1
|
Mao S, Liu ZY, Liu ZY, Liu P, Lin LC, Zhang Y, Yang JJ, Zhao JY, Tao H. Phase separation of epigenetic landscape in cardiovascular diseases. Biomed Pharmacother 2024; 181:117654. [PMID: 39522265 DOI: 10.1016/j.biopha.2024.117654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/03/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The pathogenesis of cardiovascular diseases (CVDs) is intricate, with liquid-liquid phase separation (LLPS) considered a crucial regulatory mechanism. Epigenetics is closely intertwined with cardiovascular diseases, involving mechanisms such as DNA methylation, histone modifications, and non-coding RNAs (ncRNAs) that play pivotal roles in cardiovascular disease progression and regression. It is known that specific proteins and mRNAs associated with epigenetic modifications exhibit LLPS characteristics, influencing cardiovascular diseases. Consequently, targeting epigenetic modifications to modulate LLPS emerges as a promising strategy for cardiovascular diseases treatment. This review delves into the regulatory impact of liquid-liquid phase separation on cardiovascular diseases, with a specific focus on the epigenetic landscape. The current study sought to investigate the relationship between epigenetic landscape and phase separation in cardiovascular diseases development, as well as their therapeutic implications.
Collapse
Affiliation(s)
- Sui Mao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhen-Yu Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Peng Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| |
Collapse
|
2
|
Tapia A, Liu X, Malhi NK, Yuan D, Chen M, Southerland KW, Luo Y, Chen ZB. Role of long noncoding RNAs in diabetes-associated peripheral arterial disease. Cardiovasc Diabetol 2024; 23:274. [PMID: 39049097 PMCID: PMC11271017 DOI: 10.1186/s12933-024-02327-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic disease that heightens the risks of many vascular complications, including peripheral arterial disease (PAD). Various types of cells, including but not limited to endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and macrophages (MΦs), play crucial roles in the pathogenesis of DM-PAD. Long non-coding RNAs (lncRNAs) are epigenetic regulators that play important roles in cellular function, and their dysregulation in DM can contribute to PAD. This review focuses on the developing field of lncRNAs and their emerging roles in linking DM and PAD. We review the studies investigating the role of lncRNAs in crucial cellular processes contributing to DM-PAD, including those in ECs, VSMCs, and MΦ. By examining the intricate molecular landscape governed by lncRNAs in these relevant cell types, we hope to shed light on the roles of lncRNAs in EC dysfunction, inflammatory responses, and vascular remodeling contributing to DM-PAD. Additionally, we provide an overview of the research approach and methodologies, from identifying disease-relevant lncRNAs to characterizing their molecular and cellular functions in the context of DM-PAD. We also discuss the potential of leveraging lncRNAs in the diagnosis and therapeutics for DM-PAD. Collectively, this review provides a summary of lncRNA-regulated cell functions contributing to DM-PAD and highlights the translational potential of leveraging lncRNA biology to tackle this increasingly prevalent and complex disease.
Collapse
Affiliation(s)
- Alonso Tapia
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA, 91010, USA
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Xuejing Liu
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Naseeb Kaur Malhi
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Dongqiang Yuan
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Muxi Chen
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Kevin W Southerland
- Division of Vascular and Endovascular Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yingjun Luo
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Zhen Bouman Chen
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA, 91010, USA.
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
3
|
Necroptosis Plays a Crucial Role in Vascular Injury during DVT and Is Enhanced by IL-17B. J Immunol Res 2022; 2022:6909764. [PMID: 36046722 PMCID: PMC9424031 DOI: 10.1155/2022/6909764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/16/2022] [Indexed: 12/16/2022] Open
Abstract
Background. This study investigated whether vascular endothelial necroptosis is involved in deep vein thrombosis (DVT) and how IL-17B facilitates necroptosis signaling. Methods. The DVT mouse model was induced by ligation of the IVC. The cross-sectional area of thrombus increases and the thrombus occupied the entire venous lumen at 48 h after ligation. Meanwhile, the increased expression of p-RIP3/RIP3 was most pronounced at 48 h after ligation, and the p-MLKL/MLKL peaked at 72 h. Results. Based on Illumina sequencing and KEGG pathway analyses, the activated RIP3/MLKL is associated with increased IL-17B. With thrombus formation, IL-17B was upregulated and enhanced the expression of RIP3 and MLKL in the IVC wall, as well as their phosphorylation levels (all
, the comparison group consisted of the control group, DVT group, DVT/IL-17B group, and DVT/anti-IL-17B group). The p-RIP3/RIP3 and p-MLKL/MLKL ratios were reduced by anti-IL-17B. Similarly, the weight and cross-sectional area of the thrombi were increased by IL-17B and decreased by the IL-17B antibody. IL-17B had a smaller effect on thrombosis in knockout mice compared with WT mice. In vitro, the IL-17B protein expression and the level of RIP3 and MLKL phosphorylation increased high in the OGD cells, accompanied by increased expression of IL-6 and TNF-α. IL-17B enhanced the expression of IL-6 and TNF-α but had little effect on the IL-6 and TNF-α after transfected with siRIP3 or siMLKL. Similarly, the plasma IL-17B, IL-6, and TNF-α were significantly increased after thrombosis in WT mice, and enhanced by IL-17B. But IL-17B did not increase the plasma IL-6 and TNF-α in knockout mice. Conclusions. In conclusion, those results suggest that vascular endothelial necroptosis plays a crucial role in vascular injury and IL-17B could enhance the necroptosis pathway.
Collapse
|
4
|
Liang S, Xv W, Li C, Huang Y, Qian G, Yan Y, Zou H, Li Y. Os LncRNAs Estão Envolvidos no Processo de Aterosclerose em Diversos Níveis. Arq Bras Cardiol 2022; 118:1134-1140. [PMID: 35703653 PMCID: PMC9345145 DOI: 10.36660/abc.20201383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 06/09/2021] [Indexed: 11/30/2022] Open
Abstract
A aterosclerose é a causa mais comum de doença cardiovascular em todo o mundo, ela está associada a uma alta incidência de eventos clínicos. O acúmulo de evidências elucidou que os RNAs longos não codificantes (LncRNAs) são uma nova classe de transcritos com papéis críticos nos processos fisiopatológicos da aterosclerose. Nesta revisão, resumimos o progresso recente dos LncRNAs no desenvolvimento da aterosclerose. Descrevemos principalmente os diversos mecanismos regulatórios dos LncRNAs nos níveis transcricionais e pós-transcricionais. Este estudo pode fornecer informações úteis sobre os LncRNAs como alvos terapêuticos ou biomarcadores para o tratamento da aterosclerose.
Collapse
|
5
|
Functional Implications of Intergenic GWAS SNPs in Immune-Related LncRNAs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1363:147-160. [DOI: 10.1007/978-3-030-92034-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
6
|
Emerging role of long non-coding RNAs in endothelial dysfunction and their molecular mechanisms. Biomed Pharmacother 2021; 145:112421. [PMID: 34798473 DOI: 10.1016/j.biopha.2021.112421] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are the novel class of transcripts involved in transcriptional, post-transcriptional, translational, and post-translational regulation of physiology and the pathology of diseases. Studies have evidenced that the impairment of endothelium is a critical event in the pathogenesis of atherosclerosis and its complications. Endothelial dysfunction is characterized by an imbalance in vasodilation and vasoconstriction, oxidative stress, proinflammatory factors, and nitric oxide bioavailability. Disruption of the endothelial barrier permeability, the first step in developing atherosclerotic lesions is a consequence of endothelial dysfunction. Though several factors interfere with the normal functioning of the endothelium, intrinsic epigenetic mechanisms governing endothelial function are regulated by lncRNAs and perturbations contribute to the pathogenesis of the disease. This review comprehensively addresses the biogenesis of lncRNA and molecular mechanisms underlying and regulation in endothelial function. An insight correlating lncRNAs and endothelial dysfunction-associated diseases can positively impact the development of novel biomarkers and therapeutic targets in endothelial dysfunction-associated diseases and treatment strategies.
Collapse
|
7
|
Programmed cell death in aortic aneurysm and dissection: A potential therapeutic target. J Mol Cell Cardiol 2021; 163:67-80. [PMID: 34597613 DOI: 10.1016/j.yjmcc.2021.09.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/07/2021] [Accepted: 09/23/2021] [Indexed: 12/20/2022]
Abstract
Rupture of aortic aneurysm and dissection (AAD) remains a leading cause of death. Progressive smooth muscle cell (SMC) loss is a crucial feature of AAD that contributes to aortic dysfunction and degeneration, leading to aortic aneurysm, dissection, and, ultimately, rupture. Understanding the molecular mechanisms of SMC loss and identifying pathways that promote SMC death in AAD are critical for developing an effective pharmacologic therapy to prevent aortic destruction and disease progression. Cell death is controlled by programmed cell death pathways, including apoptosis, necroptosis, pyroptosis, and ferroptosis. Although these pathways share common stimuli and triggers, each type of programmed cell death has unique features and activation pathways. A growing body of evidence supports a critical role for programmed cell death in the pathogenesis of AAD, and inhibitors of various types of programmed cell death represent a promising therapeutic strategy. This review discusses the different types of programmed cell death pathways and their features, induction, contributions to AAD development, and therapeutic potential. We also highlight the clinical significance of programmed cell death for further studies.
Collapse
|
8
|
Long Noncoding RNA OIP5-AS1 Contributes to the Progression of Atherosclerosis by Targeting miR-26a-5p Through the AKT/NF-κB Pathway. J Cardiovasc Pharmacol 2021; 76:635-644. [PMID: 32833899 DOI: 10.1097/fjc.0000000000000889] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis (AS) is a cardiovascular disease caused by multiple factors, leading to high mortality and morbidity in aged people. Some long noncoding RNAs have been reported to be associated with AS progression. However, the roles of OIP5-AS1 in AS development are still little known. In this study, the levels of OIP5-AS1 and miR-26a-5p in oxidized low-density lipoprotein (ox-LDL)-treated human umbilical vein endothelial cells (HUVECs) were determined by quantitative real-time polymerase chain reaction. Cell proliferation and apoptosis were evaluated by Cell Counting Kit-8 assay and flow cytometric analysis, respectively. The protein levels of proliferating cell nuclear antigen, B-cell lymphoma-2, cleaved caspase 3, inflammatory cytokines (IL-6 and IL-1β), protein kinase B (AKT), p-AKT, p65, p-p65, IκBα, and p-IκBα were detected by Western blot analysis. The targeting relationship between OIP5-AS1 and miR-26a-5p was verified by dual-luciferase reporter assay, RNA immunoprecipitation assay, and RNA pull-down assay. As a result, the expression of OIP5-AS1 was upregulated and miR-26a-5p was downregulated in ox-LDL-treated HUVECs. MiR-26a-5p was identified as a direct target of OIP5-AS1. OIP5-AS1 knockdown reversed the inhibitory effect on cell proliferation and the promotional effects on apoptosis and inflammation induced by ox-LDL treatment in HUVECs. Interestingly, the effects caused by OIP5-AS1 knockdown were further attenuated by miR-26a-5p inhibition. Furthermore, OIP5-AS1 knockdown blocked the AKT/NF-κB pathway by regulating miR-26a-5p expression. In conclusion, OIP5-AS1 knockdown promoted cell proliferation and suppressed apoptosis and inflammatory response in ox-LDL-treated HUVECs by targeting miR-26a-5p through blocking the AKT/NF-κB pathway, indicating a promising strategy for AS treatment.
Collapse
|
9
|
Zhu Q, Zhao C, Wang Y, Li X, Xue Y, Ma C. LncRNA NEAT1 Promote Inflammatory Responses in Coronary Slow Flow Through Regulating miR-148b-3p/ICAM-1 Axis. J Inflamm Res 2021; 14:2445-2463. [PMID: 34135616 PMCID: PMC8200141 DOI: 10.2147/jir.s312583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/13/2021] [Indexed: 12/25/2022] Open
Abstract
Background Coronary slow flow (CSF) is an angiographic phenomenon characterized by delayed coronary opacification with normal or near-normal epicardial coronary arteries. The pathogenesis of CSF is closely related to inflammatory response. Accumulating evidence shows that long non-coding RNAs (lncRNAs) play an important role in cardiovascular disease. However, the mechanism underlying the influence of the lncRNA nuclear enriched abundant transcripts 1 (NEAT1) on CSF is still unknown. Patients and Methods Forty CSF patients and forty control subjects were included in the study and underwent coronary angiography, Seattle angina questionnaire (SAQ) and echocardiography. The plasma levels of the inflammatory factors soluble intercellular adhesion molecule-1 (sICAM-1), interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) were determined by ELISA. The expression levels of NEAT1, miR-148b-3p and ICAM-1 in cells were measured by qRT-PCR or Western blotting. Cell proliferation was measured by 5‐Ethynyl‐2ʹ‐deoxyuridine (EdU) and Cell Counting Kit-8 (CCK-8) assay. Cell apoptosis was detected by apoptosis assay. The relationship between NEAT1 and miR-148b-3p was verified by luciferase reporter gene assay, RNA immunoprecipitation (RIP) assay and avidin-biotin pull-down assay. The relationship between ICAM-1 and miR-148b-3p was verified by luciferase reporter gene assay and avidin-biotin pull-down assay. Results This study showed that plasma sICAM-1, miR-148b-3p, and NEAT1 as independent predictors of a CSF diagnosis. Furthermore, plasma NEAT1 level showed superior diagnostic ability for CSF compared with sICAM-1 and miR-148b-3p. It was also shown that high expression of NEAT1 in oxygen-glucose deprivation (OGD)-treated human umbilical vein endothelial cells (HUVECs) functions as a competing endogenous RNA (ceRNA). By specifically binding miR-148b-3p, it weakened the negative regulatory effects of miR-148b-3p on the ICAM-1 target gene leading to upregulated expression of ICAM-1. This interaction was also shown to inhibit HUVEC proliferation and enhance apoptosis. Conclusion This study demonstrated for the first time the important mechanism of action of the NEAT1/miR-148b-3p/ICAM-1 axis in the progression of CSF disease, and indicated the potential of NEAT1, miR-148b-3p, and ICAM-1 as a new target for the diagnosis and treatment of CSF.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Cuiting Zhao
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yonghuai Wang
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xinxin Li
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, People's Republic of China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
10
|
Zhang L, Li Q, Chen Y, Zhu Q. LncRNA OIP5-AS1 accelerates ox-LDL-treated HUVECs injury by NF-κB pathway via miR-30c-5p. Clin Hemorheol Microcirc 2021; 78:449-460. [PMID: 33867356 DOI: 10.3233/ch-211130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Oxidized low-density lipoprotein (ox-LDL) could induce endothelial injury and played a vital role in the progression and development of atherosclerosis. This study aimed to investigate the role of Opa-interacting protein 5 antisense RNA 1 (OIP5-AS1) in ox-LDL-induced human umbilical vascular endothelial cells (HUVECs) injury and the potential mechanisms. METHODS Cell proliferation and apoptosis were evaluated by Cell Counting Kit-8 (CCK-8) assay and flow cytometry assay, respectively. The levels of lactate dehydrogenase (LDH), reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD) and nitric oxide (NO) were detected by corresponding detection kits, respectively. Quantitative real-time PCR (qRT-PCR) was conducted to measure the expression of OIP5-AS1 or microRNA-30c-5p (miR-30c-5p) in HUVECs. Binding between OIP5-AS1 and miR-30c-5p was predicted through bioinformatics analysis and confirmed by dual-luciferase reporter assay and RNA immunoprecipitation (RIP). Western blot was used to analyze p-IκB, IκB, p-p65 and p65 levels. RESULTS In HUVECs, exposure to ox-LDL led to a decrease in cell viability and an increase in LDH release and apoptosis with concomitant enhancement of oxidative stress, as evidenced by increased ROS and MDA generation, as well as decreased SOD activity and NO levels, while OIP5-AS1 knockdown or miR-30c-5p upregulation could rescue these effects above. Mechanically, OIP5-AS1 functioned as a sponge of miR-30c-5p. OIP5-AS1-induced injury and apoptosis, oxidative stress and activation of NF-κB pathway were reversed by miR-30c-5p in ox-LDL-treated HUVECs. CONCLUSION OIP5-AS1 contributed to ox-LDL-treated HUVECs injury by activation of NF-κB pathway via miR-30c-5p.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Cardiology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Qiulai Li
- Department of Cardiology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Yanxia Chen
- Department of Rehabilitation, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Qiao Zhu
- Department of Cardiology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| |
Collapse
|
11
|
Jiang Y, Sun-Waterhouse D, Chen Y, Li F, Li D. Epigenetic mechanisms underlying the benefits of flavonoids in cardiovascular health and diseases: are long non-coding RNAs rising stars? Crit Rev Food Sci Nutr 2021; 62:3855-3872. [PMID: 33427492 DOI: 10.1080/10408398.2020.1870926] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cardiovascular diseases (CVDs) rank as the first leading cause of death globally. High dietary polyphenol (especially flavonoids) intake has strongly been associated with low incidence of the primary outcome, overall mortality, blood pressure, inflammatory biomarkers, onset of new-onset type 2 diabetes mellitus (T2DM), and obesity. Phytogenic flavonoids affect the physiological and pathological processes of CVDs by modulating various biochemical signaling pathways. Non-coding RNAs (ncRNAs) have attracted increasing attention as fundamental regulator of gene expression involved in CVDs. Among the different ncRNA subgroups, long ncRNAs (lncRNAs) have recently emerged as regulatory eukaryotic transcripts and therapeutic targets with important and diverse functions in health and diseases. lncRNAs may be associated with the initiation, development and progression of CVDs by modulating acute and chronic inflammation, adipogenesis and lipid metabolism, and cellular physiology. This review summarizes this research on the modulatory effects of lncRNAs and their roles in mediating cellular processes. The mechanisms of action of flavonoids underlying their therapeutic effects on CVDs are also discussed. Based on our review, flavonoids might facilitate a significant epigenetic modification as part (if not full) of their tissue-/cell-related biological effects. This finding may be attributed to their interaction with cellular signaling pathways involved in chronic diseases. Certain lncRNAs might be the target of specific flavonoids, and some critical signaling processes involved in the intervention of CVDs might mediate the therapeutic roles of flavonoids.
Collapse
Affiliation(s)
- Yang Jiang
- College of Food Science and Engineering, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Shandong Agricultural University, Taian, PR China
| | | | - Yilun Chen
- College of Food Science and Engineering, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Shandong Agricultural University, Taian, PR China
| | - Feng Li
- College of Food Science and Engineering, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Shandong Agricultural University, Taian, PR China
| | - Dapeng Li
- College of Food Science and Engineering, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, Shandong Agricultural University, Taian, PR China
| |
Collapse
|
12
|
Aznaourova M, Schmerer N, Schmeck B, Schulte LN. Disease-Causing Mutations and Rearrangements in Long Non-coding RNA Gene Loci. Front Genet 2020; 11:527484. [PMID: 33329688 PMCID: PMC7735109 DOI: 10.3389/fgene.2020.527484] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
The classic understanding of molecular disease-mechanisms is largely based on protein-centric models. During the past decade however, genetic studies have identified numerous disease-loci in the human genome that do not encode proteins. Such non-coding DNA variants increasingly gain attention in diagnostics and personalized medicine. Of particular interest are long non-coding RNA (lncRNA) genes, which generate transcripts longer than 200 nucleotides that are not translated into proteins. While most of the estimated ~20,000 lncRNAs currently remain of unknown function, a growing number of genetic studies link lncRNA gene aberrations with the development of human diseases, including diabetes, AIDS, inflammatory bowel disease, or cancer. This suggests that the protein-centric view of human diseases does not capture the full complexity of molecular patho-mechanisms, with important consequences for molecular diagnostics and therapy. This review illustrates well-documented lncRNA gene aberrations causatively linked to human diseases and discusses potential lessons for molecular disease models, diagnostics, and therapy.
Collapse
Affiliation(s)
- Marina Aznaourova
- Institute for Lung Research, Philipps University Marburg, Marburg, Germany
| | - Nils Schmerer
- Institute for Lung Research, Philipps University Marburg, Marburg, Germany
| | - Bernd Schmeck
- Institute for Lung Research, Philipps University Marburg, Marburg, Germany.,Systems Biology Platform, German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany.,Center for Synthetic Microbiology (SYNMIKRO), Philipps University Marburg, Marburg, Germany
| | - Leon N Schulte
- Institute for Lung Research, Philipps University Marburg, Marburg, Germany.,Systems Biology Platform, German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| |
Collapse
|
13
|
Ou M, Hao S, Chen J, Zhao S, Cui S, Tu J. Downregulation of interleukin-6 and C-reactive protein underlies a novel inhibitory role of microRNA-136-5p in acute lower extremity deep vein thrombosis. Aging (Albany NY) 2020; 12:21076-21090. [PMID: 33188660 PMCID: PMC7695373 DOI: 10.18632/aging.103140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/31/2020] [Indexed: 12/23/2022]
Abstract
Deep vein thrombosis (DVT) comprises a critical and common health condition with high incidence, mortality, and long-term adverse sequelae. Several differentially expressed microRNAs (miRNAs) have emerged as promising prognostic markers in DVT. The present study intended to explore the functional relevance of miR-136-5p in acute lower extremity DVT (LEDVT). Rat models of acute LEDVT were established and miR-136-5p expression was altered by agomir or antagomir to assess its effects. In addition, in vitro gain- and loss-experiments, prior to exposure to CoCl2, were performed to investigate effects of miR-136-5p on human umbilical vein endothelial cell (HUVEC) apoptosis and levels of interleukin-6 (IL-6) and C-reactive protein (CRP). miR-136-5p was downregulated, whereas IL-6 and CRP were elevated in acute LEDVT patients. Notably, miR-136-5p was confirmed to target both IL-6 and CRP. Overexpression of miR-136-5p led to reduced length, weight, and ratio of weight to length of the venous thrombus. Furthermore, overexpressed miR-136-5p downregulated the expression of IL-6 and CRP, consequently inhibiting HUVEC apoptosis. Conjointly, our data indicate that the overexpression of miR-136-5p has the potential to bind to the 3’-UTR in the mRNAs for IL-6 and CRP and mitigate acute LEDVT, which provides a basis for new therapeutic targets in acute LEDVT treatment.
Collapse
Affiliation(s)
- Minghui Ou
- Department of Vascular Surgery, Qingdao Municipal Hospital, Qingdao 266011, P.R. China
| | - Shaobo Hao
- Department of Emergency, Qingdao Municipal Hospital, Qingdao 266011, P.R. China
| | - Jing Chen
- Department of Vascular Surgery, Qingdao Municipal Hospital, Qingdao 266011, P.R. China
| | - Shibo Zhao
- Department of Vascular Surgery, Qingdao Municipal Hospital, Qingdao 266011, P.R. China
| | - Shichao Cui
- Department of Vascular Surgery, Qingdao Municipal Hospital, Qingdao 266011, P.R. China
| | - Jie Tu
- Department of Science and Education, Qingdao Municipal Hospital, Qingdao 266011, P.R. China
| |
Collapse
|
14
|
Yuan Y, Xu L, Geng Z, Liu J, Zhang L, Wu Y, He D, Qu P. The role of non-coding RNA network in atherosclerosis. Life Sci 2020; 265:118756. [PMID: 33189816 DOI: 10.1016/j.lfs.2020.118756] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/29/2020] [Accepted: 11/10/2020] [Indexed: 12/26/2022]
Abstract
Atherosclerosis is the primary culprit of cardiovascular and cerebrovascular diseases. Also, atherogenesis and the development of atherosclerosis involve endothelial cells, monocytes/macrophages, smooth myocytes, and others. Increasingly, studies have found that non-coding RNA (ncRNA) which can regulate apoptosis, pyroptosis, autophagy, proliferation, and monocyte migration participates in atherogenesis and progress of atherosclerosis by the above. The ncRNA networks may be essential in regulating the complicated process of atherosclerosis. Accordingly, this review delves into the regulatory roles of ncRNA, which were introduced previously. The answer above is particularly crucial to explain further the regulatory mechanism of ncRNA in cardiovascular disorders. Furthermore, we discuss the possibility and related research of ncRNAs as a biomarker and therapeutic target for the prevention, diagnosis, and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yuchan Yuan
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116023, People's Republic of China
| | - Ling Xu
- Department of clinical laboratory, Xinhua Hospital Affiliated to Dalian University, Dalian 116021, People's Republic of China
| | - Zhaohong Geng
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, People's Republic of China
| | - Jingjing Liu
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116023, People's Republic of China
| | - Lijiao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, People's Republic of China
| | - Yuhang Wu
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116023, People's Republic of China
| | - Dan He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, People's Republic of China.
| | - Peng Qu
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116023, People's Republic of China; Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, People's Republic of China.
| |
Collapse
|
15
|
Lin Y, Dan H, Lu J. Overexpression of microRNA-136-3p Alleviates Myocardial Injury in Coronary Artery Disease via the Rho A/ROCK Signaling Pathway. Kidney Blood Press Res 2020; 45:477-496. [PMID: 32434208 DOI: 10.1159/000505849] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 01/08/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Coronary artery disease (CAD) is a cardiovascular disease that poses a fatal threat to human health, and the identification of potential biomarkers may help to delineate its pathophysiological mechanisms. Accumulating evidence has implicated microRNAs (miRNAs) in the pathogenesis and development of cardiovascular diseases. The present study aims to identify the expression of miRNA-136-3p (miR-136-3p) in CAD and further investigate its functional relevance in myocardial injury both in vitro and in vivo. METHODS Initially, CAD models were induced in rats by high-fat diet and intraperitoneal injection of pituitrin. Next, the effect of overexpressed miR-136-3p on cardiac function and pathological damage in myocardial tissue, cardiomyocyte apoptosis, oxidative stress and inflammatory response were assessed in CAD rats. Rat cardiac microvascular endothelial cells (CMECs) were isolated and cultured by the tissue explant method, and the CMEC injury model was induced by homocysteine (HCY). The function of miR-136-3p in vitro was further evaluated. RESULTS miR-136-3p was poorly expressed in the myocardial tissue of CAD rats and CMEC injury models. In vivo assays indicated that overexpressed miR-136-3p could improve cardiac function and alleviate pathological damage in myocardial tissue, accompanied by reduced oxidative stress and inflammatory response. Moreover,in vitro assays suggested that overexpression of miR-136-3p enhanced proliferation and migration while inhibiting apoptosis of HCY-stressed CMECs. Notably, we revealed that EIF5A2 was a target gene of miR-136-3p, and miR-136-3p inhibited EIF5A2 expression and activation of the Rho A/ROCK signaling pathway. CONCLUSION In conclusion, the overexpression of miR-136-3p could potentially impede myocardial injury in vitro and in vivo in CAD through the blockade of the Rho A/ROCK signaling pathway, highlighting a potential miR-136-3p functional relevance in the treatment of CAD.
Collapse
Affiliation(s)
- Yongbo Lin
- Department of Cardiology, People's Hospital of Dongxihu District, Wuhan, China
| | - Hanliang Dan
- Department of Cardiology, People's Hospital of Dongxihu District, Wuhan, China
| | - Jinguo Lu
- Department of Cardiology, Hospital of Traditional Chinese and Western Medicine in Hubei Province, Wuhan, China,
| |
Collapse
|
16
|
Abstract
Currently, cardiovascular diseases continue to be the leading cause of death worldwide; therefore, atherosclerosis remains one of the most crucial public health problems. This chronic and complex disease is considered to be a result of aberrant lipid homeostasis and inflammation of the inner wall of arteries that leads to plaque development. In recent years, a specific class of non-coding RNAs that are characterised by transcript lengths longer than 200 nucleotides, called long non-coding RNAs (lncRNAs), has emerged. Moreover, a growing body of evidence indicates that deregulation of lncRNA expression may contribute to the development of many diseases. Despite continuous efforts in deciphering the molecular basis of atherosclerotic plaque (AP) formation, many aspects of this process remain elusive. Therefore, continuing efforts in this area should remain the highest priority in the coming years. Establishment of a standardised experimental pipeline and validation of lncRNAs as possible relevant biomarkers for cardiovascular disease would enable the translation of gathered findings into clinical practice.
Collapse
Affiliation(s)
- Weronika Kraczkowska
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Science, 6 Święcickiego Street, 60-781, Poznan, Poland.
| | - Paweł Piotr Jagodziński
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Science, 6 Święcickiego Street, 60-781, Poznan, Poland
| |
Collapse
|
17
|
Navarro E, Mallén A, Cruzado JM, Torras J, Hueso M. Unveiling ncRNA regulatory axes in atherosclerosis progression. Clin Transl Med 2020; 9:5. [PMID: 32009226 PMCID: PMC6995802 DOI: 10.1186/s40169-020-0256-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023] Open
Abstract
Completion of the human genome sequencing project highlighted the richness of the cellular RNA world, and opened the door to the discovery of a plethora of short and long non-coding RNAs (the dark transcriptome) with regulatory or structural potential, which shifted the balance of pathological gene alterations from coding to non-coding RNAs. Thus, disease risk assessment currently has to also evaluate the expression of new RNAs such as small micro RNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), competing endogenous RNAs (ceRNAs), retrogressed elements, 3'UTRs of mRNAs, etc. We are interested in the pathogenic mechanisms of atherosclerosis (ATH) progression in patients suffering Chronic Kidney Disease, and in this review, we will focus in the role of the dark transcriptome (non-coding RNAs) in ATH progression. We will focus in miRNAs and in the formation of regulatory axes or networks with their mRNA targets and with the lncRNAs that function as miRNA sponges or competitive inhibitors of miRNA activity. In this sense, we will pay special attention to retrogressed genomic elements, such as processed pseudogenes and Alu repeated elements, that have been recently seen to also function as miRNA sponges, as well as to the use or miRNA derivatives in gene silencing, anti-ATH therapies. Along the review, we will discuss technical developments associated to research in lncRNAs, from sequencing technologies to databases, repositories and algorithms to predict miRNA targets, as well as new approaches to miRNA function, such as integrative or enrichment analysis and their potential to unveil RNA regulatory networks.
Collapse
Affiliation(s)
- Estanislao Navarro
- Independent Researcher, Barcelona, Spain. .,Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| | - Adrian Mallén
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Josep M Cruzado
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Joan Torras
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Miguel Hueso
- Department of Nephrology, Hospital Universitari Bellvitge and Bellvitge Research Institute (IDIBELL), C/Feixa Llarga, s/n; L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| |
Collapse
|
18
|
Endothelial function and T-lymphocyte subsets in patients with overlap syndrome of chronic obstructive pulmonary disease and obstructive sleep apnea. Chin Med J (Engl) 2020; 132:1654-1659. [PMID: 31283649 PMCID: PMC6759095 DOI: 10.1097/cm9.0000000000000312] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The coexistence of chronic obstructive pulmonary disease (COPD) and obstructive sleep apnea (OSA) is termed overlap syndrome (OS). COPD and OSA both have increased risks of developing cardiovascular diseases. This study aimed to explore if patients with OS exhibited a higher prevalence of cardiovascular complications, and if patients with OS exhibited vascular endothelial dysfunction and abnormalities in the cellular immune function of T lymphocytes. METHODS Totally 25 patients with stable COPD (COPD group), 25 patients with OSA (OSA group), 25 patients with OS (OS group), and 20 healthy adults (control group) were enrolled between January 2017 and December 2017 from the Respiratory Department of Tianjin Medical University General Hospital. The clinical characteristics of the four groups were collected and the expression levels of soluble vascular cell adhesion molecule-1 (sVCAM-1), tumor necrosis factor-α (TNF-α), and T-lymphocyte subsets were detected. One-way analysis of variance, χ test and Pearson correlation were used to manage the data. RESULTS The prevalence of hypertension and coronary heart disease was significantly higher in the OS group than in the control, OSA, and COPD groups (χ = 20.69, P < 0.05 and χ = 11.03, P < 0.05, respectively). The levels of sVCAM-1 and TNF-α were significantly higher in the OS group than in other groups (F = 127.40, P < 0.05 and F = 846.77, P < 0.05, respectively). The percentage of CD4+ lymphocytes and CD4+/CD8+ were both significantly lower in the OS group than in any other group (F = 25.40, P < 0.05 and F = 75.08, P < 0.05, respectively). There were significantly negative correlations in the levels of sVCAM-1 and TNF-α with CD4+/CD8+ lymphocytes (r = -0.77, P < 0.05 and r = -0.83, P < 0.05, respectively). CONCLUSIONS The prevalence of hypertension and coronary heart disease was higher in patients with OS than in patients with either OSA or COPD alone. Patients with OS exhibited more severe vascular endothelial injury, stronger inflammatory response, and lower cellular immune function.
Collapse
|
19
|
Noncoding RNAs in Vascular Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7914957. [PMID: 31998442 PMCID: PMC6969641 DOI: 10.1155/2020/7914957] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/20/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023]
Abstract
Increases in age are accompanied by vascular aging, which can lead to a variety of chronic diseases, including atherosclerosis and hypertension. Noncoding RNAs (ncRNAs) have become a research hotspot in different fields of life sciences in recent years. For example, these molecules have been found to have regulatory roles in many physiological and pathological processes. Many studies have shown that microRNAs (miRNAs) and long ncRNAs (lncRNAs) also play a regulatory role in vascular aging. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are important components of blood vessels, and the senescence of both cell types promotes the occurrence of vascular aging. This review provides a contemporary update on the molecular mechanisms underlying the senescence of ECs and VSMCs and the regulatory role of miRNAs and lncRNAs in this process.
Collapse
|
20
|
Jiang W, Agrawal DK, Boosani CS. Non-coding RNAs as Epigenetic Gene Regulators in Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:133-148. [PMID: 32285409 DOI: 10.1007/978-981-15-1671-9_7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epigenetic gene regulations can be considered as de-novo initiation of abnormal molecular signaling events whose regulation is otherwise required during normal or specific developmental stages of the organisms. Primarily, three different mechanisms have been identified to participate in epigenetic gene regulations which include, DNA methylation, non-coding RNA species (microRNAs [miRNA], and long non-coding RNAs [LNC-RNA]) and histone modifications. These de-novo epigenetic mechanisms have been associated with altered normal cellular functions which eventually facilitate normal cells to transition into an abnormal phenotype. Among the three modes of regulation, RNA species which are usually considered to be less stable, can be speculated to initiate instant alterations in gene expression compared to DNA methylation or histone modifications. However, LNC-RNAs appear to be more stable in the cells than the other RNA species. Moreover, there is increasing literature which clearly suggests that a single specific LNC-RNA can regulate multiple mechanisms and disease phenotypes. With specific focus on cardiovascular diseases, here we attempt to provide UpToDate information on the functional role of miRNAs and LNC-RNAs. Here we discuss the role of these epigenetic mediators in different components of cardiovascular disease which include physiopathological heart development, athersclerosis, retenosis, diabetic hearts, myocardial infarction, ischemia-reperfusion, heart valve disease, aortic aneurysm, osteogenesis, angiogenesis and hypoxia in the heart. While there is abundant literature support that shows the involvement of many LNC-RNAs and miRNAs in cardiovascular diseases, very few RNA species have been identified which regulate epigenetic mechanisms which is the current focus in this article. Understanding the role of these RNA species in regulating epigenetic mechanisms in different cell types causing cardiovascular disease, would advance the field and promote disease prevention approaches that are aimed to target epigenetic mechanisms.
Collapse
Affiliation(s)
- Wanlin Jiang
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Devendra K Agrawal
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Chandra Shekhar Boosani
- Department of Clinical & Translational Research, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
21
|
|
22
|
Xiong W, Qu Y, Chen H, Qian J. Insight into long noncoding RNA-miRNA-mRNA axes in myocardial ischemia-reperfusion injury: the implications for mechanism and therapy. Epigenomics 2019; 11:1733-1748. [PMID: 31701757 DOI: 10.2217/epi-2019-0119] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Emerging evidence has demonstrated that regulatory noncoding RNAs (ncRNAs), such as long noncoding RNAs (lncRNAs) and miRNAs, play crucial roles in the initiation and progress of myocardial ischemia-reperfusion injury (MIRI), which is associated with autophagy, apoptosis and necrosis of cardiomyocytes, as well as oxidative stress, inflammation and mitochondrial dysfunction. LncRNAs serve as a precursor or host of miRNAs and directly/indirectly affecting miRNAs via competitive binding or sponge effects. Simultaneously, miRNAs post-transcriptionally regulate the expression of genes by targeting various mRNA sequences due to their imperfect pairing with mRNAs. This review summarizes the potential regulatory role of lncRNA-miRNA-mRNA axes in MIRI and related molecular mechanisms of cardiac disorders, also provides insight into the potential therapies for MIRI-induced diseases.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan province 650032, PR China
| | - Yan Qu
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan province 650032, PR China.,Department of Anesthesiology, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan province 650021, PR China
| | - Hongmei Chen
- Department of Anesthesiology, Kunming Angel Women's & Children's Hospital, Kunming, Yunnan province 650108, PR China
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan province 650032, PR China
| |
Collapse
|
23
|
Shu L, Zhang W, Huang C, Huang G, Su G, Xu J. lncRNA ANRIL protects H9c2 cells against hypoxia-induced injury through targeting the miR-7-5p/SIRT1 axis. J Cell Physiol 2019; 235:1175-1183. [PMID: 31264206 DOI: 10.1002/jcp.29031] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/29/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND Acute myocardial infarction (AMI) occurred in the heart, which underwent long-term ischemia, and was mainly caused by hypoxia. Recently, studies have uncovered the participation of long noncoding RNAs (lncRNAs) in the pathogenesis of heart disease. Here, we planned to probe the role and molecular basis of ANRIL in hypoxia-induced H9c2 cell injury. METHODS Trypan blue exclusion assay and Transwell and flow cytometry assays were conducted to assess hypoxia-induced injury by determining the viability, migration, invasion, and apoptosis of H9c2 cells in different conditions, respectively. Gene expressions were evaluated by quantitative real-time polymerase chain reaction or western blot analysis as needed. RNA immunoprecipitation and luciferase reporter assays were applied to confirm the associations among genes. RESULTS ANRIL expression was dramatically enhanced in hypoxia-injured H9c2 cells, and silencing ANRIL aggravated hypoxia-induced H9c2 cell injury. ANRIL positively regulated sirtuin 1 (SIRT1) expression via competitively binding with miR-7-5p. Moreover, inhibition of miR-7-5p counteracted ANRIL depletion-exacerbated injury in hypoxic H9c2 cells, meanwhile, forced SIRT1 expression attenuated the injury-promoting effect of miR-7-5p upregulation on hypoxic H9c2 cells. CONCLUSION Our findings disclosed that ANRIL plays a protective part in hypoxia-induced H9c2 cell injury via modulating the miR-7-5p/SIRT1 axis, suggesting the great potential of ANRIL as a protective target for AMI.
Collapse
Affiliation(s)
- Liliang Shu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wanzhe Zhang
- Department of Nephrology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gongcheng Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gang Su
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
Li X, Yu M, Han L, Chen L, Zhang D, Zhou G, Zhao Q, Sun T. LINC00305 represses miR-124 expression to trigger inflammatory insults in the presence of lipopolysaccharide. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2352-2360. [PMID: 31190561 DOI: 10.1080/21691401.2019.1624559] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Xiaoyan Li
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Manqiu Yu
- Department of Trauma Reconstruction, Shandong Wendeng Orthopaedic & Traumatic Hospital Weihai, China
| | - Liang Han
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Lei Chen
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Daxue Zhang
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, China
| | - Guannan Zhou
- Emergency Department, Jining No. 1 People’s Hospital, Jining, China
| | - Qiang Zhao
- Department of Orthopaedics, Jining No.1 People’s Hospital, Jining, China
| | - Taitao Sun
- Department of Orthopaedics, Jining No.1 People’s Hospital, Jining, China
- Department of Orthopaedics, Affiliated Jining No.1 People's Hospital of Jining Medical University, Jining Medical University, Jining, China
| |
Collapse
|
25
|
Circulating Noncoding RNAs Have a Promising Future Acting as Novel Biomarkers for Colorectal Cancer. DISEASE MARKERS 2019; 2019:2587109. [PMID: 31275444 PMCID: PMC6589288 DOI: 10.1155/2019/2587109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/23/2019] [Accepted: 05/21/2019] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors worldwide, causing a large number of cancer-related deaths each year. Patients are usually diagnosed at advanced and incurable stages due to the lack of suitable screening methods for early detection. Noncoding RNAs (ncRNAs), including small and long noncoding RNAs (lncRNA), are known to have significant regulatory functions, and accumulating evidence suggests that circulating ncRNAs have potential applications as noninvasive biomarkers for diagnosing CRC, evaluating its prognosis, or predicting chemosensitivity in the general population. In this review, we summarize the origins of circulating ncRNAs and provide details of single and multiple circulating ncRNAs that might have roles as diagnostic and prognostic biomarkers in CRC. We end by discussing circulating ncRNAs that may distinguish patients with resistance to chemotherapy.
Collapse
|
26
|
Li M, Duan L, Li Y, Liu B. Long noncoding RNA/circular noncoding RNA-miRNA-mRNA axes in cardiovascular diseases. Life Sci 2019; 233:116440. [PMID: 31047893 DOI: 10.1016/j.lfs.2019.04.066] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/23/2019] [Accepted: 04/29/2019] [Indexed: 02/01/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Non-coding RNAs including long non-coding RNAs (lncRNAs), circular RNAs (circRNAs) and microRNAs (miRNAs) have been reported to participate in pathological developments of CVDs through various mechanisms. Among them, the networks among lncRNAs/circRNAs, miRNAs, and mRNAs have recently attracted attention. Understanding the molecular mechanism could aid the discovery of therapeutic targets or strategies in CVDs including atherosclerosis, myocardial infarction (MI), hypertrophy, heart failure (HF) and cardiomyopathy. In this review, we summarize the latest research involving the lncRNA/circRNA-miRNA-mRNA axis in CVDs, with emphasis on the molecular mechanism.
Collapse
Affiliation(s)
- Ming Li
- Department of Gastroenterology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China
| | - Liwei Duan
- Department of Gastroenterology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China
| | - Yangxue Li
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China.
| |
Collapse
|
27
|
Sun Y, Xiong X, Wang X. RELA promotes hypoxia-induced angiogenesis in human umbilical vascular endothelial cells via LINC01693/miR-302d/CXCL12 axis. J Cell Biochem 2019; 120:12549-12558. [PMID: 30937967 DOI: 10.1002/jcb.28521] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/02/2019] [Accepted: 01/07/2019] [Indexed: 12/17/2022]
Abstract
Hypoxia-induced angiogenesis plays a critical role in wound healing, which could be disturbed by multifactors. Upon hypoxia stimulation, CXCL12 and its receptor CXCR4 were significantly upregulated in human umbilical vascular endothelial cells (HUVECs); thus, we attempted to investigate the role and mechanism of CXCL12 in HUVEC angiogenesis under hypoxia. Via downloading and analyzing microarray profiles (GSE76743 and GSE116909), we found that LINC01693 was positively correlated with CXCL12 and upregulated by hypoxia in HUVECs, while miR-302d was downregulated by hypoxia and might target LINC01693 and CXCL12. RELA, a critical transcriptional factor for response to hypoxia, could bind to LINC01693 promoter to activate its transcription, therefore, promoting CXCL12 expression under hypoxia. LINC01693 served as a competing endogenous RNA for miR-302d to counteract miR-302d-mediated CXCL12 suppression via direct targeting. Hypoxia-induced CXCL12 upregulation and angiogenesis in HUVECs could be significantly suppressed by LINC01693 silence while enhanced by miR-302d inhibition; the effect of LINC01693 silence could be partially reversed by miR-302d inhibition. Taken together, RELA promotes the angiogenesis in HUVECs via LINC01693/miR-302d/CXCL12 axis. We provide a novel mechanism and experimental basis of CXCL12 function in hypoxia-induced HUVEC angiogenesis.
Collapse
Affiliation(s)
- Yang Sun
- Department of Plastic Surgery and Burns Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Xiong
- Department of Plastic Surgery and Burns Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiancheng Wang
- Department of Plastic Surgery and Burns Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
28
|
Xu S, Kamato D, Little PJ, Nakagawa S, Pelisek J, Jin ZG. Targeting epigenetics and non-coding RNAs in atherosclerosis: from mechanisms to therapeutics. Pharmacol Ther 2019; 196:15-43. [PMID: 30439455 PMCID: PMC6450782 DOI: 10.1016/j.pharmthera.2018.11.003] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, the principal cause of cardiovascular death worldwide, is a pathological disease characterized by fibro-proliferation, chronic inflammation, lipid accumulation, and immune disorder in the vessel wall. As the atheromatous plaques develop into advanced stage, the vulnerable plaques are prone to rupture, which causes acute cardiovascular events, including ischemic stroke and myocardial infarction. Emerging evidence has suggested that atherosclerosis is also an epigenetic disease with the interplay of multiple epigenetic mechanisms. The epigenetic basis of atherosclerosis has transformed our knowledge of epigenetics from an important biological phenomenon to a burgeoning field in cardiovascular research. Here, we provide a systematic and up-to-date overview of the current knowledge of three distinct but interrelated epigenetic processes (including DNA methylation, histone methylation/acetylation, and non-coding RNAs), in atherosclerotic plaque development and instability. Mechanistic and conceptual advances in understanding the biological roles of various epigenetic modifiers in regulating gene expression and functions of endothelial cells (vascular homeostasis, leukocyte adhesion, endothelial-mesenchymal transition, angiogenesis, and mechanotransduction), smooth muscle cells (proliferation, migration, inflammation, hypertrophy, and phenotypic switch), and macrophages (differentiation, inflammation, foam cell formation, and polarization) are discussed. The inherently dynamic nature and reversibility of epigenetic regulation, enables the possibility of epigenetic therapy by targeting epigenetic "writers", "readers", and "erasers". Several Food Drug Administration-approved small-molecule epigenetic drugs show promise in pre-clinical studies for the treatment of atherosclerosis. Finally, we discuss potential therapeutic implications and challenges for future research involving cardiovascular epigenetics, with an aim to provide a translational perspective for identifying novel biomarkers of atherosclerosis, and transforming precision cardiovascular research and disease therapy in modern era of epigenetics.
Collapse
Affiliation(s)
- Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Wooloongabba, QLD 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou 510520, 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
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo Nishi 6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Jaroslav Pelisek
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technischen Universitaet Muenchen, Germany
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
29
|
Castellanos-Rubio A, Ghosh S. Disease-Associated SNPs in Inflammation-Related lncRNAs. Front Immunol 2019; 10:420. [PMID: 30906297 PMCID: PMC6418042 DOI: 10.3389/fimmu.2019.00420] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/18/2019] [Indexed: 02/06/2023] Open
Abstract
Immune-mediated diseases, such as celiac disease, type 1 diabetes or multiple sclerosis, are a clinically heterogeneous group of diseases that share many key genetic triggers. Although the pathogenic mechanisms responsible for the development of immune mediated disorders is not totally understood, high-throughput genomic studies, such as GWAS and Immunochip, performed in the past few years have provided intriguing hints about underlying mechanisms and pathways that lead to disease. More than a hundred gene variants associated with disease susceptibility have been identified through such studies, but the progress toward understanding the underlying mechanisms has been slow. The majority of the identified risk variants are located in non-coding regions of the genome making it difficult to assign a molecular function to the SNPs. However, recent studies have revealed that many of the non-coding regions bearing disease-associated SNPs generate long non-coding RNAs (lncRNAs). LncRNAs have been implicated in several inflammatory diseases, and many of them have been shown to function as regulators of gene expression. Many of the disease associated SNPs located in lncRNAs modify their secondary structure, or influence expression levels, thereby affecting their regulatory function, hence contributing to the development of disease.
Collapse
Affiliation(s)
- Ainara Castellanos-Rubio
- Immunogenetics Research Laboratory, Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, Leioa, Spain
- Functional Studies in Celiac Disease Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Sankar Ghosh
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
30
|
Zhao H, Zhao Y, Li X, Xu L, Jiang F, Hou W, Dong L, Cao J. Effects of Antioxidant Tempol on Systematic Inflammation and Endothelial Apoptosis in Emphysematous Rats Exposed to Intermittent Hypoxia. Yonsei Med J 2018; 59:1079-1087. [PMID: 30328323 PMCID: PMC6192890 DOI: 10.3349/ymj.2018.59.9.1079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Obstructive sleep apnea and chronic obstructive pulmonary disease are independent risk factors of cardiovascular disease (CVD), and their coexistence is known as overlap syndrome (OS). Endothelial dysfunction is the initial stage of CVD; however, underlying mechanisms linking OS and CVD are not well understood. The aim of this study was to explore whether OS can lead to more severe inflammation and endothelial apoptosis by promoting endothelial dysfunction, and to assess the intervention effects of antioxidant tempol. MATERIALS AND METHODS Male Wistar rats (n=66) were exposed to normal oxygen [normal control (NC) group], intermittent hypoxia (IH group), cigarette smoke (CH group), as well as cigarette smoke and IH (OS group). Tempol intervention was assessed in OS group treated with tempol (OST group) or NaCl (OSN group). After an 8-week challenge, lung tissues, serum, and fresh blood were harvested for analysis of endothelial markers and apoptosis. RESULTS The levels of intracellular adhesion molecule-1, vascular cellular adhesion molecule-1, and apoptosis in circulating epithelial cells were the highest in OS group and the lowest in NC group. These levels were all greater in IH group than in CH group, and were lower in OST group than in OS and OSN groups (all p<0.001). CONCLUSION Synergistic effects of IH with cigarette smoke-induced emphysema produce a greater inflammatory status and endothelial apoptosis. OS-related inflammation and endothelial cell apoptosis may play important roles in promoting cardiovascular dysfunction, and antioxidant tempol could achieve a partial protective effect.
Collapse
Affiliation(s)
- Haiyan Zhao
- Respiratory Department of Tianjin Medical University General Hospital, Tianjin, China
| | - Yaping Zhao
- Respiratory Department of Tianjin Medical University General Hospital Airport Hospital, Tianjin, China
| | - Xin Li
- Respiratory Department of Tianjin Medical University General Hospital, Tianjin, China
| | - Leiqian Xu
- Respiratory Department of Tianjin Medical University General Hospital, Tianjin, China
| | - Fangxin Jiang
- Department of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Wanju Hou
- Respiratory Department of Tianjin Medical University General Hospital, Tianjin, China
| | - Lixia Dong
- Respiratory Department of Tianjin Medical University General Hospital, Tianjin, China.
| | - Jie Cao
- Respiratory Department of Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
31
|
Yuan Q, Zhang Y, Feng L, Jiang Y. Upregulated long noncoding RNA LINC01296 indicates a dismal prognosis for pancreatic ductal adenocarcinoma and promotes cell metastatic properties by affecting EMT. J Cell Biochem 2018; 120:552-561. [PMID: 30203487 DOI: 10.1002/jcb.27411] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/10/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a fatal disease that responds poorly to chemotherapy and radiotherapy and whose incidence has increased worldwide. Long noncoding RNAs have been demonstrated to play important roles in cancer initiation and progression. Long intergenic non-coding RNA 01296 (LINC01296) has been reported to be upregulated in several malignancies, but the clinical relevance and biological role of LINC01296 in PDAC are still unclear. METHODS RT-qPCR was performed to evaluate the expression of LINC01296 in 85 pared PDAC tissue samples and a panel of PDAC cell lines. The clinical value and prognostic role of LINC01296 in patients with PDAC were further explored. Furthermore, we explored the functional roles of LINC01296 depletion in PANC-1 and SW1990 cells, including cell proliferation, apoptosis, migration, invasion, and epithelial-to-mesenchymal transition (EMT). RESULTS LINC01296 was enhanced in PDAC tissues and cell lines, and this overexpression was correlated with advanced tumor stages and positive lymph node metastasis in patients with PDAC. In addition, upregulation of LINC01296 was an independent prognostic predictor for patients with PDAC after surgery. Moreover, silencing of LINC01296 followed by treatment with small interfering RNAs suppressed cell proliferation and promoted cell apoptosis by affecting the Bcl-2/caspase-3 pathway. Importantly, LINC01296 attenuation impaired the migratory and invasive potential partly by reversing EMT. CONCLUSIONS Overall, our work may help to develop a novel prognostic biomarker and therapeutic target for PDAC.
Collapse
Affiliation(s)
- Qihua Yuan
- Department of Gastrointestinal Surgery, Yidu Central Hospital of Weifang, Qingzhou, Shandong Province, China
| | - Yuxia Zhang
- Department of Gastroenterology, Yidu Central Hospital of Weifang, Qingzhou, Shandong Province, China
| | - Lijun Feng
- Department of Pharmacy Intravenous Admixture Center, Yidu Central Hospital of Weifang, Qingzhou, Shandong Province, China
| | - Yuehong Jiang
- Department of Clinical Laboratory, Heilongjiang Provincial Hospital, Harbin, Heilongjiang Province, China
| |
Collapse
|
32
|
Mou K, Liu B, Ding M, Mu X, Han D, Zhou Y, Wang LJ. lncRNA-ATB functions as a competing endogenous RNA to promote YAP1 by sponging miR-590-5p in malignant melanoma. Int J Oncol 2018; 53:1094-1104. [PMID: 29956757 PMCID: PMC6065447 DOI: 10.3892/ijo.2018.4454] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/31/2018] [Indexed: 12/23/2022] Open
Abstract
The critical long non‑coding RNAs (lncRNAs) involved in the carcinogenesis and progression of malignant melanoma (MM) have not been fully investigated. In the present study, it was identified that lncRNA activated by transforming growth factor‑β (lncRNA‑ATB) was upregulated in MM tissues and cells compared with benign nevus cells and human melanocytes, via comparative lncRNA screening from Gene Expression Omnibus datasets and reverse transcription‑quantitative polymerase chain reaction analysis. Furthermore, lncRNA‑ATB promoted the cell proliferation, cell migration, and cell invasion of MM cells in vitro, and tumor growth in vivo. It was additionally identified that lncRNA‑ATB attenuated cell cycle arrest and inhibited cellular apoptosis in MM cells. Finally, it was demonstrated that lncRNA‑ATB functions as a competing endogenous RNA (ceRNA) to enhance Yes associated protein 1 expression by competitively sponging microRNA miR‑590‑5p in MM cells. In conclusion, the present study revealed the expression and roles of lncRNA‑ATB in MM, and indicated that lncRNA‑ATB functions as a ceRNA to promote MM proliferation and invasion by sponging miR‑590‑5p.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Apoptosis/genetics
- Carcinogenesis/genetics
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation/genetics
- Disease Progression
- Female
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Humans
- Melanoma/genetics
- Melanoma/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- MicroRNAs/metabolism
- Neoplasm Invasiveness/genetics
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Small Interfering/metabolism
- Skin Neoplasms/genetics
- Skin Neoplasms/pathology
- Transcription Factors
- Xenograft Model Antitumor Assays
- YAP-Signaling Proteins
- Melanoma, Cutaneous Malignant
Collapse
Affiliation(s)
- Kuanhou Mou
- Department of Dermatology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| | - Bo Liu
- Department of Dermatology, Yan'an Traditional Chinese Medicine Hospital, Yan'an, Shaanxi 716000
| | - Meiling Ding
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Disease, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xin Mu
- Department of Dermatology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| | - Dan Han
- Department of Dermatology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| | - Yan Zhou
- Department of Dermatology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| | - Li-Juan Wang
- Department of Dermatology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| |
Collapse
|
33
|
Xu Y, Shao B. Circulating lncRNA IFNG-AS1 expression correlates with increased disease risk, higher disease severity and elevated inflammation in patients with coronary artery disease. J Clin Lab Anal 2018; 32:e22452. [PMID: 29744951 PMCID: PMC6817098 DOI: 10.1002/jcla.22452] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/23/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND This study aimed to investigate the associations of circulating long, non-coding (lncRNA) IFNG-AS1, lncRNA ANRIL and lncRNA ITSN1 relative expressions with disease risk, severity and inflammatory cytokines levels in coronary artery disease (CAD) patients. METHODS One hundred and ninety-one patients suspected of CAD who underwent coronary angiography were consecutively enrolled in this casecontrol study, and divided into CAD patients (N = 102) and controls (N = 89) according to coronary angiographic results. Blood samples of all participants were collected. Plasma lncRNA IFNG-AS1, lncRNA ANRIL and lncRNA ITSN1 expressions were detected using quantitative polymerase chain reaction (qPCR). Serum tumor necrosis factor-α (TNF-α), interleukin (IL)-1β (IL-1β), IL-6, IL-8, IL-10, and IL-17 were assessed using enzyme-linked immunosorbent assay (ELISA). Gensini Score was used to evaluate the disease severity of CAD patients. RESULTS LncRNA IFNG-AS1 relative expression in CAD patients was upregulated compared with that in controls (P < .001), and the receiver operating characteristic (ROC) curve showed that the area under curve (AUC) of lncRNA-IFNG-AS1 for predicting the risk of CAD was 0.755 (95% CI: 0.688-0.821). lncRNA IFNG-AS1 relative expression was remarkably associated with Gensini Score (r = .259, P = .009). Additionally, lncRNA IFNG-AS1 relative expression was positively associated with high-sensitivity C-reactive protein (hs-CRP) (r = .283, P = .004), TNF-α (r = .269, P = .006), and IL-6 levels (r = .425, P < .001), while it was negatively correlated with IL-10 level (r = -.263, P = .008). lncRNA ANRIL or lncRNA ITSN1 was not correlated with CA D risk, Gensini Score, hs-CRP, ESR, TNF-α, IL-1β, IL-6, IL-8, IL-10, or IL-17 levels (all P > .05). CONCLUSION Circulating lncRNA IFNG-AS1 expression correlates with increased disease risk, higher disease severity and elevated inflammation in CAD patients.
Collapse
Affiliation(s)
- Yahuan Xu
- Department of Cardiothoracic SurgeryHuangshi Central HospitalEdong Healthcare GroupAffiliated Hospital of Hubei Polytechnic UniversityHuangshiChina
| | - Bibo Shao
- Department of Intensive Care UnitHuangshi Central HospitalEdong Healthcare GroupAffiliated Hospital of Hubei Polytechnic UniversityHuangshiChina
| |
Collapse
|
34
|
Li J, Li L, Li X, Wu S. Long noncoding RNA LINC00339 aggravates doxorubicin-induced cardiomyocyte apoptosis by targeting MiR-484. Biochem Biophys Res Commun 2018; 503:3038-3043. [DOI: 10.1016/j.bbrc.2018.08.090] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/12/2018] [Indexed: 11/29/2022]
|
35
|
Halimulati M, Duman B, Nijiati J, Aizezi A. Long noncoding RNA TCONS_00024652 regulates vascular endothelial cell proliferation and angiogenesis via microRNA-21. Exp Ther Med 2018; 16:3309-3316. [PMID: 30233677 PMCID: PMC6143905 DOI: 10.3892/etm.2018.6594] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/19/2018] [Indexed: 01/02/2023] Open
Abstract
Acute coronary syndrome caused by the rupture of atherosclerotic plaques is one of the primary causes of major cardiovascular events, and neovascularization within the plaque is closely associated with its stability. Long noncoding RNA (lncRNAs) is a type of noncoding RNA that serves a crucial role in regulating vascular endothelial cells (VECs). The aim of the present study was to investigate the effect of lncRNA TCONS_00024652 on the proliferation and angiogenesis of VECs following stimulation with TNF-α. The expression of lncRNA and miRNA was measured in human umbilical vein endothelial cells (HUVECs) by reverse transcription-quantitative polymerase chain reaction. Cell proliferation was measured using a Cell Counting Kit-8 assay. Wound healing and tube formation assays were performed to determine cell migration and angiogenesis. Interactions between TCONS_00024652 and miR-21 were determined using bioinformatics and a dual-luciferase reporter assay. The results demonstrated that TCONS_00024652 is highly expressed in TNF-α-induced HUVECs. Functional assays demonstrated that the dysregulated expression of TCONS_00024652 promotes endothelial cell proliferation and angiogenesis, whereas TCONS_00024652 knockdown induces the opposite effects. Bioinformatics analysis using starBase predicted putative binding at the 3′-untranslated region of TCONS_00024652 and miR-21 and luciferase reporter assays further verified this interaction. The results of the present study suggest that the targeting of TCONS_00024652 by miR-21 may be a potential method of improving vascular endothelial dysfunction, neovascularization maturation and plaque stabilization.
Collapse
Affiliation(s)
- Muertiza Halimulati
- Department of Vascular and Thyroid Surgery, Center of Digestive and Vascular Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Bagedati Duman
- Department of Vascular and Thyroid Surgery, Center of Digestive and Vascular Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Julaiti Nijiati
- Department of Vascular and Thyroid Surgery, Center of Digestive and Vascular Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Abudoureyimu Aizezi
- Department of Vascular and Thyroid Surgery, Center of Digestive and Vascular Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| |
Collapse
|
36
|
Weirick T, Militello G, Uchida S. Long Non-coding RNAs in Endothelial Biology. Front Physiol 2018; 9:522. [PMID: 29867565 PMCID: PMC5960726 DOI: 10.3389/fphys.2018.00522] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/24/2018] [Indexed: 01/08/2023] Open
Abstract
In recent years, the role of RNA has expanded to the extent that protein-coding RNAs are now the minority with a variety of non-coding RNAs (ncRNAs) now comprising the majority of RNAs in higher organisms. A major contributor to this shift in understanding is RNA sequencing (RNA-seq), which allows a largely unconstrained method for monitoring the status of RNA from whole organisms down to a single cell. This observational power presents both challenges and new opportunities, which require specialized bioinformatics tools to extract knowledge from the data and the ability to reuse data for multiple studies. In this review, we summarize the current status of long non-coding RNA (lncRNA) research in endothelial biology. Then, we will cover computational methods for identifying, annotating, and characterizing lncRNAs in the heart, especially endothelial cells.
Collapse
Affiliation(s)
- Tyler Weirick
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, United States
| | - Giuseppe Militello
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, United States
| | - Shizuka Uchida
- Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, United States
| |
Collapse
|
37
|
He L, Chen Y, Hao S, Qian J. Uncovering novel landscape of cardiovascular diseases and therapeutic targets for cardioprotection via long noncoding RNA–miRNA–mRNA axes. Epigenomics 2018; 10:661-671. [PMID: 29692219 DOI: 10.2217/epi-2017-0176] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Protein coding sequences account for around 3% of the human genome, the rest are noncoding RNA (ncRNA) including long ncRNA (lncRNA) and miRNA. Accumulating evidence indicates that lncRNAs and miRNAs are candidate biomarkers for diagnosis, prognosis and therapy of cardiovascular diseases. The lncRNAs act as sponge-like effects on numerous miRNAs, subsequently regulating miRNAs and their targets, mRNA functions. The role of lncRNA–miRNA–mRNA axis in pathogenesis of cardiovascular diseases has been recently reported and highlighted. Herein, this review discusses emerging roles of lncRNA–miRNA–mRNA axis in cardiovascular pathophysiology and regulation, with a novel focus on cardioprotective network activities of the two subgroup ncRNAs.
Collapse
Affiliation(s)
- Liang He
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, PR China
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming Medical University, Kunming, Yunnan Province 650051, PR China
| | - Yan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, PR China
| | - Shuqing Hao
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, PR China
| | - Jinqiao Qian
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, PR China
| |
Collapse
|
38
|
Simion V, Haemmig S, Feinberg MW. LncRNAs in vascular biology and disease. Vascul Pharmacol 2018; 114:145-156. [PMID: 29425892 DOI: 10.1016/j.vph.2018.01.003] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 12/14/2022]
Abstract
Accumulating studies indicate that long non-coding RNAs (lncRNAs) play important roles in the regulation of diverse biological processes involved in homeostatic control of the vessel wall in health and disease. However, our knowledge of the mechanisms by which lncRNAs control gene expression and cell signaling pathways is still nascent. Furthermore, only a handful of lncRNAs has been functionally evaluated in response to pathophysiological stimuli or in vascular disease states. For example, lncRNAs may regulate endothelial dysfunction by modulating endothelial cell proliferation (e.g. MALAT1, H19) or angiogenesis (e.g. MEG3, MANTIS). LncRNAs have also been implicated in modulating vascular smooth muscle cell (VSMC) phenotypes or vascular remodeling (e.g. ANRIL, SMILR, SENCR, MYOSLID). Finally, emerging studies have implicated lncRNAs in leukocytes activation (e.g. lincRNA-Cox2, linc00305, THRIL), macrophage polarization (e.g. GAS5), and cholesterol metabolism (e.g. LeXis). This review summarizes recent findings on the expression, mechanism, and function of lncRNAs implicated in a range of vascular disease states from mice to human subjects. An improved understanding of lncRNAs in vascular disease may provide new pathophysiological insights and opportunities for the generation of a new class of RNA-based biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Viorel Simion
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefan Haemmig
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark W Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Pomatto MAC, Gai C, Deregibus MC, Tetta C, Camussi G. Noncoding RNAs Carried by Extracellular Vesicles in Endocrine Diseases. Int J Endocrinol 2018; 2018:4302096. [PMID: 29808089 PMCID: PMC5902008 DOI: 10.1155/2018/4302096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/16/2018] [Indexed: 02/06/2023] Open
Abstract
RNA molecules are essential and fine regulators of important biological processes. Their role is well documented also in the endocrine system, both in physiological and pathological conditions. Increasing interest is arising about the function and the importance of noncoding RNAs shuttled by extracellular vesicles (EVs). In fact, EV membrane protects nucleic acids from enzyme degradation. Nowadays, the research on EVs and their cargoes, as well as their biological functions, faces the lack of standardization in EV purification. Here, the main techniques for EV isolation are discussed and compared for their advantages and vulnerabilities. Despite the possible discrepancy due to methodological variability, EVs and their RNA content are reported to be key mediators of intercellular communication in pathologies of main endocrine organs, including the pancreas, thyroid, and reproductive system. In particular, the present work describes the role of RNAs contained in EVs in pathogenesis and progression of several metabolic dysfunctions, including obesity and diabetes, and their related manifestations. Their importance in the establishment and progression of thyroid autoimmunity disorders and complicated pregnancy is also discussed. Preliminary studies highlight the attractive possibility to use RNAs contained in EVs as biomarkers suggesting their exploitation for new diagnostic approaches in endocrinology.
Collapse
Affiliation(s)
| | - Chiara Gai
- Stem Cell Laboratory, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Maria Chiara Deregibus
- Stem Cell Laboratory, Department of Medical Sciences, University of Turin, Turin, Italy
- 2i3T Scarl, Univerity of Turin, Turin, Italy
| | - Ciro Tetta
- Unicyte AG, Oberdorf, Nidwalden, Switzerland
| | - Giovanni Camussi
- Stem Cell Laboratory, Department of Medical Sciences, University of Turin, Turin, Italy
- 2i3T Scarl, Univerity of Turin, Turin, Italy
| |
Collapse
|