1
|
Cao W, Zhao B, Gui L, Sun X, Zhang Z, Huang L. The role and mechanism of action of miR‑92a in endothelial cell autophagy. Mol Med Rep 2024; 30:172. [PMID: 39054957 PMCID: PMC11304162 DOI: 10.3892/mmr.2024.13296] [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: 03/07/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Although microRNAs (miRNAs/miRs) serve a significant role in the autophagy of vascular endothelial cells (ECs), the effect of miR‑92a on the autophagy of ECs is currently unclear. Therefore, the present study aimed to investigate the impact of miR‑92a on autophagy in ECs and the underlying molecular processes that control this biological activity. Firstly, an autophagy model of EA.hy926 cells was generated via treatment with the autophagy inducer rapamycin (rapa‑EA.hy926 cells). The expression levels of miR‑92a were then detected by reverse transcription‑quantitative PCR, and the effect of miR‑92a expression on the autophagic activity of rapa‑EA.hy926 cells was studied by overexpressing or inhibiting miR‑92a. The level of autophagy was evaluated by western blot analysis, immunofluorescence staining and transmission electron microscopy. Dual‑luciferase reporter assays were used to confirm the interaction between miR‑92a and FOXO3. The results demonstrated that the expression levels of miR‑92a were decreased in the rapa‑EA.hy926 cell autophagy model. Furthermore, overexpression and inhibition of miR‑92a revealed that upregulation of miR‑92a in these cells inhibited autophagy, whereas miR‑92a knockdown promoted it. It was also confirmed that miR‑92a directly bound to the 3'‑untranslated region of the autophagy‑related gene FOXO3 and reduced its expression. In conclusion, the present study suggested that miR‑92a inhibits autophagy activity in EA.hy926 cells by targeting FOXO3.
Collapse
Affiliation(s)
- Weili Cao
- Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Boxin Zhao
- Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Lin Gui
- Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xueyuan Sun
- Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Zhiyong Zhang
- Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Lijuan Huang
- Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
2
|
Xu M, Pang M, Wang C, An N, Chen R, Bai Y, He J, Wang C, Qi Y. MiR-92a-3p Knockdown Attenuates Transforming Growth Factor-β1-induced Tubulointerstitial Fibrosis by Targeting LIN28A-mediated EMT Pathway. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2024; 67:198-206. [PMID: 39148295 DOI: 10.4103/ejpi.ejpi-d-24-00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/11/2024] [Indexed: 08/17/2024]
Abstract
ABSTRACT The role of microRNAs in regulating tubulointerstitial fibrosis, a key feature of progressive chronic kidney disease, is of significant importance. LIN28A has been reported to attenuate renal fibrosis in obstructive nephropathy. Here, our objective was to investigate the precise biological function of the miR-92a-3p/LIN28A axis in tubulointerstitial fibrosis. The human renal proximal tubular epithelial (HK-2) cell line was exposed to transforming growth factor (TGF)-β1, establishing an in vitro model mimicking tubulointerstitial fibrosis. Luciferase reporter assay was utilized to investigate the relationship between miR-92a-3p and LIN28A. Cell transfection techniques were employed to modify the expression of miR-92a-3p and LIN28A. An in vivo model of tubulointerstitial fibrosis was created by inducing unilateral ureteral obstruction (UUO) in C57BL/6N mice. Our initial observations showed that TGF-β1 treatment of HK-2 cells and the UUO mice model led to an increase in miR-92a-3p expression and a decrease in LIN28A expression. We confirmed that miR-92a-3p directly targeted LIN28A in HK-2 cells. In TGF-β1-stimulated HK-2 cells, knocking down miR-92a-3p notably reduced the levels of alpha smooth muscle actin and vimentin and concurrently enhanced the expression of E-cadherin. These changes were counteracted upon transfection with si-LIN28A. Thus, directing interventions toward miR-92a-3p holds the potential to emerge as a viable therapeutic approach for addressing tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Mingzhi Xu
- Blood Purification Center, Hainan General Hospital, Hai-nan Affiliated Hospital of Hainan Medical University, Haikou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Li X, Peng X, Zoulikha M, Boafo GF, Magar KT, Ju Y, He W. Multifunctional nanoparticle-mediated combining therapy for human diseases. Signal Transduct Target Ther 2024; 9:1. [PMID: 38161204 PMCID: PMC10758001 DOI: 10.1038/s41392-023-01668-1] [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: 11/30/2022] [Revised: 09/14/2023] [Accepted: 10/10/2023] [Indexed: 01/03/2024] Open
Abstract
Combining existing drug therapy is essential in developing new therapeutic agents in disease prevention and treatment. In preclinical investigations, combined effect of certain known drugs has been well established in treating extensive human diseases. Attributed to synergistic effects by targeting various disease pathways and advantages, such as reduced administration dose, decreased toxicity, and alleviated drug resistance, combinatorial treatment is now being pursued by delivering therapeutic agents to combat major clinical illnesses, such as cancer, atherosclerosis, pulmonary hypertension, myocarditis, rheumatoid arthritis, inflammatory bowel disease, metabolic disorders and neurodegenerative diseases. Combinatorial therapy involves combining or co-delivering two or more drugs for treating a specific disease. Nanoparticle (NP)-mediated drug delivery systems, i.e., liposomal NPs, polymeric NPs and nanocrystals, are of great interest in combinatorial therapy for a wide range of disorders due to targeted drug delivery, extended drug release, and higher drug stability to avoid rapid clearance at infected areas. This review summarizes various targets of diseases, preclinical or clinically approved drug combinations and the development of multifunctional NPs for combining therapy and emphasizes combinatorial therapeutic strategies based on drug delivery for treating severe clinical diseases. Ultimately, we discuss the challenging of developing NP-codelivery and translation and provide potential approaches to address the limitations. This review offers a comprehensive overview for recent cutting-edge and challenging in developing NP-mediated combination therapy for human diseases.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Xiuju Peng
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Makhloufi Zoulikha
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - George Frimpong Boafo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, PR China
| | - Kosheli Thapa Magar
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China
| | - Yanmin Ju
- School of Pharmacy, China Pharmaceutical University, Nanjing, 2111198, PR China.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China.
| |
Collapse
|
4
|
Wu KL, Chen CL, Thi Nguyen MH, Tsai JC, Wang SC, Chiang WF, Hsiao PJ, Chan JS, Hou JJ, Ma N. MicroRNA regulators of vascular pathophysiology in chronic kidney disease. Clin Chim Acta 2023; 551:117610. [PMID: 37863246 DOI: 10.1016/j.cca.2023.117610] [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: 08/19/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Coronary artery disease (CAD) is a severe comorbidity in chronic kidney disease (CKD) due to heavy calcification in the medial layer and inflamed plaques. Chronic inflammation, endothelial dysfunction and vascular calcification are major contributors that lead to artherosclerosis in CKD. The lack of specific symptoms and signs of CAD and decreased accuracy of noninvasive diagnostic tools result in delayed diagnosis leading to increased mortality. MicroRNAs (miRNAs) are post-transcriptional regulators present in various biofluids throughout the body. In the circulation, miRNAs have been reported to be encapsulated in extracellular vesicles and serve as stable messengers for crosstalk among cells. miRNAs are involved in pathophysiologic mechanisms including CAD and can potentially be extended from basic research to clinical translational practice.
Collapse
Affiliation(s)
- Kun-Lin Wu
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Lung Chen
- Division of Nephrology, Department of Medicine, Landseed International Hospital, Taoyuan, Taiwan
| | - Mai-Huong Thi Nguyen
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Jen-Chieh Tsai
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan; Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Sun-Chong Wang
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Wen-Fang Chiang
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Jen Hsiao
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jenq-Shyong Chan
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ju Jung Hou
- Kaohsiung Medical University Hospital, Department of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Nianhan Ma
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan.
| |
Collapse
|
5
|
Sarakpi T, Mesic A, Speer T. Leukocyte-endothelial interaction in CKD. Clin Kidney J 2023; 16:1845-1860. [PMID: 37915921 PMCID: PMC10616504 DOI: 10.1093/ckj/sfad135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Indexed: 11/03/2023] Open
Abstract
Chronic kidney disease (CKD) represents an independent risk factor for cardiovascular diseases (CVD). Accordingly, CKD patients show a substantial increased risk of cardiovascular mortality. Inflammation represents an important link between CKD and CVD. The interaction between endothelial cells and effector cells of the innate immune system plays a central role in the development and progression of inflammation. Vascular injury causes endothelial dysfunction, leading to augmented oxidative stress, increased expression of leukocyte adhesion molecules and chronic inflammation. CKD induces numerous metabolic changes, creating a uremic milieu resulting in the accumulation of various uremic toxins. These toxins lead to vascular injury, endothelial dysfunction and activation of the innate immune system. Recent studies describe CKD-dependent changes in monocytes that promote endothelial dysfunction and thus CKD progression and CKD-associated CVD. The NLR family pyrin domain containing 3-interleukin-1β-interleukin-6 (NLRP3-IL-1β-IL-6) signaling pathway plays a pivotal role in the development and progression of CVD and CKD alike. Several clinical trials are investigating targeted inhibition of this pathway indicating that anti-inflammatory therapeutic strategies may emerge as novel approaches in patients at high cardiovascular risk and nonresolving inflammation. CKD patients in particular would benefit from targeted anti-inflammatory therapy, since conventional therapeutic regimens have limited efficacy in this population.
Collapse
Affiliation(s)
- Tamim Sarakpi
- Department of Internal Medicine 4 – Nephrology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Else Kröner-Fresenius-Zentrum for Nephrological Research, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Armir Mesic
- Department of Internal Medicine 4 – Nephrology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Else Kröner-Fresenius-Zentrum for Nephrological Research, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Thimoteus Speer
- Department of Internal Medicine 4 – Nephrology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Else Kröner-Fresenius-Zentrum for Nephrological Research, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
6
|
Zhang L, Zhang W, He L, Cui H, Wang Y, Wu X, Zhao X, Yan P, Yang C, Xiao C, Tang M, Chen L, Xiao C, Zou Y, Liu Y, Yang Y, Zhang L, Yao Y, Li J, Liu Z, Yang C, Jiang X, Zhang B. Impact of gallstone disease on the risk of stroke and coronary artery disease: evidence from prospective observational studies and genetic analyses. BMC Med 2023; 21:353. [PMID: 37705021 PMCID: PMC10500913 DOI: 10.1186/s12916-023-03072-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Despite epidemiological evidence associating gallstone disease (GSD) with cardiovascular disease (CVD), a dilemma remains on the role of cholecystectomy in modifying the risk of CVD. We aimed to characterize the phenotypic and genetic relationships between GSD and two CVD events - stroke and coronary artery disease (CAD). METHODS We first performed a meta-analysis of cohort studies to quantify an overall phenotypic association between GSD and CVD. We then investigated the genetic relationship leveraging the largest genome-wide genetic summary statistics. We finally examined the phenotypic association using the comprehensive data from UK Biobank (UKB). RESULTS An overall significant effect of GSD on CVD was found in meta-analysis (relative risk [RR] = 1.26, 95% confidence interval [CI] = 1.19-1.34). Genetically, a positive shared genetic basis was observed for GSD with stroke ([Formula: see text]=0.16, P = 6.00 × 10-4) and CAD ([Formula: see text]=0.27, P = 2.27 × 10-15), corroborated by local signals. The shared genetic architecture was largely explained by the multiple pleiotropic loci identified in cross-phenotype association study and the shared gene-tissue pairs detected by transcriptome-wide association study, but not a causal relationship (GSD to CVD) examined through Mendelian randomization (MR) (GSD-stroke: odds ratio [OR] = 1.00, 95%CI = 0.97-1.03; GSD-CAD: OR = 1.01, 95%CI = 0.98-1.04). After a careful adjustment of confounders or considering lag time using UKB data, no significant phenotypic effect of GSD on CVD was detected (GSD-stroke: hazard ratio [HR] = 0.95, 95%CI = 0.83-1.09; GSD-CAD: HR = 0.98, 95%CI = 0.91-1.06), further supporting MR findings. CONCLUSIONS Our work demonstrates a phenotypic and genetic relationship between GSD and CVD, highlighting a shared biological mechanism rather than a direct causal effect. These findings may provide insight into clinical and public health applications.
Collapse
Affiliation(s)
- Li Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Wenqiang Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Lin He
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Huijie Cui
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yutong Wang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Xueyao Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Xunying Zhao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Peijing Yan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Chao Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Changfeng Xiao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Mingshuang Tang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Lin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Chenghan Xiao
- Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yanqiu Zou
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yunjie Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yanfang Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Ling Zhang
- Department of Iatrical Polymer Material and Artificial Apparatus, School of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Yuqin Yao
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Zhenmi Liu
- Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China.
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Ben Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, and West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China.
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Dlouha D, Blaha M, Huckova P, Lanska V, Hubacek JA, Blaha V. Long-Term LDL-Apheresis Treatment and Dynamics of Circulating miRNAs in Patients with Severe Familial Hypercholesterolemia. Genes (Basel) 2023; 14:1571. [PMID: 37628623 PMCID: PMC10454435 DOI: 10.3390/genes14081571] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Lipoprotein apheresis (LA) is a therapeutic option for patients with severe hypercholesterolemia who have persistently elevated LDL-C levels despite attempts at drug therapy. MicroRNAs (miRNAs), important posttranscriptional gene regulators, are involved in the pathogenesis of atherosclerosis. Our study aimed to monitor the dynamics of twenty preselected circulating miRNAs in patients under long-term apheresis treatment. Plasma samples from 12 FH patients (men = 50%, age = 55.3 ± 12.2 years; mean LA overall treatment time = 13.1 ± 7.8 years) were collected before each apheresis therapy every sixth month over the course of four years of treatment. Eight complete follow-up (FU) samples were measured in each patient. Dynamic changes in the relative quantity of 6 miRNAs (miR-92a, miR-21, miR-126, miR-122, miR-26a, and miR-185; all p < 0.04) during FU were identified. Overall apheresis treatment time influenced circulating miR-146a levels (p < 0.04). In LDLR mutation homozygotes (N = 5), compared to heterozygotes (N = 7), we found higher plasma levels of miR-181, miR-126, miR-155, and miR-92a (all p < 0.03). Treatment with PCSK9 inhibitors (N = 6) affected the plasma levels of 7 miRNAs (miR-126, miR-122, miR-26a, miR-155, miR-125a, miR-92a, and miR-27a; all p < 0.04). Long-term monitoring has shown that LA in patients with severe familial hypercholesterolemia influences plasma circulating miRNAs involved in endothelial dysfunction, cholesterol homeostasis, inflammation, and plaque development. The longer the treatment using LA, the better the miRNA milieu depicting the potential cardiovascular risk.
Collapse
Affiliation(s)
- Dana Dlouha
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (P.H.); (J.A.H.)
| | - Milan Blaha
- 4th Department of Internal Medicine—Hematology, University Hospital Hradec Králové, 50005 Hradec Králové, Czech Republic;
- Faculty of Medicine in Hradec Králové, Charles University, 50003 Hradec Králové, Czech Republic;
| | - Pavlina Huckova
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (P.H.); (J.A.H.)
| | - Vera Lanska
- Statistical Unit, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic;
| | - Jaroslav Alois Hubacek
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (P.H.); (J.A.H.)
- 1st Faculty of Medicine, Charles University, 12108 Prague, Czech Republic
| | - Vladimir Blaha
- Faculty of Medicine in Hradec Králové, Charles University, 50003 Hradec Králové, Czech Republic;
- 3rd Department of Internal Medicine—Metabolism and Gerontology, University Hospital Hradec Králové, 50005 Hradec Králové, Czech Republic
| |
Collapse
|
8
|
Ejaz M, Usman SM, Amir S, Khan MJ. Holistic expression of miR-17-92 cluster in obesity, kidney diseases, cardiovascular diseases, and diabetes. Mol Biol Rep 2023; 50:6913-6925. [PMID: 37329480 DOI: 10.1007/s11033-023-08549-4] [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: 04/09/2023] [Accepted: 05/24/2023] [Indexed: 06/19/2023]
Abstract
miR-17-92 cluster encodes six micro RNAs (miRNAs) and plays a crucial role in the regulation of various cellular processes. Aberrant expression of this cluster may result in the onset of several diseases. Initially, the role of miR-17-92 cluster in tumorigenesis was discovered but recent research has also uncovered its role in other diseases. Members of the cluster may serve as potential biomarkers in the prognosis, diagnosis, and treatment of several diseases and their complications. In this article, we have reviewed the recent research carried out on the expression pattern of miR-17-92 cluster in non-communicable diseases i.e., obesity, cardiovascular diseases (CVD), kidney diseases (KD) and diabetes mellitus (DM). We examined miR-17-92 role in pathological processes and their potential importance as biomarkers. Each member of the cluster miR-17-92 was upregulated in obesity. miR-18a, miR-19b-3p, miR20a, and miR92a were significantly upregulated in CVD. An equal fraction of the cluster was dysregulated (upregulated and downregulated) in diabetes; however, miR-17-92 was downregulated in most studies on CKD.
Collapse
Affiliation(s)
- Maheen Ejaz
- Department of Biosciences, COMSATS University Islamabad, Park Road, Chak Shahzad Islamabad, Islamabad, 45550, Pakistan
| | - Syed Mohammad Usman
- Department of Biochemistry, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Saira Amir
- Department of Biosciences, COMSATS University Islamabad, Park Road, Chak Shahzad Islamabad, Islamabad, 45550, Pakistan
| | - Muhammad Jawad Khan
- Department of Biosciences, COMSATS University Islamabad, Park Road, Chak Shahzad Islamabad, Islamabad, 45550, Pakistan.
| |
Collapse
|
9
|
Liu P, Wang S, Li K, Yang Y, Man Y, Du F, Wang L, Tian J, Su G. Exosomal microRNA‑4516, microRNA‑203 and SFRP1 are potential biomarkers of acute myocardial infarction. Mol Med Rep 2023; 27:124. [PMID: 37203392 PMCID: PMC10206682 DOI: 10.3892/mmr.2023.13010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/20/2023] [Indexed: 05/20/2023] Open
Abstract
Acute myocardial infarction (AMI) is a serious disease which threatens public health. Exosomes (exos) contain certain genetic information and are important communication vehicles between cells. In the present study, different exosomal microRNAs (miRs), which exhibit a notable association between expression levels in plasma and AMI were assessed to support the development of new diagnostic and clinical assessment markers of patients with AMI. In total, 93 individuals, including 31 healthy controls and 62 patients with AMI, were recruited for the present study. Data on age, blood pressure, glucose levels, lipid levels and coronary angiography images were collected from the enrolled individuals, and plasma samples were collected. Plasma exos were extracted and verified using ultracentrifugation, transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and western blotting (WB). Exo‑miR‑4516 and exo‑miR‑203 in plasma exos were identified by exosomal miRNA sequencing analysis, reverse transcription‑quantitative PCR was performed to detect the levels of exo‑miR‑4516 and exo‑miR‑203 in plasma exos, and ELISA was performed to detect the levels of secretory frizzled‑related protein 1 (SFRP1) in samples. The correlation analysis between exo‑miR‑4516, exo‑miR‑203 and SFRP1 in plasma exos and AMI was presented as receiver operating characteristic curves (ROCs) of the SYNTAX score, cardiac troponin I (cTnI), low‑density lipoprotein (LDL) and each indicator separately. Kyoto Encyclopedia of Genes and Genomes enrichment analysis was performed to predict relevant enrichment pathways. Exos were successfully isolated from plasma by ultracentrifugation, which was confirmed by TEM, NTA and WB. Exo‑miR‑4516, exo‑miR‑203 and SFRP1 levels in plasma were significantly higher in the AMI group compared with the healthy control group. ROCs demonstrated that exo‑miR‑4516, exo‑miR‑203 and SFRP1 levels had a high diagnostic efficiency in predicting AMI. Exo‑miR‑4516 was positively correlated with SYNTAX score, and plasma SFRP1 was positively correlated with plasma cTnI and LDL. In conclusion, the data demonstrated that exo‑miR‑4516, exo‑miR‑203 and SFRP1 levels could be used in combination to diagnose and assess the severity of AMI. The present study was retrospectively registered (TRN, NCT02123004).
Collapse
Affiliation(s)
- Peng Liu
- Department of Cardiovascular Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan Central Hospital, Jinan, Shandong 250000, P.R. China
| | - Shuya Wang
- Department of Cardiovascular Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan Central Hospital, Jinan, Shandong 250000, P.R. China
| | - Kaiyuan Li
- Dalian Medical University, Dalian, Liaoning 116000, P.R. China
| | - Yang Yang
- Department of Cardiovascular Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Yilong Man
- Department of Cardiovascular Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan Central Hospital, Jinan, Shandong 250000, P.R. China
| | - Fengli Du
- Department of Cardiovascular Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan Central Hospital, Jinan, Shandong 250000, P.R. China
- Department of Cardiovascular Medicine, Shandong Provincial Public Health Centre, Jinan, Shandong 250000, P.R. China
| | - Lei Wang
- Department of Cardiovascular Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan Central Hospital, Jinan, Shandong 250000, P.R. China
| | - Jing Tian
- Department of Cardiovascular Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan Central Hospital, Jinan, Shandong 250000, P.R. China
| | - Guohai Su
- Department of Cardiovascular Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan Central Hospital, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
10
|
Feng Y, Lei B, Zhang H, Niu L, Li X, Luo X, Zhang F. Long noncoding RNA TUG1 induces angiogenesis of endothelial progenitor cells and dissolution of deep vein thrombosis. Thromb J 2022; 20:54. [PMID: 36163177 PMCID: PMC9511754 DOI: 10.1186/s12959-022-00413-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/03/2022] [Indexed: 03/09/2024] Open
Abstract
Objective Long non-coding RNA (lncRNA) essentially controls many physiological and pathological processes of deep vein thrombosis (DVT). Based on that, lncRNA taurine upregulated gene 1 (TUG1)-involved angiogenesis of endothelial progenitor cells (EPCs) and dissolution of DVT was explored. Methods In the in-vitro experiments, EPCs were engineered with mimic, inhibitor, siRNA, and plasmid, after which tube formation, proliferation, migration, and apoptosis were checked. In the in-vivo experiments, a DVT mouse model was established. Before the DVT operation, the mice were injected with agomir, antagomir, siRNA, and plasmid. Subsequently, thrombosis and damage to the femoral vein were pathologically evaluated. TUG1, miR-92a-3p, and 3-Hydroxy-3-methylglutaryl coenzyme A reductase (Hmgcr) expression in the femoral vein was tested. The relationship between TUG1, miR-92a-3p, and Hmgcr was validated. Results DVT mice showed suppressed TUG1 and Hmgcr expression, and elevated miR-92a-3p expression. In EPCs, TUG1 overexpression or miR-92a-3p inhibition promoted cellular angiogenesis, whereas Hmgcr silencing blocked cellular angiogenesis. In DVT mice, elevated TUG1 or inhibited miR-92a-3p suppressed thrombosis and damage to the femoral vein whilst Hmgcr knockdown acted oppositely. In both cellular and animal models, TUG1 overexpression-induced effects could be mitigated by miR-92a-3p up-regulation. Mechanically, TUG1 interacted with miR-92a-3p to regulate Hmgcr expression. Conclusion Evidently, TUG1 promotes the angiogenesis of EPCs and dissolution of DVT via the interplay with miR-92a-3p and Hmgcr. Supplementary Information The online version contains supplementary material available at 10.1186/s12959-022-00413-y.
Collapse
Affiliation(s)
- Yaping Feng
- Department of Vascular Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Bo Lei
- Anesthesia Department, Beijing Haidian Maternal & Child Health Hospital, No. 33 Haidian South Road, Haidian District, Beijing, 100080, China.
| | - Huan Zhang
- Department of Vascular Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Luyuan Niu
- Department of Vascular Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xiangtao Li
- Department of Vascular Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xiaoyun Luo
- Department of Vascular Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Fuxian Zhang
- Department of Vascular Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| |
Collapse
|
11
|
Cardiovascular Disease-Associated MicroRNAs as Novel Biomarkers of First-Trimester Screening for Gestational Diabetes Mellitus in the Absence of Other Pregnancy-Related Complications. Int J Mol Sci 2022; 23:ijms231810635. [PMID: 36142536 PMCID: PMC9501303 DOI: 10.3390/ijms231810635] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/25/2022] Open
Abstract
We assessed the diagnostic potential of cardiovascular disease-associated microRNAs for the early prediction of gestational diabetes mellitus (GDM) in singleton pregnancies of Caucasian descent in the absence of other pregnancy-related complications. Whole peripheral venous blood samples were collected within 10 to 13 weeks of gestation. This retrospective study involved all pregnancies diagnosed with only GDM (n = 121) and 80 normal term pregnancies selected with regard to equality of sample storage time. Gene expression of 29 microRNAs was assessed using real-time RT-PCR. Upregulation of 11 microRNAs (miR-1-3p, miR-20a-5p, miR-20b-5p, miR-23a-3p, miR-100-5p, miR-125b-5p, miR-126-3p, miR-181a-5p, miR-195-5p, miR-499a-5p, and miR-574-3p) was observed in pregnancies destinated to develop GDM. Combined screening of all 11 dysregulated microRNAs showed the highest accuracy for the early identification of pregnancies destinated to develop GDM. This screening identified 47.93% of GDM pregnancies at a 10.0% false positive rate (FPR). The predictive model for GDM based on aberrant microRNA expression profile was further improved via the implementation of clinical characteristics (maternal age and BMI at early stages of gestation and an infertility treatment by assisted reproductive technology). Following this, 69.17% of GDM pregnancies were identified at a 10.0% FPR. The effective prediction model specifically for severe GDM requiring administration of therapy involved using a combination of these three clinical characteristics and three microRNA biomarkers (miR-20a-5p, miR-20b-5p, and miR-195-5p). This model identified 78.95% of cases at a 10.0% FPR. The effective prediction model for GDM managed by diet only required the involvement of these three clinical characteristics and eight microRNA biomarkers (miR-1-3p, miR-20a-5p, miR-20b-5p, miR-100-5p, miR-125b-5p, miR-195-5p, miR-499a-5p, and miR-574-3p). With this, the model identified 50.50% of GDM pregnancies managed by diet only at a 10.0% FPR. When other clinical variables such as history of miscarriage, the presence of trombophilic gene mutations, positive first-trimester screening for preeclampsia and/or fetal growth restriction by the Fetal Medicine Foundation algorithm, and family history of diabetes mellitus in first-degree relatives were included in the GDM prediction model, the predictive power was further increased at a 10.0% FPR (72.50% GDM in total, 89.47% GDM requiring therapy, and 56.44% GDM managed by diet only). Cardiovascular disease-associated microRNAs represent promising early biomarkers to be implemented into routine first-trimester screening programs with a very good predictive potential for GDM.
Collapse
|
12
|
Hüsing AM, Wulfmeyer VC, Gaedcke S, Fleig SV, Rong S, DeLuca D, Haller H, Schmitt R, von Vietinghoff S. Myeloid CCR2 Promotes Atherosclerosis after AKI. J Am Soc Nephrol 2022; 33:1487-1500. [PMID: 35537780 PMCID: PMC9342642 DOI: 10.1681/asn.2022010048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/10/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The risk of cardiovascular events rises after AKI. Leukocytes promote atherosclerotic plaque growth and instability. We established a model of enhanced remote atherosclerosis after renal ischemia-reperfusion (IR) injury and investigated the underlying inflammatory mechanisms. METHODS Atherosclerotic lesions and inflammation were investigated in native and bone marrow-transplanted LDL receptor-deficient (LDLr-/- ) mice after unilateral renal IR injury using histology, flow cytometry, and gene expression analysis. RESULTS Aortic root atherosclerotic lesions were significantly larger after renal IR injury than in controls. A gene expression screen revealed enrichment for chemokines and their cognate receptors in aortas of IR-injured mice in early atherosclerosis, and of T cell-associated genes in advanced disease. Confocal microscopy revealed increased aortic macrophage proximity to T cells. Differential aortic inflammatory gene regulation in IR-injured mice largely paralleled the pattern in the injured kidney. Single-cell analysis identified renal cell types that produced soluble mediators upregulated in the atherosclerotic aorta. The analysis revealed a marked early increase in Ccl2, which CCR2+ myeloid cells mainly expressed. CCR2 mediated myeloid cell homing to the post-ischemic kidney in a cell-individual manner. Reconstitution with Ccr2-/- bone marrow dampened renal post-ischemic inflammation, reduced aortic Ccl2 and inflammatory macrophage marker CD11c, and abrogated excess aortic atherosclerotic plaque formation after renal IR. CONCLUSIONS Our data introduce an experimental model of remote proatherogenic effects of renal IR and delineate myeloid CCR2 signaling as a mechanistic requirement. Monocytes should be considered as mobile mediators when addressing systemic vascular sequelae of kidney injury.
Collapse
Affiliation(s)
- Anne M. Hüsing
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Vera C. Wulfmeyer
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Svenja Gaedcke
- German Centre for Lung Research, Hannover Medical School, Hannover, Germany
| | - Susanne V. Fleig
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany,Nephrology Section, Medical Clinic 1, University Hospital Bonn, Rheinische Friedrich Wilhelm University of Bonn, Bonn, Germany
| | - Song Rong
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - David DeLuca
- German Centre for Lung Research, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Roland Schmitt
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany
| | - Sibylle von Vietinghoff
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Hannover, Germany,Nephrology Section, Medical Clinic 1, University Hospital Bonn, Rheinische Friedrich Wilhelm University of Bonn, Bonn, Germany
| |
Collapse
|
13
|
Wang S, Duan Y. LncRNA OIP5-AS1 inhibits the lipopolysaccharide-induced inflammatory response and promotes osteogenic differentiation of human periodontal ligament cells by sponging miR-92a-3p. Bioengineered 2022; 13:12055-12066. [PMID: 35546327 PMCID: PMC9276041 DOI: 10.1080/21655979.2022.2067291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/12/2022] Open
Abstract
Periodontitis is a chronic infectious disease that affects the oral health of adults. Long non-coding RNA OIP5 antisense RNA 1 (OIP5-AS1) has been reported to downregulated in the periodontal tissue of patients with periodontitis. Therefore, the study sought to look at the possible functions of OIP5-AS1 in periodontitis and the associated underlying mechanisms. In the present study, the expression level of OIP5-AS1 and microRNA-92a-3p were analyzed using reverse transcription-quantitative PCR. The levels of osteogenic proteins were determined using western blotting and inflammatory cytokines and oxidative stress were also examined. The proliferation of human periodontal ligament stem cells (hPDLSCs) was evaluated using MTT assays. Assay of osteogenic differentiation was undertaken by means of Alkaline phosphatase staining. The possible association between OIP5-AS1 and miR-92a-3p was determined applying dual-luciferase reporter assays and verified by RNA immunoprecipitation assay. We found that OIP5-AS1 was expressed at low levels in lipopolysaccharide (LPS)-stimulated hPDLSCs. OIP5-AS1 overexpression promoted proliferation and osteogenic differentiation ability and reduced LPS-induced inflammation in hPDLSCs. Furthermore, OIP5-AS1 directly targeted and reduced miR-92a-3p expression. The overexpression of miR-92a-3p partly abolished the effects of OIP5-AS1 on LPS-induced cell proliferation and osteogenic differentiation as well as inflammation in hPDLSCs. Collectively, the results indicated that OIP5-AS1 overexpression inhibited the LPS-induced inflammatory response and promoted the osteogenic differentiation of hPDLSCs by sponging miR-92a-3p. Thus, OIP5-AS1 is probably an essential objective for research during periodontitis treatment.
Collapse
Affiliation(s)
- Shiwei Wang
- Dental Department, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi 710077, P.R. China
| | - Yao Duan
- Second Clinical Division, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, P.R. China
| |
Collapse
|
14
|
Analysis of the miRNA-mRNA Regulatory Network Reveals the Biomarker Genes in the Progression of Myocardial Ischemic Reperfusion. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:2045619. [PMID: 35463659 PMCID: PMC9020924 DOI: 10.1155/2022/2045619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/02/2022] [Accepted: 03/16/2022] [Indexed: 11/18/2022]
Abstract
Objective Cardiac injury induced by myocardial ischemic reperfusion (MI/R) is still an intractable question in clinical, and it has been confirmed as a major reason for the development of cardiovascular disease. Bioinformatics analysis has been widely used for revealing the pathogenic mechanism of diseases. This study attempted to identify the biomarkers and reveal the regulation mechanism of MI/R injury via bioinformatics analysis. Methods The GSE67308 and GSE74951 were obtained from the GEO database. The datasets were analyzed with GEO2R tool, and the genes with |logFC| > 2 and p value <0.05 were identified as the differentially expressed genes (DEGs). The enrichment analysis of the DEGs was performed with the DAVID database and R language. Moreover, the protein-protein interaction (PPI) network of DEGs was performed with the STRING database and then visualized with Cytoscape. Result The results showed that 195 downregulated mRNAs and 240 downregulated mRNAs were found in GSE67308, and 11 miRNAs were found in GSE7495. 152 common genes were screened in DEGs of GSE67308 and the targets of 11 miRNAs in GSE7495. Moreover, the enrichment analysis showed that the common genes were related with inflammatory response, immune response, PI3K/AKT, NF-κB, and TNF pathways. Besides, mmu-miR-92a-3p and mmu-miR-27b-3p were identified as the hubs miRNAs, and TNF, IL1B, and IFG1 were screened as the key nodes. Conclusion This study established a miRNA-mRNA network for cardiac injury induced by MI/R and provided the evidence concerning the molecular mechanism of MI/R injury, which provided some reference for MI/R treatment.
Collapse
|
15
|
Li H, Zhan J, Chen C, Wang D. MicroRNAs in cardiovascular diseases. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:140-168. [PMID: 37724243 PMCID: PMC10471109 DOI: 10.1515/mr-2021-0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 12/29/2021] [Indexed: 09/20/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death and disability worldwide, despite the wide diversity of molecular targets identified and the development of therapeutic methods. MicroRNAs (miRNAs) are a class of small (about 22 nucleotides) non-coding RNAs (ncRNAs) that negatively regulate gene expression at the post-transcriptional level in the cytoplasm and play complicated roles in different CVDs. While miRNA overexpression in one type of cell protects against heart disease, it promotes cardiac dysfunction in another type of cardiac cell. Moreover, recent studies have shown that, apart from cytosolic miRNAs, subcellular miRNAs such as mitochondria- and nucleus-localized miRNAs are dysregulated in CVDs. However, the functional properties of cellular- and subcellular-localized miRNAs have not been well characterized. In this review article, by carefully revisiting animal-based miRNA studies in CVDs, we will address the regulation and functional properties of miRNAs in various CVDs. Specifically, the cell-cell crosstalk and subcellular perspective of miRNAs are highlighted. We will provide the background for attractive molecular targets that might be useful in preventing the progression of CVDs and heart failure (HF) as well as insights for future studies.
Collapse
Affiliation(s)
- Huaping Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jiabing Zhan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Daowen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
16
|
Shen L, Zhou K, Liu H, Yang J, Huang S, Yu F, Huang D. Prediction of Mechanosensitive Genes in Vascular Endothelial Cells Under High Wall Shear Stress. Front Genet 2022; 12:796812. [PMID: 35087573 PMCID: PMC8787366 DOI: 10.3389/fgene.2021.796812] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/13/2021] [Indexed: 01/10/2023] Open
Abstract
Objective: The vulnerability of atherosclerotic plaques is among the leading cause of ischemic stroke. High wall shear stress (WSS) promotes the instability of atherosclerotic plaques by directly imparting mechanical stimuli, but the specific mechanisms remain unclear. We speculate that modulation of mechanosensitive genes may play a vital role in accelerating the development of plaques. The purpose of this study was to find mechanosensitive genes in vascular endothelial cells (ECs) through combining microarray data with bioinformatics technology and further explore the underlying dynamics–related mechanisms that cause the progression and destabilization of atherosclerotic plaques. Methods: Microarray data sets for human vascular ECs under high and normal WSS were retrieved from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified through the R language. The performance of enrichment analysis and protein–protein interaction (PPI) network presented the biological function and signaling pathways of the DEGs. Hub genes were identified based on the PPI network and validated by GEO data sets. Predicted transcription factor (TF) genes and miRNAs interaction with potential mechanosensitive genes were identified by NetworkAnalyst. Results: A total of 260 DEGs, 121 upregulated and 139 downregulated genes, were screened between high and normal WSS from GSE23289. A total of 10 hub genes and four cluster modules were filtered out based on the PPI network. The enrichment analysis showed that the biological functions of the hub genes were mainly involved in responses to unfolded protein and topologically incorrect protein, and t to endoplasmic reticulum stress. The significant pathways associated with the hub genes were those of protein processing in the endoplasmic reticulum, antigen processing, and presentation. Three out of the 10 hub genes, namely, activated transcription factor 3 (ATF3), heat shock protein family A (Hsp70) member 6 (HSPA6), and dual specificity phosphatase 1 (DUSP1, also known as CL100, HVH1, MKP-1, PTPN10), were verified in GSE13712. The expression of DUSP1 was higher in the senescent cell under high WSS than that of the young cell. The TF–miRNA–mechanosensitive gene coregulatory network was constructed. Conclusion: In this work, we identified three hub genes, ATF3, HSPA6, and DUSP1, as the potential mechanosensitive genes in the human blood vessels. DUSP1 was confirmed to be associated with the senescence of vascular ECs. Therefore, these three mechanosensitive genes may have emerged as potential novel targets for the prediction and prevention of ischemic stroke. Furthermore, the TF–miRNA–mechanosensitive genes coregulatory network reveals an underlying regulatory mechanism and the pathways to control disease progression.
Collapse
Affiliation(s)
- Lei Shen
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kaige Zhou
- School of Medicine, Tongji University, Shanghai, China
| | - Hong Liu
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Yang
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shuqi Huang
- Department of Neurology, Shanghai Tianyou Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fei Yu
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dongya Huang
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
17
|
Li N, Li C, Li D, Dang LH, Ren K, Du QX, Cao J, Jin QQ, Wang YY, Bai RF, Sun JH. Identifying biomarkers for evaluating wound extent and age in the contused muscle of rats using microarray analysis: a pilot study. PeerJ 2022; 9:e12709. [PMID: 35036173 PMCID: PMC8710249 DOI: 10.7717/peerj.12709] [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: 09/06/2021] [Accepted: 12/08/2021] [Indexed: 11/20/2022] Open
Abstract
Wound age estimation is still one of the most important and significant challenges in forensic practice. The extent of wound damage greatly affects the accuracy and reliability of wound age estimation, so it is important to find effective biomarkers to help diagnose wound degree and wound age. In the present study, the gene expression profiles of both mild and severe injuries in 33 rats were assayed at 0, 1, 3, 24, 48, and 168 hours using the Affymetrix microarray system to provide biomarkers for the evaluation of wound age and the extent of the wound. After obtaining thousands of differentially expressed genes, a principal component analysis, the least absolute shrinkage and selection operator, and a time-series analysis were used to select the most predictive prognostic genes. Finally, 15 genes were screened for evaluating the extent of wound damage, and the top 60 genes were also screened for wound age estimation in mild and severe injury. Selected indicators showed good diagnostic performance for identifying the extent of the wound and wound age in a Fisher discriminant analysis. A function analysis showed that the candidate genes were mainly related to cell proliferation and the inflammatory response, primarily IL-17 and the Hematopoietic cell lineage signalling pathway. The results revealed that these genes play an essential role in wound-healing and yield helpful and valuable potential biomarkers for further targeted studies.
Collapse
Affiliation(s)
- Na Li
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Chun Li
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Dan Li
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Li-Hong Dang
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Kang Ren
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Qiu-Xiang Du
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Jie Cao
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Qian-Qian Jin
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Ying-Yuan Wang
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| | - Ru-Feng Bai
- Key Laboratory of Evidence Science, China University of Political Science and Law, Beijing, China
| | - Jun-Hong Sun
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, China
| |
Collapse
|
18
|
Cheang I, Zhu Q, Liao S, Li X. Current Understanding of piRNA in Cardiovascular Diseases. FRONTIERS IN MOLECULAR MEDICINE 2022; 1:791931. [PMID: 39087079 PMCID: PMC11285661 DOI: 10.3389/fmmed.2021.791931] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/17/2021] [Indexed: 08/02/2024]
Abstract
The relationship regarding non-coding genomes and cardiovascular disease (CVD) has been explored in the past decade. As one of the leading causes of death, there remains a lack of sensitive and specific genomic biomarkers in the diagnosis and prognosis of CVD. Piwi-interacting RNA (piRNA) is a group of small non-coding RNA (ncRNA) which associated with Piwi proteins. There is an emerging strong body of evidence in support of a role for ncRNAs, including piRNAs, in pathogenesis and prognosis of CVD. This article reviews the current evidence for piRNA-regulated mechanisms in CVD, which could lead to the development of new therapeutic strategies for prevention and treatment.
Collapse
Affiliation(s)
| | | | | | - Xinli Li
- First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
19
|
Abstract
Regulatory RNAs like microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) control vascular and immune cells' phenotype and thus play a crucial role in atherosclerosis. Moreover, the mutual interactions between miRNAs and lncRNAs link both types of regulatory RNAs in a functional network that affects lesion formation. In this review, we deduce novel concepts of atherosclerosis from the analysis of the current data on regulatory RNAs' role in endothelial cells (ECs) and macrophages. In contrast to arterial ECs, which adopt a stable phenotype by adaptation to high shear stress, macrophages are highly plastic and quickly change their activation status. At predilection sites of atherosclerosis, such as arterial bifurcations, ECs are exposed to disturbed laminar flow, which generates a dysadaptive stress response mediated by miRNAs. Whereas the highly abundant miR-126-5p promotes regenerative proliferation of dysadapted ECs, miR-103-3p stimulates inflammatory activation and impairs endothelial regeneration by aberrant proliferation and micronuclei formation. In macrophages, miRNAs are essential in regulating energy and lipid metabolism, which affects inflammatory activation and foam cell formation.Moreover, lipopolysaccharide-induced miR-155 and miR-146 shape inflammatory macrophage activation through their oppositional effects on NF-kB. Most lncRNAs are not conserved between species, except a small group of very long lncRNAs, such as MALAT1, which blocks numerous miRNAs by providing non-functional binding sites. In summary, regulatory RNAs' roles are highly context-dependent, and therapeutic approaches that target specific functional interactions of miRNAs appear promising against cardiovascular diseases.
Collapse
Affiliation(s)
- Andreas Schober
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany.
| | - Saffiyeh Saboor Maleki
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Maliheh Nazari-Jahantigh
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
20
|
Zhang C, Liu M, Wang X, Chen S, Fu X, Li G, Dong N, Shang X. Mechanism of CircANKRD36 regulating cell heterogeneity and endothelial mesenchymal transition in aortic valve stromal cells by regulating miR-599 and TGF-β signaling pathway. Int J Cardiol 2022; 352:104-114. [DOI: 10.1016/j.ijcard.2022.01.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/09/2022] [Accepted: 01/19/2022] [Indexed: 02/08/2023]
|
21
|
Li X, Wang J, Wu C, Lu X, Huang J. MicroRNAs involved in the TGF-β signaling pathway in atherosclerosis. Biomed Pharmacother 2021; 146:112499. [PMID: 34959122 DOI: 10.1016/j.biopha.2021.112499] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease with a multifactorial pathogenesis. It becomes a global health concern, especially causing an array of fatal consequences among the elderly. However, the mechanisms of AS remain unexplained. The transforming growth factor-β (TGF-β) signaling pathway is widely involved in the inflammation, immune function, proliferation, differentiation,and apoptosis in vivo. Based on previous researches, it has not been confirmed whether the TGF-β pathway promotes or inhibits atherosclerosis. Furthermore, more and more studies have found that microRNAs can regulate atherosclerosis through the TGF-β signaling pathway. In this review, we summarize and discuss the role of microRNAs in the pathogenesis of atherosclerosis via the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Xiaoqing Li
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinyu Wang
- Department of Cardiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Wu
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiang Lu
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jingjing Huang
- Department of Geriatrics, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
22
|
Revealing the role of miRNA-489 as a new onco-suppressor factor in different cancers based on pre-clinical and clinical evidence. Int J Biol Macromol 2021; 191:727-737. [PMID: 34562537 DOI: 10.1016/j.ijbiomac.2021.09.089] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/17/2023]
Abstract
Recently, microRNAs (miRNAs) have shown to be potential therapeutic, diagnostic and prognostic targets in disease therapy. These endogenous non-coding RNAs contribute to regulation of different cellular events that are necessary for maintaining physiological condition. Dysregulation of miRNAs is correlated with development of various pathological events such as neurological disorders, cardiovascular diseases, and cancer. miRNA-489 is a new emerging miRNA and studies are extensively investigating its role in pathological conditions. Herein, potential function of miRNA-489 as tumor-suppressor in various cancers is described. miRNA-489 is able to sensitize cancer cells into chemotherapy by disrupting molecular pathways involved in cancer growth such as PI3K/Akt, and induction of apoptosis. The PROX1 and SUZ12 as oncogenic pathways, are affected by miRNA-489 in suppressing metastasis of cancer cells. Wnt/β-catenin as an oncogenic factor ensuring growth and malignancy of tumors is inhibited via miRNA-489 function. For enhancing drug sensitivity of tumors, restoring miRNA-489 expression is a promising strategy. The lncRNAs can modulate miRNA-489 expression in tumors and studies about circRNA role in miRNA-489 modulation should be performed. The expression level of miRNA-489 is a diagnostic tool for tumor detection. Besides, down-regulation of miRNA-489 in tumors provides unfavorable prognosis.
Collapse
|
23
|
Wang C, Li YH, Yang ZT, Cheng NT, Tang HX, Xu M. The function and mechanism of microRNA-92a-3p in lipopolysaccharide-induced acute lung injury. Immunopharmacol Immunotoxicol 2021; 44:47-57. [PMID: 34783628 DOI: 10.1080/08923973.2021.2001497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Sepsis-associated acute lung injury (ALI) is a clinically severe respiratory disorder and remains the leading cause of multiple organ failure and mortality. Herein, we used lipopolysaccharide (LPS) to generate sepsis-induced ALI and try to explore the role and mechanism of microRNA-92a-3p (miR-92a-3p) in this process. METHODS Mice were intravenously injected with miR-92a-3p agomir, antagomir and negative controls for 3 consecutive days and then were intratracheally instillated by LPS (5 mg/kg) for 12 h. To knock down the endogenous A-kinase anchoring protein 1 (AKAP1), mice were intratracheally injected with recombinant adenovirus carrying the short hairpin RNA targeting AKAP1 (shAkap1) at 1 week before LPS administration. RESULTS miR-92a-3p level was significantly upregulated in the lungs by LPS injection. miR-92a-3p antagomir reduced LPS-induced intrapulmonary inflammation and oxidative stress, thereby preventing pulmonary injury and dysfunction. In contrast, miR-92a-3p agomir aggravated LPS-induced intrapulmonary inflammation, oxidative stress, pulmonary injury and dysfunction. Moreover, we reported that AKAP1 upregulation was required for the beneficial effects of miR-92a-3p antagomir, and that AKAP1 knockdown completely abolished the anti-inflammatory and antioxidant capacities of miR-92a-3p antagomir. CONCLUSION Our data identify that miR-92a-3p modulates LPS-induced intrapulmonary inflammation, oxidative stress and ALI via AKAP1 in mice.
Collapse
Affiliation(s)
- Cong Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yang-Hao Li
- Department of Thoracic Surgery, Huangmei People's Hospital, Huanggang, China
| | - Ze-Tian Yang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ni-Tao Cheng
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - He-Xiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ming Xu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
24
|
Huang L, Cai HA, Zhang MS, Liao RY, Huang X, Hu FD. Ginsenoside Rg1 promoted the wound healing in diabetic foot ulcers via miR-489-3p/Sirt1 axis. J Pharmacol Sci 2021; 147:271-283. [PMID: 34507636 DOI: 10.1016/j.jphs.2021.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 07/07/2021] [Accepted: 07/28/2021] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Diabetic foot ulcers (DFUs) are common complications of high severity for diabetes. Ginsenoside Rg1 (Rg1) has the potential for diabetes and cardiovascular diseases therapy. This research aimed at exploring the regulation of Rg1 on DFUs treatment and the underlying mechanism. METHODS Human umbilical vein endothelial cells (HUVECs) incubated with high-glucose culture medium were established for induction of diabetes model. The MTT assay, Annexin V/PI assay and oxidative stress detection were carried out on high-glucose-induced HUVECs. Dual-luciferase reporter assay was performed to prove the interaction of miR-489-3p and Sirt1. DFUs model was established to determine the efficiency of Rg1 and miR-489-3p in wound closure of DFUs in vivo. RESULTS Rg1 promoted cell proliferation, migration and angiogenesis, and reduced cell apoptosis in high-glucose-induced HUVECs. Knockdown of miR-489-3p alleviated the high-glucose-induced damage to HUVECs, while overexpression of miR-489-3p attenuated the protection effects of Rg1. Overexpression Sirt1 promoted wound healing in DFUs and Sirt1 was a direct target of miR-489-3p. In addition, animal experiments demonstrated that Rg1 promoted wound closure by regulating miR-489-3p/Sirt1 axis. CONCLUSIONS Rg1 alleviated the DFUs by increasing Sirt1 expression via miR-489-3p downregulation and promoting activation of PI3K/AKT/eNOS signaling.
Collapse
Affiliation(s)
- Liang Huang
- Second Clinical Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, PR China; Department of Rehabilitation, Guangdong Provincial People's Hospital, Guangzhou, 510515, Guangdong Province, PR China
| | - Hua-An Cai
- Department of Rehabilitation Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan Province, PR China; Department of Sports Medicine, Institute of Translational Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan Province, PR China.
| | - Ming-Sheng Zhang
- Second Clinical Medical College, Southern Medical University, Guangzhou, 510515, Guangdong Province, PR China; Department of Rehabilitation, Guangdong Provincial People's Hospital, Guangzhou, 510515, Guangdong Province, PR China.
| | - Ruo-Yi Liao
- First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, 410004, Hunan Province, PR China
| | - Xing Huang
- Department of General Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan Province, PR China
| | - Feng-Dan Hu
- Department of Rehabilitation Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan Province, PR China
| |
Collapse
|
25
|
Dong L, Dong B. miR-489-3p overexpression inhibits lipopolysaccharide-induced nucleus pulposus cell apoptosis, inflammation and extracellular matrix degradation via targeting Toll-like receptor 4. Exp Ther Med 2021; 22:1323. [PMID: 34630677 PMCID: PMC8495590 DOI: 10.3892/etm.2021.10758] [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: 09/29/2019] [Accepted: 04/17/2020] [Indexed: 11/20/2022] Open
Abstract
Intervertebral disc degeneration (IDD) is a common disease with a high morbidity rate, which results in a significant deterioration in the quality of life of patients. MicroRNAs (miRNAs/miRs) are a class of endogenous small non-coding RNAs that influence target genes and serve critical roles in numerous biological processes. However, the role of miR-489-3p in lumbar disc degeneration is yet to be elucidated. In the present study, human NP cells were treated with 10 ng/ml lipopolysaccharide (LPS) for 24 h to investigate the role of miR-489-3p in IDD in an in vitro model. Reverse transcription-quantitative (RT-q)PCR was performed to determine the expression levels of miR-489-3p. Then, the TargetScan database was used to predict the potential binding sites between miR-489-3p and Toll-like receptor (TLR)4, and a dual-luciferase reporter assay was performed to verify the findings. Subsequently, RT-qPCR and western blotting were used to analyze the expression levels of TLR4. In addition, human nucleus pulposus (NP) cells were transfected with a miR-489-3p mimic and TLR4 overexpression plasmid to study the effects of miR-489-3p on LPS-induced human NP cells. Cell apoptosis and cell viability were also determined using flow cytometry and MTT assays, respectively. Finally, ELISAs were performed to analyze the levels of inflammatory factors. The expression levels of miR-489-3p were discovered to be downregulated in LPS-treated human NP cells. In addition, TLR4 was revealed to be a direct target gene of miR-489-3p, and its expression levels were upregulated in LPS-treated human NP cells. miR-489-3p was found to inhibit the LPS-induced decreases in cell viability and increases in apoptosis, and the concentration of inflammatory cytokines. Furthermore, miR-489-3p suppressed the LPS-induced decreases in extracellular matrix deposition via decreasing the expression levels of aggrecan and collagen type II in human NP cells. Finally, the results revealed that miR-489-3p inhibited the LPS-induced activation of the NF-κB signaling pathway in human NP cells. Conversely, all of the effects of miR-489-3p on LPS-induced human NP cells were reversed by the TLR4 overexpression plasmid. These findings suggested that miR-489-3p may represent a novel therapeutic target for the treatment of IDD.
Collapse
Affiliation(s)
- Ling Dong
- Department of Rehabilitation Medicine, Guizhou Orthopedics Hospital, Guiyang, Guizhou 550000, P.R. China
| | - Bo Dong
- Pain Rehabilitation Department of TCM Orthopedic Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
26
|
Wang JS, Tsai PH, Tseng KF, Chen FY, Yang WC, Shen MY. Sesamol Ameliorates Renal Injury-Mediated Atherosclerosis via Inhibition of Oxidative Stress/IKKα/p53. Antioxidants (Basel) 2021; 10:antiox10101519. [PMID: 34679653 PMCID: PMC8532890 DOI: 10.3390/antiox10101519] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 12/11/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are at an increased risk of premature death due to the development of cardiovascular disease (CVD) owing to atherosclerosis-mediated cardiovascular events. However, the mechanisms linking CKD and CVD are clear, and the current treatments for high-risk groups are limited. In this study, we aimed to examine the effects of sesamol, a natural compound extracted from sesame oil, on the development of atherosclerosis in a rodent CKD model, and reactive oxygen species-induced oxidative damage in an endothelial cell model. ApoE–/– mice were subjected to 5/6 nephrectomy (5/6 Nx) and administered sesamol for 8 weeks. Compared with the sham group, the 5/6 Nx ApoE–/– mice showed a significant increase in malondialdehyde levels and Oil Red O staining patterns, which significantly decreased following sesamol administration. Sesamol suppressed H2O2-induced expression of phospho-IKKα, p53, and caspase-3. Our results highlight the protective role of sesamol in renal injury-associated atherosclerosis and the pathological importance of oxidative stress burden in CKD–CVD interaction.
Collapse
Affiliation(s)
- Jie-Sian Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (J.-S.W.); (P.-H.T.); (F.-Y.C.)
- Division of Nephrology, Department of Internal Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Ping-Hsuan Tsai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (J.-S.W.); (P.-H.T.); (F.-Y.C.)
| | - Kuo-Feng Tseng
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan;
| | - Fang-Yu Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (J.-S.W.); (P.-H.T.); (F.-Y.C.)
| | - Wen-Chin Yang
- Agricultural Biotechnology Research Center, Academia Sinica, 128, Sec. 2, Academia Rd., Nankang, Taipei 115, Taiwan;
| | - Ming-Yi Shen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (J.-S.W.); (P.-H.T.); (F.-Y.C.)
- Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan
- Department of Nursing, Asia University, Taichung 41354, Taiwan
- Correspondence: or ; Tel.: +886-4-2205-3366 (ext. 5809)
| |
Collapse
|
27
|
Curcumin Antagonizes Glucose Fluctuation-Induced Renal Injury by Inhibiting Aerobic Glycolysis via the miR-489/LDHA Pathway. Mediators Inflamm 2021; 2021:6104529. [PMID: 34456629 PMCID: PMC8387199 DOI: 10.1155/2021/6104529] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/09/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
It has been considered that glucose fluctuation (GF) plays a role in renal injury and is related to diabetic nephropathy (DN) development. But the mechanism is still unclear. Aerobic glycolysis has become a topical issue in DN in recent years. There is an internal connection between GF, aerobic glycolysis, and DN. Curcumin (Cur) is a principal curcuminoid of turmeric and possesses specific protective properties in kidney functions. Cur also participates in the regulation of aerobic glycolysis switch. In this study, we first measured the levels of aerobic glycolysis and evaluated Cur's inhibitory ability in a cell model of HEK-293 under the condition of oscillating high glucose. The results indicated that GF exacerbated inflammation injury, oxidative stress, and apoptosis in HEK-293 cell, while Cur alleviated this cytotoxicity induced by GF. We found that GF increased aerobic glycolysis in HEK-293 cells and Cur presented a dose-dependent weakening effect to this exacerbation. Next, we built a panel of 17 miRNAs and 8 lncRNAs that were previously reported to mediate the Warburg effect. Our RT-qPCR results indicated that GF reduced the miR-489 content in the HEK-293 cell model and Cur could prevent this downregulation. Then, we planned to explore the character of miR-489 in Cur-triggered attenuation of the Warburg effect under GF condition. Our findings presented that Cur prevented GF-triggered aerobic glycolysis by upregulating miR-489 in HEK-293 cells. Next, we choose the miR-489/LDHA axis for further investigation. We confirmed that Cur prevented GF-triggered aerobic glycolysis via the miR-489/LDHA axis in HEK-293 cells. In conclusion, this study presented that Cur prevented GF-triggered renal injury by restraining aerobic glycolysis via the miR-489/LDHA axis in the HEK-293 cell model.
Collapse
|
28
|
Li X, Haberzettl P, Conklin DJ, Bhatnagar A, Rouchka EC, Zhang M, O’Toole TE. Exposure to Fine Particulate Matter Air Pollution Alters mRNA and miRNA Expression in Bone Marrow-Derived Endothelial Progenitor Cells from Mice. Genes (Basel) 2021; 12:1058. [PMID: 34356074 PMCID: PMC8307414 DOI: 10.3390/genes12071058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022] Open
Abstract
Exposure to fine particulate matter (PM2.5) air pollution is associated with quantitative deficits of circulating endothelial progenitor cells (EPCs) in humans. Related exposures of mice to concentrated ambient PM2.5 (CAP) likewise reduces levels of circulating EPCs and induces defects in their proliferation and angiogenic potential as well. These changes in EPC number or function are predictive of larger cardiovascular dysfunction. To identify global, PM2.5-dependent mRNA and miRNA expression changes that may contribute to these defects, we performed a transcriptomic analysis of cells isolated from exposed mice. Compared with control samples, we identified 122 upregulated genes and 44 downregulated genes in EPCs derived from CAP-exposed animals. Functions most impacted by these gene expression changes included regulation of cell movement, cell and tissue development, and cellular assembly and organization. With respect to miRNA changes, we found that 55 were upregulated while 53 were downregulated in EPCs from CAP-exposed mice. The top functions impacted by these miRNA changes included cell movement, cell death and survival, cellular development, and cell growth and proliferation. A subset of these mRNA and miRNA changes were confirmed by qRT-PCR, including some reciprocal relationships. These results suggest that PM2.5-induced changes in gene expression may contribute to EPC dysfunction and that such changes may contribute to the adverse cardiovascular outcomes of air pollution exposure.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY 40202, USA;
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA;
| | - Petra Haberzettl
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA; (P.H.); (D.J.C.); (A.B.)
- Division of Environmental Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Daniel J. Conklin
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA; (P.H.); (D.J.C.); (A.B.)
- Division of Environmental Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Aruni Bhatnagar
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA; (P.H.); (D.J.C.); (A.B.)
- Division of Environmental Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Eric C. Rouchka
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA;
- Department of Computer Science and Engineering, University of Louisville, Louisville, KY 40202, USA
| | - Mei Zhang
- Department of Medicine, University of Louisville Genomics Facility, Louisville, KY 40202, USA;
| | - Timothy E. O’Toole
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA; (P.H.); (D.J.C.); (A.B.)
- Division of Environmental Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
29
|
Xiang D, Li Y, Cao Y, Huang Y, Zhou L, Lin X, Qiao Y, Li X, Liao D. Different Effects of Endothelial Extracellular Vesicles and LPS-Induced Endothelial Extracellular Vesicles on Vascular Smooth Muscle Cells: Role of Curcumin and Its Derivatives. Front Cardiovasc Med 2021; 8:649352. [PMID: 34150863 PMCID: PMC8210670 DOI: 10.3389/fcvm.2021.649352] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/07/2021] [Indexed: 12/25/2022] Open
Abstract
Background: During the progression of atherosclerosis (AS), the vascular endothelial and smooth muscle cells are reciprocally regulated by extracellular vesicles (EVs). EVs have different effects on pathological and physiological processes due to the different cargoes contained in EVs. Purpose: To study the effects of endothelial cells-derived EVs on normal and inflammatory conditions. To investigate the effects of curcumin and curcumin derivatives (Nicotinic-curcumin) on endothelial EVs. Methods: EVs were isolated from human umbilical vein endothelial cells (HUVECs) by ultracentrifugation. To examined the effect of normal and LPS-induced endothelial cells-derived EVs on the proliferation of human aortic smooth muscle cells (HASMCs), the CCK-8 assay was performed. Transwell and wound healing assays were conducted to assess cell migration. The effects of EVs on lipid accumulation following treatment with oxidized low-density lipoprotein (Ox-LDL) were evaluated with the oil red O staining assay and HPLC. The number of EVs was calculated using the nanoparticle tracking analysis (NTA) and BCA. The expression levels of Rab27a and Rab27b that regulate the EVs secretion were measured by Western blotting assay. The differential expression of miRNAs in endothelial EVs and LPS-induced endothelial EVs was analyzed using miRNA-Sequencing (miRNA-Seq) and RT-PCR. Results: Treatment with endothelial EVs reduced the proliferation and migration of HASMCs as well as lipid accumulation in HASMCs. However, treatment with LPS-induced endothelial EVs did not inhibit the migration of HASMCs or lipid accumulation, instead it promoted the proliferation of HASMCs. Treatment with the two types of EVs induced differential expression of several miRNAs, including miR-92a-3p, miR-126-5p, miR-125a-3p, miR-143-3p, etc. Moreover, 1 μg/mL LPS induction greatly increased secretion of endothelial EVs. Treatment with curcumin and nicotinic-curcumin reduced endothelial EVs secretion, possibly by inhibiting inflammation. Conclusion: Endothelial EVs may confer beneficial effects on atherosclerosis by regulating vascular smooth muscle cell (VSMCs), whereas pro-inflammatory factors may disrupt this effect.
Collapse
Affiliation(s)
| | - Yamei Li
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Yuling Cao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Ying Huang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Lili Zhou
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Xiulian Lin
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Yong Qiao
- The Third Hospital of Changsha, Changsha, China
| | - Xin Li
- The Third Hospital of Changsha, Changsha, China
| | - Duanfang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
30
|
Vickers KC, Michell DL. HDL-small RNA Export, Transport, and Functional Delivery in Atherosclerosis. Curr Atheroscler Rep 2021; 23:38. [PMID: 33983531 DOI: 10.1007/s11883-021-00930-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW This review highlights recent advances on the mechanisms and impact of HDL-small non-coding RNAs (sRNA) on intercellular communication in atherosclerosis. RECENT FINDINGS Studies demonstrate that HDL-microRNAs (miRNA) are significantly altered in atherosclerotic cardiovascular disease (ASCVD), and are responsive to diet, obesity, and diabetes. Immune cells, pancreatic beta cells, and neurons are shown to export miRNAs to HDL. In turn, HDL can deliver functional miRNAs to recipient hepatocytes and endothelial cells regulating adhesion molecule expression, cytokines, and angiogenesis. With high-throughput sRNA sequencing, we now appreciate the full sRNA signature on circulating HDL, including the transport of rRNA and tRNA-derived fragments. Strikingly, HDL were highly enriched with exogenous microbial sRNAs. HDL transport a diverse set of host and non-host sRNAs that are altered in cardiometabolic diseases. Given the bioactivity of these sRNAs, they likely contribute to cellular communication within atherosclerotic lesions, and are potential disease biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Kasey C Vickers
- Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave. 312 Preston Research Building, Nashville, TN, 37232, USA.
| | - Danielle L Michell
- Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave. 312 Preston Research Building, Nashville, TN, 37232, USA
| |
Collapse
|
31
|
Wang Z, Liu L, Du Y, Mi Y, Wang L. The HNF1A-AS1/miR-92a-3p axis affects the radiosensitivity of non-small cell lung cancer by competitively regulating the JNK pathway. Cell Biol Toxicol 2021; 37:715-729. [PMID: 33755848 DOI: 10.1007/s10565-021-09595-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 02/21/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND It has been widely reported that long non-coding RNAs (lncRNAs) could affect the varieties of tumor response to radiotherapy. LncRNA HNF1A-AS1 is transcribed from HNF1A gene cluster's antisense strand. This work focused on the mechanism of how HNF1A-AS1 participated in the radiosensitivity of non-small cell lung cancer (NSCLC). METHODS The mRNA or protein expression of HNF1A-AS1, miR-92a-3p MAP2K4, and JNK in NSCLC cells and tissues was detected by qRT-PCR or western blotting. RNA immunoprecipitation (RIP) detection and luciferase reporting system were used to evaluate the relationship between HNFA-AS1 and miR-92a-3p or between miR-92a-3p and MAP2K4. Flow cytometry assays, colony formation, and MTT were performed to analyze the function changes in A549 and Calu-1 cells. The rescue experiment was also conducted to explore the underlying mechanisms. RESULTS HNF1A-AS1 was investigated in NSCLC cells and tissues and highly related to the advanced pathological stage. HNF1A-AS1 bound with miR-92a-3p, which was downregulated in NSCLC. It showed that miR-92a-3p was negatively related to HNF1A-AS1. Knockdown of HNF1A-AS1 impacted most cell biological behaviors in NSCLC cells, including restricting the proliferation and aggravating apoptosis. Furthermore, knockdown of HNF1A-AS1 dramatically enhanced radiotherapy sensitivity of NSCLC. Moreover, miR-92a-3p was found to target MAP2K4 and could reduce MAP2K4 expression. Inhibition of HNF1A-AS1 elevated radiotherapy sensitivity and retarded the progression of NSCLC cells, followed by decreasing expression levels of MAP2K4. Besides, MAP2K4 mimic rescued the si-HNF1A-AS1 effects on the biological behavior of NSCLC cells. CONCLUSION HNF1A-AS1 is highly expressed in NSCLC. MiR-92a-3p is the target gene of HNF1A-AS1 and involved in tumor progression by regulating the MAP2K4/JNK pathway. HNF1AS1/miR-92a-3p/MAP2K4 axis plays important roles in radiotherapy resistance of NSCLC.
Collapse
Affiliation(s)
- Zhiyu Wang
- Department of Oncology immunology, Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, 050011, People's Republic of China
| | - Liang Liu
- Tumor Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, 050011, People's Republic of China
| | - Yuankun Du
- Periodical press of Hebei Medical University, Shijiazhuang City, Hebei Province, 050011, People's Republic of China
| | - Yuan Mi
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang City, Hebei Province, 050011, People's Republic of China
| | - Lei Wang
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang City, Hebei Province, 050011, People's Republic of China.
| |
Collapse
|
32
|
MicroRNA-92a-3p enhances functional recovery and suppresses apoptosis after spinal cord injury via targeting phosphatase and tensin homolog. Biosci Rep 2021; 40:222664. [PMID: 32297644 PMCID: PMC7199448 DOI: 10.1042/bsr20192743] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 02/08/2023] Open
Abstract
Spinal cord injury (SCI) is a neurological disease commonly caused by traumatic events on spinal cords. MiRNA-92a-3p is reported to be down-regulated after SCI. Our study investigated the effects of up-regulated miR-92a-3p on SCI and the underlying mechanisms. SCI mice model was established to evaluate the functional recovery of hindlimbs of mice through open-field locomotion and scored by Basso, Beattie, and Bresnahan (BBB) locomotion scale. Apoptosis of spinal cord cells was determined by flow cytometry. The effects of miR-92a-3p on SCI were detected by intrathecally injecting miR-92a-3p agomiR (agomiR-92) into the mice prior to the establishment of SCI. Phosphatase and tensin homolog (PTEN) was predicted as a target of miR-29a-3p by TargetScan. We further assessed the effects of agomiR-92 or/and overexpressed PTEN on apoptosis rates and apoptotic protein expressions in SCI mice. Moreover, the activation of protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling was determined by Western blot. The results showed that compared with the sham-operated mice, SCI mice had much lower BBB scores, and theapoptosis rate of spinal cord cells was significantly increased. After SCI, the expression of miR-92a-3p was down-regulated, and increased expression of miR-92a-3p induced by agomiR-92 further significantly increased the BBB score and decreased apoptosis. PTEN was specifically targeted by miR-92a-3p. In addition, the phosphorylation levels of Akt and mTOR were up-regulated under the treatment of agomiR-92. Our data demonstrated that the neuroprotective effects of miR-92a-3p on spinal cord safter SCI were highly associated with the activation of the PTEN/AKT/mTOR pathway.
Collapse
|
33
|
Dong G, Huang X, Wu L, Jiang S, Tan Q, Chen S. SREBF2 triggers endoplasmic reticulum stress and Bax dysregulation to promote lipopolysaccharide-induced endothelial cell injury. Cell Biol Toxicol 2021; 38:185-201. [PMID: 33677747 DOI: 10.1007/s10565-021-09593-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/21/2021] [Indexed: 10/22/2022]
Abstract
An increased lipopolysaccharide (LPS) level in patients with cirrhosis induced the dysregulation of sterol regulatory element-binding transcription factor 2 (SREBF2), which participated in the modulation of tumor inflammatory microenvironment. However, the role of SREBF2 in the LPS-induced injury of portal vein endothelium was scarcely reported. This study aimed to investigate the effects of SREBF2 on the LPS-induced injury to endothelial cells (ECs) in vitro and in vivo and explore the underlying mechanism. In this study, we found that LPS increased SREBF2 expression through activating the TLR4/JNK/c-Jun pathway and suppressed UBE2I-mediated SREBF2 sumoylation to enhance its transcriptional activity. The dysregulation of SREBF2 induced ER stress by increasing the intracellular cholesterol level and facilitated Bax expression to cause additional damage to LPS-induced ECs. As a potential intervention, miR590-3p negatively regulated SREBF2 expression and upregulated UBE2I expression by targeting TLR4, thus alleviating LPS-induced injury. These results suggest that LPS-induced SREBF2 triggered ER stress and promoted Bax expression to injure ECs, which was reversed by miR590-3p. The mechanisms of SREBF2 mediated LPS-induced endothelial injury of portal vein, which might be the therapeutic target for PVT development in cirrhosis patients. 1. LPS promoted SREBF2 expression by activating the TLR4/JNK/c-Jun pathway and suppressed UBE2I-mediated SREBF2 sumoylation to upregulate SREBF2 transcriptional activity 2. SREBF2-mediated ER stress and Bax expression involved in LPS-induced EC injury 3. miR590-3p decreased SREBF2 expression by targeting TLR4 and mitigated LPS-induced EC injury.
Collapse
Affiliation(s)
- Gang Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaoquan Huang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Center of Evidence-Based Medicine, Fudan University, Shanghai, 200032, People's Republic of China
| | - Ling Wu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Siyu Jiang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qintian Tan
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shiyao Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Center of Evidence-Based Medicine, Fudan University, Shanghai, 200032, People's Republic of China. .,Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
34
|
Düsing P, Zietzer A, Goody PR, Hosen MR, Kurts C, Nickenig G, Jansen F. Vascular pathologies in chronic kidney disease: pathophysiological mechanisms and novel therapeutic approaches. J Mol Med (Berl) 2021; 99:335-348. [PMID: 33481059 PMCID: PMC7900031 DOI: 10.1007/s00109-021-02037-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/14/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease (CVD) is a major cause of death in patients with chronic kidney disease (CKD). Both conditions are rising in incidence as well as prevalence, creating poor outcomes for patients and high healthcare costs. Recent data suggests CKD to be an independent risk factor for CVD. Accumulation of uremic toxins, chronic inflammation, and oxidative stress have been identified to act as CKD-specific alterations that increase cardiovascular risk. The association between CKD and cardiovascular mortality is markedly influenced through vascular alterations, in particular atherosclerosis and vascular calcification (VC). While numerous risk factors promote atherosclerosis by inducing endothelial dysfunction and its progress to vascular structural damage, CKD affects the medial layer of blood vessels primarily through VC. Ongoing research has identified VC to be a multifactorial, cell-mediated process in which numerous abnormalities like mineral dysregulation and especially hyperphosphatemia induce a phenotype switch of vascular smooth muscle cells to osteoblast-like cells. A combination of pro-calcifying stimuli and an impairment of inhibiting mechanisms like fetuin A and vitamin K-dependent proteins like matrix Gla protein and Gla-rich protein leads to mineralization of the extracellular matrix. In view of recent studies, intercellular communication pathways via extracellular vesicles and microRNAs represent key mechanisms in VC and thereby a promising field to a deeper understanding of the involved pathomechanisms. In this review, we provide an overview about pathophysiological mechanisms connecting CKD and CVD. Special emphasis is laid on vascular alterations and more recently discovered molecular pathways which present possible new therapeutic targets.
Collapse
Affiliation(s)
- Philip Düsing
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Andreas Zietzer
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Philip Roger Goody
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Mohammed Rabiul Hosen
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Christian Kurts
- Institute of Experimental Immunology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, Bonn, 53127, Germany
| | - Georg Nickenig
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Felix Jansen
- Heart Center, Department of Medicine II, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
35
|
Levin-Schwartz Y, Curtin P, Flores D, Aushev VN, Tamayo-Ortiz M, Svensson K, Pantic I, Estrada-Gutierrez G, Pizano-Zárate ML, Gennings C, Satlin LM, Baccarelli AA, Tellez-Rojo MM, Wright RO, Sanders AP. Exosomal miRNAs in urine associated with children's cardiorenal parameters: a cross-sectional study. Epigenomics 2021; 13:499-512. [PMID: 33635093 PMCID: PMC8033423 DOI: 10.2217/epi-2020-0342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aims: The authors sought to examine associations between urinary exosomal miRNAs (exo-miRs), emerging biomarkers of renal health, and cardiorenal outcomes in early childhood. Materials & methods: The authors extracted exo-miRs in urine from 88 healthy Mexican children aged 4–6 years. The authors measured associations between 193 exo-miRs and cardiorenal outcomes: systolic/diastolic blood pressure, estimated glomerular filtration rate and urinary sodium and potassium levels. The authors adjusted for age, sex, BMI, socioeconomic status, indoor tobacco smoke exposure and urine specific gravity. Results: Multiple exo-miRs were identified meeting a false discovery rate threshold of q < 0.1. Specifically, three exo-miRs had increased expression with urinary sodium, 17 with urinary sodium-to-potassium ratio and one with decreased estimated glomerular filtration rate. Conclusions: These results highlight urinary exo-miRs as early-life biomarkers of children's cardiorenal health.
Collapse
Affiliation(s)
- Yuri Levin-Schwartz
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, 10029 New York, USA
| | - Paul Curtin
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, 10029 New York, USA
| | - Daniel Flores
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, 10029 NY, USA
| | - Vasily N Aushev
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, 10029 New York, USA
| | - Marcela Tamayo-Ortiz
- Center for Nutrition & Health Research, National Institute of Public Health, 62100 Cuernavaca, Morelos, Mexico.,National Council for Science & Technology, 03940 Mexico City, Mexico
| | - Katherine Svensson
- Department of Health Sciences, Karlstad University, 65188 Karlstad, Sweden
| | - Ivan Pantic
- Department of Developmental Neurobiology, National Institute of Perinatology, 11000 Mexico City, Mexico
| | | | - María L Pizano-Zárate
- Division of Community Interventions Research, National Institute of Perinatology, 11000 Mexico City, Mexico
| | - Chris Gennings
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, 10029 New York, USA
| | - Lisa M Satlin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, 10029 NY, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 10027 New York, USA
| | - Martha M Tellez-Rojo
- Center for Nutrition & Health Research, National Institute of Public Health, 62100 Cuernavaca, Morelos, Mexico
| | - Robert O Wright
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, 10029 New York, USA.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, 10029 NY, USA
| | - Alison P Sanders
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, 10029 New York, USA.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, 10029 NY, USA
| |
Collapse
|
36
|
Tao J, Xia L, Cai Z, Liang L, Chen Y, Meng J, Wang Z. Interaction Between microRNA and DNA Methylation in Atherosclerosis. DNA Cell Biol 2020; 40:101-115. [PMID: 33259723 DOI: 10.1089/dna.2020.6138] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease accompanied by complex pathological changes, such as endothelial dysfunction, foam cell formation, and vascular smooth muscle cell proliferation. Many approaches, including regulating AS-related gene expression in the transcriptional or post-transcriptional level, contribute to alleviating AS development. The DNA methylation is a crucial epigenetic modification in regulating cell function by silencing the relative gene expression. The microRNA (miRNA) is a type of noncoding RNA that plays an important role in gene post-transcriptional regulation and disease development. The DNA methylation and the miRNA are important epigenetic factors in AS. However, recent studies have found a mutual regulation between these two factors in AS development. In this study, recent insights into the roles of miRNA and DNA methylation and their interaction in the AS progression are reviewed.
Collapse
Affiliation(s)
- Jun Tao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Linzhen Xia
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Zemin Cai
- Department of Pediatrics and The First Affiliated Hospital of University of South China, Hengyang, China
| | - Lingli Liang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Yanjun Chen
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Jun Meng
- Functional Department, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
37
|
Mukushkina D, Aisina D, Pyrkova A, Ryskulova A, Labeit S, Ivashchenko A. In silico Prediction of miRNA Interactions With Candidate Atherosclerosis Gene mRNAs. Front Genet 2020; 11:605054. [PMID: 33329752 PMCID: PMC7672156 DOI: 10.3389/fgene.2020.605054] [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: 09/11/2020] [Accepted: 10/15/2020] [Indexed: 12/14/2022] Open
Abstract
The involvement of genes and miRNAs in the development of atherosclerosis is a challenging problem discussed in recent publications. It is necessary to establish which miRNAs affect the expression of candidate genes. We used known candidate atherosclerosis genes to predict associations. The quantitative characteristics of interactions of miRNAs with mRNA candidate genes were determined using the program, which identifies the localization of miRNA binding sites in mRNA, the free energy interaction of miRNA with mRNA. In mRNAs of GAS6 and NFE2L2 candidate genes, binding sites of 21 miRNAs and of 15 miRNAs, respectively, were identified. In IRS2 mRNA binding sites of 25 miRNAs were located in a cluster of 41 nt. In ADRB3, CD36, FASLG, FLT1, PLA2G7, and PPARGC1A mRNAs, clusters of miR-466, ID00436.3p-miR, and ID01030.3p-miR BS were identified. The organization of overlapping miRNA binding sites in clusters led to their compaction and caused competition among the miRNAs. The binding of 53 miRNAs to the mRNAs of 14 candidate genes with free energy interactions greater than -130 kJ/mole was determined. The miR-619-5p was fully complementary to ADAM17 and CD36 mRNAs, ID01593.5p-miR to ANGPTL4 mRNA, ID01935.5p-miR to NFE2L2, and miR-5096 to IL18 mRNA. Associations of miRNAs and candidate atherosclerosis genes are proposed for the early diagnosis of this disease.
Collapse
Affiliation(s)
- Dina Mukushkina
- Department of Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Dana Aisina
- Department of Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Anna Pyrkova
- Department of Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Alma Ryskulova
- Department of microbiology, Kazakh Medical University of Continuing Education, Almaty, Kazakhstan
| | - Siegfried Labeit
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Anatoliy Ivashchenko
- Department of Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| |
Collapse
|
38
|
Tang S, Zhong H, Xiong T, Yang X, Mao Y, Wang D. MiR-489 aggravates H2O2-induced apoptosis of cardiomyocytes via inhibiting IGF1. Biosci Rep 2020; 40:BSR20193995. [PMID: 32880387 PMCID: PMC7494985 DOI: 10.1042/bsr20193995] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 08/12/2020] [Accepted: 09/02/2020] [Indexed: 12/16/2022] Open
Abstract
Myocardial infarction (MI) is a major type of cardiovascular disorder worldwide. In the present study, we established a new microRNA (miRNA)-mRNA cross-talk network by integrating data obtained from The National Center for Biotechnology Information Gene Expression Omnibus (NCBI GEO). In addition, functional assays, including Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) analyses, were conducted using the Database for Annotation, Visualization, and Integration Discovery (DAVID). In our study, we generated a new differentially expressed miRNA (DEmiRNA)-differentially expressed gene (DEG) cross-talk network of MI composed of three miRNA (miR-489, miR-375, and miR-142-3p) nodes and 163 mRNA nodes. In vitro experiments demonstrated that miR-489 expression was increased in H2O2-treated H9c2 cardiomyocytes in vitro, mimicking myocardial injury. We observed that down-regulation of miR-489 reduced H2O2-induced apoptosis, while overexpression of miR-489 had the opposite effects, as revealed by flow cytometry and Western blot analyses. Furthermore, we confirmed the relationship between miR-489 and IGF1 through double luciferase reporter gene assays, which partly explains the antiapoptotic mechanism of miR-489. In conclusion, the experimental results of the present study could provide important clues for investigating the mechanism of MI.
Collapse
Affiliation(s)
- Shan Tang
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hongyan Zhong
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Ting Xiong
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xinquan Yang
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yongqing Mao
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Daxin Wang
- Clinical Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
39
|
Substantially Altered Expression Profile of Diabetes/Cardiovascular/Cerebrovascular Disease Associated microRNAs in Children Descending from Pregnancy Complicated by Gestational Diabetes Mellitus-One of Several Possible Reasons for an Increased Cardiovascular Risk. Cells 2020; 9:cells9061557. [PMID: 32604801 PMCID: PMC7349356 DOI: 10.3390/cells9061557] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
Gestational diabetes mellitus (GDM), one of the major pregnancy-related complications, characterized as a transitory form of diabetes induced by insulin resistance accompanied by a low/absent pancreatic beta-cell compensatory adaptation to the increased insulin demand, causes the acute, long-term, and transgenerational health complications. The aim of the study was to assess if alterations in gene expression of microRNAs associated with diabetes/cardiovascular/cerebrovascular diseases are present in whole peripheral blood of children aged 3-11 years descending from GDM complicated pregnancies. A substantially altered microRNA expression profile was found in children descending from GDM complicated pregnancies. Almost all microRNAs with the exception of miR-92a-3p, miR-155-5p, and miR-210-3p were upregulated. The microRNA expression profile also differed between children after normal and GDM complicated pregnancies in relation to the presence of overweight/obesity, prehypertension/hypertension, and/or valve problems and heart defects. Always, screening based on the combination of microRNAs was superior over using individual microRNAs, since at 10.0% false positive rate it was able to identify a large proportion of children with an aberrant microRNA expression profile (88.14% regardless of clinical findings, 75.41% with normal clinical findings, and 96.49% with abnormal clinical findings). In addition, the higher incidence of valve problems and heart defects was found in children with a prior exposure to GDM. The extensive file of predicted targets of all microRNAs aberrantly expressed in children descending from GDM complicated pregnancies indicates that a large group of these genes is involved in ontologies of diabetes/cardiovascular/cerebrovascular diseases. In general, children with a prior exposure to GDM are at higher risk of later development of diabetes mellitus and cardiovascular/cerebrovascular diseases, and would benefit from dispensarisation as well as implementation of primary prevention strategies.
Collapse
|
40
|
How do Uremic Toxins Affect the Endothelium? Toxins (Basel) 2020; 12:toxins12060412. [PMID: 32575762 PMCID: PMC7354502 DOI: 10.3390/toxins12060412] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/15/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022] Open
Abstract
Uremic toxins can induce endothelial dysfunction in patients with chronic kidney disease (CKD). Indeed, the structure of the endothelial monolayer is damaged in CKD, and studies have shown that the uremic toxins contribute to the loss of cell–cell junctions, increasing permeability. Membrane proteins, such as transporters and receptors, can mediate the interaction between uremic toxins and endothelial cells. In these cells, uremic toxins induce oxidative stress and activation of signaling pathways, including the aryl hydrocarbon receptor (AhR), nuclear factor kappa B (NF-κB), and mitogen-activated protein kinase (MAPK) pathways. The activation of these pathways leads to overexpression of proinflammatory (e.g., monocyte chemoattractant protein-1, E-selectin) and prothrombotic (e.g., tissue factor) proteins. Uremic toxins also induce the formation of endothelial microparticles (EMPs), which can lead to the activation and dysfunction of other cells, and modulate the expression of microRNAs that have an important role in the regulation of cellular processes. The resulting endothelial dysfunction contributes to the pathogenesis of cardiovascular diseases, such as atherosclerosis and thrombotic events. Therefore, uremic toxins as well as the pathways they modulated may be potential targets for therapies in order to improve treatment for patients with CKD.
Collapse
|
41
|
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: 1.3] [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
|
42
|
Groot M, Lee H. Sorting Mechanisms for MicroRNAs into Extracellular Vesicles and Their Associated Diseases. Cells 2020; 9:cells9041044. [PMID: 32331346 PMCID: PMC7226101 DOI: 10.3390/cells9041044] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EV) are secretory membranous elements used by cells to transport proteins, lipids, mRNAs, and microRNAs (miRNAs). While their existence has been known for many years, only recently has research begun to identify their function in intercellular communication and gene regulation. Importantly, cells have the ability to selectively sort miRNA into EVs for secretion to nearby or distant targets. These mechanisms broadly include RNA-binding proteins such as hnRNPA2B1 and Argonaute-2, but also membranous proteins involved in EV biogenesis such as Caveolin-1 and Neural Sphingomyelinase 2. Moreover, certain disease states have also identified dysregulated EV-miRNA content, shedding light on the potential role of selective sorting in pathogenesis. These pathologies include chronic lung disease, immune response, neuroinflammation, diabetes mellitus, cancer, and heart disease. In this review, we will overview the mechanisms whereby cells selectively sort miRNA into EVs and also outline disease states where EV-miRNAs become dysregulated.
Collapse
Affiliation(s)
- Michael Groot
- Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA;
| | - Heedoo Lee
- Department of Biology and Chemistry, Changwon National University, Changwon 51140, Korea
- Correspondence: ; Tel.: +82-55-213-3452
| |
Collapse
|
43
|
Simeoni M, Borrelli S, Garofalo C, Fuiano G, Esposito C, Comi A, Provenzano M. Atherosclerotic-nephropathy: an updated narrative review. J Nephrol 2020; 34:125-136. [PMID: 32270411 DOI: 10.1007/s40620-020-00733-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/03/2020] [Indexed: 12/13/2022]
Abstract
The increased prevalence of chronic kidney disease (CKD) in elderly patients recognizes, as main cause, the long-term exposure to atherosclerosis and hypertension. Chronic ischemic damage due to critical renal arterial stenosis induces oxidative stress and intra-renal inflammation, resulting in fibrosis and microvascular remodelling, that is the histological picture of atherosclerotic renal vascular disease (ARVD). The concomitant presence of a long history of hypertension may generate intimal thickening and luminal narrowing of renal arteries and arterioles, glomerulosclerosis, interstitial fibrosis and tubular atrophy, more typically expression of hypertensive nephropathy. These complex mechanisms contribute to the development of CKD and the progression to End Stage Kidney Disease. In elderly CKD patients, the distinction among these nephropathies may be problematic; therefore, ischemic and hypertensive nephropathies can be joined in a unique clinical syndrome defined as atherosclerotic nephropathy. The availability of novel diagnostic procedures, such as intra-vascular ultrasound and BOLD-MRI, in addition to traditional imaging, have opened new scenarios, because these tools allow to identify ischemic lesions responsive to renal revascularization. Indeed, although trials have deflated the role of renal revascularization on the renal outcomes, it should be still used to avoid dialysis initiation and/or to reduce blood pressure in selected elderly patients at high risk. Nonetheless, lifestyle modifications (smoking cessation, increased physical activity), statins and antiplatelet use, as well as cautious use of renin-angiotensin system inhibitors, remain the main therapeutic approach aimed at slowing the renal damage progression. Mesenchymal stem cells and Micro-RNA are promising target of anti-fibrotic therapy, which might provide potential benefit in ARVD patients, though safety and efficacy profile in humans is unknown too.
Collapse
Affiliation(s)
| | - Silvio Borrelli
- Nephrology and Dialysis Unit, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Carlo Garofalo
- Nephrology and Dialysis Unit, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Giorgio Fuiano
- Nephrology Units at University "Magna Graecia", Catanzaro, Italy
| | | | - Alessandro Comi
- Nephrology Units at University "Magna Graecia", Catanzaro, Italy
| | | |
Collapse
|
44
|
Morgado-Pascual JL, Rayego-Mateos S, Tejedor L, Suarez-Alvarez B, Ruiz-Ortega M. Bromodomain and Extraterminal Proteins as Novel Epigenetic Targets for Renal Diseases. Front Pharmacol 2019; 10:1315. [PMID: 31780938 PMCID: PMC6857099 DOI: 10.3389/fphar.2019.01315] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
Epigenetic mechanisms, especially DNA methylation and histone modifications, are dynamic processes that regulate the gene expression transcriptional program in normal and diseased states. The bromodomain and extraterminal (BET) protein family (BRD2, BRD3, BRD4, and BRDT) are epigenetic readers that, via bromodomains, regulate gene transcription by binding to acetylated lysine residues on histones and master transcriptional factors. Experimental data have demonstrated the involvement of some BET proteins in many pathological conditions, including tumor development, infections, autoimmunity, and inflammation. Selective bromodomain inhibitors are epigenetic drugs that block the interaction between BET proteins and acetylated proteins, thus exerting beneficial effects. Recent data have described the beneficial effect of BET inhibition on experimental renal diseases. Emerging evidence underscores the importance of environmental modifications in the origin of pathological features in chronic kidney diseases (CKD). Several cellular processes such as oxidation, metabolic disorders, cytokines, inflammation, or accumulated uremic toxins may induce epigenetic modifications that regulate key processes involved in renal damage and in other pathological conditions observed in CKD patients. Here, we review how targeting bromodomains in BET proteins may regulate essential processes involved in renal diseases and in associated complications found in CKD patients, such as cardiovascular damage, highlighting the potential of epigenetic therapeutic strategies against BET proteins for CKD treatment and associated risks.
Collapse
Affiliation(s)
- Jose Luis Morgado-Pascual
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Sandra Rayego-Mateos
- Red de Investigación Renal (REDinREN), Madrid, Spain.,Vascular and Renal Translational Research Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Lucia Tejedor
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Beatriz Suarez-Alvarez
- Red de Investigación Renal (REDinREN), Madrid, Spain.,Translational Immunology Laboratory, Health Research Institute of the Principality of Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| |
Collapse
|
45
|
Valdivielso JM, Rodríguez-Puyol D, Pascual J, Barrios C, Bermúdez-López M, Sánchez-Niño MD, Pérez-Fernández M, Ortiz A. Atherosclerosis in Chronic Kidney Disease: More, Less, or Just Different? Arterioscler Thromb Vasc Biol 2019; 39:1938-1966. [PMID: 31412740 DOI: 10.1161/atvbaha.119.312705] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Patients with chronic kidney disease (CKD) are at an increased risk of premature mortality, mainly from cardiovascular causes. The association between CKD on hemodialysis and accelerated atherosclerosis was described >40 years ago. However, more recently, it has been suggested that the increase in atherosclerosis risk is actually observed in early CKD stages, remaining stable thereafter. In this regard, interventions targeting the pathogenesis of atherosclerosis, such as statins, successful in the general population, have failed to benefit patients with very advanced CKD. This raises the issue of the relative contribution of atherosclerosis versus other forms of cardiovascular injury such as arteriosclerosis or myocardial injury to the increased cardiovascular risk in CKD. In this review, the pathophysiogical contributors to atherosclerosis in CKD that are shared with the general population, or specific to CKD, are discussed. The NEFRONA study (Observatorio Nacional de Atherosclerosis en NEFrologia) prospectively assessed the prevalence and progression of subclinical atherosclerosis (plaque in vascular ultrasound), confirming an increased prevalence of atherosclerosis in patients with moderate CKD. However, the adjusted odds ratio for subclinical atherosclerosis increased with CKD stage, suggesting a contribution of CKD itself to subclinical atherosclerosis. Progression of atherosclerosis was closely related to CKD progression as well as to the baseline presence of atheroma plaque, and to higher phosphate, uric acid, and ferritin and lower 25(OH) vitamin D levels. These insights may help design future clinical trials of stratified personalized medicine targeting atherosclerosis in patients with CKD. Future primary prevention trials should enroll patients with evidence of subclinical atherosclerosis and should provide a comprehensive control of all known risk factors in addition to testing any additional intervention or placebo.
Collapse
Affiliation(s)
- José M Valdivielso
- From the Vascular & Renal Translational Research Group and UDETMA, IRBLleida. Spanish Research Network for Renal Diseases (RedInRen. ISCIII), Lleida, Spain (J.M.V., M.B.-L.)
| | - Diego Rodríguez-Puyol
- Nephrology Unit, Fundación para la investigación del Hospital Universitario Príncipe de Asturias, RedInRen, Alcalá de Henares, Madrid, Spain (D.R.-P.)
| | - Julio Pascual
- Department of Nephrology, Institute Mar for Medical Research, Hospital del Mar, RedInRen, Barcelona, Spain (J.P., C.B.)
| | - Clara Barrios
- Department of Nephrology, Institute Mar for Medical Research, Hospital del Mar, RedInRen, Barcelona, Spain (J.P., C.B.)
| | - Marcelino Bermúdez-López
- From the Vascular & Renal Translational Research Group and UDETMA, IRBLleida. Spanish Research Network for Renal Diseases (RedInRen. ISCIII), Lleida, Spain (J.M.V., M.B.-L.)
| | - Maria Dolores Sánchez-Niño
- IIS-Fundacion Jimenez Diaz, School of Medicine, University Autonoma of Madrid, FRIAT and RedInRen, Madrid, Spain (M.D.S.-N., A.O.)
| | | | - Alberto Ortiz
- IIS-Fundacion Jimenez Diaz, School of Medicine, University Autonoma of Madrid, FRIAT and RedInRen, Madrid, Spain (M.D.S.-N., A.O.)
| |
Collapse
|