1
|
Zhang Y, Zhan L, Jiang X, Tang X. Comprehensive review for non-coding RNAs: From mechanisms to therapeutic applications. Biochem Pharmacol 2024; 224:116218. [PMID: 38643906 DOI: 10.1016/j.bcp.2024.116218] [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: 02/01/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024]
Abstract
Non-coding RNAs (ncRNAs) are an assorted collection of transcripts that are not translated into proteins. Since their discovery, ncRNAs have gained prominence as crucial regulators of various biological functions across diverse cell types and tissues, and their abnormal functioning has been implicated in disease. Notably, extensive research has focused on the relationship between microRNAs (miRNAs) and human cancers, although other types of ncRNAs, such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), are also emerging as significant contributors to human disease. In this review, we provide a comprehensive summary of our current knowledge regarding the roles of miRNAs, lncRNAs, and circRNAs in cancer and other major human diseases, particularly cancer, cardiovascular, neurological, and infectious diseases. Moreover, we discuss the potential utilization of ncRNAs as disease biomarkers and as targets for therapeutic interventions.
Collapse
Affiliation(s)
- YanJun Zhang
- College of Pharmacy and Traditional Chinese Medicine, Jiangsu College of Nursing, Huaian, Jiangsu, 223005, China
| | - Lijuan Zhan
- College of Pharmacy and Traditional Chinese Medicine, Jiangsu College of Nursing, Huaian, Jiangsu, 223005, China
| | - Xue Jiang
- College of Pharmacy and Traditional Chinese Medicine, Jiangsu College of Nursing, Huaian, Jiangsu, 223005, China.
| | - Xiaozhu Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
2
|
Seo JH, Ryu S, Cheon SY, Lee SJ, Won SJ, Yim CD, Lee HJ, Hah YS, Park JJ. Sirt6-Mediated Cell Death Associated with Sirt1 Suppression in Gastric Cancer. Cancers (Basel) 2024; 16:387. [PMID: 38254877 PMCID: PMC10814469 DOI: 10.3390/cancers16020387] [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: 12/18/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Gastric cancer, one of the leading causes of cancer-related death, is strongly associated with H. pylori infection, although other risk factors have been identified. The sirtuin (Sirt) family is involved in the tumorigenesis of gastric cancer, and sirtuins can have pro- or anti-tumorigenic effects. METHODS After determining the overall survival rate of gastric cancer patients with or without Sirt6 expression, the effect of Sirt6 upregulation was also tested using a xenograft mouse model. The regulation of Sirt6 and Sirt1, leading to the induction of mouse double minute 2 homolog (MDM2) and reactive oxygen species (ROS), was mainly analyzed using Western blotting and immunofluorescence staining, and gastric cancer cell (SNU-638) death associated with these proteins was measured using flow cytometric analysis. RESULTS Sirt6 overexpression led to Sirt1 suppression in gastric cancer cells, resulting in a higher level of gastric cancer cell death in vitro and a reduced tumor volume. ROS and MDM2 expression levels were upregulated by Sirt6 overexpression and/or Sirt1 suppression according to Western blot analysis. The upregulated ROS ultimately led to gastric cancer cell death as determined via Western blot and flow cytometric analysis. CONCLUSION We found that the upregulation of Sirt6 suppressed Sirt1, and Sirt6- and Sirt1-induced gastric cancer cell death was mediated by ROS production. These findings highlight the potential of Sirt6 and Sirt1 as therapeutic targets for treating gastric cancer.
Collapse
Affiliation(s)
- Ji Hyun Seo
- Department of Pediatrics, Institute of Health Science, College of Medicine, Gyeongsang National University, Jinju 52725, Republic of Korea;
- Institute of Medical Science, Gyeongsang National University, Jinju 52725, Republic of Korea; (S.R.); (C.D.Y.)
| | - Somi Ryu
- Institute of Medical Science, Gyeongsang National University, Jinju 52725, Republic of Korea; (S.R.); (C.D.Y.)
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| | - So Young Cheon
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea;
| | - Seong-Jun Lee
- Department of Convergence of Medical Sciences, Gyeongsang National University, Jinju 52725, Republic of Korea
| | - Seong-Jun Won
- Institute of Medical Science, Gyeongsang National University, Jinju 52725, Republic of Korea; (S.R.); (C.D.Y.)
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| | - Chae Dong Yim
- Institute of Medical Science, Gyeongsang National University, Jinju 52725, Republic of Korea; (S.R.); (C.D.Y.)
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| | - Hyun-Jin Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Chung-Ang University, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong 06973, Republic of Korea
| | - Young-Sool Hah
- Institute of Medical Science, Gyeongsang National University, Jinju 52725, Republic of Korea; (S.R.); (C.D.Y.)
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea;
| | - Jung Je Park
- Institute of Medical Science, Gyeongsang National University, Jinju 52725, Republic of Korea; (S.R.); (C.D.Y.)
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Gyeongsang National University, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea;
| |
Collapse
|
3
|
Jin Y, Jiang D. GATA6-AS1 via Sponging miR-543 to Regulate PTEN/AKT Signaling Axis Suppresses Cell Proliferation and Migration in Gastric Cancer. Mediators Inflamm 2023; 2023:9340499. [PMID: 37273453 PMCID: PMC10238141 DOI: 10.1155/2023/9340499] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/16/2022] [Accepted: 04/05/2023] [Indexed: 06/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most common and lethal cancers worldwide. In view of the prominent roles of long noncoding RNAs (lncRNAs) in cancers, we investigated the specific role and underlying mechanism of GATA binding protein 6 antisense RNA 1 (GATA6-AS1) in GC. Quantitative real-time polymerase chain reaction (qRT-PCR) detected GATA6-AS1 expression in GC cell lines. Functional assays were conducted to explore the role of GATA6-AS1 in GC. Furthermore, mechanism investigations were implemented to uncover the interaction among GATA6-AS1, microRNA-543 (miR-543), and phosphatase and tensin homolog (PTEN). In the present study, it was found that GATA6-AS1 expression is significantly downregulated in GC cell lines. Functionally, GATA6-AS1 markedly suppresses GC cell growth and migration in vitro and in vivo tumorigenesis. Besides tumor suppressor, GATA6-AS1 serves as a miR-543 sponge. Specifically speaking, GATA6-AS1 acts as a competing endogenous RNA (ceRNA) of miR-543 to upregulate the expression of PTEN, thus inactivating AKT signaling pathway to inhibit GC progression. In conclusion, this study has manifested that GATA6-AS1 inhibits GC cell proliferation and migration as a sponge of miR-543 by regulating PTEN/AKT signaling axis, offering new perspective into developing novel GC therapies.
Collapse
Affiliation(s)
- Yi Jin
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110001 Liaoning, China
| | - Daqing Jiang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110001 Liaoning, China
| |
Collapse
|
4
|
Scheper M, Iyer A, Anink JJ, Mesarosova L, Mills JD, Aronica E. Dysregulation of miR-543 in Parkinson's disease: Impact on the neuroprotective gene SIRT1. Neuropathol Appl Neurobiol 2023; 49:e12864. [PMID: 36352829 PMCID: PMC10100056 DOI: 10.1111/nan.12864] [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: 05/27/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
AIMS Parkinson's disease (PD) is a progressive and age-dependent neurodegenerative disease characterised clinically by a variety of motor symptoms and cognitive impairment. PD was initially considered to be a grey matter disease; however, recently, evidence has emerged that white matter changes in PD precede the neuronal loss seen in the grey matter. The cause of these initial white matter changes is yet to be elucidated. Here, we explored whether dysregulated miRNAs and their target mRNA could provide insight into the underlying mechanisms of early white matter changes in PD. METHODS We analysed the expression of miRNAs in three different stages of PD through RNA-sequencing and validated the differential expression of miRNAs through quantitative reverse transcription polymerase chain reaction. With bioinformatic analyses, we predicted target genes of dysregulated miRNAs and investigated their biomarker potential. Finally, in vitro, we confirmed the targetting of the gene SIRT1 by miR-543. RESULTS We identified 12 dysregulated miRNAs in PD and found that miR-543 holds potential as a biomarker for late-stage PD with dementia. We report upregulation of miR-543 in early PD white matter tissue and downregulation of SIRT1. In vitro experiments showed that the upregulation of miR-543 results in the downregulation of SIRT1 in the white matter, but not in the grey matter. CONCLUSIONS We validated SIRT1 as a target of miR-543 in the brain and showed its function as a potential biomarker. Our results highlight the idea that dysregulation of miR-543 in early PD white matter, resulting in the dysregulation of SIRT1, potentially influencing the early white matter changes observed in PD.
Collapse
Affiliation(s)
- Mirte Scheper
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Anand Iyer
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands.,Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Jasper J Anink
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Lucia Mesarosova
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - James D Mills
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Loe AKH, Zhu L, Kim TH. Chromatin and noncoding RNA-mediated mechanisms of gastric tumorigenesis. Exp Mol Med 2023; 55:22-31. [PMID: 36653445 PMCID: PMC9898530 DOI: 10.1038/s12276-023-00926-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/08/2022] [Accepted: 11/22/2022] [Indexed: 01/20/2023] Open
Abstract
Gastric cancer (GC) is one of the most common and deadly cancers in the world. It is a multifactorial disease highly influenced by environmental factors, which include radiation, smoking, diet, and infectious pathogens. Accumulating evidence suggests that epigenetic regulators are frequently altered in GC, playing critical roles in gastric tumorigenesis. Epigenetic regulation involves DNA methylation, histone modification, and noncoding RNAs. While it is known that environmental factors cause widespread alterations in DNA methylation, promoting carcinogenesis, the chromatin- and noncoding RNA-mediated mechanisms of gastric tumorigenesis are still poorly understood. In this review, we focus on discussing recent discoveries addressing the roles of histone modifiers and noncoding RNAs and the mechanisms of their interactions in gastric tumorigenesis. A better understanding of epigenetic regulation would likely facilitate the development of novel therapeutic approaches targeting specific epigenetic regulators in GC.
Collapse
Affiliation(s)
- Adrian Kwan Ho Loe
- grid.42327.300000 0004 0473 9646Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4 Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Lexin Zhu
- grid.42327.300000 0004 0473 9646Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4 Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Tae-Hee Kim
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
6
|
Badie A, Gaiddon C, Mellitzer G. Histone Deacetylase Functions in Gastric Cancer: Therapeutic Target? Cancers (Basel) 2022; 14:5472. [PMID: 36358890 PMCID: PMC9659209 DOI: 10.3390/cancers14215472] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 09/05/2023] Open
Abstract
Gastric cancer (GC) is one of the most aggressive cancers. Therapeutic treatments are based on surgery combined with chemotherapy using a combination of platinum-based agents. However, at metastatic stages of the disease, survival is extremely low due to late diagnosis and resistance mechanisms to chemotherapies. The development of new classifications has not yet identified new prognostic markers for clinical use. The studies of epigenetic processes highlighted the implication of histone acetylation status, regulated by histone acetyltransferases (HATs) and by histone deacetylases (HDACs), in cancer development. In this way, inhibitors of HDACs (HDACis) have been developed and some of them have already been clinically approved to treat T-cell lymphoma and multiple myeloma. In this review, we summarize the regulations and functions of eighteen HDACs in GC, describing their known targets, involved cellular processes, associated clinicopathological features, and impact on survival of patients. Additionally, we resume the in vitro, pre-clinical, and clinical trials of four HDACis approved by Food and Drug Administration (FDA) in cancers in the context of GC.
Collapse
Affiliation(s)
| | | | - Georg Mellitzer
- Laboratoire Streinth, Université de Strasbourg, Inserm UMR_S 1113 IRFAC, 67200 Strasbourg, France
| |
Collapse
|
7
|
Rincón-Riveros A, Rodríguez JA, Villegas VE, López-Kleine L. Identification of Two Exosomal miRNAs in Circulating Blood of Cancer Patients by Using Integrative Transcriptome and Network Analysis. Noncoding RNA 2022; 8:33. [PMID: 35645340 PMCID: PMC9149928 DOI: 10.3390/ncrna8030033] [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: 02/21/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 11/16/2022] Open
Abstract
Exosomes carry molecules of great biological and clinical interest, such as miRNAs. The contents of exosomes vary between healthy controls and cancer patients. Therefore, miRNAs and other molecules transported in exosomes are considered a potential source of diagnostic and prognostic biomarkers in cancer. Many miRNAs have been detected in recent years. Consequently, a substantial amount of miRNA-related data comparing patients and healthy individuals is available, which contributes to a better understanding of the initiation, development, malignancy, and metastasis of cancer using non-invasive sampling procedures. However, a re-analysis of available ncRNA data is rare. This study used available data about miRNAs in exosomes comparing healthy individuals and cancer patients to identify possible global changes related to the presence of cancer. A robust transcriptomic analysis identified two common miRNAs (miR-495-3p and miR-543) deregulated in five cancer datasets. They had already been implicated in different cancers but not reported in exosomes circulating in blood. The study also examined their target genes and the implications of these genes for functional processes.
Collapse
Affiliation(s)
- Andrés Rincón-Riveros
- Bioinformatics and Systems Biology Group, Universidad Nacional de Colombia, Bogotá 111221, Colombia
| | | | - Victoria E Villegas
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia
| | - Liliana López-Kleine
- Department of Statistics, Faculty of Science, Universidad Nacional de Colombia, Bogotá 111221, Colombia
| |
Collapse
|
8
|
GNL3 Regulates SIRT1 Transcription and Promotes Hepatocellular Carcinoma Stem Cell-Like Features and Metastasis. JOURNAL OF ONCOLOGY 2022; 2022:1555670. [PMID: 35432540 PMCID: PMC9010172 DOI: 10.1155/2022/1555670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/05/2022] [Indexed: 12/02/2022]
Abstract
The expression of GNL3 in hepatocellular carcinoma was detected, and its effect on the proliferation and metastasis of hepatocellular carcinoma cells was investigated. Hepatocellular carcinoma and adjacent tissues were collected. The mRNA and protein expression levels of GNL3 were detected by qRT-PCR, Western blot, and immunohistochemistry. The relationship between GNL3 and the prognosis of liver cancer was analysed using public databases. A GNL3 interfering plasmid was constructed, and the effects of GNL3 on the proliferation of HepG2 and PLC-PRF-5 hepatoma cells were detected by the CCK-8 method. Transwell chamber assays were used to detect the effects of GNL3 on the migration and invasion of hepatocellular carcinoma cells. The effects of GNL3 on SIRT1 expression and stem cell markers were analysed. The effect of GNL3 on the proliferation of hepatocellular carcinoma was detected in a subcutaneous tumor-bearing animal model. The results showed that the mRNA and protein levels of GNL3 were higher than those of adjacent tissues. The overall survival (OS) of HCC patients with high GNL3 expression was worse. In vivo and in vitro experiments confirmed that silencing GNL3 could inhibit the proliferation, migration, and invasion of hepatocellular carcinoma cells. Mechanistic studies have shown that GNL3 regulates SIRT1 expression. GNL3 mediates the stem cell-like properties of HCC cells through SIRT1. In conclusion, this study found that GNL3 increased expression in hepatocellular carcinoma, which promoted the malignant biological behavior of hepatocellular carcinoma cells and was related to the cell dry phenotype. This study has certain significance in evaluating the prognosis of HCC patients.
Collapse
|
9
|
Chen S, Guo W, Wei J, Lin H, Guo F. Knockdown of has_circ_0010452 Promotes Proliferation and Osteogenic Differentiation via Up-Regulating miR-543 in Human Bone Marrow Mesenchymal Stem Cells. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Objective: The aim of this study was to explore the role of has_circ_0010452 in the progression of osteoporosis (OP) targeting miR-543, as well as their functions in regulating proliferation and osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs).
Methods: The expression levels of circ_0010452 and miR-543 in hBMSCs at different time points of osteogenic differentiation were determined by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). After transfection of circ_0010452 siRNA or miR-543 inhibitor in hBMSCs, the relative
expression levels of osteogenic marker proteins, including oat spelt xylan (OSX), osteocalcin (OCN) and collagen I (Col-1), were determined by western blot. Cell proliferation of hBMSCs was valued by Cell Counting Kit 8 (CCK-8) assay. Dual-Luciferase reporter gene assay was performed to verify
the relationship between circ_0010452 and miR-543. Subsequently, the regulatory effects of circ_0010452 and miR-543 on osteogenic differentiation and the capability of mineralization were evaluated by alkaline phosphatase (ALP) determination and alizarin red staining, respectively. Results:
The expression of circ_0010452 decreased gradually and miR-543 increased in hBMSCs with the prolongation of osteogenic differentiation. circ_0010452 could bind to miR-543, which was negatively regulated by miR-543 in hBMSCs. Moreover, knockdown of circ_0010452 inhibited proliferation and osteogenic
differentiation by upregulating miR-543, as well as upregulating expressions of OSX, OCN and Col-1. Furthermore, knockdown of circ_0010452 markedly promoted the capability of mineralization of hBMSCs, which was further reversed by transfection of miR-543 inhibitor. The knockdown of miR-543
partially reversed the inhibitory effect of circ_0010452 on the osteogenesis of hBMSCs. Conclusions: Silence of circ_0010452 promotes the development of OP via binding to miR-543 regulating proliferation and osteogenic differentiation of hBMSCs, thus promoting the progression of osteoporosis.
Collapse
Affiliation(s)
- Siyuan Chen
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, China
| | - Weixiong Guo
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, China
| | - Jinsong Wei
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, China
| | - Han Lin
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, China
| | - Fengyan Guo
- Department of Geriatrics, Qingpu Branch, Zhongshan Hospital of Fudan University, Shanghai, 201700, China
| |
Collapse
|
10
|
Yang Q, Chen Y, Guo R, Dai Y, Tang L, Zhao Y, Wu X, Li M, Du F, Shen J, Yi T, Xiao Z, Wen Q. Interaction of ncRNA and Epigenetic Modifications in Gastric Cancer: Focus on Histone Modification. Front Oncol 2022; 11:822745. [PMID: 35155211 PMCID: PMC8826423 DOI: 10.3389/fonc.2021.822745] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 12/28/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer has developed as a very common gastrointestinal tumors, with recent effective advancements in the diagnosis and treatment of early gastric cancer. However, the prognosis for gastric cancer remains poor. As a result, there is in sore need of better understanding the mechanisms of gastric cancer development and progression to improve existing diagnostic and treatment options. In recent years, epigenetics has been recognized as an important contributor on tumor progression. Epigenetic changes in cancer include chromatin remodeling, DNA methylation and histone modifications. An increasing number of studies demonstrated that noncoding RNAs (ncRNAs) are associated with epigenetic changes in gastric cancer. Herein, we describe the molecular interactions of histone modifications and ncRNAs in epigenetics. We focus on ncRNA-mediated histone modifications of gene expression associated with tumorigenesis and progression in gastric cancer. This molecular mechanism will contribute to our deeper understanding of gastric carcinogenesis and progression, thus providing innovations in gastric cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Qingfan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yu Chen
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Rui Guo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
| | - Yalan Dai
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Liyao Tang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Yueshui Zhao
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Xu Wu
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Mingxing Li
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Fukuan Du
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Jing Shen
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Tao Yi
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Zhangang Xiao
- South Sichuan Institute of Translational Medicine, Luzhou, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| |
Collapse
|
11
|
The Role of microRNAs in the Mammary Gland Development, Health, and Function of Cattle, Goats, and Sheep. Noncoding RNA 2021; 7:ncrna7040078. [PMID: 34940759 PMCID: PMC8708473 DOI: 10.3390/ncrna7040078] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023] Open
Abstract
Milk is an integral and therefore complex structural element of mammalian nutrition. Therefore, it is simple to conclude that lactation, the process of producing milk, is as complex as the mammary gland, the organ responsible for this biochemical activity. Nutrition, genetics, epigenetics, disease pathogens, climatic conditions, and other environmental variables all impact breast productivity. In the last decade, the number of studies devoted to epigenetics has increased dramatically. Reports are increasingly describing the direct participation of microRNAs (miRNAs), small noncoding RNAs that regulate gene expression post-transcriptionally, in the regulation of mammary gland development and function. This paper presents a summary of the current state of knowledge about the roles of miRNAs in mammary gland development, health, and functions, particularly during lactation. The significance of miRNAs in signaling pathways, cellular proliferation, and the lipid metabolism in agricultural ruminants, which are crucial in light of their role in the nutrition of humans as consumers of dairy products, is discussed.
Collapse
|
12
|
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
|
13
|
An J, Guo X, Yan B. DICER-AS1 functions as competing endogenous RNA that targets CSR1 by sponging microRNA-650 and suppresses gastric cancer progression. J Int Med Res 2021; 49:3000605211041466. [PMID: 34586953 PMCID: PMC8485291 DOI: 10.1177/03000605211041466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/20/2021] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE This study explored the functional interactions between the long non-coding RNA DICER-AS1 and the cellular stress response 1 (CSR1) gene in gastric cancer. METHODS Quantitative polymerase chain reaction (qPCR) and western blotting were used to measure DICER-AS1, CSR1, and miR-650 expression levels. Gastric cancer cell line proliferation and migration abilities were analyzed using the MTT and transwell migration and invasion assays, respectively. Bioinformatic analysis and dual luciferase reporter assays were employed to study the functional interactions among miR-650, DICER-AS1, and CSR1. RESULTS DICER-AS1 and CSR1 expression levels were significantly decreased in gastric cancer tissues compared with normal tissues, and qPCR analysis showed that miR-650 was upregulated in gastric cancer tissues. Bioinformatic analysis and dual luciferase reporter assays revealed that DICER-AS1 functioned as a competing endogenous RNA that sponged miR-650, which in turn regulated CSR1 expression. Importantly, ectopic DICER-AS1 and CSR1 expression inhibited cell proliferation and migration in vitro and suppressed xenograft tumorgenicity in vivo. CONCLUSIONS These results suggest that DICER-AS1 functions as a competing endogenous RNA that regulates miR-650 to suppress proliferation and migration of gastric cancer cells by targeting CSR1. These findings indicate that targeting DICER-AS1 and miR-650 could be a novel treatment for gastric cancer.
Collapse
Affiliation(s)
- Junyan An
- Department of Gastroenterology, Weifang People’s Hospital, China
| | - Xiaoling Guo
- Infectious Diseases Department, Weifang People’s Hospital, China
| | - Bingli Yan
- Department of Gastroenterology, Weifang People’s Hospital, China
| |
Collapse
|
14
|
Zhang J, Zha W, Qian C, Ding A, Mao Z. Circular RNA circ_0001017 Sensitizes Cisplatin-Resistant Gastric Cancer Cells to Chemotherapy by the miR-543/PHLPP2 Axis. Biochem Genet 2021; 60:558-575. [PMID: 34313883 DOI: 10.1007/s10528-021-10110-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022]
Abstract
Resistance to cisplatin (CDDP) remains a major challenge for the treatment of gastric cancer (GC). Circular RNAs (circRNAs) have been implicated in the development of CDDP resistance of GC. However, the precise actions of circ_0001017 in CDDP resistance of GC remain to be elucidated. The levels of circ_0001017, microRNA (miR)-543 and PH-domain and leucine-rich repeat protein phosphatase 2 (PHLPP2) mRNA were gauged by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot was used to analyze the protein levels of Vimentin, N-cadherin, E-cadherin, and PHLPP2. Ribonuclease R (RNase R) assay was applied to evaluate the stability of circ_0001017. Cell viability and proliferation, colony formation ability, cell cycle distribution and apoptosis, and migration and invasion were detected by the Cell Counting Kit-8 (CCK-8), colony formation, flow cytometry, and transwell assays, respectively. Direct relationship between miR-543 and circ_0001017 or PHLPP2 was verified by dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. Xenograft model assay was used to assess the function of circ_0001017 in vivo. Low expression of circ_0001017 was associated with CDDP resistance of GC. Enforced expression of circ_0001017 impeded growth, metastasis, and enhanced apoptosis of HGC-27/R and AGS/R cells and sensitized them to CDDP in vitro. Circ_0001017 targeted miR-543, and circ_0001017 regulated CDDP-resistant cell behaviors and CDDP sensitivity by suppressing miR-543. PHLPP2 was a direct target of miR-543, and circ_0001017 controlled PHLPP2 expression through miR-543. Moreover, miR-543 knockdown-mediated promotion of PHLPP2 impacted CDDP-resistant cell behaviors and CDDP sensitivity in vitro. Additionally, elevated expression of circ_0001017 hindered growth of HGC-27/R cells and sensitized them to CDDP in vivo. Our findings demonstrated that enforced expression of circ_0001017 suppressed malignant behaviors and enhanced CDDP sensitivity of CDDP-resistant GC cells at least partially by the miR-543/PHLPP2 axis.
Collapse
Affiliation(s)
- Jianmin Zhang
- Department of General Surgery, First Affiliated Hospital of Soochow University, No.899 Pinghai Road, Suzhou City, 215000, Jiangsu Province, China.,Departments of General Surgery, Yancheng City No.1 People's Hospital, Yancheng City, Jiangsu Province, China
| | - Wenzhang Zha
- Departments of General Surgery, Yancheng City No.1 People's Hospital, Yancheng City, Jiangsu Province, China
| | - Changchun Qian
- Departments of General Surgery, Yancheng City No.1 People's Hospital, Yancheng City, Jiangsu Province, China
| | - Aixing Ding
- Departments of General Surgery, Yancheng City No.1 People's Hospital, Yancheng City, Jiangsu Province, China
| | - Zhongqi Mao
- Department of General Surgery, First Affiliated Hospital of Soochow University, No.899 Pinghai Road, Suzhou City, 215000, Jiangsu Province, China.
| |
Collapse
|
15
|
Wang YL, Liang RH, Wang CY, Zhang RP, Wu SY, Han X, Zhang GL. MicroRNA-543 inhibits the proliferation, migration, invasion, and epithelial-mesenchymal transition of triple-negative breast cancer cells via down-regulation of ACTL6A gene. Clin Transl Oncol 2021; 24:84-92. [PMID: 34181232 DOI: 10.1007/s12094-021-02672-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/17/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE To investigate the effect of microRNA-543 (miR-543) on the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of triple-negative breast cancer (TNBC) cells, and the associated mechanism. METHODS Human breast cancer cells (MDA-MB-231, HCC1937, and MCF-7, ZR-75-1) and normal human breast epithelial cell line (MCF10A) were transfected with miR-543 mimics or inhibitor using lipofectamine 2000. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting were used to determine the mRNA and protein expression levels of miR-543, actin-like protein 6A (ACTL6A), vimentin, Snail, and E-cadherin in breast cancer cells/tissue. Cell counting kit-8 (CCK-8), wound-healing, and Transwell assays were used to measure the effect of miR-543 on TNBC cell proliferation, invasion, and migration. Overall survival was determined using data from Gene Expression Omnibus (GEO) and Cancer Genome Atlas (TCGA) databases. Bioinformatics analysis and luciferase reporter gene assay were used to determine the regulatory effect of miR-543 on ACTL6A. RESULTS The level of expression of miR-543 was significantly lower in breast cancer cells/tissue than in normal human breast epithelial cell/tissue (p < 0.05). MicroRNA-543 expression level was significantly reduced in TNBC cells/tissue, relative to the other breast cancer cells/normal breast tissue (p < 0.05). MicroRNA-543 significantly suppressed tumor growth and the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of TNBC cells, in mouse xenograft model (p < 0.05). CONCLUSIONS miR-543 influences the biological behavior of TNBC cells by directly targeting ACTL6A gene. miR-543 could serve as a novel diagnostic and therapeutic target for TNBC.
Collapse
Affiliation(s)
- Y L Wang
- Department of Medical Oncology, Baotou Cancer Hospital, Baotou, 014030, Inner Mongolia, China
| | - R H Liang
- Department of Medical Oncology, Baotou Cancer Hospital, Baotou, 014030, Inner Mongolia, China
| | - C Y Wang
- Department of Medical Oncology, Baotou Cancer Hospital, Baotou, 014030, Inner Mongolia, China
| | - R P Zhang
- Department of Medical Oncology, Baotou Cancer Hospital, Baotou, 014030, Inner Mongolia, China
| | - S Y Wu
- Department of Medical Oncology, Baotou Cancer Hospital, Baotou, 014030, Inner Mongolia, China
| | - X Han
- Department of Breast Surgery, Baotou Cancer Hospital, No. 18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China
| | - G L Zhang
- Department of Breast Surgery, Baotou Cancer Hospital, No. 18 Tuanjie Street, Qingshan District, Baotou, 014030, Inner Mongolia, China.
| |
Collapse
|
16
|
Parol M, Gzil A, Bodnar M, Grzanka D. Systematic review and meta-analysis of the prognostic significance of microRNAs related to metastatic and EMT process among prostate cancer patients. J Transl Med 2021; 19:28. [PMID: 33413466 PMCID: PMC7788830 DOI: 10.1186/s12967-020-02644-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
The ability of tumor cells to spread from their origin place and form secondary tumor foci is determined by the epithelial-mesenchymal transition process. In epithelial tumors such as prostate cancer (PCa), the loss of intercellular interactions can be observed as a change in expression of polarity proteins. Epithelial cells acquire ability to migrate, what leads to the formation of distal metastases. In recent years, the interest in miRNA molecules as potential future treatment options has increased. In tumor microenvironment, miRNAs have the ability to regulate signal transduction pathways, where they can act as suppressors or oncogenes. MiRNAs are secreted by cancer cells, and the changes in their expression levels are closely related to a cancer progression, including epithelial-mesenchymal transition. These molecules offer new diagnostic and therapeutic possibilities. Therapeutics which make use of synthesized RNA fragments and mimic or block miRNAs affected in PCa, may lead to inhibition of tumor progression and even disease re-emission. Based on appropriate qualification criteria, we conducted a selection process to identify scientific articles describing miRNAs and their relation to epithelial-mesenchymal transition in PCa patients. The studies were published in English on Pubmed, Scopus and the Web of Science before August 08, 2019. Hazard ratios (HRs) and 95% confidence intervals (CI) as well as total Gleason score were used to assess the concordance between miRNAs and presence of metastases. A total of 13 studies were included in our meta-analysis, representing 1608 PCa patients and 15 miRNA molecules. Our study clarifies a relationship between the clinicopathological features of PCa and the aberrant expression of several miRNA as well as the complex mechanism of miRNA molecules involvement in the induction and promotion of the metastatic mechanism in PCa.
Collapse
Affiliation(s)
- Martyna Parol
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 9 Curie-Sklodowskiej Street, 85-094 Bydgoszcz, Poland
| | - Arkadiusz Gzil
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 9 Curie-Sklodowskiej Street, 85-094 Bydgoszcz, Poland
| | - Magdalena Bodnar
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 9 Curie-Sklodowskiej Street, 85-094 Bydgoszcz, Poland
| | - Dariusz Grzanka
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 9 Curie-Sklodowskiej Street, 85-094 Bydgoszcz, Poland
| |
Collapse
|
17
|
Kang T, Xing W, Xi Y, Chen K, Zhan M, Tang X, Wang Y, Zhang R, Lei M. MiR-543 regulates myoblast proliferation and differentiation of C2C12 cells by targeting KLF6. J Cell Biochem 2020; 121:4827-4837. [PMID: 32348593 DOI: 10.1002/jcb.29710] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022]
Abstract
MicroRNA-543 (miR-543) has been found to play a suppressive role in various human cancers in many studies, whereas the specific functions of miR-543 in muscle development remain poorly understood. Here, we found that the expression of miR-543 was high in skeletal muscle and increased during the differentiation of C2C12 cells. Overexpression of miR-543 repressed C2C12 cell proliferation and promoted differentiation, while knockdown of miR-543 expression produced the opposite results. During myogenesis, we predicted and verified that Krüppel-like factor 6 (KLF6), a suppressor of multiple tumor cells, was a target gene of miR-543. Then, miR-543 was found to specifically target KLF6 and repress its expression. Besides this, knockdown of KLF6 promoted the differentiation but inhibited the proliferation of C2C12 cells. Si-KLF6 can rescue the influence of miR-543 inhibitor on C2C12 cell differentiation. Our results indicate a new regulatory mechanism of miR-543 on KLF6 expression and suggest the possibility of using the miR-543/KLF6 pathway as a potential target for studying myogenesis.
Collapse
Affiliation(s)
- Tingting Kang
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, and Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wenkai Xing
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, and Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yu Xi
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, and Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Kun Chen
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, and Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Mengsi Zhan
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, and Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaoyin Tang
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, and Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yueying Wang
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, and Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ruirui Zhang
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, and Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Minggang Lei
- Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, and Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
18
|
Yuan W, Gao H, Wang G, Miao Y, Jiang K, Zhang K, Wu J. Higher miR-543 levels correlate with lower STK31 expression and longer pancreatic cancer survival. Cancer Med 2020; 9:9632-9640. [PMID: 33128354 PMCID: PMC7774731 DOI: 10.1002/cam4.3559] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 01/03/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most malignant gastrointestinal tumors and the 5‐year survival is only 9%. The expression of miRNAs in serum has been proved to be related to tumorigenesis and development of cancers. The miRNA targets and gene targets were predicted in microRNA.org, miRDB, TargetScan, and RNAInter. The expression data of STK31 (Serine/Threonine Kinase 31) and miRNAs generated from PC samples was from TCGA and the relationship of expression of STK31 and miR‐543 was confirmed in PC samples from our center. Double luciferase reporter gene assay was used to demonstrate the direct binding between miR‐543 and STK31. The effect of expression level of miRNAs on survival time was assessed by Kaplan–Meier curves. The Go Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of miR‐543‐related genes were performed. The results showed that miR‐543 had a statistically significant correlation with the expression of STK31 and contained the direct binding site with STK31. The expression level of miR‐543 may affect the survival of PC. The results of GO and KEGG pathway analysis showed that miR‐543 might play a key role in Insulin signaling pathway. MiR‐543 could be combined with STK31 and affect the expression of STK31. The expression of miR‐543 could also predict the survival of patients with PC, which suggested that miR‐543 might play an important role in PC. The GO and KEGG pathway analysis also displayed that miR‐543 was involved in several other pathways of pancreas.
Collapse
Affiliation(s)
- Weizhong Yuan
- Pancreatic Center & Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
- Pancreas Institute of Nanjing Medical UniversityNanjingJiangsuChina
- Department of General SurgeryNanjing Meishan HospitalNanjingJiangsuChina
| | - Hao Gao
- Pancreatic Center & Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
- Pancreas Institute of Nanjing Medical UniversityNanjingJiangsuChina
| | - Guangfu Wang
- Pancreatic Center & Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
- Pancreas Institute of Nanjing Medical UniversityNanjingJiangsuChina
| | - Yi Miao
- Pancreatic Center & Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
- Pancreas Institute of Nanjing Medical UniversityNanjingJiangsuChina
| | - Kuirong Jiang
- Pancreatic Center & Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
- Pancreas Institute of Nanjing Medical UniversityNanjingJiangsuChina
| | - Kai Zhang
- Pancreatic Center & Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
- Pancreas Institute of Nanjing Medical UniversityNanjingJiangsuChina
| | - Junli Wu
- Pancreatic Center & Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
- Pancreas Institute of Nanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
19
|
Jin Y, Zhang S, Liu L. Circular RNA circ_C16orf62 Suppresses Cell Growth in Gastric Cancer by miR-421/Tubulin beta-2A Chain (TUBB2A) Axis. Med Sci Monit 2020; 26:e924343. [PMID: 33006960 PMCID: PMC7537480 DOI: 10.12659/msm.924343] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background Gastric cancer (GC) is the third leading cause of cancer-associated mortality in the world. Expression of circular RNA circ_C16orf62 is reported to be low in GC. The role and mechanism of circ_C16orf62 remain unclear. Material/Methods Expression levels of circ_C16orf62 and tubulin beta-2A chain (TUBB2A) in GC tissues and cells, and microRNA-421 (miR-421) level in GC cells were detected by real-time quantitative polymerase chain reaction (RT-qPCR). The predominant cytoplasmic localization of circ_C16orf62 was identified by subcellular fractionation. The protein level of TUBB2A was detected by western blot assay. Cell proliferative ability, migration, and invasion were measured by 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT), colony formation, and several transwell assaysy. The binding relationship between miR-421 and circ_C16orf62 or TUBB2A was predicted by starBase3.0 or Targetscan, and then verified by the dual-luciferase reporter assay. The biological role ofcirc_C16orf62 was examined by xenograft tumor model in vivo. Results Circ_C16orf62 andTUBB2A were downregulated in GC tissues and cells. Circ_C16orf62 was predominantly located in the cytoplasm of GC cells, and repressed proliferation, migration, and invasion of GC cells. Mechanistically, circ_C16orf62 worked as the miR-421 sponge to upregulate TUBB2A in GC, thereby hindering GC growth. Circ_C16orf62 repressed GC tumor growth in vivo. Conclusions These findings demonstrate that circ_C16orf62 impeded proliferation, migration, and invasion in vitro and retarded tumor growth in vivo by the miR-421/TUBB2A axis in GC, providing a potential therapeutic strategy for patients with GC.
Collapse
Affiliation(s)
- Yanfeng Jin
- Department of Gastroenterology, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Shanshan Zhang
- Department of Gastroenterology, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Li Liu
- Department of Oncology, The Second Hospital of Weifang, Weifang, Shandong, China (mainland)
| |
Collapse
|
20
|
Yong H, Fu J, Gao G, Shi H, Zheng D, Zhou X. MiR-34a suppresses the proliferation and invasion of gastric cancer by modulating PDL1 in the immune microenvironment. Mol Cell Probes 2020; 53:101601. [DOI: 10.1016/j.mcp.2020.101601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/15/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
|
21
|
Wang H, Huang Y, Yang Y. LncRNA PVT1 Regulates TRPS1 Expression in Breast Cancer by Sponging miR-543. Cancer Manag Res 2020; 12:7993-8004. [PMID: 32982402 PMCID: PMC7493016 DOI: 10.2147/cmar.s263383] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/13/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Breast cancer is the most common female malignancy with high invasion and metastasis abilities. Studies have shown that long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 gene (PVT1) is an oncogene and is positively correlated with progression and metastasis of breast tumors. However, the detailed mechanism of PVT1 in breast cancer tumorigenesis is not fully understood. METHODS Real-time polymerase quantitative chain reaction (RT-qPCR) was performed to identify the expression levels of PVT1, miR-543 and trichorhinophalangeal syndrome-1 gene (TRPS1) in breast cancer tissues and cells. Cell proliferation was measured by plate clone formation and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazol-3-ium bromide (MTT) assay. Apoptosis and motility of MCF-7 and MDA-MB-436 cells were assessed with flow cytometry assay and transwell migration and invasion analyses, respectively. In addition, a model was established to probe the function of PVT1 silencing in vivo. The target relationship among PVT1, miR-543 or TRPS1 was confirmed by dual-luciferase reporter analysis, RNA immunoprecipitation (RIP) and RNA pull down assays. The protein expression level of TRPS1 was evaluated with Western blot assay. RESULTS PVT1 expression was upregulated in breast cancer tissues and cell lines. In addition, PVT1 silencing inhibited breast cancer cell growth and motility, while increased apoptosis. Meanwhile, the effects of PVT1 or miR-543 could be reversed by introducing overexpressed plasmid of miR-543 or TRPS1 in breast cancer cell lines, respectively. CONCLUSION Knockdown of PVT1 repressed breast cancer cell growth and motility, and induced apoptosis in vitro and reduced tumor volume and weight in vivo. Mechanically, the overexpression of PVT1 enhanced TRPS1 level by negatively targeted miR-543 in breast cancer.
Collapse
Affiliation(s)
- Hongtao Wang
- Department of Pharmacy, The Second Clinical Medical College, Yangtze University, Jingzhou434020, People’s Republic of China
| | - Yuanli Huang
- Department of Galactophore, The Second Clinical Medical College, Yangtze University, Jingzhou434020, People’s Republic of China
| | - Yuanrong Yang
- Department of Pharmacy, The Second Clinical Medical College, Yangtze University, Jingzhou434020, People’s Republic of China
| |
Collapse
|
22
|
He S, Wang X, Zhang J, Zhou F, Li L, Han X. TRG-AS1 is a potent driver of oncogenicity of tongue squamous cell carcinoma through microRNA-543/Yes-associated protein 1 axis regulation. Cell Cycle 2020; 19:1969-1982. [PMID: 32615889 PMCID: PMC7469544 DOI: 10.1080/15384101.2020.1786622] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The long noncoding RNA T cell receptor gamma locus antisense RNA 1 (TRG-AS1) plays an important role in glioblastoma progression. The objective of this study was to determine the expression status of TRG-AS1 in tongue squamous cell carcinoma (TSCC). The regulatory effects of TRG-AS1 depletion on the malignant processes of TSCC cells were illustrated both in vitro and in vivo. Additionally, the precise molecular mechanisms through which TRG-AS promotes TSCC oncogenicity were investigated. TRG-AS1 expression in TSCC tissues and cell lines was detected using reverse transcription-quantitative PCR. Functional experiments including Cell Counting Kit-8 assay, flow cytometric apoptotic assay, migration and invasion assays, and xenograft tumor model analysis were conducted to severally determine the effects of TRG-AS1 on TSCC cell proliferation, apoptosis, migration, and invasion in vitro and tumor growth in vivo. Herein, TRG-AS1 was highly expressed in TSCC and closely associated with advanced TNM stage, high lymph node metastasis, and poor overall survival. Functionally, TRG-AS1 depletion suppressed TSCC cell proliferation, migration, and invasion in vitro; promoted cell apoptosis; and attenuated tumor growth in vivo. Mechanistically, TRG-AS1 served as a molecular sponge for microRNA-543 (miR-543), thereby contributing to the increased expression of Yes-associated protein 1 (YAP1) - a miR-543 target. Rescue experiments confirmed that miR-543 inhibition or YAP1 overexpression abrogated the anticancer effects of TRG-AS1 silencing in TSCC cells. In conclusion, TRG-AS1 aggravates TSCC malignancy by regulating the miR-543/YAP1 axis. Identification of the TRG-AS1/miR-543/YAP1 regulatory pathway may provide novel insights into TSCC diagnosis, prognosis, and therapy.
Collapse
Affiliation(s)
- Shuwei He
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Jingjing Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Fan Zhou
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Lei Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xingmin Han
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China,CONTACT Xingmin Han
| |
Collapse
|
23
|
Zhou C, Zhao X, Duan S. The role of miR-543 in human cancerous and noncancerous diseases. J Cell Physiol 2020; 236:15-26. [PMID: 32542683 DOI: 10.1002/jcp.29860] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
MicroRNA (miRNA) is a noncoding single-stranded RNA molecule that can regulate the posttranscriptional expression level of a gene by binding to the 3'-untranslated region (3'-UTR) of the target messenger RNA. miR-543 is a kind of miRNA, which plays an important role in the occurrence and development of various human cancerous and noncancerous diseases. miR-543 directly or indirectly regulates a large number of downstream target genes and plays an important role in cellular components, biological processes, and molecular functions. In addition, many studies have verified the regulatory mechanism, physiological role, biological function, and prognostic value of miR-543. Therefore, this article reviews the papers published in the past decade and elaborates on the research progress of miR-543 from the aspects of physiology and pathology, especially in cancerous and other noncancerous diseases. In particular, we pay attention to the expression patterns, direct targets, biological functions, related pathways, and prognostic value of miR-543 reported in experimental articles. And by comparing similar research articles, we point out existing controversies in this field to date, so as to facilitate further research in the future.
Collapse
Affiliation(s)
- Cong Zhou
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Xin Zhao
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Shiwei Duan
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
24
|
Zhao H, Zheng Y, You J, Xiong J, Ying S, Xie L, Song X, Yao Y, Jin Z, Zhang C. Tumor suppressor role of miR-876-5p in gastric cancer. Oncol Lett 2020; 20:1281-1287. [PMID: 32724369 PMCID: PMC7377156 DOI: 10.3892/ol.2020.11680] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 11/07/2019] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is the second most common cancer cause of cancer-related mortality worldwide. Recent studies have demonstrated the function of microRNAs (miRNAs) in the pathogenesis of GC. miR-876-5p demonstrated an antitumor role in hepatocellular carcinoma and lung cancer; however, the function of miR-876-5p has not yet been fully identified in GC. Thus, the present study aimed to investigate the role of miR-876-5p in GC. The results of the present study demonstrated low expression levels of miR-876-5p in GC tumor tissues. Furthermore, overexpression of miR-876-5p inhibited GC cell proliferation and promoted apoptosis, whilst miR-876-5p knockdown promoted GC cell proliferation and decreased cisplatin sensitivity of GC cells. Transforming growth factor β-receptor 1 was demonstrated to be a potential target gene of miR-876-5p. Overall, the results of the present study suggest that miR-876-5p plays an antitumor role in GC.
Collapse
Affiliation(s)
- Hongwei Zhao
- Department of Emergency, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, P.R. China
| | - Yuzhu Zheng
- Oncology Department, The Third People's Hospital of Chengdu, Southwest Jiaotong University Affiliated Hospital, Chengdu, Sichuan 611756, P.R. China
| | - Jia You
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jingyuan Xiong
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shi Ying
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Linshen Xie
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Research Center for Occupational Respiratory Diseases, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xuejiao Song
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuqin Yao
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Research Center for Occupational Respiratory Diseases, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhaohui Jin
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chaoxiong Zhang
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Research Center for Occupational Respiratory Diseases, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
25
|
Ji W, Mu Q, Liu XY, Cao XC, Yu Y. ZNF281-miR-543 Feedback Loop Regulates Transforming Growth Factor-β-Induced Breast Cancer Metastasis. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:98-107. [PMID: 32512343 PMCID: PMC7281305 DOI: 10.1016/j.omtn.2020.05.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/12/2020] [Accepted: 05/18/2020] [Indexed: 12/31/2022]
Abstract
Breast cancer is the most common malignancy, and metastasis is the main cause of cancer-associated mortality in women worldwide. Transforming growth factor-β (TGF-β) signaling, an inducer of epithelial-to-mesenchymal transition (EMT), plays an important role in breast cancer metastasis. Abnormal expression of miR-543 is associated with tumorigenesis and progression of various human cancers; however, the knowledge about the role of miR-543 in breast cancer metastasis is still unknown. In this study, we demonstrated that miR-543 inhibits the EMT-like phenotype and TGF-β-induced breast cancer metastasis both in vitro and in vivo by targeting ZNF281. ZNF281 transactivates the EMT-related transcription factor ZEB1 and Snail. Furthermore, both ZEB1 and Snail can transcriptionally suppress miR-543 expression. Taken together, our data uncover the ZNF281-miR-543 feedback loop and provide a mechanism to extend the understanding of TGF-β network complexity.
Collapse
Affiliation(s)
- Wei Ji
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Qiang Mu
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; The First Department of Breast Surgery, Qingdao Central Hospital, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao 266042, China
| | - Xiang-Yu Liu
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Department of Gynecological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xu-Chen Cao
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Yue Yu
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.
| |
Collapse
|
26
|
Wang D, Cai L, Tian X, Li W. MiR-543 promotes tumorigenesis and angiogenesis in non-small cell lung cancer via modulating metastasis associated protein 1. Mol Med 2020; 26:44. [PMID: 32410569 PMCID: PMC7222519 DOI: 10.1186/s10020-020-00175-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE This study is aimed to explore the role of miR-543 in non-small cell lung cancer (NSCLC), and verify whether miR-543 targets metastasis associated protein 1 (MTA1) to affect tumorigenesis and angiogenesis in NSCLC. METHODS Firstly, miR-543 mimic and inhibitor were transfected into A549 cells and H1299 cells. The cells proliferation was tested by MTT and clone formation. The cells apoptosis was analyzed by cytometry. Tube formation assay was used to measure the vascularization of cells. qRT-PCR and Western Blot were used to measure the MTA1 expression. Dual-luciferase assay was used to analyze whether miR-543 targets MTA1. Secondly, MTA1 mimic and inhibitor were transfected into cells to analyze the effect of MTA1 on proliferation and angiogenesis in NSCLC cells. Lastly, the nude mice were used to verify the effect of miR-543 on tumorigenesis and angiogeneisis in NSCLC via modulating MATA1. RESULTS miR-543 overexpression could apparently promote cells proliferation and angiogeneisis in NSCLC cells. Meanwhile, the MTA1 expression was increased after transfecting miR-543 mimic. Dual luciferase reporter assay revealed MTA1 was a downstream target of miR-543. Further studies showed that inhibition of MTA1 weakened the role of miR-543 overexpression in NSCLC cells. Vivo experiments revealed that miR-543 promoted cells proliferation and angiogenesis in tumor tissues via modulating MTA1. CONCLUSION miR-543 could target MTA1 to promote tumorigenesis and angiogenesis in NSCLC via targeting MTA1.
Collapse
MESH Headings
- 3' Untranslated Regions
- Animals
- Apoptosis/genetics
- Biomarkers
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Disease Models, Animal
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Mice
- MicroRNAs/genetics
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- RNA Interference
- Repressor Proteins/genetics
- Trans-Activators/genetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Dawei Wang
- Department of Thoracic Surgery, Yantaishan Hospital, Yantai, 264000, Shandong, China
| | - Li Cai
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Xudong Tian
- Department of Thoracic Surgery, Liaocheng People's Hospital and Liaocheng Clinical School, No. 67 Dongchang West Road, Liaocheng, 252000, Shandong, China
| | - Wenjun Li
- Department of Thoracic Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhungding East Road, Zhifu District, Yantai, 264000, Shandong, China.
| |
Collapse
|
27
|
Arginine Supply Impacts the Expression of Candidate microRNA Controlling Milk Casein Yield in Bovine Mammary Tissue. Animals (Basel) 2020; 10:ani10050797. [PMID: 32380753 PMCID: PMC7277595 DOI: 10.3390/ani10050797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 12/21/2022] Open
Abstract
Arginine, a semi-essential functional amino acid, has been found to promote the synthesis of casein in mammary epithelial cells to some extent. Data from mouse indicated that microRNA (miRNA) are important in regulating the development of mammary gland and milk protein synthesis. Whether there are potential links among arginine, miRNA and casein synthesis in bovine mammary gland is uncertain. The objective of the present work was to detect the effects of arginine supplementation on the expression of miRNA associated with casein synthesis in mammary tissue and mammary epithelial cells (BMEC). The first study with bovine mammary epithelial cells (BMEC) focused on screening for miRNA candidates associated with the regulation of casein production by arginine. The BMEC were cultured with three different media, containing 0, 1.6 and 3.2 mM arginine, for 24 h. The expression of candidate miRNA was evaluated. Subsequently, in an in vivo study, 6 Chinese Holstein dairy cows with similar BW (mean ± SE) (512.0 ± 19.6 kg), parity (3), BCS (4.0) and DIM (190 ± 10.3 d) were randomly assigned to three experimental groups. The experimental cows received an infusion of casein, arginine (casein plus double the concentration of arginine in casein), and alanine (casein plus alanine, i.e., iso-nitrogenous to the arginine group) in a replicated 3 × 3 Latin square design with 22 d for each period (7 d for infusion and 15 d for washout). Mammary gland biopsies were obtained from each cow at the end of each infusion period. Results of the in vitro study showed differences between experimental groups and the control group for the expression of nine miRNA: miR-743a, miR-543, miR-101a, miR-760-3p, miR-1954, miR-712, miR-574-5p, miR-468 and miR-875-3p. The in vivo study showed that arginine infusion promoted milk protein content, casein yield and the expression of CSN1S1 and CSN1S2. Furthermore, the expression of miR-743a, miR-543, miR-101a, miR-760-3p, miR-1954, and miR-712 was also greater in response to arginine injection compared with the control or alanine group. Overall, results both in vivo and in vitro revealed that arginine might partly influence casein yield by altering the expression of 6 miRNAs (miR-743a, miR-543, miR-101a, miR-760-3p, miR-1954, and miR-712).
Collapse
|
28
|
Zhang Z, Nong L, Chen ML, Gu XL, Zhao WW, Liu MH, Cheng WW. Long Noncoding RNA SNHG10 Sponges miR-543 to Upregulate Tumor Suppressive SIRT1 in Nonsmall Cell Lung Cancer. Cancer Biother Radiopharm 2020; 35:771-775. [PMID: 32319822 DOI: 10.1089/cbr.2019.3334] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: Long noncoding RNA SNHG10 has been reported to promote the development of liver cancer. While by analyzing The Cancer Genome Atlas (TCGA) dataset we observed the downregulation of SNHG10 in non-small cell lung cancer (NSCLC). This study aimed to investigate the roles of SNHG10 in NSCLC. Materials and Methods: This study included 60 pairs of NSCLC and nontumor tissue samples collected from 60 NSCLC patients (males and females, 39-66 years, 50.9 ± 5.5 years). Gene expression was detected by quantitative polymerase chain reaction and western blot. Overexpression experiments were used to analyze gene interactions. Effects of cell transfections on cell proliferation were analyzed by performing CCK-8 cell proliferation assays. Results: We confirmed the downregulation of SNHG10 in NSCLC. In addition, low expression level of SNHG10 predicted the poor survival of NSCLC patients. SNHG10 can directly interact with miR-543, while overexpression of miR-543 failed to downregulate SNHG10. However, SNHG10 overexpression led to upregulation of sirtuin 1 (SIRT1), a downstream target of miR-543. Cell proliferation assay showed that SNHG10 and SIRT1 overexpression led to the decreased proliferation rate of NSCLC cells. In contrast, miR-543 over-expression played an opposite role and reduced the effects of SNHG10 and SIRT1 overexpression. Conclusions: In conclusion, SNHG10 sponges miR-543 to upregulate tumor suppressive SIRT1 in NSCLC to suppress cell proliferation.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Nong
- Department of Oncology, The Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng-Lei Chen
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Li Gu
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei-Wei Zhao
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ming-Hui Liu
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Wu Cheng
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Fuentes-Mattei E, Bayraktar R, Manshouri T, Silva AM, Ivan C, Gulei D, Fabris L, Soares do Amaral N, Mur P, Perez C, Torres-Claudio E, Dragomir MP, Badillo-Perez A, Knutsen E, Narayanan P, Golfman L, Shimizu M, Zhang X, Zhao W, Ho WT, Estecio MR, Bartholomeusz G, Tomuleasa C, Berindan-Neagoe I, Zweidler-McKay PA, Estrov Z, Zhao ZJ, Verstovsek S, Calin GA, Redis RS. miR-543 regulates the epigenetic landscape of myelofibrosis by targeting TET1 and TET2. JCI Insight 2020; 5:121781. [PMID: 31941838 PMCID: PMC7030823 DOI: 10.1172/jci.insight.121781] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 12/04/2019] [Indexed: 12/13/2022] Open
Abstract
Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by cytopenia and extramedullary hematopoiesis, resulting in splenomegaly. Multiple pathological mechanisms (e.g., circulating cytokines and genetic alterations, such as JAKV617F mutation) have been implicated in the etiology of MF, but the molecular mechanism causing resistance to JAK2V617F inhibitor therapy remains unknown. Among MF patients who were treated with the JAK inhibitor ruxolitinib, we compared noncoding RNA profiles of ruxolitinib therapy responders versus nonresponders and found miR-543 was significantly upregulated in nonresponders. We validated these findings by reverse transcription-quantitative PCR. in this same cohort, in 2 additional independent MF patient cohorts from the United States and Romania, and in a JAK2V617F mouse model of MF. Both in vitro and in vivo models were used to determine the underlying molecular mechanism of miR-543 in MF. Here, we demonstrate that miR-543 targets the dioxygenases ten-eleven translocation 1 (TET1) and 2 (TET2) in patients and in vitro, causing increased levels of global 5-methylcytosine, while decreasing the acetylation of histone 3, STAT3, and tumor protein p53. Mechanistically, we found that activation of STAT3 by JAKs epigenetically controls miR-543 expression via binding the promoter region of miR-543. Furthermore, miR-543 upregulation promotes the expression of genes related to drug metabolism, including CYP3A4, which is involved in ruxolitinib metabolism. Our findings suggest miR-543 as a potentially novel biomarker for the prognosis of MF patients with a high risk of treatment resistance and as a potentially new target for the development of new treatment options.
Collapse
Affiliation(s)
| | | | - Taghi Manshouri
- Department of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Andreia M. Silva
- Department of Experimental Therapeutics and
- Instituto de Investigação e Inovação em Saúde (i3S)
- Instituto de Engenharia Biomédica (INEB), and
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Cristina Ivan
- Department of Experimental Therapeutics and
- Center for RNA Interference and Non-coding RNAs, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Diana Gulei
- Department of Experimental Therapeutics and
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy Iuliu Hatieganu, Cluj-Napoca, Romania
- Department of Functional Genomics, The Oncology Institute, Cluj-Napoca, Romania
| | | | - Nayra Soares do Amaral
- Department of Experimental Therapeutics and
- Molecular Morphology Laboratory, Department of Investigative Pathology, AC Camargo Cancer Center, São Paulo, Brazil
| | - Pilar Mur
- Hereditary Cancer Program, Catalan Institute of Oncology, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Perez
- Department of Experimental Therapeutics and
- Mayagüez Campus, University of Puerto Rico, Mayagüez, Puerto Rico, USA
| | - Elizabeth Torres-Claudio
- Department of Experimental Therapeutics and
- University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico, USA
| | - Mihnea P. Dragomir
- Department of Experimental Therapeutics and
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy Iuliu Hatieganu, Cluj-Napoca, Romania
- Department of Surgery, Fundeni Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | | | | | - Leonard Golfman
- Department of Pediatrics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | | | - Xinna Zhang
- Center for RNA Interference and Non-coding RNAs, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Wanke Zhao
- Department of Pathology, Health Sciences Center, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Wanting Tina Ho
- Department of Pathology, Health Sciences Center, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Marcos Roberto Estecio
- Department of Epigenetics and Molecular Carcinogenesis and
- Center for Cancer Epigenetics, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | | | - Ciprian Tomuleasa
- Department of Hematology, The Oncology Institute Ion Chiricuta, University of Medicine and Pharmacy Iuliu Hatieganu, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, University of Medicine and Pharmacy Iuliu Hatieganu, Cluj-Napoca, Romania
- Department of Functional Genomics, The Oncology Institute, Cluj-Napoca, Romania
| | | | - Zeev Estrov
- Department of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Zhizhuang J. Zhao
- Department of Pathology, Health Sciences Center, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Srdan Verstovsek
- Department of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - George A. Calin
- Department of Experimental Therapeutics and
- Center for RNA Interference and Non-coding RNAs, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | | |
Collapse
|
30
|
Xia K, Zhang Y, Sun D. miR‑217 and miR‑543 downregulation mitigates inflammatory response and myocardial injury in children with viral myocarditis by regulating the SIRT1/AMPK/NF‑κB signaling pathway. Int J Mol Med 2019; 45:634-646. [PMID: 31894309 DOI: 10.3892/ijmm.2019.4442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 09/05/2019] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the expression levels and roles of microRNA (miR)‑217 and miR‑543 in viral myocarditis, and to examine their underlying mechanisms. Coxsackievirus B3 (CVB3) was used to establish in vivo and in vitro models of viral myocarditis. The levels of miR‑217 and miR‑543 were detected using reverse transcription‑quantitative PCR. The association between miR‑217 and miR‑543 and sirtuin‑1 (SIRT1) was predicted and confirmed by TargetScan and dual‑luciferase reporter assay. Cell viability was detected using Cell Counting Kit‑8 assay, and cell apoptosis was measured by analyzing the expression levels of Bcl‑2 and Bax, and by flow cytometry. In addition, the synthesis of various pro‑inflammatory factors was determined by ELISA. In addition, superoxide dismutase (SOD) activity and malondialdehyde (MDA) levels were measured in cardiomyocytes following transfection and CVB infection. miR‑217 and miR‑543 were found to be highly expressed in the peripheral blood of pediatric patients with viral myocarditis, in the peripheral blood and myocardial tissues of viral myocarditis mice and in CVB3‑infected cardiomyocytes. SIRT1 was found to be a target of both miR‑217 and miR‑543, and SIRT1 expression level was downregulated in viral myocarditis. Further analysis indicated that the reduced cell viability, increased cell apoptosis, enhanced synthesis of inflammatory factors, increased MDA content and decreased SOD activity associated with myocarditis were significantly reversed after inhibition of miR‑217 or miR‑543. Importantly, the present results showed that all the effects of miR‑217 and miR‑543 inhibition on cardiomyocytes were significantly suppressed following SIRT1 knockdown. Collectively, the present data indicated that miR‑217 and miR‑543 were significantly upregulated in viral myocarditis, and downregulation of miR‑217 and miR‑543 attenuated CVB3 infection‑induced cardiomyocyte injury by targeting SIRT1. miR‑217 and miR‑543 may be potential therapeutic targets for developing novel viral myocarditis treatments in the future.
Collapse
Affiliation(s)
- Kun Xia
- Department of Cardiovascular Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430019, P.R China
| | - Yong Zhang
- Department of Cardiovascular Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430019, P.R China
| | - Dongming Sun
- Department of Cardiovascular Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430019, P.R China
| |
Collapse
|
31
|
Wang Y, Liu G, Sun S, Qin J. miR-1294 alleviates epithelial-mesenchymal transition by repressing FOXK1 in gastric cancer. Genes Genomics 2019; 42:217-224. [PMID: 31833046 DOI: 10.1007/s13258-019-00899-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) have been reported play critical roles in regulating tumor development and progression. OBJECTIVE This study aimed to investigate the potential effect of miR-1294 in gastric cancer (GC). METHODS Reverse transcription quantitative polymerase chain reaction (RT-PCR) were performed to verify the expression level of miR-1294 and Forkhead box protein K1 (FOXK1). Overall survival data of miR-1294 for GC was analysed by log-rank test. Targetscan was used to screen potential target gene of miR-1294. Dual luciferase assay was assessed to investigate the relationship between miR-1294 and FOXK1. The miR-1294 overexpression and knockdown were designed to study the biological function of miR-1294. The migration and invasion of GC cell lines were investigated by wound healing and transwell assays. Western blotting were performed to verify the expression level of epithelial marker, mesenchymal markers and FOXK1. Overexpression of FOXK1 was designed to study the rescue effects of FOXK1 in SGC7901 cell. RESULTS miR-1294 was found downregulated in GC patients and cell lines. A higher miR-1294 expression showed a significant longer overall survival than those with a lower miR-1294 expression. miR-1294 directly targets FOXK1 and regulates the expression of FOXK1. In addition, miR-1294 regulates epithelial-mesenchymal transition (EMT) by inhibiting FOXK1 in GC cells and it can be rescued by overexpression of FOXK1. CONCLUSION miR-1294 alleviates EMT process in GC by targeting FOXK1.
Collapse
Affiliation(s)
- Yaru Wang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, Shandong, China
| | - Guangming Liu
- Department of Gastroenterology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China.
| | - Shijuan Sun
- Department of Gastroenterology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Junjie Qin
- Department of Gastroenterology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, Jilin, China
| |
Collapse
|
32
|
Wang YC, He WY, Dong CH, Pei L, Ma YL. lncRNA HCG11 regulates cell progression by targeting miR-543 and regulating AKT/mTOR pathway in prostate cancer. Cell Biol Int 2019; 43:1453-1462. [PMID: 31228307 DOI: 10.1002/cbin.11194] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 06/16/2019] [Indexed: 01/24/2023]
Abstract
Prostate cancer (PCa) is a common cancer worldwide, which mostly occurs in males over the age of 50. Accumulating evidence have determined that long non-coding RNA/microRNA (lncRNA/miRNA) axis plays a critical role in cell progression of cancers, including PCa. However, the pathogenesis of PCa has not been fully indicated. In this study, quantitative real-time polymerase chain reaction was used to detect the expression of HCG11 and miR-543. Western blot was applied to measure the protein expression of proliferating cell nuclear antigen, cleavage-caspase 3 (cle-caspase 3), N-cadherin, E-cadherin, GAPDH, P-AKT, AKT, p-mTOR, and mTOR. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), transwell invasion, and transwell migration assay were used to detect cell proliferation, invasion, and migration, respectively. The function and mechanism of lncRNA HCG11 were confirmed in PCa cell and xenograft mice models. Luciferase assay indicated that miR-543 was a target miRNA of HCG11. Further investigation revealed that overexpression of HCG11 inhibited cell proliferation, invasion, and migration, whereas induced cell apoptosis by regulating miR-543 expression in vitro and in vivo. More than that, lncRNA HCG11 inhibited phosphoinositide-3 kinase/protein kinaseB (PI3K/AKT) signaling pathway to suppress PCa progression. Our data showed the overexpression of HGC11-inhibited PI3K/AKT signaling pathway by downregulating miR-543 expression, resulting in the suppression of cell growth in PCa. This finding proved a new regulatory network in PCa and provided a novel therapeutic target of PCa.
Collapse
Affiliation(s)
- Yan-Chao Wang
- Department of Urology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
| | - Wen-Yan He
- Department of Urology, Yan'an People's Hospital, Yan'an, Shaanxi, 716000, China
| | - Chun-Hui Dong
- Department of Urology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
| | - Long Pei
- Department of Urology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
| | - Yong-Liang Ma
- Department of Urology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
| |
Collapse
|
33
|
Zhang L, Chen J, Wang L, Chen L, Du Z, Zhu L, Cui M, Zhang M, Song L. Linc-PINT acted as a tumor suppressor by sponging miR-543 and miR-576-5p in esophageal cancer. J Cell Biochem 2019; 120:19345-19357. [PMID: 31464068 DOI: 10.1002/jcb.28699] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/25/2022]
Abstract
This manuscript aimed to investigate linc-PINT's role as a tumor suppressor and its downstream microRNAs (miRNAs) in esophageal cancer. Log-rank, Cox, and nomogram were used for survival analysis. Quantitative real-time polymerase chain reaction was used to evaluate the expression. Cell counting kit-8 was used for proliferation tests. As for in vivo experiments, low expression of linc-PINT was associated with better prognosis; besides, the nomogram indicated that linc-PINT, miR-543, and miR-576-5p served well in predicting the survival rate. As for the in vitro experiments, linc-PINT could directly regulate miR-543 and miR-576-5p to inhibit the proliferation of Eca-109 cell line. In conclusion, linc-PINT-miR-543/miR-576-5p pathway could predict the prognosis and provide novel therapeutic targets for esophageal cancer.
Collapse
Affiliation(s)
- Lingmin Zhang
- Department of Anesthesiology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jingxin Chen
- Department of Pathology, SenGong Hospital of Shaanxi, Xi'an, Shaanxi, China
| | - Li Wang
- Department of Scientific Research, The Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Lin Chen
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - ZhaoZhao Du
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - Manli Cui
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - Mingxin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - Liping Song
- Department of Radiotherapy, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
34
|
miR-543 promoted the cell proliferation and invasion of nasopharyngeal carcinoma by targeting the JAM-A. Hum Cell 2019; 32:477-486. [DOI: 10.1007/s13577-019-00274-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023]
|
35
|
Wang L, Chen W, Zha J, Yan Y, Wei Y, Chen X, Zhu X, Ge L. miR‑543 acts as a novel oncogene in oral squamous cell carcinoma by targeting CYP3A5. Oncol Rep 2019; 42:973-990. [PMID: 31322243 PMCID: PMC6667884 DOI: 10.3892/or.2019.7230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/03/2019] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs/miRs) are small non-coding RNAs that can act as oncogenes or tumor-suppressor genes in human cancer. Previous studies have revealed that abnormal expression of miRNAs is closely associated with tumor cell cycle, differentiation, growth and apoptosis. miR-543 is expressed abnormally in a wide variety of cancers and has been associated with cellular proliferation, apoptosis, and invasion; however, the effect of miR-543 remains unknown in oral squamous cell carcinoma (OSCC). In the present study, the expression level of miR-543 in OSCC cell lines and tissues was investigated by RT-qPCR. A series of experiments was then performed to elucidate the functions of miR-543 in OSCC, such as CCK-8 assay, colony formation assay, flow cytometry, cell cycle distribution assay and cell apoptosis assay and Transwell assay. miR-543 expression was significantly upregulated in tumors from patients with OSCC and in OSCC cell lines. Overexpression of miR-543 promoted the proliferation, invasion and migration of OSCC cell lines, and inhibited cell apoptosis. In addition, the present study identified cytochrome P450 family 3 subfamily A member 5 (CYP3A5) as a direct target of miR-543 using software analysis and dual-luciferase reporter assays. In conclusion, the results of the present study suggest that miR-543 acts as a tumor promoter and serves a vital role in OSCC proliferation and invasion. These results confirm that miR-543 may serve as a potential novel target for the treatment of OSCC.
Collapse
Affiliation(s)
- Liping Wang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Weihong Chen
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Jun Zha
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Yongyong Yan
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Yongxiang Wei
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Xili Chen
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Xinxin Zhu
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Linhu Ge
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| |
Collapse
|
36
|
Yarahmadi S, Abdolvahabi Z, Hesari Z, Tavakoli-Yaraki M, Yousefi Z, Seiri P, Hosseinkhani S, Nourbakhsh M. Inhibition of sirtuin 1 deacetylase by miR-211-5p provides a mechanism for the induction of cell death in breast cancer cells. Gene 2019; 711:143939. [PMID: 31220581 DOI: 10.1016/j.gene.2019.06.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/06/2019] [Accepted: 06/17/2019] [Indexed: 12/24/2022]
Abstract
Sirtuin 1 is one of the regulators of cell growth and survival and its inhibition is suggested as a suitable mechanism to overcome breast cancer development. In this study we explored the role of miR-211-5p in SIRT1/p53 pathway and its influence on breast cancer cell viability and apoptosis. Cells were transfected with miR-211-5p mimic and inhibitor to modulate cellular miR-211-5p levels in breast cancer cell lines, MDA-MB-231 and MCF-7. Gene expression of miR-211-5p and SIRT1 were measured with real-time PCR. SIRT1 protein level and the acetylation of p53 as well as SIRT1 activity were evaluated by Western blotting and fluorometry, respectively. In order to explore the direct attachment of miR-211-5p to the 3'-UTR of SIRT1 mRNA, luciferase reporter assay was applied. Cell viability in response to miR-211-5p was studied by MTT assay and apoptosis was assessed by annexin V labeling followed by flow cytometry. Results showed that SIRT1 gene and protein expression were inhibited by miR-211-5p and the 3'-UTR of SIRT1 was found to be directly targeted by miR-211-5p. Inhibition of SIRT1 expression resulted in its reduced activity. Up-regulation of miR-211-5p was also followed by a significant decline in the acetylation status of p53 which was associated with remarkable decreased cell viability and induction of apoptosis in breast cancer cells. Antisense oligonucleotide of miR-211-5p acted as its inhibitor and exerted opposite effects both on SIRT1 expression and cell apoptosis. In conclusion, inhibition of SIRT1 by miR-211-5p could effectively reduce breast cancer cell survival and cause cell death and therefore might be considered a seemly mechanism for designing anticancer therapies.
Collapse
Affiliation(s)
- Sahar Yarahmadi
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zohreh Abdolvahabi
- Department of Biochemistry and Genetics, Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Zahra Hesari
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Laboratory Science, Faculty of Paramedicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeynab Yousefi
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parvaneh Seiri
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran; Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Ke N, Pi LH, Liu Q, Chen L. Long noncoding RNA SNHG7 inhibits high glucose-induced human retinal endothelial cells angiogenesis by regulating miR-543/SIRT1 axis. Biochem Biophys Res Commun 2019; 514:503-509. [DOI: 10.1016/j.bbrc.2019.04.141] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/19/2019] [Indexed: 12/12/2022]
|
38
|
Abdolvahabi Z, Nourbakhsh M, Hosseinkhani S, Hesari Z, Alipour M, Jafarzadeh M, Ghorbanhosseini SS, Seiri P, Yousefi Z, Yarahmadi S, Golpour P. MicroRNA-590-3P suppresses cell survival and triggers breast cancer cell apoptosis via targeting sirtuin-1 and deacetylation of p53. J Cell Biochem 2019; 120:9356-9368. [PMID: 30520099 DOI: 10.1002/jcb.28211] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/15/2018] [Indexed: 12/29/2022]
Abstract
Downregulation of microRNA-590-3p (miR-590-3p) is a frequently occurring, nonphysiological event which is observed in several human cancers, especially breast cancer. However, the significance of miR-590-3p still remain unclear in the progression of this disease. This study explored the role of miR-590-3p in apoptosis of breast cancer cells. Gene expression of miR-590-3p, Sirtuin-1 (SIRT1), Bcl-2 associated X protein (BAX), and p21 was evaluated with real-time polymerase chain reaction (PCR) and SIRT1 protein expression was assessed by Western blot analysis in breast cancer cell lines. Bioinformatics analysis and luciferase reporter assay were used to evaluate targeting of SIRT1 messenger RNA (mRNA) by miR-590-3p. Cells were transfected with miR-590-3p mimic and inhibitor and their effects on the expression and activity of SIRT1 were evaluated. The effects of miR-590-3p upregulation on the acetylation of p53 as well as cell viability and apoptosis were assessed by Western blot analysis, WST-1 assay, and flow cytometry, respectively. miR-590-3p expression was considerably downregulated in breast cancer cells which was accompanied by upregulation of SIRT1 expression. SIRT1 was recognized as a direct target for miR-590-3p in breast cancer cells and its protein expression and activity was dramatically inhibited by the miR-590-3p. In addition, there was an increase in p53 and its acetylated form that ultimately led to upregulation of BAX and p21 expression, suppression of cell survival, and considerable induction of apoptosis in breast cancer cells. These findings suggest that miR-590-3p exerts tumor-suppressing effects through targeting SIRT1 in breast cancer cells, which makes it a potential therapeutic target for developing more efficient treatments for breast cancer.
Collapse
Affiliation(s)
- Zohreh Abdolvahabi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Hesari
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Alipour
- Department of Nano Biotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Advanced Medical Sciences & Technologies, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Meisam Jafarzadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Parvaneh Seiri
- Department of Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeynab Yousefi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sahar Yarahmadi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pegah Golpour
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Li X, Ning L, Zhao X, Wan S. MicroRNA-543 promotes ovariectomy-induced osteoporosis through inhibition of AKT/p38 MAPK signaling pathway by targeting YAF2. J Cell Biochem 2019; 120:8561-8569. [PMID: 30506950 DOI: 10.1002/jcb.28143] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 10/31/2018] [Indexed: 01/24/2023]
Abstract
The present study aimed to determine the roles of miRNA-543 in osteoporosis in rats induced by ovariectomy. The osteoporosis rat model was established by ovariectomy induction. MiRNA-543 expression in osteoblasts was measured by quantitative real-time polymerase chain reaction. The cell proliferation and apoptosis were measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and flow cytometry assays, respectively. Western blot analysis was conducted to examine the expression of YAF-2 and AKT signaling. TargetScan analysis and dual-luciferase reporter assay were performed to determine the target gene of miRNA-543. MiRNA-543 was significantly upregulated in osteoporosis rat model. Overexpression of miRNA-543 significantly suppressed cell growth and promoted apoptosis in osteoblasts, whereas downregulation of miRNA-543 significantly enhanced cell growth and inhibited apoptosis. MiRNA-543 upregulation significantly inhibited YAF-2 expression and suppressed the phosphorylation and expression of AKT and p38 mitogen-activated protein kinases (MAPK) in osteoblasts. Furthermore, YAF-2 knockdown enhanced the effects of miRNA-543 on apoptosis in osteoblasts. AKT inhibitor MK2206 and p38 MAPK inhibitor SB203580 also enhanced the effects of miRNA-543 on apoptosis in osteoblasts. Our findings revealed that inhibition of miRNA-543 could protect osteoblasts against ovariectomy-induced osteoporosis through AKT/p38 MAPK signaling pathway by targeting YAF2.
Collapse
Affiliation(s)
- Xiang Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Lei Ning
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangde Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.,Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Shuanglin Wan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
40
|
Down-regulation of miR-543 expression increases the sensitivity of colorectal cancer cells to 5-Fluorouracil through the PTEN/PI3K/AKT pathway. Biosci Rep 2019; 39:BSR20190249. [PMID: 30842340 PMCID: PMC6430726 DOI: 10.1042/bsr20190249] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/04/2019] [Accepted: 03/04/2019] [Indexed: 12/24/2022] Open
Abstract
Resistance to chemotherapy is one of main obstacles in the treatment of colorectal cancer (CRC). However, the mechanisms are still unclear, and the treatment options are still limited. miR-543 has been indicated to act as an oncogene in some cancers, but its function in regulating chemoresistance has not been considered in CRC cells. This study investigated whether the down-regulation of miR-543 expression enhanced 5-fluorouracil (5-FU)-induced apoptosis in HCT8/FU colon cancer cells. In our study, qRT-PCR revealed that miR-543 expression was up-regulated in the HCT8/FU colon cancer cell line compared with that of HCT8 colon cancer cell line. An miR-543 inhibitor or mimic was transfected, followed by MTT assay to detect 5-FU sensitivity in HCT8 and HCT8/FU cell lines, which showed that IC50 of 5-FU was positively correlated with miR-543 expression. Further studies showed that miR-543 enhanced drug resistance by down-regulating the expression of phosphatase and tensin homolog (PTEN), which negatively regulates protein kinase B (AKT) activation. Additionally, an elevated expression of PTEN reversed the chemoresistance of miR-543-overexpressing HCT8 cells to 5-FU. These results indicate that miR-543 might be a target to increase the sensitivity of CRC cells to 5-FU through the PTEN/PI3K/AKT pathway.
Collapse
|
41
|
Gu B, Liu H, Han Y, Chen Y, Jiang H. Integrated analysis of miRNA and mRNA expression profiles in 2-, 6-, and 12-month-old Small Tail Han Sheep ovaries reveals that oar-miR-432 downregulates RPS6KA1 expression. Gene 2019; 710:76-90. [PMID: 30898702 DOI: 10.1016/j.gene.2019.02.095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/23/2019] [Accepted: 02/28/2019] [Indexed: 01/08/2023]
Abstract
Small Tail Han Sheep are an excellent local sheep breed in China, and their outstanding reproductive performance is one of their very important biological characteristics. Clarifying the ovary development process of these ewes should provide a theoretical basis for improving their reproductive efficiency. In this study, we identified the differentially expressed (DE) microRNAs (miRNAs) in 2-, 6-, and 12-month-old small-tail Han sheep ovaries by constructing and analyzing the miRNA expression profiles. These findings clarify the molecular mechanisms regulating the excellent reproductive performance of small-tail Han ewes. We used RNA-Seq technology and bioinformatic to analyze these profiles. Eleven, 13, and 19 DE miRNAs were identified in the 2- vs 6-, 6- vs 12-, and 2- vs 12-month-old ovaries, respectively. In total, 54, 37, and 198 predicted target genes of these DE miRNAs were identified in these three groups, respectively. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that in the 2- vs 6-month-old ovaries, the target genes of DE known sheep miRNAs were involved in 102 GO terms and seven signaling pathways; in the 6- vs 12-month-old ovaries, the target genes of DE known sheep miRNAs were involved in 52 GO terms and three signaling pathways; and in the 2- vs 12-month-old ovaries, the target genes of DE known sheep miRNAs were involved in 88 GO terms and six signaling pathways. Three miRNA-target regulatory networks were constructed based on these DE miRNA-target interactions. Nine miRNAs were selected to confirm to the accuracy of the miRNA sequencing data with qRT-PCR. The site at which oar-miR-432 binds RPS6KA1 was determined with a dual-luciferase system. This is the first integrated analysis the expression profiles of miRNAs and their targets during ovarian development in small-tail Han sheep. These data clarify the molecular regulatory mechanisms underlying sheep ovarian development and identify biomarkers that influence the reproductive performance of small-tail Han ewes.
Collapse
Affiliation(s)
- Bo Gu
- College of Animal Science and Technology, Jinlin Agricultural University, Changchun, Jilin, China
| | - Hang Liu
- College of Animal Science and Technology, Jinlin Agricultural University, Changchun, Jilin, China
| | - Yue Han
- College of Animal Science and Technology, Jinlin Agricultural University, Changchun, Jilin, China
| | - Yang Chen
- College of Animal Science and Technology, Jinlin Agricultural University, Changchun, Jilin, China
| | - Huaizhi Jiang
- College of Animal Science and Technology, Jinlin Agricultural University, Changchun, Jilin, China.
| |
Collapse
|
42
|
Chen F, Zhou H, Wu C, Yan H. Identification of miRNA profiling in prediction of tumor recurrence and progress and bioinformatics analysis for patients with primary esophageal cancer: Study based on TCGA database. Pathol Res Pract 2018; 214:2081-2086. [PMID: 30477645 DOI: 10.1016/j.prp.2018.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 09/13/2018] [Accepted: 10/17/2018] [Indexed: 02/07/2023]
Abstract
OBJECT This study focused on the identification of prognostic miRNAs for the prediction of tumor recurrence and progress in esophageal cancer. METHODS MiRNA profiling and clinical characteristics of esophageal cancer patients was downloaded from the TCGA database. Univariate analysis was performed to select potential prognostic miRNAs and covariates. LASSO based logistic regression was conducted to identify the prognostic miRNAs given covariates. Bioinformatics analysis including gene ontology, disease ontology and pathway enrichment analysis were performed. A nomogram was generated based on multivariate logistic regression to illustrate the association between the identified miRNAs and the risk of tumor recurrence and progress. RESULTS A total of 1881 miRNAs and 10 clinical characteristics were obtained from TCGA database. 18 miRNAs were finally identified in which 6 miRNAs were identified for the first time to be associated with the tumor recurrence and progress of esophageal cancer given covariates. Bioinformatics analysis suggested that the identified miRNAs were associated with the tumor recurrence and progress of esophageal cancer. The association between identified miRNAs and risk of tumor recurrence and progress were presented in a nomogram. CONCLUSION The 6 newly identified miRNAs may be potential biomarkers for the prediction of tumor recurrence and progress of esophageal cancer.
Collapse
Affiliation(s)
- Fangyao Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University Health Science Center, 76 Yanta Xilu Road, Xi'an, Shaanxi, 710061, China
| | - Hui Zhou
- Department of Pharmacy, First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Xilu Road, Xi'an, Shaanxi, 710061, China
| | - Chenqiuzi Wu
- First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Xilu Road, Xi'an, Shaanxi, 710061, China
| | - Hong Yan
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University Health Science Center, 76 Yanta Xilu Road, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
43
|
Yang P, Wu Z, Ma C, Pan N, Wang Y, Yan L. Endometrial miR-543 Is Downregulated During the Implantation Window in Women With Endometriosis-Related Infertility. Reprod Sci 2018; 26:900-908. [PMID: 30231774 DOI: 10.1177/1933719118799199] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Differentially expressed microRNAs (miRNAs) and their target mRNAs may lead to alterations in normal physiological status of the tissues and initiate pathological processes. The aim of this study was to investigate the expression of the most relevant miRNAs in the eutopic endometrial tissue during the window of implantation in women with endometriosis-related infertility. METHODS In the study, 76 infertile women with a regular menstrual cycle were recruited from the Center for Reproductive Medicine, Peking University Third Hospital between January 2014 and June 2016. We performed a combined messenger RNA and miRNA microarray and bioinformatics analysis of eutopic endometrium in 6 women with and without endometriosis-related infertility at the time of implantation window. Quantitative real-time polymerase chain reaction arrays were utilized to examine the expression levels of selected miRNAs (from 35 patients with endometriosis and 35 disease-free individuals at different menstrual stages). RESULTS Five differentially expressed miRNAs (miR-142-5p, miR-146a-5p, miR-1281, miR-940, and miR-4634) were significantly upregulated, whereas miR-543 was significantly downregulated in the eutopic endometrium during the window of implantation in patients with endometriosis. Further analysis showed that miR-543 was significantly upregulated at the peri-implantation phase compared with that at proliferative phase in the endometrium of disease-free patients (P < .05). However, the expression level of miR-543 was significantly decreased in patients with endometriosis (P < .05), especially downregulated at the window of implantation phase (P < .05). CONCLUSIONS miR-543 plays an important role during embryo implantation process and is associated with endometrial receptivity. Downregulation of miR-543 may affect embryo implantation, resulting in the pathogenesis of endometriosis-related infertility.
Collapse
Affiliation(s)
- Puyu Yang
- Center for Reproductive Medicine, Reproductive Medicine Center Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, North Garden Road, No. 49, Beijing, 100191, People's Republic of China
| | - Zhangxin Wu
- Center for Reproductive Medicine, Reproductive Medicine Center Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, North Garden Road, No. 49, Beijing, 100191, People's Republic of China
| | - Caihong Ma
- Center for Reproductive Medicine, Reproductive Medicine Center Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, North Garden Road, No. 49, Beijing, 100191, People's Republic of China.
| | - Ningning Pan
- Center for Reproductive Medicine, Reproductive Medicine Center Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, North Garden Road, No. 49, Beijing, 100191, People's Republic of China
| | - Yang Wang
- Center for Reproductive Medicine, Reproductive Medicine Center Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, North Garden Road, No. 49, Beijing, 100191, People's Republic of China
| | - Liying Yan
- Center for Reproductive Medicine, Reproductive Medicine Center Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, North Garden Road, No. 49, Beijing, 100191, People's Republic of China
| |
Collapse
|
44
|
Xu J, Wang F, Wang X, He Z, Zhu X. miRNA-543 promotes cell migration and invasion by targeting SPOP in gastric cancer. Onco Targets Ther 2018; 11:5075-5082. [PMID: 30174445 PMCID: PMC6110661 DOI: 10.2147/ott.s161316] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background/purpose Given the emerging role of microRNA (miRNA) in cancer progression, we investigated the role and mechanism of miRNA-543 (miR-543) in gastric cancer (GC). Materials and methods Real-time quantitative polymerase chain reaction was conducted to quantify the expression of miR-543. Luciferase reporter assay was used to confirm the association between speckle-type POZ protein (SPOP) and 3′-UTR. Moreover, the role of miR-543 and SPOP in GC was detected using transwell assays. In addition, we investigated the function of miR-543 in the epithelial–mesenchymal transition (EMT) progression. Results miR-543 was upregulated in GC. We identified SPOP as a direct target of miR-543, revealing its expression to be inversely correlated with miR-543 expression in GC tissues. Moreover, restoration of SPOP could inhibit miR-543-induced GC cell migration and invasion, whereas downregulation of miR-543 inhibited cell migration and invasion, which was partly abrogated by SPOP knockdown. Furthermore, our data also showed that miR-543 induced EMT of GC cells. Conclusion Our results demonstrated that miR-543 functions as a crucial oncogenic miRNA in GC. It exerts strong tumor-promoting effects through targeting SPOP in GC cell migration and invasion.
Collapse
Affiliation(s)
- Junfei Xu
- Department of General Surgery, First Affiliated Hospital, Soochow University, Suzhou, People's Republic of China, .,Department of General Surgery, Affiliated Hospital, Nantong University, Nantong, People's Republic of China
| | - Feiran Wang
- Department of General Surgery, Affiliated Hospital, Nantong University, Nantong, People's Republic of China
| | - Xi Wang
- Medical College of Nantong University, Nantong, People's Republic of China
| | - Zhixian He
- Department of General Surgery, Affiliated Hospital, Nantong University, Nantong, People's Republic of China
| | - Xinguo Zhu
- Department of General Surgery, First Affiliated Hospital, Soochow University, Suzhou, People's Republic of China,
| |
Collapse
|
45
|
Li S, Zhang T, Zhou X, Du Z, Chen F, Luo J, Liu Q. The tumor suppressor role of miR-155-5p in gastric cancer. Oncol Lett 2018; 16:2709-2714. [PMID: 30008945 DOI: 10.3892/ol.2018.8932] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 04/24/2018] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is the fifth most common type of malignant tumor worldwide and the most common cause of cancer-associated mortality in China. Recent studies revealed that microRNAs (miRNAs) function in the pathogenesis of GC, and that miR-155-5p expression is downregulated in GC tissues. However, the function of miR-155-5p has not been fully identified. In the present study, it was demonstrated that overexpression of miR-155-5p inhibited GC-cell proliferation and promoted apoptosis, while downregulation of miR-155-5p promoted GC-cell proliferation and decreased the cisplatin sensitivity of GC cells. Mitogen-activated protein kinase kinase kinase 10 was demonstrated to be a potential target gene of miR-155-5p. In conclusion, an antitumor role of miR-155-5p in gastric cancer was suggested.
Collapse
Affiliation(s)
- Shiqing Li
- Department of Gastroenterology, Center Hospital of Nanchong City, The Second Clinical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Tao Zhang
- Department of Gastroenterology, Center Hospital of Nanchong City, The Second Clinical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xiaoqing Zhou
- Department of Gastroenterology, Center Hospital of Nanchong City, The Second Clinical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Zonghan Du
- Department of Gastroenterology, Center Hospital of Nanchong City, The Second Clinical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Fumin Chen
- Department of Gastroenterology, Center Hospital of Nanchong City, The Second Clinical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Jun Luo
- Department of Gastroenterology, Center Hospital of Nanchong City, The Second Clinical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Qingsong Liu
- Clinical Laboratory Department, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
46
|
Ge Y, Li J, Hao Y, Hu Y, Chen D, Wu B, Fang F. MicroRNA-543 functions as an osteogenesis promoter in human periodontal ligament-derived stem cells by inhibiting transducer of ERBB2, 2. J Periodontal Res 2018; 53:832-841. [PMID: 29851072 DOI: 10.1111/jre.12572] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Previous research has indicated that altered expression of microRNAs (miRNAs) is in connection with osteogenesis of human periodontal ligament-derived stem cells (hPDLSCs). We investigated the mechanisms by which miR-543 promotes osteogenic differentiation of hPDLSCs. MATERIAL AND METHODS First, the expression of miR-543 in hPDLSCs cultured with or without an osteogenic inductive cocktail was explored. Then, the function of miR-543 during osteogenesis of hPDLSCs was investigated by overexpressing and inhibiting miR-543. Next, 3 databases were used to predict target genes of miR-543 and a luciferase report was used to validate the direct regulation of miR-543 on the target gene. Further, a rescue experiment using co-transfection of miR-543 mimic and target mimic was performed to evaluate whether overexpressing the target gene could partly rescue the efficiency of overexpressing miR-543 on osteogenesis in hPDLSCs. RESULTS miR-543 was upregulated during osteogenic differentiation of hPDLSCs. Functional experiments showed that overexpressing miR-543 could enhance osteogenesis, while inhibiting miR-543 resulted in reduced formation of mineralized nodules. The transducer of ERBB2, 2 (TOB2) was identified as a target gene of miR-543 and luciferase report revealed that miR-543 interacts directly with the 3'-untranslated repeat sequence of TOB2 mRNA. Overexpression of miR-543 inhibited the expression of TOB2 in both mRNA and protein levels while inhibiting miR-543 increased. Furthermore, the rescue experiment confirmed the promotional role of miR-543 TOB2 expression could be abrogated by overexpressing TOB2, which also had the effect of reducing osteogenic differentiation. CONCLUSION Our research confirmed that miR-543 is a promoter of osteogenesis in hPDLSCs, acting by inhibiting its target gene TOB2, which suggests that miR-543 may be a potential therapy for bone loss in periodontitis.
Collapse
Affiliation(s)
- Y Ge
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - J Li
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Y Hao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Y Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - D Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - B Wu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - F Fang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Zhang H, Guo X, Feng X, Wang T, Hu Z, Que X, Tian Q, Zhu T, Guo G, Huang W, Li X. MiRNA-543 promotes osteosarcoma cell proliferation and glycolysis by partially suppressing PRMT9 and stabilizing HIF-1α protein. Oncotarget 2018; 8:2342-2355. [PMID: 27911265 PMCID: PMC5356804 DOI: 10.18632/oncotarget.13672] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 11/06/2016] [Indexed: 01/17/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumor, occurring frequently in adolescents and possessing a high malignant severity. MicroRNAs play critical roles during OS development. Thus, elucidation of the involvement of specific microRNAs in the development of OS may provide novel therapeutic targets for OS treatment. Here, we showed that in the OS specimens from patients, the levels of miR-543 were significantly increased whereas the levels of PRMT9 were significantly decreased, compared to the paired normal bone tissue. Moreover, miR-543 and PRMT9 inversely correlated in the OS cell lines. Bioinformatics analyses predicted that miR-543 may target the 3'-UTR of PRMT9 mRNA to inhibit its translation, which was confirmed by luciferase-reporter assay. MiR-543 promoted OS cell proliferation in vitro and in vivo. Mechanistically, miR-543 inhibited PRMT9-enhanced cell oxidative phosphorylation, while miR-543 depletion promoted PRMT9-increased HIF-1α instability and inhibited glycolysis in OS cells. Clinically, miR-543 expression was negatively correlated with PRMT9 expression in OS tissues. Together, our data provide important evidence for glycolysis in OS development, and suggest that targeting glycolytic pathway through miR-543/PRMT9/HIF-1α axis may represent a potential therapeutic strategy to eradicate OS cells.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Orthopedics, Renhe Hospital, China Three Gorges University, Yichang, Hubei, China
| | - Xiaofeng Guo
- Medical College of Xiamen University, Xiamen, Fujian, China
| | - Xing Feng
- Department of Molecular Pharmacology, Rutgers University, New Brunswick, New Jersey, USA
| | - Tingting Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Zhaohua Hu
- Department of Orthopedics, Renhe Hospital, China Three Gorges University, Yichang, Hubei, China
| | - Xiangyong Que
- Department of Orthopedics, Renhe Hospital, China Three Gorges University, Yichang, Hubei, China
| | - Qingsong Tian
- Department of Medicine, Medical College, China Three Gorges University, Yichang, Hubei, China
| | - Tianbo Zhu
- Department of Medicine, Medical College, China Three Gorges University, Yichang, Hubei, China
| | - Guixian Guo
- Department of Medicine, The Second Hospital Affiliated to Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Huang
- Department of Orthopedics, Renhe Hospital, China Three Gorges University, Yichang, Hubei, China
| | - Xinzhi Li
- Department of Orthopedics, Renhe Hospital, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
48
|
Yu X, Ma C, Fu L, Dong J, Ying J. MicroRNA-139 inhibits the proliferation, migration and invasion of gastric cancer cells by directly targeting ρ-associated protein kinase 1. Oncol Lett 2018; 15:5977-5982. [PMID: 29552227 DOI: 10.3892/ol.2018.8038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 10/24/2017] [Indexed: 12/18/2022] Open
Abstract
The expression, function and underlying mechanisms of microRNA-139 (miR-139) in gastric cancer were investigated in the present study. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed to detect miR-139 expression in gastric cancer tissues and cell lines. The effects of miR-139 overexpression on gastric cancer cell proliferation, migration and invasion were evaluated. ρ-associated protein kinase 1 (ROCK1) was predicted as a downstream target of miR-139 and its role in gastric cancer was assessed by bioinformatics analysis, luciferase reporter assay, RT-qPCR and western blot analysis. ROCK1 overexpression was established to investigate if the effects of miR-139 on gastric cancer cells may be attenuated. The results indicated that miR-139 was aberrantly downregulated in gastric cancer tissues and cell lines. Increased miR-139 expression reduced gastric cancer cell proliferation, migration and invasion. ROCK1 was demonstrated to be a direct target of miR-139 in gastric cancer and ROCK1 overexpression reversed the suppressive effects on gastric cancer cell proliferation, migration and invasion induced by miR-139 overexpression. The present study provides clear evidence demonstrating the anti-oncogenic activity of miR-139 in human gastric cancer, as mediated by the targeted downregulation of ROCK1.
Collapse
Affiliation(s)
- Xuechun Yu
- Department of Gastroenterology, People's Hospital of Xuyi, Huai'an, Jiangsu 211700, P.R. China
| | - Chaojian Ma
- Department of Gastroenterology, People's Hospital of Xuyi, Huai'an, Jiangsu 211700, P.R. China
| | - Ling Fu
- Department of Gastroenterology, People's Hospital of Xuyi, Huai'an, Jiangsu 211700, P.R. China
| | - Jingwu Dong
- Department of Gastroenterology, People's Hospital of Xuyi, Huai'an, Jiangsu 211700, P.R. China
| | - Jie Ying
- Department of Infectious Diseases, People's Hospital of Xuyi, Huai'an, Jiangsu 211700, P.R. China
| |
Collapse
|
49
|
SIRT1 activator isolated from artificial gastric juice incubate of total saponins in stems and leaves of Panax ginseng. Bioorg Med Chem Lett 2018; 28:240-243. [DOI: 10.1016/j.bmcl.2017.12.067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/27/2017] [Accepted: 12/29/2017] [Indexed: 11/21/2022]
|
50
|
Zhang L, Guo X, Zhang L, Yang F, Qin L, Zhang D, Qin Y. SLC34A2 regulates miR-25-Gsk3β signaling pathway to affect tumor progression in gastric cancer stem cell-like cells. Mol Carcinog 2017; 57:440-450. [PMID: 29178392 DOI: 10.1002/mc.22768] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 11/20/2017] [Indexed: 12/18/2022]
Abstract
A novel paradigm in tumor biology suggests that gastric cancer progression is driven by gastric cancer stem cell-like cells (GCSCs), but molecular mechanisms regulating tumorigenic and self-renewal potential of GCSCs are still unclear. Here, we aim to investigate biological function of SLC34A2 in regulating sphere formation and tumorigenicity (both are the hallmark of CSCs) of GCSCs and its underlying mechanisms. Our findings testified that CD44+ cells which were derived from fresh primary gastric cancer samples and cell lines displayed stem cell-like features. Significantly, SLC34A2 is increased in CD44+ GCSCs compared with those in adherent counterpart from CD44+ GCSCs. On clinic, SLC34A2 is overexpressed in primary tumor tissues compared with adjacent counterparts. We showed that SLC34A2 regulated sphere formation and self-renewal properties of CD44+ GCSCs in vitro and in vivo. Mechanistic investigations revealed that Gsk3β was the most strikingly up-regulated gene in response to SLC34A2 knockdown in GCSCs and Wnt/β-cantenin signaling was required for SLC34A2-mediated sphere formation. Furthermore, SLC34A2 directly binds specific sites in the miR-25 promoter region and that the promoter activity is decreased after the mutation of putative SLC34A2-binding sites, indicating that SLC34A2 is required for the transcriptional induction of miR-25. Meanwhile, luciferase assays showed that miR-25 directly targeted Gsk3β in CD44+ GCSCs. Overall, our findings define a SLC34A2-miR-25-Gsk3β pathway in the regulation of GCSCs features and gastric cancer progression, with potential therapeutic applications in blocking their progression.
Collapse
Affiliation(s)
- Lanfang Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, People's Republic of China
| | - Xiaohe Guo
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, People's Republic of China
| | - Lili Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, People's Republic of China
| | - Fang Yang
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, People's Republic of China
| | - Lei Qin
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, People's Republic of China
| | - Dezhong Zhang
- Department of Gastroenterological Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, People's Republic of China
| | - Yongmei Qin
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, People's Republic of China
| |
Collapse
|