1
|
Eshraghi R, Sadati S, Bahrami A, Mirjalili SR, Farrokhian A, Mahjoubin-Tehran M, Mirzaei H. Unveiling the role of long non-coding RNA MALAT1: a comprehensive review on myocardial infarction. Front Cardiovasc Med 2024; 11:1429858. [PMID: 39171328 PMCID: PMC11335503 DOI: 10.3389/fcvm.2024.1429858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
Myocardial infarction (MI) stands at top global causes of death in developed countries, owing mostly to atherosclerotic plaque growth and endothelial injury-induced reduction in coronary blood flow. While early reperfusion techniques have improved outcomes, long-term treatment continues to be difficult. The function of lncRNAs extends to regulating gene expression in various conditions, both physiological and pathological, such as cardiovascular diseases. The objective of this research is to extensively evaluate the significance of the lncRNA called Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) in the development and management of MI. According to research, MALAT1 is implicated in processes such as autophagy, apoptosis, cell proliferation, and inflammation in the cardiovascular system. This investigation examines recent research examining the effects of MALAT1 on heart function and its potential as a mean of diagnosis and treatment for post- MI complications and ischemic reperfusion injury.
Collapse
Affiliation(s)
- Reza Eshraghi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sina Sadati
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ashkan Bahrami
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Reza Mirjalili
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alireza Farrokhian
- Department of Cardiology, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Mahjoubin-Tehran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
2
|
Bahreini F, Jabbari P, Gossing W, Aziziyan F, Frohme M, Rezaei N. The role of noncoding RNAs in pituitary adenoma. Epigenomics 2021; 13:1421-1437. [PMID: 34558980 DOI: 10.2217/epi-2021-0165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Pituitary adenomas (PAs) are common cranial tumors that affect the quality of life in patients. Early detection of PA is beneficial for avoiding clinical complications of this disease and increasing the quality of life. Noncoding RNAs, including long noncoding RNA, miRNA and circRNA, regulate protein expression, mostly by inhibiting the translation process. Studies have shown that dysregulation of noncoding RNAs is associated with PA. Hence understanding the expression pattern of noncoding RNAs can be considered a promising method for developing biomarkers. This article reviews data on the expression pattern of dysregulated noncoding RNAs involved in PA. Possible molecular mechanisms by which the dysregulated noncoding RNA could possibly induce PA are also described.
Collapse
Affiliation(s)
- Farbod Bahreini
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Parnian Jabbari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Genetics, Genomics & Bioinformatics, University of California, Riverside, CA, USA
| | - Wilhelm Gossing
- Division Molecular Biotechnology & Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, 15745, Wildau, Germany
| | - Fatemeh Aziziyan
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Marcus Frohme
- Division Molecular Biotechnology & Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, 15745, Wildau, Germany
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Ma Y, Wu H, Liang X, Zhang C, Ma Y, Wei Y, Li J, Chen H. Identification of downstream targets and signaling pathways of long non-coding RNA NR_002794 in human trophoblast cells. Bioengineered 2021; 12:6617-6628. [PMID: 34516352 PMCID: PMC8806843 DOI: 10.1080/21655979.2021.1974808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Preeclampsia (PE) is a huge threat to pregnant women. Our previous study demonstrated that long non-coding RNA (lncRNA) NR_002794 was highly expressed in placentas of PE patients and could regulate the phenotypes of trophoblast cells. However, the downstream regulatory mechanisms of NR_002794 remain unknown. In this text, some potential downstream targets or signaling pathways of NR_002794 were identified through RNA sequencing (RNA-seq) and bioinformatics analysis in SWAN71 trophoblast cells. Western blot assay demonstrated that NR_002794 inactivated protein kinase B (AKT) and extracellular signal-regulated kinase 1/2 (ERK1/2) pathways and activated cell apoptotic signaling in SWAN71 cells. Both RNA-seq and reverse transcription-quantitative PCR (RT-qPCR) outcomes showed that NR_002794 up-regulation could notably inhibit the expression of C-C motif chemokine ligand 4 like 2 (CCL4L2), interleukin 15 receptor subunit alpha (IL15RA), interleukin 32 (IL32), and tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE1), while NR_002794 knockdown induced these gene expressions in SWAN71 cells. CCK-8, BrdU, Transwell, wound healing, and flow cytometry analyses showed that NR_002794 inhibited cell proliferation and migration and induced cell apoptosis through down-regulating TIE1 in SWAN71 cells. In conclusion, lncRNA NR_002794 could exert its functions by regulating AKT and ERK1/2 pathways and TIE1 expression in human trophoblast cells.
Collapse
Affiliation(s)
- Yinyao Ma
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Hua Wu
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Xuxia Liang
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Chun Zhang
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Yanhua Ma
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Yanfen Wei
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Jing Li
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Hui Chen
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| |
Collapse
|
4
|
Sun L, Cui Y, Jiang K, Li J. Down-regulation of long non-coding RNA antisense non-coding RNA in the INK4 locus suppresses OVCAR-3 cells proliferation and induction of apoptosis by Wnt/β -catenin. J Pharm Pharmacol 2021; 73:1212-1217. [PMID: 33772549 DOI: 10.1093/jpp/rgab042] [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: 08/28/2020] [Accepted: 02/20/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Ovarian cancer is a lethal gynecological malignancy. Long non-coding RNA antisense non-coding RNA in the INK4 locus (lncRNA ANRIL) was reported to have a critical role in cancer advancement. The ANRIL-mediated oncogenic underlying molecular mechanisms are not fully understood in ovarian cancer. We aimed to study ANRIL silencing effects on the proliferation and apoptosis of OVCAR-3 cells. METHODS The ANRIL was Knockdown by transfection of OVCAR-3 cells with si-RNA against ANRIL. MTT assay and cell death ELISA kit were used to evaluate cellular proliferation and apoptosis. The expression levels of ANRIL, pro-and anti-apoptotic genes were assessed using q-RT-PCR. Western blotting was used to assess Wnt/β-catenin signalling pathway. KEY FINDINGS ANRIL down-regulating in OVCAR-3 cell lines resulted in significant inhibition of cellular proliferation, apoptosis induction, as well as suppression of cellular invasion. Besides, knockdown of ANRIL led to pro-apoptotic genes up-regulation, Bad and Bax and anti-apoptotic genes down-regulation, Bid and Bcl-2. More importantly, we observed that ANRIL inhibition suppressed the vital components expression of the Wnt/β-catenin cascade. CONCLUSION Our findings showed that down-regulation of lncRNA ANRIL resulted in the effective suppression of OVCAR-3 cell proliferation and invasion and induction of apoptosis by preventing Wnt/β-catenin signal transduction.
Collapse
Affiliation(s)
- Lingna Sun
- Department of Gynecology, Maternal and Child Health Care Hospital of Shandong Province, Jinan City, Shandong Province, China
| | - Yuping Cui
- Department of Obstetrics and Gynecology, Huantai County People's Hospital, Zibo City, Shandong Province, China
| | - Kongdi Jiang
- Department of Obstetrics and Gynecology, Huantai County People's Hospital, Zibo City, Shandong Province, China
| | - Juan Li
- Department of Gynecology, Maternal and Child Health Care Hospital of Shandong Province, Jinan City, Shandong Province, China
| |
Collapse
|
5
|
Feng T, Zhang P, Sun Y, Wang Y, Tong J, Dai H, Hua Z. High throughput sequencing identifies breast cancer-secreted exosomal LncRNAs initiating pulmonary pre-metastatic niche formation. Gene 2019; 710:258-264. [PMID: 31176731 DOI: 10.1016/j.gene.2019.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Increasing evidence indicated that cancer-secreted exosomes played an important role in tumor metastasis. However, the function of exosomes in breast cancer pulmonary metastasis remains unknown. The aim of the study was to investigate the role of exosome-derived from breast cancer-secreted long non-coding RNAs (LncRNAs) on pre-metastatic niche formation in pulmonary metastasis. METHODS Exosomes-derived from breast cancer were separated by ultracentrifugation. The high-throughput sequencing, Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway were used to detect and evaluate the differential expression of LncRNAs in lung fibroblasts with exosomes treated. And quantitative real-time polymerase chain reaction (qRT-PCR) was performed to verify candidate LncRNAs expression. RESULTS We found that exosomes-derived from breast cancer induced lung fibroblasts proliferation and migration. In addition, a large number of LncRNAs expression abnormalities were involved in the breast cancer lung metastasis microenvironment. CONCLUSION Our findings suggested that exosomal LncRNAs facilitated tumor pre-metastatic niche formation and represented a novel mechanistic insight into the molecular mechanism of cancer metastasis microenvironment.
Collapse
Affiliation(s)
- Tongbao Feng
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China; Department of Clinical Laboratory, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, PR China; Department of General Surgery, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, PR China
| | - Ping Zhang
- Department of Clinical Laboratory, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, PR China
| | - Yingxin Sun
- Department of Clinical Laboratory, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, PR China
| | - Yan Wang
- Department of Clinical Laboratory, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, PR China
| | - Jichun Tong
- Department of Cardiac Surgery, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, PR China
| | - Hong Dai
- Department of General Surgery, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, PR China.
| | - Zichun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China; Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, PR China.
| |
Collapse
|