101
|
Fan J, Li H, Xie R, Zhang X, Nie X, Shi X, Zhan J, Yin Z, Zhao Y, Dai B, Yuan S, Wen Z, Chen C, Wang DW. LncRNA ZNF593-AS Alleviates Contractile Dysfunction in Dilated Cardiomyopathy. Circ Res 2021; 128:1708-1723. [PMID: 33550812 DOI: 10.1161/circresaha.120.318437] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jiahui Fan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Rong Xie
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Xudong Zhang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Xiang Nie
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Xiaolu Shi
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China (X.S.)
| | - Jiabing Zhan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Zhongwei Yin
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Yanru Zhao
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Beibei Dai
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Shuai Yuan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Zheng Wen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.).,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China (J.F., H.L., R.X., X.Z., X.N., J.Z., Z.Y., Y.Z., B.D., S.Y., Z.W., C.C., D.W.W.)
| |
Collapse
|
102
|
Huang Y, Liu H, Guo R, Han Y, Yang Y, Zhao Y, Zheng Y, Jia L, Li W. Long Non-coding RNA FER1L4 Mediates the Autophagy of Periodontal Ligament Stem Cells Under Orthodontic Compressive Force via AKT/FOXO3 Pathway. Front Cell Dev Biol 2021; 9:631181. [PMID: 33604341 PMCID: PMC7884613 DOI: 10.3389/fcell.2021.631181] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/11/2021] [Indexed: 01/11/2023] Open
Abstract
Orthodontic tooth movement is achieved by periodontal tissue remodeling triggered by mechanical force. It is essential to investigate the reaction of periodontal ligament stem cells (PDLSCs) for improving orthodontic therapeutic approaches. Autophagy is an endogenous defense mechanism to prevent mechanical damage of environmental change. Long non-coding RNAs (lncRNAs) are key regulators in gene regulation, but their roles are still largely uncharacterized in the reaction of PDLSCs during orthodontic tooth movement. In this study, we showed that autophagy was significantly induced in PDLSCs under compressive force, as revealed by the markers of autophagy, microtubule-associated protein light chain 3 (LC3) II/I and Beclin1, and the formation of autophagosomes. After the application of compressive force, lncRNA FER1L4 was strongly upregulated. Overexpression of FER1L4 increased the formation of autophagosome and autolysosomes in PDLSCs, while knockdown of FER1L4 reversed the autophagic activity induced by mechanical force. In mechanism, FER1L4 inhibited the phosphorylation of protein kinase B (AKT) and subsequently increased the nuclear translocation of forkhead box O3 (FOXO3) and thus mediated autophagic cascades under compressive strain. In mouse model, the expression of Lc3 as well as Fer1l4 was increased in the pressure side of periodontal ligament during tooth movement. These findings suggest a novel mechanism of autophagy regulation by lncRNA during periodontal tissue remodeling of orthodontic treatment.
Collapse
Affiliation(s)
- Yiping Huang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Hao Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Runzhi Guo
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yineng Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yuhui Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yi Zhao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yunfei Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Lingfei Jia
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Weiran Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
103
|
Sumi MP, Mahajan B, Sattar RSA, Nimisha, Apurva, Kumar A, Sharma AK, Ahmad E, Ali A, Saluja SS. Elucidation of Epigenetic Landscape in Coronary Artery Disease: A Review on Basic Concept to Personalized Medicine. Epigenet Insights 2021; 14:2516865720988567. [PMID: 33598635 PMCID: PMC7863167 DOI: 10.1177/2516865720988567] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/26/2020] [Indexed: 12/11/2022] Open
Abstract
Despite extensive clinical research and management protocols applied in the field of coronary artery diseases (CAD), it still holds the number 1 position in mortality worldwide. This indicates that we need to work on precision medicine to discover the diagnostic, therapeutic, and prognostic targets to improve the outcome of CAD. In precision medicine, epigenetic changes play a vital role in disease onset and progression. Epigenetics is the study of heritable changes that do not affect the alterations of DNA sequence in the genome. It comprises various covalent modifications that occur in DNA or histone proteins affecting the spatial arrangement of the DNA and histones. These multiple modifications include DNA/histone methylation, acetylation, phosphorylation, and SUMOylation. Besides these covalent modifications, non-coding RNAs-viz. miRNA, lncRNA, and circRNA are also involved in epigenetics. Smoking, alcohol, diet, environmental pollutants, obesity, and lifestyle are some of the prime factors affecting epigenetic alterations. Novel molecular techniques such as next-generation sequencing, chromatin immunoprecipitation, and mass spectrometry have been developed to identify important cross points in the epigenetic web in relation to various diseases. The studies regarding exploration of epigenetics, have led researchers to identify multiple diagnostic markers and therapeutic targets that are being used in different disease diagnosis and management. Here in this review, we will discuss various ground-breaking contributions of past and recent studies in the epigenetic field in concert with coronary artery diseases. Future prospects of epigenetics and its implication in CAD personalized medicine will also be discussed in brief.
Collapse
Affiliation(s)
- Mamta P Sumi
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Bhawna Mahajan
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
- Department of Biochemistry, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Real Sumayya Abdul Sattar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Apurva
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Arun Kumar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Abhay Kumar Sharma
- Department of Biochemistry, All India Institute of Medical Science, Patna, Bihar, India
| | - Ejaz Ahmad
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| | - Asgar Ali
- Department of Biochemistry, All India Institute of Medical Science, Patna, Bihar, India
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Post Graduate Medical Education and Research (GIPMER), University of Delhi, New Delhi, India
| |
Collapse
|
104
|
Liao H, Qi Y, Ye Y, Yue P, Zhang D, Li Y. Mechanotranduction Pathways in the Regulation of Mitochondrial Homeostasis in Cardiomyocytes. Front Cell Dev Biol 2021; 8:625089. [PMID: 33553165 PMCID: PMC7858659 DOI: 10.3389/fcell.2020.625089] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are one of the most important organelles in cardiomyocytes. Mitochondrial homeostasis is necessary for the maintenance of normal heart function. Mitochondria perform four major biological processes in cardiomyocytes: mitochondrial dynamics, metabolic regulation, Ca2+ handling, and redox generation. Additionally, the cardiovascular system is quite sensitive in responding to changes in mechanical stress from internal and external environments. Several mechanotransduction pathways are involved in regulating the physiological and pathophysiological status of cardiomyocytes. Typically, the extracellular matrix generates a stress-loading gradient, which can be sensed by sensors located in cellular membranes, including biophysical and biochemical sensors. In subsequent stages, stress stimulation would regulate the transcription of mitochondrial related genes through intracellular transduction pathways. Emerging evidence reveals that mechanotransduction pathways have greatly impacted the regulation of mitochondrial homeostasis. Excessive mechanical stress loading contributes to impairing mitochondrial function, leading to cardiac disorder. Therefore, the concept of restoring mitochondrial function by shutting down the excessive mechanotransduction pathways is a promising therapeutic strategy for cardiovascular diseases. Recently, viral and non-viral protocols have shown potentials in application of gene therapy. This review examines the biological process of mechanotransduction pathways in regulating mitochondrial function in response to mechanical stress during the development of cardiomyopathy and heart failure. We also summarize gene therapy delivery protocols to explore treatments based on mechanical stress-induced mitochondrial dysfunction, to provide new integrative insights into cardiovascular diseases.
Collapse
Affiliation(s)
- Hongyu Liao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yan Qi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yida Ye
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Peng Yue
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, 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 Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
105
|
Long Non-Coding RNAs (lncRNAs) in Cardiovascular Disease Complication of Type 2 Diabetes. Diagnostics (Basel) 2021; 11:diagnostics11010145. [PMID: 33478141 PMCID: PMC7835902 DOI: 10.3390/diagnostics11010145] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
The discovery of non-coding RNAs (ncRNAs) has opened a new paradigm to use ncRNAs as biomarkers to detect disease progression. Long non-coding RNAs (lncRNA) have garnered the most attention due to their specific cell-origin and their existence in biological fluids. Type 2 diabetes patients will develop cardiovascular disease (CVD) complications, and CVD remains the top risk factor for mortality. Understanding the lncRNA roles in T2D and CVD conditions will allow the future use of lncRNAs to detect CVD complications before the symptoms appear. This review aimed to discuss the roles of lncRNAs in T2D and CVD conditions and their diagnostic potential as molecular biomarkers for CVD complications in T2D.
Collapse
|
106
|
Sun C, Ni M, Song B, Cao L. Circulating Circular RNAs: Novel Biomarkers for Heart Failure. Front Pharmacol 2020; 11:560537. [PMID: 33281603 PMCID: PMC7691568 DOI: 10.3389/fphar.2020.560537] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 09/21/2020] [Indexed: 12/21/2022] Open
Abstract
Heart failure (HF) is a serious, chronic disease, causing significant ill health and high mortality worldwide. The current clinical strategies emphasize reducing the transition from a healthy to a failing heart despite the shift in the clinical goal from healing to disease prevention. Recent research advancements on noncoding RNAs (ncRNAs) have demonstrated that circular RNAs (circRNAs) are significant therapeutic targets in HF. Previous studies have highlighted the potential applicability of circRNAs in the diagnosis and treatment of diseases. However, less is known regarding the potential benefits of circRNAs as novel diagnostic and treatment biomarkers for HF. In the present study, we summarize the current developments and achievements associated with the use of circRNAs as HF biomarkers. We also discuss future research directions regarding HF diagnosis and treatment.
Collapse
Affiliation(s)
- Chuan Sun
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Mingming Ni
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Bo Song
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lu Cao
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
107
|
Li H, Shi H, Zhang F, Xue H, Wang L, Tian J, Xu J, Han Q. LncRNA Tincr regulates PKCɛ expression in a miR-31-5p-dependent manner in cardiomyocyte hypertrophy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:2495-2506. [PMID: 32157348 DOI: 10.1007/s00210-020-01847-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 02/28/2020] [Indexed: 12/16/2022]
Abstract
Cardiomyocyte hypertrophy is a fatal factor in heart disease resulting in heart failure and even mortality. Although many studies have been focusing on the pathogenesis of cardiomyocyte hypertrophy, the exact molecular mechanisms are still unexclusive. In this study, we first found that the expression level of lncRNA Tincr was significantly decreased in the myocardial tissues of TAC mouse models of cardiomyocyte hypertrophy, and this result was further confirmed in H9C2 cells, a widely used rat myoblast cell lines. More intriguingly, we demonstrated that the aberration of Tincr is essential to the pathogenesis of cardiomyocyte hypertrophy, indicated by the re-induction of Tincr improving the heart functions of hypertrophic mice. In mechanism, we identified miR-31-5p as a direct target of Tincr using a widely used online bioinformatics tool StarBase, and this result was further experimentally validated using dual-luciferase reporter assay and real-time PCR. Also, we identified PRKCE as a direct target of miR-31-5p, and loss function of miR-31-5p significantly blocks the positive regulatory effect of Tincr on PRKCE expression in H9C2 cells. The knockdown of Tincr resulted in increased cardiomyocyte size, and, however, inhibition of miR-31-5p or overexpression of PRKCE significantly reversed the increased cardiomyocyte size. Taken together, our study showed that a novel Tincr-miR-31-5p axis targeting PRKCE was involved in cardiomyocyte hypertrophy, indicating that it may provide potential therapy in cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Hao Li
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Hongtao Shi
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Fan Zhang
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Honghong Xue
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Lei Wang
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- The Affiliated Cardiovascular Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Jing Tian
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Jianrong Xu
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Qinghua Han
- The First Clinical Medical School, Shanxi Medical University, No. 56 Xinjian South Road, Taiyuan, 030001, Shanxi, People's Republic of China.
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China.
| |
Collapse
|
108
|
Simion V, Zhou H, Haemmig S, Pierce JB, Mendes S, Tesmenitsky Y, Pérez-Cremades D, Lee JF, Chen AF, Ronda N, Papotti B, Marto JA, Feinberg MW. A macrophage-specific lncRNA regulates apoptosis and atherosclerosis by tethering HuR in the nucleus. Nat Commun 2020; 11:6135. [PMID: 33262333 PMCID: PMC7708640 DOI: 10.1038/s41467-020-19664-2] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 10/26/2020] [Indexed: 01/10/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are emerging regulators of pathophysiological processes including atherosclerosis. Using RNA-seq profiling of the intima of lesions, here we identify a macrophage-specific lncRNA MAARS (Macrophage-Associated Atherosclerosis lncRNA Sequence). Aortic intima expression of MAARS increases by 270-fold with atherosclerotic progression and decreases with regression by 60%. MAARS knockdown reduces atherosclerotic lesion formation by 52% in LDLR-/- mice, largely independent of effects on lipid profile and inflammation, but rather by decreasing macrophage apoptosis and increasing efferocytosis in the vessel wall. MAARS interacts with HuR/ELAVL1, an RNA-binding protein and important regulator of apoptosis. Overexpression and knockdown studies verified MAARS as a critical regulator of macrophage apoptosis and efferocytosis in vitro, in an HuR-dependent manner. Mechanistically, MAARS knockdown alters HuR cytosolic shuttling, regulating HuR targets such as p53, p27, Caspase-9, and BCL2. These findings establish a mechanism by which a macrophage-specific lncRNA interacting with HuR regulates apoptosis, with implications for a broad range of vascular disease states.
Collapse
Affiliation(s)
- Viorel Simion
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Haoyang Zhou
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Stefan Haemmig
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jacob B Pierce
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Shanelle Mendes
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yevgenia Tesmenitsky
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel Pérez-Cremades
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - James F Lee
- The Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alex F Chen
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Jarrod A Marto
- The Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Departments of Cancer Biology and Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark W Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
109
|
Zhang F, Fu X, Kataoka M, Liu N, Wang Y, Gao F, Liang T, Dong X, Pei J, Hu X, Zhu W, Yu H, Cowan DB, Hu X, Huang ZP, Wang J, Wang DZ, Chen J. Long noncoding RNA Cfast regulates cardiac fibrosis. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 23:377-392. [PMID: 33473324 PMCID: PMC7787992 DOI: 10.1016/j.omtn.2020.11.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
Cardiac fibrosis occurs in most cardiac diseases, which reduces cardiac muscle compliance, impairs both systolic and diastolic heart function and, ultimately, leads to heart failure. Long noncoding RNAs (lncRNAs) have recently emerged as important regulators of a variety of biological processes; however, little is known about the expression and function of lncRNAs in cardiac fibrosis. Using unbiased transcriptome profiling in a mouse model of myocardial infarction (MI), we identified a cardiac fibroblast-enriched lncRNA (AK048087) named cardiac fibroblast-associated transcript (Cfast), which is significantly elevated after MI. Silencing Cfast expression by small interfering RNAs (siRNAs) or lentiviral short hairpin RNAs (shRNAs) resulted in suppression of fibrosis-related gene expression and transdifferentiation of myofibroblasts into cardiac fibroblasts. Depletion of Cfast by lentiviral shRNAs in mouse hearts significantly attenuated cardiac fibrosis induced by MI or isoproterenol-infusion. Importantly, inhibition of Cfast ameliorated cardiac function following cardiac injury. RNA pull-down followed by mass spectrometry analyses identified COTL1 (coactosin-like 1) as one of the Cfast interacting proteins. Mechanistically, Cfast competitively inhibits the COTL1 interaction with TRAP1 (transforming growth factor-β receptor-associated protein 1), which enhances TGF-β signaling by augmenting SMAD2/SMAD4 complex formation. Therefore, our study identifies Cfast as a novel cardiac fibroblast-enriched lncRNA that regulates cardiac fibroblast activation in response to pathophysiological stress. Cfast could serve as a potential therapeutic target for the prevention of cardiac fibrosis and cardiac diseases.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Xuyang Fu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Masaharu Kataoka
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Ning Liu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Yingchao Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Gao
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Tian Liang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Xiaoxuan Dong
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Jianqiu Pei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Wei Zhu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Hong Yu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Douglas B Cowan
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Xinyang Hu
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, 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
| | - Jian'an Wang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Jinghai Chen
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| |
Collapse
|
110
|
Zhang X, Feng T, Zeng XXI, Liang H, Situ B, Zhang Q, Zhou F, Chen Y, Wang T, Cai D, Lin X, Xiu J, Zheng L. Identification of Transcriptional Variation in Aortic Remodeling Using a Murine Transverse Aortic Constriction (TAC) Model. Front Cardiovasc Med 2020; 7:581362. [PMID: 33304927 PMCID: PMC7693635 DOI: 10.3389/fcvm.2020.581362] [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: 07/08/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Arterial remodeling is a major pathological consequence of hypertension, which is recognized as the most common chronic non-communicable disease. However, the detailed mechanism of how arterial remodeling is induced by hypertension has not yet been fully elucidated. Evaluating the transcriptional changes in arterial tissue in response to elevated blood pressure at an early stage may provide new insights and identify novel therapeutic candidates in preventing arterial remodeling. Here, we used the ascending aorta of the transverse aortic constriction (TAC) model to induce arterial remodeling in C57BL/6 male mice. Age-matched mice were subjected to sham surgery as controls. The TAC model was only considered successful if the mice conformed to the criteria (RC/LC blood flow velocity with 5–10-fold change) 1 week after the surgery. Two weeks after surgery, the ascending aorta developed severe remodeling in TAC mice as compared to the sham group. High throughput sequencing was then applied to identify differentially expressed (DE) transcripts. In silicon analysis were then performed to systematically network transcriptional changes. A total of 1,019 mRNAs were significantly changed between TAC and the sham group at the transcriptional level. GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis revealed that stress/stimulus/immune-related biological processes played a crucial role during arterial remodeling. Our data provide a comprehensive understanding of global gene expression changes in the TAC model, which suggests that targeting inflammation and vascular smooth cell transformation are potential therapeutic strategies to interfere with the aortic remodeling at an early stage in the development of hypertension.
Collapse
Affiliation(s)
- Xinlu Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ting Feng
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin-Xin I Zeng
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Hongbin Liang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bo Situ
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiuxia Zhang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fengyun Zhou
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yejia Chen
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Du Cai
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinxin Lin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiancheng Xiu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
111
|
Gao J, Chen X, Shan C, Wang Y, Li P, Shao K. Autophagy in cardiovascular diseases: role of noncoding RNAs. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 23:101-118. [PMID: 33335796 PMCID: PMC7732971 DOI: 10.1016/j.omtn.2020.10.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular diseases (CVDs) remain the world's leading cause of death. Cardiomyocyte autophagy helps maintain normal metabolism and functioning of the heart. Importantly, mounting evidence has revealed that autophagy plays a dual role in CVD pathology. Under physiological conditions, moderate autophagy maintains cell metabolic balance by degrading and recycling damaged organelles and proteins, and it promotes myocardial survival, but excessive or insufficient autophagy is equally deleterious and contributes to disease progression. Noncoding RNAs (ncRNAs) are a class of RNAs transcribed from the genome, but most ncRNAs do not code for functional proteins. In recent years, increasingly, various ncRNAs have been identified, and they play important regulatory roles in the physiological and pathological processes of organisms, as well as in autophagy. Thus, determining whether ncRNA-regulated autophagy plays a protective role in CVDs or promotes their progression can help us to develop ncRNAs as therapeutic targets in autophagy-related CVDs. In this review, we briefly summarize the regulatory roles of several important ncRNAs, including microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), in the autophagy of various CVDs to provide a theoretical basis for the etiology and pathogenesis of CVDs and develop novel therapies to treat CVDs.
Collapse
Affiliation(s)
- Jinning Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Xiatian Chen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Chan Shan
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Kai Shao
- Department of Central Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
| |
Collapse
|
112
|
Ren K, Xu XD, Yu XH, Li MQ, Shi MW, Liu QX, Jiang T, Zheng XL, Yin K, Zhao GJ. LncRNA-modulated autophagy in plaque cells: a new paradigm of gene regulation in atherosclerosis? Aging (Albany NY) 2020; 12:22335-22349. [PMID: 33154191 PMCID: PMC7695379 DOI: 10.18632/aging.103786] [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: 04/21/2020] [Accepted: 07/14/2020] [Indexed: 12/25/2022]
Abstract
The development of atherosclerosis is accompanied by the functional deterioration of plaque cells, which leads to the escalation of endothelial inflammation, abnormal vascular smooth muscle cell phenotype switching and the accumulation of lipid-laden macrophages within vascular walls. Autophagy, a highly conserved homeostatic mechanism, is critical for the delivery of cytoplasmic substrates to lysosomes for degradation. Moderate levels of autophagy prevent atherosclerosis by safeguarding plaque cells against apoptosis, preventing inflammation, and limiting the lipid burden, whereas excessive autophagy exacerbates cell damage and inflammation and thereby accelerates the formation of atherosclerotic plaques. Increasing lines of evidence suggest that long noncoding RNAs can be either beneficial or detrimental to atherosclerosis development by regulating the autophagy level. This review summarizes the research progress related to 1) the significant role of autophagy in atherosclerosis and 2) the effects of the lncRNA-mediated modulation of autophagy on the plaque cell fate, inflammation levels, proliferative capacity, and cholesterol metabolism and subsequently on atherogenesis.
Collapse
Affiliation(s)
- Kun Ren
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, Guangdong, China.,Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Xiao-Dan Xu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiao-Hai Yu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Meng-Qi Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, Guangdong, China
| | - Meng-Wen Shi
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Qi-Xian Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Ting Jiang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, Guangdong, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, University of Calgary, Health Sciences Center, Calgary, AB, Canada.,Key Laboratory of Molecular Targets and Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| | - Guo-Jun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, Guangdong, China
| |
Collapse
|
113
|
Alexanian M, Ounzain S. Long Noncoding RNAs in Cardiac Development. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037374. [PMID: 31932317 DOI: 10.1101/cshperspect.a037374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The exquisite transcriptional control of developmental gene programs is critical for hardwiring the complex expression patterns that govern cell-fate determination and differentiation during heart development. During the past several years, studies have illuminated our understanding of a complex noncoding transcriptional landscape, primarily associated with long noncoding RNAs (lncRNAs), that is implicated in these developmental processes and has begun to reveal key functions of these transcripts. In this review, we discuss the expanding roles for lncRNAs in the earliest points of cardiac development and through differentiation and maturation of multiple cell types within the adult heart. We go on to outline the diverse mechanisms by which cardiovascular lncRNAs orchestrate these transcriptional programs, explore the challenges linked to the study of lncRNAs in developmental phenotypes, and summarize the implications for these molecules in human cardiovascular disorders.
Collapse
Affiliation(s)
| | - Samir Ounzain
- HAYA Therapeutics SA, Lausanne, Vaud 1066, Switzerland
| |
Collapse
|
114
|
Zhang Z, Wan J, Liu X, Zhang W. Strategies and technologies for exploring long noncoding RNAs in heart failure. Biomed Pharmacother 2020; 131:110572. [PMID: 32836073 DOI: 10.1016/j.biopha.2020.110572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/23/2020] [Accepted: 07/26/2020] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNA (lncRNA) was once considered to be the "noise" of genome transcription without biological function. However, increasing evidence shows that lncRNA is dynamically expressed in developmental stage or disease status, playing a regulatory role in the process of gene expression and translation. In recent years, lncRNA is considered to be a core node of functional regulatory networks that controls cardiac and also involves in multiple process of heart failure such as myocardial hypertrophy, fibrosis, angiogenesis, etc., which would be a therapeutic target for diseases. In fact, it is the development of technology that has improved our understanding of lncRNAs and broadened our perspective on heart failure. From transcriptional "noise" to star molecule, progress of lncRNAs can't be achieved without the combination of multidisciplinary technologies, especially the emergence of high-throughput approach. Thus, here, we review the strategies and technologies available for the exploration lncRNAs and try to yield insights into the prospect of lncRNAs in clinical diagnosis and treatment in heart failure.
Collapse
Affiliation(s)
- Zhen Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Jingjing Wan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Liu
- School of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Weidong Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
115
|
Qin Y, Yan G, Qiao Y, Wang D, Luo E, Hou J, Tang C. Emerging role of long non-coding RNAs in pulmonary hypertension and their molecular mechanisms (Review). Exp Ther Med 2020; 20:164. [PMID: 33093902 PMCID: PMC7571311 DOI: 10.3892/etm.2020.9293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening cardiopulmonary condition caused by several pathogenic factors. All types of PH are characterized by the excessive proliferation of pulmonary artery endothelial cells and pulmonary artery smooth muscle cells, apoptosis resistance, pulmonary vascular remodeling, sustained elevated pulmonary arterial pressure, right heart failure and even death. Over the past decade, next generation sequencing, particularly RNA-sequencing, has identified some long non-coding RNAs (lncRNAs) that may act as regulators of cell differentiation, proliferation and apoptosis. Studies have shown that lncRNAs are closely associated with the development of several diseases, including cardiovascular diseases. In addition, a number of studies have reported that lncRNAs, including maternally expressed gene 3, metastasis-associated lung adenocarcinoma transcript 1, taurine upregulated 1 and cancer susceptibility candidate 2, serve important roles in the pathogenesis of PH. Despite the development of novel drug treatments, the mortality rate of PH remains high with no evident downward trend. Therefore, certain lncRNAs may be considered as therapeutic targets for the treatment of incurable PH. The present review summarizes the latest research on lncRNAs and PH, aiming to briefly describe PH-associated lncRNAs and their mechanisms of action.
Collapse
Affiliation(s)
- Yuhan Qin
- Department of Cardiology, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yong Qiao
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Erfei Luo
- Department of Cardiology, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Jiantong Hou
- Department of Cardiology, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
116
|
Abstract
NF-κB signaling pathway has important roles in the regulation of growth and development of nervous system. This pathway has also been shown to participate in the pathogenesis of schizophrenia. Meanwhile, activity of NF-κB signaling pathway is regulated by several factors including non-coding RNAs (lncRNAs). In the current study, we evaluated expression of nine NF-κB-related lncRNAs namely DILC, ANRIL, PACER, CHAST, ADINR, DICER1-AS1, HNF1A-AS1, H19 and NKILA as well as two mRNA coding genes namely ATG5 and CEBPA in the peripheral blood of patients with schizophrenia compared with matched healthy subjects. Expressions of these genes were assessed by real time PCR technique. Expression of PACER was lower in patients with schizophrenia compared with controls (Posterior beta = − 0.684, P value = 0.049). On the other hand, expressions of CHAST, CEBPA, H19, HNF1A-AS1 and DICER1-AS1 were higher in patients compared with controls (Posterior beta = 0.39, P value = 0.005; Posterior beta = 0.844, P value < 0.0001; Posterior beta = 0.467, P value < 0.0001; Posterior beta = 1.107, P value = 0.005; Posterior beta = 0.176, P value = 0.044, respectively). We also appraised the diagnostic power of transcript quantities of CHAST, CEBPA, DICER1-AS1, H19 and HNF1A-AS1 in distinguishing between patients with schizophrenia and controls through depicting ROC curves. Based on the area under curve (AUC) values, CEBPA had the best diagnostic power (AUC = 0.948, P < 0.0001), followed by H19 (AUC = 0.815, P < 0.0001). Taken together, our study demonstrated dysregulation of NF-κB-related lncRNAs and genes in the peripheral blood of patients with schizophrenia and their potential as peripheral markers for this psychiatric condition.
Collapse
|
117
|
Wang Y, Sun X. The functions of LncRNA in the heart. Diabetes Res Clin Pract 2020; 168:108249. [PMID: 32531328 DOI: 10.1016/j.diabres.2020.108249] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 06/04/2020] [Indexed: 12/26/2022]
Abstract
Cardiovascular disease is a major cause of death and disability worldwide. Recently, increasing evidence has demonstrated that various lncRNAs play critical roles in the pathogenesis of cardiovascular diseases, including myocardial ischemia and reperfusion (I/R) injury. LncRNAs are transcripts longer than 200 nucleotides. They are considered a class of dynamic noncoding RNAs known to be involved in physiological and pathological conditions with regulatory and structural roles in numerous biological processes, including genomic imprinting, epigenetic regulation, cell proliferation, development, aging and apoptosis. They are emerging as potential key regulators of a variety of cardiovascular diseases. However, the roles of lncRNAs in the heart function remain largely unknown. The purpose of this review was to summarize the functions of lncRNAs in the heart and discuss the challenges and possible strategies of lncRNA research for cardiovascular disease.
Collapse
Affiliation(s)
- Yao Wang
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xianglan Sun
- Department of Geriatrics, Department of Geriatric Endocrinology, ShanDong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
118
|
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: 3.5] [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
|
119
|
Cheng Z, Liu L, Li Q. lncRNA ZEB2-AS1 stimulates cardiac hypertrophy by downregulating PTEN. Exp Ther Med 2020; 20:92. [PMID: 32973941 DOI: 10.3892/etm.2020.9220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiac hypertrophy (CH) is closely related to a range of cardiovascular diseases, including heart failure and sudden cardiac death. The present study aimed to elucidate the role of long non-coding RNA (lncRNA) ZEB2 antisense RNA 1 (ZEB2-AS1) in regulating the hypertrophic process of cardiomyocytes and the potential underlying mechanism. An in vivo CH mouse model was established by performing transverse aortic constriction procedures. An in vitro CH model was established in primary cardiomyocytes isolated from mice by phenylephrine (PE) treatment. The relative protein levels of BNP, ANP and PTEN in cells with different groups (CH group and control group) were determined by western blotting. Relative expression levels of ZEB2-AS1, natriuretic peptide A (ANP) and brain natriuretic peptide (BNP) were determined in both in vivo and in vitro CH models. The regulatory effects of ZEB2-AS1/phosphatase and tensin homolog (PTEN) on cell surface area, and the relative expression levels of ANP and BNP were explored. ZEB2-AS1, ANP and BNP expression levels were increased in both in vivo and in vitro CH models compared with the sham and negative control groups, respectively. ZEB2-AS1 knockdown decreased cell surface area, and downregulated ANP and BNP expression levels in PE-treated primary cardiomyocytes. Similarly, PTEN overexpression reduced cell surface area, and downregulated ANP and BNP expression levels in PE-treated primary cardiomyocytes. Moreover, PTEN reversed the regulatory effects of ZEB2-AS1 on hypertrophic cardiomyocytes. Therefore, the present study suggested that lncRNA ZEB2-AS1 may influence the progression of CH by downregulating PTEN.
Collapse
Affiliation(s)
- Zhi Cheng
- Cardiovascular Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210003, P.R. China
| | - Lingyun Liu
- Cardiovascular Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210003, P.R. China
| | - Qingguo Li
- Cardiovascular Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210003, P.R. China
| |
Collapse
|
120
|
Fang Y, Xu Y, Wang R, Hu L, Guo D, Xue F, Guo W, Zhang D, Hu J, Li Y, Zhang W, Zhang M. Recent advances on the roles of LncRNAs in cardiovascular disease. J Cell Mol Med 2020; 24:12246-12257. [PMID: 32969576 PMCID: PMC7686979 DOI: 10.1111/jcmm.15880] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/12/2020] [Accepted: 08/24/2020] [Indexed: 12/22/2022] Open
Abstract
Cardiovascular diseases are a main cause of mortality whose prevalence continues to increase worldwide. Long non-coding RNAs (lncRNAs) regulate a variety of biological processes by modifying and regulating transcription of coding genes, directly binding to proteins and even coding proteins themselves. LncRNAs play key roles in the occurrence and development of myocardial infarction, heart failure, myocardial hypertrophy, arrhythmias and other pathological processes that significantly affect the prognosis and survival of patients with cardiovascular diseases. We here review the latest research on lncRNAs in cardiovascular diseases as a basis to formulate future research on prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yexian Fang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuerong Xu
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Runze Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lang Hu
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dong Guo
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Feng Xue
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wangang Guo
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dongwei Zhang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jianqiang Hu
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wei Zhang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
121
|
The roles of long noncoding RNAs in myocardial pathophysiology. Biosci Rep 2020; 39:220734. [PMID: 31694052 PMCID: PMC6851514 DOI: 10.1042/bsr20190966] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 10/03/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023] Open
Abstract
Long noncoding RNAs (lncRNAs), more than 200 nt in length, are functional molecules found in various species. These lncRNAs play a vital role in cell proliferation, differentiation, and degeneration and are also involved in pathophysiological processes of cancer and neurodegenerative, autoimmune, and cardiovascular diseases (CVDs). In recent years, emerging challenges for intervention studies on ischemic heart diseases have received much attention. LncRNAs have a key function in the alleviation of myocardial infarction (MI) injury and myocardial ischemia–reperfusion injury. During cardiac hypertrophy (CH) and fibrosis, cardiac cells undergo structural changes and become dysfunctional due to the effects of neurohormonal factors. LncRNAs may serve as important therapeutic targets that promote cardiac remodeling and then retard the development of heart failure (HF). In addition, studies on the roles and mechanisms of action of lncRNAs participating in cardiac pathophysiology via other factors have become the focus of research worldwide. Here, we review the current knowledge on various lncRNAs and their functions in cardiac biology, particularly concentrating on ischemic heart disease, CH, and cardiac fibrosis. We next discuss the predictive value of lncRNAs as diagnostic biomarkers of CVDs.
Collapse
|
122
|
Liu L, Zhang D, Li Y. LncRNAs in cardiac hypertrophy: From basic science to clinical application. J Cell Mol Med 2020; 24:11638-11645. [PMID: 32896990 PMCID: PMC7579708 DOI: 10.1111/jcmm.15819] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/29/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac hypertrophy is a typical pathological phenotype of cardiomyopathy and a result from pathological remodelling of cardiomyocytes in humans. At present, emerging evidence demonstrated the roles of long non‐coding RNAs (lncRNAs) in regulating the pathophysiological process of cardiac hypertrophy. Herein, we would like to review the recent researches on this issue and try to analysis the potential therapeutic targets on lncRNA sites. Studies have revealed both genetic mutations related hypertrophic cardiomyopathy and the compensative cardiac hypertrophy due to pressure overload, inflammation, endocrine issues and other external stimulations, share a common molecular mechanism of ventricular hypertrophy. The emerging evidence identified the abnormal expression of lncRNAs would leading to the impairment the function of sarcomere, intracellular calcium handling and mitochondrial metabolisms. Several researches proved the therapeutic role of lncRNAs in preventing or reversing cardiac hypertrophy. With the development of delivery system for small pieces of oligonucleotide, clinicians could design gene therapy approaches to terminate the process of cardiac hypertrophy to provide better prognosis.
Collapse
Affiliation(s)
- Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
123
|
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: 3.5] [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
|
124
|
Inhibition of the long non-coding RNA NEAT1 protects cardiomyocytes from hypoxia in vitro via decreased pri-miRNA processing. Cell Death Dis 2020; 11:677. [PMID: 32826883 PMCID: PMC7442835 DOI: 10.1038/s41419-020-02854-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
While restoration of coronary blood flow to the ischemic heart is the most effective strategy for reducing infarct size, reperfusion injury represents a significant limiting factor on clinical outcomes in myocardial infarction patients. Ischemic preconditioning (IPC) has been shown to inhibit reperfusion injury and represents an attractive model for studying cardioprotective signal transduction pathways. Long non-coding RNAs (lncRNAs) are a structurally and functionally heterogenous class of RNA transcripts with unknown roles in IPC-induced cardioprotection. Through microarray-based expression profiling of 31,423 lncRNAs in cardiac tissue from IPC mice, we identified the nuclear transcript Neat1 to be rapidly and robustly decreased in response to IPC. siRNA-mediated knock down of Neat1 reduced apoptosis and necrosis in murine cardiomyocytes (CM) and human iPS-derived CMs in response to prolonged hypoxia and hypoxia-reoxygenation, assessed with Annexin V/propidium iodide-staining, a Caspase 3/7 activity assay, LDH release, and western blot for cleaved Caspase 3. Mechanistically, Neat1 was shown to regulate processing of pro-apoptotic microRNA-22 (miR-22) in murine and human CM nuclei using a luciferase reporter assay. Hypoxia-induced downregulation of Neat1 was shown to result in accumulation of unprocessed pri-miRNA and decreased availability of biologically active miRNA, including miR-22. Addition of exogenous synthetic miR-22 reversed the protective effect of Neat1 knock down in human iPS-CM. In conclusion, we have identified the nuclear lncRNA Neat1 as part of a conserved oxygen-sensitive feedback mechanism by regulation of miRNA processing and a potential target in cardioprotection.
Collapse
|
125
|
Kuwabara Y, Tsuji S, Nishiga M, Izuhara M, Ito S, Nagao K, Horie T, Watanabe S, Koyama S, Kiryu H, Nakashima Y, Baba O, Nakao T, Nishino T, Sowa N, Miyasaka Y, Hatani T, Ide Y, Nakazeki F, Kimura M, Yoshida Y, Inada T, Kimura T, Ono K. Lionheart LincRNA alleviates cardiac systolic dysfunction under pressure overload. Commun Biol 2020; 3:434. [PMID: 32792557 PMCID: PMC7426859 DOI: 10.1038/s42003-020-01164-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 07/22/2020] [Indexed: 12/05/2022] Open
Abstract
Recent high-throughput approaches have revealed a vast number of transcripts with unknown functions. Many of these transcripts are long noncoding RNAs (lncRNAs), and intergenic region-derived lncRNAs are classified as long intergenic noncoding RNAs (lincRNAs). Although Myosin heavy chain 6 (Myh6) encoding primary contractile protein is down-regulated in stressed hearts, the underlying mechanisms are not fully clarified especially in terms of lincRNAs. Here, we screen upregulated lincRNAs in pressure overloaded hearts and identify a muscle-abundant lincRNA termed Lionheart. Compared with controls, deletion of the Lionheart in mice leads to decreased systolic function and a reduction in MYH6 protein levels following pressure overload. We reveal decreased MYH6 results from an interaction between Lionheart and Purine-rich element-binding protein A after pressure overload. Furthermore, human LIONHEART levels in left ventricular biopsy specimens positively correlate with cardiac systolic function. Our results demonstrate Lionheart plays a pivotal role in cardiac remodeling via regulation of MYH6. Kuwabara et al. identify a novel long intergenic noncoding RNA (lincRNA), termed Lionheart, upregulated in pressure overloaded hearts in mice. Deleting this gene results in decreased systolic function and reduction in MYH6 protein levels following pressure overload. They demonstrate that Lionheart interacts with PURA, preventing its binding to the promoter region of Myh6 locus, leading to reduced MYH6 protein expression.
Collapse
Affiliation(s)
- Yasuhide Kuwabara
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shuhei Tsuji
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masataka Nishiga
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masayasu Izuhara
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuya Nagao
- Department of Cardiovascular Center, Osaka Red Cross Hospital, Osaka, Japan
| | - Takahiro Horie
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shin Watanabe
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Koyama
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hisanori Kiryu
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Chiba, Japan
| | - Yasuhiro Nakashima
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Baba
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tetsushi Nakao
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Nishino
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naoya Sowa
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yui Miyasaka
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Hatani
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yuya Ide
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumiko Nakazeki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshinori Yoshida
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Tsukasa Inada
- Department of Cardiovascular Center, Osaka Red Cross Hospital, Osaka, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
126
|
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: 4.3] [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
|
127
|
Non-coding RNAs: emerging players in cardiomyocyte proliferation and cardiac regeneration. Basic Res Cardiol 2020; 115:52. [PMID: 32748089 PMCID: PMC7398957 DOI: 10.1007/s00395-020-0816-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
Soon after birth, the regenerative capacity of the mammalian heart is lost, cardiomyocytes withdraw from the cell cycle and demonstrate a minimal proliferation rate. Despite improved treatment and reperfusion strategies, the uncompensated cardiomyocyte loss during injury and disease results in cardiac remodeling and subsequent heart failure. The promising field of regenerative medicine aims to restore both the structure and function of damaged tissue through modulation of cellular processes and regulatory mechanisms involved in cardiac cell cycle arrest to boost cardiomyocyte proliferation. Non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) are functional RNA molecules with no protein-coding function that have been reported to engage in cardiac regeneration and repair. In this review, we summarize the current understanding of both the biological functions and molecular mechanisms of ncRNAs involved in cardiomyocyte proliferation. Furthermore, we discuss their impact on the structure and contractile function of the heart in health and disease and their application for therapeutic interventions.
Collapse
|
128
|
Das S, Shah R, Dimmeler S, Freedman JE, Holley C, Lee JM, Moore K, Musunuru K, Wang DZ, Xiao J, Yin KJ. Noncoding RNAs in Cardiovascular Disease: Current Knowledge, Tools and Technologies for Investigation, and Future Directions: A Scientific Statement From the American Heart Association. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2020; 13:e000062. [PMID: 32812806 DOI: 10.1161/hcg.0000000000000062] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The discovery that much of the non-protein-coding genome is transcribed and plays a diverse functional role in fundamental cellular processes has led to an explosion in the development of tools and technologies to investigate the role of these noncoding RNAs in cardiovascular health. Furthermore, identifying noncoding RNAs for targeted therapeutics to treat cardiovascular disease is an emerging area of research. The purpose of this statement is to review existing literature, offer guidance on tools and technologies currently available to study noncoding RNAs, and identify areas of unmet need. METHODS The writing group used systematic literature reviews (including MEDLINE, Web of Science through 2018), expert opinion/statements, analyses of databases and computational tools/algorithms, and review of current clinical trials to provide a broad consensus on the current state of the art in noncoding RNA in cardiovascular disease. RESULTS Significant progress has been made since the initial studies focusing on the role of miRNAs (microRNAs) in cardiovascular development and disease. Notably, recent progress on understanding the role of novel types of noncoding small RNAs such as snoRNAs (small nucleolar RNAs), tRNA (transfer RNA) fragments, and Y-RNAs in cellular processes has revealed a noncanonical function for many of these molecules. Similarly, the identification of long noncoding RNAs that appear to play an important role in cardiovascular disease processes, coupled with the development of tools to characterize their interacting partners, has led to significant mechanistic insight. Finally, recent work has characterized the unique role of extracellular RNAs in mediating intercellular communication and their potential role as biomarkers. CONCLUSIONS The rapid expansion of tools and pipelines for isolating, measuring, and annotating these entities suggests that caution in interpreting results is warranted until these methodologies are rigorously validated. Most investigators have focused on investigating the functional role of single RNA entities, but studies suggest complex interaction between different RNA molecules. The use of network approaches and advanced computational tools to understand the interaction of different noncoding RNA species to mediate a particular phenotype may be required to fully comprehend the function of noncoding RNAs in mediating disease phenotypes.
Collapse
MESH Headings
- American Heart Association
- Biomarkers/metabolism
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/pathology
- Humans
- MicroRNAs/chemistry
- MicroRNAs/genetics
- MicroRNAs/metabolism
- RNA, Long Noncoding/chemistry
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Small Nucleolar/chemistry
- RNA, Small Nucleolar/genetics
- RNA, Small Nucleolar/metabolism
- RNA, Transfer/chemistry
- RNA, Transfer/genetics
- RNA, Transfer/metabolism
- RNA, Untranslated/chemistry
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- United States
Collapse
|
129
|
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: 2.3] [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
|
130
|
Yang L, Lu Y, Ming J, Pan Y, Yu R, Wu Y, Wang T. SNHG16 accelerates the proliferation of primary cardiomyocytes by targeting miRNA-770-5p. Exp Ther Med 2020; 20:3221-3227. [PMID: 32855691 PMCID: PMC7444419 DOI: 10.3892/etm.2020.9083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to clarify the influence of long non-coding RNA small nuclear host gene 16 (lncRNA SNHG16) on cardiomyocyte proliferation following ischemia/reperfusion injury (IRI) and the potential mechanism. An IRI model in mice was established by performing ligation of the anterior descending coronary artery (LAD). Primary cardiomyocytes were isolated from newborn mice and subjected to H2O2 treatment to mimic in vitro IRI. Relative levels of SNHG16 and miRNA-770-5p in both in vivo and in vitro IRI models were examined. The regulatory effects of SNHG16 and miRNA-770-5p on the proliferative ability of H2O2-treated cardiomyocytes were assessed by Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) assay. The binding relationship between SNHG16 and miRNA-770-5p was verified through dual-luciferase reporter gene assay. It is found that SNHG16 was time-dependently downregulated in the IRI models. Overexpression of SNHG16 enhanced the proliferative ability of the cardiomyocytes. miRNA-770-5p was found to be a direct target of SNHG16. Moreover, SNHG16 was able to negatively regulate the miRNA-770-5p level. Overexpression of miRNA-770-5p partially reversed the role of SNHG16 on accelerating cardiomyocyte proliferation. Collectively, SNHG16 accelerates the proliferative ability of cardiomyocytes following IRI by negatively regulating miRNA-770-5p.
Collapse
Affiliation(s)
- Linshan Yang
- Department of Cardiac Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yan Lu
- Department of Otorhinolaryngology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Jie Ming
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yuzhu Pan
- Department of Cardiac Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Renbin Yu
- Department of Cardiac Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yuhui Wu
- Department of Cardiac Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Tao Wang
- Department of Cardiac Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
131
|
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: 7.0] [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
|
132
|
Omura J, Habbout K, Shimauchi T, Wu WH, Breuils-Bonnet S, Tremblay E, Martineau S, Nadeau V, Gagnon K, Mazoyer F, Perron J, Potus F, Lin JH, Zafar H, Kiely DG, Lawrie A, Archer SL, Paulin R, Provencher S, Boucherat O, Bonnet S. Identification of Long Noncoding RNA H19 as a New Biomarker and Therapeutic Target in Right Ventricular Failure in Pulmonary Arterial Hypertension. Circulation 2020; 142:1464-1484. [PMID: 32698630 DOI: 10.1161/circulationaha.120.047626] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Right ventricular (RV) function is the major determinant for both functional capacity and survival in patients with pulmonary arterial hypertension (PAH). Despite the recognized clinical importance of preserving RV function, the subcellular mechanisms that govern the transition from a compensated to a decompensated state remain poorly understood and as a consequence there are no clinically established treatments for RV failure and a paucity of clinically useful biomarkers. Accumulating evidence indicates that long noncoding RNAs are powerful regulators of cardiac development and disease. Nonetheless, their implication in adverse RV remodeling in PAH is unknown. METHODS Expression of the long noncoding RNA H19 was assessed by quantitative PCR in plasma and RV from patients categorized as control RV, compensated RV or decompensated RV based on clinical history and cardiac index. The impact of H19 suppression using GapmeR was explored in 2 rat models mimicking RV failure, namely the monocrotaline and pulmonary artery banding. Echocardiographic, hemodynamic, histological, and biochemical analyses were conducted. In vitro gain- and loss-of-function experiments were performed in rat cardiomyocytes. RESULTS We demonstrated that H19 is upregulated in decompensated RV from PAH patients and correlates with RV hypertrophy and fibrosis. Similar findings were observed in monocrotaline and pulmonary artery banding rats. We found that silencing H19 limits pathological RV hypertrophy, fibrosis and capillary rarefaction, thus preserving RV function in monocrotaline and pulmonary artery banding rats without affecting pulmonary vascular remodeling. This cardioprotective effect was accompanied by E2F transcription factor 1-mediated upregulation of enhancer of zeste homolog 2. In vitro, knockdown of H19 suppressed cardiomyocyte hypertrophy induced by phenylephrine, while its overexpression has the opposite effect. Finally, we demonstrated that circulating H19 levels in plasma discriminate PAH patients from controls, correlate with RV function and predict long-term survival in 2 independent idiopathic PAH cohorts. Moreover, H19 levels delineate subgroups of patients with differentiated prognosis when combined with the NT-proBNP (N-terminal pro-B-type natriuretic peptide) levels or the risk score proposed by both REVEAL (Registry to Evaluate Early and Long-Term PAH Disease Management) and the 2015 European Pulmonary Hypertension Guidelines. CONCLUSIONS Our findings identify H19 as a new therapeutic target to impede the development of maladaptive RV remodeling and a promising biomarker of PAH severity and prognosis.
Collapse
Affiliation(s)
- Junichi Omura
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.)
| | - Karima Habbout
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.)
| | - Tsukasa Shimauchi
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.)
| | - Wen-Hui Wu
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.).,Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, China (W-H.W.)
| | - Sandra Breuils-Bonnet
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.)
| | - Eve Tremblay
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.)
| | - Sandra Martineau
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.)
| | - Valérie Nadeau
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.)
| | - Kassandra Gagnon
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.)
| | - Florence Mazoyer
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.)
| | - Jean Perron
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.)
| | - Francois Potus
- Department of Medicine, Queen's University, Kingston, ON, Canada (F.P., S.L.A.)
| | - Jian-Hui Lin
- Department of Infection, Immunity and Cardiovascular Science, University of Sheffield, UK (J-H.L., H.Z., D.G.K., A.L.)
| | - Hamza Zafar
- Department of Infection, Immunity and Cardiovascular Science, University of Sheffield, UK (J-H.L., H.Z., D.G.K., A.L.).,Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, UK (H.Z., D.G.K.)
| | - David G Kiely
- Department of Infection, Immunity and Cardiovascular Science, University of Sheffield, UK (J-H.L., H.Z., D.G.K., A.L.).,Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, UK (H.Z., D.G.K.)
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Science, University of Sheffield, UK (J-H.L., H.Z., D.G.K., A.L.)
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, ON, Canada (F.P., S.L.A.)
| | - Roxane Paulin
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.).,Department of Medicine, Université Laval, Québec, QC, Canada (R.P., S.P., O.B., S.B.)
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.).,Department of Medicine, Université Laval, Québec, QC, Canada (R.P., S.P., O.B., S.B.)
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.).,Department of Medicine, Université Laval, Québec, QC, Canada (R.P., S.P., O.B., S.B.)
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Center de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QC, Canada (J.O., K.H., T.S., W-H.W., S.B-B., E.T., S.M., V.N., K.G., F.M., J.P., R.P., S.P., O.B., S.B.).,Department of Medicine, Université Laval, Québec, QC, Canada (R.P., S.P., O.B., S.B.)
| |
Collapse
|
133
|
Jiang C, Ding N, Li J, Jin X, Li L, Pan T, Huo C, Li Y, Xu J, Li X. Landscape of the long non-coding RNA transcriptome in human heart. Brief Bioinform 2020; 20:1812-1825. [PMID: 29939204 DOI: 10.1093/bib/bby052] [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] [Received: 03/12/2018] [Revised: 05/02/2018] [Indexed: 12/15/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been revealed to play essential roles in the human cardiovascular system. However, information about their mechanisms is limited, and a comprehensive view of cardiac lncRNAs is lacking from a multiple tissues perspective to date. Here, the landscape of the lncRNA transcriptome in human heart was summarized. We summarized all lncRNA transcripts from publicly available human transcriptome resources (156 heart samples and 210 samples from 29 other tissues) and systematically analysed all annotated and novel lncRNAs expressed in heart. A total of 7485 lncRNAs whose expression was elevated in heart (HE lncRNAs) and 453 lncRNAs expressed in all 30 analysed tissues (EIA lncRNAs) were extracted. Using various bioinformatics resources, methods and tools, the features of these lncRNAs were discussed from various perspectives, including genomic structure, conservation, dynamic variation during heart development, cis-regulation, differential expression in cardiovascular diseases and cancers as well as regulation at transcriptional and post-transcriptional levels. Afterwards, all the features discussed above were integrated into a user-friendly resource named CARDIO-LNCRNAS (http://bio-bigdata.hrbmu.edu.cn/CARDIO-LNCRNAS/ or http://www.bio-bigdata.net/CARDIO-LNCRNAS/). This study represents the first global view of lncRNAs in the human cardiovascular system based on multiple tissues and sheds light on the role of lncRNAs in developments and heart disorders.
Collapse
Affiliation(s)
- Chunjie Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, China
| | - Na Ding
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, China
| | - Junyi Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, China
| | - Xiyun Jin
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, China
| | - Lili Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, China
| | - Tao Pan
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, China
| | - Caiqin Huo
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, China
| | - Yongsheng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, China
| |
Collapse
|
134
|
Afrough H, Ghafouri-Fard S, Yousefi H, Pakzad P, Kholghi Oskooei V, Taheri M. DICER-AS1 lncRNA: A putative culprit in the pathogenesis of gastric cancer. Exp Mol Pathol 2020; 116:104490. [PMID: 32663487 DOI: 10.1016/j.yexmp.2020.104490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/15/2020] [Accepted: 07/07/2020] [Indexed: 12/24/2022]
Abstract
The nuclear factor-κB (NF-κB) has a pivotal role in the pathogenesis of several cancers including gastric cancer. We have recently reported dysregulation of a number of NF-κB-associated lncRNAs in a variety of human disorders including breast cancer and coronary artery disease. In the current study, we evaluated expression of five NF-κB-associated lncRNAs (CHAST, ADINR, DICER1-AS1, HNF1A-AS1 and NKILA) and two NF-κB-associated-mRNA coding genes (CEBPA and ATG5) in gastric cancer tissues and their paired non-cancerous tissues using real time PCR method. Expression of DICER-AS1 was significantly down-regulated in gastric cancer tissues compared with the corresponding non-cancerous tissues (Expression ratio = 0.23, P value = .01). Expressions of other genes were not significantly different between these two sets of samples. Relative expression of DICER1-AS1 in cancer tissues versus non-cancerous tissues tended to associated with histological grade (P = .05). Tumoral expression levels of NKILA, ADINR, CEBPA and HNF1A-AS1 were significantly higher in patients with positive family history of cancer compared with those without such history (P values = .03, 0.02, 0.02 1nd 0.03, respectively). Besides, expression levels of NKILA, ADINR, DICER1-AS1, CEBPA, CHAST, HNF1A-AS1 and ATG5 were lower in H. pylori-infected tissues (P values = .01, 0.02, 0.03, 0.01, 0.004, 0.004 and 0.04, respectively). The lowest tumoral expression of DICER1-AS1 was detected in stage II cancers, while the highest expression of this lncRNA was reported in a single stage I tumor tissue. Similar pattern of expression was detected for ATG5. Significant pairwise correlations were demonstrated between expression levels of NF-ƙB-associated genes in both gastric cancer tissues and non-cancerous tissues. Expression levels of DICER1-AS1 had sensitivity and specificity values of 63.3% and 63.3% in differentiating between tumoral and non-tumoral tissues (Estimate criterion>6.96, J = 0.27, P value = .01, AUC = 0.67). Although previous studies have reported involvement of NF-κB pathway in the pathogenesis of gastric cancer, among the reported lncRNAs associated with this pathway, we could only detect differential expression of DICER1-AS1 between tumoral and non-tumoral tissues. Thus, the mechanism underlying dysregulation of this pathway might be different among various cancers.
Collapse
Affiliation(s)
- Hanieh Afrough
- College of Biology Sciences, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA
| | - Parviz Pakzad
- Department of Microbiology, School of Biology Sciences, North Branch, Islamic Azad University, Tehran, Iran
| | - Vahid Kholghi Oskooei
- Department of Laboratory Sciences, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
135
|
Hobuß L, Foinquinos A, Jung M, Kenneweg F, Xiao K, Wang Y, Zimmer K, Remke J, Just A, Nowak J, Schmidt A, Pich A, Mazlan S, Reamon-Buettner SM, Ramos GC, Frantz S, Viereck J, Loyer X, Boulanger C, Wollert KC, Fiedler J, Thum T. Pleiotropic cardiac functions controlled by ischemia-induced lncRNA H19. J Mol Cell Cardiol 2020; 146:43-59. [PMID: 32649928 DOI: 10.1016/j.yjmcc.2020.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022]
Abstract
Myocardial ischemia induces a multifaceted remodeling process in the heart. Novel therapeutic entry points to counteract maladaptive signalling include the modulation of non-coding RNA molecules such as long non-coding RNA (lncRNA). We here questioned if the lncRNA candidate H19 exhibits regulatory potential in the setting of myocardial infarction. Initial profiling of H19 expression revealed a dynamic expression profile of H19 with upregulation in the acute phase after murine cardiac ischemia. In vitro, we found that oxygen deficiency leads to H19 upregulation in several cardiac cell types. Repression of endogenous H19 caused multiple phenotypes in cultivated murine cardiomyocytes including enhanced cardiomyocyte apoptosis, at least partly through attenuated vitamin D signalling. Unbiased proteome analysis revealed further involvement of H19 in mRNA splicing and translation as well as inflammatory signalling pathways. To study H19 function more precisely, we investigated the phenotype of systemic H19 loss in a genetic mouse model of H19 deletion (H19 KO). Infarcted heart tissue of H19 KO mice showed a massive increase of pro-inflammatory cytokines after ischemia-reperfusion injury (I/R) without significant effects on scar formation or cardiac function but exaggerated cardiac hypertrophy indicating pathological cardiac remodeling. H19-dependent changes in cardiomyocyte-derived extracellular vesicle release and alterations in NF-κB signalling were evident. Cardiac cell fractionation experiments revealed that enhanced H19 expression in the proliferative phase after MI derived mainly from cardiac fibroblasts. Here further research is needed to elucidate its role in fibroblast activation and function. In conclusion, the lncRNA H19 is dynamically regulated after MI and involved in multiple pathways of different cardiac cell types including cardiomyocyte apoptosis and cardiac inflammation.
Collapse
Affiliation(s)
- Lisa Hobuß
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Ariana Foinquinos
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Ke Xiao
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Yong Wang
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Karina Zimmer
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Janet Remke
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Annette Just
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Juliette Nowak
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Arne Schmidt
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Andreas Pich
- Core Unit Mass Spectrometry and Proteomics, Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | | | | | - Gustavo Campos Ramos
- Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany; Comprehensive Heart Failure Center, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany; Comprehensive Heart Failure Center, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Janika Viereck
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Xavier Loyer
- Université de Paris, PARCC, INSERM, F-75015 Paris, France
| | | | - Kai C Wollert
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany,; REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
136
|
Talebian S, Daghagh H, Yousefi B, Ȍzkul Y, Ilkhani K, Seif F, Alivand MR. The role of epigenetics and non-coding RNAs in autophagy: A new perspective for thorough understanding. Mech Ageing Dev 2020; 190:111309. [PMID: 32634442 DOI: 10.1016/j.mad.2020.111309] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/22/2020] [Accepted: 06/28/2020] [Indexed: 12/18/2022]
Abstract
Autophagy is a major self-degradative intracellular process required for the maintenance of homeostasis and promotion of survival in response to starvation. It plays critical roles in a large variety of physiological and pathological processes. On the other hand, aberrant regulation of autophagy can lead to various cancers and neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Crohn's disease. Emerging evidence strongly supports that epigenetic signatures, related non-coding RNA profiles, and their cross-talking are significantly associated with the control of autophagic responses. Therefore, it may be helpful and promising to manage autophagic processes by finding valuable markers and therapeutic approaches. Although there is a great deal of information on the components of autophagy in the cytoplasm, the molecular basis of the epigenetic regulation of autophagy has not been completely elucidated. In this review, we highlight recent research on epigenetic changes through the expression of autophagy-related genes (ATGs), which regulate autophagy, DNA methylation, histone modifications as well as non-coding RNAs, including long non-coding RNAs (lncRNAs), microRNAs (miRNAs) and their relationship with human diseases, that play key roles in causing autophagy-related diseases.
Collapse
Affiliation(s)
- Shahrzad Talebian
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Daghagh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yusuf Ȍzkul
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Khandan Ilkhani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Seif
- Department of Immunology & Allergy, Academic Center for Education, Culture, and Research, Tehran, Iran
| | - Mohammad Reza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
137
|
An Overview of Non-coding RNAs and Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:3-45. [PMID: 32285403 DOI: 10.1007/978-981-15-1671-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease management and timely diagnosis remain a major dilemma. Delineating molecular mechanisms of cardiovascular diseases is opening horizon in the field of molecular medicines and in the development of early diagnostic markers. Non-coding RNAs are the highly functional and vibrant nucleic acids and are known to be involved in the regulation of endothelial cells, vascular and smooth muscles cells, cardiac metabolism, ischemia, inflammation and many processes in cardiovascular system. This chapter is comprehensively focusing on the overview of the non-coding RNAs including their discovery, generation, classification and functional regulation. In addition, overview regarding different non-coding RNAs as long non-coding, siRNAs and miRNAs involvement in the cardiovascular diseases is also addressed. Detailed functional analysis of this vast group of highly regulatory molecules will be promising for shaping future drug discoveries.
Collapse
|
138
|
Safa A, Arsang-Jang S, Taheri M, Omrani MD, Ghafouri-Fard S. Dysregulation of NF-κB-Associated lncRNAs in Multiple Sclerosis Patients. J Mol Neurosci 2020; 71:80-88. [DOI: 10.1007/s12031-020-01628-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/08/2020] [Indexed: 12/22/2022]
|
139
|
Lai L, Xu Y, Kang L, Yang J, Zhu G. LncRNA KCNQ1OT1 contributes to cardiomyocyte apoptosis by targeting FUS in heart failure. Exp Mol Pathol 2020; 115:104480. [PMID: 32497620 DOI: 10.1016/j.yexmp.2020.104480] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/09/2020] [Accepted: 05/30/2020] [Indexed: 11/29/2022]
Abstract
Long noncoding RNAs (lncRNAs) have recently been recognized as the important regulators in cardiac diseases. This study was aimed to investigate the role and molecular mechanism of lncRNA KCNQ1OT1 in regulating cardiomyocyte apoptosis in heart failure (HF). The mouse model of HF was induced by doxorubicin (ADR). Cell apoptosis was detected by Hoechst and TUNEL staining. Molecule expressions were determined by qRT-PCR and western blot. The interaction between KCNQ1OT1 and Fused in sarcoma (FUS) was assessed by RNA immunoprecipitation (RIP) and RNA pull-down assays. KCNQ1OT1 was up-regulated in the myocardial tissues of HF mice and the ADR-stimulated mouse myocardial cell line (HL-1). KCNQ1OT1 overexpression promoted apoptosis of ADR-stimulated HL-1 cells, while KCNQ1OT1 knockdown caused the opposite effect. The RIP and RNA pull-down results showed that KCNQ1OT1 - bound to FUS and negatively regulated its protein level. Knockdown of FUS inhibited apoptosis of ADR-stimulated HL-1 cells and reversed the effect of KCNQ1OT1 overexpression on cardiomyocyte apoptosis. In vivo experiment showed that KCNQ1OT1 ovexpression improved myocardial histopathological changes, reduced myocardial fibrosis areas, down-regulated FUS expression, and inhibited cell apoptosis of HF mice. In conclusion, KCNQ1OT1 facilitates cardiomyocyte apoptosis by - targeting FUS in ADR-induced HF.
Collapse
Affiliation(s)
- Lei Lai
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China.
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Lan Kang
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Jianmin Yang
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Gangjie Zhu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| |
Collapse
|
140
|
Prestes PR, Maier MC, Woods BA, Charchar FJ. A Guide to the Short, Long and Circular RNAs in Hypertension and Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21103666. [PMID: 32455975 PMCID: PMC7279167 DOI: 10.3390/ijms21103666] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/05/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in adults in developed countries. CVD encompasses many diseased states, including hypertension, coronary artery disease and atherosclerosis. Studies in animal models and human studies have elucidated the contribution of many genetic factors, including non-coding RNAs. Non-coding RNAs are RNAs not translated into protein, involved in gene expression regulation post-transcriptionally and implicated in CVD. Of these, circular RNAs (circRNAs) and microRNAs are relevant. CircRNAs are created by the back-splicing of pre-messenger RNA and have been underexplored as contributors to CVD. These circRNAs may also act as biomarkers of human disease, as they can be extracted from whole blood, plasma, saliva and seminal fluid. CircRNAs have recently been implicated in various disease processes, including hypertension and other cardiovascular disease. This review article will explore the promising and emerging roles of circRNAs as potential biomarkers and therapeutic targets in CVD, in particular hypertension.
Collapse
|
141
|
Abstract
While clinical gene therapy celebrates its first successes, with several products already approved for clinical use and several hundreds in the final stages of the clinical approval pipeline, there is not a single gene therapy approach that has worked for the heart. Here, we review the past experience gained in the several cardiac gene therapy clinical trials that had the goal of inducing therapeutic angiogenesis in the ischemic heart and in the attempts at modulating cardiac function in heart failure. Critical assessment of the results so far achieved indicates that the efficiency of cardiac gene delivery remains a major hurdle preventing success but also that improvements need to be sought in establishing more reliable large animal models, choosing more effective therapeutic genes, better designing clinical trials, and more deeply understanding cardiac biology. We also emphasize a few areas of cardiac gene therapy development that hold great promise for the future. In particular, the transition from gene addition studies using protein-coding cDNAs to the modulation of gene expression using small RNA therapeutics and the improvement of precise gene editing now pave the way to applications such as cardiac regeneration after myocardial infarction and gene correction for inherited cardiomyopathies that were unapproachable until a decade ago.
Collapse
Affiliation(s)
- Antonio Cannatà
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.)
| | - Hashim Ali
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (H.A., M.G.)
| | - Gianfranco Sinagra
- Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.)
| | - Mauro Giacca
- From the King's College London, British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, United Kingdom (A.C., H.A., M.G.).,Department of Medical, Surgical and Health Sciences, University of Trieste, Italy (A.C., G.S., M.G.).,Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (H.A., M.G.)
| |
Collapse
|
142
|
Trembinski DJ, Bink DI, Theodorou K, Sommer J, Fischer A, van Bergen A, Kuo CC, Costa IG, Schürmann C, Leisegang MS, Brandes RP, Alekseeva T, Brill B, Wietelmann A, Johnson CN, Spring-Connell A, Kaulich M, Werfel S, Engelhardt S, Hirt MN, Yorgan K, Eschenhagen T, Kirchhof L, Hofmann P, Jaé N, Wittig I, Hamdani N, Bischof C, Krishnan J, Houtkooper RH, Dimmeler S, Boon RA. Aging-regulated anti-apoptotic long non-coding RNA Sarrah augments recovery from acute myocardial infarction. Nat Commun 2020; 11:2039. [PMID: 32341350 PMCID: PMC7184724 DOI: 10.1038/s41467-020-15995-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 04/07/2020] [Indexed: 12/18/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) contribute to cardiac (patho)physiology. Aging is the major risk factor for cardiovascular disease with cardiomyocyte apoptosis as one underlying cause. Here, we report the identification of the aging-regulated lncRNA Sarrah (ENSMUST00000140003) that is anti-apoptotic in cardiomyocytes. Importantly, loss of SARRAH (OXCT1-AS1) in human engineered heart tissue results in impaired contractile force development. SARRAH directly binds to the promoters of genes downregulated after SARRAH silencing via RNA-DNA triple helix formation and cardiomyocytes lacking the triple helix forming domain of Sarrah show an increase in apoptosis. One of the direct SARRAH targets is NRF2, and restoration of NRF2 levels after SARRAH silencing partially rescues the reduction in cell viability. Overexpression of Sarrah in mice shows better recovery of cardiac contractile function after AMI compared to control mice. In summary, we identified the anti-apoptotic evolutionary conserved lncRNA Sarrah, which is downregulated by aging, as a regulator of cardiomyocyte survival.
Collapse
Affiliation(s)
- D Julia Trembinski
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Diewertje I Bink
- Department of Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Kosta Theodorou
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Janina Sommer
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Ariane Fischer
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Anke van Bergen
- Department of Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Chao-Chung Kuo
- Institute for Computational Genomics, Joint Research Center for Computational Biomedicine, RWTH Aachen University, Aachen, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, Joint Research Center for Computational Biomedicine, RWTH Aachen University, Aachen, Germany
| | - Christoph Schürmann
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Matthias S Leisegang
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Ralf P Brandes
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Tijna Alekseeva
- Georg Speyer Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Boris Brill
- Georg Speyer Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Astrid Wietelmann
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Christopher N Johnson
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, USA
| | | | - Manuel Kaulich
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Stanislas Werfel
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Institute of Pharmacology and Toxicology, Technical University Munich, Munich, Germany
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Munich, Germany
| | - Stefan Engelhardt
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Institute of Pharmacology and Toxicology, Technical University Munich, Munich, Germany
| | - Marc N Hirt
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kaja Yorgan
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Eschenhagen
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Luisa Kirchhof
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Patrick Hofmann
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Nicolas Jaé
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Ilka Wittig
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Functional Proteomics, Medical School, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Nazha Hamdani
- Department of Physiology, VU University Medical Center, Amsterdam, the Netherlands
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Corinne Bischof
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Jaya Krishnan
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, the Netherlands
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Reinier A Boon
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research (DZHK), Berlin, Germany.
- Department of Physiology, VU University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
143
|
Abstract
Cardiac hypertrophy is a significant risk factor for cardiovascular disease, including heart failure, arrhythmia, and sudden death. Cardiac hypertrophy involves both embryonic gene expression and transcriptional reprogramming, which are tightly regulated by epigenetic mechanisms. An increasing number of studies have demonstrated that epigenetics plays an influential role in the occurrence and development of cardiac hypertrophy. Here, we summarize the latest research progress on epigenetics in cardiac hypertrophy involving DNA methylation, histone modification, and non-coding RNA, to help understand the mechanism of epigenetics in cardiac hypertrophy. The expression of both embryonic and functional genes can be precisely regulated by epigenetic mechanisms during cardiac hypertrophy, providing a substantial number of therapeutic targets. Thus, epigenetic treatment is expected to become a novel therapeutic strategy for cardiac hypertrophy. According to the research performed to date, epigenetic mechanisms associated with cardiac hypertrophy remain far from completely understood. Therefore, epigenetic mechanisms require further exploration to improve the prevention, diagnosis, and treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Kaijun Sun
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
144
|
Hu F, Yang J, Chen X, Shen Y, Chen K, Fu X, Guo S, Jiang Z. LncRNA 1700020I14Rik/miR-297a/CGRP axis suppresses myocardial cell apoptosis in myocardial ischemia-reperfusion injury. Mol Immunol 2020; 122:54-61. [PMID: 32298875 DOI: 10.1016/j.molimm.2020.03.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/09/2020] [Accepted: 03/20/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are closely related to various human diseases, but their role in myocardial injury has not been fully elucidated. In the current study, we found that the expression of lncRNA 1700020I14Rik was significantly down-regulated in myocardial injury tissues and the underlying mechanism by which lncRNA 1700020I14Rik regulated myocardial cell injury was investigated. METHODS The model of myocardial ischemia-reperfusion (I/R) injury and myocardial cells hypoxia/reoxygenation (H/R) injury were established and the expression of 1700020I14Rik, miR-297a or CGRP was analyzed by qRT-PCR or Western blot. Moreover, myocardial cell apoptosis was assessed by TUNEL staining and the concentration of LDH in the mouse plasma sample or myocardial cell culture supernatant was measured by the LDH cytotoxicity test kit. Furthermore, the differences of myocardial cell survival rate after H/R treatment were assessed by MTT assay and the observation of CGRP expression was performed in HL-1 cells overexpressed or silenced with 1700020I14Rik or miR-297a. In addition, the regulating function of miR-297a on 1700020I14Rik and CGRP expression was analyzed by a dual luciferase reporter assay. RESULTS The expressions of 1700020I14Rik and CGRP were abnormally down-regulated in a model of myocardial I/R injury and myocardial cells H/R injury, while miR-297a was up-regulated. By TUNEL staining, the apoptotic rate of myocardial cells in the model of myocardial I/R injury was significantly increased. Furthermore, the concentrations of LDH in the mouse plasma sample or myocardial cell culture supernatant were significantly increased after myocardial cell injury. By MTT assay, the survival rate of cells was decreased after myocardial cells were treated with H/R. In addition, overexpression of 1700020I14Rik or knockdown of miR-297a could up-regulate CGRP protein level, while interference with 1700020I14Rik or overexpression of miR-297a produced the opposite result. Further study confirmed that lncRNA 1700020I14Rik/miR-297a/CGRP axis suppressed myocardial cell apoptosis in myocardial I/R injury. CONCLUSION Our results indicated that 1700020I14Rik was abnormally down-regulated in myocardial injury tissues. In-depth studies manifested that 1700020I14Rik/miR-297a/CGRP axis suppressed myocardial cell apoptosis in myocardial I/R injury.
Collapse
Affiliation(s)
- Fudong Hu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinhua Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xi Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yangyang Shen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kui Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Fu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shengcun Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Zhengming Jiang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
145
|
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: 4.0] [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
|
146
|
Abstract
RNA modulation has become a promising therapeutic approach for the treatment of several types of disease. The emerging field of noncoding RNA-based therapies has now come to the attention of cardiovascular research, in which it could provide valuable advancements in comparison to current pharmacotherapy such as small molecule drugs or antibodies. In this review, we focus on noncoding RNA-based studies conducted mainly in large-animal models, including pigs, rabbits, dogs, and nonhuman primates. The obstacles and promises of targeting long noncoding RNAs and circRNAs as therapeutic modalities in humans are specifically discussed. We also describe novel ex vivo methods based on human cells and tissues, such as engineered heart tissues and living myocardial slices that could help bridging the gap between in vivo models and clinical applications in the future. Finally, we summarize antisense oligonucleotide drugs that have already been approved by the Food and Drug Administration for targeting mRNAs and discuss the progress of noncoding RNA-based drugs in clinical trials. Additional factors, such as drug chemistry, drug formulations, different routes of administration, and the advantages of RNA-based drugs, are also included in the present review. Recently, first therapeutic miRNA-based inhibitory strategies have been tested in heart failure patients as well as healthy volunteers to study effects on wound healing (NCT04045405; NCT03603431). In summary, a combination of novel therapeutic RNA targets, large-animal models, ex vivo studies with human cells/tissues, and new delivery techniques will likely lead to significant progress in the development of noncoding RNA-based next-generation therapeutics for cardiovascular disease.
Collapse
Affiliation(s)
- Cheng-Kai Huang
- From the Institute of Molecular and Translational Therapeutic Strategies (C.-K.H., S.K.-K., T.T.), Hannover Medical School, Germany
| | - Sabine Kafert-Kasting
- From the Institute of Molecular and Translational Therapeutic Strategies (C.-K.H., S.K.-K., T.T.), Hannover Medical School, Germany
| | - Thomas Thum
- From the Institute of Molecular and Translational Therapeutic Strategies (C.-K.H., S.K.-K., T.T.), Hannover Medical School, Germany
- REBIRTH Center of Translational Regenerative Medicine (T.T.), Hannover Medical School, Germany
| |
Collapse
|
147
|
Zahid KR, Raza U, Chen J, Raj UJ, Gou D. Pathobiology of pulmonary artery hypertension: role of long non-coding RNAs. Cardiovasc Res 2020; 116:1937-1947. [PMID: 32109276 DOI: 10.1093/cvr/cvaa050] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/30/2019] [Accepted: 02/25/2020] [Indexed: 12/30/2022] Open
Abstract
Abstract
Pulmonary arterial hypertension (PAH) is a disease with complex pathobiology, significant morbidity and mortality, and remains without a cure. It is characterized by vascular remodelling associated with uncontrolled proliferation of pulmonary artery smooth muscle cells, endothelial cell proliferation and dysfunction, and endothelial-to-mesenchymal transition, leading to narrowing of the vascular lumen, increased vascular resistance and pulmonary arterial pressure, which inevitably results in right heart failure and death. There are multiple molecules and signalling pathways that are involved in the vascular remodelling, including non-coding RNAs, i.e. microRNAs and long non-coding RNAs (lncRNAs). It is only in recent years that the role of lncRNAs in the pathobiology of pulmonary vascular remodelling and right ventricular dysfunction is being vigorously investigated. In this review, we have summarized the current state of knowledge about the role of lncRNAs as key drivers and gatekeepers in regulating major cellular and molecular trafficking involved in the pathogenesis of PAH. In addition, we have discussed the limitations and challenges in translating lncRNA research in vivo and in therapeutic applications of lncRNAs in PAH.
Collapse
Affiliation(s)
- Kashif Rafiq Zahid
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
- Key Laboratory of Optoelectronic Devices, Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Umar Raza
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Khadim Abid Majeed Road, Rawalpindi, Pakistan
| | - Jidong Chen
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
| | - Usha J Raj
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
| |
Collapse
|
148
|
Inhibition of cardiomyocyte differentiation of human induced pluripotent stem cells by Ribavirin: Implication for its cardiac developmental toxicity. Toxicology 2020; 435:152422. [PMID: 32112805 DOI: 10.1016/j.tox.2020.152422] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/16/2020] [Accepted: 02/25/2020] [Indexed: 12/18/2022]
Abstract
Ribavirin has been proven to be an antiviral treatment, whereas there are still risks of hemolysis and congenital malformation. Abnormal cardiac development contributes to the occurrence and development of many heart diseases. However, there is so far no evidence that ribavirin induces human cardiac developmental toxicity. Herein, we employed the cardiac differentiation model of human induced pluripotent stem cells (hiPSCs) to determine the impact of ribavirin on heart development. Our data showed that ribavirin at clinically high concentrations (5 and 10 μM) significantly inhibited the proliferation and differentiation of hiPSCs from mesoderm to cardiac progenitor cells and cardiac progenitor cells to cardiomyocytes, but not from pluripotent status to mesoderm. Meanwhile, DCFH-DA staining revealed that ribavirin could increase ROS content in the mid-phase of differentiation. In addition, ribavirin treatment (1, 5 and 10 μM) remarkably caused DNA damage which was shown by the increase of γH2AX-positive cells and upregulation of the p53 during the differentiation of hiPSCs from mesoderm to cardiac progenitor cells. Moreover, exposuring to ribavirin (5 and 10 μM) markedly upregulated the expression of lncRNAs Gas5 in both mid-phase and late phase of differentiation and HBL1 in the mid-phase. In conclusion, our results suggest that ribavirin is detrimental in cardiac differentiation of hiPSCs, which may be associated with DNA damage, upregulated p53 and increased Gas5. It may provide the evidence for the rational clinical application of ribavirin.
Collapse
|
149
|
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: 3.3] [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
|
150
|
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
|