1
|
Cao J, Du L, Zhao X, Liu Z, Yuan J, Luo Y, Zhang S, Qin Z, Guo J. LncRNA sequencing reveals an essential role for the lncRNA-mediated ceRNA network in penile squamous cell carcinoma. Genes Immun 2024; 25:447-458. [PMID: 39242755 DOI: 10.1038/s41435-024-00295-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024]
Abstract
Penile squamous cell carcinoma (PSCC) is becoming increasingly common and posing a severe threat to men's health, particularly in developing countries. The function of long non-coding RNAs (lncRNAs) in PSCC progression remains mysterious. Therefore, we explored the significance of lncRNAs in the competing endogenous RNA (ceRNA) network in PSCC tumor progression. The 5 healthy and 6 tumor tissue samples were subjected to lncRNA sequencing. Using miRcode, LncBase, miRTarBase, miRWalk, and TargetScan, we constructed a ceRNA network of differentially expressed lncRNAs, miRNAs, and mRNAs. Our analysis resulted in a ceRNA network consisting of 4 lncRNAs, 18 miRNAs, and 38 mRNAs, whose upstream regulators, the lncRNAs MIR205HG, MIAT, HCP5, and PVT1, were all elevated in PSCC. Immunohistochemical staining confirmed that cell proliferation-related genes TFAP2C, MKI67, and TP63, positively regulated by 4 lncRNAs, were considerably overexpressed in tumor tissues. Immune analysis revealed a significant upregulation in macrophage and exhausted T cell infiltration in PSCC. Our study identified a lncRNA-miRNA-mRNA ceRNA network for PSCC, revealing possible molecular mechanisms involved in the regulation of PSCC progression by key lncRNAs and their connections to the immunosuppressive tumor microenvironment. The ceRNA network provides a novel perspective for elucidating the pathogenesis of PSCC.
Collapse
Affiliation(s)
- Jian Cao
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, China
| | - Lin Du
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xueheng Zhao
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Zhizhong Liu
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, China
| | - Junbin Yuan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanwei Luo
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shanshan Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Zailong Qin
- Guangxi Key Laboratory of Reproductive Health and Birth Defect Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
- Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
- Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
- Genetic and Metabolic Central Laboratory, Birth Defects Prevention and Control Institute of Guangxi Zhuang Autonomous Region, Nanning, China.
- Guangxi Clinical Research Center for Pediatric Diseases, Nanning, China.
| | - Jie Guo
- National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, Changsha, China.
- China National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
2
|
Luo X, Guo X, Chen N, Peng R, Pan C, Li Z, Zhao B, Ji R, Li S. miR-155 mediated regulation of PKG1 and its implications on cell invasion, migration, and apoptosis in preeclampsia through NF-κB pathway. Biol Direct 2024; 19:121. [PMID: 39587640 PMCID: PMC11590512 DOI: 10.1186/s13062-024-00526-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/02/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Preeclampsia (PE) is a severe pregnancy complication characterized by complex molecular interactions. Understanding these interactions is crucial for developing effective therapeutic strategies. METHODS This study applies a pharmacometabolomics approach to explore the roles of miR-155 and PKG1 in PE, focusing on the regulatory influence of the NF-κB signaling pathway. Blood metabolomic profiles were analyzed, and bioinformatics tools, IHC staining, Western blot (WB) analysis, and immunofluorescence (IF) localization were employed to determine the expression and function of miR-155 and PKG1. Cell invasion, migration, proliferation, and apoptosis assays were conducted to assess miR-155's modulation of PKG1. Additionally, RT-qPCR and WB analysis elucidated NF-κB-mediated regulation mechanisms. RESULTS Our findings indicate significant metabolic alterations associated with miR-155 modulation of PKG1, with NF-κB acting as a critical upstream regulator. The study demonstrates that miR-155 affects cellular functions such as invasion, migration, proliferation, and apoptosis through PKG1 modulation. Furthermore, the NF-κB signaling pathway regulates miR-155 expression, contributing to the pathological processes of PE. CONCLUSION This study provides a proof of concept for using pharmacometabolomics to understand the molecular mechanisms of PE, suggesting new therapeutic targets and advancing personalized medicine approaches. These insights highlight the potential of pharmacometabolomics to complement genomic and transcriptional data in disease characterization and treatment strategies, offering new avenues for therapeutic intervention in PE.
Collapse
Affiliation(s)
- Xiaohua Luo
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, No.7 Kangfuqian Street, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Xiaopei Guo
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, No.7 Kangfuqian Street, Erqi District, Zhengzhou, 450052, Henan, China
| | - Ningning Chen
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, No.7 Kangfuqian Street, Erqi District, Zhengzhou, 450052, Henan, China
| | - Rui Peng
- Scientific Research Department, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ci Pan
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, No.7 Kangfuqian Street, Erqi District, Zhengzhou, 450052, Henan, China
| | - Zhuyin Li
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, No.7 Kangfuqian Street, Erqi District, Zhengzhou, 450052, Henan, China
| | - Bing Zhao
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, No.7 Kangfuqian Street, Erqi District, Zhengzhou, 450052, Henan, China
| | - Ruonan Ji
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, No.7 Kangfuqian Street, Erqi District, Zhengzhou, 450052, Henan, China
| | - Siyu Li
- Department of Obstetrics and Gynaecology, The Third Affiliated Hospital of Zhengzhou University, No.7 Kangfuqian Street, Erqi District, Zhengzhou, 450052, Henan, China
| |
Collapse
|
3
|
Sun L, Yang K, Wang L, Wu S, Wen D, Wang J. LncRNA MIAT suppresses inflammation in LPS-induced J774A.1 macrophages by promoting autophagy through miR-30a-5p/SOCS1 axi. Sci Rep 2024; 14:22608. [PMID: 39349964 PMCID: PMC11442610 DOI: 10.1038/s41598-024-73607-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
Accumulated data implicate that long noncoding RNA (lncRNA) plays a pivotal role in rheumatoid arthritis (RA), potentially serving as a competitive endogenous RNA (ceRNA) for microRNAs (miRNAs). The lncRNA myocardial infarction-associated transcript (MIAT) has been demonstrated to regulate inflammation. However, the role of MIAT in the inflammation of RA remains inadequately explored. This study aims to elucidate MIAT's role in the inflammation of lipopolysaccharide (LPS)-induced macrophages and to uncover the underlying molecular mechanisms. We observed heightened MIAT expression in LPS-induced J774A.1 cells and collagen-induced arthritis mouse models, in contrast to the expression pattern of miR-30a-5p. Silencing MIAT resulted in increased expression of the inflammatory cytokines IL-1β and TNF-α. Simultaneously, MIAT interference significantly impeded macrophage autophagy, evidenced by decreased expression of autophagy-related markers LC3-II and Beclin-1, alongside increased levels of p62 in LPS-induced J774A.1 cells. Notably, MIAT functioned as a ceRNA, sponging miR-30a-5p and exerting a negative regulatory influence on its expression. SOCS1 emerged as a target of miR-30a-5p, modulated by MIAT. Mechanistically, inhibiting miR-30a-5p reversed the impact of MIAT deficiency in promoting LPS-induced inflammation, while SOCS1 knockdown countered the cytokine inhibitory effect induced by silencing miR-30a-5p. In summary, this study indicates that lncRNA MIAT suppresses inflammation in LPS-induced J774A.1 macrophages by stimulating autophagy through the miR-30a-5p/SOCS1 axis. This suggests that MIAT holds promise as a potential therapeutic target for RA inflammation.
Collapse
Affiliation(s)
- Linqian Sun
- Department of Rheumatology & Clinical Immunology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Kun Yang
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Liqin Wang
- Department of Rheumatology & Clinical Immunology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Si Wu
- Department of Infectious Disease, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Dawei Wen
- Department of Rheumatology & Clinical Immunology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Jibo Wang
- Department of Rheumatology & Clinical Immunology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
4
|
Chuang TD, Ton N, Manrique N, Rysling S, Khorram O. Targeting the long non-coding RNA MIAT for the treatment of fibroids in an animal model. Clin Sci (Lond) 2024; 138:699-709. [PMID: 38817011 PMCID: PMC11166562 DOI: 10.1042/cs20240190] [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: 01/29/2024] [Revised: 05/03/2024] [Accepted: 05/30/2024] [Indexed: 06/01/2024]
Abstract
Our previous studies indicated that there is overexpression of MIAT in fibroids and MIAT is a sponge for the miR-29 family in these tumors. The objective of the present study was to determine if the knockdown of MIAT in fibroid xenografts will increase miR-29 levels and reduce the expression of genes targeted by this miRNA such as collagen and cell cycle regulatory proteins in a mouse model for fibroids. Ovariectomized CB-17 SCID/Beige mice bearing estrogen/progesterone pellets were implanted subcutaneously in the flank with equal weight of fibroid explants which had been transduced by lentivirus for either control (empty vector) or MIAT knockdown for four weeks (n=7). Knockdown of MIAT in fibroid xenografts resulted in a 30% reduction of tumor weight and a marked increase in miR-29a, -b, and -c levels in the xenografts. There was reduced cell proliferation and expression of cell cycle regulatory genes CCND1, CDK2, and E2F1 and no significant changes in apoptosis. The xenografts with MIAT knockdown expressed lower mRNA and protein levels of FN1, COL3A1, and TGF-β3, and total collagen protein. Targeting MIAT, which sponges the pro-fibrotic miR-29 family, is an effective therapy for fibroids by reducing cell proliferation and thereby, tumor growth and accumulation of ECM, which is a hallmark of these benign gynecologic tumors.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA, U.S.A
- The Lundquist Institute for Biomedical Innovation, Torrance, CA, U.S.A
| | - Nhu Ton
- The Lundquist Institute for Biomedical Innovation, Torrance, CA, U.S.A
| | - Nathaly Manrique
- The Lundquist Institute for Biomedical Innovation, Torrance, CA, U.S.A
| | - Shawn Rysling
- The Lundquist Institute for Biomedical Innovation, Torrance, CA, U.S.A
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA, U.S.A
- The Lundquist Institute for Biomedical Innovation, Torrance, CA, U.S.A
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, U.S.A
| |
Collapse
|
5
|
Elmasri RA, Rashwan AA, Gaber SH, Rostom MM, Karousi P, Yasser MB, Kontos CK, Youness RA. Puzzling out the role of MIAT LncRNA in hepatocellular carcinoma. Noncoding RNA Res 2024; 9:547-559. [PMID: 38515792 PMCID: PMC10955557 DOI: 10.1016/j.ncrna.2024.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/31/2023] [Accepted: 01/09/2024] [Indexed: 03/23/2024] Open
Abstract
A non-negligible part of our DNA has been proven to be transcribed into non-protein coding RNA and its intricate involvement in several physiological processes has been highly evidenced. The significant biological role of non-coding RNAs (ncRNAs), including long non-coding RNAs (lncRNAs) has been variously reported. In the current review, the authors highlight the multifaceted role of myocardial infarction-associated transcript (MIAT), a well-known lncRNA, in hepatocellular carcinoma (HCC). Since its discovery, MIAT has been described as a regulator of carcinogenesis in several malignant tumors and its overexpression predicts poor prognosis in most of them. At the molecular level, MIAT is closely linked to the initiation of metastasis, invasion, cellular migration, and proliferation, as evidenced by several in-vitro and in-vivo models. Thus, MIAT is considered a possible theranostic agent and therapeutic target in several malignancies. In this review, the authors provide a comprehensive overview of the underlying molecular mechanisms of MIAT in terms of its downstream target genes, interaction with other classes of ncRNAs, and potential clinical implications as a diagnostic and/or prognostic biomarker in HCC.
Collapse
Affiliation(s)
- Rawan Amr Elmasri
- Molecular Genetics Research Team (MGRT), Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), New Administrative Capital, 11835, Cairo, Egypt
| | - Alaa A. Rashwan
- Molecular Genetics Research Team (MGRT), Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), New Administrative Capital, 11835, Cairo, Egypt
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo (AUC), 11835, Cairo, Egypt
| | - Sarah Hany Gaber
- Molecular Genetics Research Team (MGRT), Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), New Administrative Capital, 11835, Cairo, Egypt
| | - Monica Mosaad Rostom
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), 11835, Cairo, Egypt
| | - Paraskevi Karousi
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15701, Athens, Greece
| | - Montaser Bellah Yasser
- Bioinformatics Group, Center for Informatics Sciences (CIS), School of Information Technology and Computer Science (ITCS), Nile University, Giza, Egypt
| | - Christos K. Kontos
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15701, Athens, Greece
| | - Rana A. Youness
- Molecular Genetics Research Team (MGRT), Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), New Administrative Capital, 11835, Cairo, Egypt
| |
Collapse
|
6
|
Zeinelabdeen Y, Abaza T, Yasser MB, Elemam NM, Youness RA. MIAT LncRNA: A multifunctional key player in non-oncological pathological conditions. Noncoding RNA Res 2024; 9:447-462. [PMID: 38511054 PMCID: PMC10950597 DOI: 10.1016/j.ncrna.2024.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/27/2023] [Accepted: 01/14/2024] [Indexed: 03/22/2024] Open
Abstract
The discovery of non-coding RNAs (ncRNAs) has unveiled a wide range of transcripts that do not encode proteins but play key roles in several cellular and molecular processes. Long noncoding RNAs (lncRNAs) are specific class of ncRNAs that are longer than 200 nucleotides and have gained significant attention due to their diverse mechanisms of action and potential involvement in various pathological conditions. In the current review, the authors focus on the role of lncRNAs, specifically highlighting the Myocardial Infarction Associated Transcript (MIAT), in non-oncological context. MIAT is a nuclear lncRNA that has been directly linked to myocardial infarction and is reported to control post-transcriptional processes as a competitive endogenous RNA (ceRNA) molecule. It interacts with microRNAs (miRNAs), thereby limiting the translation and expression of their respective target messenger RNA (mRNA) and regulating protein expression. Yet, MIAT has been implicated in other numerous pathological conditions such as other cardiovascular diseases, autoimmune disease, neurodegenerative diseases, metabolic diseases, and many others. In this review, the authors emphasize that MIAT exhibits distinct expression patterns and functions across different pathological conditions and is emerging as potential diagnostic, prognostic, and therapeutic agent. Additionally, the authors highlight the regulatory role of MIAT and shed light on the involvement of lncRNAs and specifically MIAT in various non-oncological pathological conditions.
Collapse
Affiliation(s)
- Yousra Zeinelabdeen
- Molecular Genetics Research Team, Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), Cairo, 11835, Egypt
- Faculty of Medical Sciences/UMCG, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713 AV, the Netherlands
| | - Tasneem Abaza
- Molecular Genetics Research Team, Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), Cairo, 11835, Egypt
- Biotechnology and Biomolecular Biochemistry Program, Faculty of Science, Cairo University, Cairo, Egypt
| | - Montaser Bellah Yasser
- Bioinformatics Group, Center for Informatics Sciences (CIS), School of Information Technology and Computer Science (ITCS), Nile University, Giza, Egypt
| | - Noha M. Elemam
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Rana A. Youness
- Molecular Genetics Research Team, Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), Cairo, 11835, Egypt
| |
Collapse
|
7
|
Xia J, Huang Y, Ma M, Liu F, Cao B. Downregulating lncRNA MIAT attenuates apoptosis of podocytes exposed to high glucose. Acta Diabetol 2024; 61:451-460. [PMID: 38072843 DOI: 10.1007/s00592-023-02213-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/08/2023] [Indexed: 03/27/2024]
Abstract
AIMS Diabetic nephropathy (DN), a destructive complication of diabetes mellitus (DM), is one of the leading causes of end-stage renal disease (ESRD). This study aimed to investigate the role of long non-coding RNA (lncRNA) MIAT in high-glucose (HG)-induced podocyte injury associated with DN. METHODS Three human kidney podocyte (HKP) cultures were treated with HG to mimic DN. Expression of lncRNA MIAT, podocyte-specific and injury-related proteins, and apoptosis were assessed before and after MIAT knockdown using MIAT shRNAs. RESULTS MIAT expression was upregulated in HKPs in response to glucose stress. HG treatment resulted in a significant increase in the apoptotic rate, Bax level, and levels of injury-related proteins desmin, fibroblast-specific protein 1 (FSP-1), and smooth muscle α-actin (α-SMA), and a significant reduction in Bcl-2 levels and the levels of podocyte-specific proteins synaptopodin and podocin. Transfection of HKPs with shRNAs significantly reduced MIAT levels (p < 0.05) and attenuated apoptosis in HG-medium. Correspondingly, the levels of synaptopodin and podocin were upregulated, and desmin, FSP-1, and α-SMA were reduced (p < 0.05). Western blot analysis also showed that anti-apoptotic active caspase-3 and Bax and proapoptotic Bcl-2 were elevated and decreased, respectively, after MIAT knockdown, suggesting that apoptosis pathways are deactivated after MIAT downregulation. CONCLUSIONS High glucose upregulates MIAT level in HKPs and induces cellular injury. Knockdown of MIAT alleviates the injury likely via deactivating apoptosis pathways.
Collapse
Affiliation(s)
- Jiayi Xia
- Department of Endocrinology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Yan Huang
- Department of Medical Insurance, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Min Ma
- Department of Gynecology, Graduate School of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Fang Liu
- Department of Coloproctology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China.
| | - Bo Cao
- Department of Coloproctology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China.
| |
Collapse
|
8
|
Gandhi P, Wang Y, Li G, Wang S. The role of long noncoding RNAs in ocular angiogenesis and vascular oculopathy. Cell Biosci 2024; 14:39. [PMID: 38521951 PMCID: PMC10961000 DOI: 10.1186/s13578-024-01217-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/05/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are RNA transcripts over 200 nucleotides in length that do not code for proteins. Initially considered a genomic mystery, an increasing number of lncRNAs have been shown to have vital roles in physiological and pathological conditions by regulating gene expression through diverse mechanisms depending on their subcellular localization. Dysregulated angiogenesis is responsible for various vascular oculopathies, including diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, and corneal neovascularization. While anti-VEGF treatment is available, it is not curative, and long-term outcomes are suboptimal, and some patients are unresponsive. To better understand these diseases, researchers have investigated the role of lncRNAs in regulating angiogenesis and models of vascular oculopathies. This review summarizes recent research on lncRNAs in ocular angiogenesis, including the pro-angiogenic lncRNAs ANRIL, HOTAIR, HOTTIP, H19, IPW, MALAT1, MIAT, NEAT1, and TUG1, the anti-angiogenic lncRNAs MEG3 and PKNY, and the human/primate specific lncRNAs lncEGFL7OS, discussing their functions and mechanisms of action in vascular oculopathies.
Collapse
Affiliation(s)
- Pranali Gandhi
- Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Yuzhi Wang
- Louisiana State University School of Medicine, New Orleans, LA, 70112, USA
| | - Guigang Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei province, P.R. China.
| | - Shusheng Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA.
- Department of Ophthalmology, Tulane University, New Orleans, LA, 70112, USA.
- Tulane Personalized Health Institute, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
9
|
Weng YH, Chen J, Yu WT, Luo YP, Liu C, Yang J, Liu HB. lncRNA-MIAT rs9625066 polymorphism could be a potential biomarker for ischemic stroke. BMC Med Genomics 2024; 17:58. [PMID: 38383415 PMCID: PMC10882908 DOI: 10.1186/s12920-024-01830-w] [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: 12/16/2023] [Accepted: 02/10/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Ischemic stroke (IS) is a common and serious neurological condition that is highly fatal but so far no early diagnostic markers are available. Myocardial infarction-associated transcript (MIAT) is a long non-coding RNA (lncRNA) that could lead to IS by inducing autophagy and apoptosis in neuronal cells. However, there has been no report on the link between susceptibility to IS and the single-nucleotide polymorphisms (SNPs) of MIAT. This study aimed to investigate the association between MIAT gene polymorphisms and IS risk. METHODS A total of 320 IS patients and 310 age-, sex- and race-matched controls were included in this study. Four polymorphisms (rs2157598, rs5761664, rs1894720, and rs9625066) were genotyped by using SNPscan technique. RESULTS Among the 4 polymorphisms of MIAT, only rs9625066 was associated with IS risk (CA vs. CC: adjusted OR = 0.55, 95% CI, 0.37-0.85, P = 0.006; AA vs. CC: adjusted OR = 0.39, 95% CI, 0.16-0.94, P = 0.036; (AA + CA vs. CC: adjusted OR = 0.53, 95% CI, 0.35-0.80, P = 0.002; A vs. C adjusted OR = 0.59, 95% CI, 0.42-0.82, P = 0.002). Haplotype analysis showed a 1.32-fold increase (95% CI, 1.05-1.67, P = 0.017) in IS risk for rs2157598-rs5761664-rs1894720-rs9625066 (A-C-G-C). Logistic regression analysis identified some independent impact factors for IS including rs9625066 AA/AC, TC, TG, HDL-C (P < 0.05). CONCLUSION The rs9625066 polymorphism of MIAT might be associated with IS susceptibility in Chinese population, in which AA/CA plays a protective role in IS, whereas the CC genotype increases the risk of developing IS, suggesting it might be a marker predictive of IS risk.
Collapse
Affiliation(s)
- Yin-Hua Weng
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- School of Clinical Medicine, Guilin Medical University, Guilin, China
- College of Medical Laboratory Science, Guilin Medical University, Guilin, China
| | - Jie Chen
- Department of Laboratory Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Wen-Tao Yu
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Yan-Ping Luo
- Department of Laboratory Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
- School of Clinical Medicine, Guilin Medical University, Guilin, China
| | - Chao Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- School of Clinical Medicine, Guilin Medical University, Guilin, China
- College of Medical Laboratory Science, Guilin Medical University, Guilin, China
| | - Jun Yang
- Department of Laboratory Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China.
| | - Hong-Bo Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China.
- College of Medical Laboratory Science, Guilin Medical University, Guilin, China.
- Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, China.
| |
Collapse
|
10
|
Zhang L, Zhang C, Zheng J, Wang Y, Wei X, Yang Y, Zhao Q. miR-155-5p/Bmal1 Modulates the Senescence and Osteogenic Differentiation of Mouse BMSCs through the Hippo Signaling Pathway. Stem Cell Rev Rep 2024; 20:554-567. [PMID: 38150082 PMCID: PMC10837250 DOI: 10.1007/s12015-023-10666-3] [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] [Accepted: 12/18/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND The core clock gene brain and muscle ARNT like-1 (Bmal1) is involved in the regulation of bone tissue aging. However, current studies are mostly limited to the establishment of the association between Bmal1 and bone senescence, without in-depth exploration of its main upstream and downstream regulatory mechanisms. METHODS The luciferase reporter assay, RT-qPCR and Western blotting were performed to detect the interaction between miR-155-5p and Bmal1. The effects of miR-155-5p and Bmal1 on the aging and osteogenic differentiation ability of mouse bone marrow mesenchymal stem cells (BMSCs) were investigated by cell counting kit-8 (CCK-8) assay, flow cytometry, β-gal staining, alkaline phosphatase quantitative assay and alizarin red staining in vitro. The potential molecular mechanism was identified by ChIP-Seq, RNA-seq database analysis and immunofluorescence staining. RESULTS The expression of Bmal1 declined with age, while the miR-155-5p was increased. miR-155-5p and Bmal1 repressed each other's expression, and miR-155-5p targeted the Bmal1. Besides, miR-155-5p inhibited the proliferation and osteogenic differentiation of BMSCs, promoted cell apoptosis and senescence, inhibited the expression and nuclear translocation of YAP and TAZ. However, Bmal1 facilitated the osteogenic differentiation and suppressed the aging of BMSCs, meanwhile inactivated the Hippo pathway. Moreover, YAP inhibitors abrogated the positive regulation of aging and osteogenic differentiation in BMSCs by miR-155-5p and Bmal1. CONCLUSION In mouse BMSCs, miR-155-5p and Bmal1 regulated the aging and osteogenic differentiation ability of BMSCs mainly through the Hippo signaling pathway. Our findings provide new insights for the interventions in bone aging.
Collapse
Affiliation(s)
- Lanxin Zhang
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Chengxiaoxue Zhang
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Jiawen Zheng
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Yuhong Wang
- Department of Stomatology, West China Fourth Hospital, Sichuan University, 18, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Xiaoyu Wei
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Yuqing Yang
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China
| | - Qing Zhao
- Department of Orthodontics, State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases, West China School & Hospital of Stomatology, Sichuan University, 14, 3Rd Section of Ren Min Nan Rd, Chengdu, 610041, China.
| |
Collapse
|
11
|
Fawzy MS, Ibrahiem AT, Osman DM, Almars AI, Alshammari MS, Almazyad LT, Almatrafi NDA, Almazyad RT, Toraih EA. Angio-Long Noncoding RNA MALAT1 (rs3200401) and MIAT (rs1061540) Gene Variants in Ovarian Cancer. EPIGENOMES 2024; 8:5. [PMID: 38390896 PMCID: PMC10885055 DOI: 10.3390/epigenomes8010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/04/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
The genotyping of long non-coding RNA (lncRNA)-related single-nucleotide polymorphisms (SNPs) could be associated with cancer risk and/or progression. This study aimed to analyze the angiogenesis-related lncRNAs MALAT1 (rs3200401) and MIAT (rs1061540) variants in patients with ovarian cancer (OC) using "Real-Time allelic discrimination polymerase chain reaction" in 182 formalin-fixed paraffin-embedded (FFPE) samples of benign, borderline, and primary malignant ovarian tissues. Differences in the genotype frequencies between low-grade ovarian epithelial tumors (benign/borderline) and malignant tumors and between high-grade malignant epithelial tumors and malignant epithelial tumors other than high-grade serous carcinomas were compared. Odds ratios (ORs)/95% confidence intervals were calculated as measures of the association strength. Additionally, associations of the genotypes with the available pathological data were analyzed. The heterozygosity of MALAT1 rs3200401 was the most common genotype (47.8%), followed by C/C (36.3%). Comparing the study groups, no significant differences were observed regarding this variant. In contrast, the malignant epithelial tumors had a higher frequency of the MIAT rs1061540 C/C genotype compared to the low-grade epithelial tumor cohorts (56.7% vs. 37.6, p = 0.031). The same genotype was significantly higher in high-grade serous carcinoma than its counterparts (69.4% vs. 43.8%, p = 0.038). Multivariate Cox regression analysis showed that the age at diagnosis was significantly associated with the risk of OC development. In contrast, the MIAT T/T genotype was associated with a low risk of malignant epithelial tumors under the homozygote comparison model (OR = 0.37 (0.16-0.83), p = 0.017). Also, MIAT T allele carriers were less likely to develop high-grade serous carcinoma under heterozygote (CT vs. CC; OR = 0.33 (0.12-0.88), p = 0.027) and homozygote (TT vs. CC; OR = 0.26 (0.07-0.90), p = 0.034) comparison models. In conclusion, our data provide novel evidence for a potential association between the lncRNA MIAT rs1061540 and the malignant condition of ovarian cancer, suggesting the involvement of such lncRNAs in OC development.
Collapse
Affiliation(s)
- Manal S Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 73213, Saudi Arabia
- Unit of Medical Research and Postgraduate Studies, Faculty of Medicine, Northern Border University, Arar 73213, Saudi Arabia
| | - Afaf T Ibrahiem
- Department of Pathology, Faculty of Medicine, Northern Border University, Arar 73213, Saudi Arabia
| | - Dalia Mohammad Osman
- Department of Medical Laboratories Technology, Faculty of Applied Medical Sciences, Northern Border University, Arar 73213, Saudi Arabia
| | - Amany I Almars
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | | | | | | | - Renad Tariq Almazyad
- Faculty of Applied Medical Sciences, Northern Border University, Arar 73213, Saudi Arabia
| | - Eman A Toraih
- Division of Endocrine and Oncologic Surgery, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
12
|
Khachigian LM. The MEK-ERK-Egr-1 axis and its regulation in cardiovascular disease. Vascul Pharmacol 2023; 153:107232. [PMID: 37734428 DOI: 10.1016/j.vph.2023.107232] [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: 08/10/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
Cardiovascular disease (CVD) is the primary cause of morbidity and mortality in the Western world. Multiple molecular and cellular processes underpinning the pathogenesis of CVD are regulated by the zinc finger transcription factor and product of an immediate-early gene, early growth response-1 (Egr-1). Egr-1 regulates multiple pro-inflammatory processes that underpin the manifestation of CVD. The activity of Egr-1 itself is influenced by a range of post-translational modifications including sumoylation, ubiquitination and acetylation. Egr-1 also undergoes phosphorylation by protein kinases, such as extracellular-signal regulated kinase (ERK) which is itself phosphorylated by MEK. This article reviews recent progress on the MEK-ERK-Egr-1 cascade, notably regulation in conjunction with factors and agents such as TET2, TRIB2, MIAT, SphK1, cAMP, teneligliptin, cholinergic drugs, red wine and flavonoids, wogonin, febuxostat, docosahexaenoic acid and AT1R blockade. Such insights should provide new opportunity for therapeutic intervention in CVD.
Collapse
Affiliation(s)
- Levon M Khachigian
- Vascular Biology and Translational Research, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
13
|
Li Z, Duan Y, Yan S, Zhang Y, Wu Y. The miR-302/367 cluster: Aging, inflammation, and cancer. Cell Biochem Funct 2023; 41:752-766. [PMID: 37555645 DOI: 10.1002/cbf.3836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023]
Abstract
MicroRNAs (miRNAs) are a class of noncoding RNAs that occupy a significant role in biological processes as important regulators of intracellular homeostasis. First, we will discuss the biological genesis and functions of the miR-302/367 cluster, including miR-302a, miR-302b, miR-302c, miR-302d, and miR-367, as well as their roles in physiologically healthy tissues. The second section of this study reviews the progress of the miR-302/367 cluster in the treatment of cancer, inflammation, and diseases associated with aging. This cluster's aberrant expression in cells and/or tissues exhibits similar or different effects in various diseases through molecular mechanisms such as proliferation, apoptosis, cycling, drug resistance, and invasion. This article also discusses the upstream and downstream regulatory networks of miR-302/367 clusters and their related mechanisms. Particularly because studies on the upstream regulatory molecules of miR-302/367 clusters, which include age-related macular degeneration, myocardial infarction, and cancer, have become more prevalent in recent years. MiR-302/367 cluster can be an important therapeutic target and the use of miRNAs in combination with other molecular markers may improve diagnostic or therapeutic capabilities, providing unique insights and a more dynamic view of various diseases. It is noted that miRNAs can be an important bio-diagnostic target and offer a promising method for illness diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Zhou Li
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi Province, China
| | - Yan Duan
- Department of Stomatology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi Province, China
| | - Shaofu Yan
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi Province, China
| | - Yao Zhang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi Province, China
| | - Yunxia Wu
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi Province, China
- Department of Stomatology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
14
|
Hosseini SA, Haddadi MH, Fathizadeh H, Nemati F, Aznaveh HM, Taraj F, Aghabozorgizadeh A, Gandomkar G, Bazazzadeh E. Long non-coding RNAs and gastric cancer: An update of potential biomarkers and therapeutic applications. Biomed Pharmacother 2023; 163:114407. [PMID: 37100014 DOI: 10.1016/j.biopha.2023.114407] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 04/28/2023] Open
Abstract
The frequent metastasis of gastric cancer (GC) complicates the cure and therefore the development of effective diagnostic and therapeutic approaches is urgently necessary. In recent years, lncRNA has emerged as a drug target in the treatment of GC, particularly in the areas of cancer immunity, cancer metabolism, and cancer metastasis. This has led to the demonstration of the importance of these RNAs as prognostic, diagnostic and therapeutic agents. In this review, we provide an overview of the biological activities of lncRNAs in GC development and update the latest pathological activities, prognostic and diagnostic strategies, and therapeutic options for GC-related lncRNAs.
Collapse
Affiliation(s)
- Sayedeh Azimeh Hosseini
- Department of Medical Biotechnology, School of Advanced Technology, Shahrekord University of Medical Sciences, Shahrekord, Iran; Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; USERN office, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Hadis Fathizadeh
- Student Research Committee, Sirjan School of Medical Sciences, Sirjan, Iran; Department of Laboratory sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Foroogh Nemati
- Department of Microbiology, Kashan University of Medical Sciences, Kashan, Iran
| | - Hooman Mahmoudi Aznaveh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| | - Farima Taraj
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - AmirArsalan Aghabozorgizadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Golmaryam Gandomkar
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Elaheh Bazazzadeh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| |
Collapse
|
15
|
Yu Y, Dong Y, Deng B, Yang T. IncRNA MIAT Accelerates Keloid Formation by miR-411-5p/JAG1 Axis. Crit Rev Eukaryot Gene Expr 2023; 33:81-92. [PMID: 36734859 DOI: 10.1615/critreveukaryotgeneexpr.2022044734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The long non-coding RNA (lncRNA) myocardial infarction-associated transcript (MIAT) regulates the biological functions of many kinds of cells. The aim of this study is to explore the mechanism of MIAT and how it affects keloid progression. The expressions of MIAT, JAG1, and miR-411-5p in keloid tissues and keloid fibroblasts (KEL FIBs) were quantified by conducting Western blot and quantitative reverse transcription polymerase chain reaction analyses. The influences of MIAT, JAG1, and miR-411-5p on the abilities of KEL FIBs to proliferate, migrate, and invade were assessed by means of the CCK-8, wound healing, and Transwell experiments. To determine the binding relationship among MIAT, JAG1, and miR-411-5p, we performed luciferase reporter and RIP experiments. In keloid tissues and KEL FIBs, MIAT and JAG1 were upregulated while miR-411-5p was downregulated. Knocking-down MIAT or JAG1 significantly inhibited proliferation, migration and invasion. On the contrary, suppressing miR-411-5p expression produced an opposite effect. With regard to mechanisms, MIAT sponged miR-411-5p, which targeted JAG1. MIAT accelerates keloid formation by modulating the miR-411-5p/JAG1 axis.
Collapse
Affiliation(s)
- Yingyan Yu
- Department of Dermatology, University of Electronic Science and Technology of China Hospital, Chengdu 611731, Sichuan, China
| | - Yujie Dong
- Department of Dermatology, Kun Ming Li Du Medical Beauty Hospital, Kunming 650000, Yunnan, China
| | - Benyuan Deng
- Department of General Surgery, West China Health Care Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Ting Yang
- Department of Plastic Surgery and Cosmetic Dermatology, West China School/Hospital of Stomatology Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
16
|
Zhang M, Zhang Z, Hu J, Zhou S, Ai W. Knockdown of long noncoding RNA MIAT attenuates hypoxia-induced cardiomyocyte injury by regulating the miR-488-3p/Wnt/β-catenin pathway. Cell Biol Int 2023; 47:63-74. [PMID: 36273414 DOI: 10.1002/cbin.11945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 08/01/2022] [Accepted: 08/12/2022] [Indexed: 12/31/2022]
Abstract
Dysfunction of cardiomyocytes contributes to the development of acute myocardial infarction (AMI). Nonetheless, the regulatory mechanism of lncRNA myocardial infarction-associated transcript (MIAT) in cardiomyocyte injury remains largely unclear. The cardiomyocyte injury was assessed via cell viability and apoptosis using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) and flow cytometry, respectively. The levels of MIAT, microRNA (miR)-488-3p, and Wnt5a were detected via quantitative real-time polymerase chain reaction and Western blot. After bioinformatical analysis, the binding between miR-488-3p and MIAT or Wnt5a was confirmed via dual-luciferase reporter, RNA immunoprecipitation, and RNA pull-down assays. Our results showed that MIAT expression was increased in AC16 cells after hypoxia treatment. Silencing of MIAT alleviated hypoxia-induced viability reduction, apoptosis increase, and Wnt/β-catenin pathway activation. MIAT directly targeted miR-488-3p. MiR-488-3p might repress hypoxia-induced cardiomyocyte injury, and its knockdown reversed the effect of MIAT depletion on cardiomyocyte injury. Wnt5a was validated as a target of miR-488-3p. Wnt5a expression restoration attenuated the influence of MIAT knockdown on hypoxia-triggered cardiomyocyte injury. Our findings demonstrated that downregulation of MIAT might mitigate hypoxia-induced cardiomyocyte injury partly through miR-488-3p mediated Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Ming Zhang
- General Practice Medicine Department, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, China
| | - Zhiling Zhang
- General Practice Medicine Department, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, China
| | - Jie Hu
- General Practice Medicine Department, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, China
| | - Shulan Zhou
- General Practice Medicine Department, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, China
| | - Wenwei Ai
- General Practice Medicine Department, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
17
|
LncRNA MIAT Upregulates NEGR1 by Competing for miR-150-5p as a Competitive Endogenous RNA in SCIRI Rats. Int J Genomics 2022; 2022:2942633. [PMID: 36620092 PMCID: PMC9812626 DOI: 10.1155/2022/2942633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 12/30/2022] Open
Abstract
Objective Spinal cord ischemia-reperfusion injury (SCIRI) can cause a pathological state of irreversible delayed death of neurons in the spinal cord tissue and a range of complications, such as spinal cord dysfunction and motor function impairment. This study aimed to determine whether the long-stranded non-coding ribonucleic acid (lncRNA), myocardial infarction-associated transcript (MIAT), could upregulate neuronal growth regulator 1 (NEGR1) by competing for miR-150-5p as a competitive endogenous RNA in a rat SCIRI model. Methods The MIAT knockdown vector or the corresponding blank vector was injected into the spinal cord of healthy sprague Dawley (SD) rats. Administration of the MIAT knockdown vector led to the establishment of the SCIRI rat model. Basso, Beattie & Bresnahan locomotor rating scale (BBB) assessment of hind limb motion. Pathological changes in the spinal cord were observed via hematoxylin and eosin staining and eosin staining. Quantitative polymerase chain reaction was performed to determine the expression levels of the candidate microRNAs and predicted candidate genes, and the relationship between them. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL) staining was used to detect apoptosis in the spinal cord tissue of rats in each group. Western blotting was performed to determine the expression of the apoptosis-related proteins, caspase-9, caspase-3, and BCL2-Associated X (Bax)/B-cell lymphoma-2 (Bcl-2). The luciferase reporter gene was used to assess the interaction among the lncRNA, MIAT, and miR-150-5, and the interaction between miR-150-5 and NEGR1. Results The sh-lncRNA, MIAT, improved exercise status, and pathological changes in the spinal cord of SCIRI rats, inhibited apoptosis, increased the expression of miR-150-5p, and reduced the expression of NEGR1. Compared with mimics-NC, the transfection of miR-150-5p significantly decreased the relative fluorescence activity ratio of MIAT 3'-untranslated region (3'-UTR) wild-type Human embryonic kidney cells 293 (HEK-293 cells). Compared with mimics-negative control (NC), the transfection of miR-150-5p significantly decreased the relative fluorescence activity ratio of NEGR1 3'-UTR wild-type HEK-293 cells. Conclusion MIAT can affect the symptoms of SCIRI in rats. Furthermore, as a competitive endogenous RNA, MIAT upregulates NEGR1 by competing with miR-150-5p in SCIRI rats.
Collapse
|
18
|
Silencing lncRNA 93358 Inhibits the Apoptosis of Myocardial Cells in Myocardial Infarction Rats by Inducing the Expression of SLC8A1. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1138709. [PMID: 35845941 PMCID: PMC9283055 DOI: 10.1155/2022/1138709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/18/2022]
Abstract
Objective. To explore the inhibitor effects and mechanism of lncRNA 93358 against the apoptosis of myocardial cells in rats with myocardial infarction. Methods. The myocardial infarction model was established in rats, which were identified by cardiac ultrasound. TTC staining was used to evaluate the degree of heart infarction, and HE staining was utilized to determine the pathological state in myocardial tissues. The apoptotic state in myocardial tissues was confirmed by TUNEL assay. lncRNA 93358 was screened out using a high-throughput sequencing which was confirmed by RT-qPCR. The interaction between miR-466c-3p and SLC8A1 was identified using the dual-luciferase reporter assay. The expression level of Bax, Bcl-2, and SLC8A1 was determined in lncRNA 93358 knockdown cells using RT-qPCR and Western blotting Results. Massive myocardial necrosis was observed in model rats according to the results of TTC staining, HE staining, and TUNEL assay. lncRNA 93358 and Bax were found significantly upregulated, and Bcl-2 and SLC8A1 were greatly downregulated in model rats, which were dramatically reversed by the knockdown of lncRNA 93358, accompanied by the decline area of myocardial necrosis and decreased apoptotic myocardial cells. Conclusion. Silencing lncRNA 93358 inhibits the apoptosis of myocardial cells in rats with myocardial infarction by inducing the expression of SLC8A1.
Collapse
|
19
|
Keshavarz Alikhani H, Pourhamzeh M, Seydi H, Shokoohian B, Hossein-khannazer N, Jamshidi-adegani F, Al-Hashmi S, Hassan M, Vosough M. Regulatory Non-Coding RNAs in Familial Hypercholesterolemia, Theranostic Applications. Front Cell Dev Biol 2022; 10:894800. [PMID: 35813199 PMCID: PMC9260315 DOI: 10.3389/fcell.2022.894800] [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: 03/12/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Familial hypercholesterolemia (FH) is a common monogenic disease which is associated with high serum levels of low-density lipoprotein cholesterol (LDL-C) and leads to atherosclerosis and cardiovascular disease (CVD). Early diagnosis and effective treatment strategy can significantly improve prognosis. Recently, non-coding RNAs (ncRNAs) have emerged as novel biomarkers for the diagnosis and innovative targets for therapeutics. Non-coding RNAs have essential roles in the regulation of LDL-C homeostasis, suggesting that manipulation and regulating ncRNAs could be a promising theranostic approach to ameliorate clinical complications of FH, particularly cardiovascular disease. In this review, we briefly discussed the mechanisms and pathophysiology of FH and novel therapeutic strategies for the treatment of FH. Moreover, the theranostic effects of different non-coding RNAs for the treatment and diagnosis of FH were highlighted. Finally, the advantages and disadvantages of ncRNA-based therapies vs. conventional therapies were discussed.
Collapse
Affiliation(s)
- Hani Keshavarz Alikhani
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Pourhamzeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Homeyra Seydi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bahare Shokoohian
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nikoo Hossein-khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Jamshidi-adegani
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Sulaiman Al-Hashmi
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- *Correspondence: Massoud Vosough,
| |
Collapse
|
20
|
Li J, Yang K, Pan X, Peng H, Hou C, Xiao J, Wang Q. Long Noncoding RNA MIAT Regulates Hyperosmotic Stress-Induced Corneal Epithelial Cell Injury via Inhibiting the Caspase-1-Dependent Pyroptosis and Apoptosis in Dry Eye Disease. J Inflamm Res 2022; 15:3269-3283. [PMID: 35676970 PMCID: PMC9169976 DOI: 10.2147/jir.s361541] [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: 02/14/2022] [Accepted: 05/20/2022] [Indexed: 12/21/2022] Open
Abstract
Purpose The biological role and mechanism of long noncoding RNA (lncRNA) myocardial infarction-associated transcript (MIAT) in dry eye remain to be illustrated. Pyroptosis is a noticeable form of inflammatory activation, which is characteristic of gasdermin D (GSDMD)-driven cell death. The present study was designed to explore the role of MIAT in pyroptosis and apoptosis induced by hyperosmolarity stress (HS) in human corneal epithelial cells (HCECs). Methods HCECs were cultured in 70–120 mM hyperosmotic medium for 24 h to create a dry eye model in vitro. The level of the pyroptosis marker GSDMD was measured, and the cell inflammatory response was evaluated by detecting IL-1β and IL-18 levels. Exogenous caspase-1 inhibitor Ac-YVAD-CHO was used. The pyroptosis in HCECs was examined by caspase-1 activity, immunofluorescent staining, and Western blotting. Flow cytometry was performed to test the apoptosis rate of HCECs. Cell migration and proliferation were detected. The expression of the lncRNA MIAT in HCECs was detected by quantitative real-time PCR. MIAT was knocked down by small interfering RNA (siRNA) transfection. The effects of caspase-1 inhibition on pyroptosis, apoptosis, migration, and proliferation were observed. Results HS promoted pyroptosis in HCECs by elevating caspase-1, GSDMD, and the active cleavage of GSDMD (N-terminal domain, N-GSDMD), and increased the release of IL-1β, IL-18, LDH and the rate of apoptosis, with reduced cell migration. These changes were prevented by the inhibition of caspase-1. The expression of MIAT was significantly increased in HCECs exposed to a hyperosmotic medium. Silencing MIAT increased the expression of GSDMD, caspase-1, and inflammatory chemokines IL-1β and IL-18, and promoted apoptosis while inhibiting migration and proliferation in HCECs. Conclusion The lncRNA MIAT is involved in HS-induced pyroptosis and apoptosis and the inflammatory response of HCECs and provides a new understanding of the pathogenesis of dry eye.
Collapse
Affiliation(s)
- Jinjian Li
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Kun Yang
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Xinghui Pan
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Hui Peng
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Chenting Hou
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Jie Xiao
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
| | - Qing Wang
- Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, 266500, People’s Republic of China
- Correspondence: Qing Wang, Ophthalmology, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266500, People’s Republic of China, Tel +86 17853290636, Fax +86 532 82911747, Email
| |
Collapse
|
21
|
Dissecting the Biological Relevance and Clinical Impact of lncRNA MIAT in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13215518. [PMID: 34771679 PMCID: PMC8583301 DOI: 10.3390/cancers13215518] [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: 09/29/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary The interest in the biological role and clinical impact of long non-coding RNAs (lncRNAs) in multiple myeloma (MM) is continuously expanding. Many studies, mainly involving solid tumors, have strongly suggested the MIAT oncogenic role; more recently, it has been suggested that MIAT may have a role in inducing Bortezomib resistance in MM. However, data concerning MIAT deregulation in MM are virtually absent. In this context, we investigated the expression pattern and the clinical relevance of the lncRNA MIAT in MM, taking advantage of the publicly available CoMMpass database. Our findings prompt further studies to elucidate better the significance of MIAT in MM. Abstract The biological impact of long non-coding RNAs (lncRNAs) in multiple myeloma (MM) is becoming an essential aspect of the investigation, which may contribute to understanding the disease’s complex pathobiology, providing novel potential therapeutic targets. Herein, we investigated the expression pattern and the clinical relevance of the lncRNA MIAT in MM, taking advantage of the publicly available CoMMpass database. MIAT expression in MM is highly heterogeneous and significantly associated with specific molecular lesions frequently occurring in MM. Transcriptome analyses of MM PCs from patients included in the CoMMpass database indicated a potential involvement of MIAT in different signaling pathways and ribosome biogenesis and assembly. These findings suggest that MIAT deregulation may play a pathogenetic role in MM by affecting both proliferation pathways and, indirectly, the translational process. Although MIAT expression levels seem not to be significantly associated with clinical outcome in multivariate analyses, high MIAT expression levels are associated with bortezomib resistance, this suggesting that MIAT targeting could overcome drug resistance in MM. These findings strongly prompt for further studies investigating the significance of MIAT in MM.
Collapse
|