiple significant biological processes and pathways were involved in HF pathogenesis, such as extracellular matrix structural constituent, proteinaceous extracellular matrix, positive regulation of apoptotic process and integrin signaling pathway. Nine DElncRNAs, 3 DEmiRNAs and 25 DEmRNAs were filtrated out to construct a ceRNA network, which visually displayed their regulatory roles with therapeutic target potential. Conclusions The present study identified differentially expressed RNAs that might be involved in the pathogenesis and progression of HF. The outcomes shed lights into the underlying mechanisms for HF and provided candidate biomarkers and intervention targets for further research.
Collapse
Affiliation(s)
- Yongli He
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhe Feng
- The First Affiliated Hospital of Henan University of Science and Technology, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jianjun Lu
- The Second Clinical Medical School, Southern Medical University, Guangzhou, China.,Department of Medical Services, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rui Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Cheng Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yingling Zhou
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
28
|
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: 1.4] [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
|
29
|
Li X, Zuo C, Sun D, Zhao T, Zhang Z. Arsenite Increases Linc-ROR in Human Bronchial Epithelial Cells that Can Be Inhibited by Antioxidant Factors. Biol Trace Elem Res 2020; 198:131-141. [PMID: 32030632 DOI: 10.1007/s12011-020-02065-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/30/2020] [Indexed: 02/07/2023]
Abstract
Oxidative stress is the main mechanism of arsenite toxicity. Long intergenic non-coding RNA regulator of reprogramming is a newly found stress-response long non-coding RNA that is activated in various stress conditions. However, whether long intergenic non-coding RNA, regulator of reprogramming (linc-ROR) is involved in arsenite-induced oxidative stress has not been explored. In this study, we found that arsenite dose responsively increased the expression of linc-ROR in human bronchial epithelial (HBE) cells, along with elevated oxidative stress demonstrated by increased intracellular reactive oxygen species (ROS) and DNA damage, as well as decreased antioxidant glutathione and superoxide dismutase. We further found that the pre-treatment with N-acetylcysteine, a widely used ROS scavenger, and the over-expression of antioxidant NRF2 protein, both significantly reduced arsenite-induced oxidative stress in arsenite-treated HBE cells, and the linc-ROR over-expression was also inhibited, suggesting that oxidative stress is a key factor for the increase of linc-ROR in arsenite-treated HBE cells. Moreover, our results of bio-informatic analysis showed that arsenite-induced oxidative stress might modulate linc-ROR expression via 3 genes and the up-regulated linc-ROR in arsenite-induced oxidative stress may get involved in cellular processes such as cellular stress response, RNA metabolism, and DNA repair. Collectively, our study demonstrates that oxidative stress plays the key role in arsenite-induced over-expression of linc-ROR, and linc-ROR may be a new clue for exploring the mechanism of arsenite toxicity.
Collapse
Affiliation(s)
- Xinyang Li
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin Nanlu, 610041, Chengdu, People's Republic of China
| | - Chao Zuo
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin Nanlu, 610041, Chengdu, People's Republic of China
| | - Donglei Sun
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin Nanlu, 610041, Chengdu, People's Republic of China
| | - Tianhe Zhao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin Nanlu, 610041, Chengdu, People's Republic of China
| | - Zunzhen Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin Nanlu, 610041, Chengdu, People's Republic of China.
| |
Collapse
|
30
|
Zhang C, Han B, Xu T, Li D. The biological function and potential mechanism of long non-coding RNAs in cardiovascular disease. J Cell Mol Med 2020; 24:12900-12909. [PMID: 33052009 PMCID: PMC7701533 DOI: 10.1111/jcmm.15968] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (lncRNAs), as part of the family of non-protein-coding transcripts, are implicated in the occurrence and progression of several cardiovascular diseases (CVDs). With recent advances in lncRNA research, these molecules are purported to regulate gene expression at multiple levels, thereby producing beneficial or detrimental biological effects during CVD pathogenesis. At the transcriptional level, lncRNAs affect gene expression by interacting with DNA and proteins, for example, components of chromatin-modifying complexes, or transcription factors affecting chromatin status. These potential mechanisms suggest that lncRNAs guide proteins to specific gene loci (eg promoter regions), or forestall proteins to specific genomic sites via DNA binding. Additionally, some lncRNAs are required for correct chromatin conformation, which occurs via chromatin looping in enhancer-like models. At the post-transcriptional level, lncRNAs interact with RNA molecules, mainly microRNAs (miRNAs) and mRNAs, potentially regulating CVD pathophysiological processes. Moreover, lncRNAs appear to post-transcriptionally modulate gene expression by participating in mRNA splicing, stability, degradation and translation. Thus, the purpose of this review is to provide a comprehensive summary of lncRNAs implicated in CVD biological processes, with an emphasis on potential mechanisms of action.
Collapse
Affiliation(s)
- Chengmeng Zhang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Bing Han
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou, China
| | - Tongda Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
31
|
Cao H, Yang B, Zhao Y, Deng X, Shen X. The pro-apoptosis and pro-inflammation role of LncRNA HIF1A-AS1 in Coxsackievirus B3-induced myocarditis via targeting miR-138. Cardiovasc Diagn Ther 2020; 10:1245-1255. [PMID: 33224748 DOI: 10.21037/cdt-20-545] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Cardiomyocyte apoptosis and inflammation induced by Coxsackievirus B3 (CVB3) are key pathogenetic mechanisms in viral myocarditis. Alterations in microRNAs and lncRNAs are associated with cardiac remodeling. However, whether the microRNA and lncRNA interact to regulate cardiomyocyte apoptosis and inflammation is unknown. Methods BALB/c mice were infected with CVB3 to generate acute viral myocarditis model. The expression levels of lncRNA HIF1A-AS1 and miR-138 were examined in mouse myocardium and primary cardiomyocytes with CVB3 infection. We then knocked down HIF1A-AS1 by siRNA and upregulated miR-138 by microRNA mimics in cardiomyocytes. Results The expression of lncRNA HIF1A-AS1was significantly increased in CVB3-induced myocardium and cardiomyocytes. As documented by flow cytometry, silencing of HIF1A-AS1 alleviated late apoptosis (5.1%±2.8% vs. 17.2%±4.2%, P<0.01) and ROS production (68.73%±2.78% vs. 90.40%±2.86%, P<0.01) compared to their levels in cardiomyocytes transfected with control siRNA. The content of proinflammatory cytokines was substantially decreased by HIF1A-AS1 siRNA. Furthermore, we identified that HIF1A-AS1 bound to miR-138 and significantly suppressed miR-138 expression. Blocking HIF1A-AS1 attenuated IκBα phosphorylation and NF-κB activity, while cotransfection with miR-138 mimics markedly reversed its effect. Conclusions In conclusion, lncRNA HIF1A-AS1 promotes NF-κB signaling and subsequently aggravates cardiomyocyte apoptosis and inflammation via targeting miR-138.
Collapse
Affiliation(s)
- Huili Cao
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Bin Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yajing Zhao
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xingqiang Deng
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaoyu Shen
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
32
|
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: 3.6] [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
|
33
|
Wang W, Li Y, Zhi S, Li J, Miao J, Ding Z, Peng Y, Huang Y, Zheng R, Yu H, Qi P, Wang J, Fu X, Hu M, Chen S. LncRNA-ROR/microRNA-185-3p/YAP1 axis exerts function in biological characteristics of osteosarcoma cells. Genomics 2020; 113:450-461. [PMID: 32898639 DOI: 10.1016/j.ygeno.2020.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/21/2020] [Accepted: 09/03/2020] [Indexed: 11/19/2022]
Abstract
AIM The co-expression network of long non-coding RNA ROR (lncRNA-ROR) and microRNA-185-3p (miR-185-3p) has not been focused on osteosarcoma. Therein, this work was initiated to uncover lncRNA-ROR and miR-185-3p functions in osteosarcoma. METHODS LncRNA-ROR, miR-185-3p and Yes-associated protein 1 (YAP1) expression in osteosarcoma tissues and cells were detected. The screened cells (MG63 and U2OS) were transfected with decreased and/or increased lncRNA-ROR and miR-185-3p to explore osteosarcoma progression. Tumor growth was detected by tumor xenografts in mice. RESULTS Up-regulated lncRNA-ROR and YAP1 and down-regulated miR-185-3p were found in osteosarcoma. LncRNA ROR knockdown or miR-185-3p overexpression inhibited osteosarcoma cell progression while lncRNA ROR elevation or miR-185-3p inhibition presented the opposite effects. Function of lncRNA ROR was rescued by miR-185-3p and regulated the growth and metastasis of osteosarcoma cells via modulating YAP1, the target gene of miR-185-3p. CONCLUSION This work illustrates that lncRNA-ROR down-regulation or miR-185-3p up-regulation inhibits osteosarcoma progression via YAP1 repression.
Collapse
Affiliation(s)
- Weiguo Wang
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yuezhan Li
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Shuang Zhi
- Four Gynecological Wards, Ningbo Women & Children's Hospital, Ningbo 315000, Zhejiang, China
| | - Jinsong Li
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Jinglei Miao
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Zhiyu Ding
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yi Peng
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yan Huang
- The Second Xiangya Hospital of Central South University, Changsha 410013, Hunan, China
| | - Ruping Zheng
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China
| | - Haiyang Yu
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China
| | - Pei Qi
- Department of pharmacy, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, China
| | - Jianlong Wang
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Xingchang Fu
- Department of Orthopedics, Hunan Aerospace hospital, Changsha 410205, Hunan, China
| | - Minghua Hu
- Department of Anatomy, Histology and Embryology, Changsha Medical University, Changsha 410219, China
| | - Shijie Chen
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
34
|
Chen C, Zong M, Lu Y, Guo Y, Lv H, Xie L, Fu Z, Cheng Y, Si Y, Ye B, Fan L. Differentially expressed lnc-NOS2P3-miR-939-5p axis in chronic heart failure inhibits myocardial and endothelial cells apoptosis via iNOS/TNFα pathway. J Cell Mol Med 2020; 24:11381-11396. [PMID: 32844595 PMCID: PMC7576245 DOI: 10.1111/jcmm.15740] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 07/06/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Inflammatory cytokine‐induced cell apoptosis is important for initiation and progression of chronic heart failure (CHF). Non‐coding RNAs, including long non‐coding RNAs and microRNAs, have emerged as critical regulators of this pathological process. The role in regulating inflammation and induction to cell apoptosis in CHF is not well understood. This study found CHF patients had elevated serum miR‐939‐5p, with greater increase in New York Heart Association (NYHA) I‐II patients than in NYHA III‐IV. Moreover, miR‐939‐5p was positively correlated with B‐type natriuretic peptide (BNP) in NYHA III‐IV patients, while not in NYHA I‐II. Further study showed miR‐939‐5p mimics promoted cell proliferation and inhibited inflammatory cytokine‐induced apoptosis of HUVECs and H9C2, while inhibition of endogenous miR‐939‐5p produced the opposite effects. Induced nitric oxide synthase (iNOS) and tumour necrosis factor α (TNFα) were identified as target genes of miR‐939‐5p. Additionally, lncRNA‐NOS2P3 acted as an endogenous sponge RNA to inhibit miR‐939‐5p expression, regulate the expression of iNOS/TNFα and control inflammation‐induced cells apoptosis. These suggest that CHF patients exhibited elevated serum miR‐939‐5p level especially in NYHA I‐II grades. And lnc‐NOS2P3‐miR‐939‐5p‐iNOS/TNFα pathway regulated inflammatory cytokine‐induced endothelial and myocardial cells apoptosis and provided a promising strategy for diagnosis and treatment of CHF.
Collapse
Affiliation(s)
- Cuncun Chen
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ming Zong
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Lu
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yide Guo
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Honggen Lv
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lihong Xie
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiyan Fu
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Cheng
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuying Si
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bei Ye
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lieying Fan
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
35
|
Collins L, Binder P, Chen H, Wang X. Regulation of Long Non-coding RNAs and MicroRNAs in Heart Disease: Insight Into Mechanisms and Therapeutic Approaches. Front Physiol 2020; 11:798. [PMID: 32754048 PMCID: PMC7365882 DOI: 10.3389/fphys.2020.00798] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide and there is an increasing need to identify new therapeutic targets that could be used to prevent or treat these diseases. Due to recent scientific advances, non-coding RNAs are widely accepted as important regulators of cellular processes, and the identification of an axis of interaction between long non-coding RNAs (lncRNAs) and micro RNAs (miRNAs) has provided another platform through which cardiovascular disease could be targeted therapeutically. Increasing evidence has detailed the importance of these non-coding RNAs, both individually and in an axis of regulation, in the processes and diseases involving the heart. However, further investigation into the consequences of targeting this mechanism, as well as refinement of how the system is targeted, are required before a treatment can be provided in clinic. This level of genomic regulation provides an exciting potential novel therapeutic strategy for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Lucy Collins
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Pablo Binder
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Hongshan Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, China
| | - Xin Wang
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
36
|
Bektik E, Cowan DB, Wang DZ. Long Non-Coding RNAs in Atrial Fibrillation: Pluripotent Stem Cell-Derived Cardiomyocytes as a Model System. Int J Mol Sci 2020; 21:ijms21155424. [PMID: 32751460 PMCID: PMC7432754 DOI: 10.3390/ijms21155424] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is a type of sustained arrhythmia in humans often characterized by devastating alterations to the cardiac conduction system as well as the structure of the atria. AF can lead to decreased cardiac function, heart failure, and other complications. Long non-coding RNAs (lncRNAs) have been shown to play important roles in the cardiovascular system, including AF; however, a large group of lncRNAs is not conserved between mouse and human. Furthermore, AF has complex networks showing variations in mechanisms in different species, making it challenging to utilize conventional animal models to investigate the functional roles and potential therapeutic benefits of lncRNAs for AF. Fortunately, pluripotent stem cell (PSC)-derived cardiomyocytes (CMs) offer a reliable platform to study lncRNA functions in AF because of certain electrophysiological and molecular similarities with native human CMs. In this review, we first summarize the broad aspects of lncRNAs in various heart disease settings, then focus on their potential roles in AF development and pathophysiology. We also discuss current uses of PSCs in AF research and describe how these studies could be developed into novel therapeutics for AF and other cardiovascular diseases.
Collapse
Affiliation(s)
- Emre Bektik
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
| | - Douglas B. Cowan
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood, Boston, MA 02115, USA; (E.B.); (D.B.C.)
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Correspondence:
| |
Collapse
|
37
|
Lozano-Vidal N, Bink DI, Boon RA. Long noncoding RNA in cardiac aging and disease. J Mol Cell Biol 2020; 11:860-867. [PMID: 31152659 PMCID: PMC6884711 DOI: 10.1093/jmcb/mjz046] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/18/2019] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the main cause of morbidity and mortality in Western society and present an important age-related risk. With the constant rise in life expectancy, prevalence of CVD in the population will likely increase further. New therapies, especially in the elderly, are needed to combat CVD. This review is focused on the role of long noncoding RNA (lncRNA) in CVD. RNA sequencing experiments in the past decade showed that most RNA does not code for protein, but many RNAs function as ncRNA. Here, we summarize the recent findings of lncRNA regulation in the diseased heart. The potential use of these RNAs as biomarkers of cardiac disease prediction is also discussed.
Collapse
Affiliation(s)
- Noelia Lozano-Vidal
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam Universitair Medische Centra, Vrije Universiteit Amsterdam, 1081 HZ, Amsterdam, the Netherlands
| | - Diewertje I Bink
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam Universitair Medische Centra, Vrije Universiteit Amsterdam, 1081 HZ, Amsterdam, the Netherlands
| | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam Universitair Medische Centra, Vrije Universiteit Amsterdam, 1081 HZ, Amsterdam, the Netherlands.,Institute of Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Berlin, Germany
| |
Collapse
|
38
|
The Expression of microRNA in Adult Rat Heart with Isoproterenol-Induced Cardiac Hypertrophy. Cells 2020; 9:cells9051173. [PMID: 32397324 PMCID: PMC7290591 DOI: 10.3390/cells9051173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac hypertrophy is a common pathological condition and an independent risk factor that triggers cardiovascular morbidity. As an important epigenetic regulator, miRNA is widely involved in many biological processes. In this study, miRNAs expressed in rat hearts that underwent isoprenaline-induced cardiac hypertrophy were identified using high-throughput sequencing, and functional verification of typical miRNAs was performed using rat primary cardiomyocytes. A total of 623 miRNAs were identified, of which 33 were specifically expressed in cardiac hypertrophy rats. The enriched pathways of target genes of differentially expressed miRNAs included the FoxO signaling pathway, dopaminergic synapse, Wnt signaling pathway, MAPK (mitogen-activated protein kinase) signaling pathway, and Hippo signaling pathway. Subsequently, miR-144 was the most differentially expressed miRNA and was subsequently selected for in vitro validation. Inhibition of miR-144 expression in primary myocardial cells caused up-regulation of cardiac hypertrophy markers atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). The dual luciferase reporter system showed that ANP may be a target gene of miR-144. Long non-coding RNA myocardial infarction associated transcript (LncMIAT) is closely related to heart disease, and here, we were the first to discover that LncMIAT may act as an miR-144 sponge in isoproterenol-induced cardiac hypertrophy. Taken together, these results enriched the understanding of miRNA in regulating cardiac hypertrophy and provided a reference for preventing and treating cardiac hypertrophy.
Collapse
|
39
|
Tang R, Long T, Lui KO, Chen Y, Huang ZP. A Roadmap for Fixing the Heart: RNA Regulatory Networks in Cardiac Disease. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 20:673-686. [PMID: 32380417 PMCID: PMC7210385 DOI: 10.1016/j.omtn.2020.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/16/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023]
Abstract
With the continuous development of RNA biology and massive genome-wide transcriptome analysis, more and more RNA molecules and their functions have been explored in the last decade. Increasing evidence has demonstrated that RNA-related regulatory networks play an important role in a variety of human diseases, including cardiovascular diseases. In this review, we focus on RNA regulatory networks in heart disease, most of which are devastating conditions with no known cure. We systemically summarize recent discoveries of important new components of RNA regulatory networks, including microRNAs, long non-coding RNAs, and circular RNAs, as well as multiple regulators that affect the activity of these networks in cardiac physiology and pathology. In addition, this review covers emerging micropeptides, which represent short open reading frames (sORFs) in long non-coding RNA transcripts that may modulate cardiac physiology. Based on the current knowledge of RNA regulatory networks, we think that ongoing discoveries will not only provide us a better understanding of the molecular mechanisms that underlie heart disease, but will also identify novel biomarkers and therapeutic targets for the diagnosis and treatment of cardiac disease.
Collapse
Affiliation(s)
- Rong Tang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Tianxin Long
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Kathy O Lui
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR 999077, China
| | - Yili Chen
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
40
|
Yeh CF, Chang YCE, Lu CY, Hsuan CF, Chang WT, Yang KC. Expedition to the missing link: Long noncoding RNAs in cardiovascular diseases. J Biomed Sci 2020; 27:48. [PMID: 32241300 PMCID: PMC7114803 DOI: 10.1186/s12929-020-00647-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/27/2020] [Indexed: 12/31/2022] Open
Abstract
With the advances in deep sequencing-based transcriptome profiling technology, it is now known that human genome is transcribed more pervasively than previously thought. Up to 90% of the human DNA is transcribed, and a large proportion of the human genome is transcribed as long noncoding RNAs (lncRNAs), a heterogenous group of non-coding transcripts longer than 200 nucleotides. Emerging evidence suggests that lncRNAs are functional and contribute to the complex regulatory networks involved in cardiovascular development and diseases. In this article, we will review recent evidence on the roles of lncRNAs in the biological processes of cardiovascular development and disorders. The potential applications of lncRNAs as biomarkers and targets for therapeutics are also discussed.
Collapse
Affiliation(s)
- Chih-Fan Yeh
- Graduate Institute and Department of Pharmacology, National Taiwan University School of Medicine, No.1, Sec. 1, Ren-Ai Rd, 1150R, Taipei, Taiwan.,Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, No.1, Sec. 1, Ren-Ai Rd, 1150R, Taipei, Taiwan
| | - Yu-Chen Eugene Chang
- Graduate Institute and Department of Pharmacology, National Taiwan University School of Medicine, No.1, Sec. 1, Ren-Ai Rd, 1150R, Taipei, Taiwan
| | - Cheng-Yuan Lu
- Graduate Institute and Department of Pharmacology, National Taiwan University School of Medicine, No.1, Sec. 1, Ren-Ai Rd, 1150R, Taipei, Taiwan
| | - Chin-Feng Hsuan
- Division of Cardiology, Department of Internal Medicine, E-Da Dachang Hospital, Kaohsiung, Taiwan.,Department of Medicine, I-Shou University School of Medicine, Kaohsiung, Taiwan
| | - Wei-Tien Chang
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kai-Chien Yang
- Graduate Institute and Department of Pharmacology, National Taiwan University School of Medicine, No.1, Sec. 1, Ren-Ai Rd, 1150R, Taipei, Taiwan. .,Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, No.1, Sec. 1, Ren-Ai Rd, 1150R, Taipei, Taiwan.
| |
Collapse
|
41
|
Jusic A, Devaux Y. Mitochondrial noncoding RNA-regulatory network in cardiovascular disease. Basic Res Cardiol 2020; 115:23. [PMID: 32140778 DOI: 10.1007/s00395-020-0783-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/17/2020] [Indexed: 12/22/2022]
Abstract
Mitochondrial function and integrity are vital for the maintenance of cellular homeostasis, particularly in high-energy demanding cells. Cardiomyocytes have a large number of mitochondria, which provide a continuous and bulk supply of the ATP necessary for cardiac mechanical function. More than 90% of the ATP consumed by the heart is derived from the mitochondrial oxidative metabolism. Decreased energy supply as the main consequence of mitochondrial dysfunction is closely linked to cardiovascular disease (CVD). The discovery of noncoding RNA (ncRNAs) in the mitochondrial compartment has changed the traditional view of molecular pathways involved in the regulatory network of CVD. Mitochondrial ncRNAs participate in controlling cardiovascular pathogenesis by regulating glycolysis, mitochondrial energy status, and the expression of genes involved in mitochondrial metabolism. Understanding the underlying mechanisms of the association between impaired mitochondrial function resulting from fluctuation in expression levels of ncRNAs and specific disease phenotype can aid in preventing and treating CVD. This review presents an overview of the role of mitochondrial ncRNAs in the complex regulatory network of the cardiovascular pathology. We will summarize and discuss (1) mitochondrial microRNAs (mitomiRs) and long noncoding RNAs (lncRNAs) encoded either by nuclear or mitochondrial genome which are involved in the regulation of mitochondrial metabolism; (2) the role of mitomiRs and lncRNAs in the pathogenesis of several CVD such as hypertension, cardiac hypertrophy, acute myocardial infarction and heart failure; (3) the biomarker and therapeutic potential of mitochondrial ncRNAs in CVD; (4) and the challenges inherent to their translation into clinical application.
Collapse
Affiliation(s)
- Amela Jusic
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, 1A-B rue Edison, 1445, Strassen, Luxembourg.
| | | |
Collapse
|
42
|
Xu L, Wang H, Jiang F, Sun H, Zhang D. LncRNA AK045171 protects the heart from cardiac hypertrophy by regulating the SP1/MG53 signalling pathway. Aging (Albany NY) 2020; 12:3126-3139. [PMID: 32087602 PMCID: PMC7066930 DOI: 10.18632/aging.102668] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 12/24/2019] [Indexed: 12/15/2022]
Abstract
Hearts often undergo abnormal remodelling and hypertrophic growth in response to pathological stress. Long non-coding RNAs (LncRNAs) can change cardiac function and participate in regulation of cardiac hypertrophy. The present study aims to identify the role of AK045171 in cardiac hypertrophy and the underlying mechanism in hypertrophic cascades. Mice with cardiac hypertrophy were established through transverse aortic constriction (TAC). Cardiac hypertrophy in cardiomyocytes was induced by angiotensin II (angII). The expression of AK045171 and its target gene SP1 was examined in cardiomyocytes transfected with miRNA. The AK045171 expression level was downregulated in mice after TAC surgery. Overexpression of AK045171 attenuated cardiac hypertrophy both in vitro and in vivo. The mechanism study indicated that AK045171 binds with SP1, which promotes transcription activation of MEG3. It is suggested that overexpression of AK045171 might have clinical potential to suppress cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Li Xu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Hongjiang Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Feng Jiang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Hao Sun
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Dapeng Zhang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
43
|
Fang Q, Liu T, Yu C, Yang X, Shao Y, Shi J, Ye X, Zheng X, Yan J, Xu D, Zou X. LncRNA TUG1 alleviates cardiac hypertrophy by targeting miR-34a/DKK1/Wnt-β-catenin signalling. J Cell Mol Med 2020; 24:3678-3691. [PMID: 32057178 PMCID: PMC7131932 DOI: 10.1111/jcmm.15067] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/29/2019] [Accepted: 01/06/2020] [Indexed: 02/06/2023] Open
Abstract
The current study was designed to explore the role and underlying mechanism of lncRNA taurine up‐regulated gene 1 (TUG1) in cardiac hypertrophy. Mice were treated by transverse aortic constriction (TAC) surgery to induce cardiac hypertrophy, and cardiomyocytes were treated by phenylephrine (PE) to induce hypertrophic phenotype. Haematoxylin‐eosin (HE), wheat germ agglutinin (WGA) and immunofluorescence (IF) were used to examine morphological alterations. Real‐time PCR, Western blots and IF staining were used to detect the expression of RNAs and proteins. Luciferase assay and RNA pull‐down assay were used to verify the interaction. It is revealed that TUG1 was up‐regulated in the hearts of mice treated by TAC surgery and in PE‐induced cardiomyocytes. Functionally, overexpression of TUG1 alleviated cardiac hypertrophy both in vivo and in vitro. Mechanically, TUG1 sponged and sequestered miR‐34a to increase the Dickkopf 1 (DKK1) level, which eventually inhibited the activation of Wnt/β‐catenin signalling. In conclusion, the current study reported the protective role and regulatory mechanism of TUG1 in cardiac hypertrophy and suggested that TUG1 may serve as a novel molecular target for treating cardiac hypertrophy.
Collapse
Affiliation(s)
- Qingxia Fang
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ting Liu
- Department of pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenhuan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Xiuli Yang
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yanfei Shao
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jiana Shi
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiaolan Ye
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiaochun Zheng
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jieping Yan
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Danfeng Xu
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiaozhou Zou
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou, China.,People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
44
|
Gomes CPDC, Schroen B, Kuster GM, Robinson EL, Ford K, Squire IB, Heymans S, Martelli F, Emanueli C, Devaux Y, On behalf of the EU-CardioRNA COST Action (CA17129). Regulatory RNAs in Heart Failure. Circulation 2020; 141:313-328. [PMID: 31986093 PMCID: PMC7012349 DOI: 10.1161/circulationaha.119.042474] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
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
|
45
|
Rahmani Z, Mojarrad M, Moghbeli M. Long non-coding RNAs as the critical factors during tumor progressions among Iranian population: an overview. Cell Biosci 2020; 10:6. [PMID: 31956395 PMCID: PMC6961246 DOI: 10.1186/s13578-020-0373-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cancer is associated with various genetic and environmental risk factors. Beside the mutations or aberrant expression of protein-coding genes, the genetic deregulation of non-coding RNAs has also an important role during tumor progression and metastasis. Long non-coding RNAs (lncRNAs) are a class of ncRNAs larger than 200 nucleotides that may function as tumor-suppressor or oncogene. MAIN BODY There is a raising trend of cancer incidence among Iranian population during the last decades. Therefore, it is required to prepare a general population specific panel of genetic markers for the early detection of cancer in this population. The tissue-specific expression characteristics and high stability in body fluids highlight the lncRNAs as efficient diagnostic and prognostic noninvasive biomarkers in cancer. In present review we summarized all of the lncRNAs which have been reported until now in different tumors among Iranian patients. CONCLUSIONS This review paves the way of introducing a population based noninvasive diagnostic panel of lncRNAs for the early detection of tumor cells among Iranian population.
Collapse
Affiliation(s)
- Zahra Rahmani
- Department of Medical Genetics, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Mojarrad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
46
|
Gabriel AF, Costa MC, Enguita FJ. Interactions Among Regulatory Non-coding RNAs Involved in Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:79-104. [PMID: 32285406 DOI: 10.1007/978-981-15-1671-9_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Non-coding RNAs (ncRNAs) are important regulatory players in human cells that have been shown to modulate different cellular processes and biological functions through controlling gene expression, being also involved in pathological conditions such as cardiovascular diseases. Among them, long non-coding RNAs (lncRNAs) and circular (circRNAs) could act as competing endogenous RNAs (ceRNAs) sequestering other ncRNAs. This entangled network of interactions has been reported to trigger the decay of the targeted ncRNAs having important roles in gene regulation. Growing evidences have been demonstrated that the regulatory mechanism underlying the crosstalk between different ncRNA species, namely lncRNAs, circRNAs and miRNAs has also an important role in the pathophysiological processes of cardiovascular diseases. In this chapter, the main regulatory relationship among lncRNAs, circRNAs and miRNAs were summarized and their role in the control and development of cardiovascular diseases was highlighted.
Collapse
Affiliation(s)
- André F Gabriel
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Cardiomics Unit, Centro de Cardiologia da Universidade de Lisboa (CCUL), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Marina C Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Cardiomics Unit, Centro de Cardiologia da Universidade de Lisboa (CCUL), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Francisco J Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal. .,Cardiomics Unit, Centro de Cardiologia da Universidade de Lisboa (CCUL), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
47
|
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: 1.0] [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
|
48
|
Sun Y, Fan W, Xue R, Dong B, Liang Z, Chen C, Li J, Wang Y, Zhao J, Huang H, Jiang J, Wu Z, Dai G, Fang R, Yan Y, Yang T, Huang ZP, Dong Y, Liu C. Transcribed Ultraconserved Regions, Uc.323, Ameliorates Cardiac Hypertrophy by Regulating the Transcription of CPT1b (Carnitine Palmitoyl transferase 1b). Hypertension 2019; 75:79-90. [PMID: 31735087 DOI: 10.1161/hypertensionaha.119.13173] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transcribed ultraconserved regions (T-UCRs) are a novel class of long noncoding RNAs transcribed from UCRs, which exhibit 100% DNA sequence conservation among humans, mice, and rats. However, whether T-UCRs regulate cardiac hypertrophy remains unclear. We aimed to explore the effects of T-UCRs on cardiac hypertrophy. First, we performed long noncoding RNA microarray analysis on hearts of mice subjected to sham surgery or aortic banding and found that the T-UCR uc.323 was decreased significantly in mice with aortic banding-induced cardiac hypertrophy. In vitro loss- and gain-of-function experiments demonstrated that uc.323 protected cardiomyocytes against hypertrophy induced by phenylephrine. Additionally, we discovered that mammalian target of rapamycin 1 contributed to phenylephrine-induced uc.323 downregulation and uc.323-mediated cardiomyocyte hypertrophy. We further mapped the possible target genes of uc.323 through global microarray mRNA expression analysis after uc.323 knockdown and found that uc.323 regulated the expression of cardiac hypertrophy-related genes such as CPT1b (Carnitine Palmitoyl transferase 1b). Then, chromatin immunoprecipitation proved that EZH2 (enhancer of zeste homolog 2) bound to the promoter of CPT1b via H3K27me3 (trimethylation of lysine 27 of histone H3) to induce CPT1b downregulation. And overexpression of CPT1b could block uc.323-mediated cardiomyocyte hypertrophy. Finally, we found that uc.323 deficiency induced cardiac hypertrophy. Our results reveal that uc.323 is a conserved T-UCR that inhibits cardiac hypertrophy, potentially by regulating the transcription of CPT1b via interaction with EZH2.
Collapse
Affiliation(s)
- Yu Sun
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,Department of Cardiology, the Second People's Hospital of Guangdong Province, Guangzhou, Guangdong, China (Y.S.).,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Wendong Fan
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Ruicong Xue
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Bin Dong
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Zhuomin Liang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Chen Chen
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Jiayong Li
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Yan Wang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Jingjing Zhao
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Huiling Huang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Jingzhou Jiang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Zexuan Wu
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Gang Dai
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Rong Fang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Youchen Yan
- Department of Cardiology, Center for Translational Medicine (Y.Y., T.Y.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Tiqun Yang
- Department of Cardiology, Center for Translational Medicine (Y.Y., T.Y.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhan-Peng Huang
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Yugang Dong
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| | - Chen Liu
- From the Department of Cardiology (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.), the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, Guangdong, China (Y.S., W.F., R.X., B.D., Z.L., C.C., J.L., Y.W., J.Z., H.H., J.J., Z.W., G.D., R.F., Z.-p.H., Y.D., C.L.)
| |
Collapse
|
49
|
Sweta S, Dudnakova T, Sudheer S, Baker AH, Bhushan R. Importance of Long Non-coding RNAs in the Development and Disease of Skeletal Muscle and Cardiovascular Lineages. Front Cell Dev Biol 2019; 7:228. [PMID: 31681761 PMCID: PMC6813187 DOI: 10.3389/fcell.2019.00228] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022] Open
Abstract
The early mammalian embryo is characterized by the presence of three germ layers-the outer ectoderm, middle mesoderm and inner endoderm. The mesoderm is organized into paraxial, intermediate and lateral plate mesoderm. The musculature, vasculature and heart of the adult body are the major derivatives of mesoderm. Tracing back the developmental process to generate these specialized tissues has sparked much interest in the field of regenerative medicine focusing on generating specialized tissues to treat patients with degenerative diseases. Several Long Non-Coding RNAs (lncRNAs) have been identified as regulators of development, proliferation and differentiation of various tissues of mesodermal origin. A better understanding of lncRNAs that can regulate the development of these tissues will open potential avenues for their therapeutic utility and enhance our knowledge about disease progression and development. In this review, we aim to summarize the functions and mechanisms of lncRNAs regulating the early mesoderm differentiation, development and homeostasis of skeletal muscle and cardiovascular system with an emphasis on their therapeutic potential.
Collapse
Affiliation(s)
- Sweta Sweta
- Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, India
| | - Tatiana Dudnakova
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Smita Sudheer
- Department of Genomic Science, Central University of Kerala, Kasaragod, India
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Raghu Bhushan
- Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, India
| |
Collapse
|
50
|
Cresci S, Pereira NL, Ahmad F, Byku M, de las Fuentes L, Lanfear DE, Reilly CM, Owens AT, Wolf MJ. Heart Failure in the Era of Precision Medicine: A Scientific Statement From the American Heart Association. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2019; 12:458-485. [PMID: 31510778 DOI: 10.1161/hcg.0000000000000058] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One of 5 people will develop heart failure over his or her lifetime. Early diagnosis and better understanding of the pathophysiology of this disease are critical to optimal treatment. The "omics"-genomics, pharmacogenomics, epigenomics, proteomics, metabolomics, and microbiomics- of heart failure represent rapidly expanding fields of science that have, to date, not been integrated into a single body of work. The goals of this statement are to provide a comprehensive overview of the current state of these omics as they relate to the development and progression of heart failure and to consider the current and potential future applications of these data for precision medicine with respect to prevention, diagnosis, and therapy.
Collapse
|