1
|
Bao Z, Yu X, Zhang L. The circ_0003928/miR-31-5p/MAPK6 cascade affects high glucose-induced inflammatory response, fibrosis and oxidative stress in HK-2 cells. Transpl Immunol 2024; 86:102078. [PMID: 38964515 DOI: 10.1016/j.trim.2024.102078] [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: 12/21/2023] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a severe diabetic complication disorder. Circular RNAs (circRNAs) actively participate in DN pathogenesis. In this report, we sought to define a new mechanism of circ_0003928 in regulating high glucose (HG)-induced HK-2 cells. METHODS To construct a DN cell model, we treated HK-2 cells with HG. Cell viability and apoptosis were detected by CCK-8 and flow cytometry, respectively. The inflammatory cytokines were quantified by ELISA. Protein analysis was performed by immunoblotting, and mRNA expression was detected by quantitative PCR. The circ_0003928/miR-31-5p and miR-31-5p/MAPK6 relationships were validated by RNA pull-down and luciferase assays. RESULTS HG promoted HK-2 cell apoptosis, fibrosis and oxidative stress. Circ_0003928 and MAPK6 levels were enhanced and miR-31-5p level was decreased in HK-2 cells after HG treatment. Circ_0003928 disruption promoted cell growth and inhibited apoptosis, inflammatory response, fibrosis and oxidative stress in HG-induced HK-2 cells. Circ_0003928 targeted miR-31-5p, and MAPK6 was a target of miR-31-5p. Circ_0003928 regulated MAPK6 expression through miR-31-5p. The functions of circ_0003928 disruption in HG-induced HK-2 cells were reversed by miR-31-5p downregulation or MAPK6 upregulation. CONCLUSION Circ_0003928 exerts regulatory impacts on HG-induced apoptosis, inflammation, fibrosis and oxidative stress in human HK-2 cells by the miR-31-5p/MAPK6 axis.
Collapse
Affiliation(s)
- Zheng Bao
- Department of Nephrology, Hanchuan People's Hospital, Hanchuan City 431600, Hubei, China
| | - Xia Yu
- Department of Nephrology, Hanchuan People's Hospital, Hanchuan City 431600, Hubei, China
| | - Lixia Zhang
- Department of Nephrology, Hanchuan People's Hospital, Hanchuan City 431600, Hubei, China.
| |
Collapse
|
2
|
Gluba-Sagr A, Franczyk B, Rysz-Górzyńska A, Olszewski R, Rysz J. The Role of Selected lncRNAs in Lipid Metabolism and Cardiovascular Disease Risk. Int J Mol Sci 2024; 25:9244. [PMID: 39273193 PMCID: PMC11395304 DOI: 10.3390/ijms25179244] [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: 03/12/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 09/15/2024] Open
Abstract
Lipid disorders increase the risk for the development of cardiometabolic disorders, including type 2 diabetes, atherosclerosis, and cardiovascular disease. Lipids levels, apart from diet, smoking, obesity, alcohol consumption, and lack of exercise, are also influenced by genetic factors. Recent studies suggested the role of long noncoding RNAs (lncRNAs) in the regulation of lipid formation and metabolism. Despite their lack of protein-coding capacity, lncRNAs are crucial regulators of various physiological and pathological processes since they affect the transcription and epigenetic chromatin remodelling. LncRNAs act as molecular signal, scaffold, decoy, enhancer, and guide molecules. This review summarises available data concerning the impact of lncRNAs on lipid levels and metabolism, as well as impact on cardiovascular disease risk. This relationship is significant because altered lipid metabolism is a well-known risk factor for cardiovascular diseases, and lncRNAs may play a crucial regulatory role. Understanding these mechanisms could pave the way for new therapeutic strategies to mitigate cardiovascular disease risk through targeted modulation of lncRNAs. The identification of dysregulated lncRNAs may pose promising candidates for therapeutic interventions, since strategies enabling the restoration of their levels could offer an effective means to impede disease progression without disrupting normal biological functions. LncRNAs may also serve as valuable biomarker candidates for various pathological states, including cardiovascular disease. However, still much remains unknown about the functions of most lncRNAs, thus extensive studies are necessary elucidate their roles in physiology, development, and disease.
Collapse
Affiliation(s)
- Anna Gluba-Sagr
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland
| | - Aleksandra Rysz-Górzyńska
- Department of Ophthalmology and Visual Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland
| | - Robert Olszewski
- Department of Gerontology, Public Health and Didactics, National Institute of Geriatrics, Rheumatology and Rehabilitation in Warsaw, 02-637 Warsaw, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland
| |
Collapse
|
3
|
Zheng G, Sun S, Zhang G, Liang X. miR-144 affects the immune response and activation of inflammatory responses in Cynoglossus semilaevis by regulating the expression of CsMAPK6. FISH & SHELLFISH IMMUNOLOGY 2024; 149:109578. [PMID: 38670413 DOI: 10.1016/j.fsi.2024.109578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/02/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
MicroRNAs are increasingly recognized for their pivotal role in the immune system, yet the specific regulatory functions of fish-derived microRNAs remain largely unexplored. In this research, we discovered a novel miRNA, Cse-miR-144, in the Chinese tongue sole (Cynoglossus semilaevis), characterized by a 73-base pair precursor and a 21-nucleotide mature sequence. Our findings revealed that the expression of Cse-miR-144 was notably inhibited by various Vibrio species. Utilizing bioinformatics and dual-luciferase assay techniques, we established that the pro-inflammatory cytokine gene CsMAPK6 is a direct target of Cse-miR-144. Subsequent in vitro and in vivo western blotting analyses confirmed that Cse-miR-144 can effectively reduce the protein levels of CsMAPK6 post-transcriptionally. Moreover, CsMAPK6 is known to be involved in the activation of the Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-kB). Additional investigations using qPCR and ELISA demonstrated that suppression of Cse-miR-144 leads to an upsurge in the liver mRNA levels of various immune genes (including MYD88, TRAF6, NF-κB, TRAF2, TRAF3, and TNF), alongside a marked increase in the production and secretion of pro-inflammatory cytokines (IL-1β, IL-6, and IL-8) in the bloodstream of C. semilaevis. These findings collectively underscore the potential of Cse-miR-144 as a key inhibitor of CsMAPK and its crucial role in modulating the immune and inflammatory responses in teleost fish. Compared to the siRNA, miRNA is a better tool in controlling the expression of target gene with a lower cost.
Collapse
Affiliation(s)
- Guiliang Zheng
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Siqi Sun
- Biodesign Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Guosong Zhang
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China.
| | - Xia Liang
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China.
| |
Collapse
|
4
|
Sahadevan P, Dingar D, Nawaito SA, Nair RS, Trépanier J, Sahmi F, Shi Y, Gillis MA, Sirois MG, Meloche S, Tardif JC, Allen BG. ERK3 is involved in regulating cardiac fibroblast function. Physiol Rep 2024; 12:e16108. [PMID: 38872461 PMCID: PMC11176743 DOI: 10.14814/phy2.16108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
ERK3/MAPK6 activates MAP kinase-activated protein kinase (MK)-5 in selected cell types. Male MK5 haplodeficient mice show reduced hypertrophy and attenuated increase in Col1a1 mRNA in response to increased cardiac afterload. In addition, MK5 deficiency impairs cardiac fibroblast function. This study determined the effect of reduced ERK3 on cardiac hypertrophy following transverse aortic constriction (TAC) and fibroblast biology in male mice. Three weeks post-surgery, ERK3, but not ERK4 or p38α, co-immunoprecipitated with MK5 from both sham and TAC heart lysates. The increase in left ventricular mass and myocyte diameter was lower in TAC-ERK3+/- than TAC-ERK3+/+ hearts, whereas ERK3 haploinsufficiency did not alter systolic or diastolic function. Furthermore, the TAC-induced increase in Col1a1 mRNA abundance was diminished in ERK3+/- hearts. ERK3 immunoreactivity was detected in atrial and ventricular fibroblasts but not myocytes. In both quiescent fibroblasts and "activated" myofibroblasts isolated from adult mouse heart, siRNA-mediated knockdown of ERK3 reduced the TGF-β-induced increase in Col1a1 mRNA. In addition, intracellular type 1 collagen immunoreactivity was reduced following ERK3 depletion in quiescent fibroblasts but not myofibroblasts. Finally, knocking down ERK3 impaired motility in both atrial and ventricular myofibroblasts. These results suggest that ERK3 plays an important role in multiple aspects of cardiac fibroblast biology.
Collapse
Affiliation(s)
- Pramod Sahadevan
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Dharmendra Dingar
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Sherin A Nawaito
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Reshma S Nair
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Joëlle Trépanier
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Fatiha Sahmi
- Montreal Heart Institute, Montréal, Québec, Canada
| | - Yanfen Shi
- Montreal Heart Institute, Montréal, Québec, Canada
| | | | - Martin G Sirois
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada
| | - Sylvain Meloche
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Bruce G Allen
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
5
|
Elkhadragy L, Myers A, Long W. Role of the Atypical MAPK ERK3 in Cancer Growth and Progression. Cancers (Basel) 2024; 16:1381. [PMID: 38611058 PMCID: PMC11011113 DOI: 10.3390/cancers16071381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/23/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Extracellular signal-regulated kinase 3 (ERK3) is an atypical mitogen-activated protein kinase (MAPK) whose structural and regulatory features are distinct from those of conventional MAPKs, such as ERK1/2. Since its identification in 1991, the regulation, substrates and functions of ERK3 have remained largely unknown. However, recent years have witnessed a wealth of new findings about ERK3 signaling. Several important biological functions for ERK3 have been revealed, including its role in neuronal morphogenesis, inflammation, metabolism, endothelial cell tube formation and epithelial architecture. In addition, ERK3 has been recently shown to play important roles in cancer cell proliferation, migration, invasion and chemoresistance in multiple types of cancers. Furthermore, accumulating studies have uncovered various molecular mechanisms by which the expression level, protein stability and activity of ERK3 are regulated. In particular, several post-translational modifications (PTMs), including ubiquitination, hydroxylation and phosphorylation, have been shown to regulate the stability and activity of ERK3 protein. In this review, we discuss recent findings regarding biochemical and cellular functions of ERK3, with a main focus on its roles in cancers, as well as the molecular mechanisms of regulating its expression and activity.
Collapse
Affiliation(s)
- Lobna Elkhadragy
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA; (L.E.); (A.M.)
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Amanda Myers
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA; (L.E.); (A.M.)
| | - Weiwen Long
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA; (L.E.); (A.M.)
| |
Collapse
|
6
|
Jiang X, Zhang M. The roles of long noncoding RNA NEAT1 in cardiovascular diseases. Hypertens Res 2024; 47:735-746. [PMID: 38177287 DOI: 10.1038/s41440-023-01551-0] [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: 07/02/2023] [Revised: 11/09/2023] [Accepted: 11/21/2023] [Indexed: 01/06/2024]
Abstract
The morbidity of cardiovascular diseases (CVDs) gradually increases worldwide. Long noncoding RNAs (lncRNAs) are a large class of non-(protein)-coding RNAs with lengths beyond 200 nucleotides. Increasing evidence suggests that lncRNA NEAT1 plays important roles in the pathogenesis of CVDs, such as myocardial infarction, heart failure, myocardial ischemia-reperfusion (I/R) injury, atherosclerosis, hypertension, cardiomyopathy, and others. We summarized the current studies of NEAT1 in CVDs, which shed light on the understanding of the molecular mechanisms of CVDs and understanding the therapeutic potential of NEAT1.
Collapse
Affiliation(s)
- Xiaoying Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| | - Mingjuan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| |
Collapse
|
7
|
He J, Yu J, Han C, Yang W, Zhang C, Hao W, Duan Y. The SNHG10-miR-495-3p-PTEN axis is involved in sevoflurane-mediated protective effects in cardiomyocytes against hypoxia/reoxygenation injury. Toxicol In Vitro 2024; 94:105724. [PMID: 37884162 DOI: 10.1016/j.tiv.2023.105724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 08/14/2023] [Accepted: 10/22/2023] [Indexed: 10/28/2023]
Abstract
Myocardial infarction (MI) has been considered a leading cause of death worldwide. Relieving ischemia-reperfusion myocardial damage is one of the major roles in treating MI. Sevoflurane postconditioning provides myocardial protection, and this study probes the mechanism of sevoflurane-mediated protective effects. A hypoxia/reoxygenation (H/R) model was constructed in cardiomyocytes, which were pretreated with 2.4% sevoflurane. Alterations in SNHG10, miR-495-3p, and PTEN levels were determined, and gain- or loss-of functional assays were conducted to confirm the role of the SNHG10/miR-495-3p axis, which is potentially regulated by sevoflurane. Cell viability, oxidative stress, and inflammatory reactions were all evaluated. The results indicated that sevoflurane post-conditioning attenuated H/R-induced cardiomyocyte damage and reduced the SHNH10 level. SNHG10 overexpression reversed sevoflurane-mediated protective effects on cardiomyocytes. Moreover, SNHG10 targeted miR-495-3p and restrained its expression, while miR-495-3p targeted PTEN, suppressed PTEN levels, and promoted HIF-1α expression. miR-495-3p overexpression decreased SNHG10-mediated myocardial injury and enhanced HIF-1α levels. However, no additional protection was found when sevoflurane was administered to H/R-exposed cardiomyocytes following treatment with the HIF-1α inhibitor LW6. Overall, sevoflurane protects cardiomyocytes from H/R by modulating the SNHG10-miR-495-3p-PTEN-HIF-1α axis.
Collapse
Affiliation(s)
- Jiandong He
- Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, Shanxi, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jing Yu
- Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, Shanxi, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Chongfang Han
- Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, Shanxi, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Wenqu Yang
- Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, Shanxi, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Chunmin Zhang
- Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, Shanxi, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Weihong Hao
- Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, Shanxi, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Yinglei Duan
- Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, Shanxi, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| |
Collapse
|
8
|
Spanos M, Gokulnath P, Chatterjee E, Li G, Varrias D, Das S. Expanding the horizon of EV-RNAs: LncRNAs in EVs as biomarkers for disease pathways. EXTRACELLULAR VESICLE 2023; 2:100025. [PMID: 38188000 PMCID: PMC10768935 DOI: 10.1016/j.vesic.2023.100025] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Extracellular vesicles (EVs) are membrane-bound nanoparticles with different types of cargo released by cells and postulated to mediate functions such as intercellular communications. Recent studies have shown that long non-coding RNAs (lncRNAs) or their fragments are present as cargo within EVs. LncRNAs are a heterogeneous group of RNA species with a length exceeding 200 nucleotides with diverse functions in cells based on their localization. While lncRNAs are known for their important functions in cellular regulation, their presence and role in EVs have only recently been explored. While certain studies have observed EV-lncRNAs to be tissue-and disease-specific, it remains to be determined whether or not this is a global observation. Nonetheless, these molecules have demonstrated promising potential to serve as new diagnostic and prognostic biomarkers. In this review, we critically evaluate the role of EV-derived lncRNAs in several prevalent diseases, including cancer, cardiovascular diseases, and neurodegenerative diseases, with a specific focus on their role as biomarkers.
Collapse
Affiliation(s)
- Michail Spanos
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Priyanka Gokulnath
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dimitrios Varrias
- Albert Einstein College of Medicine/Jacobi Medical Center, The Bronx, NY, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Song W, Qiu N. MiR-495-3p depletion contributes to myocardial ischemia/reperfusion injury in cardiomyocytes by targeting TNC. Regen Ther 2022; 21:380-388. [PMID: 36161101 PMCID: PMC9478495 DOI: 10.1016/j.reth.2022.08.007] [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: 05/05/2022] [Revised: 08/08/2022] [Accepted: 08/25/2022] [Indexed: 11/24/2022] Open
Abstract
Background Tenascin-C (TNC) has been found to abnormally express in myocardial ischemia/reperfusion injury (MI/RI), but its effect on cardiomyocytes apoptosis is unknown and is worthy of investigation. Methods H9C2 cells were given hypoxia/reoxygenation (H/R) treatment to obtain the replica of MI/RI in vitro. The effect of H/R on viability, apoptosis and inflammation was studied by CCK-8 assay, flow cytometry, mitochondrial membrane potential (MMP) and Ca2+ measurements as well as enzyme linked immunosorbent assay. We applied bioinformatics analysis and luciferase reporter assay to screened and validated TNC-targeting miR-495-3p which was then mechanistically investigated by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. With the assistance of cell transfection, rescue assays were conducted. Results H9C2 cells showed diminished viability, accelerated apoptosis, elevated tumour necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β), and TNC overexpression in response to H/R induction, while silencing of TNC partially reversed the effect of H/R treatment on the H9C2 cells. TNC silencing reduced Ca2+ level and enhanced MMP level in the H/R-stimulated cells. MiR-495-3p targeted TNC and showed a low expression in the H/R-stimulated cells. The expression of TNC was negatively regulated by miR-495-3p. Inhibition of miR-495-3p repressed viability and MMP level, and facilitated apoptosis and levels of Ca2+, TNF-α and IL-1β in the H/R-stimulated cells. The effect of TNC silencing and miR-495-3p depletion on H/R-induced cardiomyocyte injury was mutually reversed in vitro. Conclusion MiR-495-3p targeted TNC to regulate the apoptosis and inflammation of cardiomyocytes in H/R induction, which was associated with Ca2+ overload.
Collapse
Affiliation(s)
- Wei Song
- Department of Cardiology, Taizhou Central Hospital (Taizhou University Hospital), 999 Donghai Avenue, Jiaojiang District, Taizhou, Zhejiang Province, China
| | - Naiyan Qiu
- Department of Cardiology, The Fifth People's Hospital of Jinan, No. 24297 Jingshi Road, Huaiyin District, Jinan, Shandong Province, 250021, China
| |
Collapse
|
10
|
Chen S, Wang M, Jian R, Li H, Liu G, Zhou C, Xiong Y, Wang W. Circ_HIPK3 Inhibits H 2O 2-Induced Lens Epithelial Cell Injury in Age-Related Cataract Depending on the Regulation of miR-495-3p/HDAC4 Pathway. Biochem Genet 2022; 61:565-577. [PMID: 36002666 DOI: 10.1007/s10528-022-10266-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022]
Abstract
Age-related cataract (ARC) is one of the most common chronic diseases. Circular RNA (circ)_HIPK3 is reported to be involved in the advancement of ARC, but its molecular mechanism has not been clarified. Our study provides a new perspective on the clinical treatment of ARC. Our data showed that the expression levels of circ_HIPK3 and histone deacetylase 4 (HDAC4) were downregulated, while microRNA (miR)-495-3p level was increased in ARC tissues and H2O2-induced SRA01/04 cells. Functional experiments showed that circ_HIPK3 and HDAC4 overexpression could inhibit H2O2-induced lens epithelial cell apoptosis and fibrosis. In terms of mechanism, we found that circ_HIPK3 could sponge miR-495-3p, miR-495-3p could target HDAC4. Besides, we confirmed that circ_HIPK3 sponged miR-495-3p to positively regulate HDAC4. Additionally, miR-495-3p overexpression or HDAC4 knockdown reversed the inhibition effect of circ_HIPK3 on H2O2-induced lens epithelial cell injury. In conclusion, our data showed that circ_HIPK3 suppressed H2O2-induced lens epithelial cell injury by regulating miR-495-3p/HDAC4 axis.
Collapse
Affiliation(s)
- Sihui Chen
- Department of Ophthalmology, Jingmen No.1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| | - Minghong Wang
- Department of Ophthalmology, Jingmen No.1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| | - Rui Jian
- Department of Ophthalmology, Jingmen No.1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| | - Hui Li
- Department of Ophthalmology, Jingmen No.1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| | - Guoli Liu
- Department of Ophthalmology, Jingmen No.1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| | - Cuiyun Zhou
- Department of Ophthalmology, Jingmen No.1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| | - Yan Xiong
- Department of Ophthalmology, Jingmen No.1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| | - Wenqian Wang
- Department of Ophthalmology, Jingmen No.1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China.
| |
Collapse
|
11
|
Chen YE, Yang H, Pang HB, Shang FQ. Circ-CBFB exacerbates hypoxia/reoxygenation-triggered cardiomyocyte injury via regulating miR-495-3p in a VDAC1-dependent manner. J Biochem Mol Toxicol 2022; 36:e23189. [PMID: 35920438 DOI: 10.1002/jbt.23189] [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: 12/28/2021] [Revised: 05/31/2022] [Accepted: 07/25/2022] [Indexed: 11/09/2022]
Abstract
A large body of literature has identified that circular RNAs play critical roles in regulating the occurrence and development of cardiovascular disease. In the present study, we intended to provide new ideas and perspectives on the functional role of circ-CBFB in hypoxia/reoxygenation (H/R)-injured cardiomyocytes. We observed that circ-CBFB expression was enhanced which was accompanied by a miR-495-3p reduction in response to H/R exposure. Functionally, deletion of circ-CBFB obviously potentiated cell viability and restrained cell apoptosis, which was accompanied by a remarkable elevation of antiapoptotic Bcl-2 but the repression of proapoptotic Bax and cleaved caspase-3 in response to H/R. Additionally, the absence of circ-CBFB dramatically prohibited H/R-evoked cardiomyocyte oxidative stress, as revealed by a decrease in reactive oxygen species overproduction, diminution in MAD content, and enhancement in SOD, CAT, and GSH-Px activities. Moreover, elimination of circ-CBFB resulted in improvement of mitochondrial dysfunction, as assessed by mitochondrial membrane potential, adenosine triphosphate production, and the release of cyto-c. Interestingly, circ-CBFB inversely regulated miR-495-3p expression via acting as a competing endogenous RNA. VDAC1 was identified to be a functional target of miR-495-3p and positively modulated by circ-CBFB. Mechanically, dissipation of miR-495-3p or augmentation of VDAC1 manifestly counteracted the beneficial effects of circ-CBFB knockdown on H/R-elicited cardiomyocyte insult. Collectively, these observations demonstrated that absence of circ-CBFB offered cardio-protection against H/R-triggered cardiomyocyte injury by relieving apoptosis, oxidative stress, and mitochondria dysfunction through miR-495-3p/VDAC1 axis. This work unveiled an innovative axis of circ-CBFB/miR-495-3p/VDAC1 in H/R-challenged cardiomyocyte damage, exerting its potential in providing new thoughts in acute myocardial infarction management.
Collapse
Affiliation(s)
- Yue-E Chen
- Translational Medicine Center, Xi'an Chest Hospital, The Affiliated Chest Hospital of Xi'an Jiaotong University Medical School, Xi'an, Shaanxi, China
| | - Han Yang
- Translational Medicine Center, Xi'an Chest Hospital, The Affiliated Chest Hospital of Xi'an Jiaotong University Medical School, Xi'an, Shaanxi, China
| | - Hu-Bo Pang
- Department of Pneumology, People's Hospital of Zhenba County, Zhenba, Shaanxi, China
| | - Fen-Qing Shang
- Translational Medicine Center, Xi'an Chest Hospital, The Affiliated Chest Hospital of Xi'an Jiaotong University Medical School, Xi'an, Shaanxi, China
| |
Collapse
|
12
|
Liang ZY, Xu XJ, Rao J, Yang ZL, Wang CH, Chen CM. Mesenchymal Stem Cell-Derived Exosomal MiRNAs Promote M2 Macrophages Polarization: Therapeutic Opportunities for Spinal Cord Injury. Front Mol Neurosci 2022; 15:926928. [PMID: 35903172 PMCID: PMC9319398 DOI: 10.3389/fnmol.2022.926928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Spinal cord injury (SCI) is an enormous public health concern affecting approximately 250,000–500,000 people worldwide each year. It is mostly irreversible considering the limitations of currently available treatments, and its prevention and management have been the prime focus of many studies. Mesenchymal stem cell (MSC) transplantation is one of the most promising treatments for SCI. The role of MSCs in SCI has been studied extensively, and MSCs have been shown to have many limitations. Moreover, the therapeutic effects of MSCs are more likely related to paracrine effects. In SCIs, macrophages from peripheral sources differentiate into M1 macrophages, promoting inflammation and aggravating neuronal damage; however, studies have shown that MSC-derived exosomes can induce the polarization of macrophages from the M1 to the M2 phenotype, thereby promoting nerve function recovery in patients with SCI. In this review, we discussed the research progress of MSC-derived exosomal miRNAs in promoting M2 macrophage differentiation in the SCI, and introduced some exosomal miRNAs that can regulate the differentiation of M2 macrophages in non-SCI; it is hoped that the regulatory role of these exosome-derived miRNAs can be confirmed in SCI.
Collapse
Affiliation(s)
- Ze-Yan Liang
- *Correspondence: Ze-Yan Liang Chun-Hua Wang Chun-Mei Chen
| | | | | | | | - Chun-Hua Wang
- *Correspondence: Ze-Yan Liang Chun-Hua Wang Chun-Mei Chen
| | - Chun-Mei Chen
- *Correspondence: Ze-Yan Liang Chun-Hua Wang Chun-Mei Chen
| |
Collapse
|
13
|
Yang P, Liang K, Wang W, Zhou D, Chen Y, Jiang X, Fu R, Zhu B, Lin X. LncRNA SOX2-OTinhibitionprotects against myocardialischemia/reperfusion-inducedinjury via themicroRNA-186-5p (miR-186-5p)/Yin Yang 1 (YY1)pathway. Bioengineered 2022; 13:280-290. [PMID: 34967264 PMCID: PMC8805857 DOI: 10.1080/21655979.2021.2000229] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/27/2021] [Indexed: 01/15/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) exert essential effects in regulating myocardial ischemia/reperfusion (MI/R)-induced injury. This work intended to explore the functions of lncRNA SOX2-OT and its regulatory mechanism within MI/R-induced injury. In this study, gene expression was determined by RT-qPCR. Western blotting was applied for the detection of protein levels. Pro-inflammatory cytokine concentrations, cardiomyocyte viability, and apoptosis were detected via ELISA, CCK-8 and flow cytometry. In the in vitro model, SOX2-OT and YY1 were both upregulated, while miR-186-5p was downregulated. SOX2-OT knockdown attenuated oxygen-glucose deprivation/reoxygenation (OGD/R)-induced cardiomyocyte dysregulation through relieving inflammation, promoting proliferation, and reducing apoptosis in OGD/R-treated H2C9 cells. SOX2-OT positively regulated YY1 expression via miR-186-5p. Moreover, miR-186-5p inhibition or YY1 upregulation abolished the effects of SOX2-OT blocking on the inflammatory responses, proliferation, and apoptosis of OGD/R-challenged H2C9 cells. In conclusion, our results, for the first time, demonstrated that SOX2-OT inhibition attenuated MI/R injury in vitro via regulating the miR-186-5p/YY1 axis, offering potential therapeutic targets for MI/R injury treatment.
Collapse
Affiliation(s)
- Pengjie Yang
- Department of Thoracic Surgery, Affiliated People’s Hospital of Inner Mongolia Medical University
| | - Kun Liang
- Geriatric Medical Center, Inner Mongolia People’s Hospital, Hohhot, China
| | - Weisong Wang
- Department of Dispensary, Affiliated Hospital of Inner Mongolia Medical University
| | - Dehua Zhou
- Department of Emergency, People’s Hospital of Inner Mongolia Autonomous Region
| | - Yuan Chen
- Department of Pharmacy, Affiliated People’s Hospital of Inner Mongolia Medical University, Hohhot, P.R.China
| | - Xueyan Jiang
- Department of Pharmacy, Affiliated People’s Hospital of Inner Mongolia Medical University, Hohhot, P.R.China
| | - Rong Fu
- Department of Pharmacy, Affiliated People’s Hospital of Inner Mongolia Medical University, Hohhot, P.R.China
| | - Benben Zhu
- Department of Pharmacy, Affiliated People’s Hospital of Inner Mongolia Medical University, Hohhot, P.R.China
| | - Xuefeng Lin
- Department of Cardiovascular Medicine, First Affiliated Hospital of Baotou Medical College, Baotou, P.R.China
| |
Collapse
|
14
|
Yao T, Song Y, Li S, Gu J, Yan X. Inhibition of lncRNA NEAT1 protects endothelial cells against hypoxia/reoxygenation‑induced NLRP3 inflammasome activation by targeting the miR‑204/BRCC3 axis. Mol Med Rep 2021; 25:32. [PMID: 34850961 PMCID: PMC8669661 DOI: 10.3892/mmr.2021.12548] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 08/27/2020] [Indexed: 12/02/2022] Open
Abstract
Cardiovascular ischemia/reperfusion (I/R) injury is primarily caused by oxygen recovery after prolonged hypoxia. Previous studies found that the long non coding RNA (lncRNA) nuclear enriched abundant transcript 1 (NEAT1) was involved in cardiovascular pathology, and that NOD-like receptor protein 3 (NLRP3) inflammasome activation-dependent pyroptosis played a key role in cardiovascular I/R injury. The present study aimed to explore the molecular mechanism of I/R pathogenesis in order to provide novel insights for potential future therapies. Cell viability and lactate dehydrogenase enzyme activity assays were used to detect cell injury after human umbilical vein endothelial cells (HUVECs) were subjected to hypoxia/reoxygenation (H/R). The expression of the NEAT1/microRNA (miR)-204/BRCA1/BRCA2-containing complex subunit 3 (BRCC3) axis was examined by reverse transcription-quantitative PCR, and the associations among genes were confirmed by luciferase reporter assays. Western blotting and ELISA were used to measure the level of NLRP3 inflammasome activation-dependent pyroptosis. The results demonstrated that NEAT1, BRCC3 expression and NLRP3 inflammasome activation-dependent pyroptosis were significantly increased in H/R-injured HUVECs, whereas silencing BRCC3 or NEAT1 attenuated H/R-induced injury and pyroptosis. NEAT1 positively regulated BRCC3 expression via competitively binding with miR-204. Moreover, NEAT1 overexpression counteracted miR-204 mimic-induced injury, BRCC3 expression and NLRP3 inflammasome activation-dependent pyroptosis. Taken together, these findings demonstrated that inhibition of lncRNA NEAT1 protects HUVECs against H/R-induced NLRP3 inflammasome activation by targeting the miR-204/BRCC3 axis.
Collapse
Affiliation(s)
- Tao Yao
- Department of Anesthesiology, Shenzhen Bao'an Maternity and Child Health Hospital, Shenzhen, Guangdong 518100, P.R. China
| | - Yiting Song
- Department of Anesthesiology, Shenzhen Bao'an Maternity and Child Health Hospital, Shenzhen, Guangdong 518100, P.R. China
| | - Shutao Li
- Department of Anesthesiology, Shenzhen Bao'an Maternity and Child Health Hospital, Shenzhen, Guangdong 518100, P.R. China
| | - Jing Gu
- Department of Anesthesiology, Shenzhen Bao'an Maternity and Child Health Hospital, Shenzhen, Guangdong 518100, P.R. China
| | - Xuetao Yan
- Department of Anesthesiology, Shenzhen Bao'an Maternity and Child Health Hospital, Shenzhen, Guangdong 518100, P.R. China
| |
Collapse
|
15
|
The Role of Long Non-coding RNA, Nuclear Enriched Abundant Transcript 1 (NEAT1) in Cancer and Other Pathologies. Biochem Genet 2021; 60:843-867. [PMID: 34689290 DOI: 10.1007/s10528-021-10138-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/23/2021] [Indexed: 02/08/2023]
Abstract
Nuclear enriched abundant transcript 1 (NEAT1), consisting of two kinds of lncRNAs of 3.7 kB NEAT1-1 and 23 kB NEAT1-2, can be highly expressed in organs and tissues such as the ovary, prostate, colon, and pancreas, and is involved in paraspeckle formation and mRNA editing and gene expression. Therefore, NEAT1 is a potential biomarker for the treatment of a variety of diseases, which may be caused by two factors (isoforms of NEAT1 and NEAT1 sponging miRNA as ceRNA). However, there is still much confusion about the mechanism and downstream effector between the abnormal expression of NEAT1 and various diseases. This review summarizes recent research progress on NEAT1 in cancer and other pathologies and provides a more reliable theoretical basis for the treatment of related diseases.
Collapse
|
16
|
Downregulation of lncRNA NEAT1 Ameliorates LPS-Induced Inflammatory Responses by Promoting Macrophage M2 Polarization via miR-125a-5p/TRAF6/TAK1 Axis. Inflammation 2021; 43:1548-1560. [PMID: 32388658 DOI: 10.1007/s10753-020-01231-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The lncRNA nuclear enriched abundant transcript 1 (NEAT1) promotes sepsis-inflammatory responses and acute kidney injury (AKI), but little known about the underlying mechanisms. This study aims to investigate the roles of NEAT1 in regulating macrophage polarization and its potential for alleviating inflammatory responses during sepsis pathogenesis. Mouse RAW264.7 macrophages were treated with lipopolysaccharide (LPS) as a cellular inflammatory model. NEAT1 shRNA, miR-125a-5p mimics, and TRAF6-overexpressing vector were used to transfect RAW264.7 cells. NEAT1, miR-125a-5p, and mRNA levels of functional genes were detected by quantitative RT-PCR. Protein abundances were analyzed by western blotting. Macrophage polarization was evaluated by flow cytometry. The bindings of miR-125a-5p with NEAT1 or TRAF6 gene were validated by dual luciferase reporter assay. LPS treatment promoted NEAT1 and suppressed miR-125a-5p expression in mouse macrophage cells. NEAT1 silencing by shRNAs promoted macrophage M2 polarization under LPS treatment, which upregulated miR-125a-5p expression, repressed TRAF6 expression and TAK1 protein phosphorylation in macrophages. These cellular and molecular changes induced by NEAT1 shRNAs were abrogated by miR-125a-5p inhibitors. Moreover, miR-125a-5p mimics suppressed TRAF6 expression and TAK1 protein phosphorylation in LPS-treated macrophages, thus causing macrophage M2 polarization under LPS treatment. TRAF6 overexpression abrogated the miR-125a-5p mimics-induced macrophage M2 polarization. miR-125a-5p could directly bind to NEAT1 or TRAF6 gene in macrophages. lncRNA NEAT1 knockdown ameliorates LPS-induced inflammation by promoting macrophage M2 polarization via miR-125a-5p/TRAF6/TAK1 axis.
Collapse
|
17
|
Li J, Zhang Y, Zhang D, Li Y. The Role of Long Non-coding RNAs in Sepsis-Induced Cardiac Dysfunction. Front Cardiovasc Med 2021; 8:684348. [PMID: 34041287 PMCID: PMC8141560 DOI: 10.3389/fcvm.2021.684348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
Sepsis is a syndrome with life-threatening organ dysfunction induced by a dysregulated host response to infection. The heart is one of the most commonly involved organs during sepsis, and cardiac dysfunction, which is usually indicative of an extremely poor clinical outcome, is a leading cause of death in septic cases. Despite substantial improvements in the understanding of the mechanisms that contribute to the origin and responses to sepsis, the prognosis of sepsis-induced cardiac dysfunction (SICD) remains poor and its molecular pathophysiological changes are not well-characterized. The recently discovered group of mediators known as long non-coding RNAs (lncRNAs) have presented novel insights and opportunities to explore the mechanisms and development of SICD and may provide new targets for diagnosis and therapeutic strategies. LncRNAs are RNA transcripts of more than 200 nucleotides with limited or no protein-coding potential. Evidence has rapidly accumulated from numerous studies on how lncRNAs function in associated regulatory circuits during SICD. This review outlines the direct evidence of the effect of lncRNAs on SICD based on clinical trials and animal studies. Furthermore, potential functional lncRNAs in SICD that have been identified in sepsis studies are summarized with a proven biological function in research on other cardiovascular diseases.
Collapse
Affiliation(s)
- Jiawen Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yulin Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Bai XF, Niu RZ, Liu J, Pan XD, Wang F, Yang W, Wang LQ, Sun LZ. Roles of noncoding RNAs in the initiation and progression of myocardial ischemia-reperfusion injury. Epigenomics 2021; 13:715-743. [PMID: 33858189 DOI: 10.2217/epi-2020-0359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The morbidity and mortality of myocardial ischemia-reperfusion injury (MIRI) have increased in modern society. Noncoding RNAs (ncRNAs), including lncRNAs, circRNAs, piRNAs and miRNAs, have been reported in a variety of studies to be involved in pathological initiation and developments of MIRI. Hence this review focuses on the current research regarding these ncRNAs in MIRI. We comprehensively introduce the important features of lncRNAs, circRNAs, piRNA and miRNAs and then summarize the published studies of ncRNAs in MIRI. A clarification of lncRNA-miRNA-mRNA, lncRNA-transcription factor-mRNA and circRNA-miRNA-mRNA axes in MIRI follows, to further elucidate the crucial roles of ncRNAs in MIRI. Bioinformatics analysis has revealed the biological correlation of mRNAs with MIRI. We provide a comprehensive perspective for the roles of these ncRNAs and their related networks in MIRI, providing a theoretical basis for preclinical and clinical studies on ncRNA-based gene therapy for MIRI treatment.
Collapse
Affiliation(s)
- Xiang-Feng Bai
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China.,Department of Cardiovascular Surgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Rui-Ze Niu
- Department of Animal Zoology, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Jia Liu
- Department of Animal Zoology, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Xu-Dong Pan
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Feng Wang
- Department of Animal Zoology, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Wei Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Lu-Qiao Wang
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Li-Zhong Sun
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| |
Collapse
|
19
|
Long Noncoding RNAs in Myocardial Ischemia-Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8889123. [PMID: 33884101 PMCID: PMC8041529 DOI: 10.1155/2021/8889123] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/17/2020] [Accepted: 03/23/2021] [Indexed: 12/19/2022]
Abstract
Following an acute myocardial infarction, reperfusion therapy is currently the most effective way to save the ischemic myocardium; however, restoring blood flow may lead to a myocardial ischemia-reperfusion injury (MIRI). Recent studies have confirmed that long-chain noncoding RNAs (LncRNAs) play important roles in the pathophysiology of MIRIs. These LncRNA-mediated roles include cardiomyocyte apoptosis, autophagy, necrosis, oxidative stress, inflammation, mitochondrial dysfunction, and calcium overload, which are regulated through the expression of target genes. Thus, LncRNAs may be used as clinical diagnostic markers and therapeutic targets to treat or prevent MIRI. This review evaluates the research on LncRNAs involved in MIRIs and provides new ideas for preventing and treating this type of injury.
Collapse
|
20
|
Wang L, Qu P, Yin W, Sun J. Lnc-NEAT1 induces cell apoptosis and inflammation but inhibits proliferation in a cellular model of hepatic ischemia/reperfusion injury. J Int Med Res 2021; 49:300060519887251. [PMID: 33682508 PMCID: PMC7944539 DOI: 10.1177/0300060519887251] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE We aimed to investigate the effect of long non-coding RNA nuclear-enriched abundant transcript 1 (lnc-NEAT1) on regulating hepatocyte proliferation, apoptosis, and inflammation during hepatic ischemia/reperfusion (I/R) injury. METHODS Human liver cells (HL-7702) were cultured under glucose-free and oxygen-free conditions to construct the I/R injury model. Expression of lnc-NEAT1 was detected in this model and in normal cells. Plasmids of control overexpression [NC(+)], lnc-NEAT1 overexpression [NEAT1(+)], control short hairpin (sh)RNA [NC(-)], and lnc-NEAT1 shRNA [NEAT1(-)] were transfected into HL-7702 cells and subsequently subjected to I/R treatment. Cell proliferation, apoptosis, apoptosis-related proteins, and inflammatory cytokines were assessed. RESULTS Lnc-NEAT1 expression was elevated in the I/R group compared with the normal group. Cell proliferation was decreased in the NEAT1(+) group compared with the NC(+) group but increased in NEAT1(-) compared with NC(-). The apoptosis rate increased in the NEAT1(+) group compared with the NC(+) group but decreased in NEAT1(-) compared with NC(-). Western blot assay (detection of apoptosis-related proteins) showed similar results. Expression of interleukin-1β, interleukin-6, and tumor necrosis factor-α increased in the NEAT1(+) group compared with NC(+) but decreased in NEAT1(-) compared with NC(-). CONCLUSION Lnc-NEAT1 is overexpressed, induces cell apoptosis and inflammation, and inhibits proliferation during hepatic I/R injury.
Collapse
Affiliation(s)
- Liu Wang
- General Department 2, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Houhu General Department, Wuhan, China
| | - Pan Qu
- General Department 3, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanling Yin
- Department of Geratology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiao Sun
- General Department 2, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Houhu General Department, Wuhan, China
| |
Collapse
|
21
|
Silenced long non-coding RNA activated by DNA damage elevates microRNA-495-3p to suppress atherosclerotic plaque formation via reducing Krüppel-like factor 5. Exp Cell Res 2021; 401:112519. [PMID: 33636159 DOI: 10.1016/j.yexcr.2021.112519] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/22/2021] [Accepted: 02/09/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Atherosclerosis (AS) is an inflammatory disease and the formation of atherosclerotic plaque plays a critical role in AS progression. We aimed to investigate the effect of long non-coding RNA (lncRNA) activated by DNA damage (NORAD)/microRNA-495-3p (miR-495-3p)/Krüppel-like factor 5 (KLF5) axis on atherosclerotic plaque formation. METHODS The ApoE-/- mice were fed a high-fat diet to construct AS mouse models and the modeled mice were treated with altered NORAD, miR-495-3p or KLF5. NORAD, miR-495-3p and KLF5 expression in mouse aorta tissues were evaluated, and the levels of inflammatory factors, oxidative stress factors, endothelial function indices and blood lipid in mice were all determined. The atherosclerotic plaque area, lipid deposition area, collagen fibers and CD68 expression in mouse aorta tissues were assessed. The regulatory relation between NORAD and miR-495-3p, and the target relation between miR-495-3p and KLF5 were confirmed. RESULTS NORAD and KLF5 were increased whereas miR-495-3p was decreased in atherosclerotic mouse aortas. Inhibited NORAD or elevated miR-495-3p suppressed inflammation, oxidative stress, endothelial dysfunction, blood lipid level, atherosclerotic plaque area, collagen fibers and CD68 expression in atherosclerotic mouse aortas. Effects of elevated miR-495-3p on atherosclerotic mice could be reversed by up-regulation of KLF5. NORAD served as a sponge of miR-495-3p and miR-495-3p directly targeted KLF5. CONCLUSION Silenced NORAD elevated miR-495-3p to suppress atherosclerotic plaque formation via reducing KLF5. Findings in our research may be helpful for exploring molecular mechanisms of AS.
Collapse
|
22
|
Xu L, Song Q, Ouyang Z, Zhang X, Zhang C. let7f‑5p attenuates inflammatory injury in i n vitro pneumonia models by targeting MAPK6. Mol Med Rep 2020; 23:95. [PMID: 33300070 PMCID: PMC7723174 DOI: 10.3892/mmr.2020.11734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Pneumonia accounts for ~1.3 million mortalities in children per year worldwide. MicroRNAs are implicated in several diseases, including cancer and pneumonia; however, the role of let7f-5p in pneumonia is not completely understood. In the present study, lipopolysaccharide (LPS) was used to establish an in vitro pneumonia model in A549 and WI-38 cells. The reverse transcription-quantitative PCR (RT-qPCR) and western blotting results demonstrated that let7f-5p expression levels were significantly decreased, whereas MAPK6 expression levels were significantly increased in the peripheral venous blood of patients with pneumonia and in LPS-induced A549 and WI-38 cells compared with healthy volunteers and control cells, respectively. Furthermore, the dual-luciferase reporter assay demonstrated that let7f-5p targeted the 3′-untranslated region of MAPK6. The ELISA and RT-qPCR results demonstrated that let7f-5p mimic ameliorated LPS-induced inflammatory injury in A549 and WI-38 cells, as demonstrated by decreased expression levels of proinflammatory cytokines, including TNF-α and IL-6. In addition, the Cell Counting Kit-8 assay results indicated that let7f-5p mimic ameliorated LPS-induced reductions in cell viability, and the western blotting results demonstrated that let7f-5p mimic reversed LPS-induced activation of the STAT3 signaling pathway. Notably, the aforementioned let7f-5p-mediated effects were reversed by MAPK6 overexpression. Collectively, the results of the present study suggested that let7f-5p inhibited inflammation by targeting MAPK6 in the in vitro pneumonia model, thus let7f-5p may serve as a potential novel therapeutic target for pneumonia.
Collapse
Affiliation(s)
- Lin Xu
- Department of Biomedical Science, Guizhou University Medical College, Guiyang, Guizhou 550025, P.R. China
| | - Qingying Song
- Department of Anesthesiology, The Second Affiliated Hospital of Guizhou College of Traditional Chinese Medicine, Guiyang, Guizhou 550003, P.R. China
| | - Zhanghong Ouyang
- Development Planning Division, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Xiangyan Zhang
- Department of Biomedical Science, Guizhou University Medical College, Guiyang, Guizhou 550025, P.R. China
| | - Cheng Zhang
- Department of Biomedical Science, Guizhou University Medical College, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
23
|
Chen YQ, Yang X, Xu W, Yan Y, Chen XM, Huang ZQ. Knockdown of lncRNA TTTY15 alleviates myocardial ischemia-reperfusion injury through the miR-374a-5p/FOXO1 axis. IUBMB Life 2020; 73:273-285. [PMID: 33296140 DOI: 10.1002/iub.2428] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/13/2020] [Accepted: 11/30/2020] [Indexed: 01/11/2023]
Abstract
Myocardial ischemia/reperfusion (I/R) injury greatly contributes to myocardial tissue damage in patients with coronary disease, which eventually leads to heart failure. Long noncoding RNAs (lncRNAs) have an emerging role in the process of myocardial I/R injury. Our previous work revealed the protective role of miR-374a-5p against myocardial I/R injury. In this study, we explored the role of lncRNA TTTY15 and its potential interaction mechanisms with miR-374a-5p in myocardial I/R injury. The expression of TTTY15 was increased both in vitro and in vivo after myocardial I/R injury models according to quantitative real-time polymerase chain reaction. Various assays were conducted to evaluate the regulatory relationship among TTTY15, miR-374a-5p, FOXO1, and autophagy in H9c2 and HL-1 cells. The results showed that TTTY15 suppresses autophagy and myocardial I/R injury by targeting miR-374a-5p. We found that TTTY15 regulates miR-374a-5p, thus affecting FOXO1 expression and autophagy in myocytes during I/R. Furthermore, in an in vivo mouse model of myocardial I/R injury, suppression of TTTY15 successfully alleviated myocardial I/R injury. Our results reveal a novel feedback mechanism in which TTTY15 regulates miRNA processing and a potential target in myocardial I/R injury. TTTY15 is a promising therapeutic target for treating myocardial I/R injury.
Collapse
Affiliation(s)
- Yong-Quan Chen
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Xin Yang
- Department of Geriatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Wei Xu
- Department of Cardiology, Huadu District People's Hospital of Guangzhou, Guangzhou, PR China
| | - Yi Yan
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Xi-Ming Chen
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Zhao-Qi Huang
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| |
Collapse
|
24
|
Gong C, Zhou X, Lai S, Wang L, Liu J. Long Noncoding RNA/Circular RNA-miRNA-mRNA Axes in Ischemia-Reperfusion Injury. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8838524. [PMID: 33299883 PMCID: PMC7710414 DOI: 10.1155/2020/8838524] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/30/2020] [Accepted: 10/30/2020] [Indexed: 12/18/2022]
Abstract
Ischemia-reperfusion injury (IRI) elicits tissue injury involved in a wide range of pathologies. Multiple studies have demonstrated that noncoding RNAs (ncRNAs), including long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), and microRNAs (miRNAs), participate in the pathological development of IRI, and they may act as biomarkers, therapeutic targets, or prognostic indicators. Nonetheless, the specific molecular mechanisms of ncRNAs in IRI have not been completely elucidated. Regulatory networks among lncRNAs/circRNAs, miRNAs, and mRNAs have been the focus of attention in recent years. Studies on the underlying molecular mechanisms have contributed to the discovery of therapeutic targets or strategies in IRI. In this review, we comprehensively summarize the current research on the lncRNA/circRNA-miRNA-mRNA axes and highlight the important role of these axes in IRI.
Collapse
Affiliation(s)
- Chengwu Gong
- Department of Cardiothoracic Surgery, Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xueliang Zhou
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Songqing Lai
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Lijun Wang
- Department of Cardiothoracic Surgery, Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jichun Liu
- Department of Cardiothoracic Surgery, Second Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
25
|
Song XL, Zhang FF, Wang WJ, Li XN, Dang Y, Li YX, Yang Q, Shi MJ, Qi XY. LncRNA A2M-AS1 lessens the injury of cardiomyocytes caused by hypoxia and reoxygenation via regulating IL1R2. Genes Genomics 2020; 42:1431-1441. [PMID: 33057899 DOI: 10.1007/s13258-020-01007-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Myocardial ischemia and reperfusion injury (MI/RI) is a complex pathophysiological process, which can lead to severe myocardial injury. The long noncoding RNA alpha-2-macroglobulin antisense RNA 1 (A2M-AS1) has been revealed to be abnormally expressed in MI, However, its function in MI and the potential mechanism are still unclear. OBJECTIVE To evaluate the functional role of A2M-AS1 in hypoxia/reoxygenation (H/R)-induced neonatal cardiomyocytes and its potential molecular mechanism. METHODS Dataset GSE66360 was obtained from GEO database for analyzing the RNA expression of A2M-AS1 and interleukin 1 receptor type 2 (IL1R2). KEGG pathway enrichment analysis of the genes that co-expressed with A2M-AS1 was performed. Human neonatal cardiomyocytes were subjected to H/R to construct in vitro models. QRT-PCR and Western blot were adopted to test the levels of mRNA and protein. The viability and apoptosis of cardiomyocytes were tested by CCK-8 and flow cytometry assays, respectively. RESULTS The expression of A2M-AS1 was notably downregulated in H/R-treated cardiomyocytes. Overexpression of A2M-AS1 can notably enhance the cell viability of H/R-damaged cardiomyocytes, whereas knockdown of A2M-AS1 showed the opposite outcomes. Besides, a negative correlation was showed between A2M-AS1 and IL1R2 expression. In H/R-treated cardiomyocytes, overexpression of IL1R2 weakened the promoting proliferation and anti-apoptosis effects caused by overexpressing A2M-AS1, however, IL1R2-knockdown abolished the anti-proliferation and pro-apoptosis effects caused by silencing A2M-AS1. CONCLUSION This study demonstrates the potential regulatory role of A2M-AS1/ IL1R2 axis in cardiomyocytes suffered from H/R, and provides insight into the protection of MI/RI.
Collapse
Affiliation(s)
- Xue-Lian Song
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Fei-Fei Zhang
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Wen-Jing Wang
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Xin-Ning Li
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Yi Dang
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Ying-Xiao Li
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Qian Yang
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Mei-Jing Shi
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Xiao-Yong Qi
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China.
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China.
| |
Collapse
|
26
|
Chen Z, Yan Y, Wu J, Qi C, Liu J, Wang J. Expression level and diagnostic value of exosomal NEAT1/miR-204/MMP-9 in acute ST-segment elevation myocardial infarction. IUBMB Life 2020; 72:2499-2507. [PMID: 32916037 DOI: 10.1002/iub.2376] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/09/2020] [Accepted: 08/16/2020] [Indexed: 12/21/2022]
Abstract
Acute myocardium infarction (AMI) is one of the main causes of cardiovascular death, and timely intervention and diagnosis are essential. Owing to the irreversible apoptosis and death of myocardial cells, which ultimately causes heart failure, the problem of myocardial repair after myocardial infarction needs to be urgently addressed. Exosomes can act as messengers between cells, delivering large amounts of proteins, RNA, and lipids to receptor cells, and regulating target cell functions. Studies have shown that exosomes can repair infarcted myocardium. We aimed to investigate the relationship between long non-coding RNA NEAT1 in serum exosomes of patients and AMI and its underlying mechanism. Subjects were divided into control, UA, and STEMI groups. RNA was extracted from the serum exosomes, and the expressions of lncRNA NEAT1 and miR-204 were detected by qRT-PCR. MMP-9 was detected by western blot, Spearman test was used to analyze the correlation among the three. Logistic regression and Receiver-operating characteristic curve (ROC) were used to evaluate the prediction of acute myocardial infarction. The expressions of NEAT1 and MMP-9 in serum exosomes of patients with acute ST-segment elevation myocardial infarction were up-regulated and positively correlated, miR-204 expression was down-regulated, there were no correlations between miR-204 with NEAT1, or MMP-9. Exosomal NEAT1, miR-204, and MMP-9 displayed potent biomarkers for diagnosis of acute ST-segment elevation myocardial infarction.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Junduo Wu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Chao Qi
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Jia Liu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
27
|
Zong L, Wang W. CircANXA2 Promotes Myocardial Apoptosis in Myocardial Ischemia-Reperfusion Injury via Inhibiting miRNA-133 Expression. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8590861. [PMID: 32685535 PMCID: PMC7334784 DOI: 10.1155/2020/8590861] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This project is aimed at investigating whether CircANXA2 can promote the apoptosis of myocardial cells by inhibiting miR-133 expression and thereby participate in the development of myocardial ischemia-reperfusion injury. Materials and Method. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the expression level of CircANXA2 in H9c2 cells after hypoxia/reoxygenation (H/R) treatment. Evaluation of myocardial injury markers in H9c2 cells was performed using commercial kits, including lactate dehydrogenase (LDH), malonaldehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidation (GSH-PX). MTT analysis and flow cytometry were used to detect myocardial cell proliferation and apoptosis, respectively. Western blot was used to detect the protein expression of apoptosis-related genes. RESULT qRT-PCR results showed that compared with the control, the expression of CircANXA2 was upregulated and the expression level of miR-133 was significantly decreased in H/R-treated H9c2 cells. CircANXA2 overexpression increased LDH, MDA, SOD, and GSH-PX activity in H/R-treated H9c2 cells. At the same time, CircANXA2 overexpression inhibited the proliferation of H/R-treated cells, and CircANXA2 was able to induce cardiomyocyte apoptosis. Western blot results showed that after overexpression of CircANXA2, the proapoptotic genes Bax and cytochrome C was upregulated, while the antiapoptotic gene Bcl-2 was downregulated. In H9c2 cells, upregulating miR-133 can reverse the inhibition of proliferation induced by CircANXA2 overexpression and increase apoptosis. CONCLUSIONS CircANXA2 promotes cardiomyocyte apoptosis in myocardial ischemia-reperfusion injury by inhibiting the expression of miR-133. CircANXA2 may be a potential target for myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Liang Zong
- Department of Cardiac Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Weixin Wang
- Department of Cardiac Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| |
Collapse
|
28
|
Ren L, Wang Q, Ma L, Wang D. MicroRNA-760-mediated low expression of DUSP1 impedes the protective effect of NaHS on myocardial ischemia-reperfusion injury. Biochem Cell Biol 2020; 98:378-385. [PMID: 32160475 DOI: 10.1139/bcb-2019-0310] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is the leading cause of the poor prognosis for patients undergoing clinical cardiac surgery. Micro-RNAs are involved in MIRI; however, the effect of miR-760 on MIRI and the molecular mechanisms behind it have not yet been described. For our in-vivo experiments, 20 rats were randomly distributed between 2 groups (n = 10): the sham-treatment group and the ischemia-reperfusion (I/R) group. For our in-vitro experiments, H9C2 cells were subjected to hypoxia for 6 h, and then reoxygenated to establish an hypoxia-reoxygenation (H/R) model. High expression levels of of miR-760 were observed in the rats subjected to MIRI and the H9C2 cells subjected to H/R. Further, the levels of lactate dehydrogenase (LDH) and malonaldehyde (MDA) were increased, and the size of the myocardial infarct was notably greater in the rats subjected to MIRI, suggesting that miR-760 worsens the effects of MIRI. The inhibitory effects from NaHS on apoptosis were enhanced, as were the expression levels of cleaved caspase 3 and cleaved PARP in H9C2 cells exposed to H/R, and with low-expression levels of miR-760. TargetScan and dual luciferase reporter assays further confirmed the targeted relationship between dual-specificity protein phosphatase (DUSP1) and miR-760. Additionally, miR-760 overexpression and H/R treatment of H9C2 cells inhibited the expression of DUSP1, which further promoted apoptosis. Furthermore, DUSP1 enhanced the anti-apoptotic effects of NaHS in rats subjected to MIRI. Taken together, these findings suggest that miR-760 inhibits the protective effect of NaHS against MIRI.
Collapse
Affiliation(s)
- Lin Ren
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang City, Hebei Province 050011, China.,Department of Cardiology, First Hospital of Qinhuangdao, Qinhuangdao City, Hebei Province 066000, China
| | - Qian Wang
- Department of Geriatrics, First Hospital of Qinhuangdao, Qinhuangdao City, Hebei Province 066000, China
| | - Lixiang Ma
- Department of Cardiology, First Hospital of Qinhuangdao, Qinhuangdao City, Hebei Province 066000, China
| | - Dongmei Wang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang City, Hebei Province 050011, China
| |
Collapse
|