1
|
Zareifar P, Ahmed HM, Ghaderi P, Farahmand Y, Rahnama N, Esbati R, Moradi A, Yazdani O, Sadeghipour Y. miR-142-3p/5p role in cancer: From epigenetic regulation to immunomodulation. Cell Biochem Funct 2024; 42:e3931. [PMID: 38379239 DOI: 10.1002/cbf.3931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/01/2024] [Accepted: 01/08/2024] [Indexed: 02/22/2024]
Abstract
MicroRNAs (miRNAs) play critical roles in cancer pathobiology, acting as regulators of gene expression and pivotal drivers of tumorigenesis. It is believed that miRNAs act through canonical mechanisms, involving the binding of mature miRNAs to target messenger RNAs (mRNAs) and subsequent repression of protein translation or degradation of target mRNAs. miR-142-3p/5p has been extensively studied and established as a key regulator in various malignancies. Recent discoveries have revealed miR-142-3p/5p serve as either oncogene or tumor suppressor in cancer. By targeting epigenetic factor and cancer-related signaling pathway, miR-142-3p/5p can regulate wide range of downstream genes. The immune modulatory role of miR-142-3p/5p has been shown in various cancers, which provides significant insight into immunosuppression and tumor escape from the immune response. Exosomes with miR-142-3p/5p facilitate cell communication and can affect cancer cell behavior, offering potential therapeutic, and diagnosis applications in cancer therapy. In this review, for the first time, we comprehensively summarize the current knowledge regarding mentioned functions of miR-142-3p/5p in cancer pathobiology.
Collapse
Affiliation(s)
- Parisa Zareifar
- Golestan University of Medical Science, Gorgan, Golestan, Iran
| | | | - Pouya Ghaderi
- Department of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Yalda Farahmand
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Negin Rahnama
- Department of Internal Medicine and Health Services, Semnan University of Medical Sciences, Semnan, Iran
| | - Romina Esbati
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Ali Moradi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Yazdani
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Yasin Sadeghipour
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Hussen BM, Saleem SJ, Abdullah SR, Mohamadtahr S, Hidayat HJ, Rasul MF, Taheri M, Kiani A. Current landscape of miRNAs and TGF-β signaling in lung cancer progression and therapeutic targets. Mol Cell Probes 2023; 72:101929. [PMID: 37683829 DOI: 10.1016/j.mcp.2023.101929] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
Lung cancer (LC) is the primary reason for cancer-associated fatalities globally. Due to both tumor-suppressing and tumor-promoting activities, the TGF-β family of growth factors is extremely essential to tumorigenesis. A non-coding single-stranded short RNA called microRNA (miRNA), which is made up of about 22 nt and is encoded by endogenous genes, can control normal and pathological pathways in various kinds of cancer, including LC. Recent research demonstrated that the TGF-β signaling directly can affect the synthesis of miRNAs through suppressor of mothers against decapentaplegic (SMAD)-dependent activity or other unidentified pathways, which could generate allostatic feedback as a result of TGF-β signaling stimulation and ultimately affect the destiny of cancer tissues. In this review, we emphasize the critical functions of miRNAs in lung cancer progression and, more critically, how they affect the TGF-β signaling pathway, and explore the role of both the TGF-β signaling pathway and miRNAs as potential therapeutic targets for improving the treatments of LC patients.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq; Department of Biomedical Sciences, Cihan University-Erbil, Erbil, Kurdistan Region, 44001, Iraq
| | - Safeen Jasim Saleem
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Snur Rasool Abdullah
- Medical Laboratory Science, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Sayran Mohamadtahr
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University-Erbil, Kurdistan Region, Iraq
| | - Mohammed Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany; Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arda Kiani
- Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Lung Research and Developmental Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Duan B, Zhang H, Zhu Z, Yan X, Ji Z, Li J. LncRNA LINC01871 sponging miR-142-3p to modulate ZYG11B promotes the chemoresistance of colorectal cancer cells by inducing autophagy. Anticancer Drugs 2023; 34:827-836. [PMID: 36847071 PMCID: PMC10344439 DOI: 10.1097/cad.0000000000001478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 08/22/2022] [Indexed: 03/01/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a malignant tumor in the digestive tract. Increasing evidence indicated that chemoresistance leads to a poor prognosis of CRC. Herein, we aimed to uncover the potential mechanism by which long intergenic noncoding RNA-1871 (LINC01871) affects the chemoresistance of CRC cells. METHODS Relative level of LINC01871 in CRC tissues was assessed by reverse transcription quantitative PCR (RT-qPCR). Kaplan-Meier analysis was conducted to determine the relevance of LINC01871 and the prognosis of CRC patients. Cell Counting Kit-8 (CCK-8) and colony formation assay were used to evaluate the proliferation of SW480 cells. Expression levels of proteins and their genes were assessed by western blot, immunofluorescence staining and RT-qPCR. In addition, the interaction of LINC01871, miR-142-3p and protein zyg-11 homolog B (ZYG11B) were analyzed via dual-luciferase reporter assays. RESULTS LINC01871 was low-expressed in CRC tissues and cell lines. Patients with a low level of LINC01871 showed significantly lower survival rate. pcDNA-LINC01871 significantly reduced the viability of SW480 cells ( P < 0.01), elevated SW480 cells sensitivity to 5-FU ( P < 0.01), reduced LC3 punctate aggregates ( P < 0.01) and downregulated the relative mRNA expression level of autophagy related protein 9A, autophagy related protein 4B and high mobility group box 1 ( P < 0.01) in SW480 cells. Moreover, LINC01871 was found to sponge miR-142-3p, and ZYG11B was the target of miR-142-3p. MiR-142-3p mimic significantly recovered the effect of pcDNA-LINC001871, whereas pcDNA-ZYG11B reversed the recovery effect of the miR-142-3p mimic. CONCLUSION LINC01871/miR-142-3p/ ZYG11B axis regulates the chemoresistance of CRCs by inducing autophagy.
Collapse
Affiliation(s)
- Bensong Duan
- Department of Gastroenterology, Endoscopy Center
| | - Haibin Zhang
- Department of Gastroenterology, Endoscopy Center
| | | | - Xiaohan Yan
- Department of Gastroenterology, Endoscopy Center
| | - Zhonghua Ji
- Department of Anesthesia, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jingze Li
- Department of Gastroenterology, Endoscopy Center
| |
Collapse
|
4
|
Zhang X, Li X, Wang C, Wang S, Zhuang Y, Liu B, Lian X. Identification of markers for predicting prognosis and endocrine metabolism in nasopharyngeal carcinoma by miRNA-mRNA network mining and machine learning. Front Endocrinol (Lausanne) 2023; 14:1174911. [PMID: 37538797 PMCID: PMC10396331 DOI: 10.3389/fendo.2023.1174911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/21/2023] [Indexed: 08/05/2023] Open
Abstract
Background Nasopharyngeal cancer (NPC) has a high incidence in Southern China and Asia, and its survival is extremely poor in advanced patients. MiRNAs play critical roles in regulating gene expression and serve as therapeutic targets in cancer. This study sought to disclose key miRNAs and target genes responsible for NPC prognosis and endocrine metabolism. Materials and methods Three datasets (GSE32960, GSE70970, and GSE102349) of NPC samples came from Gene Expression Omnibus (GEO). Limma and WGCNA were applied to identify key prognostic miRNAs. There were 12 types of miRNA tools implemented to study potential target genes (mRNAs) of miRNAs. Univariate Cox regression and stepAIC were introduced to construct risk models. Pearson analysis was conducted to analyze the correlation between endocrine metabolism and RiskScore. Single-sample gene set enrichment analysis (ssGSEA), MCP-counter, and ESTIMATE were performed for immune analysis. The response to immunotherapy was predicted by TIDE and SubMap analyses. Results Two key miRNAs (miR-142-3p and miR-93) were closely involved in NPC prognosis. The expression of the two miRNAs was dysregulated in NPC cell lines. A total of 125 potential target genes of the key miRNAs were screened, and they were enriched in autophagy and mitophagy pathways. Five target genes (E2F1, KCNJ8, SUCO, HECTD1, and KIF23) were identified to construct a prognostic model, which was used to divide patients into high group and low group. RiskScore was negatively correlated with most endocrine-related genes and pathways. The low-risk group manifested higher immune infiltration, anticancer response, more activated immune-related pathways, and higher response to immunotherapy than the high-risk group. Conclusions This study revealed two key miRNAs that were highly contributable to NPC prognosis. We delineated the specific links between key miRNAs and prognostic mRNAs with miRNA-mRNA networks. The effectiveness of the five-gene model in predicting NPC prognosis as well as endocrine metabolism provided a guidance for personalized immunotherapy in NPC patients.
Collapse
Affiliation(s)
- Xixia Zhang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao Li
- Department Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Caixia Wang
- Department Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuang Wang
- Department Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuan Zhuang
- Department Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bing Liu
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xin Lian
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Jiang Y, Liu Y, Zhang Y, Ouyang J, Feng Y, Li S, Wang J, Zhang C, Tan L, Zhong J, Zou L. MicroRNA-142-3P suppresses the progression of papillary thyroid carcinoma by targeting FN1 and inactivating FAK/ERK/PI3K signaling. Cell Signal 2023:110792. [PMID: 37406787 DOI: 10.1016/j.cellsig.2023.110792] [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: 04/24/2023] [Revised: 06/25/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
OBJECTIVES miR-142-3P is a tumor suppressor in various malignant cancers. However, the function of miR-142-3P in papillary thyroid carcinoma (PTC) remains to be elucidated. The aim of this study was to explore the function and mechanism of miR-142-3P in PTC. METHODS Real Time Quantitative PCR (RT-qPCR) was used to assess the expression of miR-142-3P and Fibronectin 1 (FN1) in PTC. The correlation between FN1 and miR-142-3P expression was analyzed by Spearman's correlation analysis. Cell Counting Kit 8 (CCK8), 5-ethynyl-2'-deoxyuridine (EDU) assay, cell migration and invasion assay and wound healing measures evaluated the effect of miR-142-3P and FN1 on cell proliferation, migration and invasion. Dural Luciferase reported gene assay evaluated the interaction between miR-142-3P and 3' untranslated region (UTR) of FN1. The Epithelial-Mesenchymal-Transition (EMT) and apoptosis related marker genes were measured using western blot analysis (WB). RESULTS miR-142-3P was significantly decreased in both PTC specimens and relevant cell lines. Functionally, miR-142-3P inhibited cell proliferation, migration, invasion and EMT, and induced the cell apoptosis in PTC. In addition, miR-142-3P bound directly with 3' UTR of FN1 and negatively regulated the expression of FN1 in PTC. FN1 expression is elevated in PTC, and its aberrant high correlated with declines in recurrence-free survival (RFS). Moreover, FN1 promoted cell proliferation, migration, invasion and EMT, induced cell apoptosis in PTC cells. Depletion of FN1 rescues the effect of miR-142-3P inhibitor on cell proliferation, invasion, apoptosis and EMT via inactivating Focal Adhesion Kinase (FAK)/Extracellular Signal-Regulated Kinase (ERK) / Phosphoinostide 3-kinase (P13K) signaling. CONCLUSION miR-142-3P suppressed cell proliferation, migration, invasion and EMT through modulating FN1/FAK/ERK/PI3K signaling in PTC, suggesting it as a potential therapeutic target for PTC.
Collapse
Affiliation(s)
- Yufei Jiang
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan Province 410005, People's Republic of China; Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China; Aculty of Healty Science, University of Macau, Macau 999078, People's Republic of China
| | - Yarong Liu
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan Province 410005, People's Republic of China; Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Yiyuan Zhang
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Jielin Ouyang
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan Province 410005, People's Republic of China; Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Yang Feng
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan Province 410005, People's Republic of China; Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Shumei Li
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan Province 410005, People's Republic of China; Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Jingjing Wang
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Chaojie Zhang
- Department of Papillary Thyroid Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China
| | - Lihong Tan
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China.
| | - Jie Zhong
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China.
| | - Lianhong Zou
- Institute of Clinical and Translational Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province 410005, People's Republic of China.
| |
Collapse
|
6
|
Gu Y, Becker MA, Müller L, Reuss K, Umlauf F, Tang T, Menger MD, Laschke MW. MicroRNAs in Tumor Endothelial Cells: Regulation, Function and Therapeutic Applications. Cells 2023; 12:1692. [PMID: 37443725 PMCID: PMC10340284 DOI: 10.3390/cells12131692] [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: 05/18/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Tumor endothelial cells (TECs) are key stromal components of the tumor microenvironment, and are essential for tumor angiogenesis, growth and metastasis. Accumulating evidence has shown that small single-stranded non-coding microRNAs (miRNAs) act as powerful endogenous regulators of TEC function and blood vessel formation. This systematic review provides an up-to-date overview of these endothelial miRNAs. Their expression is mainly regulated by hypoxia, pro-angiogenic factors, gap junctions and extracellular vesicles, as well as long non-coding RNAs and circular RNAs. In preclinical studies, they have been shown to modulate diverse fundamental angiogenesis-related signaling pathways and proteins, including the vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) pathway; the rat sarcoma virus (Ras)/rapidly accelerated fibrosarcoma (Raf)/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway; the phosphoinositide 3-kinase (PI3K)/AKT pathway; and the transforming growth factor (TGF)-β/TGF-β receptor (TGFBR) pathway, as well as krüppel-like factors (KLFs), suppressor of cytokine signaling (SOCS) and metalloproteinases (MMPs). Accordingly, endothelial miRNAs represent promising targets for future anti-angiogenic cancer therapy. To achieve this, it will be necessary to further unravel the regulatory and functional networks of endothelial miRNAs and to develop safe and efficient TEC-specific miRNA delivery technologies.
Collapse
Affiliation(s)
- Yuan Gu
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Saar, Germany; (M.A.B.); (L.M.); (K.R.); (F.U.); (T.T.); (M.D.M.); (M.W.L.)
| | | | | | | | | | | | | | | |
Collapse
|
7
|
TIF1γ inhibits lung adenocarcinoma EMT and metastasis by interacting with the TAF15/TBP complex. Cell Rep 2022; 41:111513. [DOI: 10.1016/j.celrep.2022.111513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/24/2022] [Accepted: 09/26/2022] [Indexed: 11/22/2022] Open
|
8
|
Najafipour R, Mohammadi D, Estaki Z, Zarabadi K, Jalilvand M, Moghbelinejad S. Screening for differentially expressed microRNAs in BALF and blood samples of infected COVID-19 ARDS patients by small RNA deep sequencing. J Clin Lab Anal 2022; 36:e24672. [PMID: 36166345 PMCID: PMC9539155 DOI: 10.1002/jcla.24672] [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: 01/08/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 01/08/2023] Open
Abstract
Background The pandemic COVID‐19 has caused a high mortality rate and poses a significant threat to the population of the entire world. Due to the novelty of this disease, the pathogenic mechanism of the disease and the host cell's response are not yet fully known, so lack of evidence prevents a definitive conclusion about treatment strategies. The current study employed a small RNA deep‐sequencing approach for screening differentially expressed microRNA (miRNA) in blood and bronchoalveolar fluid (BALF) samples of acute respiratory distress syndrome (ARDS) patients. Methods In this study, BALF and blood samples were taken from patients with ARDS (n = 5). Control samples were those with suspected lung cancer candidates for lung biopsy (n = 3). Illumina high‐throughput (HiSeq 2000) sequencing was performed to identify known and novel miRNAs differentially expressed in the blood and BALFs of ARDS patients compared with controls. Results Results showed 2234 and 8324 miRNAs were differentially expressed in blood and BALF samples, respectively. In BALF samples, miR‐282, miR‐15‐5p, miR‐4485‐3p, miR‐483‐3p, miR‐6891‐5p, miR‐200c, miR‐4463, miR‐483‐5p, and miR‐98‐5p were upregulated and miR‐15a‐5p, miR‐548c‐5p, miR‐548d‐3p, miR‐365a‐3p, miR‐3939, miR‐514‐b‐5p, miR‐513a‐3p, miR‐513a‐5p, miR‐664a‐3p, and miR‐766‐3p were downregulated. On the contrary, in blood samples miR‐15b‐5p, miR‐18a‐3p, miR‐486‐3p, miR‐486‐5p, miR‐146a‐5p, miR‐16‐2‐3p, miR‐6501‐5p, miR‐365‐3p, miR‐618, and miR‐623 were top upregulated miRNAs and miR‐21‐5p, miR‐142a‐3p, miR‐181‐a, miR‐31‐5p, miR‐99‐5p, miR‐342‐5p, miR‐183‐5p, miR‐627‐5p, and miR‐144‐3p were downregulated miRNAs. Network functional analysis for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), in ARDS patients' blood and BALF samples, showed that the target genes were more involved in activating inflammatory and apoptosis process. Conclusion Based on our results, the transcriptome profile of ARDS patients would be a valuable source for understanding molecular mechanisms of host response and developing clinical guidance on anti‐inflammatory medication.
Collapse
Affiliation(s)
- Reza Najafipour
- Genetics Research Center, The University of Social Welfare and Rehabilitation Science, Tehran, Iran
| | - Davood Mohammadi
- Department of Surgery, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Zohreh Estaki
- Department of Pediatric Dentistry, School of Dentistry, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kiana Zarabadi
- Department of Medical Genetics, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Manijeh Jalilvand
- Research Institute for Prevention of Non-Communicable Diseases, Cellular and Molecular Research Centre, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sahar Moghbelinejad
- Research Institute for Prevention of Non-Communicable Diseases, Cellular and Molecular Research Centre, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
9
|
Plousiou M, De Vita A, Miserocchi G, Bandini E, Vannini I, Melloni M, Masalu N, Fabbri F, Serra P. Growth Inhibition of Retinoblastoma Cell Line by Exosome-Mediated Transfer of miR-142-3p. Cancer Manag Res 2022; 14:2119-2131. [PMID: 35791342 PMCID: PMC9250773 DOI: 10.2147/cmar.s351979] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 06/09/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Retinoblastoma (Rb) is the most common ocular paediatric malignancy and is caused by a mutation of the two alleles of the tumor suppressor gene, RB1. The tumor microenvironment (TME) represents a complex system whose function is not yet well defined and where microvesicles, such as exosomes, play a key role in intercellular communication. Micro-RNAs (mRNAs) have emerged as important modifiers of biological mechanisms involved in cancer and been able to regulate tumor progression. Methods Co-culture of monocytes with retinoblastoma cell lines, showed a significant growth decrease. Given the interaction between Rb cells and monocytes, we investigated the role of the supernatant in the cross-talk between cell lines, by taking the product of the co-culture and then using it as a culture medium for Rb cells. Results miR-142-3p showed to be particularly over-expressed both in the Rb cell line and in the medium used for their culture, comparing to control cell line and the normal supernatant, respectively. Therefore, we provided evidence that miR-142-3p is released by monocytes in the co-culture medium’s exosomes and that it is subsequently up-taken by Rb cells, causing the inhibition of proliferation of Rb cell line by affecting cell cycle progression. Conclusion This study highlights the role of exosomic miR-142-3p in the TME of Rb and identifies new molecular targets, which are able to control tumor growth aiming the development of a forward-looking miR-based strategy.
Collapse
Affiliation(s)
- Meropi Plousiou
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Alessandro De Vita
- Osteoncology Unit, Bioscience Laboratory IRCCS Istituto Romagnolo Per lo Studio dei Tumori (IRST), "Dino Amadori", 47014 Meldola, Italy
| | - Giacomo Miserocchi
- Osteoncology Unit, Bioscience Laboratory IRCCS Istituto Romagnolo Per lo Studio dei Tumori (IRST), "Dino Amadori", 47014 Meldola, Italy
| | - Erika Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Ivan Vannini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Mattia Melloni
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Nestory Masalu
- Unit of Biostatistics and Clinical Trials, Bioscience Laboratory IRCCS Istituto Romagnolo Per lo Studio dei Tumori (IRST), "Dino Amadori", 47014 Meldola, Italy
| | - Francesco Fabbri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Patrizia Serra
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Scientifico Romagnolo Per lo Studio dei Tumori (IRST), "Dino Amadori", Meldola, Italy
| |
Collapse
|
10
|
Fan C, Xiong F, Tang Y, Li P, Zhu K, Mo Y, Wang Y, Zhang S, Gong Z, Liao Q, Li G, Zeng Z, Guo C, Xiong W, Huang H. Construction of a lncRNA–mRNA Co-Expression Network for Nasopharyngeal Carcinoma. Front Oncol 2022; 12:809760. [PMID: 35875165 PMCID: PMC9302896 DOI: 10.3389/fonc.2022.809760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 06/06/2022] [Indexed: 11/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) widely regulate gene expression and play important roles in the pathogenesis of human diseases, including malignant tumors. However, the functions of most lncRNAs remain to be elucidated. In order to study and screen novel lncRNAs with important functions in the carcinogenesis of nasopharyngeal carcinoma (NPC), we constructed a lncRNA expression profile of 10 NPC tissues and 6 controls through a gene microarray. We identified 1,276 lncRNAs, of which most are unknown, with different expression levels in the healthy and NPC tissues. In order to shed light on the functions of these unknown lncRNAs, we first constructed a co-expression network of lncRNAs and mRNAs using bioinformatics and systematic biological approach. Moreover, mRNAs were clustered and enriched by their biological functions, and those lncRNAs have similar expression trends with mRNAs were defined as functional molecules with potential biological significance. The module may help identify key lncRNAs in the carcinogenesis of NPC and provide clues for in-depth study of their functions and associated signaling pathways. We suggest the newly identified lncRNAs may have clinic value as biomarkers and therapeutic targets for NPC diagnosis and treatment.
Collapse
Affiliation(s)
- Chunmei Fan
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Fang Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanyan Tang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Panchun Li
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Kunjie Zhu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yongzhen Mo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Yumin Wang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Shanshan Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaojiang Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Can Guo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
- *Correspondence: Wei Xiong, ; He Huang,
| | - He Huang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
- *Correspondence: Wei Xiong, ; He Huang,
| |
Collapse
|
11
|
Wan X, Yan Z, Tan Z, Cai Z, Qi Y, Lu L, Xu Y, Chen J, Lei T. MicroRNAs in Dopamine Agonist-Resistant Prolactinoma. Neuroendocrinology 2022; 112:417-426. [PMID: 34034260 DOI: 10.1159/000517356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/23/2021] [Indexed: 11/19/2022]
Abstract
Dopamine agonists (DAs) are preferred for the treatment of prolactinomas and are usually very effective. Nonetheless, 20-30% of bromocriptine- and approximately 10% of cabergoline-treated individuals exhibit resistance to DAs. In addition, the mechanism underlying this phenomenon remains elusive. In this study, we summarize the major findings regarding the role of microRNAs (miRNAs) in the pathogenesis of DA-resistant prolactinoma (DARP). Currently available evidence suggests that miRNAs are usually dysregulated in DARP and that, although controversial, the dysregulated miRNAs target the transforming growth factor (TGF)-β, dopamine 2 receptor (D2R), or estradiol (E2)/estrogen receptor (ER) signaling pathways to mediate the therapeutic effect of DAs. These findings provide new incentives for research on innovative strategies for predicting patients' responsiveness to dopamine therapies and for developing treatment approaches. Unfortunately, recent studies tended to focus exclusively on the differential miRNA expression profiles between DARP and dopamine-sensitive prolactinoma, and no definitive consensus has been reached regarding the role of these miRNAs in the modulation mechanism. Therefore, current and future efforts should be directed toward the exploration of the mechanism underlying the dysregulation of miRNAs as well as of the target proteins that are affected by the dysregulated miRNAs. Furthermore, the modulation of the expression of dysregulated miRNAs, which target the D2R, TGF-β, or E2/ER signaling pathways, might be a promising alternative to treat patients with DARP and improve their prognosis.
Collapse
Affiliation(s)
- Xueyan Wan
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zisheng Yan
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhoubin Tan
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Cai
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiwei Qi
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Lu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Xu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Wang B, Ji D, Xing W, Li F, Huang Z, Zheng W, Xue J, Zhu Y, Yang X. miR-142-3p and HMGB1 Are Negatively Regulated in Proliferation, Apoptosis, Migration, and Autophagy of Cartilage Endplate Cells. Cartilage 2021; 13:592S-603S. [PMID: 33955243 PMCID: PMC8804737 DOI: 10.1177/19476035211012444] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cartilage endplate (CEP) degeneration plays a vital role in the pathological process of intervertebral disc degeneration. It has been previously reported that microRNAs may participate in the occurrence and development of intervertebral disc degeneration through regulating its target genes directly. The regulatory roles of miR-142-3p/HMGB1 in some orthopedic diseases have been determined successively, but there was no report about the degeneration of CEP. Therefore, we aimed to determine the regulation of miR-142-3p/HMGB1 or potential molecular mechanisms on proliferation, apoptosis, migration, and autophagy of CEP cells. METHODS The target gene of miR-142-3p was determined by double luciferase assay. We selected ATDC5 cell lines. CCK-8 method was used to detect cell proliferation. Real-time fluorescence quantitative polymerase chain reaction was used to determine gene expression levels, and western blot analysis was used to determine protein expression levels. We chose flow cytometry to measure cell apoptosis and cell cycle. RESULTS The result of luciferase detection showed that the target gene of miR-142-3p in CEP cells was HMGB1. Knockdown of the miR-142-3p inhibited the expression level of HMGB1, the proliferation and migration of CEP cells, but it promoted apoptosis of CEP cells. In addition, the detection results of the proteins related to apoptosis or autophagy showed that knockdown of miR-142-3p promoted apoptosis and autophagy. CONCLUSION The negative regulation of miR-142-3p/HMGB1 can affect the proliferation, apoptosis, migration, and autophagy of CEP cells. Our results provide a new idea for the targeted treatment of CEP degeneration by inhibiting the expression of HMGB1.
Collapse
Affiliation(s)
- Bo Wang
- School of Graduate, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.,Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Demin Ji
- School of Graduate, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.,Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Wenhua Xing
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Feng Li
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Zhi Huang
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Wenkai Zheng
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Jianmin Xue
- School of Graduate, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.,Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yong Zhu
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xuejun Yang
- Surgical Department of Thoracolumbar, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
13
|
Identification of Prognostic and Chemopredictive microRNAs for Non-Small-Cell Lung Cancer by Integrating SEER-Medicare Data. Int J Mol Sci 2021; 22:ijms22147658. [PMID: 34299277 PMCID: PMC8306800 DOI: 10.3390/ijms22147658] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/08/2021] [Accepted: 07/15/2021] [Indexed: 12/14/2022] Open
Abstract
This study developed a novel methodology to correlate genome-scale microRNA (miRNA) expression profiles in a lung squamous cell carcinoma (LUSC) cohort (n = 57) with Surveillance, Epidemiology, and End Results (SEER)-Medicare LUSC patients (n = 33,897) as a function of composite tumor progression indicators of T, N, and M cancer stage and tumor grade. The selected prognostic and chemopredictive miRNAs were extensively validated with miRNA expression profiles of non-small-cell lung cancer (NSCLC) patient samples collected from US hospitals (n = 156) and public consortia including NCI-60, The Cancer Genome Atlas (TCGA; n = 1016), and Cancer Cell Line Encyclopedia (CCLE; n = 117). Hsa-miR-142-3p was associated with good prognosis and chemosensitivity in all the studied datasets. Hsa-miRNA-142-3p target genes (NUP205, RAN, CSE1L, SNRPD1, RPS11, SF3B1, COPA, ARCN1, and SNRNP200) had a significant impact on proliferation in 100% of the tested NSCLC cell lines in CRISPR-Cas9 (n = 78) and RNA interference (RNAi) screening (n = 92). Hsa-miR-142-3p-mediated pathways and functional networks in NSCLC short-term survivors were elucidated. Overall, the approach integrating SEER-Medicare data with comprehensive external validation can identify miRNAs with consistent expression patterns in tumor progression, with potential implications for prognosis and prediction of chemoresponse in large NSCLC patient populations.
Collapse
|
14
|
Huang R, Cho WC, Sun Y, Katie Chan KH. The Lung Cancer Associated MicroRNAs and Single Nucleotides Polymorphisms: a Mendelian Randomization Analysis. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:2346-2352. [PMID: 33018478 DOI: 10.1109/embc44109.2020.9176344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Lung cancer is a major public health burden and among the highest incidence and mortality rates of the cancers. MicroRNAs (miRNAs) play an important role in the development of lung cancer. The aim of this study was to investigate whether there was a potential causal relation between miRNAs and non-small-cell lung cancer (NSCLC). 1,026 patients with NSCLC from The Cancer Genome Atlas (TCGA) were analyzed. NSCLC associated SNPs' allele scores were established, and candidate miRNAs were filtered from differential expression analysis. Mendelian randomization (MR) analysis was conducted for 5 candidate miRNA (hsa-miR-135b, hsa-miR-142, hsa-miR-182, hsa-miR-183 and hsa-miR-3607) and 76 candidate SNPs in lung adenocarcinoma (LUAD) group. According to the core assumptions of MR, there was no clear evidence of a causal relation between the 5 candidate miRNAs and LUAD. The reads per million miRNAs mapped (RPM) level of candidate miRNAs changed less than 3% per allele score. To our knowledge, this is the first study using the TCGA data set to investigate the causal relation between miRNAs and lung cancer using the MR approach, and also one of the first MR studies to use miRNA expression as an exposure factor, with the SNPs as instrumental variables.
Collapse
|
15
|
Pan L, Xiao X, Zhao Y, Yin L, Fu M, Zhang X, Jiang P. The functional roles of long noncoding RNA DANCR in Human Cancers. J Cancer 2020; 11:6970-6981. [PMID: 33123287 PMCID: PMC7591992 DOI: 10.7150/jca.44384] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) have been wildly explored in various cellular processes and their aberrant expression could lead to tumorigenesis, development and progression. Differentiation antagonizing non-protein coding RNA (DANCR), a well-known lncRNA that is aberrant expression in various tumors, including hepatocellular carcinoma, gastric cancer, colorectal cancer, breast cancer, lung cancer and glioma and so on, in which it functions as oncogene mainly, contributing to cancer development and progression. High expressed DANCR is correlated with poor prognosis. In the present review, we summarize recent progression concerning the role, potential clinical utilities and underlying molecular mechanisms of DANCR related to occurrence and development of multiple cancers.
Collapse
Affiliation(s)
- Lei Pan
- Department of Breast Surgery, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu 212002, China
| | - Xiudi Xiao
- Department of Breast Surgery, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu 212002, China
| | - Yuan Zhao
- Department of Breast Surgery, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu 212002, China
| | - Liang Yin
- Department of Breast Surgery, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu 212002, China
| | - Min Fu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China.,Department of General Surgery, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu 212002, China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu 212013, China
| | - Pengcheng Jiang
- Department of General Surgery, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu 212002, China
| |
Collapse
|
16
|
Urbanek-Trzeciak MO, Galka-Marciniak P, Nawrocka PM, Kowal E, Szwec S, Giefing M, Kozlowski P. Pan-cancer analysis of somatic mutations in miRNA genes. EBioMedicine 2020; 61:103051. [PMID: 33038763 PMCID: PMC7648123 DOI: 10.1016/j.ebiom.2020.103051] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/16/2020] [Accepted: 09/16/2020] [Indexed: 02/08/2023] Open
Abstract
Background miRNAs are considered important players in oncogenesis, serving either as oncomiRs or suppressormiRs. Although the accumulation of somatic alterations is an intrinsic aspect of cancer development and many important cancer-driving mutations have been identified in protein-coding genes, the area of functional somatic mutations in miRNA genes is heavily understudied. Methods Here, based on the analysis of large genomic datasets, mostly the whole-exome sequencing of over 10,000 cancer/normal sample pairs deposited within the TCGA repository, we undertook an analysis of somatic mutations in miRNA genes. Findings We identified and characterized over 10,000 somatic mutations and showed that some of the miRNA genes are overmutated in Pan-Cancer and/or specific cancers. Nonrandom occurrence of the identified mutations was confirmed by a strong association of overmutated miRNA genes with KEGG pathways, most of which were related to specific cancer types or cancer-related processes. Additionally, we showed that mutations in some of the overmutated genes correlate with miRNA expression, cancer staging, and patient survival. Interpretation Our study is the first comprehensive Pan-Cancer study of cancer somatic mutations in miRNA genes. It may help to understand the consequences of mutations in miRNA genes and the identification of miRNA functional mutations. The results may also be the first step (form the basis and provide the resources) in the development of computational and/or statistical approaches/tools dedicated to the identification of cancer-driver miRNA genes. Funding This work was supported by research grants from the Polish National Science Centre 2016/22/A/NZ2/00184 and 2015/17/N/NZ3/03629.
Collapse
Affiliation(s)
| | | | - Paulina M Nawrocka
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Ewelina Kowal
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Sylwia Szwec
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Maciej Giefing
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Piotr Kozlowski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland.
| |
Collapse
|
17
|
Li Y, Xiang Y, Song Y, Wan L, Yu G, Tan L. Dysregulated miR-142, -33b and -423 in granulosa cells target TGFBR1 and SMAD7: a possible role in polycystic ovary syndrome. Mol Hum Reprod 2020; 25:638-646. [PMID: 30865275 DOI: 10.1093/molehr/gaz014] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/10/2019] [Accepted: 03/03/2019] [Indexed: 12/15/2022] Open
Abstract
It is well established that microRNA (miRNA) expression profiles are altered in patients with polycystic ovary syndrome (PCOS). In addition, abnormal transforming growth factor beta (TGFB) signaling in granulosa cells is related to the pathological conditions of PCOS. However, the function of dysregulated miRNAs in PCOS is still unclear. In this study, we aimed to elucidate the roles of specific miRNAs in PCOS. We collected follicular fluid from 46 patients with PCOS and 32 healthy controls. Granulosa cells (GCs) were separated and the levels of six candidate miRNAs were determined by quantitative RT-PCR. The direct targets of three dysregulated miRNAs were predicted using bioinformatic tools and confirmed using a dual luciferase assay and immunoblotting. The biological function of three dysregulated miRNAs in primary GCs was determined using a cell proliferation assay and flow cytometry. We found that miR-423 expression was downregulated (P = 0.038), and the levels of miR-33b (P = 0.032) and miR-142 (P = 0.021) were upregulated in GCs from patients with PCOS, compared to controls. miR-423 directly repressed SMAD family member 7 (SMAD7) expression, while transforming growth factor beta receptor 1 (TGFBR1) was a direct target of both miR-33b and miR-142. An RNA oligonucleotide mixture containing miR-423 inhibitor, miR-33b mimic, and miR-142 mimic repressed TGFB signaling, promoted cell proliferation (P = 0.0098), repressed apoptosis (P = 0.027), and increased S phase cell numbers (P = 0.0036) in primary cultures of GCs, compared to the cells treated with a sequence scrambled control RNA oligonucleotide. This study unveiled the possible roles of three miRNAs in PCOS and might provide candidate biomarkers for PCOS diagnosis while in vivo functional studies, using transgenic or knockout mouse models, are expected to confirm the roles of dysregulated miRNAs in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Yan Li
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yungai Xiang
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuxia Song
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lijing Wan
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guo Yu
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Tan
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
18
|
Yang M, He X, Huang X, Wang J, He Y, Wei L. LncRNA MIR4435-2HG-mediated upregulation of TGF-β1 promotes migration and proliferation of nonsmall cell lung cancer cells. ENVIRONMENTAL TOXICOLOGY 2020; 35:582-590. [PMID: 31875359 DOI: 10.1002/tox.22893] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/08/2019] [Accepted: 12/16/2019] [Indexed: 06/10/2023]
Abstract
The roles of long noncoding RNAs (lncRNAs) have been shown to play critical roles in tumor progression. Here, it was identified that lncRNA MIR4435-2HG was highly expressed in lung cancer tissues, especially in nonsmall cell lung cancer (NSCLC). A consistent result was obtained in lung cancer cells. Functional experiments showed that knockdown of MIR4435-2HG reduced the proliferation and migration ability of NSCLC cells. Transcriptome-sequencing analysis indicated that TGF-β signaling was mostly enriched in NSCLC cells with MIR4435-2HG knockdown. Furthermore, MIR4435-2HG was identified as an miRNA sponge for TGF-β1 and thus activated TGF-β signaling. Additionally, re-activation of TGF-β1 rescued MIR4435-2HG knockdown-mediated inhibition on the progression of NSCLC cells. Therefore, this work indicates a novel MIR4435-2HG/TGF-β1 axis responsible for NSCLC cell progression.
Collapse
Affiliation(s)
- Mei Yang
- Department of Respiratory and Critical Care, Yunnan Second People's Hospital, Kunming City, Yunnan Province, China
| | - Xiaohua He
- Department of Respiratory and Critical Care, Yunnan Second People's Hospital, Kunming City, Yunnan Province, China
| | - Xiaoxian Huang
- Department of Respiratory and Critical Care, Yunnan Second People's Hospital, Kunming City, Yunnan Province, China
| | - Jiyang Wang
- Department of Cardiovascular Surgery, Yunnan Second People's Hospital, Kunming City, Yunnan Province, China
| | - Yuxin He
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Li Wei
- Department of Respiratory and Critical Care, Yunnan Second People's Hospital, Kunming City, Yunnan Province, China
| |
Collapse
|
19
|
Zhang Y, Roth JA, Yu H, Ye Y, Xie K, Zhao H, Chang DW, Huang M, Li H, Qu J, Wu X. A 5-microRNA signature identified from serum microRNA profiling predicts survival in patients with advanced stage non-small cell lung cancer. Carcinogenesis 2020; 40:643-650. [PMID: 30428030 DOI: 10.1093/carcin/bgy132] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/11/2018] [Indexed: 12/19/2022] Open
Abstract
Circulating microRNAs (miRNAs) are potential biomarkers for cancer diagnosis, screening and prognosis. This study aimed to identify serum miRNAs as predictors of survival in patients with advanced non-small cell lung cancer (NSCLC). We profiled serum miRNAs in a pilot set of four patients with good survival (>24 months) and four patients with poor survival (<6 months). We selected 140 stably detectable miRNAs and 42 miRNAs reported in literature for further analysis. Expression of these 182 miRNAs was measured using high-throughput polymerase chain reaction assay, and their association with 3-year survival in the discovery (n = 345) and validation (n = 177) cohorts was assessed. Five serum miRNAs (miR-191, miR-28-3p, miR-145, miR-328 and miR-18a) were significantly associated with 3-year overall survival in both cohorts. A combined 5-miRNA risk score was created to assess the cumulative impact of these miRNAs on risk of death. Quartile analysis of the risk score showed significant association with 3-year death risk, with a 4.6-, 6.8- and 9.3-month reduction in median survival time for the second, third and fourth quartiles, respectively. Survival tree analysis also identified distinct risk groups with different 3-year survival durations. Data from The Cancer Genome Atlas revealed all five miRNAs were differentially expressed (P < 0.0001) in paired tumor and normal tissues. Pathway analysis indicated that target genes of these five miRNAs were mainly enriched in inflammatory/immune response pathways and pathways implicated in resistance to chemoradiotherapy and/or targeted therapy. Our results suggested that the 5-miRNA signature could serve as a prognostic predictor in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Yajie Zhang
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hao Yu
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuanqing Ye
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kunlin Xie
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hua Zhao
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David W Chang
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maosheng Huang
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xifeng Wu
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
20
|
High Mobility Group A (HMGA): Chromatin Nodes Controlled by a Knotty miRNA Network. Int J Mol Sci 2020; 21:ijms21030717. [PMID: 31979076 PMCID: PMC7038092 DOI: 10.3390/ijms21030717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/11/2022] Open
Abstract
High mobility group A (HMGA) proteins are oncofoetal chromatin architectural factors that are widely involved in regulating gene expression. These proteins are unique, because they are highly expressed in embryonic and cancer cells, where they play a relevant role in cell proliferation, stemness, and the acquisition of aggressive tumour traits, i.e., motility, invasiveness, and metastatic properties. The HMGA protein expression levels and activities are controlled by a connected set of events at the transcriptional, post-transcriptional, and post-translational levels. In fact, microRNA (miRNA)-mediated RNA stability is the most-studied mechanism of HMGA protein expression modulation. In this review, we contribute to a comprehensive overview of HMGA-targeting miRNAs; we provide detailed information regarding HMGA gene structural organization and a comprehensive evaluation and description of HMGA-targeting miRNAs, while focusing on those that are widely involved in HMGA regulation; and, we aim to offer insights into HMGA-miRNA mutual cross-talk from a functional and cancer-related perspective, highlighting possible clinical implications.
Collapse
|
21
|
Olafsdottir TA, Theodors F, Bjarnadottir K, Bjornsdottir US, Agustsdottir AB, Stefansson OA, Ivarsdottir EV, Sigurdsson JK, Benonisdottir S, Eyjolfsson GI, Gislason D, Gislason T, Guðmundsdóttir S, Gylfason A, Halldorsson BV, Halldorsson GH, Juliusdottir T, Kristinsdottir AM, Ludviksdottir D, Ludviksson BR, Masson G, Norland K, Onundarson PT, Olafsson I, Sigurdardottir O, Stefansdottir L, Sveinbjornsson G, Tragante V, Gudbjartsson DF, Thorleifsson G, Sulem P, Thorsteinsdottir U, Norddahl GL, Jonsdottir I, Stefansson K. Eighty-eight variants highlight the role of T cell regulation and airway remodeling in asthma pathogenesis. Nat Commun 2020; 11:393. [PMID: 31959851 PMCID: PMC6971247 DOI: 10.1038/s41467-019-14144-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 12/05/2019] [Indexed: 12/28/2022] Open
Abstract
Asthma is one of the most common chronic diseases affecting both children and adults. We report a genome-wide association meta-analysis of 69,189 cases and 702,199 controls from Iceland and UK biobank. We find 88 asthma risk variants at 56 loci, 19 previously unreported, and evaluate their effect on other asthma and allergic phenotypes. Of special interest are two low frequency variants associated with protection against asthma; a missense variant in TNFRSF8 and 3‘ UTR variant in TGFBR1. Functional studies show that the TNFRSF8 variant reduces TNFRSF8 expression both on cell surface and in soluble form, acting as loss of function. eQTL analysis suggests that the TGFBR1 variant acts through gain of function and together with an intronic variant in a downstream gene, SMAD3, points to defective TGFβR1 signaling as one of the biological perturbations increasing asthma risk. Our results increase the number of asthma variants and implicate genes with known role in T cell regulation, inflammation and airway remodeling in asthma pathogenesis. Asthma is a common allergic airway disease with significant inter-individual heterogeneity. Here, Olafsdottir et al. report a genome-wide meta-analysis of two large population-based cohorts to identify sequence variants that associate with asthma risk and perform follow-up functional analyses on a protective loss-of-function variant in TNFRSF8.
Collapse
Affiliation(s)
- Thorunn A Olafsdottir
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | - Unnur Steina Bjornsdottir
- Department of Medicine, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland.,The Medical Center Mjodd, Reykjavik, Iceland
| | | | | | - Erna V Ivarsdottir
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | - David Gislason
- The Medical Center Mjodd, Reykjavik, Iceland.,Department of Respiratory Medicine and Sleep, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Thorarinn Gislason
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Department of Sleep, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | | | | | - Bjarni V Halldorsson
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland.,School of Science and Engineering, Reykjavik University, Reykjavík, Iceland
| | | | | | | | - Dora Ludviksdottir
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Department of Respiratory Medicine and Sleep, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Bjorn R Ludviksson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | | | | | - Pall T Onundarson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Department of Laboratory Hematology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Olof Sigurdardottir
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Department of Clinical Biochemistry, Akureyri Hospital, Akureyri, Iceland
| | | | | | - Vinicius Tragante
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland.,Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland.,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Ingileif Jonsdottir
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland. .,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc., Reykjavik, Iceland. .,Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
22
|
Tan YF, Chen ZY, Wang L, Wang M, Liu XH. MiR-142-3p functions as an oncogene in prostate cancer by targeting FOXO1. J Cancer 2020; 11:1614-1624. [PMID: 32047567 PMCID: PMC6995382 DOI: 10.7150/jca.41888] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 12/08/2019] [Indexed: 12/20/2022] Open
Abstract
Prostate cancer (PCa) is a heterogeneous malignancy, and is a primary cause of cancer-related death in males. Forkhead box transcription factor O1 (FOXO1) exerts antitumor effects in various cancers, including PCa. However, the regulatory mechanism of miR-142-3p on FOXO1 expression in human PCa has not been characterized. In this study, we showed that FOXO1 protein levels were downregulated in PCa tissues and cells. Moreover, FOXO1 expression was a predictor of disease-free survival in patients with PCa and was a predictor of prognosis. Increased expression of FOXO1 suppressed cellular proliferation and induced cell cycle arrest at G0/G1 in vitro. However, FOXO1 mRNA and protein levels were inconsistent in human PCa tissues and cell lines. We showed that miR-142-3p levels were negatively correlated with FOXO1 protein levels in PCa. We also showed that miR-142-3p suppressed FOXO1 expression by directly targeting its 3′-untranslated region. Furthermore, suppression of miR-142-3p inhibited cell proliferation and induced cell cycle arrest, and these effects were blocked by FOXO1 knockdown. In vivo experiments showed that miR-142-3p knockout impaired tumor growth. Our results validate that FOXO1 acted as a tumor suppressor in PCa and demonstrated that FOXO1 was regulated by miR-142-3p, and miR-142-3p may be a potential target for treatment of PCa.
Collapse
Affiliation(s)
- Yi-Fan Tan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhi-Yuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Min Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xiu-Heng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
23
|
Carofino BL, Dinshaw KM, Ho PY, Cataisson C, Michalowski AM, Ryscavage A, Alkhas A, Wong NW, Koparde V, Yuspa SH. Head and neck squamous cancer progression is marked by CLIC4 attenuation in tumor epithelium and reciprocal stromal upregulation of miR-142-3p, a novel post-transcriptional regulator of CLIC4. Oncotarget 2019; 10:7251-7275. [PMID: 31921386 PMCID: PMC6944452 DOI: 10.18632/oncotarget.27387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023] Open
Abstract
Chloride intracellular channel 4 (CLIC4) is a tumor suppressor implicated in processes including growth arrest, differentiation, and apoptosis. CLIC4 protein expression is diminished in the tumor parenchyma during progression in squamous cell carcinoma (SCC) and other neoplasms, but the underlying mechanisms have not been identified. Data from The Cancer Genome Atlas suggest this is not driven by genomic alterations. However, screening and functional assays identified miR-142-3p as a regulator of CLIC4. CLIC4 and miR-142-3p expression are inversely correlated in head and neck (HN) SCC and cervical SCC, particularly in advanced stage cancers. In situ localization revealed that stromal immune cells, not tumor cells, are the predominant source of miR-142-3p in HNSCC. Furthermore, HNSCC single-cell expression data demonstrated that CLIC4 is lower in tumor epithelial cells than in stromal fibroblasts and endothelial cells. Tumor-specific downregulation of CLIC4 was confirmed in an SCC xenograft model concurrent with immune cell infiltration and miR-142-3p upregulation. These findings provide the first evidence of CLIC4 regulation by miRNA. Furthermore, the distinct localization of CLIC4 and miR-142-3p within the HNSCC tumor milieu highlight the limitations of bulk tumor analysis and provide critical considerations for both future mechanistic studies and use of miR-142-3p as a HNSCC biomarker.
Collapse
Affiliation(s)
- Brandi L. Carofino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kayla M. Dinshaw
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Pui Yan Ho
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Aleksandra M. Michalowski
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Andrew Ryscavage
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Nathan W. Wong
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Vishal Koparde
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Stuart H. Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
24
|
Li M, Jin Y, Li Y. LncRNA TP73-AS1 Activates TGF-β1 to Promote the Migration and Invasion of Colorectal Cancer Cell. Cancer Manag Res 2019; 11:10523-10529. [PMID: 31908524 PMCID: PMC6924583 DOI: 10.2147/cmar.s228490] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/26/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose LncRNA TP73-AS1 has been demonstrated to promote the developments of several types of human cancer. However, its role in colorectal cancer (CRC) is unknown. Methods All CRC patients (n=70, 40 males and 30 females, 38 to 66 years’ old, 52.1 ± 5.3 years’ old) in this study were enrolled in the Affiliated Hospital of Southwest Medical University from July 2012 to January 2014. Cells, vectors, and transient transfections, RT-qPCR, western-blotting, as well as measurements of cell migration and invasion abilities were carried out during the research. Results In the present study, we found that TP73-AS1 was upregulated in CRC tissues compared with adjacent non-CRC tissues in CRC patients. Upregulation of TP73-AS1 was closely correlated with poor prognosis. TGF-β1 was also upregulated in CRC tissues and positive correlated with TP73-AS1. TP73-AS1 overexpression caused upregulated TGF-β1 in CRC cells, while TGF-β1 overexpression showed no significant effect on TP73-AS1. TP73-AS1 and TGF-β1 overexpressions caused enhanced migration and invasion of CRC cells. TGF-β inhibitor treatment caused suppressed migration and invasion of CRC cells and attenuated effects of TP73-AS1 and TGF-β1 overexpression. Conclusion Therefore, TP73-AS1 may inactivate TGF-β1 to inhibit the migration and invasion of CRC cells.
Collapse
Affiliation(s)
- Mingli Li
- Department of Chinese traditional medicine, Jining University, Qufu City, Shandong Province, 273155, People's Republic of China
| | - Yuanyuan Jin
- The First People's Hospital of Yuzhong County, Lanzhou City, Gansu Province 730100, People's Republic of China
| | - Yuanzhi Li
- Department of Anorectal Hemorrhoid Fistula, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province 646000, People's Republic of China
| |
Collapse
|
25
|
Sun X, Cui S, Fu X, Liu C, Wang Z, Liu Y. MicroRNA-146-5p promotes proliferation, migration and invasion in lung cancer cells by targeting claudin-12. Cancer Biomark 2019; 25:89-99. [PMID: 31006666 DOI: 10.3233/cbm-182374] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
MicroRNAs (miRNAs) have been regarded as important regulators in different pathological processes of cells. Abnormal expression of miRNAs is frequently associated with cell proliferation, metastasis and apoptosis in various cancers. This study aimed to explore the effect of miR-146-5p on cell growth, metastasis and its mechanism in lung cancer cells. The expressions of miR-146-5p and claudin-12 in A549 and WI-38 cells were altered by transient transfection. Cisplatin was used to develop cells for regulation of cisplatin sensitivity. Cell viability, migration, invasion, and apoptosis were analyzed by CCK-8, Transwell and flow cytometry assays. The protein expressions of Wnt/β-catenin and PI3K/AKT/MAPK pathway-related factors were detected. miR-146-5p suppression inhibited cell viability, migration and invasion but promoted apoptosis in A549 cells. Moreover, overexpression of miR-146-5p reduced the sensitivity of A549 cells and WI-38 cells to cisplatin. In addition, claudin-12 was a direct target of miR-146-5p and was negatively regulated by miR-146-5p. Claudin-12 silence significantly reversed miR-146-5p suppression-mediated anti-tumor effects in A549 cells. Furthermore, miR-146-5p overexpression activated Wnt/β-catenin and PI3K/AKT/MAPK signal pathways via down-regulation of claudin-12. The results indicated that miR-146-5p promoted cell viability, migration and invasion, inhibited apoptosis and activated Wnt/β-catenin and PI3K/AKT/MAPK signal pathways by regulating claudin-12 expression in lung cancer cells.
Collapse
Affiliation(s)
- Xianghong Sun
- Department of Outpatient (Tumor), The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| | - Shichao Cui
- Department of Respiratory Medicine, The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| | - Xiaofeng Fu
- Department of Outpatient, The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| | - Chuan Liu
- Department of Surgery, Qingdao West Coast New District No.2 People's Hospital, Qingdao, Shandong, China
| | - Zhi Wang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University (Shinan Branch), Qingdao, Shandong, China
| | - Yuanwei Liu
- Department of Cancer Comprehensive Therapy, The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| |
Collapse
|
26
|
Moradi-Marjaneh R, Khazaei M, Ferns GA, Aghaee-Bakhtiari SH. The Role of TGF-β Signaling Regulatory MicroRNAs in the Pathogenesis of Colorectal Cancer. Curr Pharm Des 2019; 24:4611-4618. [PMID: 30636580 DOI: 10.2174/1381612825666190110150705] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/24/2018] [Accepted: 12/31/2018] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is one of the most common cancers globally and is associated with a high mortality rate. The transforming growth factor beta (TGF-β) signaling pathway plays an important role in normal intestinal tissue function, but has also been implicated in the development of CRC. MicroRNAs (miRNAs) have also recently emerged as important regulators of cancer development and progression. They act by targeting multiple signaling pathways including the TGF-β signaling pathway. There is growing evidence demonstrating that miRNAs target various components of the TGF-β signaling pathway, including TGF-β1, TGF-β2, regulatory SMADs (SMAD1, 2, 3, 5 and 9), co-mediator SMAD4, inhibitory SMADs (SMAD6 and 7) and the TGF-β receptors, and thereby alter the proliferation and migration of CRC cells. In this review, we summarize the data concerning the interaction between TGF-β signaling pathway and miRNAs with the aim to better understanding the CRC molecular mechanisms and hence better management of this disease.
Collapse
Affiliation(s)
- Reyhaneh Moradi-Marjaneh
- Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, United Kingdom
| | - Seyed H Aghaee-Bakhtiari
- Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
27
|
Xu T, He BS, Pan B, Pan YQ, Sun HL, Liu XX, Xu XN, Chen XX, Zeng KX, Xu M, Wang SK. MiR-142-3p functions as a tumor suppressor by targeting RAC1/PAK1 pathway in breast cancer. J Cell Physiol 2019; 235:4928-4940. [PMID: 31674013 DOI: 10.1002/jcp.29372] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/07/2019] [Indexed: 12/24/2022]
Abstract
MicroRNA-142-3p (miR-142-3p) was previously investigated in various cancers, whereas, it's role in breast cancer (BC) remains far from understood. In this study, we found that miR-142-3p was markedly decreased both in cell lines and BC tumor tissues. Elevated miR-142-3p expression suppressed growth and metastasis of BC cell lines via gain-of-function assay in vitro and in vivo. Mechanistically, miR-142-3p could regulate the ras-related C3 botulinum toxin substrate 1 (RAC1) expression in protein level, which simultaneously suppressed the epithelial-to-mesenchymal transition related protein levels and the activity of PAK1 phosphorylation, respectively. In addition, rescue experiments revealed RAC1 overexpression could reverse tumor-suppressive role of miR-142-3p. Our results showed miR-142-3p could function as a tumor suppressor via targeting RAC1/PAK1 pathway in BC, suggesting a potent therapeutic target for BC treatment.
Collapse
Affiliation(s)
- Tao Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Bang-Shun He
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Bei Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Qin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hui-Ling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiang-Xiang Liu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xue-Ni Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Medical College, Southeast University, Nanjing, China
| | - Xiao-Xiang Chen
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Medical College, Southeast University, Nanjing, China
| | - Kai-Xuan Zeng
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Medical College, Southeast University, Nanjing, China
| | - Mu Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shu-Kui Wang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
28
|
Wang S, Huang M, Wang Z, Wang W, Zhang Z, Qu S, Liu C. MicroRNA‑133b targets TGFβ receptor I to inhibit TGF‑β‑induced epithelial‑to‑mesenchymal transition and metastasis by suppressing the TGF‑β/SMAD pathway in breast cancer. Int J Oncol 2019; 55:1097-1109. [PMID: 31545407 PMCID: PMC6776189 DOI: 10.3892/ijo.2019.4879] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/04/2019] [Indexed: 01/01/2023] Open
Abstract
Breast cancer (BC) is one of the most common types of cancer and the leading cause of cancer-associated mortality among women worldwide. Accumulating evidence indicates that microRNA (miR)-133b inhibits the proliferation and invasion of cancer cells. Considering that transforming growth factor (TGF)-β signaling plays a key role in cellular epithelial-to-mesenchymal transition (EMT) and cancer metastasis, it is crucial to explore the roles and underlying molecular mechanisms of miR-133b in regulating TGF-β-induced EMT during progression of BC. In the present study, an inverse correlation was observed between the expression of miR-133b and TGFβ receptor I (TGFβR1) mRNA in BC cells and tissues. Furthermore, miR-133b expression was found to be decreased in the BC tissues of patients with lymph node metastasis and advanced tumor-node-metastasis stage, while the expression of TGFβR1 was upregulated. Overexpression of miR-133b significantly decreased the expression of TGFβR1, an indispensable receptor of TGF-β/SMAD signaling, and suppressed TGF-β-induced EMT and BC cell invasion in vitro, whereas miR-133b knockdown exerted the opposite effects. Mechanistically, TGFβR1 was verified as a direct target of miR-133b as determined by bioinformatics analysis and a dual-luciferase reporter assay. In addition, small interfering RNA-mediated knockdown of TGFβR1 mimicked the phenotype of miR-133b overexpression in BC cells. Furthermore, miR-133b overexpression suppressed BC cell invasion in vivo. Collectively, the findings of the present study indicated that miR-133b acts as a tumor suppressor, inhibiting TGF-β-induced EMT and metastasis by directly targeting TGFβR1, and suppressing the TGF-β/SMAD pathway. Therefore, miR-133b may be of value as a diagnostic biomarker of BC.
Collapse
Affiliation(s)
- Shengjie Wang
- Laboratory Animal Center of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Mingliang Huang
- Medical College of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Zichen Wang
- Medical College of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Wan Wang
- Medical College of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Zhiyuan Zhang
- Medical College of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Shuting Qu
- Medical College of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Chun Liu
- Laboratory Animal Center of Nantong University, Nantong, Jiangsu 226000, P.R. China
| |
Collapse
|
29
|
Xu Y, Wang L, Jiang L, Zhang X. Novel MicroRNA Biomarkers, miR-142-5p, miR-550a, miR-1826, and miR-1201, Were Identified for Primary Melanoma. J Comput Biol 2019; 27:815-824. [PMID: 31526187 DOI: 10.1089/cmb.2019.0198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This study was aimed to identify novel miRNA biomarkers and explore the cooperative function of multi-RNAs in the progress of primary melanoma. The miRNA expression profile GSE62370 generated from 9 congenital nevi and 92 primary melanoma samples was downloaded from the Gene Expression Omnibus database. Differentially expressed miRNAs between primary melanoma and congenital nevi were compared and the target genes of them were selected. Pathway enrichment analysis and protein/protein interaction (PPI) network of miRNA target genes were performed. In addition, the differential expression of miRNAs to identify the tumor stage-dependent differences in miRNA expression was analyzed. Differentially expressed miRNAs, including 6 upregulated and 23 downregulated, were found in primary melanoma. Besides, the miRNA-associated gene regulatory network revealed 274 nodes, including miR-142-5p and miR-125b, and 307 miRNA-target pairs. miRNA-related Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, such as melanoma, was found. Target genes in the PPI module were mainly enriched in cancer-related pathways. Finally, the melanoma stage-related overexpressed miR-142-5p and the downregulated miR-550, miR-1826, miR-1201, miR-205, and miR-125b were identified. Some validated miRNAs, including miR-125a/b, let-7a/b, and miR-205, were found and illustrated the reliability of our study. Four novel miRNAs, including miR-142-5p, miR-550a, miR-1826, and miR-1201, were considered to have potential prognostic values for primary melanoma.
Collapse
Affiliation(s)
- Yangchun Xu
- Department of Dermatology, Second Hospital of Jilin University, Changchun, China
| | - Ling Wang
- Department of Gynecology, Second Hospital of Jilin University, Changchun, China
| | - Lanxiang Jiang
- Department of Dermatology, Second Hospital of Jilin University, Changchun, China
| | - Xuan Zhang
- Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Berrien-Elliott MM, Sun Y, Neal C, Ireland A, Trissal MC, Sullivan RP, Wagner JA, Leong JW, Wong P, Mah-Som AY, Wong TN, Schappe T, Keppel CR, Cortez VS, Stamatiades EG, Li MO, Colonna M, Link DC, French AR, Cooper MA, Wang WL, Boldin MP, Reddy P, Fehniger TA. MicroRNA-142 Is Critical for the Homeostasis and Function of Type 1 Innate Lymphoid Cells. Immunity 2019; 51:479-490.e6. [PMID: 31402259 PMCID: PMC6750984 DOI: 10.1016/j.immuni.2019.06.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/16/2019] [Accepted: 06/20/2019] [Indexed: 02/07/2023]
Abstract
Natural killer (NK) cells are cytotoxic type 1 innate lymphoid cells (ILCs) that defend against viruses and mediate anti-tumor responses, yet mechanisms controlling their development and function remain incompletely understood. We hypothesized that the abundantly expressed microRNA-142 (miR-142) is a critical regulator of type 1 ILC biology. Interleukin-15 (IL-15) signaling induced miR-142 expression, whereas global and ILC-specific miR-142-deficient mice exhibited a cell-intrinsic loss of NK cells. Death of NK cells resulted from diminished IL-15 receptor signaling within miR-142-deficient mice, likely via reduced suppressor of cytokine signaling-1 (Socs1) regulation by miR-142-5p. ILCs persisting in Mir142-/- mice demonstrated increased expression of the miR-142-3p target αV integrin, which supported their survival. Global miR-142-deficient mice exhibited an expansion of ILC1-like cells concurrent with increased transforming growth factor-β (TGF-β) signaling. Further, miR-142-deficient mice had reduced NK-cell-dependent function and increased susceptibility to murine cytomegalovirus (MCMV) infection. Thus, miR-142 critically integrates environmental cues for proper type 1 ILC homeostasis and defense against viral infection.
Collapse
Affiliation(s)
- Melissa M Berrien-Elliott
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Yaping Sun
- Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Carly Neal
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Aaron Ireland
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Maria C Trissal
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ryan P Sullivan
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Julia A Wagner
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jeffrey W Leong
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Pamela Wong
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Annelise Y Mah-Som
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Terrence N Wong
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Timothy Schappe
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Catherine R Keppel
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Victor S Cortez
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | | | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Daniel C Link
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Anthony R French
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Wei-Le Wang
- Department of Molecular and Cellular Biology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Mark P Boldin
- Department of Molecular and Cellular Biology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Pavan Reddy
- Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Todd A Fehniger
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
31
|
Feng H, Jia XM, Gao NN, Tang H, Huang W, Ning N. Overexpressed VEPH1 inhibits epithelial-mesenchymal transition, invasion, and migration of human cutaneous melanoma cells through inactivating the TGF-β signaling pathway. Cell Cycle 2019; 18:2860-2875. [PMID: 31599708 DOI: 10.1080/15384101.2019.1638191] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Malignant melanoma has a profound influence on populations around the world, with the underlying mechanisms controlling this disease yet to be fully identified. Hence, the current study aimed to investigate effects associated with VEPH1 on epithelial-mesenchymal transition (EMT), proliferation, invasion, migration and the apoptosis of human cutaneous melanoma (CM) cells through the TGF-β signaling pathway. Microarray-based gene analysis was initially performed to screen the CM-related differentially expressed genes. The expression of VEPH1, TGF-β signaling pathway- and EMT-related genes in CM tissues and cell lines was subsequently evaluated. Gain-of- and loss-of-function experiments were conducted to examine the effects of VEPH1 and the TGF-β signaling pathway on the expression of EMT-related genes, cell proliferation, migration, invasion, cell cycle and apoptosis in vitro. Finally, tumor formation in nude mice was conducted. VEPH1 was lowly expressed and regulated the progression of CM with involvement in the TGF-β signaling pathway. Human CM tissues were noted to activate the TGF-β signaling pathway and EMT. A375 cells treated with overexpressed VEPH1 plasmids or/and TGF-β signaling pathway inhibitor SB-431542 displayed diminished TGF-β, SMAD4, Vimentin and N-cadherin expression while the expression of E-cadherin was elevated, accompanied by decreased cell proliferation, migration, invasion, inhibited cell cycle entry. However, si-VEPH1 or TGF-β signaling pathway activator contributed to reverse results. Taken together, the key findings of the current study present evidence suggesting that VEPH1 protects against human CM by inhibiting the activation of the TGF-β signaling pathway, highlighting its potential as a target for the prognosis and diagnosis of CM.
Collapse
Affiliation(s)
- Hao Feng
- Department of Dermatology, The First Affiliated Hospital of Hunan Normal University (Hunan Province People's Hospital) , Changsha , P.R. China
| | - Xiao-Min Jia
- Department of Pathology, The First Affiliated Hospital of Hunan Normal University (Hunan Province People's Hospital) , Changsha , P.R. China
| | - Ni-Na Gao
- Department of Pathology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital , Changsha , P.R. China
| | - Hua Tang
- Department of Dermatology, The First Affiliated Hospital of Hunan Normal University (Hunan Province People's Hospital) , Changsha , P.R. China
| | - Wei Huang
- Department of Gynaecology, The First Affiliated Hospital of Hunan Normal University (Hunan Province People's Hospital) , Changsha , P.R. China
| | - Ning Ning
- Department of Medical Administration, The First Affiliated Hospital of Hunan Normal University (Hunan Province People's Hospital) , Changsha , P.R. China
| |
Collapse
|
32
|
Zeng SG, Lin X, Liu JC, Zhou J. Hypoxia‑induced internalization of connexin 26 and connexin 43 in pulmonary epithelial cells is involved in the occurrence of non‑small cell lung cancer via the P53/MDM2 signaling pathway. Int J Oncol 2019; 55:845-859. [PMID: 31485592 PMCID: PMC6741836 DOI: 10.3892/ijo.2019.4867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 04/16/2019] [Indexed: 12/18/2022] Open
Abstract
Reports have highlighted an association between connexins (CXs) or gap junction proteins and non-small cell lung cancer (NSCLC). In the present study, it was aimed to elucidate the regulatory mechanism of CX26 and CX43 under hypoxic conditions in NSCLC. Clinical samples were collected for analysis of CX26 and CX43 expression and clinical cancerization followed by quantification of CX26 and CX43 expression. Following the establishment of an in vitro hypoxia model, P53/murine double minute-2 (MDM2) signaling pathway-, proliferation- and epithelial-mesenchymal transition (EMT)-related genes were quantified to evaluate the influence of CX26 and CX43 on the biological functions of pulmonary epithelial cells in NSCLC. In addition, the proliferation and tumorigenicity of cancer cells were assessed by EdU staining and xenograft tumors, respectively. Decreased expression of CX26 and CX43 was found in cancer tissues compared with surrounding normal tissue. Hypoxia was shown to activate the P53/MDM2 axis and stimulate the downregulation, ubiquitination and degradation of CX26 and CX43, which were translocated from the membrane to the cytoplasm. Low levels of CX26 and CX43 were demonstrated to further promote EMT and the induction of the proliferation and tumorigenicity of cancer cells. These results were reflected by decreased E-cadherin expression and increased N-cadherin expression, along with increased cell migration, promoted cell proliferation ability and elevated relative protein expression of Oct4 and Nanog, and accelerated tumor growth, accompanied by a higher number of metastatic nodes. Taken together, the key observations of the present study demonstrate that the internalization of CX26 and CX43 promoted proliferation, EMT and migration and thus induced NSCLC via aberrant activation of the P53/MDM2 signaling pathway under hypoxic conditions.
Collapse
Affiliation(s)
- Shang-Gan Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiang Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ji-Chun Liu
- Departments of Cardio‑Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jin Zhou
- Department of Respiratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
33
|
Mansoori B, Mohammadi A, Gjerstorff MF, Shirjang S, Asadzadeh Z, Khaze V, Holmskov U, Kazemi T, Duijf PHG, Baradaran B. miR-142-3p is a tumor suppressor that inhibits estrogen receptor expression in ER-positive breast cancer. J Cell Physiol 2019; 234:16043-16053. [PMID: 30741415 DOI: 10.1002/jcp.28263] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/16/2019] [Accepted: 01/22/2019] [Indexed: 01/24/2023]
Abstract
Estrogen receptors (ERs) are involved in the development of many types of malignant tumors, in particular, breast cancer. Among others, ERs affect cell growth, proliferation, and differentiation. The microRNA (miRNA) miR-142-3p has been shown to inhibit carcinogenesis by regulating various cellular processes, including cell cycle progression, cell migration, apoptosis, and invasion. It does so via targeting molecules involved in a range of signaling pathways. We surgically collected 20 ER-positive breast cancer samples, each with matched adjacent normal breast tissue, and measured the expression of miR-142-3p via quantitative real-time polymerase chain reaction (qRT-PCR). Bioinformatics methods, luciferase reporter assay, qRT-PCR, and western blot analysis were used to assess whether miR-142-3p could target ESR1, which encodes the estrogen receptor, in ER-positive breast cancer cells and patient samples. We also restored miRNA expression and performed cell viability, cytotoxicity, and colony formation assays. Western blot analysis and qRT-PCR were used to study the expression of apoptosis and stemness markers. We found that miR-142-3p is downregulated in ER-positive breast cancers. Restoration of miR-142-3p expression in ER-positive breast cancer cells reduced cell viability, induced apoptosis via the intrinsic pathway and decreased both colony formation and the expression of stem cell markers. Bioinformatic analysis predicted miR-142-3p could bind to 3'-untranslated region ESR1 messenger RNA (mRNA). Consistently, we demonstrated that miR-142-3p reduced luciferase activity in ER-positive breast cancer cells, and decreased ESR1 expression in both mRNA and protein levels. The results revealed miR-142-3p and ESR1 expression correlated negatively in ER-positive breast cancer samples. The results suggest miR-142-3p acts as a tumor suppressor via multiple mechanisms. Thus, restoration of miR-142-3p expression, for example, via miRNA replacement therapy, may represent an effective strategy for the treatment of ER-positive breast cancer patients.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morten F Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Uffe Holmskov
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Jin C, Xiao L, Zhou Z, Zhu Y, Tian G, Ren S. MiR-142-3p suppresses the proliferation, migration and invasion through inhibition of NR2F6 in lung adenocarcinoma. Hum Cell 2019; 32:437-446. [PMID: 31168689 DOI: 10.1007/s13577-019-00258-0] [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] [Received: 09/20/2018] [Accepted: 05/06/2019] [Indexed: 12/09/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide and lung adenocarcinoma is its main type. MicroRNAs are small, non-coding and single-strand RNAs that regulate gene expression in human cancers. The aim of our study is to investigate the underlying molecular mechanism of miR-142-3p in NSCLC. The expression of miR-142-3p in lung adenocarcinoma tissues and cells was detected by RT-qPCR. Next, cell proliferation, migration, invasion and apoptosis were examined by CCK-8, scratch assay, transwell assay and flow cytometry in A549 and HCC827 cells, respectively. Then, the target of miR-142-3p was predicted by targetscanHuman 7.2 and confirmed using dual-luciferase reporter assay. Additionally, RT-qPCR and western blot were used to detect the expression of NR2F6, MMP2, MMP9 and caspase-3. The results showed that miR-142-3p expression was significantly decreased in tumor tissues and cells. Overexpression of miR-142-3p inhibited the proliferation, migration, invasion and promoted cell apoptosis in vitro, while knockdown of miR-142-3p had reversed function. Furthermore, NR2F6 was identified as a direct target of miR-142-3p, which was negatively correlated with miR-142-3p expression. Finally, miR-142-3p overexpression suppressed the expression of NR2F6, MMP2 and MMP9, but improved caspase-3 expression, while miR-142-3p knockdown got the opposite expression results. Suppressing MMP2 and MMP9 activities inhibited cell invasion. In summary, these findings indicated that miR-142-3p inhibits lung adenocarcinoma cell proliferation, migration and invasion, and enhances cell apoptosis by targeting NR2F6, suggesting that miR-142-3p may be a novel therapeutic target for lung adenocarcinoma treatment.
Collapse
Affiliation(s)
- Chang'e Jin
- Department of Respiratory and Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, Guangdong, China
| | - Liang Xiao
- Department of Surgery and Oncology, Shenzhen Second People's Hospital, First Affiliated Hospital to Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Zeqiang Zhou
- Department of Surgery and Oncology, Shenzhen Second People's Hospital, First Affiliated Hospital to Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Yan Zhu
- Department of Surgery and Oncology, Shenzhen Second People's Hospital, First Affiliated Hospital to Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Geng Tian
- Department of Surgery and Oncology, Shenzhen Second People's Hospital, First Affiliated Hospital to Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Shuhua Ren
- Department of Thoracic Surgery, Tangshan Gongren Hospital, No. 27, Wenhua Road, Tangshan, 063000, Hebei, China.
| |
Collapse
|
35
|
Weidle UH, Birzele F, Nopora A. MicroRNAs as Potential Targets for Therapeutic Intervention With Metastasis of Non-small Cell Lung Cancer. Cancer Genomics Proteomics 2019; 16:99-119. [PMID: 30850362 PMCID: PMC6489690 DOI: 10.21873/cgp.20116] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 02/08/2023] Open
Abstract
The death toll of non-small cell lung cancer (NSCLC) patients is primarily due to metastases, which are poorly amenable to therapeutic intervention. In this review we focus on miRs associated with metastasis of NSCLC as potential new targets for anti-metastatic therapy. We discuss miRs validated as therapeutic targets by in vitro data, identification of target(s) and pathway(s) and in vivo efficacy data in at least one clinically-relevant metastasis-related model. A few of the discussed miRs correlate with the clinical status of NSCLC patients. Using miRs as therapeutic agents has the advantage that targeting a single miR can potentially interfere with several metastatic pathways. Depending on their mode of action, the corresponding miRs can be up- or down-regulated compared to normal matching tissues. Here, we describe therapeutic approaches for reconstitution therapy and miR inhibition, general principles of anti-metastatic therapy as well as current technical pitfalls.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Fabian Birzele
- Roche Innovation Center Basel, F. Hofman La Roche, Basel, Switzerland
| | - Adam Nopora
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
36
|
Gao P, Wang H, Yu J, Zhang J, Yang Z, Liu M, Niu Y, Wei X, Wang W, Li H, Wang Y, Sun G. miR-3607-3p suppresses non-small cell lung cancer (NSCLC) by targeting TGFBR1 and CCNE2. PLoS Genet 2018; 14:e1007790. [PMID: 30557355 PMCID: PMC6312350 DOI: 10.1371/journal.pgen.1007790] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 12/31/2018] [Accepted: 10/25/2018] [Indexed: 01/07/2023] Open
Abstract
Accumulating evidence indicates that miRNAs can be promising diagnostic and/or prognostic markers for various cancers. In this study, we identified a novel miRNA, miR-3607-3p, and its targets in non-small cell lung cancer (NSCLC). The expression of miR-3607-3p was measured and its correlation with patient prognosis was determined. Ectopic expression in NSCLC cells, xenografts, and metastasis models was used to evaluate the effects of miR-3607-3p on proliferation and migration of NSCLC. Luciferase assay and western blotting were performed to validate the potential targets of miR-3607-3p after preliminary screening by microarray analysis and computer-aided algorithms. We demonstrated that miR-3607-3p was downregulated in NSCLC tissues and that miR-3607-3p might act as an independent predictor for overall survival in NSCLC. Moreover, serum miR-3607-3p may be a novel and stable marker for NSCLC. We found that overexpression of miR-3607-3p inhibited cell proliferation, colony formation, migration and invasion, and hampered the cell cycle of NSCLC cell lines in vitro. Our results suggested that miR-3607-3p directly targets TGFBR1 and CCNE2. In accordance with in vitro studies, we confirmed that miR-3607-3p functions as a potent suppressor miRNA of NSCLC. We showed that miR-3607-3p agomir could reduce tumor growth and inhibit TGFBR1 and CCNE2 protein expression. Taken together, our findings indicate that miR-3607-3p can inhibit NSCLC cell growth and metastasis by targeting TGFBR1 and CCNE2 protein expression, and provide new evidence of miR-3607-3p as a potential non-invasive biomarker and therapeutic target for NSCLC.
Collapse
MESH Headings
- Aged
- Animals
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Cell Cycle/genetics
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation/genetics
- Cyclins/antagonists & inhibitors
- Cyclins/genetics
- Down-Regulation
- Female
- Gene Knockdown Techniques
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Male
- Mice
- Mice, Nude
- Middle Aged
- Neoplasm Invasiveness/genetics
- Neoplasm Invasiveness/prevention & control
- Neoplasm Metastasis
- Prognosis
- RNA, Small Nucleolar/antagonists & inhibitors
- RNA, Small Nucleolar/blood
- RNA, Small Nucleolar/genetics
- Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors
- Receptor, Transforming Growth Factor-beta Type I/genetics
Collapse
Affiliation(s)
- Peng Gao
- Department of Radiation Oncology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| | - Huan Wang
- Department of Radiation Oncology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| | - Jiarui Yu
- Department of Radiation Oncology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| | - Jie Zhang
- Department of pathology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| | - Zhao Yang
- Department of Radiation Oncology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| | - Meiyue Liu
- Department of Radiation Oncology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| | - Yi Niu
- Department of Radiation Oncology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| | - Xiaomei Wei
- Department of Radiation Oncology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| | - Wei Wang
- Department of Radiation Oncology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| | - Hongmin Li
- Department of pathology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| | - Yadi Wang
- Department of Radiation Oncology, PLA Army General Hospital, Beijing, China
| | - Guogui Sun
- Department of Radiation Oncology, North China University of Science and Technology Affiliated People’s Hospital, Tangshan, China
| |
Collapse
|
37
|
Lawson J, Dickman C, Towle R, Jabalee J, Javer A, Garnis C. Extracellular vesicle secretion of miR-142-3p from lung adenocarcinoma cells induces tumor promoting changes in the stroma through cell-cell communication. Mol Carcinog 2018; 58:376-387. [PMID: 30362621 DOI: 10.1002/mc.22935] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 10/10/2018] [Accepted: 10/23/2018] [Indexed: 11/10/2022]
Abstract
Extracellular vesicles (EVs) are mediators of communication between cancer cells and the surrounding tumor microenvironment. EV content is able to influence key tumorigenic changes including invasion, metastasis, and inducing pro-tumor changes in the stroma. MiR-142-3p is a known tumor suppressor in LAC and was recently shown to be enriched within LAC EVs, indicating its potential as a key signaling miRNA. Our research demonstrates the role EV associated miR-142-3p plays when transferred from LAC cells to both endothelial and fibroblast cells. We demonstrate that transfer of miR-142-3p in LAC EVs to endothelial cells promotes angiogenesis through inhibition of TGFβR1. Additionally, we show EV associated miR-142-3p promotes the cancer-associated fibroblast phenotype in lung fibroblast cells which we show is independent of TGFβ signaling. These findings suggest that miR-142-3p within LAC EVs can be transferred from LAC cells to both endothelial and fibroblast cells to promote tumor associated changes.
Collapse
Affiliation(s)
- James Lawson
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Christopher Dickman
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Rebecca Towle
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - James Jabalee
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Ariana Javer
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Cathie Garnis
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
38
|
Shabani P, Izadpanah S, Aghebati-Maleki A, Baghbani E, Baghbanzadeh A, Fotouhi A, Bakhshinejad B, Aghebati-Maleki L, Baradaran B. Role of miR-142 in the pathogenesis of osteosarcoma and its potential as therapeutic approach. J Cell Biochem 2018; 120:4783-4793. [PMID: 30450580 DOI: 10.1002/jcb.27857] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023]
Abstract
Osteosarcoma (OS) is the most common primary malignant tumor of the bone with a strong tendency to early metastasis, and occurs in growing bones more commonly in children and adolescents. Considering the limited therapeutic methods and lack of 100% success of these methods, developing innovative therapies with high efficacy and lower side effects is needed. Meanwhile, miRNAs and the studies indicating the involvement of miRNAs in OS development have attracted attentions as a result of the frequent abnormalities in expression of miRNAs in cancer. miRNAs are noncoding short sequences with lengths ranging from 18 to 25 nucleotides that play a very important role in cellular processes, such as proliferation, differentiation, migration, and apoptosis. MiRNAs can have either oncogenic or tumor suppressive role based on cellular function and targets. This review aimed to have overview on miR-142 as a tumor suppressor in OS. Moreover, the genes involved in the disease, such as RAC1, HMAG1, MMP9, MMP2, and E-cadherin, which have irregularities as a result of change in miR-142 expression, and, thereby, result in increasing the proliferation, invasion, and metastasis of the cells in the tissues and OS cells will be discussed.
Collapse
Affiliation(s)
- Parastoo Shabani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sama Izadpanah
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Genetics and Molecular Medicine, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Fotouhi
- Department of Orthopedic Surgery, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Bakhshinejad
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
39
|
Wang L, Tong X, Zhou Z, Wang S, Lei Z, Zhang T, Liu Z, Zeng Y, Li C, Zhao J, Su Z, Zhang C, Liu X, Xu G, Zhang HT. Circular RNA hsa_circ_0008305 (circPTK2) inhibits TGF-β-induced epithelial-mesenchymal transition and metastasis by controlling TIF1γ in non-small cell lung cancer. Mol Cancer 2018; 17:140. [PMID: 30261900 PMCID: PMC6161470 DOI: 10.1186/s12943-018-0889-7] [Citation(s) in RCA: 265] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 09/13/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND TGF-β promotes tumor invasion and metastasis through inducing epithelial-mesenchymal transition (EMT) in non-small cell lung cancer (NSCLC). Circular RNAs (circRNAs) are recognized as functional non-coding RNAs involved in human cancers. However, whether and how circRNAs contribute to TGF-β-induced EMT and metastasis in NSCLC remain vague. Here, we investigated the regulation and function of Circular RNA hsa_circ_0008305 (circPTK2) in TGF-β-induced EMT and tumor metastasis, as well as a link between circPTK2 and transcriptional intermediary factor 1 γ (TIF1γ) in NSCLC. METHODS Circular RNAs were determined by human circRNA Array analysis, real-time quantitative reverse transcriptase PCR and northern blot. Luciferase reporter, RNA-binding protein immunoprecipitation (RIP), RNA pull-down and fluorescence in situ hybridization (FISH) assays were employed to test the interaction between circPTK2 and miR-429/miR-200b-3p. Ectopic overexpression and siRNA-mediated knockdown of circPTK2, TGF-β-induced EMT, Transwell migration and invasion in vitro, and in vivo experiment of metastasis were used to evaluate the function of circPTK2. Transcription and prognosis analyses were done in public databases. RESULTS CircPTK2 and TIF1γ were significantly down-regulated in NSCLC cells undergoing EMT induced by TGF-β. CircPTK2 overexpression augmented TIF1γ expression, inhibited TGF-β-induced EMT and NSCLC cell invasion, whereas circPTK2 knockdown had the opposite effects. CircPTK2 functions as a sponge of miR-429/miR-200b-3p, and miR-429/miR-200b-3p promote TGF-β-induced EMT and NSCLC cell invasion by targeting TIF1γ. CircPTK2 overexpression inhibited the invasion-promoting phenotype of endogenous miR-429/miR-200b-3p in NSCLC cells in response to TGF-β. CircPTK2 overexpression significantly decreased the expression of Snail, an important downstream transcriptional activator of TGF-β/Smad signaling. In an in vivo experiment of metastasis, circPTK2 overexpression suppressed NSCLC cell metastasis. Moreover, circPTK2 expression was dramatically down-regulated and positively correlated with TIF1γ expression in human NSCLC tissues. Especially, circPTK2 was significantly lower in metastatic NSCLC tissues than non-metastatic counterparts. CONCLUSION Our findings show that circPTK2 (hsa_circ_0008305) inhibits TGF-β-induced EMT and metastasis by controlling TIF1γ in NSCLC, revealing a novel mechanism by which circRNA regulates TGF-β-induced EMT and tumor metastasis, and suggesting that circPTK2 overexpression could provide a therapeutic strategy for advanced NSCLC.
Collapse
Affiliation(s)
- Longqiang Wang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, 199 Ren’ai Road, Suzhou, 215123 Jiangsu China
| | - Xin Tong
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, 199 Ren’ai Road, Suzhou, 215123 Jiangsu China
| | - Zhengyu Zhou
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, 199 Ren’ai Road, Suzhou, 215123 Jiangsu China
- Laboratory Animal Center, Medical College of Soochow University, Suzhou, 215123 Jiangsu China
| | - Shengjie Wang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, 199 Ren’ai Road, Suzhou, 215123 Jiangsu China
- Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang, 222000 China
| | - Zhe Lei
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, 199 Ren’ai Road, Suzhou, 215123 Jiangsu China
| | - Tianze Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086 Heilongjiang China
| | - Zeyi Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Suzhou, 215006 Jiangsu China
| | - Yuanyuan Zeng
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Suzhou, 215006 Jiangsu China
| | - Chang Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Suzhou, 215006 Jiangsu China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Suzhou, 215006 Jiangsu China
| | - Zhiyue Su
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, 199 Ren’ai Road, Suzhou, 215123 Jiangsu China
| | - Cuijuan Zhang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, 199 Ren’ai Road, Suzhou, 215123 Jiangsu China
| | - Xia Liu
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, 199 Ren’ai Road, Suzhou, 215123 Jiangsu China
| | - Guangquan Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, 150086 Heilongjiang China
| | - Hong-Tao Zhang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, 199 Ren’ai Road, Suzhou, 215123 Jiangsu China
- Suzhou Key Laboratory for Molecular Cancer Genetics, Suzhou, 215123 Jiangsu China
| |
Collapse
|
40
|
Shi X, Liu Q, Li N, Tu W, Luo R, Mei X, Ma Y, Xu W, Chu H, Jiang S, Du Z, Zhao H, Zhao L, Jin L, Wu W, Wang J. MiR-3606-3p inhibits systemic sclerosis through targeting TGF-β type II receptor. Cell Cycle 2018; 17:1967-1978. [PMID: 30145936 PMCID: PMC6224271 DOI: 10.1080/15384101.2018.1509621] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/26/2018] [Accepted: 07/28/2018] [Indexed: 12/24/2022] Open
Abstract
Systemic sclerosis (SSc) is a multisystemic fibrotic disease characterized by excessive collagen deposition and extracellular matrix synthesis. Though transforming growth factor-β (TGF-β) plays a fundamental role in the pathogenesis of SSc, the mechanism by which TGF-β signaling acts in SSc remains largely unclear. Here, we showed that TGF-β type II receptor (TGFBR2) was significantly upregulated in both human SSc dermal tissues and primary fibroblasts. In fibroblasts, siRNA-induced knockdown of TGFBR2 resulted in a reduction of p-SMAD2/3 levels and reduced production of type I collagen. Additionally, functional experiments revealed that downregulation of TGFBR2 yielded an anti-growth effect on fibroblasts through inhibiting cell cycle progression. Further studies showed that miR-3606-3p could directly target the 3'-UTR of TGFBR2 and significantly decrease the levels of both TGFBR2 mRNA and protein. Furthermore, SSc dermal tissues and primary fibroblasts contain significantly reduced amounts of miR-3606-3p, and the overexpression of miR-3606-3p in fibroblasts replicates the phenotype of TGFBR2 downregulation. Collectively, our findings demonstrated that increased TGFBR2 could be responsible for the hyperactive TGF-β signaling observed in SSc. Moreover, we identified a pivotal role for miR-3606-3p in SSc, which acts, at least partly, through the attenuation of TGF-β signaling via TGFBR2 repression, suggesting that the regulation of miR-3606-3p/TGFBR2 could be a promising therapeutic target that could improve the treatment strategy for fibrosis.
Collapse
Affiliation(s)
- Xiangguang Shi
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
- Human Phenome Institute, Fudan University, Shanghai, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qingmei Liu
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Na Li
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Wenzhen Tu
- Division of Rheumatology, Shanghai TCM-Integrated Hospital, Shanghai, China
| | - Ruoyu Luo
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Xueqian Mei
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Yanyun Ma
- Human Phenome Institute, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Weihong Xu
- The Clinical Laboratory of Shanghai Tongren Hosipital, Jiaotong University, Shanghai, China
| | - Haiyan Chu
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Shuai Jiang
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Zhimin Du
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Han Zhao
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Liang Zhao
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, P. R. China
- Human Phenome Institute, Fudan University, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Wang S, Lan F, Xia Y. lncRA ANCR Inhibits Non-Small Cell Lung Cancer Cell Migration and Invasion by Inactivating TGF-β Pathway. Med Sci Monit 2018; 24:6002-6009. [PMID: 30154397 PMCID: PMC6126415 DOI: 10.12659/msm.911492] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background lncRNA ANCR is proved to be a tumor suppressor gene only in colorectal cancer and breast cancer. Our study aimed to explore the possible involvement of ACNR in non-small cell lung cancer (NSCLC). Material/Methods In this study, we first detected the expression of ACNR in lung biopsies and plasma of both NSCLC patients and healthy controls. The diagnostic value of ANCR for NSCLC was analyzed by ROC curve analysis. Follow-up data of NSCLC patients was analyzed and the prognostic value of ANCR was analyzed by survival curve analysis. ANCR expression vector was transfected into cells of human NSCLC cell lines, and the effects on cell migration and invasion were explored by Transwell migration and invasion assays, respectively. TGF-β1 expression after ANCR overexpression was detected by Western blot analysis. Results ANCR was significantly downregulated in NSCLC patients compared with healthy controls in lung biopsies and plasma. Downregulated expression of ANCR distinguishes NSCLC patients from healthy controls and low NSCLC expression level indicates shorter postoperative survival time of NSCLC patients. ANCR overexpression inhibited NSCLC cell migration and invasion and downregulated TGF-β1 expression, while TGF-β1 treatment showed no significant effects on ANCR expression but promoted NSCLC cell migration and invasion. In addition, TGF-β1 treatment also attenuated the inhibitory effects of ANCR overexpression on NSCLC cell migration and invasion. Conclusions ANCR can inhibit NSCLC cell migration and invasion by downregulating TGF-β1 expression.
Collapse
Affiliation(s)
- Shaobin Wang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Fen Lan
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Yang Xia
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
42
|
Trissal MC, Wong TN, Yao JC, Ramaswamy R, Kuo I, Baty J, Sun Y, Jih G, Parikh N, Berrien-Elliott MM, Fehniger TA, Ley TJ, Maillard I, Reddy PR, Link DC. MIR142 Loss-of-Function Mutations Derepress ASH1L to Increase HOXA Gene Expression and Promote Leukemogenesis. Cancer Res 2018; 78:3510-3521. [PMID: 29724719 PMCID: PMC6030481 DOI: 10.1158/0008-5472.can-17-3592] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/12/2018] [Accepted: 04/23/2018] [Indexed: 12/22/2022]
Abstract
Point mutations in the seed sequence of miR-142-3p are present in a subset of acute myelogenous leukemia (AML) and in several subtypes of B-cell lymphoma. Here, we show that mutations associated with AML result both in loss of miR-142-3p function and in decreased miR-142-5p expression. Mir142 loss altered the hematopoietic differentiation of multipotent hematopoietic progenitors, enhancing their myeloid potential while suppressing their lymphoid potential. During hematopoietic maturation, loss of Mir142 increased ASH1L protein expression and consequently resulted in the aberrant maintenance of Hoxa gene expression in myeloid-committed hematopoietic progenitors. Mir142 loss also enhanced the disease-initiating activity of IDH2-mutant hematopoietic cells in mice. Together these data suggest a novel model in which miR-142, through repression of ASH1L activity, plays a key role in suppressing HOXA9/A10 expression during normal myeloid differentiation. AML-associated loss-of-function mutations of MIR142 disrupt this negative signaling pathway, resulting in sustained HOXA9/A10 expression in myeloid progenitors/myeloblasts and ultimately contributing to leukemic transformation.Significance: These findings provide mechanistic insights into the role of miRNAs in leukemogenesis and hematopoietic stem cell function. Cancer Res; 78(13); 3510-21. ©2018 AACR.
Collapse
Affiliation(s)
- Maria C Trissal
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Terrence N Wong
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Juo-Chin Yao
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Rahul Ramaswamy
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Iris Kuo
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Jack Baty
- Division of Biostatistics, Washington University, St. Louis, Missouri
| | - Yaping Sun
- Division of Hematology-Oncology, University of Michigan, Ann Arbor, Michigan
| | - Gloria Jih
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | - Nishi Parikh
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | | | - Todd A Fehniger
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Timothy J Ley
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Ivan Maillard
- Division of Hematology-Oncology, University of Michigan, Ann Arbor, Michigan
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Pavan R Reddy
- Division of Hematology-Oncology, University of Michigan, Ann Arbor, Michigan
| | - Daniel C Link
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
43
|
Lee YY, Yarmishyn AA, Wang ML, Chen HY, Chiou SH, Yang YP, Lin CF, Huang PI, Chen YW, Ma HI, Chen MT. MicroRNA-142-3p is involved in regulation of MGMT expression in glioblastoma cells. Cancer Manag Res 2018; 10:775-785. [PMID: 29695934 PMCID: PMC5903834 DOI: 10.2147/cmar.s157261] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most malignant brain tumor, and there is no effective treatment strategy. Patients with GBM have a median overall survival of only 14.6 months. Current treatment consists of safe and maximal surgical excision, followed by concurrent chemoradiotherapy and maintenance chemotherapy. There are several obstacles that hinder the effectiveness of this aggressive treatment. Temozolomide (TMZ) is an oral alkylating drug that acts through alkylating the O6 position of guanine in DNA that leads to cell death. However, the expression and enzymatic activity of the DNA repair protein MGMT limits the therapeutic benefit from treatment with TMZ. MGMT reduces the efficacy of alkylating drugs by removing the methyl or alkyl group from damaged O6-methylguanine. Expression levels of MGMT play an important role in the outcome of GBM patients. miRNAs are a group of small regulatory RNAs that control target gene expression by binding to mRNAs. miR-142-3p has been found to be an important factor in the development and maintenance of the oncogenic state. Results In this study, we sought to investigate whether miR-142-3p can regulate MGMT gene expression in GBM cells. Here, we show that miR-142-3p downregulates MGMT expression through binding to the 3′-UTR of MGMT mRNA, thus affecting protein translation. Responsiveness to TMZ was significantly enhanced after transfection with miR-142-3p. Overexpression of miR-142-3p also sensitized GBM cells to alkylating drugs. Conclusion Above all, our findings demonstrate that miR-142-3p plays a critical role in regulating MGMT expression, has great potential for future clinical applications, and acts as a new diagnostic marker for this intractable disease.
Collapse
Affiliation(s)
- Yi-Yen Lee
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital.,Faculty of Medicine, National Yang-Ming University
| | - Aliaksandr A Yarmishyn
- Institute of Pharmacology, National Yang-Ming University.,Department of Medical Research, Taipei Veterans General Hospital
| | - Mong-Lien Wang
- Institute of Pharmacology, National Yang-Ming University.,Department of Medical Research, Taipei Veterans General Hospital
| | - Hsiao-Yun Chen
- Department of Medical Research, Taipei Veterans General Hospital.,Institute of Clinical Medicine, National Yang Ming University
| | - Shih-Hwa Chiou
- Institute of Pharmacology, National Yang-Ming University.,Department of Medical Research, Taipei Veterans General Hospital.,Institute of Clinical Medicine, National Yang Ming University
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital.,Institute of Clinical Medicine, National Yang Ming University
| | - Chun-Fu Lin
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital.,Faculty of Medicine, National Yang-Ming University
| | - Pin-I Huang
- Faculty of Medicine, National Yang-Ming University.,Cancer Center, Radiation Oncology Division, Taipei Veterans General Hospital
| | - Yi-Wei Chen
- Faculty of Medicine, National Yang-Ming University.,Cancer Center, Radiation Oncology Division, Taipei Veterans General Hospital
| | - Hsin-I Ma
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Teh Chen
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital.,Faculty of Medicine, National Yang-Ming University
| |
Collapse
|
44
|
MicroRNA‑142‑5p modulates breast cancer cell proliferation and apoptosis by targeting phosphatase and tensin homolog. Mol Med Rep 2018; 17:7529-7536. [PMID: 29620260 PMCID: PMC5983952 DOI: 10.3892/mmr.2018.8812] [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: 06/15/2017] [Accepted: 01/12/2018] [Indexed: 12/11/2022] Open
Abstract
A total of 60 breast cancer (BC) tissues and adjacent healthy tissues from patients who underwent surgery in Renmin Hospital of Wuhan University were collected for analysis in the present study. Results from reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) demonstrated that, compared with the adjacent healthy tissues, the expression levels of microRNA (miR)‑142‑5p were significantly elevated in BC tissues. Bioinformatics analysis was performed using TargetScan for the prediction of potential target sites that matched the seed region of miR‑142‑5p; phosphatase and tensin homolog (PTEN) exhibited the highest score and was selected for further analysis. Results of RT‑qPCR analysis demonstrated that, compared with the adjacent healthy tissues, the mRNA expression levels of PTEN were significantly decreased in breast cancer tissues. miR‑142‑5p and PTEN expression levels were positively and negatively associated, respectively, with patient tumor size and metastasis. MDA‑MB‑231 cells were divided into three groups including the Control group, the miR‑NC inhibitor group and the miR‑142‑5p inhibitor group. As for alterations in cell behavior, including cell viability and cell apoptosis, and protein expression levels, there were no significant differences between Control and miR‑NC inhibitor groups. MTT assay results revealed that, compared with Control and miR‑NC inhibitor groups, miR‑142‑5p inhibitor reduced MDA‑MB‑231 cell proliferation. Flow cytometric analysis demonstrated that, compared with Control and miR‑NC inhibitor groups, miR‑142‑5p inhibitor treatment induced MDA‑MB‑231 cell apoptosis. Western blotting results demonstrated that, compared with Control and miR‑NC inhibitor groups, miR‑142‑5p inhibitor treatment significantly increased the expression of PTEN, reduced the activation of phosphatidylinositol‑4,5‑bisphosphate 3‑kinase/RACα serine/threonine‑protein kinase signaling. Finally, PTEN was demonstrated to interact with miR‑142‑5p from the results of dual‑luciferase reporter assay in the present study. The findings of the present study suggested that miR‑142‑5p may be a potential therapeutic target for the future investigations and insights for breast cancer.
Collapse
|
45
|
Wang Y, Cao Z, Wang L, Liu S, Cai J. Downregulation of microRNA-142-3p and its tumor suppressor role in gastric cancer. Oncol Lett 2018; 15:8172-8180. [PMID: 29849811 DOI: 10.3892/ol.2018.8330] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/23/2018] [Indexed: 12/17/2022] Open
Abstract
An increasing number of studies indicate that microRNAs (miRNAs) may exert an oncogenic or tumor suppressive role in diverse types of cancer. MicroRNA (miR)-142-3p has been detected to be downregulated in a number of cancer types, and it may function as a tumor suppressor. However, the expression profile and potential role of miR-142-3p in gastric cancer remain unknown. In the present study, the expression of miR-142-3p in numerous gastric cancer samples was investigated. It was observed that miR-142-3p was markedly downregulated in cancer tissues compared with normal tissues. Furthermore, a low expression level of miR-142-3p was associated with higher tumor stages. The overexpression of miR-142-3p was able to inhibit the proliferation, invasion and migration of gastric cancer cells. A further investigation into the mechanism underlying the effect of miR-142-3p identified cyclin T2 (CCNT2) as a target of miR-142-3p in gastric cancers. miR-142-3p may exert its tumor suppressor role partially by downregulating CCNT2. These results suggested that the abnormal downregulation of miR-142-3p and the subsequent increase in CCNT2 expression may have an important role in gastric cancer carcinogenesis.
Collapse
Affiliation(s)
- Yi Wang
- Department of VIP, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, P.R. China
| | - Zhidong Cao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China.,Department of Orthopedics, The Emergency Medical Center of Chongqing, Chongqing 400014, P.R. China
| | - Lanlan Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100005, P.R. China
| | - Siqi Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100005, P.R. China
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, P.R. China
| |
Collapse
|
46
|
Li C, Wan L, Liu Z, Xu G, Wang S, Su Z, Zhang Y, Zhang C, Liu X, Lei Z, Zhang HT. Long non-coding RNA XIST promotes TGF-β-induced epithelial-mesenchymal transition by regulating miR-367/141-ZEB2 axis in non-small-cell lung cancer. Cancer Lett 2018; 418:185-195. [PMID: 29339211 DOI: 10.1016/j.canlet.2018.01.036] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 12/19/2022]
Abstract
Growing evidence shows that lncRNA XIST functions as an oncogene accelerating tumor progression. Transforming growth factor β (TGF-β)-induced epithelial-mesenchymal transition (EMT) plays a key role in tumor metastasis. However, it is still unclear whether lncRNA XIST is implicated in TGF-β-induced EMT and influences cell invasion and metastasis in non-small-cell lung cancer (NSCLC). Here, we observed increased expression of lncRNA XIST and ZEB2 mRNA in metastatic NSCLC tissues. Knockdown of lncRNA XIST inhibited ZEB2 expression, and repressed TGF-β-induced EMT and NSCLC cell migration and invasion. Being in consistent with the in vitro findings, the in vivo experiment of metastasis showed that knockdown of lncRNA XIST inhibited pulmonary metastasis of NSCLC cells in mice. In addition, knockdown of ZEB2 expression can inhibit TGF-β-induced EMT and NSCLC cell migration and invasion. Mechanistically, lncRNA XIST and ZEB2 were targets of miR-367 and miR-141. Furthermore, both miR-367 and miR-141 expression can be upregulated by knockdown of lncRNA XIST. Taken together, our study reveals that lncRNA XIST can promote TGF-β-induced EMT and cell invasion and metastasis by regulating miR-367/miR-141-ZEB2 axis in NSCLC.
Collapse
Affiliation(s)
- Chang Li
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Genetics, School of Biology and Basic Medical Science, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Liang Wan
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Genetics, School of Biology and Basic Medical Science, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zeyi Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Guangquan Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, China
| | - Shengjie Wang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Genetics, School of Biology and Basic Medical Science, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang, 222000, China
| | - Zhiyue Su
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Genetics, School of Biology and Basic Medical Science, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yingxi Zhang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Genetics, School of Biology and Basic Medical Science, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Cuijuan Zhang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Genetics, School of Biology and Basic Medical Science, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xia Liu
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Genetics, School of Biology and Basic Medical Science, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhe Lei
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Genetics, School of Biology and Basic Medical Science, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Suzhou Key Laboratory for Molecular Cancer Genetics, Suzhou, Jiangsu, 215123, China.
| | - Hong-Tao Zhang
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Department of Genetics, School of Biology and Basic Medical Science, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Suzhou Key Laboratory for Molecular Cancer Genetics, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
47
|
Yang Z, He J, Gao P, Niu Y, Zhang J, Wang L, Liu M, Wei X, Liu C, Zhang C, Wang W, Du J, Li H, Hu W, Sun G. miR-769-5p suppressed cell proliferation, migration and invasion by targeting TGFBR1 in non-small cell lung carcinoma. Oncotarget 2017; 8:113558-113570. [PMID: 29371929 PMCID: PMC5768346 DOI: 10.18632/oncotarget.23060] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/14/2017] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs (miRNAs) are key regulators of multiple cancers, including non-small cell lung carcinoma (NSCLC). The aim of this study was to determine the expression pattern of miR-769-5p in NSCLC and to investigate its biological role during tumorigenesis. We showed that miR-769-5p was significantly downregulated and predicted poor prognosis in NSCLC compared with corresponding normal tissues. We then investigated its function and found that miR-769-5p significantly inhibited cell proliferation, migration and invasion in vitro and reduced tumor growth and metastasis in vivo. Furthermore, we explored the molecular mechanisms by which miR-769-5p contributes to NSCLC suppression and identified TGFBR1 as a direct target gene of miR-769-5p. Finally, we showed that TGFBR1 had opposite effects to those of miR-769-5p on lung cancer cells, suggesting that miR-769-5p might inhibit lung tumorigenesis by silencing TGFBR1. Taken together, our results demonstrated that miR-769-5p plays a pivotal role in NSCLC by inhibiting cell proliferation, migration and invasion by targeting TGFBR1.
Collapse
Affiliation(s)
- Zhao Yang
- 1 Department of Radiation Oncology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Jin He
- 2 Department of Hepatobiliary Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Cencer, Tianjin, China
| | - Peng Gao
- 1 Department of Radiation Oncology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Yi Niu
- 1 Department of Radiation Oncology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Jie Zhang
- 3 Department of Pathology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Lei Wang
- 3 Department of Pathology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Meiyue Liu
- 1 Department of Radiation Oncology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Xiaomei Wei
- 1 Department of Radiation Oncology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Chunling Liu
- 3 Department of Pathology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Chao Zhang
- 1 Department of Radiation Oncology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Wei Wang
- 1 Department of Radiation Oncology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Jiayi Du
- 3 Department of Pathology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Hongmin Li
- 3 Department of Pathology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Wanning Hu
- 1 Department of Radiation Oncology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| | - Guogui Sun
- 1 Department of Radiation Oncology, North China University of Science and Technology Affiliated People's Hospital, Tangshan, China
| |
Collapse
|
48
|
Yu Q, Xiang L, Yin L, Liu X, Yang D, Zhou J. Loss-of-function of miR-142 by hypermethylation promotes TGF-β-mediated tumour growth and metastasis in hepatocellular carcinoma. Cell Prolif 2017; 50:e12384. [PMID: 28963738 PMCID: PMC6529086 DOI: 10.1111/cpr.12384] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/18/2017] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Hypermethylation-induced epigenetic silencing of tumour suppressor genes (TSGs) are frequent events during carcinogenesis. MicroRNA-142 (miR-142) is found to be dysregulated in cancer patients to participate into tumour growth, metastasis and angiogenesis. However, the tumour suppressive role of miR-142 and the status of methylation are not fully understood in hepatocellular carcinoma (HCC). METHODS Hepatocellular carcinoma tissues and corresponding non-neoplastic tissues were collected. The expression and function of miR-142 and TGF-β in two HCC cell lines were determined. The miRNA-mRNA network of miR-142 was analysed in HCC cell lines. RESULTS We found that the miR-142 expression was reduced in tumour tissues and two HCC cell lines HepG2 and SMMC7721, which correlated to higher TNM stage, metastasis and differentiation. Moreover, miR-142 was identified to directly target and inhibit transforming growth factor β (TGF-β), leading to decreased cell vitality, proliferation, EMT and the ability of pro-angiogenesis in TGF-β-dependent manner. Interestingly, the status of methylation of miR-142 was analysed and the results found the hypermethylated miR-142 in tumour patients and cell lines. The treatment of methylation inhibitor 5-Aza could restore the expression of miR-142 to suppress the TGF-β expression, which impaired TGF-β-induced tumour growth. CONCLUSION These findings implicated that miR-142 was a tumour suppressor gene in HCC and often hyermethylated to increase TGF-β-induced development of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Qiangfeng Yu
- Department of Hepatobiliary SurgeryNanfang Hospital Affiliated to Southern Medical UniversityGuangzhouChina
- Department of Hepatobiliary SurgeryThe Second Hospital of LongyanLongyanChina
| | - Leyang Xiang
- Department of Hepatobiliary SurgeryNanfang Hospital Affiliated to Southern Medical UniversityGuangzhouChina
| | - Libo Yin
- Department of Hepatobiliary SurgeryChangzhou No. 2 People's HospitalNanjing Medical UniversityChangzhouJiangsuChina
| | - Xincheng Liu
- The Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Dinghua Yang
- Department of Hepatobiliary SurgeryNanfang Hospital Affiliated to Southern Medical UniversityGuangzhouChina
| | - Jianyin Zhou
- Department of Hepatobiliary and Pancreatic SurgeryZhongshan HospitalXiamen UniversityXiamenChina
| |
Collapse
|
49
|
Genome-wide Profiling of Urinary Extracellular Vesicle microRNAs Associated With Diabetic Nephropathy in Type 1 Diabetes. Kidney Int Rep 2017; 3:555-572. [PMID: 29854963 PMCID: PMC5976846 DOI: 10.1016/j.ekir.2017.11.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/15/2017] [Accepted: 11/27/2017] [Indexed: 01/01/2023] Open
Abstract
Introduction Diabetic nephropathy (DN) is a form of progressive kidney disease that often leads to end-stage renal disease (ESRD). It is initiated by microvascular complications due to diabetes. Although microalbuminuria (MA) is the earliest clinical indication of DN among patients with type 1 diabetes (T1D), it lacks the sensitivity and specificity to detect the early onset of DN. Recently, microRNAs (miRNAs) have emerged as critical regulators in diabetes as well as various forms of kidney disease, including renal fibrosis, acute kidney injury, and progressive kidney disease. Additionally, circulating extracellular miRNAs, especially miRNAs packaged in extracellular vesicles (EVs), have garnered significant attention as potential noninvasive biomarkers for various diseases and health conditions. Methods As part of the University of Pittsburgh Epidemiology of Diabetes Complications (EDC) study, urine was collected from individuals with T1D with various grades of DN or MA (normal, overt, intermittent, and persistent) over a decade at prespecified intervals. We isolated EVs from urine and analyzed the small-RNA using NextGen sequencing. Results We identified a set of miRNAs that are enriched in urinary EVs compared with EV-depleted samples, and identified a number of miRNAs showing concentration changes associated with DN occurrence, MA status, and other variables, such as hemoglobin A1c levels. Conclusion Many of the miRNAs associated with DN occurrence or MA status directly target pathways associated with renal fibrosis (including transforming growth factor-β and phosphatase and tensin homolog), which is one of the major contributors to the pathology of DN. These miRNAs are potential biomarkers for DN and MA.
Collapse
|
50
|
Joo JH, Hong IK, Kim NK, Choi E. Trichosanthes kirilowii extract enhances repair of UVB radiation‑induced DNA damage by regulating BMAL1 and miR‑142‑3p in human keratinocytes. Mol Med Rep 2017; 17:877-883. [PMID: 29115465 PMCID: PMC5780168 DOI: 10.3892/mmr.2017.7932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 05/19/2017] [Indexed: 12/31/2022] Open
Abstract
Ultraviolet B (UVB) radiation induces DNA damage, oxidative stress and inflammation, and suppresses the immune system in the skin, which collectively contribute to skin aging and carcinogenesis. The DNA damage response, including DNA repair, can be regulated by the circadian clock and microRNA (miRNA) expression. The aim of the present study was to evaluate the reparative action of Trichosanthes kirilowii extract (TKE) against UVB irradiation-induced DNA damage in human keratinocytes. TKE demonstrated low cytotoxicity in normal HaCaT keratinocytes at low doses (up to 100 µg/ml). The results of a comet assay revealed that TKE enhanced the repair of UVB-induced DNA damage. TKE significantly upregulated the expression of the core clock protein, brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein-1 (BMAL1), and downregulated the expression of miRNA (miR)-142-3p, as demonstrated using western blotting and the reverse transcription-quantitative polymerase chain reaction. Furthermore, the suppression of miR-142-3p by a specific inhibitor positively correlated with the repair activity. Overall, the data obtained demonstrated that TKE enhanced the repair of UVB-induced DNA damage by regulating the expression of BMAL1 and miR-142-3p. Consequently, TKE can be considered a potential candidate for the treatment of skin diseases associated with UVB-induced damage.
Collapse
Affiliation(s)
- Ji-Hye Joo
- Research and Development Center, Greensolutions Co., Ltd., Chuncheon, Gangwon 24232, Republic of Korea
| | - In-Kee Hong
- Research and Development Center, Radiant Co., Ltd., Chuncheon, Gangwon 24398, Republic of Korea
| | - Nam Kyoung Kim
- Research and Development Center, Greensolutions Co., Ltd., Chuncheon, Gangwon 24232, Republic of Korea
| | - Eunmi Choi
- Research and Development Center, Greensolutions Co., Ltd., Chuncheon, Gangwon 24232, Republic of Korea
| |
Collapse
|