101
|
Epigenetics, HIV, and Cardiovascular Disease Risk. Curr Probl Cardiol 2020; 46:100615. [PMID: 32507271 DOI: 10.1016/j.cpcardiol.2020.100615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022]
Abstract
Human immunodeficiency virus (HIV) is currently considered a risk factor for cardiovascular disease (CVD). With the advent of antiretroviral treatment and prevention, HIV-related morbidity and mortality rates have decreased significantly. Prolonged life expectancy heralded higher prevalence of diseases of aging, including CVD-associated morbidity and mortality, having an earlier onset in people living with HIV (PLHIV) compared to their noninfected counterparts. Several epigenetic biomarkers are now available as predictors of health and disease, with DNA methylation being one of the most widely studied. Epigenetic biomarkers are changes in gene expression without alterations to the intrinsic DNA sequence, with the potential to predict risk of future CVD, as well as the outcome and response to therapy among PLHIV. We sought to review the available literature referencing epigenetic markers to determine underlying biomechanism predisposing high-risk PLHIV to CVD, elucidating areas of possible intervention.
Collapse
|
102
|
Jan MI, Ali T, Ishtiaq A, Mushtaq I, Murtaza I. Prospective Advances in Non-coding RNAs Investigation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:385-426. [PMID: 32285426 DOI: 10.1007/978-981-15-1671-9_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Non-coding RNAs (ncRNAs) play significant roles in numerous physiological cellular processes and molecular alterations during pathological conditions including heart diseases, cancer, immunological disorders and neurological diseases. This chapter is focusing on the basis of ncRNA relation with their functions and prospective advances in non-coding RNAs particularly miRNAs investigation in the cardiovascular disease management.The field of ncRNAs therapeutics is a very fascinating and challenging too. Scientists have opportunity to develop more advanced therapeutics as well as diagnostic approaches for cardiovascular conditions. Advanced studies are critically needed to deepen the understanding of the molecular biology, mechanism and modulation of ncRNAs and chemical formulations for managing CVDs.
Collapse
Affiliation(s)
- Muhammad Ishtiaq Jan
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Tahir Ali
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ayesha Ishtiaq
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Iram Mushtaq
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Iram Murtaza
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
103
|
Wang Q, Yang Y, Fu X, Wang Z, Liu Y, Li M, Zhang Y, Li Y, Li PF, Yu T, Chu XM. Long noncoding RNA XXYLT1-AS2 regulates proliferation and adhesion by targeting the RNA binding protein FUS in HUVEC. Atherosclerosis 2020; 298:58-69. [PMID: 32171981 DOI: 10.1016/j.atherosclerosis.2020.02.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/06/2020] [Accepted: 02/25/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS The endothelium is crucially involved in the pathogenesis of atherosclerosis according to accumulating evidence. Moreover, recent studies have showed that lncRNAs could serve as biomarkers of cardiovascular diseases, in particular atherosclerosis. However, the underlying mechanism of endothelial dysfunction involving lncRNAs in atherosclerosis remains unknown. This study investigated the mechanism of lncRNA XXYLT1-AS2 in endothelial dysfunction in atherosclerosis. METHODS The levels of lncRNA XXYLT1-AS2, FUS, VCAM-1, MCP-1, p-AKT, and p-P65 were measured in arteries and HUVEC cell lines via quantitative real-time PCR or Western blot. FISH assay demonstrated that XXYLT1-AS2 and FUS are localized in the nucleus. HUVECs were transfected with si-XXYLT1-AS2 or XXYLT1-AS2 to further assess cell proliferation, migration, and adhesion. Furthermore, bioinformatics analysis, RNA immunoprecipitation and immunofluorescence were performed to investigate the target genes of XXYLT1-AS2 and possible signal pathways. RESULTS Overexpression of XXYLT1-AS2 inhibited cell proliferation and migration, reduced the expression of adhesion molecules (VCAM-1) and chemoattractant proteins (MCP-1), and restrained monocyte adhesion to endothelial cells. Mechanistic investigations indicated that XXYLT1-AS2 directly interacts with the target gene FUS/cyclin D1 and modulates the proliferation and migration of endothelial cells (ECs). Moreover, XXYLT1-AS2 exerts a protective role against the inflammatory response in atherosclerosis by blocking NF-κB activity. Clinically, the involvement of XXYLT1-AS2/FUS was also observed in human arteries and the results were consistent with the in vitro analysis. CONCLUSIONS Our study identified a novel long non-coding RNA (XXYLT1-AS2) and suggests that it might act as an underlying therapeutic target in atherosclerosis-related diseases by regulating ECs functions.
Collapse
Affiliation(s)
- Qi Wang
- Department of Cardiology, The Affiliated hospital of Qingdao University, Qingdao, 266000, China
| | - Yanyan Yang
- Institute for translational medicine, Qingdao University, No. 38 Dengzhou Road, 266021, China
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated hospital of Qingdao University, Qingdao, 266000, China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated hospital of Qingdao University, Qingdao, 266000, China
| | - Yan Liu
- Institute for translational medicine, Qingdao University, No. 38 Dengzhou Road, 266021, China
| | - Min Li
- Institute for translational medicine, Qingdao University, No. 38 Dengzhou Road, 266021, China
| | - Yinfeng Zhang
- Institute for translational medicine, Qingdao University, No. 38 Dengzhou Road, 266021, China
| | - Yonghong Li
- Department of Cardiology, The Affiliated hospital of Qingdao University, Qingdao, 266000, China
| | - Pei-Feng Li
- Institute for translational medicine, Qingdao University, No. 38 Dengzhou Road, 266021, China
| | - Tao Yu
- Institute for translational medicine, Qingdao University, No. 38 Dengzhou Road, 266021, China; Department of Cardiac Ultrasound, The Affiliated hospital of Qingdao University, Qingdao, 266000, China.
| | - Xian-Ming Chu
- Department of Cardiology, The Affiliated hospital of Qingdao University, Qingdao, 266000, China; Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, No. 5 Zhiquan Road, Qingdao, 266000, China.
| |
Collapse
|
104
|
Feng X, Wang Y, Chen W, Xu S, Li L, Geng Y, Shen A, Gao H, Zhang L, Liu S. SIRT3 inhibits cardiac hypertrophy by regulating PARP-1 activity. Aging (Albany NY) 2020; 12:4178-4192. [PMID: 32139662 PMCID: PMC7093179 DOI: 10.18632/aging.102862] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 01/24/2020] [Indexed: 01/14/2023]
Abstract
Sirtuin 3 (SIRT3) is a type III histone deacetylase that inhibits cardiac hypertrophy. It is mainly localized in the mitochondria and is thus implicated in mitochondrial metabolism. Recent studies have shown that SIRT3 can also accumulate in the nuclear under stressed conditions, and participated in histone deacetylation of target proteins. Poly [ADP-ribose] polymerase 1 (PARP-1) functions as an important PARP isoform that was involved in cardiac hypertrophy. Our experiments showed that SIRT3 accumulated in the nuclear of cardiomyocytes treated with isoproterenol or SIRT3 overexpression. Moreover, overexpression of SIRT3 by adenovirus inhibited the expression of cardiac hypertrophic genes-ANF and BNP, as well as abrogating PARP-1 activation induced by isoproterenol or phenylephrine. In addition, co-immunoprecipitation experiments revealed that SIRT3 could interact with PARP-1, and overexpression of SIRT3 could decrease the acetylation level of PARP-1. Our results indicate that SIRT3 exerts protective effects against cardiac hypertrophy by reducing the level of acetylation and activity of PARP-1, thus providing novel mechanistic insights into SIRT3-mediated cardiprotective actions.
Collapse
Affiliation(s)
- Xiaojun Feng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China
| | - Yanan Wang
- Department of Pharmaceutics, College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, PR. China
| | - Wenxu Chen
- Department of Pharmaceutics, College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, PR. China
| | - Suowen Xu
- Aab Cardiovascular Research Institute, University of Rochester, West Henrietta, NY 14586, USA
| | - Lingli Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China
| | - Yadi Geng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China
| | - Aizong Shen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China.,Anhui Provincial Cardiovascular Institute, Hefei, Anhui, PR. China
| | - Hui Gao
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, PR. China
| | - Lei Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China
| | - Sheng Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR. China
| |
Collapse
|
105
|
Wei X, Yi X, Zhu XH, Jiang DS. Histone methylation and vascular biology. Clin Epigenetics 2020; 12:30. [PMID: 32070413 PMCID: PMC7027016 DOI: 10.1186/s13148-020-00826-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/09/2020] [Indexed: 12/20/2022] Open
Abstract
The vasculature not only transports oxygenated blood, metabolites, and waste products but also serves as a conduit for hormonal communication between distant tissues. Therefore, it is important to maintain homeostasis within the vasculature. Recent studies have greatly expanded our understanding of the regulation of vasculature development and vascular-related diseases at the epigenetic level, including by protein posttranslational modifications, DNA methylation, and noncoding RNAs. Integrating epigenetic mechanisms into the pathophysiologic conceptualization of complex and multifactorial vascular-related diseases may provide promising therapeutic approaches. Several reviews have presented detailed discussions of epigenetic mechanisms not including histone methylation in vascular biology. In this review, we primarily discuss histone methylation in vascular development and maturity, and in vascular diseases.
Collapse
Affiliation(s)
- Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, China
- NHC Key Laboratory of Organ Transplantation, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xue-Hai Zhu
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, China
- NHC Key Laboratory of Organ Transplantation, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, China.
- NHC Key Laboratory of Organ Transplantation, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
106
|
Epigenetic Signaling and RNA Regulation in Cardiovascular Diseases. Int J Mol Sci 2020; 21:ijms21020509. [PMID: 31941147 PMCID: PMC7014325 DOI: 10.3390/ijms21020509] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/14/2022] Open
Abstract
RNA epigenetics is perhaps the most recent field of interest for translational epigeneticists. RNA modifications create such an extensive network of epigenetically driven combinations whose role in physiology and pathophysiology is still far from being elucidated. Not surprisingly, some of the players determining changes in RNA structure are in common with those involved in DNA and chromatin structure regulation, while other molecules seem very specific to RNA. It is envisaged, then, that new small molecules, acting selectively on RNA epigenetic changes, will be reported soon, opening new therapeutic interventions based on the correction of the RNA epigenetic landscape. In this review, we shall summarize some aspects of RNA epigenetics limited to those in which the potential clinical translatability to cardiovascular disease is emerging.
Collapse
|
107
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
108
|
Hulshoff MS, del Monte-Nieto G, Kovacic J, Krenning G. Non-coding RNA in endothelial-to-mesenchymal transition. Cardiovasc Res 2019; 115:1716-1731. [PMID: 31504268 PMCID: PMC6755356 DOI: 10.1093/cvr/cvz211] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/17/2019] [Accepted: 08/29/2019] [Indexed: 02/06/2023] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT) is the process wherein endothelial cells lose their typical endothelial cell markers and functions and adopt a mesenchymal-like phenotype. EndMT is required for development of the cardiac valves, the pulmonary and dorsal aorta, and arterial maturation, but activation of the EndMT programme during adulthood is believed to contribute to several pathologies including organ fibrosis, cardiovascular disease, and cancer. Non-coding RNAs, including microRNAs, long non-coding RNAs, and circular RNAs, modulate EndMT during development and disease. Here, we review the mechanisms by which non-coding RNAs facilitate or inhibit EndMT during development and disease and provide a perspective on the therapeutic application of non-coding RNAs to treat fibroproliferative cardiovascular disease.
Collapse
Affiliation(s)
- Melanie S Hulshoff
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), Groningen, The Netherlands
- Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August University, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site, Göttingen, Germany
| | | | - Jason Kovacic
- Dept. Cardiology, Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), Groningen, The Netherlands
| |
Collapse
|
109
|
Feng X, Zhang L, Xu S, Shen AZ. ATP-citrate lyase (ACLY) in lipid metabolism and atherosclerosis: An updated review. Prog Lipid Res 2019; 77:101006. [PMID: 31499095 DOI: 10.1016/j.plipres.2019.101006] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/17/2019] [Accepted: 08/18/2019] [Indexed: 12/21/2022]
Abstract
ATP citrate lyase (ACLY) is an important enzyme linking carbohydrate to lipid metabolism by generating acetyl-CoA from citrate for fatty acid and cholesterol biosynthesis. Mendelian randomization of large human cohorts has validated ACLY as a promising target for low-density-lipoprotein-cholesterol (LDL-C) lowering and cardiovascular protection. Among current ACLY inhibitors, Bempedoic acid (ETC-1002) is a first-in-class, prodrug-based direct competitive inhibitor of ACLY which regulates lipid metabolism by upregulating hepatic LDL receptor (LDLr) expression and activity. ACLY deficiency in hepatocytes protects from hepatic steatosis and dyslipidemia. In addition, pharmacological inhibition of ACLY by bempedoic acid, prevents dyslipidemia and attenuates atherosclerosis in hypercholesterolemic ApoE-/- mice, LDLr-/- mice, and LDLr-/- miniature pigs. Convincing data from clinical trials have revealed that bempedoic acid significantly lowers LDL-C as monotherapy, combination therapy, and add-on with statin therapy in statin-intolerant patients. More recently, a phase 3 CLEAR Harmony clinical trial ("Safety and Efficacy of Bempedoic Acid to Reduce LDL Cholesterol") has shown that bempedoic acid reduces the level of LDL-C in hypercholesterolemic patients receiving guideline-recommended statin therapy with a good safety profile. Hereby, we provide a updated review of the expression, regulation, genetics, functions of ACLY in lipid metabolism and atherosclerosis, and highlight the therapeutic potential of ACLY inhibitors (such as bempedoic acid, SB-204990, and other naturally-occuring inhibitors) to treat atherosclerotic cardiovascular diseases. It must be pointed out that long-term large-scale clinical trials in high-risk patients, are warranted to validate whether ACLY represent a promising therapeutic target for pharmaceutic intervention of dyslipidemia and atherosclerosis; and assess the safety and efficacy profile of ACLY inhibitors in improving cardiovascular outcome of patients.
Collapse
Affiliation(s)
- Xiaojun Feng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, PR China
| | - Lei Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, PR China
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | - Ai-Zong Shen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, PR China.
| |
Collapse
|
110
|
Long Noncoding Competing Endogenous RNA Networks in Age-Associated Cardiovascular Diseases. Int J Mol Sci 2019; 20:ijms20123079. [PMID: 31238513 PMCID: PMC6627372 DOI: 10.3390/ijms20123079] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the most serious health problem in the world, displaying high rates of morbidity and mortality. One of the main risk factors for CVDs is age. Indeed, several mechanisms are at play during aging, determining the functional decline of the cardiovascular system. Aging cells and tissues are characterized by diminished autophagy, causing the accumulation of damaged proteins and mitochondria, as well as by increased levels of oxidative stress, apoptosis, senescence and inflammation. These processes can induce a rapid deterioration of cellular quality-control systems. However, the molecular mechanisms of age-associated CVDs are only partially known, hampering the development of novel therapeutic strategies. Evidence has emerged indicating that noncoding RNAs (ncRNAs), such as long ncRNAs (lncRNAs) and micro RNAs (miRNAs), are implicated in most patho-physiological mechanisms. Specifically, lncRNAs can bind miRNAs and act as competing endogenous-RNAs (ceRNAs), therefore modulating the levels of the mRNAs targeted by the sponged miRNA. These complex lncRNA/miRNA/mRNA networks, by regulating autophagy, apoptosis, necrosis, senescence and inflammation, play a crucial role in the development of age-dependent CVDs. In this review, the emerging knowledge on lncRNA/miRNA/mRNA networks will be summarized and the way in which they influence age-related CVDs development will be discussed.
Collapse
|
111
|
Skuratovskaia D, Vulf M, Komar A, Kirienkova E, Litvinova L. Promising Directions in Atherosclerosis Treatment Based on Epigenetic Regulation Using MicroRNAs and Long Noncoding RNAs. Biomolecules 2019; 9:E226. [PMID: 31212708 PMCID: PMC6627269 DOI: 10.3390/biom9060226] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/03/2019] [Accepted: 06/07/2019] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is one of the leading causes of mortality from cardiovascular disease (CVD) and is a chronic inflammatory disease of the middle and large arteries caused by a disruption of lipid metabolism. Noncoding RNA (ncRNA), including microRNA (miRNA), small interfering RNA (siRNA) and long noncoding RNA (lncRNA), was investigated for the treatment of atherosclerosis. Regulation of the expression of noncoding RNA targets the constituent element of the pathogenesis of atherosclerosis. Currently, miRNA therapy commonly employs miRNA antagonists and mimic compounds. In this review, attention is focused on approaches to correcting molecular disorders based on the genetic regulation of the transcription of key genes responsible for the development of atherosclerosis. Promising technologies were considered for the treatment of atherosclerosis, and examples are given for technologies that have been shown to be effective in clinical trials.
Collapse
Affiliation(s)
- Daria Skuratovskaia
- Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| | - Maria Vulf
- Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| | - Aleksandra Komar
- Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| | - Elena Kirienkova
- Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| | - Larisa Litvinova
- Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| |
Collapse
|
112
|
Zhang M, Zheng Y, Sun Y, Li S, Chen L, Jin X, Hou X, Liu X, Chen Q, Li J, Liu M, Zheng X, Zhang Y, Wu J, Yu B. Knockdown of NEAT1 induces tolerogenic phenotype in dendritic cells by inhibiting activation of NLRP3 inflammasome. Am J Cancer Res 2019; 9:3425-3442. [PMID: 31281488 PMCID: PMC6587165 DOI: 10.7150/thno.33178] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
Rationale: Tolerogenic dendritic cells (tol-DCs) play essential roles in immune-related diseases and induce immune tolerance by shaping T-cell responses. Accumulating evidence suggests that long noncoding RNAs (lncRNAs) play important regulatory roles in the immune system. However, the potential roles and underlying mechanisms of lncRNAs in tol-DCs remain unclear. Methods: RNA in-situ hybridization, histochemistry, and qRT-PCR were performed to determine the distribution and expression of NEAT1 in DCs. Flow cytometry was used to analyze the tolerogenic function of DCs. Small sequencing, followed by bioinformatic analysis, was performed to determine the target genes of NEAT1. The mechanism of NEAT1 was explored using a luciferase reporter, chromatin immunoprecipitation assays, and Immunofluorescence. In-vivo experiments were used to investigate the induction of immune tolerance via NEAT1-knockdown DCs. Results: Our results show that lncRNA NEAT1 can induce tolerogenic phenotype in DCs. Mechanistically, small RNA-seq analysis revealed that NEAT1 knockdown preferentially affected the expression of miR-3076-3p. Furthermore, NEAT1 used the NLRP3 inflammasome as a molecular decoy for miR-3076-3p, thus facilitating the expression of tolerogenic phenotype in DCs. Moreover, the transcription factor E2F1 acted as a repressor of NEAT1 transcription via activity of H3K27ac. Our results also indicate that NEAT1 knockdown in DCs can induce immune tolerance in models of experimental autoimmune myocarditis and heart transplantation. Conclusions: Taken together, our study shows the mechanism used by NEAT1 in inducing tol-DCs and highlights the therapeutic potential of targeting NEAT1 for the treatment of immune-related diseases.
Collapse
|
113
|
Guillot A, Tacke F. Liver Macrophages: Old Dogmas and New Insights. Hepatol Commun 2019; 3:730-743. [PMID: 31168508 PMCID: PMC6545867 DOI: 10.1002/hep4.1356] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/28/2019] [Indexed: 12/12/2022] Open
Abstract
Inflammation is a hallmark of virtually all liver diseases, such as liver cancer, fibrosis, nonalcoholic steatohepatitis, alcoholic liver disease, and cholangiopathies. Liver macrophages have been thoroughly studied in human disease and mouse models, unravelling that the hepatic mononuclear phagocyte system is more versatile and complex than previously believed. Liver macrophages mainly consist of liver‐resident phagocytes, or Kupffer cells (KCs), and bone marrow‐derived recruited monocytes. Although both cell populations in the liver demonstrate principal functions of macrophages, such as phagocytosis, danger signal recognition, cytokine release, antigen processing, and the ability to orchestrate immune responses, KCs and recruited monocytes retain characteristic ontogeny markers and remain remarkably distinct on several functional aspects. While KCs dominate the hepatic macrophage pool in homeostasis (“sentinel function”), monocyte‐derived macrophages prevail in acute or chronic injury (“emergency response team”), making them an interesting target for novel therapeutic approaches in liver disease. In addition, recent data acquired by unbiased large‐scale techniques, such as single‐cell RNA sequencing, unraveled a previously unrecognized complexity of human and murine macrophage polarization abilities, far beyond the old dogma of inflammatory (M1) and anti‐inflammatory (M2) macrophages. Despite tremendous progress, numerous challenges remain in deciphering the full spectrum of macrophage activation and its implication in either promoting liver disease progression or repairing injured liver tissue. Being aware of such heterogeneity in cell origin and function is of crucial importance when studying liver diseases, developing novel therapeutic interventions, defining macrophage‐based prognostic biomarkers, or designing clinical trials. Growing knowledge in gene expression modulation and emerging technologies in drug delivery may soon allow shaping macrophage populations toward orchestrating beneficial rather than detrimental inflammatory responses.
Collapse
Affiliation(s)
- Adrien Guillot
- Laboratory of Liver Diseases National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health Bethesda MD.,Department of Hepatology/Gastroenterology Charité University Medical Center Berlin Germany
| | - Frank Tacke
- Department of Hepatology/Gastroenterology Charité University Medical Center Berlin Germany
| |
Collapse
|
114
|
Feng X, Sureda A, Jafari S, Memariani Z, Tewari D, Annunziata G, Barrea L, Hassan ST, Šmejkal K, Malaník M, Sychrová A, Barreca D, Ziberna L, Mahomoodally MF, Zengin G, Xu S, Nabavi SM, Shen AZ. Berberine in Cardiovascular and Metabolic Diseases: From Mechanisms to Therapeutics. Theranostics 2019; 9:1923-1951. [PMID: 31037148 PMCID: PMC6485276 DOI: 10.7150/thno.30787] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/05/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular and metabolic diseases (CVMD) are the leading causes of death worldwide, underscoring the urgent necessity to develop new pharmacotherapies. Berberine (BBR) is an eminent component of traditional Chinese and Ayurvedic medicine for more than 2000 years. Recently, BBR has attracted much interest for its pharmacological actions in treating and/or managing CVMD. Recent discoveries of basic, translational and clinical studies have identified many novel molecular targets of BBR (such as AMPK, SIRT1, LDLR, PCSK9, and PTP1B) and provided novel evidences supporting the promising therapeutic potential of BBR to combat CVMD. Thus, this review provides a timely overview of the pharmacological properties and therapeutic application of BBR in CVMD, and underlines recent pharmacological advances which validate BBR as a promising lead drug against CVMD.
Collapse
|
115
|
Rutaecarpine: A promising cardiovascular protective alkaloid from Evodia rutaecarpa (Wu Zhu Yu). Pharmacol Res 2019; 141:541-550. [DOI: 10.1016/j.phrs.2018.12.019] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022]
|
116
|
Niu N, Xu S, Xu Y, Little PJ, Jin ZG. Targeting Mechanosensitive Transcription Factors in Atherosclerosis. Trends Pharmacol Sci 2019; 40:253-266. [PMID: 30826122 DOI: 10.1016/j.tips.2019.02.004] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/23/2019] [Accepted: 02/05/2019] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is the primary underlying cause of cardiovascular disease which preferentially develops at arterial regions exposed to disturbed flow (DF), but much less at regions of unidirectional laminar flow (UF). Recent studies have demonstrated that DF and UF differentially regulate important aspects of endothelial function, such as vascular inflammation, oxidative stress, vascular tone, cell proliferation, senescence, mitochondrial function, and glucose metabolism. DF and UF regulate vascular pathophysiology via differential regulation of mechanosensitive transcription factors (MSTFs) (KLF2, KLF4, NRF2, YAP/TAZ/TEAD, HIF-1α, NF-κB, AP-1, and others). Emerging studies show that MSTFs represent promising therapeutic targets for the prevention and treatment of atherosclerosis. We present here a comprehensive overview of the role of MSTFs in atherosclerosis, and highlight future directions for developing novel therapeutic agents by targeting MSTFs.
Collapse
Affiliation(s)
- Niu Niu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - Yanni Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; National Health Commission (NHC) Key Laboratory of Biotechnology of Antibiotics, National Center for Drug (Microbiology) Screening Laboratory, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS and PUMC), Beijing, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Wooloongabba, QLD 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-Sen University, Guangzhou 510520, China
| | - Zheng-Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
117
|
Wang J, Niu N, Xu S, Jin ZG. A simple protocol for isolating mouse lung endothelial cells. Sci Rep 2019; 9:1458. [PMID: 30728372 PMCID: PMC6365507 DOI: 10.1038/s41598-018-37130-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/26/2018] [Indexed: 12/13/2022] Open
Abstract
Endothelial dysfunction is the common molecular basis of multiple human diseases, such as atherosclerosis, diabetes, hypertension, and acute lung injury. Therefore, primary isolation of high-purity endothelial cells (ECs) is crucial to study the mechanisms of endothelial function and disease pathogenesis. Mouse lung ECs (MLECs) are widely used in vascular biology and lung cell biology studies such as pulmonary inflammation, angiogenesis, vessel permeability, leukocyte/EC interaction, nitric oxide production, and mechanotransduction. Thus, in this paper, we describe a simple, and reproducible protocol for the isolation and culture of MLECs from adult mice using collagenase I-based enzymatic digestion, followed by sequential sorting with PECAM1 (also known as CD31)- and ICAM2 (also known as CD102)-coated microbeads. The morphology of isolated MLECs were observed with phase contrast microscope. MLECs were authenticated by CD31 immunoblotting, and immunofluorescent staining of established EC markers VE-cadherin and von Willebrand factor (vWF). Cultured MLECs also showed functional characteristics of ECs, evidenced by DiI-oxLDL uptake assay and THP-1 monocyte adhesion assay. Finally, we used MLECs from endothelium-specific enhancer of zeste homolog 2 (EZH2) knockout mice to show the general applicability of our protocol. To conclude, we describe here a simple and reproducible protocol to isolate highly pure and functional ECs from adult mouse lungs. Isolation of ECs from genetically engineered mice is important for downstream phenotypic, genetic, or proteomic studies.
Collapse
Affiliation(s)
- Jinping Wang
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, 14623, USA
- Department of Pharmacy, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Niu Niu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, 14623, USA
| | - Suowen Xu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, 14623, USA
| | - Zheng Gen Jin
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, 14623, USA.
| |
Collapse
|
118
|
Wang G, Zheng X, Zheng Y, Cao R, Zhang M, Sun Y, Wu J. Construction and analysis of the lncRNA‑miRNA‑mRNA network based on competitive endogenous RNA reveals functional genes in heart failure. Mol Med Rep 2018; 19:994-1003. [PMID: 30569169 PMCID: PMC6323221 DOI: 10.3892/mmr.2018.9734] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) is a principal cause of morbidity and mortality worldwide, affecting an estimated 38 million people. Although significant progress has been made with respect to the underlying molecular mechanisms, the role of the competing endogenous RNA (ceRNA) network in the pathogenesis of HF remains largely unknown. In this study, an HF-associated ceRNA network was constructed based on the differentially expressed long noncoding RNAs (lncRNAs), microRNAs (miRNAs) and mRNAs obtained, respectively, from the GSE77399, GSE104150 and GSE84796 datasets. The ceRNA network consisted of 12 lncRNA nodes, 43 miRNA nodes, 343 mRNA nodes and 530 edges. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis demonstrated that the ceRNA network was primarily enriched in the immune response, inflammatory response and T cell and B cell receptor signaling pathways. In addition, three lncRNAs (growth arrest specific 5, taurine upregulated 1 and HOX transcript antisense RNA) and three miRNAs [hsa-miRNA (miR)-26b-5p, hsa-miR-8485 and hsa-miR-940] with higher node degrees compared with other genes were selected as hub nodes. The expression of hub nodes in patients with HF was verified by reverse transcription-quantitative polymerase chain reaction analysis. The present study provided further insights into the important roles of the ceRNA network in HF development, and indicated the potential use of these hub nodes as diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Guohong Wang
- Department of Cardiovascular Center of Beijing Tongren Hospital, Affiliated to Capital Medical University, Beijing 100730, P.R. China
| | - Xianghui Zheng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150010, P.R. China
| | - Yang Zheng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150010, P.R. China
| | - Rui Cao
- Cardiovascular Department, The First Hospital of Harbin, Harbin, Heilongjiang 150010, P.R. China
| | - Maomao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150010, P.R. China
| | - Yong Sun
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150010, P.R. China
| | - Jian Wu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150010, P.R. China
| |
Collapse
|