51
|
Wang H, Xue W, Ouyang W, Jiang X, Jiang X. miR-23a-3p/SIX1 regulates glucose uptake and proliferation through GLUT3 in head and neck squamous cell carcinomas. J Cancer 2020; 11:2529-2539. [PMID: 32201523 PMCID: PMC7066005 DOI: 10.7150/jca.30995] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/15/2020] [Indexed: 02/06/2023] Open
Abstract
SIX1 overexpression has been reported in several cancers. However, its involvement in head and neck squamous cell carcinoma (HNSCC) remains unclear. In this study we investigated the clinical significance and biological roles of SIX1 in HNSCC. SIX1 expression was upregulated in HNSCC and correlated with TNM stage and nodal metastasis. Analysis of TCGA dataset demonstrated that high SIX1 expression correlated with poor patient prognosis. Overexpression of SIX1 in the Fadu cell line upregulated cell proliferation, colony formation, glucose uptake and ATP production. In contrast, SIX1 depletion in the Detroit562 cell line downregulated cell proliferation, colony formation, glucose uptake and ATP production. We analyzed a series of genes involved in glucose metabolism and found that SIX1 overexpression upregulated GLUT3, an important glucose transporter, at both mRNA and protein levels. Using the TRANSFAC database, we found that SIX1 had potential binding sites on the GLUT3 promoter, which was validated by chromatin immunoprecipitation (ChIP) assays. Next, we focused on miR-23a-3p, which could target SIX1 in HNSCC cells. The miR-23a-3p mimic downregulated SIX1 expression while the miR-23a-3p inhibitor upregulated SIX1 expression. The binding of miR-23a-3p to the 3'-UTR of SIX1 was confirmed using the luciferase reporter assay. Analysis of TCGA dataset showed a negative correlation between the miR-23a-3p and SIX1. Furthermore, the miR-23a-3p mimic inhibited cell proliferation, ATP production and glucose uptake, which could be rescued by transfection with the SIX1 plasmid. In summary, our study demonstrated that SIX1 facilitated HNSCC cell growth through regulation of GLUT3 and glucose uptake. miR-23a-3p targeted the SIX1/GLUT3 axis and suppressed glucose uptake and proliferation in HNSCC.
Collapse
Affiliation(s)
- Hongming Wang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Weishuang Xue
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wunyu Ouyang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaoze Jiang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuejun Jiang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
52
|
Parichatikanond W, Luangmonkong T, Mangmool S, Kurose H. Therapeutic Targets for the Treatment of Cardiac Fibrosis and Cancer: Focusing on TGF-β Signaling. Front Cardiovasc Med 2020; 7:34. [PMID: 32211422 PMCID: PMC7075814 DOI: 10.3389/fcvm.2020.00034] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/24/2020] [Indexed: 12/22/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a common mediator of cancer progression and fibrosis. Fibrosis can be a significant pathology in multiple organs, including the heart. In this review, we explain how inhibitors of TGF-β signaling can work as antifibrotic therapy. After cardiac injury, profibrotic mediators such as TGF-β, angiotensin II, and endothelin-1 simultaneously activate cardiac fibroblasts, resulting in fibroblast proliferation and migration, deposition of extracellular matrix proteins, and myofibroblast differentiation, which ultimately lead to the development of cardiac fibrosis. The consequences of fibrosis include a wide range of cardiac disorders, including contractile dysfunction, distortion of the cardiac structure, cardiac remodeling, and heart failure. Among various molecular contributors, TGF-β and its signaling pathways which play a major role in carcinogenesis are considered master fibrotic mediators. In fact, recently the inhibition of TGF-β signaling pathways using small molecule inhibitors, antibodies, and gene deletion has shown that the progression of several cancer types was suppressed. Therefore, inhibitors of TGF-β signaling are promising targets for the treatment of tissue fibrosis and cancers. In this review, we discuss the molecular mechanisms of TGF-β in the pathogenesis of cardiac fibrosis and cancer. We will review recent in vitro and in vivo evidence regarding antifibrotic and anticancer actions of TGF-β inhibitors. In addition, we also present available clinical data on therapy based on inhibiting TGF-β signaling for the treatment of cancers and cardiac fibrosis.
Collapse
Affiliation(s)
| | - Theerut Luangmonkong
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Hitoshi Kurose
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
53
|
Xiao Y, Qing J, Li B, Chen L, Nong S, Yang W, Tang X, Chen Z. TIM-3 Participates in the Invasion and Metastasis of Nasopharyngeal Carcinoma via SMAD7/SMAD2/SNAIL1 Axis-Mediated Epithelial-Mesenchymal Transition. Onco Targets Ther 2020; 13:1993-2006. [PMID: 32184631 PMCID: PMC7064287 DOI: 10.2147/ott.s237222] [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: 11/04/2019] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Background T-cell immunoglobulin and mucin domain-containing molecule-3 (TIM-3) was originally found to negatively regulate immune response and mediate immune escape in tumors. Subsequently, an increasing body of evidence has shown that TIM-3 exerts positive functions in the development and progression of several tumors. However, the role of TIM-3 in nasopharyngeal carcinoma (NPC) remains unknown. Methods Data from the Cancer Genome Atlas-head and neck squamous cell carcinoma and immunohistochemistry were analyzed to compare the expression of TIM-3 in NPC and non-cancerous nasopharyngitis tissues. Cell proliferation was evaluated using the Cell counting kit-8 in vitro and xenograft experiment in nude mice in vivo. Flow cytometry was used to evaluate the cell cycle. The migration and invasion of NPC cells were assessed through wound healing and Transwell assays. In addition, Western blotting was used to analyze the expression of specific proteins. Results Higher expression of TIM-3 was detected in NPC tissues than normal nasopharyngeal tissues and positively correlated with the clinical stage and T classification; however, it was not correlated with gender, age, and N classification. Furthermore, overexpression of TIM-3 using lentiviral vectors increased the malignancy of 6-10B and CNE-2 cell lines that lowly express TIM-3, by promoting cell proliferation, migration, and invasion in vitro and in vivo. In addition, overexpression of TIM-3 was associated with upregulation of matrix metalloproteinase 9 (MMP9) and MMP2, and led to epithelial-mesenchymal transition (EMT) by increasing the levels of mesenchymal markers (ie, N-cadherin, Vimentin) and decreasing those of the epithelial marker E-cadherin. Further study showed that SMAD7 was downregulated in the TIM-3 overexpression group. Relatively, phosphorylated SMAD2 and downstream molecule SNAIL1 were also upregulated in this group. Conclusion TIM-3 exerts a tumor-promoting function in NPC by mediating changes in the SMAD7/SMAD2/SNAIL1 axis. These findings provide a new idea for the study of invasion, metastasis, and treatment of NPC.
Collapse
Affiliation(s)
- Yangyang Xiao
- Department of Clinical Laboratory, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China
| | - Jilin Qing
- Center for Reproductive Medicine and Genetics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China
| | - Baoxuan Li
- Department of Ophthalmology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, People's Republic of China
| | - Liuyan Chen
- Department of Clinical Laboratory, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China
| | - Shengzhou Nong
- Department of Clinical Laboratory, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China
| | - Wenhui Yang
- Department of Clinical Laboratory, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China
| | - Xiaogang Tang
- Department of Intensive Care Unit, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China
| | - Zhizhong Chen
- Department of Clinical Laboratory, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China
| |
Collapse
|
54
|
Giannoudis A, Clarke K, Zakaria R, Varešlija D, Farahani M, Rainbow L, Platt-Higgins A, Ruthven S, Brougham KA, Rudland PS, Jenkinson MD, Young LS, Falciani F, Palmieri C. A novel panel of differentially-expressed microRNAs in breast cancer brain metastasis may predict patient survival. Sci Rep 2019; 9:18518. [PMID: 31811234 PMCID: PMC6897960 DOI: 10.1038/s41598-019-55084-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer brain metastasis (BCBM) is an area of unmet clinical need. MicroRNAs (miRNAs) have been linked to the metastatic process in breast cancer (BC). In this study, we aim to determine differentially-expressed miRNAs utilising primary BCs that did not relapse (BCNR, n = 12), primaries that relapsed (BCR) and their paired (n = 40 pairs) brain metastases (BM) using the NanoString™ nCounter™ miRNA Expression Assays. Significance analysis of microarrays identified 58 and 11 differentially-expressed miRNAs between BCNR vs BCR and BCR vs BM respectively and pathway analysis revealed enrichment for genes involved in invasion and metastasis. Four miRNAs, miR-132-3p, miR-199a-5p, miR-150-5p and miR-155-5p, were differentially-expressed within both cohorts (BCNR-BCR, BCR-BM) and receiver-operating characteristic curve analysis (p = 0.00137) and Kaplan-Meier survival method (p = 0.0029, brain metastasis-free survival; p = 0.0007, overall survival) demonstrated their potential use as prognostic markers. Ingenuity pathway enrichment linked them to the MET oncogene, and the cMET protein was overexpressed in the BCR (p < 0.0001) and BM (p = 0.0008) cases, compared to the BCNRs. The 4-miRNAs panel identified in this study could be potentially used to distinguish BC patients with an increased risk of developing BCBM and provide potential novel therapeutic targets, whereas cMET-targeting warrants further investigation in the treatment of BCBM.
Collapse
Affiliation(s)
- Athina Giannoudis
- Institute of Translational Medicine, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Kim Clarke
- Computational Biology Facility, University of Liverpool, Liverpool, UK
| | - Rasheed Zakaria
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Damir Varešlija
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mosavar Farahani
- Institute of Translational Medicine, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Lucille Rainbow
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | | | - Stuart Ruthven
- Department of Pathology, Royal Liverpool University Hospital, Liverpool, UK
| | | | - Philip S Rudland
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Michael D Jenkinson
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
- Institute of Translational Medicine, Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Leonie S Young
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Carlo Palmieri
- Institute of Translational Medicine, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
- The Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, UK.
| |
Collapse
|
55
|
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
|
56
|
Abolghasemi M, Tehrani SS, Yousefi T, Karimian A, Mahmoodpoor A, Ghamari A, Jadidi-Niaragh F, Yousefi M, Kafil HS, Bastami M, Edalati M, Eyvazi S, Naghizadeh M, Targhazeh N, Yousefi B, Safa A, Majidinia M, Rameshknia V. MicroRNAs in breast cancer: Roles, functions, and mechanism of actions. J Cell Physiol 2019; 235:5008-5029. [PMID: 31724738 DOI: 10.1002/jcp.29396] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022]
Abstract
Breast cancer is one of the most lethal malignancies in women in the world. Various factors are involved in the development and promotion of the malignancy; most of them involve changes in the expression of certain genes, such as microRNAs (miRNAs). MiRNAs can regulate signaling pathways negatively or positively, thereby affecting tumorigenesis and various aspects of cancer progression, particularly breast cancer. Besides, accumulating data demonstrated that miRNAs are a novel tool for prognosis and diagnosis of breast cancer patients. Herein, we will review the roles of these RNA molecules in several important signaling pathways, such as transforming growth factor, Wnt, Notch, nuclear factor-κ B, phosphoinositide-3-kinase/Akt, and extracellular-signal-regulated kinase/mitogen activated protein kinase signaling pathways in breast cancer.
Collapse
Affiliation(s)
- Maryam Abolghasemi
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Iran.,Student Research Committee, Babol University of medical sciences, Babol, Iran
| | - Sadra Samavarchi Tehrani
- Departmant of Clinical Biochemistry, Tehran University of Medical Sciences, Tehran, Iran.,Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Tooba Yousefi
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Iran.,Student Research Committee, Babol University of medical sciences, Babol, Iran
| | - Ansar Karimian
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Iran.,Student Research Committee, Babol University of medical sciences, Babol, Iran
| | - Ata Mahmoodpoor
- Anesthesiology Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aliakbar Ghamari
- Anesthesiology Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Bastami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Edalati
- Department of Laboratory Sciences, Paramedical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Eyvazi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Naghizadeh
- Departmant of Clinical Biochemistry, Tehran University of Medical Sciences, Tehran, Iran.,Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloufar Targhazeh
- Student Research Committee, Babol University Of Medical Sciences, Babol, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Safa
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Vahid Rameshknia
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Medicine, Islamic Azad University, Tabriz, Iran
| |
Collapse
|
57
|
Cantini L, Bertoli G, Cava C, Dubois T, Zinovyev A, Caselle M, Castiglioni I, Barillot E, Martignetti L. Identification of microRNA clusters cooperatively acting on epithelial to mesenchymal transition in triple negative breast cancer. Nucleic Acids Res 2019; 47:2205-2215. [PMID: 30657980 PMCID: PMC6412120 DOI: 10.1093/nar/gkz016] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/17/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs play important roles in many biological processes. Their aberrant expression can have oncogenic or tumor suppressor function directly participating to carcinogenesis, malignant transformation, invasiveness and metastasis. Indeed, miRNA profiles can distinguish not only between normal and cancerous tissue but they can also successfully classify different subtypes of a particular cancer. Here, we focus on a particular class of transcripts encoding polycistronic miRNA genes that yields multiple miRNA components. We describe 'clustered MiRNA Master Regulator Analysis (ClustMMRA)', a fully redesigned release of the MMRA computational pipeline (MiRNA Master Regulator Analysis), developed to search for clustered miRNAs potentially driving cancer molecular subtyping. Genomically clustered miRNAs are frequently co-expressed to target different components of pro-tumorigenic signaling pathways. By applying ClustMMRA to breast cancer patient data, we identified key miRNA clusters driving the phenotype of different tumor subgroups. The pipeline was applied to two independent breast cancer datasets, providing statistically concordant results between the two analyses. We validated in cell lines the miR-199/miR-214 as a novel cluster of miRNAs promoting the triple negative breast cancer (TNBC) phenotype through its control of proliferation and EMT.
Collapse
Affiliation(s)
- Laura Cantini
- Institut Curie, 26 rue d'Ulm, F-75005 Paris, France.,PSL Research University, F-75005 Paris, France.,Inserm, U900, F-75005, Paris France.,Mines Paris Tech, F-77305 cedex Fontainebleau, France.,Computational Systems Biology Team, Institut de Biologie de l'Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Ecole Normale Supérieure, Paris Sciences et Lettres Research University, 75005 Paris, France
| | - Gloria Bertoli
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Italy
| | - Claudia Cava
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Italy
| | - Thierry Dubois
- Institut Curie, 26 rue d'Ulm, F-75005 Paris, France.,PSL Research University, F-75005 Paris, France.,Institut Curie, PSL Research University, Department of Translational Research, Breast Cancer Biology Group, Paris, France
| | - Andrei Zinovyev
- Institut Curie, 26 rue d'Ulm, F-75005 Paris, France.,PSL Research University, F-75005 Paris, France.,Inserm, U900, F-75005, Paris France.,Mines Paris Tech, F-77305 cedex Fontainebleau, France
| | - Michele Caselle
- Department of Physics and INFN, Università degli Studi di Torino, Turin, Italy
| | - Isabella Castiglioni
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Italy
| | - Emmanuel Barillot
- Institut Curie, 26 rue d'Ulm, F-75005 Paris, France.,PSL Research University, F-75005 Paris, France.,Inserm, U900, F-75005, Paris France.,Mines Paris Tech, F-77305 cedex Fontainebleau, France
| | - Loredana Martignetti
- Institut Curie, 26 rue d'Ulm, F-75005 Paris, France.,PSL Research University, F-75005 Paris, France.,Inserm, U900, F-75005, Paris France.,Mines Paris Tech, F-77305 cedex Fontainebleau, France
| |
Collapse
|
58
|
Li HL, Sun JJ, Ma H, Liu SJ, Li N, Guo SJ, Shi Y, Xu YY, Qi ZY, Wang YQ, Wang F, Guo RM, Liu D, Xue FX. MicroRNA-23a inhibits endometrial cancer cell development by targeting SIX1. Oncol Lett 2019; 18:3792-3802. [PMID: 31579409 PMCID: PMC6757317 DOI: 10.3892/ol.2019.10694] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/05/2019] [Indexed: 12/27/2022] Open
Abstract
The present study focused on exploring the inhibitory mechanism of microRNA (miR)-23a in endometrial cancer. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) was used to investigate miR-23a expression in endometrial tissues and endometrial cancer cells. A colony formation assay using crystal violet staining was performed to compare cell proliferation, while wound-healing and Transwell assays were performed to compare cell migration and invasion. Subsequently, bioinformatics and a luciferase reporter gene assay were used to investigate the effect of miR-23a on sine oculis homeobox homolog 1 (SIX1) expression, and the biological function of SIX1 was analyzed. Additionally, a nude mouse tumorigenicity assay was performed to test the inhibitory effect of miR-23a and Taxol® therapy in endometrial cancer. Finally, immunohistochemistry and RT-qPCR were used to explore the association between miR-23a and SIX1 expression in endometrial cancer tissues. miR-23a was underexpressed in endometrial cancer tissues compared with in para-carcinoma tissues, and the overexpression of miR-23a inhibited proliferation and invasion of endometrial cancer cells. Furthermore, SIX1 was demonstrated to be a downstream target of miR-23a, and miR-23a reduced SIX1 expression. Additionally, SIX1 inversely promoted cell proliferation, migration and invasion. In addition, the effects of reduced cell proliferation and increased cell invasion following miR-23a overexpression could be reversed by adding SIX1 to in vitro culture. Furthermore, the inhibitory effect of miR-23a and Taxol therapy, which reduced SIX1 expression in endometrial cancer, was demonstrated in vivo. Finally, a negative association between miR-23a and SIX1 expression was demonstrated in endometrial cancer tissues. The results of the present study revealed that miR-23a may inhibit endometrial cancer development by targeting SIX1.
Collapse
Affiliation(s)
- Hong-Lin Li
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Jun-Jie Sun
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Hui Ma
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Shen-Jia Liu
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Na Li
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Su-Jie Guo
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Yang Shi
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Yan-Ying Xu
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Zhi-Ying Qi
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Yu-Quan Wang
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Fang Wang
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Rui-Meng Guo
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Dong Liu
- Department of Obstetrics and Gynecology, The Secondary Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Feng-Xia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
59
|
Xia X, Lu H, Li C, Huang Y, Wang Y, Yang X, Zheng JC. miR-106b regulates the proliferation and differentiation of neural stem/progenitor cells through Tp53inp1-Tp53-Cdkn1a axis. Stem Cell Res Ther 2019; 10:282. [PMID: 31547867 PMCID: PMC6755702 DOI: 10.1186/s13287-019-1387-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/28/2019] [Accepted: 08/16/2019] [Indexed: 12/22/2022] Open
Abstract
Background Recent studies suggested that miR-17~106 family was involved in the regulation of neural stem/progenitor cells (NPCs). However, distinct function of each family member was reported in regulating stem cells within and without the brain. Hence, to investigate the roles of individual miRNAs in miR-17~106 family and mechanisms underlying their effects on neurogenesis is important to extend our understanding in the CNS development. Methods Here, we examined the influence of miR-106a/b on the proliferation, differentiation, and survival of embryonic NPCs using specific mimics and inhibitor. The targets of miR-106a/b were identified from miRNA target prediction database and confirmed by luciferase assay. Specific siRNAs were utilized to erase the effects of miR-106a/b on the expression levels of target genes. Results A positive correlation was observed between the temporal reduction of miR-106a/b expression levels and the decline of NPC pools in vivo and in vitro. The perturbation of miR-106’s function approaches revealed that miR-106b, but not miR-106a, facilitated the maintenance of NPCs and repressed the generation of both neuronal and glial cells, without preference to a particular lineage. No effect was observed for miR-106a/b in NPCs’ survival. The influence of miR-106b on NPCs’ proliferation and differentiation is likely achieved by directly inhibiting the expression of Tp53inp1 and Cdkn1a, key components of Tp53inp1-Tp53-Cdkn1a axis. Conclusion Our study demonstrated a novel axis, miR-106b-Tp53inp1-Tp53-Cdkn1a, in regulating the proliferation and differentiation of NPCs. Electronic supplementary material The online version of this article (10.1186/s13287-019-1387-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Hongfang Lu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Chunhong Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Yunlong Huang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072, China.,Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5930, USA
| | - Yi Wang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| | - Xiaoyu Yang
- Department of Anesthesiology, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, 200065, China
| | - Jialin C Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072, China. .,Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, 200092, China. .,Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5930, USA. .,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-5930, USA.
| |
Collapse
|
60
|
Pfau ML, Menard C, Cathomas F, Desland F, Kana V, Chan KL, Shimo Y, LeClair K, Flanigan ME, Aleyasin H, Walker DM, Bouchard S, Mack M, Hodes GE, Merad MM, Russo SJ. Role of Monocyte-Derived MicroRNA106b∼25 in Resilience to Social Stress. Biol Psychiatry 2019; 86:474-482. [PMID: 31101319 PMCID: PMC6717005 DOI: 10.1016/j.biopsych.2019.02.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 02/13/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Clinical studies suggest that heightened peripheral inflammation contributes to the pathogenesis of stress-related disorders, including major depressive disorder. However, the molecular mechanisms within peripheral immune cells that mediate enhanced stress vulnerability are not well known. Because microRNAs (miRs) are important regulators of immune response, we sought to examine their role in mediating inflammatory and behavioral responses to repeated social defeat stress (RSDS), a mouse model of stress vulnerability that produces susceptible and resilient phenotypes. METHODS We isolated Ly6chigh monocytes via fluorescence-activated cell sorting in the blood of susceptible and resilient mice following RSDS and profiled miR expression via quantitative real-time polymerase chain reaction. Bone marrow chimeric mice were generated to confirm a causal role of the miR-106b∼25 cluster in bone marrow-derived leukocytes in mediating stress resilience versus susceptibility. RESULTS We found that RSDS produces an increase in circulating Ly6chigh inflammatory monocytes in both susceptible and resilient mice. We next investigated whether intrinsic leukocyte posttranscriptional mechanisms contribute to individual differences in stress response and the resilient phenotype. Of the miRs profiled in our panel, eight were significantly regulated by RSDS within Ly6chigh monocytes, including miR-25-3p, a member of the miR-106b∼25 cluster. Selective knockout of the miR-106b∼25 cluster in peripheral leukocytes promoted behavioral resilience to RSDS. CONCLUSIONS Our results identify the miR-106b∼25 cluster as a key regulator of stress-induced inflammation and depression that may represent a novel therapeutic target for drug development.
Collapse
Affiliation(s)
- Madeline L Pfau
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Caroline Menard
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry and Neuroscience, Faculty of Medicine and Cervo Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fiona Desland
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Oncological Science, Tisch Cancer Institute and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Veronika Kana
- Department of Oncological Science, Tisch Cancer Institute and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kenny L Chan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yusuke Shimo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Katherine LeClair
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Meghan E Flanigan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hossein Aleyasin
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Deena M Walker
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sylvain Bouchard
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Georgia E Hodes
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Miriam M Merad
- Department of Oncological Science, Tisch Cancer Institute and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
61
|
Yin W, Chen J, Wang G, Zhang D. MicroRNA‑106b functions as an oncogene and regulates tumor viability and metastasis by targeting LARP4B in prostate cancer. Mol Med Rep 2019; 20:951-958. [PMID: 31173237 PMCID: PMC6625195 DOI: 10.3892/mmr.2019.10343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 03/07/2019] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer (PCa) is the most common malignancy among males worldwide, and is one of the leading causes of cancer‑related mortality. MicroRNAs (miRs) are a type of endogenous, noncoding RNA that serve a key role in pathological processes, and have been demonstrated to be involved in the formation and progression of PCa. Previous studies have reported that miR‑106b acts as an oncogene; however, the specific effects of miR‑106b on PCa have not been fully elucidated. The present study aimed to investigate the role and underlying molecular mechanisms of miR‑106b in the initiation and progression of PCa. In this study, miR‑106b was reported to be overexpressed and la‑related protein 4B (LARP4B) was downregulated in PCa tissues compared with paracancerous tissues. In addition, LARP4B was identified as a target gene of miR‑106b by bioinformatics prediction analysis and a dual luciferase reporter gene assay. Furthermore, MTT, wound healing and Transwell assays were performed to evaluate PCa cell viability, and migration and invasive abilities. The data revealed that inhibition of miR‑106b significantly suppressed the viability, migration and invasion of PCa cells. In addition, inhibition of miR‑106b significantly suppressed the mRNA and protein expression of cancer‑related genes, including matrix metalloproteinase‑2, cluster of differentiation 44 and Ki‑67, and increased that of the tumor suppressor, mothers against decapentaplegic homolog 2. Collectively, the findings of the present study indicated that miR‑106b may target LAR4B to inhibit cancer cell viability, migration and invasion, and may be considered as a novel therapeutic target in PCa.
Collapse
Affiliation(s)
- Weiqi Yin
- Department of Urology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Junfeng Chen
- Department of Urology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Guoyao Wang
- Department of Urology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Dongxu Zhang
- Department of Urology, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
62
|
Li F, Huang J, Liu J, Xu W, Yuan Z. Multivariate analysis of clinicopathological and prognostic significance of miRNA 106b~25 cluster in gastric cancer. Cancer Cell Int 2019; 19:200. [PMID: 31384175 PMCID: PMC6664745 DOI: 10.1186/s12935-019-0918-7] [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: 04/09/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022] Open
Abstract
Background miRNA 106b~25 cluster were demonstrated to be an oncogene. In previous study, we had analyzed the diagnostic significance of miRNA 106b~25 based on its carcinogenesis effect. The significance of miRNA 106b~25 for prognosis of gastric cancer were not researched. Methods We applied multivariate analysis of PCA, PLS-DA and Cox Regression for clinicopathological features and survival time to explore the significance of miRNA 106b~25 expression in plasma and cancer tissues for gastric cancer. Results The expression of miRNA 106b, miRNA 93 and miRNA 25 in plasma were positively correlated with their expression in tumor tissues. Via PCA analysis, it was found that miRNA 106b~25 expression in plasma and tumor, T, N and TNM stage were correlated with each other. Via PLS-DA analysis, we identified that T, N and TNM stage were important factors for miRNA 106b~25 expression both in plasma and tumor (all VIP value > 1.2). According to loading weights of variables for the first and second components, it was found that the importance of the miRNA 106b~25s expression carried with the progressed stage of gastric cancer. In the survival analysis, COX regression showed that T stage, plasma miRNA 106b and tumor miRNA 93 were significant risk factors for overall survival [HR: 0.400 (0.205–0.780); P = 0.007; HR: 0.371 (0.142–0.969), P = 0.043; 0.295 (0.134–0.650), P = 0.002]. Conclusion Plasma and tumor miRNA 106b~25 expression correlated with T, N and TNM stage. Increased miRNA 106b~25 expression was important characters carried with gastric cancer progression. T stage, plasma miRNA106b and tumor miRNA 93 significant risk factors for overall survival. Electronic supplementary material The online version of this article (10.1186/s12935-019-0918-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fangxuan Li
- 1Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhuxi Road, Hexi District, Tianjin, 300060 China.,2Department of Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Hexi District, Tianjin, 300060 China
| | - Jinchao Huang
- 2Department of Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Hexi District, Tianjin, 300060 China.,3Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Hexi District, Tianjin, 300060 China
| | - Juntian Liu
- 2Department of Cancer Prevention, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Hexi District, Tianjin, 300060 China
| | - Wengui Xu
- 3Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Hexi District, Tianjin, 300060 China
| | - Zhiyong Yuan
- 1Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Huanhuxi Road, Hexi District, Tianjin, 300060 China
| |
Collapse
|
63
|
Wan W, Wan W, Long Y, Li Q, Jin X, Wan G, Zhang F, Lv Y, Zheng G, Li Z, Zhu Y. MiR-25-3p promotes malignant phenotypes of retinoblastoma by regulating PTEN/Akt pathway. Biomed Pharmacother 2019; 118:109111. [PMID: 31336343 DOI: 10.1016/j.biopha.2019.109111] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/29/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
Aberrant expression of microRNAs plays an important role in the pathogenesis and progression of retinoblastoma. MiR-25, a member of the miR-106b˜25 cluster, has been reported to be abnormally expressed in retinoblastoma, but the exact role of it remains unclear. In our study, we found that miR-25-3p was upregulated in retinoblastoma tissues and cell lines. Enforced expression of miR-25-3p in retinoblastoma cell line WERI-RB-1 increased cell growth, colony formation, anchorage-independent growth, cell migration and invasion in vitro and tumor xenograft growth in vivo. In contrast, inhibited miR-25-3p expression in retinoblastoma cell line Y79 suppressed cell growth, colony formation, anchorage-independent growth, cell migration and invasion. Through luciferase reporter assay, we found that phosphatase and tensin homolog (PTEN) was a direct target of miR-25-3p. This was verified by western blot that miR-25-3p overexpression suppressed PTEN and activated Akt signaling. In addition, miR-25-3p was found to promote epithelial-mesenchymal transition (EMT) of WERI-RB-1 cells through PTEN/Akt pathway. Western blot analysis revealed that miR-25-3p overexpression increased Vimentin and Snail expression, and suppressed E-cadherin expression, but this could be reversed by restoring PTEN. Moreover, LY294002 treatment or restoring PTEN expression abolished the effects of miR-25-3p on cell invasion, colony formation and anchorage-independent growth in vitro and tumor xenograft growth in vivo. Taken together, our results suggested that miR-25-3p promotes malignant transformation of retinoblastoma cells by suppressing PTEN.
Collapse
Affiliation(s)
- Wencui Wan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China
| | - Weiwei Wan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China
| | - Yang Long
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China
| | - Qiuming Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China.
| | - Xuemin Jin
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China
| | - Guangming Wan
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China
| | - Fengyan Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China
| | - Yong Lv
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China
| | - Guangying Zheng
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China
| | - Zhigang Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China
| | - Yu Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou 450052, China.
| |
Collapse
|
64
|
Yeh HW, Lee SS, Chang CY, Lang YD, Jou YS. A New Switch for TGFβ in Cancer. Cancer Res 2019; 79:3797-3805. [PMID: 31300476 DOI: 10.1158/0008-5472.can-18-2019] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 10/17/2018] [Accepted: 05/08/2019] [Indexed: 11/16/2022]
Abstract
The TGFβ cytokine plays dichotomous roles during tumor progression. In normal and premalignant cancer cells, the TGFβ signaling pathway inhibits proliferation and promotes cell-cycle arrest and apoptosis. However, the activation of this pathway in late-stage cancer cells could facilitate the epithelial-to-mesenchymal transition, stemness, and mobile features to enhance tumorigenesis and metastasis. The opposite functions of TGFβ signaling during tumor progression make it a challenging target to develop anticancer interventions. Nevertheless, the recent discovery of cellular contextual determinants, especially the binding partners of the transcription modulators Smads, is critical to switch TGFβ responses from proapoptosis to prometastasis. In this review, we summarize the recently identified contextual determinants (such as PSPC1, KLF5, 14-3-3ζ, C/EBPβ, and others) and the mechanisms of how tumor cells manage the context-dependent autonomous TGFβ responses to potentiate tumor progression. With the altered expression of some contextual determinants and their effectors during tumor progression, the aberrant molecular prometastatic switch might serve as a new class of theranostic targets for developing anticancer strategies.
Collapse
Affiliation(s)
- Hsi-Wen Yeh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Szu-Shuo Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Chieh-Yu Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Yaw-Dong Lang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuh-Shan Jou
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan. .,Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| |
Collapse
|
65
|
Shen DW, Li YL, Hou YJ, Xu ZD, Li YZ, Chang JY. MicroRNA-543 promotes cell invasion and impedes apoptosis in pituitary adenoma via activating the Wnt/β-catenin pathway by negative regulation of Smad7. Biosci Biotechnol Biochem 2019; 83:1035-1044. [PMID: 30973065 DOI: 10.1080/09168451.2019.1591260] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
ABSTRACT
Pituitary adenomas (PA) are commonly occurring benign neoplasms. Identification of molecular pathway resulting in pituitary tumorigenesis remains challenges in endocrine oncology. The present study was conducted with aim of investigating the role of microRNA-543 (miR-543) in PA development. Up-regulated miR-543 and downregulated Smad7 were observed in PA tissues. Afterwards, the specific mechanism of miR-543 and Smad7 in PA were determined with the use of ectopic expression, depletion and reporter assay experiments. Smad7 was confirmed as a target gene of miR-543. HP75 cells treated with overexpressed miR-543 exhibited increased cell proliferation, migration and invasion, while decreased cell apoptosis as well as expression of Cleaved caspase-3 and Cleaved caspase-8 were observed. Suppression of miR-543 contributed to an opposite trend to the above findings. Based on the findings, the inhibition of miR-543 was found to play a tumor suppressive role in PA through the down-regulation of Wnt/β-catenin pathway by negatively regulating Smad7.
Collapse
Affiliation(s)
- Da-Wei Shen
- Department of Neurosurgery, Yidu Central Hospital of Weifang, Weifang, P. R. China
| | - Yun-Long Li
- Department of Neurosurgery, Yidu Central Hospital of Weifang, Weifang, P. R. China
| | - Yu-Jie Hou
- Department of Hand and Foot Surgery, Yidu Central Hospital of Weifang, Weifang, P. R. China
| | - Zhi-Dan Xu
- Pharmacology Teaching and Research Section, Weifang Nursing Vocational College, Weifang, P. R. China
| | - Yong-Zhe Li
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, P. R. China
| | - Jian-Yong Chang
- Department of Neurosurgery, Weifang People’s Hospital, Weifang, P. R. China
| |
Collapse
|
66
|
Zhu M, Zhang N, He S. Transcription factor KLF4 modulates microRNA-106a that targets Smad7 in gastric cancer. Pathol Res Pract 2019; 215:152467. [PMID: 31146975 DOI: 10.1016/j.prp.2019.152467] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/12/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023]
Abstract
Mounting evidence has revealed that microRNAs (miRNAs, miRNA) play oncogenic or anti-oncogenic roles in many cancer types. Our previous studies have found the ectopic expression of miR-106a in gastric cancer. However, its deregulation and some potential targets have not yet been fully explored. In this investigation, we identified that the upstream transcriptional factor krüppel-like factor 4 (KLF4), a novel regulator, directly bound to the promoter sequence of miR-106a and was responsible for its deregulation. Using real-time PCR and immunohistochemistry, we further verified that the expression level of KLF4 was negatively correlated with the miR-106a expression in tissue samples. Moreover, the downstream locus was also screened and small mothers against decapentaplegic 7 (Smad7) was revealed to be a direct target of miR-106a, with its 3'-UTR region complementarily bound to miR-106a and the protein expression was mediated by miR-106a in gastric cancer cells, which was confirmed by luciferase assay and Western blot. The role of KLF4-miR-106a-Smad7 in gastric cancer invasion was assessed by real-time PCR and transwell assay. The promoting effect of miR-106a on gastric cancer invasion was significantly abolished by the overexpression of KLF4. The silencing of Smad7 partially promoted the cell invasion when miR-106a was suppressed. In conclusion, we suggest that the ectopic expression of miR-106a is modulated by the upstream transcriptional factor KLF4, which influences the invasive ability of gastric cancer through the downstream target Smad7. MiR-106a should, therefore, be considered as a potential molecular phenotype in gastric cancer.
Collapse
Affiliation(s)
- Meng Zhu
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Shaanxi, Xi'an, 710061, China
| | - Ning Zhang
- Department of Pathology, General Hospital of Ningxia Medical University, 804 Shengli Street, Ningxia, Yinchuan, 750004, China
| | - Shuixiang He
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Shaanxi, Xi'an, 710061, China.
| |
Collapse
|
67
|
Guo J, Li S, Li Y, Yan C, Wan Q, Wang Z. HSP90 inhibitor 17-AAG prevents apoptosis of cardiomyocytes via miR-93-dependent mitigation of endoplasmic reticulum stress. J Cell Biochem 2019; 120:7888-7896. [PMID: 30556167 DOI: 10.1002/jcb.28064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/22/2018] [Indexed: 01/24/2023]
Abstract
Heart failure accounts for substantial morbidity and mortality worldwide. Accumulating evidence suggests that aberrant cardiac cell death caused by endoplasmic reticulum stress (ERS) is often associated with structural or functional cardiac abnormalities that lead to insufficient cardiac output. The detailed molecular mechanism underlying the pathological death of cardiomyocytes still remains poorly understood. We found that 17-AAG (tanespimycin), an HSP90 (heat shock protein 90) inhibitor often used to kill cancer cells, could potently inhibit tunicamycin-induced ERS and the downstream nuclear factor kappa B activity in neonatal rat cardiomyocytes, leading to diminished apoptotic signaling and thus enhanced cell survival. Interestingly, the antiapoptotic effect of 17-AAG on cardiomyocytes required normal expression of miR-93, an oncogenic microRNA known to promote cell survival and growth. Our study implicated a new pharmacological role of 17-AAG in supporting the miR-93-associated oncogenic signaling to prevent the pathological death of cardiomyocytes. The results opened opportunities for exploring new strategies in the development of therapeutic agents.
Collapse
Affiliation(s)
- Jingjing Guo
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| | - Shengnan Li
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| | - Yanming Li
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| | - Chenyun Yan
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| | - Qilin Wan
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| | - Zhizhong Wang
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| |
Collapse
|
68
|
Tomar D, Yadav AS, Kumar D, Bhadauriya G, Kundu GC. Non-coding RNAs as potential therapeutic targets in breast cancer. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1863:194378. [PMID: 31048026 DOI: 10.1016/j.bbagrm.2019.04.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/15/2019] [Accepted: 04/23/2019] [Indexed: 12/15/2022]
Abstract
Paradigm shifting studies especially involving non-coding RNAs (ncRNAs) during last few decades have significantly changed the scientific perspectives regarding the complexity of cellular signalling pathways. Several studies have shown that the non-coding RNAs, initially ignored as transcriptional noise or products of erroneous transcription; actually regulate plethora of biological phenomena ranging from developmental processes to various diseases including cancer. Current strategies that are employed for the management of various cancers including that of breast fall short when their undesired side effects like Cancer Stem Cells (CSC) enrichment, low recurrence-free survival and development of drug resistance are taken into consideration. This review aims at exploring the potential role of ncRNAs as therapeutics in breast cancer, by providing a comprehensive understanding of their mechanism of action and function and their crucial contribution in regulating various aspects of breast cancer progression such as cell proliferation, angiogenesis, EMT, CSCs, drug resistance and metastasis. In addition, we also provide information about various strategies that can be employed or are under development to explore them as potential moieties that may be used for therapeutic intervention in breast cancer.
Collapse
Affiliation(s)
- Deepti Tomar
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, India.
| | - Amit S Yadav
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, India.
| | - Dhiraj Kumar
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| | - Garima Bhadauriya
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, India
| | - Gopal C Kundu
- Laboratory of Tumor Biology, Angiogenesis and Nanomedicine Research, National Centre for Cell Science (NCCS), Pune, India.
| |
Collapse
|
69
|
Bahiraee A, Ebrahimi R, Halabian R, Aghabozorgi AS, Amani J. The role of inflammation and its related microRNAs in breast cancer: A narrative review. J Cell Physiol 2019; 234:19480-19493. [PMID: 31025369 DOI: 10.1002/jcp.28742] [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] [Received: 12/25/2018] [Revised: 03/27/2019] [Accepted: 04/10/2019] [Indexed: 12/21/2022]
Abstract
Breast cancer is recognized as the most common type of cancer among women with a high rate of mortality all over the world. Over the past years, growing attention has been regarded to realize more about the mechanisms underlying the disease process. It is revealed that the progression of breast cancer may be strongly linked to chronic inflammation owing to the role of inflammatory factors in genetic instability and subsequent cancer predisposition. Although the association between breast cancer and inflammatory pathways has been well-defined now, only recent evidence pointed towards the inflammation-related microRNAs (miRNAs) as potential biomarkers and therapeutic targets involved in the crosstalk of multiple pathways during breast cancer development. Moreover, the practical interactions between these miRNAs and inflammatory factors are also a little characterized. In this review, we intended to describe the effects of predominant inflammatory pathways such as cytokines, phosphoinositide 3-kinase/protein kinase B, and nuclear factor kappa B in association with tumor promoting and tumor suppressing miRNAs on breast cancer progression. Providing new studies in the field of combining biomarkers for early diagnosis, prognosis, and monitoring breast cancer are very important. Notably, understanding the underlying mechanisms of miRNAs as a possible link between inflammation and tumorigenesis may offer a novel insight for combating this epidemic.
Collapse
Affiliation(s)
- Alireza Bahiraee
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amirsaeed Sabeti Aghabozorgi
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
70
|
miR-106b-5p and miR-17-5p could predict recurrence and progression in breast ductal carcinoma in situ based on the transforming growth factor-beta pathway. Breast Cancer Res Treat 2019; 176:119-130. [PMID: 30989460 PMCID: PMC6548759 DOI: 10.1007/s10549-019-05192-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/02/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Ductal carcinoma in situ (DCIS) is well-known precursor of invasive ductal carcinoma (IDC). Parts of patients show recurrence as DCIS or IDC after local treatment, but there are no established markers predicting relapse. We analyzed changes in miRNA and oncogene expression during DCIS progression/evolution to identify potential markers predicting recurrence. METHODS Forty archival tissues diagnosed as primary or recurrent DCIS and DCIS adjacent to IDC were analyzed. MiRNA hierarchical clustering showed up-regulation of miR-17-5p and miR-106b-5p in recurrent DCIS and DCIS adjacent to IDC. Target genes were predicted based on pre-formed miRNA databases and PanCancer Pathway panel. MiRNAs were transfected into MCF-10A and MCF-7 cells; western blot analysis was performed with MCF-7 cell line to evaluate the effects on TGF-β downstream pathway. RESULTS miRNA hierarchical clustering showed 17 dysregulated miRNAs, including miR-17-5p and miR-106b-5p. Based on miRNA database and nCounter Pancancer pathway analysis, TGFβRII was selected as target of miR-106b-5p and miR-17-5p. MiR-106b-5p- and miR-17-5p-transfected MCF-7 cells showed decreased expression of TGFβRII, especially in cells transfected with both miRNAs. CONCLUSION miR-106b-5p and miR-17-5p might have a role in breast cancer recurrence and progression by suppressing TGF-β activity, leading to early breast cancer carcinogenesis.
Collapse
|
71
|
Hu B, Mao Z, Du Q, Jiang X, Wang Z, Xiao Z, Zhu D, Wang X, Zhu Y, Wang H. miR-93-5p targets Smad7 to regulate the transforming growth factor-β1/Smad3 pathway and mediate fibrosis in drug-resistant prolactinoma. Brain Res Bull 2019; 149:21-31. [PMID: 30946881 DOI: 10.1016/j.brainresbull.2019.03.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/23/2019] [Accepted: 03/28/2019] [Indexed: 12/29/2022]
Abstract
Prolactinoma is a common subtype of pituitary tumors. Dopamine receptor agonists are the preferred treatment for prolactinoma; however, with this therapy, drug resistance often occurs. In our previous work, we found that partial resistant prolactinomas showed increased fibrosis and that the transforming growth factor (TGF)-β1/Smad3 signaling pathway mediated fibrosis and was involved in drug resistance. Additionally, the success of surgery is known to be heavily influenced by the consistency of the pituitary adenoma. Therefore, in this study, we aimed to clarify the mechanisms of fibrosis in prolactinoma. Using high-throughput sequencing for analysis of microRNAs, we found that miR-93-5p was significantly upregulated in prolactinoma samples with a high degree of fibrosis compared with that in samples without fibrosis. Furthermore, we found that miR-93-5p was negatively correlated with the relative expression of Smad7 and positively correlated with the relative expression of TGF-β1 in clinical prolactinoma samples. In addition, luciferase reporter assays showed that miR-93-5p could downregulate the Smad7 gene, an important inhibitor of the TGF-β1/Smad3 signaling pathway, and activate TGF-β1/Smad3 signaling-mediated fibrosis in a feed-forward loop. Moreover, miR-93-5p could enhance the drug resistance of prolactinoma cells by regulation of TGF-β1/Smad3-dependent fibrosis. Taken together, our findings demonstrated that miR-93-5p may be a potential therapeutic target for inhibiting fibrosis and reducing drug resistance in prolactinoma cells.
Collapse
Affiliation(s)
- Bin Hu
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhigang Mao
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiu Du
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Zongming Wang
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zheng Xiao
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dimin Zhu
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xin Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yonghong Zhu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Haijun Wang
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
72
|
Oh JG, Jang SP, Yoo J, Lee MA, Lee SH, Lim T, Jeong E, Kho C, Kook H, Hajjar RJ, Park WJ, Jeong D. Role of the PRC2-Six1-miR-25 signaling axis in heart failure. J Mol Cell Cardiol 2019; 129:58-68. [DOI: 10.1016/j.yjmcc.2019.01.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/25/2018] [Accepted: 01/21/2019] [Indexed: 01/14/2023]
|
73
|
Kingsbury TJ, Kim M, Civin CI. Regulation of cancer stem cell properties by SIX1, a member of the PAX-SIX-EYA-DACH network. Adv Cancer Res 2019; 141:1-42. [PMID: 30691681 DOI: 10.1016/bs.acr.2018.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The PAX-SIX-EYA-DACH network (PSEDN) is a central developmental transcriptional regulatory network from Drosophila to humans. The PSEDN is comprised of four conserved protein families; including paired box (PAX), sine oculis (SIX), eyes absent (EYA), and dachshund (DACH). Aberrant expression of PSEDN members, particularly SIX1, has been observed in multiple human cancers, where SIX1 expression correlates with increased aggressiveness and poor prognosis. In conjunction with its transcriptional activator EYA, the SIX1 transcription factor increases cancer stem cell (CSC) numbers and induces epithelial-mesenchymal transition (EMT). SIX1 promotes multiple hallmarks and enabling characteristics of cancer via regulation of cell proliferation, senescence, apoptosis, genome stability, and energy metabolism. SIX1 also influences the tumor microenvironment, enhancing recruitment of tumor-associated macrophages and stimulating angiogenesis, to promote tumor development and progression. EYA proteins are multifunctional, possessing a transcriptional activation domain and tyrosine phosphatase activity, that each contributes to cancer stem cell properties. DACH proteins function as tumor suppressors in solid cancers, opposing the actions of SIX-EYA and reducing CSC prevalence. Multiple mechanisms can lead to increased SIX1 expression, including loss of SIX1-targeting tumor suppressor microRNAs (miRs), whose expression correlates inversely with SIX1 expression in cancer patient samples. In this review, we discuss the major mechanisms by which SIX1 confers CSC and EMT features and other important cancer cell characteristics. The roles of EYA and DACH in CSCs and cancer progression are briefly highlighted. Finally, we summarize the clinical significance of SIX1 in cancer to emphasize the potential therapeutic benefits of effective strategies to disrupt PSEDN protein interactions and functions.
Collapse
|
74
|
Manzanarez-Ozuna E, Flores DL, Gutiérrez-López E, Cervantes D, Juárez P. Model based on GA and DNN for prediction of mRNA-Smad7 expression regulated by miRNAs in breast cancer. Theor Biol Med Model 2018; 15:24. [PMID: 30594253 PMCID: PMC6310970 DOI: 10.1186/s12976-018-0095-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/30/2018] [Indexed: 01/06/2023] Open
Abstract
Background The Smad7 protein is negative regulator of the TGF-β signaling pathway, which is upregulated in patients with breast cancer. miRNAs regulate proteins expressions by arresting or degrading the mRNAs. The purpose of this work is to identify a miRNAs profile that regulates the expression of the mRNA coding for Smad7 in breast cancer using the data from patients with breast cancer obtained from the Cancer Genome Atlas Project. Methods We develop an automatic search method based on genetic algorithms to find a predictive model based on deep neural networks (DNN) which fit the set of biological data and apply the Olden algorithm to identify the relative importance of each miRNAs. Results A computational model of non-linear regression is shown, based on deep neural networks that predict the regulation given by the miRNA target transcripts mRNA coding for Smad7 protein in patients with breast cancer, with R2 of 0.99 is shown and MSE of 0.00001. In addition, the model is validated with the results in vivo and in vitro experiments reported in the literature. The set of miRNAs hsa-mir-146a, hsa-mir-93, hsa-mir-375, hsa-mir-205, hsa-mir-15a, hsa-mir-21, hsa-mir-20a, hsa-mir-503, hsa-mir-29c, hsa-mir-497, hsa-mir-107, hsa-mir-125a, hsa-mir-200c, hsa-mir-212, hsa-mir-429, hsa-mir-34a, hsa-let-7c, hsa-mir-92b, hsa-mir-33a, hsa-mir-15b, hsa-mir-224, hsa-mir-185 and hsa-mir-10b integrate a profile that critically regulates the expression of the mRNA coding for Smad7 in breast cancer. Conclusions We developed a genetic algorithm to select best features as DNN inputs (miRNAs). The genetic algorithm also builds the best DNN architecture by optimizing the parameters. Although the confirmation of the results by laboratory experiments has not occurred, the results allow suggesting that miRNAs profile could be used as biomarkers or targets in targeted therapies. Electronic supplementary material The online version of this article (10.1186/s12976-018-0095-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Edgar Manzanarez-Ozuna
- Universidad Autónoma de Baja California, Carretera Transpeninsular Ensenada-Tijuana 3917 Colonia Playitas, C.P. 22860, Ensenada, B.C., Mexico
| | - Dora-Luz Flores
- Universidad Autónoma de Baja California, Carretera Transpeninsular Ensenada-Tijuana 3917 Colonia Playitas, C.P. 22860, Ensenada, B.C., Mexico.
| | - Everardo Gutiérrez-López
- Universidad Autónoma de Baja California, Carretera Transpeninsular Ensenada-Tijuana 3917 Colonia Playitas, C.P. 22860, Ensenada, B.C., Mexico
| | - David Cervantes
- Universidad Autónoma de Baja California, Carretera Transpeninsular Ensenada-Tijuana 3917 Colonia Playitas, C.P. 22860, Ensenada, B.C., Mexico
| | - Patricia Juárez
- Centro de Investigación Científica y de Educación Superior de Ensenada, Carretera Ensenada-Tijuana No. 3918, Zona Playitas, C.P. 22860, Ensenada, B.C., Mexico
| |
Collapse
|
75
|
Li X, Wu X. MiR-21-5p promotes the progression of non-small-cell lung cancer by regulating the expression of SMAD7. Onco Targets Ther 2018; 11:8445-8454. [PMID: 30568467 PMCID: PMC6276624 DOI: 10.2147/ott.s172393] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective The objective of this study was to detect the expression of MiR-21-5p in non-small-cell lung cancer (NSCLC) tissues, and to investigate the effect of its expression on the progression of NSCLC. Methods Real-time fluorescent quantitative PCR was used to detect the relative expression of MiR-21-5p in 118 NSCLC tumor tissues to their adjacent normal tissues. The expressions of SMAD7, MMP-9, E-cadherin, and vimentin proteins were detected by Western blotting or immunohistochemistry. Cell colony formation, scratch, and Transwell assays were used to detect the proliferation, migration, and invasion ability of A549 cells, respectively. Results MiR-21-5p was highly expressed in the tumor tissues of NSCLC patients, and its expression was significantly correlated with the clinical classification of NSCLC patients (χ2=7.154, P=0.007), tumor size (χ2=4.372, P=0.037), differentiation (χ2=13.713, P=0.001), lymph node metastasis (χ2=5.101, P=0.024), distant metastasis (χ2=12.599, P=0.000), and TNM stage (χ2=6.344, P=0.012), whereas it was positively correlated with the expression of SMAD7 protein (r=0.669, P<0.05). The results of the luciferase gene reporter system showed that MiR-21-5p targeted and promoted the expression of SMAD7 gene, which enhanced NSCLC cell proliferation. Furthermore, MiR-21-5p promoted the expressions of MMP-9 and vimentin proteins as well as inhibited the expression of E-cadherin protein, which is associated with an elevated SMAD7 protein expression and enhanced the invasion/migration ability of NSCLC cells. Conclusion MiR-21-5p was highly expressed in NSCLC tumor tissues, and its high expression could promote NSCLC progression by targeting the expression of SMAD7.
Collapse
Affiliation(s)
- Xiangpan Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuchang, Wuhan, Hubei Province, People's Republic of China
| | - Xiaofei Wu
- Department of Neurology, Chinese People's Liberation Army, Wuhan General Hospital, Wuchang District, Wuhan, Hubei Province, People's Republic of China,
| |
Collapse
|
76
|
Wu S, Wang Y, Yuan Z, Wang S, Du H, Liu X, Wang Q, Zhu X. Human adipose‑derived mesenchymal stem cells promote breast cancer MCF7 cell epithelial‑mesenchymal transition by cross interacting with the TGF‑β/Smad and PI3K/AKT signaling pathways. Mol Med Rep 2018; 19:177-186. [PMID: 30483746 PMCID: PMC6297785 DOI: 10.3892/mmr.2018.9664] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 10/19/2018] [Indexed: 01/14/2023] Open
Abstract
The influence and underlying mechanisms of human adipose-derived stem cells (Hu-ADSCs) on breast cancer cells in the tumor microenvironment remain unclear. Understanding the association between Hu-ADSCs and cancer cells may provide targets for breast cancer treatment and reference for the clinical application of stem cells. Therefore, a Hu-ADSC and breast cancer MCF7 cell coculture system was established to investigate the paracrine effects of Hu-ADSCs on MCF7 cell migration and invasion, in addition to the potential mechanism of action by reverse transcription-quantitative polymerase chain reaction and western blotting. Hu-ADSCs enhanced MCF7 cell migration and invasion by decreasing the expression of epithelial marker E-cadherin, and increasing the expression of interstitial marker N-cadherin and epithelial-mesenchymal transition (EMT) transcription factors in vitro. The EMT effect of cocultured MCF7 cells was inhibited with the addition of anti-transforming growth factor (TGF)-β1 or phosphoinositide 3-kinase (PI3K) inhibitor LY294002, accompanied by a significant decrease in phosphorylated (p)-mothers against decapentaplegic homolog (Smad) and p-protein kinase B (AKT) expression. The data suggested that the paracrine effect of Hu-ADSCs in the tumor microenvironment promoted the EMT of MCF7 cells by cross interacting with the TGF-β/Smad and PI3K/AKT pathways.
Collapse
Affiliation(s)
- Simeng Wu
- Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yajun Wang
- Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhe Yuan
- Cord Blood Bank, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Siliang Wang
- Department of Medical Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Hongmei Du
- Department of Medical Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xue Liu
- Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Qiushi Wang
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xike Zhu
- Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
77
|
Zhai W, Li S, Zhang J, Chen Y, Ma J, Kong W, Gong D, Zheng J, Xue W, Xu Y. Sunitinib-suppressed miR-452-5p facilitates renal cancer cell invasion and metastasis through modulating SMAD4/SMAD7 signals. Mol Cancer 2018; 17:157. [PMID: 30419914 PMCID: PMC6231268 DOI: 10.1186/s12943-018-0906-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Although microRNAs (miRNAs) were revealed as crucial modulators in tumor metastasis and target therapy, our understanding of their roles in metastatic renal cell carcinoma (mRCC) and Sunitinib treatment was limited. Here we sought to identify human miRNAs that acted as key regulators in renal cancer metastasis and Sunitinib treatment. EXPERIMENTAL DESIGN We focused on 2 published microarray data to select out our anchored miRNA and then explored the roles of miR-452-5p both in vitro and in vivo, which was downregulated after Sunitinib treatment while upregulated in metastasis renal cell carcinoma (RCC) tissues. RESULTS Here, we discovered that treating with Sunitinib, the targeted receptor tyrosine kinase inhibitor (TKI), inhibited renal cancer cell migration and invasion via attenuating the expression of miR-452-5p. The novel identified miR-452-5p was upregulated and associated with poor prognosis in RCC. Preclinical studies using multiple RCC cells and xenografts model illustrated that miR-452-5p could promote RCC cell migration and invasion in vitro and in vivo. Mechanistically, P65 could directly bind to the miR-452-5p promoter and thus transcriptionally induce miR-452-5p expression, which led to post-transcriptionally abrogate SMAD4 expression, thus inhibition of its downstream gene SMAD7. CONCLUSION Our study presented a road map for targeting this newly identified miR-452-5p and its SMAD4/SMAD7 signals pathway, which imparted a new potential therapeutic strategy for mRCC treatment.
Collapse
Affiliation(s)
- Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Pudong District, Shanghai, 200127, China.
| | - Saiyang Li
- Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Jin Zhang
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Pudong District, Shanghai, 200127, China
| | - Yonghui Chen
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Pudong District, Shanghai, 200127, China
| | - Junjie Ma
- Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Wen Kong
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Pudong District, Shanghai, 200127, China
| | - Dongkui Gong
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, 200072, China
| | - Junhua Zheng
- Department of Urology, Shanghai First People's Hospital, School of Medicine in Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Pudong District, Shanghai, 200127, China.
| | - Yunfei Xu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, 200072, China.
| |
Collapse
|
78
|
Gao Y, Deng K, Liu X, Dai M, Chen X, Chen J, Chen J, Huang Y, Dai S, Chen J. Molecular mechanism and role of microRNA-93 in human cancers: A study based on bioinformatics analysis, meta-analysis, and quantitative polymerase chain reaction validation. J Cell Biochem 2018; 120:6370-6383. [PMID: 30390344 DOI: 10.1002/jcb.27924] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/27/2018] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Currently, studies have shown that microRNA-93 (miR-93) can be an oncogene or a tumor suppressor in different kinds of cancers. The role of miR-93 in human cancers is inconsistent and the underlying mechanism on the aberrant expression of miR-93 is complicated. METHODS We first conducted gene enrichment analysis to give insight into the prospective mechanism of miR-93. Second, we performed a meta-analysis to evaluate the clinical value of miR-93. Finally, a validation test based on quantitative polymerase chain reaction (qPCR) was performed to further investigate the role of miR-93 in pan-cancer. RESULTS Gene Ontology (GO) enrichment analysis results showed that the target genes of miR-93 were closely related to transcription, and MAPK1, RBBP7 and Smad7 became the hub genes. In the diagnostic meta-analysis, the overall sensitivity, specificity, and area under the curve were 0.76 (0.64-0.85), 0.82 (0.64-0.92), and 0.85 (0.82-0.88), respectively, which suggested that miR-93 had excellent performance on the diagnosis for human cancers. In the prognostic meta-analysis, dysregulated miR-93 was found to be associated with poor OS in cancer patients. In the qPCR validation test, the serum levels of miR-93 were upregulated in breast cancer, breast hyperplasia, lung cancer, chronic obstructive pulmonary disease, nasopharyngeal cancer, hepatocellular cancer, gastric ulcer, endometrial cancer, esophageal cancer, laryngeal cancer, and prostate cancer compared with healthy controls. CONCLUSIONS miR-93 could act as an effective diagnostic and prognostic factor for cancer patients. Its clinical value for cancer early diagnosis and survival prediction is promising.
Collapse
Affiliation(s)
- Yun Gao
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Kaifeng Deng
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Xuexiang Liu
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Meiyu Dai
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Xiaoli Chen
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Jifei Chen
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Jianming Chen
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Yujie Huang
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Shengming Dai
- Medical Science Laboratory, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Jingfan Chen
- Department of General Surgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| |
Collapse
|
79
|
Emerging ways to treat breast cancer: will promises be met? Cell Oncol (Dordr) 2018; 41:605-621. [PMID: 30259416 DOI: 10.1007/s13402-018-0409-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common cancer among women and it is responsible for more than 40,000 deaths in the United States and more than 500,000 deaths worldwide each year. In previous decades, the development of improved screening, diagnosis and treatment methods has led to decreases in BC mortality rates. More recently, novel targeted therapeutic options, such as the use of monoclonal antibodies and small molecule inhibitors that target specific cancer cell-related components, have been developed. These components include ErbB family members (HER1, HER2, HER3 and HER4), Ras/MAPK pathway components (Ras, Raf, MEK and ERK), VEGF family members (VEGFA, VEGFB, VEGFC, VEGF and PGF), apoptosis and cell cycle regulators (BAK, BAX, BCL-2, BCL-X, MCL-1 and BCL-W, p53 and PI3K/Akt/mTOR pathway components) and DNA repair pathway components such as BRCA1. In addition, long noncoding RNA inhibitor-, microRNA inhibitor/mimic- and immunotherapy-based approaches are being developed for the treatment of BC. Finally, a novel powerful technique called CRISPR-Cas9-based gene editing is emerging as a precise tool for the targeted treatment of cancer, including BC. CONCLUSIONS Potential new strategies that are designed to specifically target BC are presented. Several clinical trials using these strategies are already in progress and have shown promising results, but inherent limitations such as off-target effects and low delivery efficiencies still have to be resolved. By improving the clinical efficacy of current therapies and exploring new ones, it is anticipated that novel ways to overcome BC may become attainable.
Collapse
|
80
|
Mota MSV, Jackson WP, Bailey SK, Vayalil P, Landar A, Rostas JW, Mulekar MS, Samant RS, Shevde LA. Deficiency of tumor suppressor Merlin facilitates metabolic adaptation by co-operative engagement of SMAD-Hippo signaling in breast cancer. Carcinogenesis 2018; 39:1165-1175. [PMID: 29893810 PMCID: PMC6148973 DOI: 10.1093/carcin/bgy078] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/15/2018] [Accepted: 06/08/2018] [Indexed: 12/17/2022] Open
Abstract
The NF2 gene encodes the tumor and metastasis suppressor protein Merlin. Merlin exerts its tumor suppressive role by inhibiting proliferation and inducing contact-growth inhibition and apoptosis. In the current investigation, we determined that loss of Merlin in breast cancer tissues is concordant with the loss of the inhibitory SMAD, SMAD7, of the TGF-β pathway. This was reflected as dysregulated activation of TGF-β signaling that co-operatively engaged with effectors of the Hippo pathway (YAP/TAZ/TEAD). As a consequence, the loss of Merlin in breast cancer resulted in a significant metabolic and bioenergetic adaptation of cells characterized by increased aerobic glycolysis and decreased oxygen consumption. Mechanistically, we determined that the co-operative activity of the Hippo and TGF-β transcription effectors caused upregulation of the long non-coding RNA Urothelial Cancer-Associated 1 (UCA1) that disengaged Merlin's check on STAT3 activity. The consequent upregulation of Hexokinase 2 (HK2) enabled a metabolic shift towards aerobic glycolysis. In fact, Merlin deficiency engendered cellular dependence on this metabolic adaptation, endorsing a critical role for Merlin in regulating cellular metabolism. This is the first report of Merlin functioning as a molecular restraint on cellular metabolism. Thus, breast cancer patients whose tumors demonstrate concordant loss of Merlin and SMAD7 may benefit from an approach of incorporating STAT3 inhibitors.
Collapse
Affiliation(s)
- Mateus S V Mota
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - William P Jackson
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - Sarah K Bailey
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - Praveen Vayalil
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - Aimee Landar
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - Jack W Rostas
- Department of Surgery, University of Louisville, Louisville, KY, USA
| | - Madhuri S Mulekar
- Department of Mathematics and Statistics, University of South Alabama, Mobile, AL, USA
| | - Rajeev S Samant
- Department of Pathology, University of Louisville, Louisville, KY, USA
- UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, University of Louisville, Louisville, KY, USA
- UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
81
|
Mehlich D, Garbicz F, Włodarski PK. The emerging roles of the polycistronic miR-106b∼25 cluster in cancer - A comprehensive review. Biomed Pharmacother 2018; 107:1183-1195. [PMID: 30257332 DOI: 10.1016/j.biopha.2018.08.097] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are short, non-coding RNA molecules that regulate gene expression at the post-transcriptional level by inhibiting translation and decreasing the stability of the targeted transcripts. Over the last two decades, miRNAs have been recognized as important regulators of cancer cell biology, acting either as oncogenes or tumor suppressors. The polycistronic miR-106b∼25 cluster, located within an intron of MCM7 gene, consists of three highly conserved miRNAs: miR-25, miR-93 and miR-106b. A constantly growing body of evidence indicates that these miRNAs are overexpressed in numerous human malignancies and regulate multiple cellular processes associated with cancer development and progression, including: cell proliferation and survival, invasion, metastasis, angiogenesis and immune evasion. Furthermore, recent studies revealed that miR-106b∼25 cluster miRNAs modulate cancer stem cells characteristics and might promote resistance to anticancer therapies. In light of these novel discoveries, miRNAs belonging to the miR-106b∼25 cluster have emerged as key oncogenic drivers as well as potential biomarkers and plausible therapeutic targets in different tumor types. Herein, we comprehensively review novel findings on the roles of miR-106b∼25 cluster in human cancer, and provide a broad insight into the molecular mechanisms underlying its oncogenic properties.
Collapse
Affiliation(s)
- Dawid Mehlich
- Laboratory of Centre for Preclinical Research, Department of Methodology, Medical University of Warsaw, 1B Banacha Str., 02-091 Warsaw, Poland; Laboratory of Experimental Medicine, Centre of New Technologies, University of Warsaw, 2C Banacha Str., 02-097, Warsaw, Poland
| | - Filip Garbicz
- Laboratory of Centre for Preclinical Research, Department of Methodology, Medical University of Warsaw, 1B Banacha Str., 02-091 Warsaw, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, 61 Żwirki i Wigury Str., 02-091 Warsaw, Poland; Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, 14 Indiry Gandhi Str., 02-776 Warsaw, Poland
| | - Paweł K Włodarski
- Laboratory of Centre for Preclinical Research, Department of Methodology, Medical University of Warsaw, 1B Banacha Str., 02-091 Warsaw, Poland.
| |
Collapse
|
82
|
Li Q, Li B, Li Q, Wei S, He Z, Huang X, Wang L, Xia Y, Xu Z, Li Z, Wang W, Yang L, Zhang D, Xu Z. Exosomal miR-21-5p derived from gastric cancer promotes peritoneal metastasis via mesothelial-to-mesenchymal transition. Cell Death Dis 2018; 9:854. [PMID: 30154401 PMCID: PMC6113299 DOI: 10.1038/s41419-018-0928-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/25/2018] [Accepted: 07/30/2018] [Indexed: 02/08/2023]
Abstract
Peritoneal metastasis is a primary metastatic route for gastric cancers, and the mechanisms underlying this process are still unclear. Peritoneal mesothelial cells (PMCs) undergo mesothelial-to-mesenchymal transition (MMT) to provide a favorable environment for metastatic cancer cells. In this study, we investigated how the exosomal miR-21-5p induces MMT and promotes peritoneal metastasis. Gastric cancer (GC)-derived exosomes were identified by transmission electron microscopy and western blot analysis, then the uptake of exosomes was confirmed by PKH-67 staining. The expression of miR-21-5p and SMAD7 were measured by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot, and the interactions between miR-21-5p and its target genes SMAD7 were confirmed by Luciferase reporter assays. The MMT of PMCs was determined by invasion assays, adhesion assays, immunofluorescent assay, and western blot. Meanwhile, mouse model of tumor peritoneal dissemination model was performed to investigate the role of exosomal miR-21-5p in peritoneal metastasis in vivo. We found that PMCs could internalize GC-derived exosomal miR-21-5p and led to increased levels of miR-21-5p in PMCs. Through various types of in vitro and in vivo assays, we confirmed that exosomal miR-21-5p was able to induce MMT of PMCs and promote tumor peritoneal metastasis. Moreover, our study revealed that this process was promoted by exosomal miR-21-5p through activating TGF-β/Smad pathway via targeting SMAD7. Altogether, our data suggest that exosomal miR-21-5p induces MMT of PMCs and promote cancer peritoneal dissemination by targeting SMAD7. The exosomal miR-21-5p may be a novel therapeutic target for GC peritoneal metastasis.
Collapse
Affiliation(s)
- Qiang Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Qing Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Song Wei
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Zhongyuan He
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Xiaoxu Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Lu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Yiwen Xia
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Zhipeng Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Weizhi Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Li Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China.
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| |
Collapse
|
83
|
Ni S, Weng W, Xu M, Wang Q, Tan C, Sun H, Wang L, Huang D, Du X, Sheng W. miR-106b-5p inhibits the invasion and metastasis of colorectal cancer by targeting CTSA. Onco Targets Ther 2018; 11:3835-3845. [PMID: 30013364 PMCID: PMC6038879 DOI: 10.2147/ott.s172887] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Although miR-106b-5p has been reported to play a pivotal role in various human malignancies, its role in colorectal cancer (CRC) remains unknown. In this study, we comprehensively investigated miR-106b-5p expression and biologic functions in CRC and the molecular mechanism involved. Patients and methods miR-106b-5p expression was detected in CRC tissues and cell lines by quantitative reverse transcription-polymerase chain reaction. The effects of miR-106b-5p on metastasis were determined in vitro using transwell assays, and in vivo effects were evaluated using a mouse tail vein injection model. Downstream targets of miR-106b-5p were confirmed using bioinformatics programs, luciferase assays, and rescue experiments. Target gene expression and clinicopathologic parameters were also analyzed. Results miR-106b-5p expression was lower in CRC tissues than in corresponding nontumorous tissues (P=0.009), and miR-106b-5p downregulation was negatively associated with lymph node metastasis (P=0.006). Functional assays demonstrated that miR-106b-5p overexpression suppressed CRC cell migration and invasion in vitro and lung metastasis formation in vivo. In addition, luciferase assays confirmed that miR-106b-5p directly bound to the 3' untranslated region of cathepsin A (CTSA) and that miR-106b-5p suppressed CRC cell migration and invasion by targeting CTSA. Clinicopathologic analysis showed that CTSA was significantly upregulated in CRC, and increased CTSA was negatively associated with lymph node metastasis (P=0.012). Conclusion Our findings revealed that miR-106b-5p inhibits CRC metastasis by upregulating CTSA expression, which may lead to novel therapeutic strategies for CRC patients.
Collapse
Affiliation(s)
- Shujuan Ni
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Weiwei Weng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Qifeng Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Cong Tan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Lei Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Xiang Du
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China, .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China,
| |
Collapse
|
84
|
Guarnieri AL, Towers CG, Drasin DJ, Oliphant MUJ, Andrysik Z, Hotz TJ, Vartuli RL, Linklater ES, Pandey A, Khanal S, Espinosa JM, Ford HL. The miR-106b-25 cluster mediates breast tumor initiation through activation of NOTCH1 via direct repression of NEDD4L. Oncogene 2018; 37:3879-3893. [PMID: 29662198 PMCID: PMC6043359 DOI: 10.1038/s41388-018-0239-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 02/01/2018] [Accepted: 03/03/2018] [Indexed: 01/07/2023]
Abstract
Tumor-initiating cells (TIC) represent a subset of tumor cells with increased self-renewal capability. TICs display resistance to frontline cancer treatment and retain the ability to repopulate a tumor after therapy, leading to cancer relapse. NOTCH signaling has been identified as an important driver of the TIC population, yet mechanisms governing regulation of this pathway in cancer remain to be fully elucidated. Here we identify a novel mechanism of NOTCH regulation and TIC induction in breast cancer via the miR-106b-25 miRNA cluster. We show that the miR-106b-25 cluster upregulates NOTCH1 in multiple breast cancer cell lines, representing both estrogen receptor (ER+) and triple negative breast cancer (TNBC) through direct repression of the E3 ubiquitin ligase, NEDD4L. We further show that upregulation of NOTCH1 is necessary for TIC induction downstream of miR-106b-25 in both ER + and TNBC breast cancer cells, and that re-expression of NEDD4L is sufficient to reverse miR106b-25-mediated NOTCH1 upregulation and TIC induction. Importantly, we demonstrate a significant positive correlation between miR-106b-25 and NOTCH1 protein, yet a significant inverse correlation between miR-106b-25 and NEDD4L mRNA in human breast cancer, suggesting a critical role for the miR106b-25/NEDD4L/NOTCH1 axis in the disease. Further, we show for the first time that NEDD4L expression alone is significantly associated with a better relapse-free prognosis for breast cancer patients. These data expand our knowledge of the mechanisms underlying NOTCH activation and TIC induction in breast cancer, and may provide new avenues for the development of therapies targeting this resistant subset of tumor cells.
Collapse
Affiliation(s)
- A L Guarnieri
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - C G Towers
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - D J Drasin
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - M U J Oliphant
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Z Andrysik
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - T J Hotz
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - R L Vartuli
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - E S Linklater
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - A Pandey
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - S Khanal
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - J M Espinosa
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - H L Ford
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
85
|
Kabekkodu SP, Shukla V, Varghese VK, D' Souza J, Chakrabarty S, Satyamoorthy K. Clustered miRNAs and their role in biological functions and diseases. Biol Rev Camb Philos Soc 2018; 93:1955-1986. [PMID: 29797774 DOI: 10.1111/brv.12428] [Citation(s) in RCA: 228] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 04/20/2018] [Accepted: 04/26/2018] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are endogenous, small non-coding RNAs known to regulate expression of protein-coding genes. A large proportion of miRNAs are highly conserved, localized as clusters in the genome, transcribed together from physically adjacent miRNAs and show similar expression profiles. Since a single miRNA can target multiple genes and miRNA clusters contain multiple miRNAs, it is important to understand their regulation, effects and various biological functions. Like protein-coding genes, miRNA clusters are also regulated by genetic and epigenetic events. These clusters can potentially regulate every aspect of cellular function including growth, proliferation, differentiation, development, metabolism, infection, immunity, cell death, organellar biogenesis, messenger signalling, DNA repair and self-renewal, among others. Dysregulation of miRNA clusters leading to altered biological functions is key to the pathogenesis of many diseases including carcinogenesis. Here, we review recent advances in miRNA cluster research and discuss their regulation and biological functions in pathological conditions.
Collapse
Affiliation(s)
- Shama P Kabekkodu
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Vaibhav Shukla
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Vinay K Varghese
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Jeevitha D' Souza
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| |
Collapse
|
86
|
Qi Z, Tang T, Sheng L, Ma Y, Liu Y, Yan L, Qi S, Ling L, Zhang Y. Salidroside inhibits the proliferation and migration of gastric cancer cells via suppression of Src‑associated signaling pathway activation and heat shock protein 70 expression. Mol Med Rep 2018; 18:147-156. [PMID: 29749547 PMCID: PMC6059663 DOI: 10.3892/mmr.2018.8958] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 04/09/2018] [Indexed: 12/02/2022] Open
Abstract
Salidroside, an active ingredient extracted from the Rhodiola rosea plant, has potential anti-tumor effects. However, the effects of salidroside on gastric cancer cell proliferation and migration remain unclear. In the present study, the inhibitory effects of salidroside on gastric cancer cell proliferation, migration and invasion and the molecular mechanisms underlying these effects were investigated. The human gastric cancer cell line, BGC-823, was treated with different concentrations of salidroside (200, 400 and 600 µg/ml). Cell proliferation was determined with Cell Counting Kit-8 and colony formation assays, and the migration and invasion of cells was detected by a wound healing and Transwell assay, respectively. Western blotting was performed to detect the levels of N-cadherin, E-cadherin and heat shock protein (HSP)70. In addition, the phosphorylation of proto-oncogene tyrosine-protein kinase Src (Src), protein kinase B (Akt), mitogen activated protein kinase 1 (ERK), signal transducer and activator of transcription (STAT)3 and focal adhesion kinase 1 (FAK) was examined by western blotting. The levels of matrix metalloproteinase (MMP)-2 and MMP-9 were determined by enzyme-linked immunosorbent assay kits. Levels of reactive oxygen species (ROS) in cells were measured by a fluorescence plate reader with dichloro-dihydro-fluorescein diacetate. The results indicated that salidroside significantly suppressed cell proliferation and colony formation, inhibited cell migration and invasion, increased E-cadherin expression and decreased N-cadherin, MMP-2 and MMP-9 expression. Furthermore, salidroside suppressed ROS production and subsequently reduced the phosphorylation of Src, Akt, ERK and FAK. Salidroside also inhibited HSP70 expression, and HSP70 overexpression reversed the inhibitory effects of salidroside on BGC-823 cell proliferation, migration and invasion. In conclusion, the present study revealed that salidroside inhibited the proliferation, migration and invasion of BGC-823 cells by downregulating ROS-mediated Src-associated signaling pathway activation and HSP70 expression.
Collapse
Affiliation(s)
- Zhilin Qi
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Tuo Tang
- Anhui Province Key Laboratory of Active Biological Macromolecules, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Lili Sheng
- Department of Oncology, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Yunfei Ma
- Anhui Province Key Laboratory of Active Biological Macromolecules, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Yinhua Liu
- Department of Pathology, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Liang Yan
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Shimei Qi
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Liefeng Ling
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Yao Zhang
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
87
|
Chen KHE, Bustamante K, Nguyen V, Walker AM. Involvement of miR-106b in tumorigenic actions of both prolactin and estradiol. Oncotarget 2018; 8:36368-36382. [PMID: 28422740 PMCID: PMC5482661 DOI: 10.18632/oncotarget.16755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
Abstract
Prolactin promotes a variety of cancers by an array of different mechanisms. Here, we have investigated prolactin's inhibitory effect on expression of the cell cycle-regulating protein, p21. Using a miRNA array, we identified a number of miRNAs upregulated by prolactin treatment, but one in particular that was strongly induced by prolactin and predicted to bind to the 3′UTR of p21 mRNA, miR-106b. By creating a p21 mRNA 3′UTR-luciferase mRNA construct, we demonstrated degradation of the construct in response to prolactin in human breast, prostate and ovarian cancer cell lines. Increased expression of miR-106b replicated, and anti-miR-106b counteracted, the effects of prolactin on degradation of the 3′UTR construct, p21 mRNA levels, and cell proliferation in breast (T47D) and prostate (PC3) cancer cells. Increased expression of miR-106b also stimulated migration of the very epithelioid T47D cell line. By contrast, anti-miR-106b dramatically decreased expression of the mesenchymal markers, SNAIL-2, TWIST-2, VIMENTIN, and FIBRONECTIN. Using signaling pathway inhibitors and the 3′UTR construct, induction of miR-106b by prolactin was determined to be mediated through the MAPK/ERK and PI3K/Akt pathways and not through Jak2/Stat5 in both T47D and PC3 cells. Prolactin activation of MAPK/ERK and PI3K/Akt also activates ERα in the absence of an ERα ligand. 17β-estradiol promoted degradation of the construct in both cell lines and pre-incubation in the estrogen antagonist, Fulvestrant, blocked the ability of both prolactin and 17β-estradiol to induce the construct-degrading activity. Together, these data support a convergence of the prolactin and 17β-estradiol miR-106b-elevating signaling pathways at ERα.
Collapse
Affiliation(s)
- Kuan-Hui Ethan Chen
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Karissa Bustamante
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Vi Nguyen
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Ameae M Walker
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| |
Collapse
|
88
|
Yao W, Pan Z, Du X, Zhang J, Li Q. miR-181b-induced SMAD7 downregulation controls granulosa cell apoptosis through TGF-β signaling by interacting with the TGFBR1 promoter. J Cell Physiol 2018; 233:6807-6821. [PMID: 29319157 DOI: 10.1002/jcp.26431] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/05/2018] [Indexed: 12/25/2022]
Abstract
SMAD7 disrupts the TGF-β signaling pathway by influencing TGFBR1 stability and by blocking the binding of TGFBR1 to SMAD2/3. In this study, we showed that SMAD7 attenuated the TGF-β signaling pathway in ovarian granulosa cells (GCs) by regulating TGFBR1 transcriptional activity. To function as a transcription factor, SMAD7 downregulated the mRNA levels of TGFBR1 via direct binding to the SMAD-binding elements (SBEs) within the promoter region of pig TGFBR1. We also showed that SMAD7 enhanced porcine GC apoptosis by interrupting TGFBR1 and the TGF-β signaling pathway. Interestingly, miR-181b, a microRNA that is downregulated during porcine follicular atresia, was identified to be directly targeting SMAD7 at its 3'-UTR. By inhibiting SMAD7, miR-181b could inhibit GC apoptosis by activating the TGF-β signaling pathway. Our findings provide new insights into the mechanisms underlying the regulation of the TGF-β signaling pathway by SMAD7 and miR-181b.
Collapse
Affiliation(s)
- Wang Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zengxiang Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xing Du
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jinbi Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
89
|
Xue B, Lu QY, Massie L, Qualls C, Mao JT. Grape seed procyanidin extract against lung cancer: the role of microrna-106b, bioavailability, and bioactivity. Oncotarget 2018; 9:15579-15590. [PMID: 29643994 PMCID: PMC5884649 DOI: 10.18632/oncotarget.24528] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 02/10/2018] [Indexed: 12/19/2022] Open
Abstract
MiR-106b is an oncomir and a potential target for anti-cancer therapy. We hypothesize that grape seed procyanidin extract (GSE) exerts antineoplastic effects on lung cancer through modulations of miR-106b and its downstream target. We found that GSE significantly down-regulated miR-106b in a variety of lung neoplastic cells and increased cyclin-dependent kinase inhibitor 1A (CDKN1A) mRNA and protein (p21) levels. Transfection of miR-106b mimics reversed the up-regulations of CDKN1A mRNA and p21, abrogated the GSE induced anti-proliferative and anti-invasive properties in lung cancer cells. Oral gavage of leucoselect phytosome (LP), a standardized GSE to athymic nude mice down-regulated MIR106B mRNA and miR-106b expressions, and increased CDKN1A mRNA expression in tumor xenografts, correlating to significant reduction of tumor growth. To assess bioavailability, GSE and metabolites in plasma levels, between 60-90 minutes after gavage of LP were measured by LC/MS at treatment week 4 and 8. A novel bioactivity assay was also developed using lung homogenates from treated mice co-cultured with human lung cancer cells. LP-treated mouse lung homogenates significantly reduced proliferations of various lung cancer cells. Our findings reveal novel antineoplastic mechanisms by GSE, further define the pharmacokinetics and pharmacodynamics of LP, and support the continued investigation of LP against lung cancer.
Collapse
Affiliation(s)
- Bingye Xue
- Pulmonary, Critical Care, and Sleep Section, New Mexico Veterans Administration Health Care System, University of New Mexico, Biomedical Research Institute of New Mexico, Albuquerque, NM, USA
| | - Qing-Yi Lu
- UCLA Center for Human Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Larry Massie
- Pathology and Clinical Laboratory Services, New Mexico Veterans Administration Health Care System, University of New Mexico, Albuquerque, NM, USA
| | - Clifford Qualls
- Biomedical Research Institute of New Mexico, New Mexico Veterans Administration Health Care System, University of New Mexico, Albuquerque, NM, USA
| | - Jenny T. Mao
- Pulmonary, Critical Care, and Sleep Section, New Mexico Veterans Administration Health Care System, University of New Mexico, Biomedical Research Institute of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
90
|
PIM1 mediates epithelial-mesenchymal transition by targeting Smads and c-Myc in the nucleus and potentiates clear-cell renal-cell carcinoma oncogenesis. Cell Death Dis 2018; 9:307. [PMID: 29472550 PMCID: PMC5833424 DOI: 10.1038/s41419-018-0348-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/16/2017] [Accepted: 01/23/2018] [Indexed: 01/03/2023]
Abstract
Emerging evidence has shown that the PIM serine/threonine kinase family, including PIM1, PIM2 and PIM3, is associated with tumour progression towards metastasis. PIM1, an attractive molecular target, has been identified as a potential prognostic biomarker for haematological and epithelial malignancies. However, to date, the potential regulatory roles and molecular mechanisms by which PIM1 affects the development and progression of cancers, including clear-cell renal-cell carcinoma (ccRCC), remain largely unknown. Herein, we present the first evidence that PIM1 is aberrantly overexpressed in human ccRCC tissues and cell lines and positively correlated with human ccRCC progression. In our study, depletion of PIM1 attenuated ccRCC cell proliferation, colony formation, migration, invasion and angiogenesis, suggesting that PIM1 expression may be a cancer-promoting event in ccRCC. Mechanistically, we observed that PIM1 could interact with Smad2 or Smad3 in the nucleus and subsequently phosphorylate Smad2 and Smad3 to induce the expression of transcription factors, including ZEB1, ZEB2, Snail1, Snail2 and Twist, to promote epithelial-mesenchymal transition (EMT). In addition, PIM1-mediated phosphorylation of c-Myc activates the expression of the above transcription factors to synergistically promote EMT but does not activate Smads. Collectively, our results demonstrate that aberrant expression of PIM1 contributes to ccRCC development and progression. Moreover, our data reveal a potential molecular mechanism in which PIM1 mediates crosstalk between signalling pathways, including different Smad proteins and c-Myc, which target downstream transcription factors (ZEB1, ZEB2, Snail1, Snail2 and Twist) to trigger EMT. Together, our data suggest that PIM1 may be a potential therapeutic target for ccRCC patients.
Collapse
|
91
|
Fang C, Dai CY, Mei Z, Jiang MJ, Gu DN, Huang Q, Tian L. microRNA-193a stimulates pancreatic cancer cell repopulation and metastasis through modulating TGF-β2/TGF-βRIII signalings. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:25. [PMID: 29433538 PMCID: PMC5809917 DOI: 10.1186/s13046-018-0697-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/02/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Pancreatic cancer characterizes high recurrence and poor prognosis. In clinical practice, radiotherapy is widely used for pancreatic cancer treatment. However, the outcome remains undesirable due to tumor repopulation and following recurrence and metastasis after radiation. So, it is highly needed to explore the underlying molecular mechanisms and accordingly develop therapeutic strategies. Our previous studies revealed that dying cells from chemoradiation could stimulate repopulation of surviving pancreatic cancer cells. However, we still knew little how dying cells provoke pancreatic cancer cell repopulation. We herein would explore the significance of TGF-β2 changes and investigate the modulation of microRNA-193a (miR-193a), and identify their contributions to pancreatic cancer repopulation and metastasis. METHODS In vitro and in vivo repopulation models were established to mimic the biological processes of pancreatic cancer after radiation. Western blot, real-time PCR and dual-luciferase reporter assays were accordingly used to detect miR-193a and TGF-β2/TGF-βRIII signalings at the level of molecular, cellular and experimental animal model, respectively. Flow cytometry analysis, wound healing and transwell assay, vascular endothelial cell penetration experiment, and bioluminescence imaging were employed to assessthe biological behaviors of pancreatic cancer after different treatments. Patient-derived tumor xenograft (PDX) mice models were established to evaluate the therapeutic potential of miR-193a antagonist on pancreatic cancer repopulation and metastasis after radiation. RESULTS miR-193a was highly expressed in the irradiated pancreatic cancer dying cells, accordingly elevated the level of miR-193a in surviving cells, and further promoted pancreatic cancer repopulation and metastasis in vitro and in vivo. miR-193a accelerated pancreatic cancer cell cycle and stimulated cell proliferation and repopulation through inhibiting TGF-β2/TGF-βRIII/SMADs/E2F6/c-Myc signaling, and even destroyed normal intercellular junctions and promoted metastasis via repressing TGF-β2/TGF-βRIII/ARHGEF15/ABL2 pathway. Knockdown of miR-193a or restoration of TGF-β2/TGF-βRIII signaling in pancreatic cancer cells was found to block pancreatic cancer repopulation and metastasis after radiation. In PDX models, the treatment in combination with miR-193a antagonist and radiation was found to dramatically inhibit pancreatic cancer cell repopulation and metastasis, and further improved the survival after radiation. CONCLUSIONS Our findings demonstrated that miR-193a stimulated pancreatic cancer cell repopulation and metastasis through modulating TGF-β2/TGF-βRIII signalings, and miR-193a might be a potential therapeutic target for pancreatic cancer repopulation and metastasis.
Collapse
Affiliation(s)
- Chi Fang
- Institute of Translational Medicine, Science bldg. Rm 205, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, New Songjiang Rd No.650, Songjiang District, Shanghai, 201620, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Yun Dai
- Institute of Translational Medicine, Science bldg. Rm 205, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, New Songjiang Rd No.650, Songjiang District, Shanghai, 201620, China
| | - Zhu Mei
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Jie Jiang
- Institute of Translational Medicine, Science bldg. Rm 205, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, New Songjiang Rd No.650, Songjiang District, Shanghai, 201620, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dian-Na Gu
- Institute of Translational Medicine, Science bldg. Rm 205, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, New Songjiang Rd No.650, Songjiang District, Shanghai, 201620, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Huang
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,The Comprehensive Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Tian
- Institute of Translational Medicine, Science bldg. Rm 205, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, New Songjiang Rd No.650, Songjiang District, Shanghai, 201620, China. .,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
92
|
Dong X, Hu X, Chen J, Hu D, Chen LF. BRD4 regulates cellular senescence in gastric cancer cells via E2F/miR-106b/p21 axis. Cell Death Dis 2018; 9:203. [PMID: 29434197 PMCID: PMC5833665 DOI: 10.1038/s41419-017-0181-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 12/27/2022]
Abstract
Small molecules targeting bromodomains of BET proteins possess strong anti-tumor activities and have emerged as potential therapeutics for cancer. However, the underlying mechanisms for the anti-proliferative activity of these inhibitors are still not fully characterized. In this study, we demonstrated that BET inhibitor JQ1 suppressed the proliferation and invasiveness of gastric cancer cells by inducing cellular senescence. Depletion of BRD4, which was overexpressed in gastric cancer tissues, but not other BET proteins recapitulated JQ1-induced cellular senescence with increased cellular SA-β-Gal activity and elevated p21 levels. In addition, we showed that the levels of p21 were regulated at the post-transcriptional level by BRD4-dependent expression of miR-106b-5p, which targets the 3'-UTR of p21 mRNA. Overexpression of miR-106b-5p prevented JQ1-induced p21 expression and BRD4 inhibition-associated cellular senescence, whereas miR-106b-5p inhibitor up-regulated p21 and induced cellular senescence. Finally, we demonstrated that inhibition of E2F suppressed the binding of BRD4 to the promoter of miR-106b-5p and inhibited its transcription, leading to the increased p21 levels and cellular senescence in gastric cancer cells. Our results reveal a novel mechanism by which BRD4 regulates cancer cell proliferation by modulating the cellular senescence through E2F/miR-106b-5p/p21 axis and provide new insights into using BET inhibitors as potential anticancer drugs.
Collapse
Affiliation(s)
- Xingchen Dong
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Xiangming Hu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jinjing Chen
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Dan Hu
- Department of Pathology, Fujian Provincial Cancer Hospital, The Affiliated Hospital of Fujian Medical University, Fujian, China, 350108
| | - Lin-Feng Chen
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China, 350108.
- Department of Medical Biochemistry, College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
93
|
Jafari SH, Saadatpour Z, Salmaninejad A, Momeni F, Mokhtari M, Nahand JS, Rahmati M, Mirzaei H, Kianmehr M. Breast cancer diagnosis: Imaging techniques and biochemical markers. J Cell Physiol 2018; 233:5200-5213. [PMID: 29219189 DOI: 10.1002/jcp.26379] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 12/04/2017] [Indexed: 12/13/2022]
Abstract
Breast cancer is a complex disease which is found as the second cause of cancer-associated death among women. Accumulating of evidence indicated that various factors (i.e., gentical and envirmental factors) could be associated with initiation and progression of breast cancer. Diagnosis of breast cancer patients in early stages is one of important aspects of breast cancer treatment. Among of various diagnosis platforms, imaging techniques are main diagnosis approaches which could provide valuable data on patients with breast cancer. It has been showed that various imaging techniques such as mammography, magnetic resonance imaging (MRI), positron-emission tomography (PET), Computed tomography (CT), and single-photon emission computed tomography (SPECT) could be used for diagnosis and monitoring patients with breast cancer in various stages. Beside, imaging techniques, utilization of biochemical biomarkers such as proteins, DNAs, mRNAs, and microRNAs could be employed as new diagnosis and therapeutic tools for patients with breast cancer. Here, we summarized various imaging techniques and biochemical biomarkers could be utilized as diagnosis of patients with breast cancer. Moreover, we highlighted microRNAs and exosomes as new diagnosis and therapeutic biomarkers for monitoring patients with breast cancer.
Collapse
Affiliation(s)
- Seyed Hamed Jafari
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Saadatpour
- Radiology Specialist at Bozorgmehr Imaging Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arash Salmaninejad
- Drug Applied Research Center, Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
| | - Fatemeh Momeni
- General Practitioner, Medical Researcher, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mojgan Mokhtari
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Javid Sadri Nahand
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Rahmati
- Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Kianmehr
- Department of Medical Physics, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
94
|
Verboon LJ, Obulkasim A, de Rooij JDE, Katsman-Kuipers JE, Sonneveld E, Baruchel A, Trka J, Reinhardt D, Pieters R, Cloos J, Kaspers GJL, Klusmann JH, Zwaan CM, Fornerod M, van den Heuvel-Eibrink MM. MicroRNA-106b~25 cluster is upregulated in relapsed MLL-rearranged pediatric acute myeloid leukemia. Oncotarget 2018; 7:48412-48422. [PMID: 27351222 PMCID: PMC5217027 DOI: 10.18632/oncotarget.10270] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 06/08/2016] [Indexed: 12/13/2022] Open
Abstract
The most important reason for therapy failure in pediatric acute myeloid leukemia (AML) is relapse. In order to identify miRNAs that contribute to the clonal evolution towards relapse in pediatric AML, miRNA expression profiling of 127 de novo pediatric AML cases were used. In the diagnostic phase, no miRNA signatures could be identified that were predictive for relapse occurrence, in a large pediatric cohort, nor in a nested mixed lineage leukemia (MLL)-rearranged pediatric cohort. AML with MLL- rearrangements are found in 15-20% of all pediatric AML samples, and reveal a relapse rate up to 50% for certain translocation partner subgroups. Therefore, microRNA expression profiling of six paired initial diagnosis-relapse MLL-rearranged pediatric AML samples (test cohort) and additional eight paired initial diagnosis-relapse samples with MLL-rearrangements (validation cohort) was performed. A list of 53 differentially expressed miRNAs was identified of which the miR-106b~25 cluster, located in intron 13 of MCM7, was the most prominent. These differentially expressed miRNAs however could not predict a relapse in de novo AML samples with MLL-rearrangements at diagnosis. Furthermore, higher mRNA expression of both MCM7 and its upstream regulator E2F1 was found in relapse samples with MLL-rearrangements. In conclusion, we identified the miR-106b~25 cluster to be upregulated in relapse pediatric AML with MLL-rearrangements.
Collapse
Affiliation(s)
- Lonneke J Verboon
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital Rotterdam, Rotterdam, The Netherlands
| | - Askar Obulkasim
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital Rotterdam, Rotterdam, The Netherlands
| | - Jasmijn D E de Rooij
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital Rotterdam, Rotterdam, The Netherlands
| | - Jenny E Katsman-Kuipers
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital Rotterdam, Rotterdam, The Netherlands
| | - Edwin Sonneveld
- Dutch Childhood Oncology Group (DCOG), The Hague, The Netherlands
| | - André Baruchel
- Department of Hematology, Hopital Saint- Louis, Paris, France
| | - Jan Trka
- Department of Pediatric Hematology/Oncology, 2nd Medical School, Charles University, Prague, Czech Republic
| | - Dirk Reinhardt
- Clinic for Pediatrics III, University Hospital Essen, Essen, Germany
| | - Rob Pieters
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jacqueline Cloos
- Paediatric Oncology/Haematology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Gertjan J L Kaspers
- Paediatric Oncology/Haematology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Jan-Henning Klusmann
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Christian Michel Zwaan
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital Rotterdam, Rotterdam, The Netherlands
| | - Maarten Fornerod
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital Rotterdam, Rotterdam, The Netherlands
| | - Marry M van den Heuvel-Eibrink
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital Rotterdam, Rotterdam, The Netherlands.,Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
95
|
Majumder M, Dunn L, Liu L, Hasan A, Vincent K, Brackstone M, Hess D, Lala PK. COX-2 induces oncogenic micro RNA miR655 in human breast cancer. Sci Rep 2018; 8:327. [PMID: 29321644 PMCID: PMC5762661 DOI: 10.1038/s41598-017-18612-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 12/14/2017] [Indexed: 01/16/2023] Open
Abstract
We show that Cyclooxygenase-2 over-expression induces an oncogenic microRNA miR655 in human breast cancer cells by activation of EP4. MiR655 expression positively correlated with COX-2 in genetically disparate breast cancer cell lines and increased in all cell lines when grown as spheroids, implicating its link with stem-like cells (SLCs). Ectopic miR655 over-expression in MCF7 and SKBR3 cells resulted in increased proliferation, migration, invasion, spheroid formation and Epithelial to Masenchymal transition (EMT). Conversely, knocking down miR655 in aggressive MCF7-COX2 and SKBR3-COX2 cells reverted these phenotypes. MCF7-miR655 cells displayed upregulated NOTCH/WNT genes; both pathway inhibitors abrogated miR655-induced spheroid formation, linking miR655 with SLC-related pathways. MiR655 expression was dependent on EP4 activity and EP4 downstream signaling pathways PI3K/AKT, ERK and NF-kB and led to TGFβ resistance for Smad3 phosphorylation. Tail vein injection of MCF7-miR655 and SKBR3-miR655 cells in NOD/SCID/GUSB-null mice revealed increased lung colony growth and micrometastases to liver and spleen. MiR655 expression was significantly high in human breast tumors (n = 105) compared to non-tumor tissues (n = 20) and associated with reduced patient survival. Thus miR655 could serve as a prognostic breast cancer biomarker.
Collapse
Affiliation(s)
- Mousumi Majumder
- Department of Biology, Brandon University, Brandon, Manitoba, Canada
| | - Leanna Dunn
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Ling Liu
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Asma Hasan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Krista Vincent
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Muriel Brackstone
- Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Lawson Research Institute, London, Ontario, Canada
| | - David Hess
- Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, London, Ontario, Canada
| | - Peeyush K Lala
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
- Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
96
|
Chen H, Pan H, Qian Y, Zhou W, Liu X. MiR-25-3p promotes the proliferation of triple negative breast cancer by targeting BTG2. Mol Cancer 2018; 17:4. [PMID: 29310680 PMCID: PMC5759260 DOI: 10.1186/s12943-017-0754-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/26/2017] [Indexed: 12/11/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is highly invasive and aggressive and lacks specific molecular targets to improve the prognosis. MiR-25-3p promotes proliferation of many tumors and its role and underlying mechanisms in TNBC remain to be well elucidated. Methods Differential expression of miR-25-3p in TNBC was measured with quantitative real-time PCR (qRT-PCR) in both TNBC tissues and cell lines and was validated in the Cancer Genome Atlas (TCGA) database. The effects of miR-25-3p on proliferation, apoptosis capacity of TNBC were evaluated using Cell counting kit-8 (CCK-8), colony formation assay and Annexin V-FITC/PI analyses. The tumor growth in vivo was observed in xenograft model. Luciferase reporter assay, qPCR and western blot were performed to validate a potential target of miR-25-3p in TNBC. Involvement of the AKT and MAPK pathways was investigated by western blot. Results MiR-25-3p was found to be upregulated in TNBC in tissues and cell lines. MiR-25-3p promoted TNBC cell proliferation in vitro and tumor growth in xenograft model, while suppression of miR-25-3p induced cell apoptosis. The luciferase reporter assay confirmed that B-cell translocation gene 2 (BTG2) might be a direct target of miR-25-3p, and its expression was negatively regulated by miR-25-3p. Moreover, inhibition of BTG2 expression accounted for the role of miR-25-3p in TNBC. Furthermore, BTG2 suppression might indirectly activate the AKT and ERK-MAPK signaling pathways to mediate the downstream effects of miR-25-3p. Conclusions This study demonstrates that miR-25-3p promotes proliferation by targeting tumor suppressor BTG2 and may identify new diagnostic and therapeutic targets in TNBC. Electronic supplementary material The online version of this article (10.1186/s12943-017-0754-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hua Chen
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Hong Pan
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Yi Qian
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Wenbin Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Xiaoan Liu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China.
| |
Collapse
|
97
|
TGF-β1 targets a microRNA network that regulates cellular adhesion and migration in renal cancer. Cancer Lett 2018; 412:155-169. [DOI: 10.1016/j.canlet.2017.10.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 01/07/2023]
|
98
|
Wang M, Yang YO, Jin Q, Shang L, Zhang J. Function of miR-25 in the invasion and metastasis of esophageal squamous carcinoma cells and bioinformatical analysis of the miR-106b-25 cluster. Exp Ther Med 2017; 15:440-446. [PMID: 29250158 DOI: 10.3892/etm.2017.5358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 09/06/2017] [Indexed: 01/07/2023] Open
Abstract
MicroRNAs (miRNAs/miRs) are a class of small, non-coding RNA molecules that serve a key function in carcinogenesis and tumor progression. Recent evidence indicates that miRNAs may act as powerful regulators of migration and invasion. The present study aimed to investigate the effect of miR-25 on the invasion and metastasis of KYSE-150 and EC109 esophageal squamous cell carcinoma (ESCC) cells, and predict the mechanism of this effect by bioinformatically analyzing the miR-106b-25 cluster. In order to alter the expression of miR-25 in the two cell lines, a miR-25 inhibitor or mimic were transfected into the cells, which were then studied via Transwell migration and invasion assays. Subsequently, the target genes of the miR-106b-25 cluster were predicted using miRanda, PicTar, TargetScan and miRTarbase, and the functions of the target genes were predicted via Gene Ontology term and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses. Then, a protein-protein interaction (PPI) network was produced using the Search Tool for the Retrieval of Interacting Genes. The results revealed that overexpressing miR-25 led to significantly increased cell migration and invasion in KYSE150 and EC109 cells. Suppressing miR-25 resulted in significantly decreased cell migration and invasion in KYSE150 cells, while the result was not significant in EC109 cells. Target genes of the miR-106b-25 cluster were significantly enriched in the biological process regulation of cellular metabolic process and several cancer-associated pathways, such as those for glioma and melanoma. The PPI network revealed that PTEN, TP53, MDM2, E2F1, PRMT5, MCM2, RB1, CDKN1A, SHAD7 and EZH2 may serve core roles within the network and associate with one another during the pathogenesis of ESCC. These results indicate that a high expression of miR-25 promotes the invasion and metastasis of ESCC cells, while the influence of low expression of miR-25 differs with cells with different degrees of differentiation. Invasion and metastasis are not effected in cells with poor differentiation, while they were decreased in well differentiated cells. Furthermore, PTEN, TP53, MDM2, E2F1, PRMT5, MCM2, RB1, CDKN1A, SHAD7 and EZH2 may be targeted by the miR-106b-25 cluster, and act together to regulate the development of ESCC.
Collapse
Affiliation(s)
- Meng Wang
- Medical Department, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Yangyang Ou Yang
- Department of Gastroenterology, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Qingtao Jin
- Department of Gastroenterology, Jiaxiang People's Hospital, Jining, Shandong 272400, P.R. China
| | - Linlin Shang
- Department of Pharmacy, Jining No. 2 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Jian Zhang
- Department of Gastroenterology, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
99
|
miR-143 and miR-145 inhibit gastric cancer cell migration and metastasis by suppressing MYO6. Cell Death Dis 2017; 8:e3101. [PMID: 29022908 PMCID: PMC5682659 DOI: 10.1038/cddis.2017.493] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/26/2017] [Accepted: 08/28/2017] [Indexed: 01/07/2023]
Abstract
Metastasis is a major clinical obstacle responsible for the high mortality and poor prognosis of gastric cancer (GC). MicroRNAs (miRNAs) are critical mediators of metastasis that act by modulating their target genes. In this study, we found that miR-143 and miR-145 act via a common target gene, MYO6, to regulate the epithelial–mesenchymal transition (EMT) and inhibit metastasis. We determined that miR-143 and miR-145 were downregulated in GC, and the ectopic expression of miR-143 and/or miR-145 inhibited GC cell migration and metastasis. Furthermore, MYO6 was identified as a direct common target of miR-143 and miR-145 and was elevated in GC. Silencing of MYO6 resulted in a metastasis-suppressive activity similar to that of miR-143 and miR-145, while restoring MYO6 attenuated the anti-metastatic or anti-EMT effects caused by miR-143 and miR-145. Clinically, an inverse correlation was observed between miR-143/145 levels and MYO6 levels in GC tissues, and either miR-143/145 downregulation or MYO6 upregulation was associated with more malignant phenotypes in patients with GC. In conclusion, miR-143 and miR-145 suppress GC cell migration and metastasis by inhibiting MYO6 expression and the EMT, which provides a novel mechanism and promising therapeutic target for the treatment of GC metastasis.
Collapse
|
100
|
Liu J, Zhu G, Xu S, Liu S, Lu Q, Tang Z. Analysis of miRNA expression profiling in human umbilical vein endothelial cells affected by heat stress. Int J Mol Med 2017; 40:1719-1730. [PMID: 29039486 PMCID: PMC5716433 DOI: 10.3892/ijmm.2017.3174] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/28/2017] [Indexed: 12/20/2022] Open
Abstract
To investigate the regulation of endothelial cell (EC) microRNAs (miRNAs) altered by heat stress, miRNA microarrays and bioinformatics methods were used to determine changes in miRNA profiles and the pathophysiological characteristics of differentially expressed miRNAs. A total of 31 differentially expressed miRNAs were identified, including 20 downregulated and 11 upregulated miRNAs. Gene Ontology (GO) enrichment analysis revealed that the validated targets of the differentially expressed miRNAs were significantly enriched in gene transcription regulation. The pathways were also significantly enriched in the Kyoto Encyclopedia of Genes and Genomes analysis, and most were cancer-related, including the mitogen-activated protein kinase signaling pathway, pathways involved in cancer, the Wnt signaling pathway, the Hippo signaling pathway, proteoglycans involved in cancer and axon guidance. The miRNA-gene and miRNA-GO network analyses revealed several hub miRNAs, genes and functions. Notably, miR-3613-3p played a dominant role in both networks. MAP3K2, MGAT4A, TGFBR1, UBE2R2 and SMAD4 were most likely to be controlled by the altered miRNAs in the miRNA-gene network. The miRNA-GO network analysis revealed significantly complicated associations between miRNAs and different functions, and that the significantly enriched functions targeted by the differentially expressed miRNAs were mostly involved in regulating gene transcription. The present study demonstrated that miRNAs are involved in the pathophysiology of heat-treated ECs. Understanding the functions of miRNAs may provide novel insights into the molecular mechanisms underlying the heat-induced pathophysiology of ECs.
Collapse
Affiliation(s)
- Jie Liu
- Department of Emergency, Wuhan General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| | - Guoguo Zhu
- Department of Emergency, Wuhan General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| | - Siya Xu
- Department of Emergency, Wuhan General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| | - Shixin Liu
- Department of Emergency, Wuhan General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| | - Qiping Lu
- Department of General Surgery, Wuhan General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| | - Zhongzhi Tang
- Department of Emergency, Wuhan General Hospital of People's Liberation Army of China, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|