1
|
Wang A, Wang C, Xuan B, Sun Y, Li B, Zhao Q, Yu R, Wang X, Zhu M, Wei J. The role of splicing events in the inflammatory response of atherosclerosis: molecular mechanisms and modulation. Front Immunol 2024; 15:1507420. [PMID: 39742258 PMCID: PMC11685076 DOI: 10.3389/fimmu.2024.1507420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by persistent inflammatory responses throughout all stages of its progression. Modulating these inflammatory responses is a promising avenue for the development of cardiovascular disease therapies. Splicing events modulate gene expression and diversify protein functionality, exerting pivotal roles in the inflammatory mechanisms underlying atherosclerosis. These insights may provide novel opportunities for developing anti-inflammatory therapies for this disease. This article systematically discusses the diverse splice variants and how splicing events impact the inflammatory response in atherosclerosis via endothelial cells, macrophages, and vascular smooth muscle cells, highlighting their underlying molecular mechanisms and implications. Furthermore, this study summarizes clinical evidence supporting splicing-related molecules as diagnostic biomarkers and therapeutic targets in atherosclerosis. Lastly, we outline the current challenges and future research directions concerning splicing events and inflammatory responses in atherosclerosis. This offers a novel perspective and evidence for formulating new therapeutic strategies aimed at lowering the risk of atherosclerosis.
Collapse
Affiliation(s)
- Aolong Wang
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Chengzhi Wang
- Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Bihan Xuan
- College of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yanqin Sun
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Bin Li
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Henan Evidence-Based Medicine Center of Chinese Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Qifei Zhao
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Rui Yu
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xinlu Wang
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Mingjun Zhu
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jingjing Wei
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
2
|
Soubeyrand S, Lau P, Nikpay M, Ma L, Bjorkegren JLM, McPherson R. Long Noncoding RNA TRIBAL Links the 8q24.13 Locus to Hepatic Lipid Metabolism and Coronary Artery Disease. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004674. [PMID: 39624902 DOI: 10.1161/circgen.124.004674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 10/11/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Genome-wide association studies identified a 20-Kb region of chromosome 8 (8q24.13) associated with plasma lipids, hepatic steatosis, and risk for coronary artery disease. The region is proximal to TRIB1, and given its well-established role in lipid regulation in animal models, TRIB1 has been proposed to mediate the contribution of the 8q24.13 locus to these traits. This region overlaps a gene encoding the primate-specific long noncoding RNA transcript TRIBAL/TRIB1AL (TRIB1-associated locus), but the contribution of TRIBAL to coronary artery disease risk remains untested. METHODS Using recently available expression quantitative trait loci data and hepatocyte models, we further investigated this locus by Mendelian randomization analysis. Following antisense oligonucleotide targeting of TRIBAL, transcription array, quantitative reverse transcription polymerase chain reaction, and enrichment analyses were performed and effects on apoB and triglyceride secretion were determined. RESULTS Mendelian randomization analysis supports a causal relationship between genetically determined hepatic TRIBAL expression and markers of hepatic steatosis and coronary artery disease risk. By contrast, expression data sets did not support expression quantitative trait loci relationships between coronary artery disease-associated variants and TRIB1. TRIBAL suppression reduced the expression of key regulators of triglyceride metabolism and bile acid synthesis. Enrichment analyses identified patterns consistent with impaired metabolic functions, including reduced triglyceride and cholesterol handling ability. Furthermore, TRIBAL suppression was associated with reduced hepatocyte secretion of triglycerides. CONCLUSIONS This work identifies TRIBAL as a gene bridging the genotype-phenotype relationship at the 8q24.13 locus with effects on genes regulating hepatocyte lipid metabolism and triglyceride secretion.
Collapse
Affiliation(s)
- Sébastien Soubeyrand
- Atherogenomics Laboratory (S.S., P.L., M.N., R.M.), University of Ottawa Heart Institute, Canada
| | - Paulina Lau
- Atherogenomics Laboratory (S.S., P.L., M.N., R.M.), University of Ottawa Heart Institute, Canada
| | - Majid Nikpay
- Atherogenomics Laboratory (S.S., P.L., M.N., R.M.), University of Ottawa Heart Institute, Canada
| | - Lijiang Ma
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (L.M., J.L.M.B.)
| | - Johan L M Bjorkegren
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (L.M., J.L.M.B.)
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden (J.L.M.B.)
| | - Ruth McPherson
- Atherogenomics Laboratory (S.S., P.L., M.N., R.M.), University of Ottawa Heart Institute, Canada
- Division of Cardiology, Ruddy Canadian Cardiovascular Genetics Centre (R.M.), University of Ottawa Heart Institute, Canada
| |
Collapse
|
3
|
Vilaça A, Jesus C, Lino M, Hayman D, Emanueli C, Terracciano CM, Fernandes H, de Windt LJ, Ferreira L. Extracellular vesicle transfer of lncRNA H19 splice variants to cardiac cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102233. [PMID: 38974998 PMCID: PMC11225836 DOI: 10.1016/j.omtn.2024.102233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/29/2024] [Indexed: 07/09/2024]
Abstract
The delivery of therapeutic long non-coding RNAs (lncRNA) to the heart by extracellular vesicles (EVs) is promising for heart repair. H19, a lncRNA acting as a major regulator of gene expression within the cardiovascular system, is alternatively spliced, but the loading of its different splice variants into EVs and their subsequent uptake by recipient cardiac cells remain elusive. Here, we dissected the cellular expression of H19 splice variants and their loading into EVs secreted by Wharton-Jelly mesenchymal stromal/stem cells (WJ-MSCs). We demonstrated that overexpression of the mouse H19 gene in WJ-MSCs induces the expression of H19 splice variants at different levels. Interestingly, EVs isolated from the H19-transfected WJ-MSCs (EV-H19) showed similar expression levels for all tested splice variant sets. In vitro, we further demonstrated that EV-H19 was taken up by cardiomyocytes, fibroblasts, and endothelial cells (ECs). Finally, analysis of EV tropism in living rat myocardial slices indicated that EVs were internalized mostly by cardiomyocytes and ECs. Collectively, our results indicated that EVs can be loaded with different lncRNA splice variants and successfully internalized by cardiac cells.
Collapse
Affiliation(s)
- Andreia Vilaça
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Institute of Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
- Department of Cardiology, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, the Netherlands
- PhD Program in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Carlos Jesus
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
| | - Miguel Lino
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Institute of Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
- Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
| | - Danika Hayman
- Imperial College London, National Heart and Lung Institute, London, UK
| | - Costanza Emanueli
- Imperial College London, National Heart and Lung Institute, London, UK
| | | | - Hugo Fernandes
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
- Multidisciplinary Institute of Ageing (MIA-Portugal), University of Coimbra, Coimbra, Portugal
| | - Leon J. de Windt
- Department of Cardiology, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
4
|
Jiang J, Wu H, Ji Y, Han K, Tang JM, Hu S, Lei W. Development and disease-specific regulation of RNA splicing in cardiovascular system. Front Cell Dev Biol 2024; 12:1423553. [PMID: 39045460 PMCID: PMC11263117 DOI: 10.3389/fcell.2024.1423553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/17/2024] [Indexed: 07/25/2024] Open
Abstract
Alternative splicing is a complex gene regulatory process that distinguishes itself from canonical splicing by rearranging the introns and exons of an immature pre-mRNA transcript. This process plays a vital role in enhancing transcriptomic and proteomic diversity from the genome. Alternative splicing has emerged as a pivotal mechanism governing complex biological processes during both heart development and the development of cardiovascular diseases. Multiple alternative splicing factors are involved in a synergistic or antagonistic manner in the regulation of important genes in relevant physiological processes. Notably, circular RNAs have only recently garnered attention for their tissue-specific expression patterns and regulatory functions. This resurgence of interest has prompted a reevaluation of the topic. Here, we provide an overview of our current understanding of alternative splicing mechanisms and the regulatory roles of alternative splicing factors in cardiovascular development and pathological process of different cardiovascular diseases, including cardiomyopathy, myocardial infarction, heart failure and atherosclerosis.
Collapse
Affiliation(s)
- Jinxiu Jiang
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Hongchun Wu
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yabo Ji
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Kunjun Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Jun-Ming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| |
Collapse
|
5
|
Ouyang S, Zhou ZX, Liu HT, Ren Z, Liu H, Deng NH, Tian KJ, Zhou K, Xie HL, Jiang ZS. LncRNA-mediated Modulation of Endothelial Cells: Novel Progress in the Pathogenesis of Coronary Atherosclerotic Disease. Curr Med Chem 2024; 31:1251-1264. [PMID: 36788688 DOI: 10.2174/0929867330666230213100732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/06/2022] [Accepted: 11/17/2022] [Indexed: 02/16/2023]
Abstract
Coronary atherosclerotic disease (CAD) is a common cardiovascular disease and an important cause of death. Moreover, endothelial cells (ECs) injury is an early pathophysiological feature of CAD, and long noncoding RNAs (lncRNAs) can modulate gene expression. Recent studies have shown that lncRNAs are involved in the pathogenesis of CAD, especially by regulating ECs. In this review, we summarize the novel progress of lncRNA-modulated ECs in the pathogenesis of CAD, including ECs proliferation, migration, adhesion, angiogenesis, inflammation, apoptosis, autophagy, and pyroptosis. Thus, as lncRNAs regulate ECs in CAD, lncRNAs will provide ideal and novel targets for the diagnosis and drug therapy of CAD.
Collapse
Affiliation(s)
- Shao Ouyang
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
- Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, Department of Cardiovascular Medicine, Hengyang Medical School, The Second Affiliated Hospital, Clinical Medicine Research Center of Arteriosclerotic Disease of Hunan Province, University of South China, Hunan 421001, China
| | - Zhi-Xiang Zhou
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Hui-Ting Liu
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Zhong Ren
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Huan Liu
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Nian-Hua Deng
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Kai-Jiang Tian
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Kun Zhou
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Hai-Lin Xie
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Zhi-Sheng Jiang
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| |
Collapse
|
6
|
Rodríguez-Esparragón F, Torres-Mata LB, Cazorla-Rivero SE, Serna Gómez JA, González Martín JM, Cánovas-Molina Á, Medina-Suárez JA, González-Hernández AN, Estupiñán-Quintana L, Bartolomé-Durán MC, Rodríguez-Pérez JC, Varas BC. Analysis of ANRIL Isoforms and Key Genes in Patients with Severe Coronary Artery Disease. Int J Mol Sci 2023; 24:16127. [PMID: 38003316 PMCID: PMC10671206 DOI: 10.3390/ijms242216127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
ANRIL (Antisense Noncoding RNA in the INK4 Locus), also named CDKN2B-AS1, is a long non-coding RNA with outstanding functions that regulates genes involved in atherosclerosis development. ANRIL genotypes and the expression of linear and circular isoforms have been associated with coronary artery disease (CAD). The CDKN2A and the CDKN2B genes at the CDKN2A/B locus encode the Cyclin-Dependent Kinase inhibitor protein (CDKI) p16INK4a and the p53 regulatory protein p14ARF, which are involved in cell cycle regulation, aging, senescence, and apoptosis. Abnormal ANRIL expression regulates vascular endothelial growth factor (VEGF) gene expression, and upregulated Vascular Endothelial Growth Factor (VEGF) promotes angiogenesis by activating the NF-κB signaling pathway. Here, we explored associations between determinations of the linear, circular, and linear-to-circular ANRIL gene expression ratio, CDKN2A, VEGF and its receptor kinase insert domain-containing receptor (KDR) and cardiovascular risk factors and all-cause mortality in high-risk coronary patients before they undergo coronary artery bypass grafting surgery (CABG). We found that the expression of ANRIL isoforms may help in the prediction of CAD outcomes. Linear isoforms were correlated with a worse cardiovascular risk profile while the expression of circular isoforms of ANRIL correlated with a decrease in oxidative stress. However, the determination of the linear versus circular ratio of ANRIL did not report additional information to that determined by the evaluation of individual isoforms. Although the expressions of the VEFG and KDR genes correlated with a decrease in oxidative stress, in binary logistic regression analysis it was observed that only the expression of linear isoforms of ANRIL and VEGF significantly contributed to the prediction of the number of surgical revascularizations.
Collapse
Affiliation(s)
- Francisco Rodríguez-Esparragón
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias de la Universidad de La Laguna, 38296 San Cristobal de La Laguna, Tenerife, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura B. Torres-Mata
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Department of Specific Didactics, University of Las Palmas de Gran Canaria, 35004 Las Palmas de Gran Canaria, Gran Canaria, Spain
| | - Sara E. Cazorla-Rivero
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Department of Internal Medicine, University of La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Jaime A. Serna Gómez
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Deparment of Cardiovascular Surgery, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
| | - Jesús M. González Martín
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ángeles Cánovas-Molina
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Chronic Pain Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
| | - José A. Medina-Suárez
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Department of Specific Didactics, University of Las Palmas de Gran Canaria, 35004 Las Palmas de Gran Canaria, Gran Canaria, Spain
| | - Ayose N. González-Hernández
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Deparment of Neurology and Clinical Neurophysiology, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
| | - Lidia Estupiñán-Quintana
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
| | - María C. Bartolomé-Durán
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
| | - José C. Rodríguez-Pérez
- Vice Chancellor of Research, Universidad Fernando Pessoa Canarias, 35002 Santa María de Guía de Gran Canaria, Gran Canaria, Spain;
| | - Bernardino Clavo Varas
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain; (L.B.T.-M.); (S.E.C.-R.); (J.A.S.G.); (J.M.G.M.); (Á.C.-M.); (J.A.M.-S.); (A.N.G.-H.); (L.E.-Q.); (M.C.B.-D.)
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias de la Universidad de La Laguna, 38296 San Cristobal de La Laguna, Tenerife, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Chronic Pain Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Radiation Oncology Department, Hospital Universitario de Gran Canaria Dr. Negrín, 35010 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Universitary Institute for Research in Biomedicine and Health (iUIBS), Molecular and Translational Pharmacology Group, University of Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Gran Canaria, Spain
- Spanish Group of Clinical Research in Radiation Oncology (GICOR), 28290 Madrid, Spain
| |
Collapse
|
7
|
Sanchez A, Lhuillier J, Grosjean G, Ayadi L, Maenner S. The Long Non-Coding RNA ANRIL in Cancers. Cancers (Basel) 2023; 15:4160. [PMID: 37627188 PMCID: PMC10453084 DOI: 10.3390/cancers15164160] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
ANRIL (Antisense Noncoding RNA in the INK4 Locus), a long non-coding RNA encoded in the human chromosome 9p21 region, is a critical factor for regulating gene expression by interacting with multiple proteins and miRNAs. It has been found to play important roles in various cellular processes, including cell cycle control and proliferation. Dysregulation of ANRIL has been associated with several diseases like cancers and cardiovascular diseases, for instance. Understanding the oncogenic role of ANRIL and its potential as a diagnostic and prognostic biomarker in cancer is crucial. This review provides insights into the regulatory mechanisms and oncogenic significance of the 9p21 locus and ANRIL in cancer.
Collapse
Affiliation(s)
| | | | | | - Lilia Ayadi
- CNRS, Université de Lorraine, IMoPA, F-54000 Nancy, France
| | | |
Collapse
|
8
|
Hu K, Huang MJ, Ling S, Li YX, Cao XY, Chen YF, Lei JM, Fu WZ, Tan BF. LncRNA CASC11 upregulation promotes HDAC4 to alleviate oxidized low-density lipoprotein-induced injury of cardiac microvascular endothelial cells. Kaohsiung J Med Sci 2023; 39:758-768. [PMID: 37096653 DOI: 10.1002/kjm2.12687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/15/2023] [Accepted: 03/14/2023] [Indexed: 04/26/2023] Open
Abstract
Long noncoding RNAs (LncRNAs) are essential to regulate the pathogenesis of coronary artery disease (CAD). This study was conducted to analyze the functionality of long noncoding RNA cancer susceptibility candidate 11 (lncRNA CASC11) in oxidized low-density lipoprotein (ox-LDL)-induced injury of cardiac microvascular endothelial cells (CMECs). CMECs were treated with ox-LDL to induce the CAD cell model. The cellular expression levels of CASC11 and histone deacetylase 4 (HDAC4) were determined by real-time quantitative polymerase chain reaction or Western blot assay. Cell absorbance, apoptosis, angiogenesis, and inflammation were evaluated by cell counting kit-8, flow cytometry, tube formation, and enzyme-linked immunosorbent assays. The subcellular localization of CASC11 was examined by the nuclear/cytoplasmic fractionation assay. The binding of human antigen R (HuR) to CASC11 and HDAC4 was analyzed by RNA immunoprecipitation. HDAC4 stability was determined after actinomycin D treatment. CASC11 was found to be decreased in the CAD cell model. CASC11 upregulation increased cell viability and angiogenesis and reduced apoptosis and inflammation. CASC11 bound to HuR and improved HDAC4 expression. HDAC4 downregulation counteracted the protective role of CASC11 overexpression in CMECs. In summary, CASC11 alleviated ox-LDL-induced injury of CMECs by binding to HuR and stabilizing HDAC4.
Collapse
Affiliation(s)
- Ke Hu
- Medical College, Hunan University of Medicine, Huaihua, China
| | - Min-Jiang Huang
- Medical College, Hunan University of Medicine, Huaihua, China
| | - Sha Ling
- Department of Cardiology, First Affiliated Hospital, Hunan University of Medicine, Huaihua, China
| | - Yu-Xian Li
- Medical College, Hunan University of Medicine, Huaihua, China
| | - Xiang-Yu Cao
- Medical College, Hunan University of Medicine, Huaihua, China
| | - Yue-Fu Chen
- Medical College, Hunan University of Medicine, Huaihua, China
| | - Jian-Ming Lei
- Department of Cardiology, First Affiliated Hospital, Hunan University of Medicine, Huaihua, China
| | - Wen-Zhe Fu
- Department of Cardiology, First Affiliated Hospital, Hunan University of Medicine, Huaihua, China
| | - Bi-Feng Tan
- Department of Cardiology, First Affiliated Hospital, Hunan University of Medicine, Huaihua, China
| |
Collapse
|
9
|
Wu X, Li J, Sun G, Yang J, Peng Y, Bai X, Wang L. Role of LncRNAs in the Pathogenesis of Coronary Artery Disease. Rev Cardiovasc Med 2023; 24:96. [PMID: 39076276 PMCID: PMC11273030 DOI: 10.31083/j.rcm2404096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 07/31/2024] Open
Abstract
Coronary artery disease (CAD), caused by coronary artery occlusion, is a common cardiovascular disease worldwide. Long non-coding RNAs (lncRNAs) are implicated in the regulation of endothelial cell injury, angiogenesis, plaque formation, and other pathological mechanisms in CAD by acting on different cell types. Some lncRNAs are significantly upregulated in CAD patients; however, other lncRNAs are significantly downregulated. Differential expression of lncRNAs in CAD patients enables them to be exploited as potential biomarkers to evaluate disease progression and diagnosis/prognosis in CAD patients. In this study, we reviewed the role of lncRNAs in the development of different clinical subtypes of CAD.
Collapse
Affiliation(s)
- Xinyu Wu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan, China
| | - Jingru Li
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan, China
| | - Guihu Sun
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan, China
| | - Yunzhu Peng
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan, China
| | - Xiangfeng Bai
- Department of Cardiac Surgery, The First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan, China
| | - Luqiao Wang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, 650032 Kunming, Yunnan, China
| |
Collapse
|
10
|
Azizidoost S, Nasrolahi A, Sheykhi-Sabzehpoush M, Akiash N, Assareh AR, Anbiyaee O, Antosik P, Dzięgiel P, Farzaneh M, Kempisty B. Potential roles of endothelial cells-related non-coding RNAs in cardiovascular diseases. Pathol Res Pract 2023; 242:154330. [PMID: 36696805 DOI: 10.1016/j.prp.2023.154330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Endothelial dysfunction is identified by a conversion of the endothelium toward decreased vasodilation and prothrombic features and is known as a primary pathogenic incident in cardiovascular diseases. An insight based on particular and promising biomarkers of endothelial dysfunction may possess vital clinical significances. Currently, non-coding RNAs due to their participation in critical cardiovascular processes like initiation and progression have gained much attention as possible diagnostic as well as prognostic biomarkers in cardiovascular diseases. Emerging line of proof has demonstrated that abnormal expression of non-coding RNAs is nearly correlated with the pathogenesis of cardiovascular diseases. In the present review, we focus on the expression and functional effects of various kinds of non-coding RNAs in cardiovascular diseases and negotiate their possible clinical implications as diagnostic or prognostic biomarkers and curative targets.
Collapse
Affiliation(s)
- Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Nehzat Akiash
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ahmad Reza Assareh
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Omid Anbiyaee
- Cardiovascular Research Center, Nemazi Hospital, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paweł Antosik
- Institute of Veterinary Medicine, Department of Veterinary Surgery, Nicolaus Copernicus University, Torun, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Bartosz Kempisty
- Institute of Veterinary Medicine, Department of Veterinary Surgery, Nicolaus Copernicus University, Torun, Poland; Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wrocław, Poland; North Carolina State University College of Agriculture and Life Sciences, Raleigh, NC 27695, USA.
| |
Collapse
|
11
|
Liu Y, Zhu ZX, Zboinski EK, Qiu W, Lian J, Liu S, Van Dyke TE, Johansson HE, Tu Q, Luo E, Chen JJ. Long non-coding RNA APDC plays important regulatory roles in metabolism of bone and adipose tissues. RNA Biol 2023; 20:836-846. [PMID: 37953645 PMCID: PMC10653663 DOI: 10.1080/15476286.2023.2268489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 11/14/2023] Open
Abstract
The long noncoding RNA (lncR) ANRIL in the human genome is an established genetic risk factor for atherosclerosis, periodontitis, diabetes, and cancer. However, the regulatory role of lncR-ANRIL in bone and adipose tissue metabolism remains unclear. To elucidate the function of lncRNA ANRIL in a mouse model, we investigated its ortholog, AK148321 (referred to as lncR-APDC), located on chr4 of the mouse genome, which is hypothesized to have similar biological functions to ANRIL. We initially revealed that lncR-APDC in mouse bone marrow cells (BMSCs) and lncR-ANRIL in human osteoblasts (hFOBs) are both increased during early osteogenesis. Subsequently, we examined the osteogenesis, adipogenesis, osteoclastogenesis function with lncR-APDC deletion/overexpression cell models. In vivo, we compared the phenotypic differences in bone and adipose tissue between APDC-KO and wild-type mice. Our findings demonstrated that lncR-APDC deficiency impaired osteogenesis while promoting adipogenesis and osteoclastogenesis. Conversely, the overexpression of lncR-APDC stimulated osteogenesis, but impaired adipogenesis and osteoclastogenesis. Furthermore, KDM6B was downregulated with lncR-APDC deficiency and upregulated with overexpression. Through binding-site analysis, we identified miR-99a as a potential target of lncR-APDC. The results suggest that lncR-APDC exerts its osteogenic function via miR-99a/KDM6B/Hox pathways. Additionally, osteoclasto-osteogenic imbalance was mediated by lncR-APDC through MAPK/p38 and TLR4/MyD88 activation. These findings highlight the pivotal role of lncR-APDC as a key regulator in bone and fat tissue metabolism. It shows potential therapeutic for addressing imbalances in osteogenesis, adipogenesis, and osteoclastogenesis.
Collapse
Affiliation(s)
- Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Zoe Xiaofang Zhu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Elissa K. Zboinski
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Wei Qiu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, USA
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junxiang Lian
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, USA
- Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Thomas E. Van Dyke
- Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection, and Immunity, Faculty of Medicine, Harvard University, Boston, MA, USA
| | - Hans E. Johansson
- Research and Development, LGC Biosearch Technologies, Petaluma, CA, USA
| | - Qisheng Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, USA
| | - En Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jake Jinkun Chen
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, USA
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
12
|
Tan AQ, Zheng YF. The Roles of SNHG Family in Osteoblast Differentiation. Genes (Basel) 2022; 13:2268. [PMID: 36553535 PMCID: PMC9777675 DOI: 10.3390/genes13122268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/17/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Small nucleolar RNA host genes (SNHGs), members of long-chain noncoding RNAs (lncRNAs), have received increasing attention regarding their roles in multiple bone diseases. Studies have revealed that SNHGs display unique expression profile during osteoblast differentiation and that they could act as promising biomarkers of certain bone diseases, such as osteoporosis. Osteogenesis of mesenchymal stem cells (MSCs) is an important part of bone repair and reconstruction. Moreover, studies confirmed that the SNHG family participate in the regulation of osteogenic differentiation of MSCs in part by regulating important pathways of osteogenesis, such as Wnt/β-catenin signaling. Based on these observations, clarifying the SNHG family's roles in osteogenesis (especially in MSCs) and their related mechanisms would provide novel ideas for possible applications of lncRNAs in the diagnosis and treatment of bone diseases. After searching, screening, browsing and intensive reading, we uncovered more than 30 papers related to the SNHG family and osteoblast differentiation that were published in recent years. Here, our review aims to summarize these findings in order to provide a theoretical basis for further research.
Collapse
Affiliation(s)
| | - Yun-Fei Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| |
Collapse
|
13
|
Li Z, Qiao J, Ma W, Zhou J, Gu L, Deng D, Zhang B. P14AS upregulates gene expression in the CDKN2A/2B locus through competitive binding to PcG protein CBX7. Front Cell Dev Biol 2022; 10:993525. [PMID: 36176277 PMCID: PMC9513069 DOI: 10.3389/fcell.2022.993525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/24/2022] [Indexed: 12/27/2022] Open
Abstract
Background: It is well known that P16 INK4A , P14 ARF , P15 INK4B mRNAs, and ANRIL lncRNA are transcribed from the CDKN2A/2B locus. LncRNA P14AS is a lncRNA transcribed from antisense strand of P14 ARF promoter to intron-1. Our previous study showed that P14AS could upregulate the expression level of ANRIL and P16 INK4A and promote the proliferation of cancer cells. Because polycomb group protein CBX7 could repress P16 INK4A expression and bind ANRIL, we wonder whether the P14AS-upregulated ANRIL and P16 INK4A expression is mediated with CBX7. Results: In this study, we found that the upregulation of P16 INK4A , P14 ARF , P15 INK4B and ANRIL expression was induced by P14AS overexpression only in HEK293T and HCT116 cells with active endogenous CBX7 expression, but not in MGC803 and HepG2 cells with weak CBX7 expression. Further studies showed that the stable shRNA-knockdown of CBX7 expression abolished the P14AS-induced upregulation of these P14AS target genes in HEK293T and HCT116 cells whereas enforced CBX7 overexpression enabled P14AS to upregulate expression of these target genes in MGC803 and HepG2 cells. Moreover, a significant association between the expression levels of P14AS and its target genes were observed only in human colon cancer tissue samples with high level of CBX7 expression (n = 38, p < 0.05), but not in samples (n = 37) with low level of CBX7 expression, nor in paired surgical margin tissues. In addition, the results of RNA immunoprecipitation (RIP)- and chromatin immunoprecipitation (ChIP)-PCR analyses revealed that lncRNA P14AS could competitively bind to CBX7 protein which prevented the bindings of CBX7 to both lncRNA ANRIL and the promoters of P16 INK4A , P14 ARF and P15 INK4B genes. The amounts of repressive histone modification H3K9m3 was also significantly decreased at the promoters of these genes by P14AS in CBX7 actively expressing cells. Conclusions: CBX7 expression is essential for P14AS to upregulate the expression of P16 INK4A , P14 ARF , P15 INK4B and ANRIL genes in the CDKN2A/2Blocus. P14AS may upregulate these genes' expression through competitively blocking CBX7-binding to ANRIL lncRNA and target gene promoters.
Collapse
Affiliation(s)
| | | | | | | | | | - Dajun Deng
- *Correspondence: Dajun Deng, ; Baozhen Zhang,
| | | |
Collapse
|
14
|
de Gonzalo-Calvo D, Sopić M, Devaux Y. Methodological considerations for circulating long noncoding RNA quantification. Trends Mol Med 2022; 28:616-618. [PMID: 35701316 DOI: 10.1016/j.molmed.2022.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022]
Abstract
In the past decade, significant resources have been invested in long noncoding RNA (lncRNA) research. Despite the knowledge available, we are far from incorporation of lncRNA into clinical practice. Here, we emphasize the technical challenges in the field, hoping to provoke a response leading to new consensus and guidelines.
Collapse
Affiliation(s)
- David de Gonzalo-Calvo
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain; CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain.
| | - Miron Sopić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia.
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | | |
Collapse
|
15
|
Bai N, Liu W, Xiang T, Zhou Q, Pu J, Zhao J, Luo D, Liu X, Liu H. Genetic association of ANRIL with susceptibility to Ischemic stroke: A comprehensive meta-analysis. PLoS One 2022; 17:e0263459. [PMID: 35653368 PMCID: PMC9162336 DOI: 10.1371/journal.pone.0263459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 01/19/2022] [Indexed: 11/18/2022] Open
Abstract
Background
Ischemic stroke (IS) is a complex polygenic disease with a strong genetic background. The relationship between the ANRIL (antisense non-coding RNA in the INK4 locus) in chromosome 9p21 region and IS has been reported across populations worldwide; however, these studies have yielded inconsistent results. The aim of this study is to clarify the types of single-nucleotide polymorphisms on the ANRIL locus associated with susceptibility to IS using meta-analysis and comprehensively assess the strength of the association.
Methods
Relevant studies were identified by comprehensive and systematic literature searches. The quality of each study was assessed using the Newcastle-Ottawa Scale. Allele and genotype frequencies were extracted from each of the included studies. Odds ratios with corresponding 95% confidence intervals of combined analyses were calculated under three genetic models (allele frequency comparison, dominant model, and recessive model) using a random-effects or fixed-effects model. Heterogeneity was tested using the chi-square test based on the Cochran Q statistic and I2 metric, and subgroup analyses and a meta-regression model were used to explore sources of heterogeneity. The correction for multiple testing used the false discovery rate method proposed by Benjamini and Hochberg. The assessment of publication bias employed funnel plots and Egger’s test.
Results
We identified 25 studies (15 SNPs, involving a total of 11,527 cases and 12,216 controls maximum) and performed a meta-analysis. Eight SNPs (rs10757274, rs10757278, rs2383206, rs1333040, rs1333049, rs1537378, rs4977574, and rs1004638) in ANRIL were significantly associated with IS risk. Six of these SNPs (rs10757274, rs10757278, rs2383206, rs1333040, rs1537378, and rs4977574) had a significant relationship to the large artery atherosclerosis subtype of IS. Two SNPs (rs2383206 and rs4977574) were associated with IS mainly in Asians, and three SNPs (rs10757274, rs1333040, and rs1333049) were associated with susceptibility to IS mainly in Caucasians. Sensitivity analyses confirmed the reliability of the original results. Ethnicity and individual studies may be the main sources of heterogeneity in ANRIL.
Conclusions
Our results suggest that some single-nucleotide polymorphisms on the ANRIL locus may be associated with IS risk. Future studies with larger sample numbers are necessary to confirm this result. Additional functional analyses of causal effects of these polymorphisms on IS subtypes are also essential.
Collapse
Affiliation(s)
- Na Bai
- Department of Neurology, The Third People’s Hospital of Chengdu & The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Wei Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
- Department of Neurology, Nanbu People’s Hospital, Nanbu, Sichuan, China
| | - Tao Xiang
- Department of Neurology, The Third People’s Hospital of Chengdu & The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Qiang Zhou
- Department of Neurology, The Third People’s Hospital of Chengdu & The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jun Pu
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jing Zhao
- Department of Neurology, Nanbu People’s Hospital, Nanbu, Sichuan, China
| | - Danyang Luo
- Nuclear Industry 416 Hospital & The Second Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xindong Liu
- Nuclear Industry 416 Hospital & The Second Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Hua Liu
- Department of Neurology, The Third People’s Hospital of Chengdu & The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
16
|
Liu Z, Zhang Z, Song G, Wang X, Xing H, Wang C. Resveratrol Alleviates Skeletal Muscle Insulin Resistance by Downregulating Long Noncoding RNA. Int J Endocrinol 2022; 2022:2539519. [PMID: 35096054 PMCID: PMC8791716 DOI: 10.1155/2022/2539519] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/15/2021] [Accepted: 01/04/2022] [Indexed: 01/04/2023] Open
Abstract
Long noncoding RNA (lncRNA) is a crucial factor in the progression of insulin resistance (IR). Resveratrol (RSV) exhibits promising therapeutic potential for IR. However, there are few studies on whether RSV improves IR through lncRNA. This study aimed to determine whether RSV could influence the expression of lncRNA and to elucidate the underlying mechanism. Mice were divided into three groups: control group, high-fat diet (HFD) group, and HFD + RSV group. We conducted a high-throughput sequencing analysis to detect lncRNA and mRNA expression signatures and the ceRNA-network in the skeletal muscles of mice that were fed an HFD to induce IR. Hierarchical clustering, gene enrichment, and gene ceRNA-network analyses were subsequently conducted. Differentially expressed lncRNAs were selected and validated via reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The biological functions of the selected lncRNAs were investigated by silencing the target genes via lentivirus transfection of C2C12 mouse myotube cells. RSV treatment reversed the expression of 338 mRNAs and 629 lncRNAs in the skeletal muscles of mice with HFD-induced IR. The results of the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes database analyses indicated that the differentially expressed mRNAs modulated type II diabetes mellitus. After validating randomly selected lncRNAs via RT-qPCR, we identified a novel lncRNA, NONMMUT044897.2, which was upregulated in the HFD group and reversed with RSV treatment. Additionally, NONMMUT044897.2 was proven to function as a ceRNA of microRNA- (miR-) 7051-5p. Suppressor of Cytokine Signaling 1 (SOCS1) was confirmed as a target of miR-7051-5p. We further performed lentivirus transfection to knock down NONMMUT044897.2 in vitro and found that NONMMUT044897.2 silenced SOCS1 and potentiated the insulin signaling pathway. Hence, RSV mimicked the silencing effect of lentivirus transfection on NONMMUT044897.2. Our study revealed that RSV reduced IR in mouse skeletal muscles via the regulation of NONMMUT044897.2.
Collapse
Affiliation(s)
- Zhihong Liu
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang 050017, China
- Endocrinology Department, Hebei General Hospital, Shijiazhuang 050051, China
- Endocrinology Department, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Zhimei Zhang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang 050017, China
- Endocrinology Department, Hebei General Hospital, Shijiazhuang 050051, China
| | - Guangyao Song
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang 050017, China
- Endocrinology Department, Hebei General Hospital, Shijiazhuang 050051, China
| | - Xing Wang
- Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang 050051, China
| | - Hanying Xing
- Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang 050051, China
| | - Chao Wang
- Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang 050051, China
| |
Collapse
|
17
|
Long non-coding RNA GAS5 inhibits osteogenic differentiation through miR-382-3p/ TAF1 signaling. Mol Cell Biol 2021; 42:e0054120. [PMID: 34898279 DOI: 10.1128/mcb.00541-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) have been confirmed as important regulators during osteogenic differentiation. Previous researches have disclosed that growth arrest-specific transcript 5 (GAS5) can promote the osteogenic differentiation of human bone marrow mesenchyml stem cells (hBMSCs), but the underlying regulatory mechanism of GAS5 during the osteogenic differentiation of hBMSCs is unclear. Methods: Osteogenic differentiation was induced in hBMSCs by using osteogenic medium (OM). Gene expression was assessed by RT-qPCR or western blot assays as needed. ALP activity, ALP staining and ARS staining assays were performed to evaluate the impact of GAS5, microRNA-382-3p (miR-382-3p) and TATA-box binding protein associated factor 1 (TAF1) on osteogenic differentiation in vitro. The interaction among GAS5, miR-382-3p and TAF1 was determined by RIP, ChIP and luciferase reporter assays. Results: Expression of GAS5 (transcript variant 2) was down-regulated during the osteogenic differentiation of hBMSCs and its overexpression retarded the osteogenic differentiation of hBMSCs. GAS5 inhibited miR-382-3p through targeting RNA-directed microRNA degradation (TDMD). MiR-382-3p down-regulation partially offset the promoted osteogenic differentiation of hBMSCs upon GAS5 silencing. TAF1 negatively modulated osteogenic differentiation and it activated GAS5 transcription so as to form a positive GAS5/miR-382-3p/TAF1 feedback loop in hBMSCs. Conclusion: This research was the first to reveal that the GAS5/miR-382-3p/TAF1 feedback loop inhibited the osteogenic differentiation of hBMSCs, which provided new clues for exploring the mechanism of osteogenic differentiation and disclosed the potential of GAS5 as a promising target during osteogenic differentiation.
Collapse
|
18
|
Zhu L, Zhao S, Zhao W. Potential Regulatory Role of lncRNA-miRNA-mRNA in Coronary Artery Disease (CAD). Int Heart J 2021; 62:1369-1378. [PMID: 34789641 DOI: 10.1536/ihj.21-156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Coronary artery disease (CAD) is a high-incidence of heart disease. We aimed to identify potential biomarkers linked to the progression of CAD using multiple sets of data mining analysis methods. The long noncoding RNA (lncRNA) + messenger RNA (mRNA) data set GSE113079 and microRNA (miRNA) data set GSE28858 were downloaded from Gene Expression Omnibus. After data preprocessing, differentially expressed mRNA, lncRNA, and miRNA were identified using limma software. In addition, weighted gene co-expression network analysis (WGCNA) was used for the construction and screening of modules related to disease states. Besides, key mRNAs and lncRNAs were extracted for protein-protein interaction (PPI) network construction and lncRNA-mRNA co-expression analysis. Additionally, the final integration resulted in the lncRNA-miRNA-mRNA relationship pairs (competing endogenous RNA (ceRNA) network). Finally, CTD 2020 update database was used for the verification of the expression level of the candidate genes. A total of 1319 differentially expressed mRNAs and 1983 lncRNAs were screened. After WGCNA, a total of 234 mRNAs and 546 lncRNAs were identified. A PPI network including 127 mRNA corresponding proteins was constructed. The ceRNA network included 24 up-regulated lncRNAs, 16 down-regulated miRNAs, and 42 up-regulated mRNAs. Through the validation of CTD 2020 update database, 21 CAD related mRNAs, and four important ceRNAs those may be participated in the pathogenesis of CAD were obtained. In this study, through multiple sets of data mining methods, the regulatory relationship of lncRNA, miRNA, and mRNA was comprehensively analyzed, and the important role of lncRNA-miRNA-mRNA in the pathogenesis of CAD was emphasized.
Collapse
Affiliation(s)
- Liyuan Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University
| | - Shuiping Zhao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University
| | - Wang Zhao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University
| |
Collapse
|
19
|
Long Non-Coding RNA Regulation of Epigenetics in Vascular Cells. Noncoding RNA 2021; 7:ncrna7040062. [PMID: 34698214 PMCID: PMC8544676 DOI: 10.3390/ncrna7040062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/02/2021] [Accepted: 09/14/2021] [Indexed: 01/16/2023] Open
Abstract
The vascular endothelium comprises the interface between the circulation and the vessel wall and, as such, is under the dynamic regulation of vascular signalling, nutrients, and hypoxia. Understanding the molecular drivers behind endothelial cell (EC) and vascular smooth muscle cell (VSMC) function and dysfunction remains a pivotal task for further clinical progress in tackling vascular disease. A newly emerging era in vascular biology with landmark deep sequencing approaches has provided us with the means to profile diverse layers of transcriptional regulation at a single cell, chromatin, and epigenetic level. This review describes the roles of major vascular long non-coding RNA (lncRNAs) in the epigenetic regulation of EC and VSMC function and discusses the recent progress in their discovery, detection, and functional characterisation. We summarise new findings regarding lncRNA-mediated epigenetic mechanisms—often regulated by hypoxia—within the vascular endothelium and smooth muscle to control vascular homeostasis in health and disease. Furthermore, we outline novel molecular techniques being used in the field to delineate the lncRNA subcellular localisation and interaction with proteins to unravel their biological roles in the epigenetic regulation of vascular genes.
Collapse
|
20
|
Xu Y, Chen J, Wang M, Yu R, Zou W, Shen W. Mechanism of lncRNA-ANRIL/miR-181b in autophagy of cardiomyocytes in mice with uremia by targeting ATG5. PLoS One 2021; 16:e0256734. [PMID: 34469488 PMCID: PMC8410126 DOI: 10.1371/journal.pone.0256734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 08/13/2021] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVES This study is to investigate whether the cardiac microvascular endothelial cells (CMECs) can regulate the autophagy of cardiomyocytes (CMs) by secreting lncRNA-ANRIL/miR-181b exosomes, thus participating in the occurrence of uremic cardiovascular disease (CVD). METHODS A 5/6 nephrectomy uremia model was established, with the mice injected with ANRIL-shRNA lentivirus vector, miR-181b agomir, and related control reagents, containing the serum creatinine and urea nitrogen measured. The renal tissue sections of mice were stained with Periodic Acid-Schiff (PAS), TUNEL, and Hematoxylin-Eosin (HE) performed on myocardial tissue sections of mice. ANRIL-shRNA, miR-181b mimics, and related control reagents were transfected into CMECs, in which the exosomes were extracted and co-cultured with CMs. The expressions of ANRIL, miR-181b and ATG5 were detected by qRT-PCR, and the expressions of autophagy related proteins by Western blot, as well as the binding of ANRIL and miR-181b by the double luciferase reporter gene experiment. RESULTS ANRIL down-regulation or miR-181b up-regulation can increase the weight of mice with uremia, as well as the expressions of p62 and miR-181b, and reduce the content of serum creatinine and urea nitrogen, the damage of kidney and myocardial tissues, the number of apoptotic cells in myocardial tissues, as well as the expressions of ANRIL, ATG5, Beclin1, and LC3. CMs can absorb the exosomes of CMECs. Compared with IS+ CMEC-Exo group, the expressions of ANRIL and ATG5 in CMs of IS+ CMEC-Exo + sh lncRNA ANRIL and IS+CMEC-Exo+miR-181b mimics groups was down-regulated, as well as the expressions of ATG5, Beclin1, and LC3, while miR-181b expression was up-regulated as well as P62 expression. CONCLUSIONS CMECs can regulate autophagy of CMs by releasing exosomes containing ANRIL and miR-181b.
Collapse
Affiliation(s)
- Ying Xu
- Department of Urology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Zhejiang, China
| | - Jing Chen
- Department of Urology, Tongde Hospital of Zhejiang Province, Zhejiang, China
| | - Minmin Wang
- Department of Nephrology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Zhejiang, China
| | - Rizhen Yu
- Department of Nephrology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Zhejiang, China
| | - Wenly Zou
- Department of Nephrology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Zhejiang, China
| | - Wei Shen
- Department of Nephrology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Zhejiang, China
| |
Collapse
|
21
|
Huang SF, Peng XF, Jiang L, Hu CY, Ye WC. LncRNAs as Therapeutic Targets and Potential Biomarkers for Lipid-Related Diseases. Front Pharmacol 2021; 12:729745. [PMID: 34421622 PMCID: PMC8371450 DOI: 10.3389/fphar.2021.729745] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/26/2021] [Indexed: 12/30/2022] Open
Abstract
Lipid metabolism is an essential biological process involved in nutrient adjustment, hormone regulation, and lipid homeostasis. An irregular lifestyle and long-term nutrient overload can cause lipid-related diseases, including atherosclerosis, myocardial infarction (MI), obesity, and fatty liver diseases. Thus, novel tools for efficient diagnosis and treatment of dysfunctional lipid metabolism are urgently required. Furthermore, it is known that lncRNAs based regulation like sponging microRNAs (miRNAs) or serving as a reservoir for microRNAs play an essential role in the progression of lipid-related diseases. Accordingly, a better understanding of the regulatory roles of lncRNAs in lipid-related diseases would provide the basis for identifying potential biomarkers and therapeutic targets for lipid-related diseases. This review highlighted the latest advances on the potential biomarkers of lncRNAs in lipid-related diseases and summarised current knowledge on dysregulated lncRNAs and their potential molecular mechanisms. We have also provided novel insights into the underlying mechanisms of lncRNAs which might serve as potential biomarkers and therapeutic targets for lipid-related diseases. The information presented here may be useful for designing future studies and advancing investigations of lncRNAs as biomarkers for diagnosis, prognosis, and therapy of lipid-related diseases.
Collapse
Affiliation(s)
- Shi-Feng Huang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Xiao-Fei Peng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Lianggui Jiang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Ching Yuan Hu
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Wen-Chu Ye
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| |
Collapse
|
22
|
Li J, Zhang Y, Zhang D, Li Y. The Role of Long Non-coding RNAs in Sepsis-Induced Cardiac Dysfunction. Front Cardiovasc Med 2021; 8:684348. [PMID: 34041287 PMCID: PMC8141560 DOI: 10.3389/fcvm.2021.684348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
Sepsis is a syndrome with life-threatening organ dysfunction induced by a dysregulated host response to infection. The heart is one of the most commonly involved organs during sepsis, and cardiac dysfunction, which is usually indicative of an extremely poor clinical outcome, is a leading cause of death in septic cases. Despite substantial improvements in the understanding of the mechanisms that contribute to the origin and responses to sepsis, the prognosis of sepsis-induced cardiac dysfunction (SICD) remains poor and its molecular pathophysiological changes are not well-characterized. The recently discovered group of mediators known as long non-coding RNAs (lncRNAs) have presented novel insights and opportunities to explore the mechanisms and development of SICD and may provide new targets for diagnosis and therapeutic strategies. LncRNAs are RNA transcripts of more than 200 nucleotides with limited or no protein-coding potential. Evidence has rapidly accumulated from numerous studies on how lncRNAs function in associated regulatory circuits during SICD. This review outlines the direct evidence of the effect of lncRNAs on SICD based on clinical trials and animal studies. Furthermore, potential functional lncRNAs in SICD that have been identified in sepsis studies are summarized with a proven biological function in research on other cardiovascular diseases.
Collapse
Affiliation(s)
- Jiawen 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
| | - Yulin Zhang
- 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
|