1
|
Huang Y, Zhan W, Song C, Tan M, Wu L, Wu S. The lncRNA SENCR knockdown alleviates vascular calcification via miR-4731-5p by suppressing endoplasmic reticulum stress. PLoS One 2025; 20:e0323058. [PMID: 40354352 PMCID: PMC12068569 DOI: 10.1371/journal.pone.0323058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/01/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Accumulation of calcium phosphate crystals is associated with vascular calcification (VC); however, the mechanism that promotes VC remains unclear. Accumulating evidence indicates that smooth muscle and endothelial cell-enriched migration/differentiation-associated lncRNA (SENCR) exerts a critical role in VC. This work focuses on the molecules involved in β-glycerophosphate-induced osteogenic differentiation of vascular smooth muscle cells (VSMCs) through SENCR epigenetic modification of Runx2 in an endoplasmic reticulum stress (ERS)-dependent manner. METHODS We cultured VSMCs to explore the relationship among β-glycerophosphate, SENCR, and VC and also investigate the function of SENCR in β-glycerophosphate-induced osteogenic differentiation and VC in vitro. RESULTS Our findings indicate that β-glycerophosphate enhanced SENCR, MSH homeobox 2, Runx2, ERS-related markers, alkaline phosphatase activity, and cellular calcium deposition and suppressed the expression of α-SMA, SM 22α, and miR-4731-5p. SENCR silencing increased miR-4731-5p expression, which subsequently inhibited β-glycerophosphate-associated endoplasmic reticulum stress at the post-transcriptional level. Critically, the facts that direct interplay between SENCR and miR-4731-5p, and the downregulation of miR-4731-5p efficiently reversed the suppression of ERS-induced by SENCR silencing were observed. Collectively, the present study clarifies a novel mechanism by which downregulation of SRNRC contributes to the ERS-dependent osteogenic differentiation of VSMCs and VC by sponging miR-4731-5p. This study demonstrates that SENCR/miR-4731-5p axis is involved in β-glycerophosphate-mediated VC in vitro.
Collapse
MESH Headings
- Endoplasmic Reticulum Stress/genetics
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/pathology
- Glycerophosphates/pharmacology
- Core Binding Factor Alpha 1 Subunit/genetics
- Core Binding Factor Alpha 1 Subunit/metabolism
- Animals
- Cell Differentiation/genetics
- Cell Differentiation/drug effects
- Osteogenesis/genetics
- Osteogenesis/drug effects
- Myocytes, Smooth Muscle/metabolism
- Cells, Cultured
- Rats
- Gene Knockdown Techniques
Collapse
Affiliation(s)
- Yongpan Huang
- School of Medicine, Changsha Social Work College, Changsha, Hunan, China
| | - Wei Zhan
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Chong Song
- School of Medicine, Changsha Social Work College, Changsha, Hunan, China
| | - Meihua Tan
- Hunan Carnation Endowment Industry Investment Real Estate Co., Ltd, Changsha, China
| | - Li Wu
- Hunan Carnation Endowment Industry Investment Real Estate Co., Ltd, Changsha, China
| | - Sina Wu
- Department of Respiratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
2
|
Kong X, Li F, Wang Y. Emerging Roles of Long Non-Coding RNAs in Cardiovascular Diseases. J Cell Mol Med 2025; 29:e70453. [PMID: 40032652 PMCID: PMC11875779 DOI: 10.1111/jcmm.70453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/26/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Over the past decade, studies have demonstrated that circulating long non-coding RNAs (lncRNAs)-recognised for their stability and ease of detection-serve as crucial regulators and potential biomarkers in multiple diseases. LncRNAs regulate key processes, including endothelial function, vascular remodelling, and myocardial hypertrophy, all of which influence CVD progression. Additionally, lncRNAs display cell-, tissue-, and disease-specific expression patterns, making them ideal therapeutic targets or tools. This review presents a comprehensive overview of the current understanding of lncRNAs in CVDs, examining their mechanisms of action and recent research advances. It also addresses the use of lncRNAs as diagnostic and prognostic markers, as well as potential applications of RNA therapeutics in novel treatment strategies.
Collapse
Affiliation(s)
- Xiangyue Kong
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling‐Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Beijing Institute of Heart, Lung, and Blood Vessel DiseasesBeijingChina
| | - Fengjuan Li
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling‐Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Beijing Institute of Heart, Lung, and Blood Vessel DiseasesBeijingChina
| | - Yuan Wang
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling‐Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
- Beijing Institute of Heart, Lung, and Blood Vessel DiseasesBeijingChina
| |
Collapse
|
3
|
Brown SD, Klimi E, Bakker WAM, Beqqali A, Baker AH. Non-coding RNAs to treat vascular smooth muscle cell dysfunction. Br J Pharmacol 2025; 182:246-280. [PMID: 38773733 DOI: 10.1111/bph.16409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/19/2024] [Accepted: 03/14/2024] [Indexed: 05/24/2024] Open
Abstract
Vascular smooth muscle cell (vSMC) dysfunction is a critical contributor to cardiovascular diseases, including atherosclerosis, restenosis and vein graft failure. Recent advances have unveiled a fascinating range of non-coding RNAs (ncRNAs) that play a pivotal role in regulating vSMC function. This review aims to provide an in-depth analysis of the mechanisms underlying vSMC dysfunction and the therapeutic potential of various ncRNAs in mitigating this dysfunction, either preventing or reversing it. We explore the intricate interplay of microRNAs, long-non-coding RNAs and circular RNAs, shedding light on their roles in regulating key signalling pathways associated with vSMC dysfunction. We also discuss the prospects and challenges associated with developing ncRNA-based therapies for this prevalent type of cardiovascular pathology. LINKED ARTICLES: This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
MESH Headings
- Animals
- Humans
- Cardiovascular Diseases/drug therapy
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- RNA, Circular/genetics
- RNA, Circular/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- RNA, Untranslated/pharmacology
- RNA, Untranslated/therapeutic use
Collapse
Affiliation(s)
- Simon D Brown
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Eftychia Klimi
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Abdelaziz Beqqali
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Andrew H Baker
- BHF Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
4
|
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
|
5
|
Tao Y, Li G, Yang Y, Wang Z, Wang S, Li X, Yu T, Fu X. Epigenomics in aortic dissection: From mechanism to therapeutics. Life Sci 2023; 335:122249. [PMID: 37940070 DOI: 10.1016/j.lfs.2023.122249] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Aortic dissection (AD) has an unfavorable prognosis. It requires early diagnosis, appropriate treatment strategies, and suspicion to recognize symptoms; thus, it is commonly described as an acute aortic emergency. The clinical manifestations of painless AD are complex and variable. However, there is no effective treatment to prevent the progression of AD. Therefore, study of the molecular targets and mechanisms of AD to enable prevention or early intervention is particularly important. Although multiple gene mutations have been proposed as linked to AD development, evidence that multiple epigenetic elements are strongly associated is steadily increasing. These epigenetic processes include DNA methylation, N6-methyladenosine, histone modification, non-histone posttranslational modification, and non-coding RNAs (ncRNAs). Among these processes, resveratrol targeting Sirtuin 1 (SIRT1), 5-azacytidine (5azaC) targeting DNA methyltransferase (DNMT), and vitamin C targeting ten-eleven translocation 2 (Tet2) showed unique advantages in improving AD and vascular dysfunction. Finally, we explored potential epigenetic drugs and diagnostic methods for AD, which might provide options for the future.
Collapse
Affiliation(s)
- Yan Tao
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Gang Li
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, Shandong 250021, China; Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, People's Republic of China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Shizhong Wang
- The department of Cardiology surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China.
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China.
| |
Collapse
|
6
|
Zhang J, Han L, Liu H, Zhang H, An Z. Metabolomic analysis reveals the metabolic disturbance in aortic dissection: Subtype difference and accurate diagnosis. Nutr Metab Cardiovasc Dis 2023; 33:1556-1564. [PMID: 37263915 DOI: 10.1016/j.numecd.2023.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 04/12/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND AND AIMS Aortic dissection (AD), a severe clinical emergency with high mortality, is easily misdiagnosed as are other cardiovascular diseases. This study aimed at discovering plasma metabolic markers with the potential to diagnose AD and clarifying the metabolic differences between two subtypes of AD. METHODS AND RESULTS To facilitate the diagnosis of AD, we investigated the plasma metabolic profile by metabolomic approach. A total 482 human subjects were enrolled in the study: 80 patients with AD (50 with Stanford type A and 30 with Stanford type B), 198 coronary artery disease (CAD) patients, and 204 healthy individuals. Plasma samples were submitted to targeted metabolomic analysis. The partial least-squares discriminant analysis models were constructed to illustrate clear discrimination of AD patients with CAD patients and healthy control. Subsequently, the metabolites that were clinically relevant to the disturbances in AD were identified. Twenty metabolites induced the separation of AD patients and healthy control, 9 of which caused the separation of CAD patients and healthy control. There are 11 metabolites specifically down-regulated in AD group. Subgroup analysis showed that the levels of glycerol and uridine were dramatically lower in the plasma of patients with Stanford type A AD than those in the healthy control or Stanford type B AD groups. CONCLUSION This study characterized metabolomic profiles specifically associated with the pathogenesis and development of AD. The findings of this research may potentially lead to earlier diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Jinghui Zhang
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100029, China
| | - Lu Han
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100029, China; Beijing Lab for Cardiovascular Precision Medicine, Beijing, 100069, China; Key Laboratory of Medical Engineering for Cardiovascular Disease, Beijing, 100069, China
| | - Hongchuan Liu
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100029, China
| | - Hongjia Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Lab for Cardiovascular Precision Medicine, Beijing, 100069, China; Key Laboratory of Medical Engineering for Cardiovascular Disease, Beijing, 100069, China.
| | - Zhuoling An
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
7
|
Wang G, Luo Y, Gao X, Liang Y, Yang F, Wu J, Fang D, Luo M. MicroRNA regulation of phenotypic transformations in vascular smooth muscle: relevance to vascular remodeling. Cell Mol Life Sci 2023; 80:144. [PMID: 37165163 PMCID: PMC11071847 DOI: 10.1007/s00018-023-04793-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/10/2023] [Accepted: 04/27/2023] [Indexed: 05/12/2023]
Abstract
Alterations in the vascular smooth muscle cells (VSMC) phenotype play a critical role in the pathogenesis of several cardiovascular diseases, including hypertension, atherosclerosis, and restenosis after angioplasty. MicroRNAs (miRNAs) are a class of endogenous noncoding RNAs (approximately 19-25 nucleotides in length) that function as regulators in various physiological and pathophysiological events. Recent studies have suggested that aberrant miRNAs' expression might underlie VSMC phenotypic transformation, appearing to regulate the phenotypic transformations of VSMCs by targeting specific genes that either participate in the maintenance of the contractile phenotype or contribute to the transformation to alternate phenotypes, and affecting atherosclerosis, hypertension, and coronary artery disease by altering VSMC proliferation, migration, differentiation, inflammation, calcification, oxidative stress, and apoptosis, suggesting an important regulatory role in vascular remodeling for maintaining vascular homeostasis. This review outlines recent progress in the discovery of miRNAs and elucidation of their mechanisms of action and functions in VSMC phenotypic regulation. Importantly, as the literature supports roles for miRNAs in modulating vascular remodeling and for maintaining vascular homeostasis, this area of research will likely provide new insights into clinical diagnosis and prognosis and ultimately facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Gang Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yulin Luo
- GCP Center, Affiliated Hospital (Traditional Chinese Medicine) of Southwest Medical University, Luzhou, China
| | - Xiaojun Gao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu Liang
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Feifei Yang
- School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Jianbo Wu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Dan Fang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China.
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Longmatan District, No. 1, Section 1, Xianglin Road, Luzhou, Sichuan, China.
- Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
- Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
8
|
Han C, Qi Y, She Y, Zhang M, Xie H, Zhang J, Zhao Z, Peng C, Liu Y, Lin Y, Wang J, Zeng D. Long noncoding RNA SENCR facilitates the progression of acute myeloid leukemia through the miR-4731-5p/IRF2 pathway. Pathol Res Pract 2023; 245:154483. [PMID: 37120908 DOI: 10.1016/j.prp.2023.154483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a type of hematological tumor caused by malignant clone hematopoietic stem cells. The relationship between lncRNAs and tumor occurrence and progression has been gaining attention. Research has shown that Smooth muscle and endothelial cell-enriched migration/differentiation-associated lncRNA (SENCR) is abnormally expressed in various diseases, whereas its role in AML is still poorly understood. METHODS The expression of SENCR, microRNA-4731-5p (miR-4731-5p) and Interferon regulatory factor 2 (IRF2) were measured using qRT-PCR. The proliferation, cycle and apoptosis of AML cells with or without knockdown of SENCR were detected by CCK-8 assay, EdU assay, flow cytometry, western blotting and TUNEL assay, respectively. Consistently, SENCR knockdown was impaired the AML progression in immunodeficient mice. In addition, the binding of miR-4731-5p to SENCR or IRF2 was confirmed by luciferase reporter genes assay. Finally, rescue experiments were conducted to confirm the role of SENCR/miR-4731-5p/IRF2 axis in AML. RESULTS SENCR is highly expressed in AML patients and cell lines. The patients with high SENCR expression had poorer prognosis compared with those with low SENCR expression. Interestingly, knockdown of SENCR inhibits the growth of AML cells. Further results demonstrated that the reduction of SENCR slows the progression of AML in vivo. SENCR could function as a competing endogenous RNA (ceRNA) to negatively regulate miR-4731-5p in AML cells. Furthermore, IRF2 was validated as a direct target gene of miR-4731-5p in AML cells. CONCLUSIONS Our findings underscore the important role of SENCR in regulating the malignant phenotype of AML cells by targeting the miR-4731-5p/IRF2 axis.
Collapse
Affiliation(s)
- Changhao Han
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Yan Qi
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Yuanting She
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Meijuan Zhang
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Huan Xie
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Jing Zhang
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Zhongyue Zhao
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Cuicui Peng
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Yu Liu
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Yizhang Lin
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Jin Wang
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China
| | - Dongfeng Zeng
- Department of Hematology, Daping Hospital, Third Military Medical University, No. 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, China.
| |
Collapse
|
9
|
Xun M, Zhang J, Wu M, Chen Y. Long non-coding RNAs: The growth controller of vascular smooth muscle cells in cardiovascular diseases. Int J Biochem Cell Biol 2023; 157:106392. [PMID: 36828237 DOI: 10.1016/j.biocel.2023.106392] [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: 10/05/2022] [Revised: 02/13/2023] [Accepted: 02/19/2023] [Indexed: 02/25/2023]
Abstract
The active proliferation and migration of vascular smooth muscle cells supports the healing of vessel damage while their abnormal aggression or destitution contribute to the aberrant intima-medial structure and function in various cardiovascular diseases, so the understanding of the proliferation disorders of vascular smooth muscle cell and the related mechanism is the basis of effective intervention and control for cardiovascular diseases. Recently, long non-coding RNAs have stood out as upstream switchers for multiple proliferative signaling pathways and molecules, and many of them have been shown to conduce to the dysregulated proliferation and apoptosis of vascular smooth muscle cells under various pathogenic stimuli. This article discusses the long non-coding RNAs disclosed and linked to atherosclerosis, pulmonary hypertension, and aneurysms, and focuses upon their modulation of vascular smooth muscle cell population affecting three deadly cardiovascular diseases.
Collapse
Affiliation(s)
- Min Xun
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Jie Zhang
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Meichun Wu
- Hengyang Medical School, University of South China, Hengyang 421001, China; School of Nursing, University of South China, Hengyang 421001, China
| | - Yuping Chen
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China; Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
10
|
Chen H, Yu Y, Zhou L, Chen J, Li Z, Tan X. Cuproptosis-related LncRNAs signature as biomarker of prognosis and immune infiltration in pancreatic cancer. Front Genet 2023; 14:1049454. [PMID: 36713077 PMCID: PMC9880288 DOI: 10.3389/fgene.2023.1049454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/02/2023] [Indexed: 01/14/2023] Open
Abstract
Background: Pancreatic cancer (PC) is a malignant gastrointestinal tumor with a terrible prognosis. Cuproptosis is a recently discovered form of cell death. This study is intended to explore the relationship between cuproptosis-related lncRNAs (CRLncs) signature with the prognosis and the tumor microenvironment (TME) of PC. Methods: Transcript sequencing data of PC samples with clinical information were obtained from the Cancer Genome Atlas (TCGA). Univariate Cox regression analysis and LASSO regression analysis were employed to construct the prognostic signature based on CRLncs associated with PC survival. A nomogram was created according to this signature, and the signaling pathway enrichment was analyzed. Subsequently, we explored the link between this prognostic signature with the mutational landscape and TME. Eventually, drug sensitivity was predicted based on this signature. Results: Forty-six of 159 CRLncs were most significantly relevant to the prognosis of PC, and a 6-lncRNA prognostic signature was established. The expression level of signature lncRNAs were detected in PC cell lines. The AUC value of the ROC curve for this risk score predicting 5-year survival in PC was .944, which was an independent prognostic factor for PC. The risk score was tightly related to the mutational pattern of PC, especially the driver genes of PC. Single-sample gene set enrichment analysis (ssGSEA) demonstrated a significant correlation between signature with the TME of PC. Ultimately, compounds were measured for therapy in high-risk and low-risk PC patients, respectively. Conclusion: A prognostic signature of CRLncs for PC was established in the current study, which may serve as a promising marker for the outcomes of PC patients and has important forecasting roles for gene mutations, immune cell infiltration, and drug sensitivity in PC.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaodong Tan
- Department of General Surgery, Pancreatic, and Thyroid Ward, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
11
|
Li Y, Ma X, Mei S, Ji Y, Wang D, He L, Sun D, Yan J. mRNA, lncRNA, and circRNA expression profiles in a new aortic dissection murine model induced by hypoxia and Ang II. Front Cardiovasc Med 2022; 9:984087. [PMID: 36386298 PMCID: PMC9643159 DOI: 10.3389/fcvm.2022.984087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Background and aims Aortic dissection (AD) is a cardiovascular emergency with degeneration of the aortic media. Mounting evidence indicates obstructive sleep apnea (OSA) as an independent risk factor for AD development with unknown mechanisms. This study aims to establish a stable murine model of OSA-related AD (OSA-AD) and uncover the potential changes in gene transcripts in OSA-AD. Materials and methods ApoE–/– mice were exposed to the chronic intermittent hypoxia (CIH) system combined with Ang II administration to establish the OSA-AD model. Pathological staining was performed to exhibit the physiological structure of the mouse aorta. The SBC mouse ceRNA microarray was used to identify significantly differentially expressed (DE) mRNAs, DE long-non-coding RNAs (DElncRNAs), and DE circular RNAs (DEcircRNAs) in OSA-AD tissues. Subsequently, bioinformatics analysis, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genome (KEGG), and protein–protein interaction (PPI) analyses, were performed to evaluate the function of the significantly differentially expressed transcripts (DETs). The hub genes were confirmed using quantitative real-time polymerase chain reaction (qRT-PCR). Results ApoE–/– mice exposed to CIH and Ang II showed a high ratio of aortic accident (73.33%) and significant aortic diameter dilatation (1.96 ± 0.175 mm). A total of 1,742 mRNAs, 2,625 lncRNAs, and 537 circRNAs were identified as DETs (LogFC ≥ 1.5 or ≤ –1.5, P < 0.05). GO and KEGG analyses demonstrated that the differentially expressed mRNAs (DEmRNAs) were most enriched in cell proliferation, migration, apoptosis, inflammation, and hypoxia-related terms, which are closely related to aortic structural homeostasis. The PPI network contained 609 nodes and 934 connections, the hub genes were highlighted with the CytoHubba plugin and confirmed by qRT-PCR in AD tissues. KEGG pathway analysis revealed that the cis-regulated genes of DElncRNAs and circRNAs-host genes were enriched in aortic structural homeostasis-related pathways. Conclusion Our findings help establish a de novo OSA-AD animal model using ApoE–/– mice. Many DEmRNAs, DElncRNAs, and DEcircRNAs were screened for the first time in OSA-AD tissues. Our findings provide useful bioinformatics data for understanding the molecular mechanism of OSA-AD and developing potential therapeutic strategies for OSA-AD.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhu Ma
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Mei
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yueping Ji
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | - Dong Wang
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | - Liqun He
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
- Liqun He,
| | - Dating Sun
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, China
- Dating Sun,
| | - Jiangtao Yan
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jiangtao Yan,
| |
Collapse
|
12
|
Emerging Role of Non-Coding RNAs in Aortic Dissection. Biomolecules 2022; 12:biom12101336. [PMID: 36291545 PMCID: PMC9599213 DOI: 10.3390/biom12101336] [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: 09/01/2022] [Revised: 09/12/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
Aortic dissection (AD) is a fatal cardiovascular acute disease with high incidence and mortality, and it seriously threatens patients’ lives and health. The pathogenesis of AD mainly includes vascular inflammation, extracellular matrix degradation, and phenotypic conversion as well as apoptosis of vascular smooth muscle cells (VSMCs); however, its detailed mechanisms are still not fully elucidated. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), are an emerging class of RNA molecules without protein-coding ability, and they play crucial roles in the progression of many diseases, including AD. A growing number of studies have shown that the dysregulation of ncRNAs contributes to the occurrence and development of AD by modulating the expression of specific target genes or the activity of related proteins. In addition, some ncRNAs exhibit great potential as promising biomarkers and therapeutic targets in AD treatment. In this review, we systematically summarize the recent findings on the underlying mechanism of ncRNA involved in AD regulation and highlight their clinical application as biomarkers and therapeutic targets in AD treatment. The information reviewed here will be of great benefit to the development of ncRNA-based therapeutic strategies for AD patients.
Collapse
|