1
|
Zhang Y, Zhao H, Tian L, Yang Z, Zheng L, Zhang H, Zhu Y, Ma Y, Xu Y, Liu Y. SPARC: a key mediator of apoptosis in human umbilical vein endothelial cells and its role in hypertension mechanism. In Vitro Cell Dev Biol Anim 2025; 61:374-388. [PMID: 40229525 DOI: 10.1007/s11626-025-01026-1] [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: 12/13/2024] [Accepted: 01/19/2025] [Indexed: 04/16/2025]
Abstract
Hypertensionis a leading global health issue associated with high mortality and severe complications. Understanding its molecular mechanisms is essential for identifying novel therapeutic targets. Secreted protein acidic and rich in cysteine (SPARC) is associated with cell migration, disease pathophysiology, and inflammation; however, its role in hypertension remains under investigation. This study investigates the role of SPARC in hypertension, focusing on its impact on endothelial dysfunction.Using the GSE75815 dataset from the GEO database, we identified 71 differentially expressed genes (DEGs) associated with hypertension. Pathway analyses and protein-protein interaction networks constructed through the STRING database highlighted six hub genes, with further evaluation based on Comparative Toxicogenomics Database (CTD) scores. Immune cell profiling via ImmuCellAI revealed an increase in naive B cells, positively correlating with hub gene expression.Experimental validation in human umbilical vein endothelial cells (HUVECs) treated with angiotensin II demonstrated that SPARC downregulation reduced apoptosis and BAX expression. Silencing SPARC enhanced endothelial cell proliferation, migration, and nitric oxide production, counteracting angiotensin II-induced damage. Notably, angiotensin II upregulated SPARC secretion, suggesting its critical role in mediating endothelial dysfunction.These findings establish SPARC as a key contributor to the molecular pathways underlying hypertension. Targeting SPARC may represent a novel therapeutic strategy to mitigate endothelial dysfunction and improve outcomes for hypertensive patients.Our findings highlight SPARC as a key player in the molecular pathways of hypertension. Modulating SPARC expression may offer a promising therapeutic strategy to counteract endothelial dysfunction and improve outcomes in hypertensive patients.
Collapse
Affiliation(s)
- Yingyue Zhang
- Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, P.R. China
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Haijing Zhao
- Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, P.R. China
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Liuyang Tian
- Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, P.R. China
- Beijing an Zhen Hospital, Affiliated of Capital University of Medical Sciences, Beijing, 100029, P.R. China
| | - Zengao Yang
- Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, P.R. China
- School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Li Zheng
- Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, P.R. China
- School of Medicine, Nankai University, Tianjin, 300071, P.R. China
| | - Honghong Zhang
- Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, P.R. China
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Yue Zhu
- Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, P.R. China
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Yuhan Ma
- Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, P.R. China
- School of Medicine, Nankai University, Tianjin, 300071, P.R. China
| | - Yong Xu
- Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, P.R. China.
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, P.R. China.
| | - Yuqi Liu
- Department of Cardiology, The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, P.R. China.
- National Key Laboratory of Kidney Diseases, Beijing, 100853, P.R. China.
- Department of Cardiology & National Clinical Research Center of Geriatric Disease, Beijing, 100853, P.R. China.
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Beijing, 100853, P.R. China.
| |
Collapse
|
2
|
Zhang X, Yao S, Wang Y, Chu C, Du M, Mu J. Associations of microRNA Gene Polymorphisms With Salt Sensitivity, Longitudinal Blood Pressure Changes, and Hypertension Incidence in the Chinese Population. J Clin Hypertens (Greenwich) 2025; 27:e70019. [PMID: 39994918 PMCID: PMC11850435 DOI: 10.1111/jch.70019] [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: 11/08/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025]
Abstract
MicroRNAs (miRNAs) are small endogenous RNA molecules that play an essential role in various disease processes including elevated blood pressure (BP). Although the effects of dietary salt and potassium intake on BP regulation have been established, their co-interaction with miRNAs are still unclear. The purpose of the current study was to explore the connection between miRNA gene polymorphisms and BP response to salt and potassium intake, and the relationship between miRNA gene polymorphisms and long-term BP changes and hypertension development. A total of 333 participants underwent a chronic sodium-potassium dietary intervention trial, which included a 3-day normal diet, followed by a 7-day low-salt diet, then a 7-day high-salt diet, and finally a 7-day high-salt with potassium-supplemented diet. This cohort was subsequently followed for up to 14 years. Single-nucleotide polymorphisms (SNPs) rs115254818 in miR-26b-3p, rs11191676 and rs2292807 in miR-1307-5p, and rs4143957 in miR-382-5p were significantly correlated with systolic BP (SBP) and mean arterial pressure (MAP) responses to high-salt intake, whereas rs11191676 and rs2292807 in miR-1307-5p exhibited significant associations with SBP response to potassium-supplemented diet. Furthermore, SNPs rs2070960 in miR-3620-5p and rs12364149 in miR-210-3p demonstrated significant correlations with diastolic BP and MAP alterations at 14 years of follow-up. Generalized linear mixed model analysis revealed a significant association between rs2070960 in miR-3620-5p and hypertension development over a 14-year period. Our study indicates that miRNA gene polymorphisms are pivotal in the salt and potassium sensitivity of BP, as well as in the longitudinal BP progression and hypertension incidence. Trial Registration: ClinicalTrials.gov identifier: NCT02734472.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Shi Yao
- Guangdong Key Laboratory of Age‐Related Cardiac and Cerebral DiseasesAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdongChina
| | - Yang Wang
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Chao Chu
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Mingfei Du
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| | - Jianjun Mu
- Department of Cardiovascular MedicineFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Key Laboratory of Molecular Cardiology of Shaanxi ProvinceXi'anChina
| |
Collapse
|
3
|
Xu ZJ, Shen RM, Hu WM, Shen JY, Wu XY, Lv LC. Association between "Life's Essential 8" cardiovascular health and apparent treatment-resistant hypertension among US adults from the NHANES, 2005 to 2018. Front Cardiovasc Med 2024; 11:1453563. [PMID: 39735865 PMCID: PMC11671479 DOI: 10.3389/fcvm.2024.1453563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/02/2024] [Indexed: 12/31/2024] Open
Abstract
Background The association between healthy lifestyle and American Heart Association (AHA) Life's Essential 8 (LE8) score and apparent treatment-resistant hypertension(aTRH)remains uncertain. We aimed to explore the association between healthy lifestyle and higher LE8 score and apparent treatment-resistant hypertension in the general population. Methods Using NHANES data from 2005 to 2018, we included and analyzed information on 7,474 participants eligible for this study. The association between LE8 and aTRH was explored using logistic regression models, and the association between LE8 and antihypertension drugs uncontrolled hypertension was further explored using logistic regression models. Results Participants with higher LE8 scores tended to be non-Hispanic white and married or living with a partner; have low income and higher education; and be without Chronic kidney disease (CKD)(all p-values <0.001). Compared to subjects with low CVH, participants with moderate and high CVH exhibited lower risks of 47% and 76%, respectively. After adjusting for covariates, there was no evidence of a nonlinear association between LE8 and aTRH (p for nonlinearity = 0.456). Physical activity (PA), body mass index (BMI), and blood glucose were associated with aTRH (all p-values < 0.05), while diet, nicotine exposure, sleep, and blood lipids were not significantly associated with aTRH. Compared to the low LE8 group, the ORs for the high LE8 group were 0.46 (95% CI, 0.28 to 0.76) and 0.07 (95% CI, 0.02 to 0.20) for uncontrolled hypertension with 1-2 and 3-4 antihypertensive drugs, respectively. In the sensitivity analysis, subgroup analyses were performed on all covariates, and the results remained stable. Conclusion In our study, we found a significant association between higher LE8 scores and a lower risk of aTRH. Our findings suggest that implementing various healthy lifestyle practices and managing known cardiovascular risk factors could be a feasible comprehensive preventive approach to aTRH.
Collapse
Affiliation(s)
| | | | | | | | | | - Ling-chun Lv
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Zhejiang, China
| |
Collapse
|
4
|
Yaacoub S, Boudaka A, AlKhatib A, Pintus G, Sahebkar A, Kobeissy F, Eid AH. The pharmaco-epigenetics of hypertension: a focus on microRNA. Mol Cell Biochem 2024; 479:3255-3271. [PMID: 38424404 PMCID: PMC11511726 DOI: 10.1007/s11010-024-04947-9] [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: 11/26/2023] [Accepted: 01/20/2024] [Indexed: 03/02/2024]
Abstract
Hypertension is a major harbinger of cardiovascular morbidity and mortality. It predisposes to higher rates of myocardial infarction, chronic kidney failure, stroke, and heart failure than most other risk factors. By 2025, the prevalence of hypertension is projected to reach 1.5 billion people. The pathophysiology of this disease is multifaceted, as it involves nitric oxide and endothelin dysregulation, reactive oxygen species, vascular smooth muscle proliferation, and vessel wall calcification, among others. With the advent of new biomolecular techniques, various studies have elucidated a gaping hole in the etiology and mechanisms of hypertension. Indeed, epigenetics, DNA methylation, histone modification, and microRNA-mediated translational silencing appear to play crucial roles in altering the molecular phenotype into a hypertensive profile. Here, we critically review the experimentally determined associations between microRNA (miRNA) molecules and hypertension pharmacotherapy. Particular attention is given to the epigenetic mechanisms underlying the physiological responses to antihypertensive drugs like candesartan, and other relevant drugs like clopidogrel, aspirin, and statins among others. Furthermore, how miRNA affects the pharmaco-epigenetics of hypertension is especially highlighted.
Collapse
Affiliation(s)
- Serge Yaacoub
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ammar Boudaka
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ali AlKhatib
- Department of Nutrition and Food Sciences, Lebanese International University, Beirut, Lebanon
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100, Sassari, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics and Biomarkers (CNMB), Morehouse School of Medicine, Neuroscience Institute, Atlanta, GA, USA
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
5
|
Huang ZM, Kang JQ, Chen PZ, Deng LF, Li JX, He YX, Liang J, Huang N, Luo TY, Lan QW, Chen HK, Guo XG. Identifying the Interaction Between Tuberculosis and SARS-CoV-2 Infections via Bioinformatics Analysis and Machine Learning. Biochem Genet 2024; 62:2606-2630. [PMID: 37991568 DOI: 10.1007/s10528-023-10563-x] [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: 08/08/2023] [Accepted: 10/25/2023] [Indexed: 11/23/2023]
Abstract
The number of patients with COVID-19 caused by severe acute respiratory syndrome coronavirus 2 is still increasing. In the case of COVID-19 and tuberculosis (TB), the presence of one disease affects the infectious status of the other. Meanwhile, coinfection may result in complications that make treatment more difficult. However, the molecular mechanisms underpinning the interaction between TB and COVID-19 are unclear. Accordingly, transcriptome analysis was used to detect the shared pathways and molecular biomarkers in TB and COVID-19, allowing us to determine the complex relationship between COVID-19 and TB. Two RNA-seq datasets (GSE114192 and GSE163151) from the Gene Expression Omnibus were used to find concerted differentially expressed genes (DEGs) between TB and COVID-19 to identify the common pathogenic mechanisms. A total of 124 common DEGs were detected and used to find shared pathways and drug targets. Several enterprising bioinformatics tools were applied to perform pathway analysis, enrichment analysis and networks analysis. Protein-protein interaction analysis and machine learning was used to identify hub genes (GAS6, OAS3 and PDCD1LG2) and datasets GSE171110, GSE54992 and GSE79362 were used for verification. The mechanism of protein-drug interactions may have reference value in the treatment of coinfection of COVID-19 and TB.
Collapse
Affiliation(s)
- Ze-Min Huang
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Jia-Qi Kang
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The First Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Pei-Zhen Chen
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Lin-Fen Deng
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Jia-Xin Li
- Department of Clinical Medicine, The First Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Ying-Xin He
- Clinical Laboratory Medicine, Guangzhou Medical University, Guangzhou, 510006, China
| | - Jie Liang
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Nan Huang
- Clinical Laboratory Medicine, Guangzhou Medical University, Guangzhou, 510006, China
| | - Tian-Ye Luo
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Qi-Wen Lan
- Department of Clinical Medicine, The Second Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Hao-Kai Chen
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Xu-Guang Guo
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 510000, China.
| |
Collapse
|
6
|
Yang Q, Murata K, Ikeda T, Minatoya K, Masumoto H. miR-124-3p downregulates EGR1 to suppress ischemia-hypoxia reperfusion injury in human iPS cell-derived cardiomyocytes. Sci Rep 2024; 14:14811. [PMID: 38926457 PMCID: PMC11208498 DOI: 10.1038/s41598-024-65373-x] [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: 02/04/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024] Open
Abstract
Ischemic heart diseases are a major global cause of death, and despite timely revascularization, heart failure due to ischemia-hypoxia reperfusion (IH/R) injury remains a concern. The study focused on the role of Early Growth Response 1 (EGR1) in IH/R-induced apoptosis in human cardiomyocytes (CMs). Human induced pluripotent stem cell (hiPSC)-derived CMs were cultured under IH/R conditions, revealing higher EGR1 expression in the IH/R group through quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting (WB). Immunofluorescence analysis (IFA) showed an increased ratio of cleaved Caspase-3-positive apoptotic cells in the IH/R group. Using siRNA for EGR1 successfully downregulated EGR1, suppressing cleaved Caspase-3-positive apoptotic cell ratio. Bioinformatic analysis indicated that EGR1 is a plausible target of miR-124-3p under IH/R conditions. The miR-124-3p mimic, predicted to antagonize EGR1 mRNA, downregulated EGR1 under IH/R conditions in qRT-PCR and WB, as confirmed by IFA. The suppression of EGR1 by the miR-124-3p mimic subsequently reduced CM apoptosis. The study suggests that treatment with miR-124-3p targeting EGR1 could be a potential novel therapeutic approach for cardioprotection in ischemic heart diseases in the future.
Collapse
Affiliation(s)
- Qiaoke Yang
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kozue Murata
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Tadashi Ikeda
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hidetoshi Masumoto
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
7
|
Xu D, Gareev I, Beylerli O, Pavlov V, Le H, Shi H. Integrative bioinformatics analysis of miRNA and mRNA expression profiles and identification of associated miRNA-mRNA network in intracranial aneurysms. Noncoding RNA Res 2024; 9:471-485. [PMID: 38511055 PMCID: PMC10950608 DOI: 10.1016/j.ncrna.2024.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 03/22/2024] Open
Abstract
Background Intracranial aneurysms (IAs) represent protrusions in the vascular wall, with their growth and wall thinning influenced by various factors. These processes can culminate in the rupture of the aneurysm, leading to subarachnoid hemorrhage (SAH). Unfortunately, over half of the patients prove unable to withstand SAH, succumbing to adverse outcomes despite intensive therapeutic interventions, even in premier medical facilities. This study seeks to discern the pivotal microRNAs (miRNAs) and genes associated with the formation and progression of IAs. Methods The investigation gathered expression data of miRNAs (from GSE66240) and mRNAs (from GSE158558) within human aneurysm tissue and superficial temporal artery (STA) samples, categorizing them into IA and normal groups. This classification was based on the Gene Expression Omnibus (GEO) database. Results A total of 70 differentially expressed microRNAs (DEMs) and 815 differentially expressed mRNAs (DEGs) were pinpointed concerning IA. Subsequently, a miRNA-mRNA network was constructed, incorporating 9 significantly upregulated DEMs and 211 significantly downregulated DEGs. Simultaneously, functional enrichment and pathway analyses were conducted on both DEMs and DEGs. Through protein-protein interaction (PPI) network analysis and functional enrichment, 9 significantly upregulated DEMs (hsa-miR-188-5p, hsa-miR-590-5p, hsa-miR-320b, hsa-miR-423-5p, hsa-miR-140-5p, hsa-miR-486-5p, hsa-miR-320a, hsa-miR-342-3p, and hsa-miR-532-5p) and 50 key genes (such as ATP6V1G1, KBTBD6, VIM, PA2G4, DYNLL1, METTL21A, MDH2, etc.) were identified, suggesting their potential significant role in IA. Among these genes, ten were notably negatively regulated by at least two key miRNAs. Conclusions The findings of this study provide valuable insights into the potential pathogenic mechanisms underlying IA by elucidating a miRNA-mRNA network. This comprehensive approach sheds light on the intricate interplay between miRNAs and genes, offering a deeper understanding of the molecular dynamics involved in IA development and progression.
Collapse
Affiliation(s)
- Dongxiao Xu
- Department of Neurosurgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Ilgiz Gareev
- Central Research Laboratory, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 3 Lenin street, 450008, Russia
| | - Ozal Beylerli
- Central Research Laboratory, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 3 Lenin street, 450008, Russia
| | - Valentin Pavlov
- Department of Urology, Bashkir State Medical University, 3 Lenin Street, 450008, Ufa, Russia
| | - Huang Le
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Huaizhang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| |
Collapse
|
8
|
Lin Y, He J, Mou Z, Chen H, You W, Guan T, Chen L. Ferroptosis-related genes, a novel therapeutic target for focal segmental glomerulosclerosis. BMC Nephrol 2024; 25:58. [PMID: 38368317 PMCID: PMC10874534 DOI: 10.1186/s12882-024-03490-5] [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: 07/06/2023] [Accepted: 02/01/2024] [Indexed: 02/19/2024] Open
Abstract
Recent studies have suggested that ferroptosis participates in various renal diseases. However, its effect on focal segmental glomerulosclerosis remains unclear. This study analyzed the GSE125779 and GSE121211 datasets to identify the differentially expressed genes (DEGs) in renal tubular samples with and without FSGS. The Cytoscape was used to construct the protein-protein interaction network. Moreover, the ferroptosis-related genes (FRGs) were obtained from the ferroptosis database, while ferroptosis-related DEGs were obtained by intersection with DEGs. The target genes were analyzed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The GSE108112 dataset was used to verify the expression of target FRGs. Besides, we built the mRNA-miRNA network regarding FRGs using the NetworkAnalyst database, and circRNAs corresponding to key miRNAs were predicted in the ENCORI database. In this study, 16 ferroptosis-related DEGs were identified between FSGS and healthy subjects, while five co-expressed genes were obtained by three topological algorithms in Cytoscape. These included the most concerned Hub genes JUN, HIF1A, ALB, DUSP1 and ATF3. The KEGG enrichment analysis indicated that FRGs were associated with mitophagy, renal cell carcinoma, and metabolic pathways. Simultaneously, the co-expressed hub genes were analyzed to construct the mRNA-miRNA interaction network and important miRNAs such as hsa-mir-155-5p, hsa-mir-1-3p, and hsa-mir-124-3p were obtained. Finally, 75 drugs targeting 54 important circRNAs and FRGs were predicted. This study identified the Hub FRGs and transcriptomic molecules from FSGS in renal tubules, thus providing novel diagnostic and therapeutic targets for FSGS.
Collapse
Affiliation(s)
- Yanbin Lin
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jinxuan He
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhixiang Mou
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | | | | | - Tianjun Guan
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lan Chen
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- Fujian Medical University, Fuzhou, China.
- Xiamen Municipal Health Commission, Xiamen, China.
| |
Collapse
|
9
|
Zheng T, Wu Y, Guo KX, Tan ZJ, Yang T. The process of hypertension induced by high-salt diet: Association with interactions between intestinal mucosal microbiota, and chronic low-grade inflammation, end-organ damage. Front Microbiol 2023; 14:1123843. [PMID: 36925479 PMCID: PMC10011071 DOI: 10.3389/fmicb.2023.1123843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/27/2023] [Indexed: 03/06/2023] Open
Abstract
Inflammation and immunity play a major role in the development of hypertension, and a potential correlation between host mucosal immunity and inflammatory response regulation. We explored the changes of intestinal mucosal microbiota in hypertensive rats induced by high-salt diet and the potential link between the intestinal mucosal microbiota and inflammation in rats. Therefore, we used PacBio (Pacific Bioscience) SMRT sequencing technology to determine the structure of intestinal mucosal microbiota, used enzyme-linked immunosorbent assay (ELISA) to determined the proinflammatory cytokines and hormones associated with hypertension in serum, and used histopathology methods to observe the kidney and vascular structure. We performed a potential association analysis between intestinal mucosal characteristic bacteria and significantly different blood cytokines in hypertensive rats induced by high-salt. The results showed that the kidney and vascular structures of hypertensive rats induced by high salt were damaged, the serum concentration of necrosis factor-α (TNF-α), angiotensin II (AngII), interleukin-6 (IL-6), and interleukin-8 (IL-8) were significantly increased (p < 0.05), and the coefficient of immune organ spleen was significantly changed (p < 0.05), but there was no significant change in serum lipids (p > 0.05). From the perspective of gut microbiota, high-salt diet leads to significant changes in intestinal mucosal microbiota. Bifidobacterium animalis subsp. and Brachybacterium paraconglomeratum were the dominant differential bacteria in intestinal mucosal, with the AUC (area under curve) value of Bifidobacterium animalis subsp. and Brachybacterium paraconglomeratum were 1 and 0.875 according to ROC (receiver operating characteristic) analysis. Correlation analysis showed that Bifidobacterium animalis subsp. was correlated with IL-6, IL-8, TNF-α, and Ang II. Based on our results, we can speculated that high salt diet mediated chronic low-grade inflammation through inhibited the growth of Bifidobacterium animalis subsp. in intestinal mucosa and caused end-organ damage, which leads to hypertension.
Collapse
Affiliation(s)
- Tao Zheng
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yi Wu
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Kang-xiao Guo
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Zhou-jin Tan
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Tao Yang
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| |
Collapse
|
10
|
Ahmadi Badi S, Malek A, Paolini A, Rouhollahi Masoumi M, Seyedi SA, Amanzadeh A, Masotti A, Khatami S, Siadat SD. Downregulation of ACE, AGTR1, and ACE2 genes mediating SARS-CoV-2 pathogenesis by gut microbiota members and their postbiotics on Caco-2 cells. Microb Pathog 2022; 173:105798. [PMID: 36174833 PMCID: PMC9511898 DOI: 10.1016/j.micpath.2022.105798] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Coronavirus disease-2019 (COVID-19) is a complex infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that can cause also gastrointestinal symptoms. There are various factors that determine the host susceptibility and severity of infection, including the renin-angiotensin system, the immune response, and the gut microbiota. In this regard, we aimed to investigate the gene expression of ACE, AGTR1, ACE2, and TMPRSS2, which mediate SARS-CoV-2 pathogenesis by Akkermansia muciniphila, Faecalibacterium prausnitzii, Bacteroides thetaiotaomicron, and Bacteroides fragilis on Caco-2 cells. Also, the enrichment analysis considering the studied genes was analyzed on raw data from the microarray analysis of COVID-19 patients. MATERIALS AND METHODS Caco-2 cells were treated with live, heat-inactivated form and cell free supernatants of A. muciniphila, F. prausnitzii, B. thetaiotaomicron and B. fragilis for overnight. After RNA extraction and cDNA synthesis, the expression of studied genes was assessed by RT-qPCR. DNA methylation of studied genes was analyzed by Partek® Genomics Suite® software on the GSE174818 dataset. We used GSE164805 and GSE166552 datasets from COVID-19 patients to perform enrichment analysis by considering the mentioned genes via GEO2R, DAVID. Finally, the related microRNAs to GO terms concerned on the studied genes were identified by miRPath. RESULTS The downregulation of ACE, AGTR1, and ACE2 genes by A. muciniphila, F. prausnitzii, B. thetaiotaomicron, and B. fragilis in live, heat-inactivated, and cell-free supernatants was reported for the first time. These genes had hypomethylated DNA status in COVID-19 patients' raw data. The highest fold enrichment in upregulated RAS pathways and immune responses belonged to ACE, AGTR1, and ACE2 by considering the protein-protein interaction network. The common miRNAs targeting the studied genes were reported as miR-124-3p and miR-26b-5p. In combination with our experimental data and bioinformatic analysis, we showed the potential of A. muciniphila, F. prausnitzii, B. thetaiotaomicron, and B. fragilis and postbiotics to reduce ACE, ATR1, and ACE2 expression, which are essential genes that drive upregulated biological processes in COVID-19 patients. CONCLUSION Accordingly, due to the potential of studied bacteria on the alteration of ACE, AGTR1, ACE2 genes expression, understanding their correlation with demonstrated miRNAs expression could be valuable. These findings suggest the importance of considering targeted gut microbiota intervention when designing the possible therapeutic strategy for controlling the COVID-19.
Collapse
Affiliation(s)
- Sara Ahmadi Badi
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran; Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran.
| | - Amin Malek
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran; Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran.
| | - Alessandro Paolini
- Children's Hospital Bambino Gesù-IRCCS, Research Laboratories, V.le di San Paolo 15, 00146, Rome, Italy.
| | - Mahya Rouhollahi Masoumi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Seyed Amirhesam Seyedi
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran; Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran.
| | - Amir Amanzadeh
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran.
| | - Andrea Masotti
- Children's Hospital Bambino Gesù-IRCCS, Research Laboratories, V.le di San Paolo 15, 00146, Rome, Italy.
| | - Shohreh Khatami
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran.
| | - Seyed Davar Siadat
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran; Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
11
|
Anik MI, Mahmud N, Masud AA, Khan MI, Islam MN, Uddin S, Hossain MK. Role of Reactive Oxygen Species in Aging and Age-Related Diseases: A Review. ACS APPLIED BIO MATERIALS 2022; 5:4028-4054. [PMID: 36043942 DOI: 10.1021/acsabm.2c00411] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Research on the role of reactive oxygen species (ROS) in the aging process has advanced significantly over the last two decades. In light of recent findings, ROS takes part in the aging process of cells along with contributing to various physiological signaling pathways. Antioxidants being cells' natural defense mechanism against ROS-mediated alteration, play an imperative role to maintain intracellular ROS homeostasis. Although the complete understanding of the ROS regulated aging process is yet to be fully comprehended, current insights into various sources of cellular ROS and their correlation with the aging process and age-related diseases are portrayed in this review. In addition, results on the effect of antioxidants on ROS homeostasis and the aging process as well as their advances in clinical trials are also discussed in detail. The future perspective in ROS-antioxidant dynamics on antiaging research is also marshaled to provide future directions for ROS-mediated antiaging research fields.
Collapse
Affiliation(s)
- Muzahidul I Anik
- Department of Chemical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Niaz Mahmud
- Department of Biomedical Engineering, Military Institute of Science and Technology, Dhaka 1216, Bangladesh
| | - Abdullah Al Masud
- Department of Chemical Engineering, Bangladesh University of Engineering and Technology, Dhaka 1000, Bangladesh
| | - Md Ishak Khan
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Md Nurul Islam
- Department of Bioregulatory Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Shihab Uddin
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, Fukuoka 819-0395, Japan
| | - M Khalid Hossain
- Institute of Electronics, Atomic Energy Research Establishment, Bangladesh Atomic Energy Commission, Dhaka 1349, Bangladesh
- Interdisciplinary Graduate School of Engineering Science, Kyushu University, Fukuoka 816-8580, Japan
| |
Collapse
|
12
|
Han J, Yang J, Wang Q, Yin X, Sun Z, Huang C, Chen G, Zheng L, Jiang D. Ibandronate promotes autophagy by inhibiting Rac1–mTOR signaling pathway in vitro and in vivo. Cell Death Dis 2022; 8:186. [PMID: 35397636 PMCID: PMC8994753 DOI: 10.1038/s41420-022-00995-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/02/2022]
Abstract
We previously reported that ibandronate (IBAN) could improve endothelial function in spontaneously hypertensive rats. However, the mechanism by which IBAN improves endothelial function is unclear. The IBAN-induced autophagic process in vitro experiments were determined by detection of LC3, Beclin1, and P62 protein levels via western blotting. The autophagy flux was detected by confocal microscopy and transmission electron microscopy. For in vivo experiments, spontaneously hypertensive rats were orally administered with IBAN. Utilizing angiotensin II (Ang II) to stimulate the human umbilical vein endothelial cells (HUVECs) and human pulmonary microvascular endothelial cells (HPMECs) as a model of endothelial cell injury in hypertension, we found that IBAN promoted autophagy and protected cell viability in Ang II-treated-endothelial cells while these effects could be reversed by autophagy inhibitor. In terms of mechanism, IBAN treatment decreased the levels of Rac1 and mammalian target of rapamycin (mTOR) pathway. Activating either Rac1 or mTOR could reverse IBAN-induced autophagy. Furthermore, the in vivo experiments also indicated that IBAN promotes autophagy by downregulating Rac1-mTOR. Taken together, our results firstly revealed that IBAN enhances autophagy via inhibiting Rac1-mTOR signaling pathway, and thus alleviates Ang II-induced injury in endothelial cells.
Collapse
|
13
|
Xu Y, Zheng Y, Shen P, Zhou L. Role of long noncoding RNA KCNQ1 overlapping transcript 1/microRNA-124-3p/BCL-2-like 11 axis in hydrogen peroxide (H 2O 2)-stimulated human lens epithelial cells. Bioengineered 2022; 13:5035-5045. [PMID: 35170373 PMCID: PMC8973598 DOI: 10.1080/21655979.2022.2032966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Age-related cataract (ARC) is one of the most common causes of vision loss in aging people. This research analyzed the functions and mechanism of long noncoding RNA KCNQ1 overlapping transcript 1 (KCNQ1OT1) in hydrogen peroxide (H2O2)-stimulated human lens epithelial cells (SRA01/04 cells) in ARC. SRA01/04 cells were stimulated with 200 µM H2O2 to establish oxidative damage in the ARC model. A MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay and flow cytometry analysis were conducted to evaluate cell growth and apoptosis. The relevance between KCNQ1OT1 and microRNA (miR)-124-3p or miR-124-3p and BCL-2-like 11 (BCL2L11) was measured through Starbase and a dual luciferase reporter gene assay. The levels of KCNQ1OT1 and miR-124-3p were assessed via quantitative real-time polymerase chain reaction (qRT-PCR). We observed that KCNQ1OT1 was over-expressed and miR-124-3p was low-expressed in H2O2-stimulated SRA01/04 cells. KCNQ1OT1 interacted with miR-124-3p and negatively mediated its levels. In addition, KCNQ1OT1-siRNA reversed the effects of H2O2 on SRA01/04 cells, evidenced by enhanced cell viability, inhibited apoptotic cells, promoted Bcl-2 expression, and reduced Bax levels. Nevertheless, these observations were inverted after miR-124-3p inhibitor treatment. Likewise, miR-124-3p mimic had a protective effect on H2O2-stimulated SRA01/04 cells. Our data suggested that BCL2L11 targeted miR-124-3p directly. In summary, the data indicated that lncRNA KCNQ1OT1 down-regulation protected SRA01/04 cells from oxidative stress stimulated damage via the miR-124-3p/BCL2L11 pathway.
Collapse
Affiliation(s)
- Yue Xu
- Department of Ophthalmology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Yanhua Zheng
- Department of Ophthalmology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Pincheng Shen
- Department of Ophthalmology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Liping Zhou
- Department of Ophthalmology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
14
|
MiR-124 and miR-506 are involved in the decline of protein C in children with extra-hepatic portal vein obstruction. Sci Rep 2021; 11:12320. [PMID: 34112885 PMCID: PMC8192904 DOI: 10.1038/s41598-021-91862-4] [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: 10/29/2020] [Accepted: 06/02/2021] [Indexed: 12/13/2022] Open
Abstract
The deficiency of protein C (PROC) can be partly rescued by Rex shunt through restoring portal blood flow in children with extra-hepatic portal venous obstruction (EHPVO). However, the decline of PROC is still found in some patients with a normal portal blood flow after Rex shunt. The aim of this study was to identify the candidate miRNAs involving in the decline of PROC and their mechanism. The protein level of PROC was detected by the ELISA assay, and was compared between sick and healthy groups. The expressions of miRNAs and PROC mRNA were measured using qRT-PCR, and were compared between sick and healthy groups. The correlation between PROC and candidate miRNAs was analysed by a Pearson correlation analysis to identify the most significant miRNAs. The expression of PROC mRNA was detected by qRT-PCR in HL-7702 and LX-2 cells tansfected with miRNAs mimics or inhibitors and negative control (NC) mimics, which was compared among the different groups. The rates of liver cells’ proliferation and apoptosis were detected in HL-7702 and LX-2 cells tansfected with miRNAs mimics or inhibitors or with overexpressing PROC and negative control mimics by CKK8 assay and flow cytometry, which were compared among the different groups. The expressions of COX-2 and VEGF were measured by qRT-PCR, and were compared between the miRNAs groups and NC group. Western blot was assayed for detecting the protein levels of PROC, COX-2, VEGF, Bcl-2 and Bax, which were compared between the miRNAs groups and NC group. The expression of PROC mRNA was lower, and the expressions of miR-506-3p and miR-124-3p were higher in children with EHPVO than healthy group. PROC mRNA was negatively correlated with the expression of miR-506-3p and miR-124-3p. Compared to the NC group, the transcription activity of PROC was lower after exposure of miR-506 and miR-124 mimics in HL-7702 and LX-2 cells, but this phenomenon was reversed after inhibiting miR-506 and miR-124. The rate of cell proliferation was lower after exposure of miR-506 and miR-124 than the NC group, which was increased after inhibiting miR-506 and miR-124 in HL-7702 cells and overexpressing PROC in LX-2 cells. The apoptotic rate was higher after exposure of miR-506 and miR-124 than the NC group, which was decreased after inhibiting miR-506 and miR-124 in HL-7702 cells and overexpressing PROC in LX-2 cells. The mRNA levels of COX-2 and VEGF were significantly higher after exposure of miR-506 and miR-124 mimics than those in the NC group. The protein levels of PROC and Bcl-2 were down-regulated, and the levels of COX-2, Bax and VEGF were up-regulated after exposure of miR-506 and miR-124 in HL-7702 cells, but this phenomenon was reversed after inhibiting miR-506 and miR-124. MiR-506-3p and miR-124-3p may involve in the decline of PROC in protein and transcriptional level, in which the anti-proliferation and pro-apoptosis role of miR-506-3p and miR-124-3p for liver cells may involve in this mechanism.
Collapse
|
15
|
Li Q, Liu S, Yan J, Sun MZ, Greenaway FT. The potential role of miR-124-3p in tumorigenesis and other related diseases. Mol Biol Rep 2021; 48:3579-3591. [PMID: 33877528 DOI: 10.1007/s11033-021-06347-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/07/2021] [Indexed: 01/16/2023]
Abstract
MicroRNAs (miRNAs) are a class of single-stranded noncoding and endogenous RNA molecules with a length of 18-25 nucleotides. Previous work has shown that miR-124-3p leads to malignant progression of cancer including cell apoptosis, migration, invasion, drug resistance, and also recovers neural function, affects adipogenic differentiation, facilitates wound healing through control of various target genes. miR-124-3p has been mainly previously characterized as a tumor suppressor regulating tumorigenesis and progression in several cancers, such as hepatocellular carcinoma (HCC), gastric cancer (GC), bladder cancer, ovarian cancer (OC), and leukemia, as a tumor promotor in breast cancer (BC), and it has been also widely studied in a variety of neurological diseases, like Parkinson's disease (PD), dementia and Alzheimer's disease (AD), and cardiovascular diseases, ulcerative colitis (UC), acute respiratory distress syndrome (ARDS). To lay the groundwork for future therapeutic strategies, in this review we mainly focus on the most recent years of literature on the functions of miR-124-3p in related major cancers, as well as its downstream target genes. Although current work as yet provides an incomplete picture, miR-124-3p is still worthy of more attention as a practical and effective clinical biomarker.
Collapse
Affiliation(s)
- Qian Li
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun Southern Road, Dalian, 116044, China.,Department of Hematology, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Shuqing Liu
- Department of Biochemistry, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun Southern Road, Dalian, 116044, China. .,Department of Hematology, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China.
| | - Jinsong Yan
- Department of Hematology, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Ming-Zhong Sun
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun Southern Road, Dalian, 116044, China. .,Department of Hematology, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China.
| | - Frederick T Greenaway
- Carlson School of Chemistry and Biochemistry, Clark University, Worcester, MA, 01610, USA
| |
Collapse
|