1
|
Liu G, Song Y, Yin S, Zhang B, Han P. Machine learning using scRNA-seq Combined with bulk-seq to identify lactylation-related hub genes in carotid arteriosclerosis. Sci Rep 2025; 15:17794. [PMID: 40404675 PMCID: PMC12098907 DOI: 10.1038/s41598-025-00834-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 04/30/2025] [Indexed: 05/24/2025] Open
Abstract
Atherosclerosis is a chronic inflammatory disease, this study aims to investigate the immune landscape in carotid atherosclerotic plaque formation and explore diagnostic biomarkers of lactylation-associated genes, so as to gain new insights into underlying molecular mechanisms and provide new perspectives for disease detection and treatment. Single cell transcriptome data and Bulk transcriptome data of carotid atherosclerosis samples were obtained from the Gene Expression Omnibus (GEO). Eleven cell types were identified by scRNA-seq data. Lactylation scores were significantly higher in γδT cells than in cells of other subtypes, but lower in plasma cells than in cells of other subtypes. The scores of malignant related pathways were significantly increased in cells with high lactylation scores. scRNA-seq combined with bulk-seq identified differentially expressed lactylation genes in carotid atherosclerosis. A diagnostic model was constructed by combining 10 machine learning algorithms and 101 algorithms, SOD1, DDX42 and PDLIM1 as core genes. Further analysis revealed that the expression levels of core genes were significantly correlated with immune cell infiltration, and their regulatory networks were constructed. Clinical samples verified that the expression of core gene in unstable plaque was significantly lower than that in stable plaque, suggesting that it has protective effect on atherosclerosis. By combining scRNA-seq and Bulk transcriptome data in this study, three lactylation-associated genes SOD1, DDX42 and PDLIM1 were identified in carotid atherosclerosis samples, providing targets for the diagnosis and treatment of carotid atherosclerosis samples.
Collapse
Affiliation(s)
- Gaoyan Liu
- Department of Vascular Surgery, First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Ye Song
- Department of General Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Shanxue Yin
- Department of Vascular Surgery, First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Bo Zhang
- Department of Vascular Surgery, First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Peng Han
- Department of Vascular Surgery, First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
2
|
Bolha L, Hočevar A, Jurčić V. Current state of epigenetics in giant cell arteritis: Focus on microRNA dysregulation. Autoimmun Rev 2025; 24:103739. [PMID: 39732382 DOI: 10.1016/j.autrev.2024.103739] [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: 09/18/2024] [Revised: 12/23/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
Giant cell arteritis (GCA) is a primary systemic vasculitis affecting the elderly, characterized by a granulomatous vessel wall inflammation of large- and medium-sized arteries. The immunopathology of GCA is complex, involving both the innate and adaptive arms of the immune system, where a maladaptive inflammatory-driven vascular repair process ultimately results in vessel wall thickening, intramural vascular smooth muscle cell proliferation, neovascularization and vessel lumen occlusion, which can lead to serious ischemic complications such as visual loss and ischemic stroke. Over the past decade, microRNA (miRNA) dysregulation has been highlighted as an important contributing factor underlying the pathogenesis of GCA. Since current understanding of miRNA involvement in GCA remains largely based on extrapolation of previously determined miRNA functions in vitro or in loss- or gain-of-function studies, an overall insight into the role of miRNA alteration in GCA pathophysiology remains limited. In this narrative review, we summarize the current knowledge on aberrantly expressed miRNAs in GCA and thoroughly discuss the impact of their altered regulatory role in the context of GCA setting. Furthermore, we address challenges and future perspectives in utilization of miRNA-based diagnostic and prognostic biomarkers of GCA in clinical settings.
Collapse
Affiliation(s)
- Luka Bolha
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| | - Alojzija Hočevar
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vesna Jurčić
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
3
|
Chen X, Wang S, Hou W, Zhang Y, Hou Y, Tong H, Zhang X, Liu Y, Yang R, Li X, Fang Q, Fan J. Decellularized adipose matrix hydrogel-based in situ delivery of antagomiR-150-5p for rat abdominal aortic aneurysm therapy. Mater Today Bio 2024; 29:101350. [PMID: 39677522 PMCID: PMC11638622 DOI: 10.1016/j.mtbio.2024.101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 12/17/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a progressive aortic disease featured by inflammation, vascular smooth muscle cells (VSMCs) depletion, and elastin degradation. MicroRNAs were related to AAA formation, which bring the approach for precise and targeted drug therapy for AAA. We developed a new strategy based on decellularized adipose matrix (DAM) hydrogel immobilized on the adventitia to release antagomiR-150-5p for preventing the AAA development. In this study, Cacl2-induced and elastase-induced rat AAA models were established. We found that miR-150-5p was upregulated while Notch3 was downregulated in two rat AAA models. Then a mold was designed for shaping hydrogel for miR-150-5p delivery around the abdominal aorta. Interestingly, inhibition of miR-150-5p in AAA by local release of antagomiR-150-5p with DAM hydrogel significantly prevented aortic dilation and elastin degradation. Moreover, inflammatory cell infiltration, the expression of inflammatory cytokines (MCP-1, TNF-α, and NF-κB (p65)), and matrix metalloproteinases (MMP-2, MMP-9) were increased while Notch3 and α-SMA were decreased in rat AAA, which can be attenuated by antagomiR-150-5p treatment. In VSMCs with TNF-α stimulation, we further demonstrated that inhibition of miR-150-5p downregulated NF-κB (p65), MMP-2, and MMP-9 and upregulated elastin via Notch3. This work presents a translational potential strategy for AAA repair via DAM hydrogel sustained release of antagomiR-150-5p, and highlights the mechanism of miR-150-5p during AAA progression by regulating Notch3.
Collapse
Affiliation(s)
- Xin Chen
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Shoushuai Wang
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
- Department of Radiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, 570311, PR China
| | - Weijian Hou
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Yanhui Zhang
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Yapeng Hou
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Hao Tong
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Xiaoxin Zhang
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Yue Liu
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Ruoxuan Yang
- Department of Dental Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning Province, PR China
| | - Xiang Li
- Department of Cell Biology, School of Life Sciences, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| | - Qin Fang
- Cardiac Surgery, First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, PR China
| | - Jun Fan
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, PR China
| |
Collapse
|
4
|
Zhao H, Jin Z, Li J, Fang J, Wu W, Fang JF. Novel insights of disulfidptosis-mediated immune microenvironment regulation in atherosclerosis based on bioinformatics analyses. Sci Rep 2024; 14:27336. [PMID: 39521794 PMCID: PMC11550432 DOI: 10.1038/s41598-024-78392-5] [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: 06/05/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Atherosclerosis (AS) is the leading cause of coronary heart disease, which is the primary cause of death worldwide. Recent studies have identified disulfidptosis as a new type of cell death that may be involved in onset and development of many diseases. However, the role of disulfidptosis in AS is not clear. In this study, bioinformatics analysis and experiments in vivo and in vitro were performed to evaluate the potential relationship between disulfidptosis and AS. AS-related sequencing data were obtained from the Gene Expression Omnibus (GEO). Bioinformatics techniques were used to evaluate differentially expressed genes (DEGs) associated with disulfidptosis-related AS. Hub genes were screened using least absolute shrinkage and selection operator (LASSO) and random forests (RF) methods. In addition, we established a foam cell model in vitro and an AS mouse model in vivo to verify the expressions of hub genes. In addition, we constructed a diagnostic nomogram with hub genes to predict progression of AS. Finally, the consensus clustering method was used to establish two different subtypes, and associations between subtypes and immunity were explored. As the results, 9 disulfidptosis-related AS DEGs were identified from GSE28829 and GSE43292 datasets. Evaluation of DEGs using LASSO and RF methods resulted in identification of 4 hub genes (CAPZB, DSTN, MYL6, PDLIM1), which were analyzed for diagnostic value using ROC curve analysis and verified in vitro and in vivo. Furthermore, a nomogram including hub genes was established that accurately predicted the occurrence of AS. The consensus clustering algorithm was used to separate patients with early atherosclerotic plaques and patients with advanced atherosclerotic plaques into two disulfidptosis subtypes. Cluster B displayed higher levels of infiltrating immune cells, which indicated that patients in cluster B may have a positive immune response for progression of AS. In summary, disulfidptosis-related genes including CAPZB, DSTN, MYL6, and PDLIM1 may be diagnostic markers and therapeutic targets for AS. In addition, these genes are closely related to immune cells, which may inform immunotherapy for AS.
Collapse
Affiliation(s)
- Huanyi Zhao
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Zheng Jin
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Junlong Li
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Junfeng Fang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Wei Wu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| | - J F Fang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| |
Collapse
|
5
|
Deng Y, Jiang S, Lin X, Wang B, Chen B, Tong J, Shi W, Yu B, Tang J. Differential expression profile of miRNAs between stable and vulnerable plaques of carotid artery stenosis patients. Genes Genet Syst 2023. [PMID: 37121730 DOI: 10.1266/ggs.22-00123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Plaque vulnerability is associated with the degree of carotid artery stenosis (CS) and the risk of stroke. MicroRNAs (miRNAs) exert critical functions in disease progression, although only a few miRNAs have been well identified in CS. Therefore, this study aimed to investigate the differential expression profile of miRNAs and their potential functions in plaques of CS patients. Three CS patients with stable plaques and three patients with vulnerable plaques who underwent carotid endarterectomy were enrolled in this study. Differentially expressed miRNAs (DEmiRNAs) between patients with stable and vulnerable plaques were determined using small RNA sequencing. Target genes of DEmiRNAs were predicted and submitted to functional analyses. Validation of dysregulated DEmiRNAs was determined using quantitative real-time polymerase chain reaction (qRT-PCR). After sequencing, 76 DEmiRNAs were identified in vulnerable plaques, including 53 upregulated miRNAs and 23 downregulated miRNAs. Next, 23,495 target genes of the identified DEmiRNAs were predicted and functionally analyzed. This indicated that the target genes of the identified DEmiRNAs were mainly enriched in protein phosphorylation, transcription, nitrogen compound metabolism, endocytosis and autophagy, and related to signaling pathways of Hippo, MAPK, insulin, TGF-β, FoxO, AMPK and p53. Furthermore, qRT-PCR results for six miRNAs showed that five (83%) of them (hsa-miR-511-5p, hsa-miR-150-5p, hsa-miR-378a-5p, hsa-miR-365b-5p and hsa-miR-6511b-5p) were consistent with the sequencing results. Differential expression profiles and potential function of miRNAs associated with plaque stability in CS patients are identified for the first time, which should help to understand the regulatory mechanism of plaque stability in CS.
Collapse
Affiliation(s)
- Ying Deng
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Shuai Jiang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Xueguang Lin
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Bo Wang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Bo Chen
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Jindong Tong
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Weijun Shi
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Bo Yu
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| | - Jingdong Tang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center
- Fudan Zhangjiang Institute
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling
| |
Collapse
|
6
|
Rozhkov AN, Shchekochikhin DY, Ashikhmin YI, Mitina YO, Evgrafova VV, Zhelankin AV, Gognieva DG, Akselrod AS, Kopylov PY. The Profile of Circulating Blood microRNAs in Outpatients with Vulnerable and Stable Atherosclerotic Plaques: Associations with Cardiovascular Risks. Noncoding RNA 2022; 8:ncrna8040047. [PMID: 35893230 PMCID: PMC9326687 DOI: 10.3390/ncrna8040047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Non-coding RNAs reflect many biological processes in the human body, including athero-sclerosis. In a cardiology outpatient department cohort (N = 83), we aimed to compare the levels of circulating microRNAs in groups with vulnerable plaques (N = 22), stable plaques (N = 23) and plaque-free (N = 17) depending on coronary computed tomography angiography and to evaluate associations of microRNA levels with calculated cardiovascular risks (CVR), based on the SCORE2 (+OP), ACC/AHA, ATP-III and MESA scales. Coronary computed tomography was performed on a 640-slice computed tomography scanner. Relative plasma levels of microRNA were assessed via a real-time polymerase chain reaction. We found significant differences in miR-143-3p levels (p = 0.0046 in plaque-free vs. vulnerable plaque groups) and miR-181b-5p (p = 0.0179 in stable vs. vulnerable plaques groups). Analysis of microRNA associations with CVR did not show significant differences for SCORE2 (+OP) and ATPIII scales. MiR-126-5p and miR-150-5p levels were significantly higher (p < 0.05) in patients with ACC/AHA risk >10% and miR-145-5p had linear relationships with ACC/AHA score (adjusted p = 0.0164). The relative plasma level of miR-195 was higher (p < 0.05) in patients with MESA risk > 7.5% and higher (p < 0.05) in patients with zero coronary calcium index (p = 0.036). A linear relationship with coronary calcium was observed for miR-126-3p (adjusted p = 0.0484). A positive correlation with high coronary calcium levels (> 100 Agatson units) was found for miR-181-5p (p = 0.036). Analyzing the biological pathways of these microRNAs, we suggest that miR-143-3p and miR-181-5p can be potential markers of the atherosclerosis process. Other miRNAs (miR-126-3p, 126-5p, 145-5p, 150-5p, 195-5p) can be considered as potential cardiovascular risk modifiers, but it is necessary to validate our results in a large prospective trial.
Collapse
Affiliation(s)
- Andrey N. Rozhkov
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (D.G.G.); (P.Y.K.)
- Correspondence: ; Tel.: +7-915-085-32-95
| | - Dmitry Yu. Shchekochikhin
- Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (D.Y.S.); (V.V.E.); (A.S.A.)
| | - Yaroslav I. Ashikhmin
- International Medical Cluster, 40 Bolshoy Boulevard Skolkovo Innovation Center, 121205 Moscow, Russia;
| | - Yulia O. Mitina
- Skolkovo Institute of Science and Technology, 121205 Moscow, Russia;
| | - Veronika V. Evgrafova
- Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (D.Y.S.); (V.V.E.); (A.S.A.)
| | - Andrey V. Zhelankin
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia;
| | - Daria G. Gognieva
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (D.G.G.); (P.Y.K.)
- Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (D.Y.S.); (V.V.E.); (A.S.A.)
| | - Anna S. Akselrod
- Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (D.Y.S.); (V.V.E.); (A.S.A.)
| | - Philippe Yu. Kopylov
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (D.G.G.); (P.Y.K.)
- Department of Cardiology, Functional and Ultrasound Diagnostics, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (D.Y.S.); (V.V.E.); (A.S.A.)
| |
Collapse
|
7
|
Azari ZD, Aljubran F, Nothnick WB. Inflammatory MicroRNAs and the Pathophysiology of Endometriosis and Atherosclerosis: Common Pathways and Future Directions Towards Elucidating the Relationship. Reprod Sci 2022; 29:2089-2104. [PMID: 35476352 DOI: 10.1007/s43032-022-00955-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/19/2022] [Indexed: 11/25/2022]
Abstract
Emerging data indicates an association between endometriosis and subclinical atherosclerosis, with women with endometriosis at a higher risk for cardiovascular disease later in life. Inflammation is proposed to play a central role in the pathophysiology of both diseases and elevated levels of systemic pro-inflammatory cytokines including macrophage migration inhibitory factor (MIF), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) are well documented. However, a thorough understanding on the mediators and mechanisms which contribute to altered cytokine expression in both diseases remain poorly understood. MicroRNAs (miRNAs) are important post-transcriptional regulators of inflammatory pathways and numerous studies have reported altered circulating levels of miRNAs in both endometriosis and atherosclerosis. Potential contribution of miRNA-mediated inflammatory cascades common to the pathophysiology of both diseases has not been evaluated but could offer insight into common pathways and early manifestation relevant to both diseases which may help understand cause and effect. In this review, we discuss and summarize differentially expressed inflammatory circulating miRNAs in endometriosis subjects, compare this profile to that of circulating levels associated with atherosclerosis when possible, and then discuss mechanistic studies focusing on these miRNAs in relevant cell, tissue, and animal models. We conclude by discussing the potential utility of targeting the relevant miRNAs in the MIF-IL-6-TNF-α pathway as therapeutic options and offer insight into future studies which will help us better understand not only the role of these miRNAs in the pathophysiology of both endometriosis and atherosclerosis but also commonality between both diseases.
Collapse
Affiliation(s)
- Zubeen D Azari
- Kansas City University of Medicine and Biosciences, Kansas City, MO, 64106, USA
| | - Fatimah Aljubran
- Department of Molecular and Integrative Physiology, Institute for Reproductive and Perinatal Sciences, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Warren B Nothnick
- Department of Molecular and Integrative Physiology, Institute for Reproductive and Perinatal Sciences, University of Kansas Medical Center, Kansas City, KS, 66160, USA. .,Department of Obstetrics and Gynecology, Institute for Reproductive and Perinatal Sciences, University of Kansas Medical Center, Kansas City, KS, 66160, USA. .,Center for Reproductive Sciences, Institute for Reproductive and Perinatal Sciences, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
8
|
Nazarenko MS, Koroleva IA, Zarubin AA, Sleptcov AA. miRNA Regulome in Different Atherosclerosis Phenotypes. Mol Biol 2022. [DOI: 10.1134/s0026893322020108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
9
|
Macchi C, Greco MF, Favero C, Dioni L, Cantone L, Hoxha M, Vigna L, Solazzo G, Corsini A, Banach M, Pesatori AC, Bollati V, Ruscica M. Associations Among PCSK9 Levels, Atherosclerosis-Derived Extracellular Vesicles, and Their miRNA Content in Adults With Obesity. Front Cardiovasc Med 2022; 8:785250. [PMID: 35071356 PMCID: PMC8782054 DOI: 10.3389/fcvm.2021.785250] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/13/2021] [Indexed: 01/16/2023] Open
Abstract
Background: Extracellular vesicles (EV) concentration is generally increased in patients with cardiovascular diseases, although the protective role of EVs in atherosclerosis has been reported. Among the specific cargo of EVs, miRNAs contribute to different stages of atherosclerosis. Aim of the present report has been to investigate, in individuals with obesity, the interplay among EVs derived from cells relevant for the atherosclerotic process (i.e., platelets, endothelium, monocytes/macrophages, and neutrophils), their miRNA content and proprotein convertase subtilisin/kexin type 9 (PCSK9), one of the main regulators of low-density lipoprotein receptor (LDLR). Methods and Results: EVs have been isolated from 936 individuals with obesity (body mass index = 33.6 ± 5.6 Kg/m2) and a raised cardiovascular risk (e.g., LDL-C = 131.6 ± 36.4 mg/dL, HOMA-IR = 3.1, and roughly 50% on anti-hypertensive medications). PCSK9 levels were negatively associated with EV count in the range 150–400 nm and with those derived from macrophages (CD14+), endothelium (CD105+), and neutrophils (CD66+). The association between PCSK9 and platelet-derived EVs (CD61+) was modified by platelet counts. PCSK9 was significantly associated with five EV-derived miRNAs (hsa-miRNA−362−5p,−150,−1244,−520b-3p,−638). Toll-like receptor 4 and estrogen receptor 1 were targeted by all five miRNAs and LDLR by four. The effect on LDLR expression is mainly driven by hsa-miR-150. Considering the implication of EV in atherosclerosis onset and progression, our findings show a potential role of PCSK9 to regulate EV-derived miRNAs, especially those involved in inflammation and expression of low-density lipoprotein receptor (LDLR) receptor.
Collapse
Affiliation(s)
- Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Maria Francesca Greco
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Favero
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Laura Dioni
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Laura Cantone
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Mirjam Hoxha
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Luisella Vigna
- Occupational Medicine Unit, Fondazione Cà Granda, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulia Solazzo
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.,Istituto di Ricovero e Cura a Carattere ScientificoI (RCCS) Multimedica, Milan, Italy
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Lodz, Poland.,Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Angela C Pesatori
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Valentina Bollati
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
10
|
Bian Y, Cai W, Lu H, Tang S, Yang K, Tan Y. miR-150-5p affects AS plaque with ASMC proliferation and migration by STAT1. Open Med (Wars) 2021; 16:1642-1652. [PMID: 34761115 PMCID: PMC8569285 DOI: 10.1515/med-2021-0357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/09/2021] [Accepted: 08/18/2021] [Indexed: 11/15/2022] Open
Abstract
We explore miR-150-5p in atherosclerosis (AS). The AS model was constructed using Apo E-/- mice with an injection of the miR-150-5p mimic or an inhibitor. Pathological characteristics were assessed using Oil red O staining and Masson staining. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot were used to analyze the expressions of microRNA-150-5p (miR-150-5p), STAT1, α-SMA (α-smooth muscle actin) and proliferating cell nuclear antigen (PCNA). Targetscan and dual-luciferase reporter assay were used to analyze the interaction between miR-150-5p and STAT1. The viability, migration, cell cycle and α-SMA and PCNA expressions in oxidized low-density lipoprotein (ox-LDL)-stimulated primary human aortic smooth muscle cells (ASMCs) were assessed using molecular experiments. miR-150-5p was reduced in both AS mice and ox-LDL-stimulated human aortic smooth muscle cells but STAT1 had the opposite effect. The miR-150-5p inhibitor alleviated the increase of lipid plaque and reduced collagen accumulation in the aortas during AS. Upregulation of α-SMA and PCNA was reversed by miR-150-5p upregulation. STAT1 was targeted by miR-150-5p, and overexpressed miR-150-5p weakened the ox-LDL-induced increase of viability and migration abilities and blocked cell cycle in ASMCs, but overexpressed STAT1 blocked the effect of the miR-150-5p mimic. This paper demonstrates that miR-150-5p has potential as a therapeutic target in AS, with plaque stabilization by regulating ASMC proliferation and migration via STAT1.
Collapse
Affiliation(s)
- Yuan Bian
- Department of Neurosurgery, Guigang City People's Hospital, Guigang, 537100, China
| | - Wenqiang Cai
- Department of Neurosurgery, Guigang City People's Hospital, Guigang, 537100, China
| | - Hongying Lu
- Department of Neurosurgery, Guigang City People's Hospital, Guigang, 537100, China
| | - Shuhong Tang
- Department of Neurosurgery, Guigang City People's Hospital, Guigang, 537100, China
| | - Keqin Yang
- Department of Neurosurgery, Guigang City People's Hospital, Guigang, 537100, China
| | - Yan Tan
- Department of Neurosurgery, Guigang City People's Hospital, No. 1, Zhongshan Middle Road, Guigang, 537100, China
| |
Collapse
|
11
|
Li X, Yang Y, Wang Z, Jiang S, Meng Y, Song X, Zhao L, Zou L, Li M, Yu T. Targeting non-coding RNAs in unstable atherosclerotic plaques: Mechanism, regulation, possibilities, and limitations. Int J Biol Sci 2021; 17:3413-3427. [PMID: 34512156 PMCID: PMC8416736 DOI: 10.7150/ijbs.62506] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/23/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) caused by arteriosclerosis are the leading cause of death and disability worldwide. In the late stages of atherosclerosis, the atherosclerotic plaque gradually expands in the blood vessels, resulting in vascular stenosis. When the unstable plaque ruptures and falls off, it blocks the vessel causing vascular thrombosis, leading to strokes, myocardial infarctions, and a series of other serious diseases that endanger people's lives. Therefore, regulating plaque stability is the main means used to address the high mortality associated with CVDs. The progression of the atherosclerotic plaque is a complex integration of vascular cell apoptosis, lipid metabolism disorders, inflammatory cell infiltration, vascular smooth muscle cell migration, and neovascular infiltration. More recently, emerging evidence has demonstrated that non-coding RNAs (ncRNAs) play a significant role in regulating the pathophysiological process of atherosclerotic plaque formation by affecting the biological functions of the vasculature and its associated cells. The purpose of this paper is to comprehensively review the regulatory mechanisms involved in the susceptibility of atherosclerotic plaque rupture, discuss the limitations of current approaches to treat plaque instability, and highlight the potential clinical value of ncRNAs as novel diagnostic biomarkers and potential therapeutic strategies to improve plaque stability and reduce the risk of major cardiovascular events.
Collapse
Affiliation(s)
- Xiaoxin Li
- Institute for translational medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, 266021, People's Republic of China
| | - Yanyan Yang
- Institute for translational medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, 266021, People's Republic of China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shaoyan Jiang
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, No. 5 Zhiquan Road, Qingdao 266000, China
| | - Yuanyuan Meng
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xiaoxia Song
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Liang Zhao
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Lu Zou
- Institute for translational medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, 266021, People's Republic of China
| | - Min Li
- Institute for translational medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, 266021, People's Republic of China
| | - Tao Yu
- Institute for translational medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, 266021, People's Republic of China.,Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
12
|
Wan W, Long Y, Jin X, Li Q, Wan W, Liu H, Zhu Y. Protective Role of microRNA-200a in Diabetic Retinopathy Through Downregulation of PDLIM1. J Inflamm Res 2021; 14:2411-2424. [PMID: 34113148 PMCID: PMC8187036 DOI: 10.2147/jir.s303540] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/04/2021] [Indexed: 12/26/2022] Open
Abstract
Background Diabetic retinopathy (DR) is a most common microvascular complication and regarded as the leading cause of blindness in the working age population. The involvement of miR-200a in various disorders has become recognized, and the objective of this study was to identify the protective effect of miR-200a in the development of DR. Methods The contents of miR-200a and its potential target gene, PDZ and LIM domain protein 1 (PDLIM1), were detected in both in-vivo and in-vitro DR models. Retinal leakage and inflammatory factor concentrations were detected after vitreous injections of miR-200a/PDLIM1 vectors in mice. The cellular viability, apoptosis and cellular migration were investigated using trypan blue staining, flow cytometry and transwell assay with human retinal microvascular endothelial cells (HRMECs). Besides, the prediction and confirmation of miR-200a targeting PDLIM1 were conducted with bioinformation analyses and dual-luciferase reporter assay. Results Lower miR-200a and higher PDLIM1 levels were detected in both in-vivo and in-vitro DR models. Besides, it was found that miR-200a treatment would significantly inhibit retinal permeability and inflammatory factors. Through targeting PDLIM1, it was found that miR-200a could improve cellular viability, remit apoptotic status and reduce cellular migration significantly in high glucose-treated HRMECs. Conclusion Our results demonstrated that miR-200a could be used as a potential therapy target through down-regulating PDLIM1 in DR.
Collapse
Affiliation(s)
- Wencui Wan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yang Long
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xuemin Jin
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Qiuming Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Weiwei Wan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Hongzhuo Liu
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yu Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
13
|
Elchaninov A, Nikitina M, Vishnyakova P, Lokhonina A, Makarov A, Sukhikh G, Fatkhudinov T. Macro- and microtranscriptomic evidence of the monocyte recruitment to regenerating liver after partial hepatectomy in mouse model. Biomed Pharmacother 2021; 138:111516. [PMID: 33765583 DOI: 10.1016/j.biopha.2021.111516] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/11/2021] [Accepted: 03/14/2021] [Indexed: 02/07/2023] Open
Abstract
Macrophages are important regulators of liver repair. Participation of migratory monocytes/macrophages in regeneration of hepatic tissues after resection remains disputable. In mouse the resection promotes migration of Ly6C+CD11b+ monocytes/macrophages to the remnant liver accompanied by a reduction in its CD206 + macrophage content. Macrophage proliferation within the liver reaches maximum on day 3 after the surgery. Corresponding macro- and microtranscriptomic profiles of macrophages in regeneration liver cannot be unambiguously defined as pro- or anti-inflammatory. Their typical features include elevated expression of leukocyte chemoattractant factors, and many of the differentially expressed sequences are related to the control of cell growth and metabolic processes in the liver. These findings revealed essential roles of immigration of monocytes/macrophages and macrophages proliferation in maintenance of macrophage populations in the mouse liver during its recovery from a massive resection.
Collapse
Affiliation(s)
- Andrey Elchaninov
- Laboratory of Regenerative Medicine, FSBI National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow 117997, Russia; Histology Department, Medical Institute, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia.
| | - Maria Nikitina
- Laboratory of Growth and Development, FSBSI Scientific Research Institute of Human Morphology, Moscow 117418, Russia
| | - Polina Vishnyakova
- Laboratory of Regenerative Medicine, FSBI National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow 117997, Russia; Histology Department, Medical Institute, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - Anastasia Lokhonina
- Laboratory of Regenerative Medicine, FSBI National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow 117997, Russia; Histology Department, Medical Institute, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - Andrey Makarov
- Laboratory of Regenerative Medicine, FSBI National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow 117997, Russia; Histology Department, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, Moscow 117997 Russia
| | - Gennady Sukhikh
- Laboratory of Regenerative Medicine, FSBI National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow 117997, Russia
| | - Timur Fatkhudinov
- Laboratory of Growth and Development, FSBSI Scientific Research Institute of Human Morphology, Moscow 117418, Russia; Histology Department, Medical Institute, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
| |
Collapse
|
14
|
Zhang Y, Wu H, He R, Ye C, Chen H, Wang J, Li Z. Dickkopf-2 knockdown protects against classic macrophage polarization and lipid loading by activation of Wnt/β-catenin signaling. J Cardiol 2021; 78:328-333. [PMID: 34030936 DOI: 10.1016/j.jjcc.2021.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Wnt/β-catenin signaling pathway plays an important role in regulation of macrophage activation implicated in the development of atherosclerosis. However, as a negative regulator of Wnt/β-catenin, the potential role of Dickkopf-2 (Dkk2) on macrophage activation remains unexplored. MATERIALS AND METHODS Bone marrow-derived macrophages (BMDMs) and mouse peritoneal macrophages (MPMs) collected from ApoE knockout mice upon oxidation low lipoprotein (Ox-LDL) administration were performed to test the expression of Dkk2. The loss-of-function strategy using siRNA-Dkk2 was further utilized for the function of Dkk2. Inhibition of β-catenin with XAV939 (a β-catenin specific inhibitor) was further used for testing its effect on macrophage activation mediated by Dkk2 knockdown. RESULTS AND CONCLUSION In the current study, real time-polymerase chain reaction analysis demonstrated that an up-regulated Dkk2 expression was observed in BMDMs and MPMs of ApoE knockout mice upon Ox-LDL administration, which was confirmed by western blot. The double immunofluorescence staining further exhibited that Dkk2 showed a strong immunoreactivity in BMDMs and primarily located in cytoplasm of macrophages. Dkk2 knockdown significantly decreased the genes related to classic M1 polarized macrophage but increased alternative M2 polarized macrophage markers. Moreover, Dkk2 silencing dramatically attenuated foam cell formation which was contributed by promoted markers' expression associated with cholesterol efflux but attenuated markers to cholesterol influx. Mechanistically, we observed that Dkk2 knockdown activated Wnt/β-catenin signaling by promoting β-catenin to translocate into the nuclei of macrophages, and XAV939 reversed the ameliorated effect of Dkk2 silencing macrophage activation. Taken together, these results suggested that downregulated Dkk2 expression in macrophages was responsible for the inactivation of macrophage through targeting Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yuan Zhang
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Hongkun Wu
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Rui He
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Changlun Ye
- Department of Cardiology, Chongqing Qijiang District People's Hospital, Chongqing, PR China
| | - Hao Chen
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Jiao Wang
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Zhenggong Li
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China.
| |
Collapse
|
15
|
Sapp RM, Chesney CA, Springer CB, Laskowski MR, Singer DB, Eagan LE, Mascone SE, Evans WS, Prior SJ, Hagberg JM, Ranadive SM. Race-specific changes in endothelial inflammation and microRNA in response to an acute inflammatory stimulus. Am J Physiol Heart Circ Physiol 2021; 320:H2371-H2384. [PMID: 33961505 DOI: 10.1152/ajpheart.00991.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Both aberrant vascular reactivity to acute cardiovascular stress and epigenetic mechanisms such as microRNA (miR) may underlie the increased propensity for African Americans (AA) to develop cardiovascular disease. This study assessed racial differences in acute induced endothelial inflammation and related miRs. Cultured human umbilical vein endothelial cells (HUVECs) derived from AA and Caucasian Americans (CA) were exposed to influenza vaccine to determine changes in inflammatory markers, endothelial nitric oxide synthase (eNOS), and miR expression/release. Endothelial function [flow-mediated dilation (FMD)], circulating IL-6, and circulating miR were also measured in young, healthy AA and CA individuals before and after receiving the influenza vaccine. There were no significant racial differences in any parameters at baseline. The vaccine induced increases in IL-6 release (24%, P = 0.02) and ICAM-1 mRNA (40%, P = 0.03), as well as reduced eNOS mRNA (24%, P = 0.04) in AA HUVECs, but not in CA HUVECs (all P > 0.05). Intracellular levels of anti-inflammatory miR-221-3p and miR-222-3p increased specifically in CA HUVECs (72% and 53%, P = 0.04 and P = 0.06), whereas others did not change in either race. HUVEC secretion of several miRs decreased in both races, whereas the release of anti-inflammatory miR-150-5p was decreased only by AA cells (-30%, P = 0.03). In individuals of both races, circulating IL-6 increased approximately twofold 24 h after vaccination (both P < 0.01) and returned to baseline levels by 48 h, whereas FMD remained unchanged. Although macrovascular function was unaffected by acute inflammation in AA and CA individuals, AA endothelial cells exhibited increased susceptibility to acute inflammation and unique changes in related miR.NEW & NOTEWORTHY Used as an acute inflammatory stimulus, the influenza vaccine induced an inflammatory response and decreased eNOS gene expression in endothelial cells derived from African Americans, but not Caucasian Americans. Race-specific changes in intracellular expression and release of specific microRNAs also occurred and may contribute to an exaggerated inflammatory response in African Americans. In vivo, the vaccine caused similar systemic inflammation but had no effect on endothelial function or circulating microRNAs in individuals of either race.
Collapse
Affiliation(s)
- Ryan M Sapp
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Catalina A Chesney
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Catherine B Springer
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Matthew R Laskowski
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland
| | - Daniel B Singer
- Department of Biology, University of Maryland, College Park, Maryland
| | - Lauren E Eagan
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Sara E Mascone
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - William S Evans
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Steven J Prior
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland.,Baltimore Veterans Affairs Geriatric Research, Education and Clinical Center, Baltimore, Maryland
| | - James M Hagberg
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Sushant M Ranadive
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| |
Collapse
|
16
|
Seneviratne A, Cave L, Hyde G, Moestrup SK, Carling D, Mason JC, Haskard DO, Boyle JJ. Metformin directly suppresses atherosclerosis in normoglycaemic mice via haematopoietic adenosine monophosphate-activated protein kinase. Cardiovasc Res 2021; 117:1295-1308. [PMID: 32667970 PMCID: PMC8064441 DOI: 10.1093/cvr/cvaa171] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 06/03/2018] [Accepted: 06/22/2020] [Indexed: 12/31/2022] Open
Abstract
AIMS Atherosclerotic vascular disease has an inflammatory pathogenesis. Heme from intraplaque haemorrhage may drive a protective and pro-resolving macrophage M2-like phenotype, Mhem, via AMPK and activating transcription factor 1 (ATF1). The antidiabetic drug metformin may also activate AMPK-dependent signalling. Hypothesis: Metformin systematically induces atheroprotective genes in macrophages via AMPK and ATF1, thereby suppresses atherogenesis. METHODS AND RESULTS Normoglycaemic Ldlr-/- hyperlipidaemic mice were treated with oral metformin, which profoundly suppressed atherosclerotic lesion development (P < 5 × 10-11). Bone marrow transplantation from AMPK-deficient mice demonstrated that metformin-related atheroprotection required haematopoietic AMPK [analysis of variance (ANOVA), P < 0.03]. Metformin at a clinically relevant concentration (10 μM) evoked AMPK-dependent and ATF1-dependent increases in Hmox1, Nr1h2 (Lxrb), Abca1, Apoe, Igf1, and Pdgf, increases in several M2-markers and decreases in Nos2, in murine bone marrow macrophages. Similar effects were seen in human blood-derived macrophages, in which metformin-induced protective genes and M2-like genes, suppressible by si-ATF1-mediated knockdown. Microarray analysis comparing metformin with heme in human macrophages indicated that the transcriptomic effects of metformin were related to those of heme, but not identical. Metformin-induced lesional macrophage expression of p-AMPK, p-ATF1, and downstream M2-like protective effects. CONCLUSION Metformin activates a conserved AMPK-ATF1-M2-like pathway in mouse and human macrophages, and results in highly suppressed atherogenesis in hyperlipidaemic mice via haematopoietic AMPK.
Collapse
Affiliation(s)
| | - Luke Cave
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Gareth Hyde
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - David Carling
- MRC London Institute of Medical Sciences, Imperial College London, UK
| | - Justin C Mason
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Dorian O Haskard
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
17
|
Fasolo F, Di Gregoli K, Maegdefessel L, Johnson JL. Non-coding RNAs in cardiovascular cell biology and atherosclerosis. Cardiovasc Res 2020; 115:1732-1756. [PMID: 31389987 DOI: 10.1093/cvr/cvz203] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/14/2019] [Accepted: 08/05/2019] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis underlies the predominant number of cardiovascular diseases and remains a leading cause of morbidity and mortality worldwide. The development, progression and formation of clinically relevant atherosclerotic plaques involves the interaction of distinct and over-lapping mechanisms which dictate the roles and actions of multiple resident and recruited cell types including endothelial cells, vascular smooth muscle cells, and monocyte/macrophages. The discovery of non-coding RNAs (ncRNAs) including microRNAs, long non-coding RNAs, and circular RNAs, and their identification as key mechanistic regulators of mRNA and protein expression has piqued interest in their potential contribution to atherosclerosis. Accruing evidence has revealed ncRNAs regulate pivotal cellular and molecular processes during all stages of atherosclerosis including cell invasion, growth, and survival; cellular uptake and efflux of lipids, expression and release of pro- and anti-inflammatory intermediaries, and proteolytic balance. The expression profile of ncRNAs within atherosclerotic lesions and the circulation have been determined with the aim of identifying individual or clusters of ncRNAs which may be viable therapeutic targets alongside deployment as biomarkers of atherosclerotic plaque progression. Consequently, numerous in vivo studies have been convened to determine the effects of moderating the function or expression of select ncRNAs in well-characterized animal models of atherosclerosis. Together, clinicopathological findings and studies in animal models have elucidated the multifaceted and frequently divergent effects ncRNAs impose both directly and indirectly on the formation and progression of atherosclerosis. From these findings' potential novel therapeutic targets and strategies have been discovered which may pave the way for further translational studies and possibly taken forward for clinical application.
Collapse
Affiliation(s)
- Francesca Fasolo
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar-Technical University Munich, Biedersteiner Strasse 29, Munich, Germany
| | - Karina Di Gregoli
- Laboratory of Cardiovascular Pathology, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar-Technical University Munich, Biedersteiner Strasse 29, Munich, Germany.,Molecular Vascular Medicine, Karolinska Institute, Center for Molecular Medicine L8:03, 17176 Stockholm, Sweden.,German Center for Cardiovascular Research (DZHK), Partner Site Munich (Munich Heart Alliance), Munich, Germany
| | - Jason L Johnson
- Laboratory of Cardiovascular Pathology, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
18
|
Chen G, Gao J, Sheng Y, Han X, Ji X, Zhao M, Wu J. Diagnostic value of miR-92a in asymptomatic carotid artery stenosis patients and its ability to predict cerebrovascular events. Diagn Pathol 2020; 15:74. [PMID: 32522208 PMCID: PMC7285548 DOI: 10.1186/s13000-020-00987-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 05/28/2020] [Indexed: 03/13/2023] Open
Abstract
Background Early diagnosis of asymptomatic carotid artery stenosis (ACAS) is important to prevent the incidence of cerebrovascular events. This study aimed to investigate the circulating expression of microRNA-92a (miR-92a) in ACAS patients and evaluate its diagnostic value for ACAS and predictive value for cerebrovascular events. Methods Circulating expression of miR-92a was measured using quantitative real-time PCR. Chi-square test was used to analyze the association of miR-92a with ACAS patients’ clinical characteristics. A receiver operating characteristic (ROC) was used to evaluate the diagnostic value of miR-92a, and the Kaplan-Meier method and Cox regression analysis were used to assess the predictive value of miR-92a for cerebrovascular events. Results Serum expression of miR-92a was higher in ACAS patients than that in the healthy controls (P < 0.001), and associated with patients’ degree of carotid stenosis (P = 0.013). The elevated miR-92a expression could distinguish ACAS patients from healthy individual, and was an independent predictive factor for the occurrence of cerebrovascular events (P = 0.015). Conclusion The data from this study indicated that circulating increased miR-92a may serve as a noninvasive diagnostic biomarker for ACAS and a potential risk factor for the future onset of cerebrovascular events.
Collapse
Affiliation(s)
- Gang Chen
- Department of Vascular Interventional, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Jianwei Gao
- Department of Vascular Interventional, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Yuguo Sheng
- Department of Vascular Interventional, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Xinqiang Han
- Department of Vascular Interventional, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Xingang Ji
- Department of Vascular Interventional, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Mengpeng Zhao
- Department of Vascular Interventional, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Jian Wu
- Department of Vascular Interventional, Binzhou Medical University Hospital, Binzhou, 256603, China.
| |
Collapse
|
19
|
ALK5 deficiency inhibits macrophage inflammation and lipid loading by targeting KLF4. Biosci Rep 2020; 40:222146. [PMID: 32065217 PMCID: PMC7056445 DOI: 10.1042/bsr20194188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 11/17/2022] Open
Abstract
The transforming growth factor type-β (TGF-β) has been demonstrated to play an important role in the development of atherosclerosis through binding to the serine/threonine kinase transmembrane type I and type II receptors. However, as a key type I receptor for TGF-β, the exact role and the underlying mechanism of Activin receptor-like kinase 5 (ALK5) on macrophage activation involved in atherogenesis remain unclear. In the present study, enhanced ALK5 expression was found in bone marrow derived macrophages (BMDMs) upon OX-LDL stimulation tested by RT-PCR and Western blot, which was further verified by co-immunofluorescence staining. Next, the loss-of-function of ALK5 used AdshALK5 transfection was performed to test the effect of ALK5 on macrophage activation. We observed that ALK5 silencing inhibited pro-inflammatory but promoted anti-inflammatory macrophage markers expression. Moreover, decreased foam cell formation was found in ALK5 knockdown macrophages accompanied by increased cholesterol efflux. Mechanistically, ALK5 knockdown significantly increased KLF4 expression that was responsible for the attenuated macrophage activation induced by ALK5 knockdown. Collectively, these findings suggested that neutralization of ALK5 may act as a promising strategy for the management of atherosclerosis.
Collapse
|
20
|
Gong FH, Xiao XQ, Zhang XP, Long L, Huang S, Wang XS, Shu ZL, Yang YS. Association Between Unstable Angina and CXCL17: a New Potential Biomarker. Open Med (Wars) 2020; 14:939-944. [PMID: 31934638 PMCID: PMC6947758 DOI: 10.1515/med-2019-0080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis and chemokines are strongly related, but the role of the chemokine CXCL17 in atherogenesis is still poorly understood. We aim to investigate the serum CXCL17 levels in different stages of patients with coronary heart disease and explore whether these differences contribute to atherosclerosis. In the current prospective study, we enrolled 48 patients with unstable angina (UA), 51 patients with stable angina (SA) and 41 patients for the control group (CG). All subjects were diagnosed by coronary angiography and Gensini score was used to evaluate the severity of coronary artery disease. The CXCL17 levels were determined using ELISA, while lipid metabolism indicators and high sensitivity C-reactive protein (hs-CRP) were detected by automatic biochemical analyzer. We observed that the unstable angina group had higher CXCL17 levels compared with the stable angina and the control group. The logistic regression analysis showed that CXCL17 was an independent risk factor for unstable angina. Our results showed that CXCL17 was also statistically correlated with hs-CRP, while it was irrelevant with Gensini score. CXCL17 levels were associated with activity of inflammatory response and the instability of atherosclerotic plaques. These results suggest that CXCL17 elevation may be a potential new biomarker of unstable angina.
Collapse
Affiliation(s)
- Fu-Han Gong
- Department of Cardiology, Tongren Municipal People's Hospital, No.120 Taoyuan Avenue, Chuandong Education park, Bijiang District, Tongren City 554300, China
| | - Xiao-Qiang Xiao
- Department of Cardiology, Tongren Municipal People's Hospital, No.120 Taoyuan Avenue, Chuandong Education park, Bijiang District, Tongren City 554300, China
| | - Xue-Ping Zhang
- Department of Cardiology, Tongren Municipal People's Hospital, No.120 Taoyuan Avenue, Chuandong Education park, Bijiang District, Tongren City 554300, China
| | - Li Long
- Department of Clinical Laboratory, Tongren Municipal People's Hospital, Tongren 554300, China
| | - Sheng Huang
- Department of Ophthalmology, Tongren Municipal People's Hospital, Tongren 554300, China
| | - Xue-Sheng Wang
- Department of Cardiology, Tongren Municipal People's Hospital, No.120 Taoyuan Avenue, Chuandong Education park, Bijiang District, Tongren City 554300, China
| | - Zhen-Lin Shu
- Department of Cardiology, Tongren Municipal People's Hospital, No.120 Taoyuan Avenue, Chuandong Education park, Bijiang District, Tongren City 554300, China
| | - Yong-Sheng Yang
- Department of Cardiology, Tongren Municipal People's Hospital, No.120 Taoyuan Avenue, Chuandong Education park, Bijiang District, Tongren City 554300, China
| |
Collapse
|
21
|
MicroRNA-150 deficiency accelerates intimal hyperplasia by acting as a novel regulator of macrophage polarization. Life Sci 2020; 240:116985. [DOI: 10.1016/j.lfs.2019.116985] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 10/11/2019] [Accepted: 10/17/2019] [Indexed: 11/18/2022]
|
22
|
Cai T, Cui X, Zhang K, Zhang A, Liu B, Mu JJ. LncRNA TNK2-AS1 regulated ox-LDL-stimulated HASMC proliferation and migration via modulating VEGFA and FGF1 expression by sponging miR-150-5p. J Cell Mol Med 2019; 23:7289-7298. [PMID: 31468685 PMCID: PMC6815783 DOI: 10.1111/jcmm.14575] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/24/2019] [Accepted: 06/29/2019] [Indexed: 12/12/2022] Open
Abstract
Long non‐coding RNAs (lncRNAs) have been indicated for the regulatory roles in cardiovascular diseases. This study determined the expression of lncRNA TNK2 antisense RNA 1 (TNK2‐AS1) in oxidized low‐density lipoprotein (ox‐LDL)‐stimulated human aortic smooth muscle cells (HASMCs) and examined the mechanistic role of TNK2‐AS1 in the proliferation and migration of HASMCs. Our results demonstrated that ox‐LDL promoted HASMC proliferation and migration, and the enhanced proliferation and migration in ox‐LDL‐treated HASMCs were accompanied by the up‐regulation of TNK2‐AS1. In vitro functional studies showed that TNK2‐AS1 knockdown suppressed cell proliferation and migration of ox‐LDL‐stimulated HASMCs, while TNK2‐AS1 overexpression enhanced HASMC proliferation and migration. Additionally, TNK2‐AS1 inversely regulated miR‐150‐5p expression via acting as a competing endogenous RNA (ceRNA), and the enhanced effects of TNK2‐AS1 overexpression on HASMC proliferation and migration were attenuated by miR‐150‐5p overexpression. Moreover, miR‐150‐5p could target the 3’ untranslated regions of vascular endothelial growth factor A (VEGFA) and fibroblast growth factor 1 (FGF1) to regulate FGF1 and VEGFA expression in HASMCs, and the inhibitory effects of miR‐150‐5p overexpression in ox‐LDL‐stimulated HASMCs were attenuated by enforced expression of VEGFA and FGF1. Enforced expression of VEGFA and FGF1 also partially restored the suppressed cell proliferation and migration induced by TNK2‐AS1 knockdown in ox‐LDL‐stimulated HASMCs, while the enhanced effects of TNK2‐AS1 overexpression on HASMC proliferation and migration were attenuated by the knockdown of VEGFA and FGF1. Collectively, our findings showed that TNK2‐AS1 exerted its action in ox‐LDL‐stimulated HASMCs via regulating VEGFA and FGF1 expression by acting as a ceRNA for miR‐150‐5p.
Collapse
Affiliation(s)
- Tianzhi Cai
- Department of Cardiology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Cardiology, the First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Xiuzhen Cui
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Kelin Zhang
- Department of Cardiology, the First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Anji Zhang
- Department of Cardiology, the First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Baixue Liu
- Department of Cardiology, the First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jian-Jun Mu
- Department of Cardiology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
23
|
Analysis of the microRNA Expression Profile of Bovine Monocyte-derived Macrophages Infected with Mycobacterium avium subsp. Paratuberculosis Reveals that miR-150 Suppresses Cell Apoptosis by Targeting PDCD4. Int J Mol Sci 2019; 20:ijms20112708. [PMID: 31159463 PMCID: PMC6600136 DOI: 10.3390/ijms20112708] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/21/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
M. avium subsp. paratuberculosis (MAP) is the causative pathogen of Johne’s disease, a chronic granulomatous enteritis that principally affects ruminants and can survive, proliferate and disseminate in macrophages. MicroRNAs (miRNAs) are important regulators of gene expression and can impact the processes of cells. To investigate the role of miRNAs in monocyte-derived macrophages (MDMs) during MAP infection, we used high-throughput sequencing technology to analyze small RNA libraries of MAP-infected and control MDMs. The results showed that a total of 21 miRNAs were differentially expressed in MDMs after MAP infection, and 8864 target genes were predicted. A functional analysis showed that the target genes were mainly involved in the MAPK signaling pathway, Toll-like receptor signaling pathway, NF-kappa B signaling pathway and apoptosis. In addition, using a dual-luciferase reporter assay, flow cytometry, and a small interfering (si)RNA knockdown assay, the role of miR-150 in regulating macrophage apoptosis by targeting the programmed cell death protein-4 (PDCD4) was demonstrated. These results provide an experimental basis to reveal the regulatory mechanism of MAP infection and suggest the potential of miRNAs as biomarkers for the diagnosis of Johne’s disease in bovines.
Collapse
|
24
|
Affiliation(s)
| | - Alan Daugherty
- Graduate Center for Nutritional Sciences
- Saha Cardiovascular Research Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Hong Lu
- Graduate Center for Nutritional Sciences
- Saha Cardiovascular Research Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
- Hong Lu, MD, PhD, Saha Cardiovascular Research Center , University of Kentucky, 741 South Limestone, BBSRB, Room 249, Lexington, KY 40536-0509, USA, Phone: +1 859 323 4639, Fax: +1 859 257 3235,
| |
Collapse
|
25
|
MiR-150 predicts survival in patients with sepsis and inhibits LPS-induced inflammatory factors and apoptosis by targeting NF-κB1 in human umbilical vein endothelial cells. Biochem Biophys Res Commun 2018; 500:828-837. [PMID: 29689269 DOI: 10.1016/j.bbrc.2018.04.168] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 12/14/2022]
Abstract
MiR-150 is involved into some pathological processes, such as tumorigenesis and autoimmune diseases. However, little is known about the involvement of miR-150 in human sepsis. In this study, plasma miR-150 level had a diagnostic and independent prognostic value in patients with sepsis, and negatively correlated with renal dysfunction and 28-day survival as well as plasma levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α). MiR-150 expression was also significantly decreased in human umbilical vein endothelial cells (HUVECs) and C57BL/6 mice with sepsis after lipopolysaccharides (LPS) treatment. In-vitro, miR-150 over-expression protected HUVECs from LPS-induced apoptosis and the expressions of nuclear factor-κB1 (NF-κB1), IL-6, TNF-α, intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and E-selectin. Furthermore, NF-κB1 was identified as a direct target of miR-150. Restored NF-κB1 expression antagonized the protective effects of miR-150, while suppression of NF-κB1 enhanced these protective effects. Our findings indicate miR-150 predicts survival in patients with sepsis and inhibits LPS-induced inflammatory factors and apoptosis by targeting NF-κB1 in human umbilical vein endothelial cells. Thus, miR-150 may be a useful biomarker or target in the diagnosis, prognosis and treatment of patients with sepsis.
Collapse
|