101
|
Lei Z, Chai N, Tian M, Zhang Y, Wang G, Liu J, Tian Z, Yi X, Chen D, Li X, Yu P, Hu H, Xu B, Jian C, Bian Z, Guo H, Wang J, Peng S, Nie Y, Huang N, Hu S, Wu K. Novel peptide GX1 inhibits angiogenesis by specifically binding to transglutaminase-2 in the tumorous endothelial cells of gastric cancer. Cell Death Dis 2018; 9:579. [PMID: 29785022 PMCID: PMC5962530 DOI: 10.1038/s41419-018-0594-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/21/2018] [Accepted: 04/12/2018] [Indexed: 12/28/2022]
Abstract
The clinical application of GX1, an optimal gastric cancer (GC) targeting peptide, is greatly limited because its receptor in the GC vasculature is unknown. In this study, we screened the candidate receptor of GX1, transglutaminase-2(TGM2), by co-immunoprecipitation (co-IP) combined with mass spectrometry. We found that TGM2 was up-regulated in GC vascular endothelial cells and that GX1 receptor expression was suppressed correspondingly after TGM2 downregulation. A highly consistent co-localization of GX1 receptor and TGM2 was detected at both the cellular and tissue levels. High TGM2 expression was evident in GC tissues from patients with poor prognosis. After TGM2 downregulation, the GX1-mediated inhibition of proliferation and migration and the induction of the apoptosis of GC vascular endothelial cells were weakened or even reversed. Finally, we observed that GX1 could inhibit the GTP-binding activity of TGM2 by reducing its intracellular distribution and downregulating its downstream molecular targets (nuclear factor-kappa B, NF-κB; hypoxia-inducible factor 1-α, HIF1α) in GC vascular endothelial cells. Our study confirms that peptide GX1 can inhibit angiogenesis by directly binding to TGM2, subsequently reducing the GTP-binding activity of TGM2 and thereby suppressing its downstream pathway(NF-κB/HIF1α). Our conclusions suggest that GX1/TGM2 may provide a new target for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Zhijie Lei
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Na Chai
- Department of Radiology, Xjing Hospital of Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Miaomiao Tian
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Ying Zhang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Guodong Wang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Jian Liu
- Department of Radiology, Xjing Hospital of Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Zuhong Tian
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Xiaofang Yi
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Di Chen
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Xiaowei Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Pengfei Yu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Hao Hu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Bing Xu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Chao Jian
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Zhenyuan Bian
- Department of Hepatobiliary Surgery, Xjing Hospital of Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Hao Guo
- Department of Neurosurgery, Tangdu Hospital of Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, People's Republic of China
| | - Jinpeng Wang
- Department of Orthopedics, Xjing Hospital of Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Shiming Peng
- National Institute of Biological Sciences, Beijing, 102206, People's Republic of China
| | - Yongzhan Nie
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China
| | - Niu Huang
- National Institute of Biological Sciences, Beijing, 102206, People's Republic of China.
| | - Sijun Hu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China.
| | - Kaichun Wu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, Shaanxi Province, People's Republic of China.
| |
Collapse
|
102
|
[MicroRNA-218 promotes osteosarcoma cell apoptosis by down-regulating oncogene B lymphoma mouse Moloney leukemia virus insertion region 1]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38. [PMID: 29891444 PMCID: PMC6743892 DOI: 10.3969/j.issn.1673-4254.2018.05.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To investigate the tumor-suppressing effect of microRNA-218 (miR-218) in osteosarcoma (OS) and explore its molecular mechanism. METHODS We examined the expression levels of miR-218 in 68 pairs of OS and adjacent tissue samples using qRT-PCR.Cultured human OS cell line Saos-2 was transfected with miR-218 mimics or anti-miR-218 mimics, and the cell apoptosis was assessed using CCK-8 assay, annexin V-FITC staining and Western blotting.We also analyzed the potential functional targets of miR-218 in Saos-2 cells using luciferase assay, qRT-PCR and Western blotting. RESULTS The expression level of miR-218 was lowered by at least 8 folds in OS tissues as compared with the adjacent tissues.In cultured Saos-2 cells, transfection with miR-218 mimics for 24, 36, and 48 h resulted in a significant reduction in the cell viability, while transfection with anti-miR-218 mimics significantly increased the cell viability.The cells transfected with miR-218 mimics showed an obviously enhanced expression of cleaved poly (ADP-ribose) polymerase (C-PARP) as compared with the cells transfected with anti-miR-218 mimics and the control cells.Flow cytometry demonstrated obviously increased apoptosis of the cells following miR-218 mimics transfection.We identified the oncogene B lymphoma mouse Moloney leukemia virus insertion region 1 (BMI-1) as a specific target of miR-218 in Saos-2 cells. BMI-1 expressions at both the mRNA and protein levels were significantly reduced in Saos-2 cells overexpressing miR-218 but increased in the cells with miR-218 knockdown as compared to the control cells.Luciferase reporter assay indicated that miR-218 directly inhibited the expression of BMI-1 via binding to its 3'-UTR in OS cells. CONCLUSION miR-218 can promote OS cell apoptosis and plays the role as a tumor suppressor by down-regulating BMI-1.
Collapse
|
103
|
Volinia S, Bertagnolo V, Grassilli S, Brugnoli F, Manfrini M, Galasso M, Scatena C, Mazzanti CM, Lessi F, Naccarato G, Caligo A, Bianchini E, Piubello Q, Orvieto E, Rugge M, Natali C, Reale D, Vecchione A, Warner S, Croce CM, Capitani S. Levels of miR-126 and miR-218 are elevated in ductal carcinoma in situ (DCIS) and inhibit malignant potential of DCIS derived cells. Oncotarget 2018; 9:23543-23553. [PMID: 29805754 PMCID: PMC5955110 DOI: 10.18632/oncotarget.25261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/06/2018] [Indexed: 12/21/2022] Open
Abstract
A substantial number of ductal carcinoma in situ (DCIS) detected by mammography never progress to invasive ductal carcinoma (IDC) and current approaches fail to identify low-risk patients not at need of adjuvant therapies. We aimed to identify the key miRNAs protecting DCIS from malignant evolution, that may constitute markers for non-invasive lesions. We studied 100 archived DCIS samples, including pure DCIS, DCIS with adjacent IDC and pure DCIS from patients with subsequent IDC in contralateral breast or no recurrence. A DCIS derived cell line was used for molecular and cellular studies. A genome wide study revealed that pure DCIS has higher miR-126 and miR-218 expression than DCIS with adjacent IDC lesions or than IDC. The down-regulation of miR-126 and miR-218 promoted invasiveness in vitro and, in patients with pure DCIS, was associated with later onset of IDC. Survival studies of independent cohorts indicated that both miRNAs play a protective role in IDC. The clinical findings are in agreement with the miRNAs' roles in cell adhesion, differentiation and proliferation. We propose that miR-126 and miR-218 have a protective role in DCIS and represent novel biomarkers for the risk assessment in women with early detection of breast cancer.
Collapse
Affiliation(s)
- Stefano Volinia
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44121, Italy.,LTTA Centre, University of Ferrara, Ferrara 44121, Italy
| | - Valeria Bertagnolo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44121, Italy
| | - Silvia Grassilli
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44121, Italy
| | - Federica Brugnoli
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44121, Italy
| | - Marco Manfrini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44121, Italy
| | - Marco Galasso
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44121, Italy
| | - Cristian Scatena
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56126, Italy
| | | | | | - Giuseppe Naccarato
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56126, Italy
| | - Adelaide Caligo
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56126, Italy
| | - Enzo Bianchini
- Pathology Division, S. Anna University Hospital, Ferrara 44124, Italy
| | - Quirino Piubello
- Department of Diagnostic and Pathology, Azienda Ospedaliera Universitaria Integrata di Verona, Verona 37126, Italy
| | - Enrico Orvieto
- Department of Medicine DIMED, University of Padova, Padova 35121, Italy
| | - Massimo Rugge
- Department of Medicine DIMED, University of Padova, Padova 35121, Italy
| | - Cristina Natali
- Pathology Division, Santa Maria della Misericordia Hospital, Rovigo 45100, Italy
| | - Domenico Reale
- Pathology Division, Santa Maria della Misericordia Hospital, Rovigo 45100, Italy
| | - Andrea Vecchione
- Department of Pathology, St. Andrea University Hospital, University of Rome, La Sapienza, Rome 00185, Italy
| | - Sarah Warner
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Carlo Maria Croce
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Silvano Capitani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44121, Italy.,LTTA Centre, University of Ferrara, Ferrara 44121, Italy
| |
Collapse
|
104
|
Li X, Jiang M, Chen D, Xu B, Wang R, Chu Y, Wang W, Zhou L, Lei Z, Nie Y, Fan D, Shang Y, Wu K, Liang J. miR-148b-3p inhibits gastric cancer metastasis by inhibiting the Dock6/Rac1/Cdc42 axis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:71. [PMID: 29587866 PMCID: PMC5872400 DOI: 10.1186/s13046-018-0729-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/08/2018] [Indexed: 12/19/2022]
Abstract
Background Our previous work showed that some Rho GTPases, including Rho, Rac1 and Cdc42, play critical roles in gastric cancer (GC); however, how they are regulated in GC remains largely unknown. In this study, we aimed to investigate the roles and molecular mechanisms of Dock6, an atypical Rho guanine nucleotide exchange factor (GEF), in GC metastasis. Methods The expression levels of Dock6 and miR-148b-3p in GC tissues and paired nontumor tissues were determined by immunohistochemistry (IHC) and in situ hybridization (ISH), respectively. The correlation between Dock6/miR-148b-3p expression and the overall survival of GC patients was calculated by the Kaplan-Meier method and log-rank test. The roles of Dock6 and miR-148b-3p in GC were investigated by in vitro and in vivo functional studies. Rac1 and Cdc42 activation was investigated by GST pull-down assays. The inhibition of Dock6 transcription by miR-148b-3p was determined by luciferase reporter assays. Results A significant increase in Dock6 expression was found in GC tissues compared with nontumor tissues, and its positive expression was associated with lymph node metastasis and a higher TNM stage. Patients with positive Dock6 expression exhibited shorter overall survival periods than patients with negative Dock6 expression. Dock6 promoted GC migration and invasion by increasing the activation of Rac1 and Cdc42. miR-148b-3p expression was negatively correlated with Dock6 expression in GC, and it decreased the motility of GC cells by inhibiting the Dock6/Rac1/Cdc42 axis. Conclusions Dock6 was over-expressed in GC tissues, and its positive expression was associated with GC metastasis and indicated poor prognosis of GC patients. Targeting of Dock6 by miR-148b-3p could activate Rac1 and Cdc42, directly affecting the motility of GC cells. Targeting the Dock6-Rac1/Cdc42 axis could serve as a new therapeutic strategy for GC treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-0729-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaowei Li
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Mingzuo Jiang
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Di Chen
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Bing Xu
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China.,Department of Gastroenterology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710004, China
| | - Rui Wang
- National-Local Joint Engineering Research Center of Biodiagnostics & Biotheraphy, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710032, China
| | - Yi Chu
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Weijie Wang
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Lin Zhou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Zhijie Lei
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Yulong Shang
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China.
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China.
| | - Jie Liang
- State Key Laboratory of Cancer Biology & National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
105
|
Guan B, Wu K, Zeng J, Xu S, Mu L, Gao Y, Wang K, Ma Z, Tian J, Shi Q, Guo P, Wang X, He D, Du Y. Tumor-suppressive microRNA-218 inhibits tumor angiogenesis via targeting the mTOR component RICTOR in prostate cancer. Oncotarget 2018; 8:8162-8172. [PMID: 28030804 PMCID: PMC5352391 DOI: 10.18632/oncotarget.14131] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 11/01/2016] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs, a kind of small non-coding RNAs, can regulate gene expression by targeting mRNAs for translational repression or degradation. Much evidence has suggested that miR-218 was a tumor suppressor in many human cancers including prostate cancer. However, the underlying role of miR-218 in tumor angiogenesis and the mechanisms in PCa and other cancers remains to be unclear. Here in this present study, we demonstrated that miR-218 inhibited the tumor angiogenesis of PCa cells in vitro and in vivo. RICTOR, the mTOR component 2, was a direct target of miR-218 and miR218-RICTOR-VEGFA axis was the mechanism inhibiting the tumor angiogenesis of PCa cells. RICTOR knockdown phenocopied miR-218 overexpression in inhibiting prostate cancer angiogenesis. Altogether, our findings indicate that down-regulation of miR-218 contributes to tumor angiogenesis through RICTOR/VEGFA axis in PCa, providing new insights into the potential mechanisms of PCa oncogenesis and revealing the potential of miR-218 as a useful serum biomarker and a new therapeutic target for human PCa.
Collapse
Affiliation(s)
- Bing Guan
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Kaijie Wu
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Laboratory, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Jin Zeng
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Laboratory, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Shan Xu
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Laboratory, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Lijun Mu
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yang Gao
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ke Wang
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Laboratory, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Zhenkun Ma
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Laboratory, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Juanhua Tian
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qi Shi
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Peng Guo
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Laboratory, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Xinyang Wang
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Laboratory, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Dalin He
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Laboratory, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yuefeng Du
- Department of Urology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Oncology Research Laboratory, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| |
Collapse
|
106
|
Zhang G, Chen L, Khan AA, Li B, Gu B, Lin F, Su X, Yan J. miRNA-124-3p/neuropilin-1(NRP-1) axis plays an important role in mediating glioblastoma growth and angiogenesis. Int J Cancer 2018; 143:635-644. [PMID: 29457830 DOI: 10.1002/ijc.31329] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/29/2017] [Accepted: 01/30/2018] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is the most lethal brain malignancy which involves multi-gene abnormality. Unfortunately, effective therapy against GBM remains lacking. Previously, we found that NRP-1 and its downstream NRP-1/GIPC1 pathway played an important role in GBM. In our study, we further investigated the upstream signaling of NRP-1 to understand how it is regulated. First, we identified that hsa-miR-124-3p was miRNA differentially expressed in GBM and in normal brain tissues by high-throughput sequencing. Then, by dual luciferase reporter gene, we found miR-124-3p can specially bind to the 3'UTR region of the NRP-1 thus suppresses its expression. Moreover, miR-124-3p overexpression significantly inhibited GBM cell proliferation, migration and tumor angiogenesis which resulted in GBM apoptosis and cell cycle arrest, putatively via NRP-1 mediated PI3K/Akt/NFκB pathways activation in GBM cells. Meanwhile, miR-124-3p overexpression also suppressed tumor growth and reduced tumor angiogenesis when targeted by NRP-1 in a PDX model. Furthermore, NRP-1 mAb exerted synergistic inhibitory effects with miR-124-3p overexpression in GBM. Thus, we discovered that miR-124-3p acts as the upstream suppressor of NRP-1 which promotes GBM cell development and growth by PI3K/Akt/NFκB pathway. The miR-124-3p/NRP-1/GIPC1 pathway as a new pathway has a vital role in GBM, and it could be considered as the potential target for malignant gliomas in future.
Collapse
Affiliation(s)
- Guilong Zhang
- Department of Neurosurgery, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Lukui Chen
- Department of Neurosurgery, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Ahsan Ali Khan
- Department of Neurosurgery, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Bingqian Li
- Department of Neurosurgery, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Bin Gu
- Department of Neurosurgery, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Fan Lin
- Department of Cell Biology, Nanjing Medical University, Nanjing, 210029, China.,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 210029, China
| | - Xinhui Su
- Department of Nuclear Medicine, Zhongshan Hospital Xiamen University, Xiamen, 361004, China
| | - Jianghua Yan
- Medical College, Cancer Research Center, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
107
|
Zhang L, Zhang Y, Wong SH, Law PTY, Zhao S, Yu J, Chan MTV, Wu WKK. Common Deregulation of Seven Biological Processes by MicroRNAs in Gastrointestinal Cancers. Sci Rep 2018; 8:3287. [PMID: 29459716 PMCID: PMC5818544 DOI: 10.1038/s41598-018-21573-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 02/07/2018] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs are frequently dysregulated in human neoplasms, including gastrointestinal cancers. Nevertheless, the global influence of microRNA dysregulation on cellular signaling is still unknown. Here we sought to elucidate cellular signaling dysregulation by microRNAs in gastrointestinal cancers at the systems biology level followed by experimental validation. Signature dysregulated microRNAs in gastric, colorectal and liver cancers were defined based on our previous studies. Targets of signature dysregulated miRNAs were predicted using multiple computer algorithms followed by gene enrichment analysis to identify biological processes perturbed by dysregulated microRNAs. Effects of microRNAs on endocytosis were measured by epidermal growth factor (EGF) internalization assay. Our analysis revealed that, aside from well-established cancer-related signaling pathways, several novel pathways, including axon guidance, neurotrophin/nerve growth factor signaling, and endocytosis, were found to be involved in the pathogenesis of gastrointestinal cancers. The regulation of EGF receptor (EGFR) endocytosis by two predicted miRNAs, namely miR-17 and miR-145, was confirmed experimentally. Functionally, miR-145, which blocked EGFR endocytosis, prolonged EGFR membrane signaling and altered responsiveness of colon cancer cells to EGFR-targeting drugs. In conclusion, our analysis depicts a comprehensive picture of cellular signaling dysregulation, including endocytosis, by microRNAs in gastrointestinal cancers.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuchen Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Sunny H Wong
- Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Priscilla T Y Law
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Shan Zhao
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China.
| | - William K K Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China. .,Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences and Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
108
|
Taipaleenmäki H, Farina NH, van Wijnen AJ, Stein JL, Hesse E, Stein GS, Lian JB. Antagonizing miR-218-5p attenuates Wnt signaling and reduces metastatic bone disease of triple negative breast cancer cells. Oncotarget 2018; 7:79032-79046. [PMID: 27738322 PMCID: PMC5346696 DOI: 10.18632/oncotarget.12593] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 09/19/2016] [Indexed: 01/02/2023] Open
Abstract
Wnt signaling is implicated in bone formation and activated in breast cancer cells promoting primary and metastatic tumor growth. A compelling question is whether osteogenic miRNAs that increase Wnt activity for bone formation are aberrantly expressed in breast tumor cells to support metastatic bone disease. Here we report that miR-218-5p is highly expressed in bone metastases from breast cancer patients, but is not detected in normal mammary epithelial cells. Furthermore, inhibition of miR-218-5p impaired the growth of bone metastatic MDA-MB-231 cells in the bone microenvironment in vivo. These findings indicate a positive role for miR-218-5p in bone metastasis. Bioinformatic and biochemical analyses revealed a positive correlation between aberrant miR-218-5p expression and activation of Wnt signaling in breast cancer cells. Mechanistically, miR-218-5p targets the Wnt inhibitors Sclerostin (SOST) and sFRP-2, which highly enhances Wnt signaling. In contrast, delivery of antimiR-218-5p decreased Wnt activity and the expression of metastasis-related genes, including bone sialoprotein (BSP/IBSP), osteopontin (OPN/SPP1) and CXCR-4, implicating a Wnt/miR-218-5p regulatory network in bone metastatic breast cancer. Furthermore, miR-218-5p also mediates the Wnt-dependent up-regulation of PTHrP, a key cytokine promoting cancer-induced osteolysis. Antagonizing miR-218-5p reduced the expression of PTHrP and Rankl, inhibited osteoclast differentiation in vitro and in vivo, and prevented the development of osteolytic lesions in a preclinical metastasis model. We conclude that pathological elevation of miR-218-5p in breast cancer cells activates Wnt signaling to enhance metastatic properties of breast cancer cells and cancer-induced osteolytic disease, suggesting that miR-218-5p could be an attractive therapeutic target for preventing disease progression.
Collapse
Affiliation(s)
- Hanna Taipaleenmäki
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA, USA.,Heisenberg-Group for Molecular Skeletal Biology, Department of Trauma, Hand & Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicholas H Farina
- Department of Biochemistry & Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| | - Andre J van Wijnen
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Janet L Stein
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Biochemistry & Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| | - Eric Hesse
- Heisenberg-Group for Molecular Skeletal Biology, Department of Trauma, Hand & Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gary S Stein
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Biochemistry & Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| | - Jane B Lian
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Biochemistry & Cancer Center, University of Vermont College of Medicine, Burlington, VT, USA
| |
Collapse
|
109
|
Jiang Z, Song Q, Zeng R, Li J, Li J, Lin X, Chen X, Zhang J, Zheng Y. MicroRNA-218 inhibits EMT, migration and invasion by targeting SFMBT1 and DCUN1D1 in cervical cancer. Oncotarget 2018; 7:45622-45636. [PMID: 27285984 PMCID: PMC5216747 DOI: 10.18632/oncotarget.9850] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 05/20/2016] [Indexed: 01/08/2023] Open
Abstract
Repeated infection with high-risk HPV is a major cause for the development and metastasis of human cervical cancer, even though the mechanism of the metastasis is still not completely understood. Here, we reported that miR-218 (microRNA-218) was downregulated in cervical cancer tissues, especially in metastatic cancer tissues. We found that miR-218 expression was associated with clinicopathological characteristics of patients with cervical cancer. MiR-218 overexpression inhibited Epithelial-Mesenchymal Transition (EMT), migration and invasiveness of cervical cancer cells in vitro. Moreover, miR-218 repressed the expression of SFMFBT1 (Scm-like with four MBT domains 1) and DCUN1D1 (defective in cullin neddylation 1, domain containing 1) by direct binding to the 3′UTRs of the mRNAs. The overexpression of SFMBT1 induced EMT and increased the migration and invasiveness of cervical cancer cells, while the overexpression of DCUN1D1 increased the migration and invasiveness of these cells, but did not induce EMT. An inverse correlation was observed between the expression of miR-218 and DCUN1D1 protein in cervical cancer tissues. Importantly, HPV16 E6 downregulated the expression of miR-218 in cervical cancer, while miR-218 rescued the promotion effect of HPV16 E6 on the expression of SFMBT1 and DCUN1D1. Taken together, our results revealed that HPV16 E6 promoted EMT and invasion in cervical cancer via the repression of miR-218, while miR-218 inhibited EMT and invasion in cervical cancer by targeting SFMBT1 and DCUN1D1.
Collapse
Affiliation(s)
- Zhaojing Jiang
- Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qiancheng Song
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Rong Zeng
- Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Li
- Department of Oncology, Xuzhou Cancer Hospital, Xuzhou, China
| | - Jingyu Li
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaochun Lin
- Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xing Chen
- Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiren Zhang
- Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yanfang Zheng
- Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
110
|
MACC1 is post-transcriptionally regulated by miR-218 in colorectal cancer. Oncotarget 2018; 7:53443-53458. [PMID: 27462788 PMCID: PMC5288198 DOI: 10.18632/oncotarget.10803] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/13/2016] [Indexed: 02/07/2023] Open
Abstract
Metastasis is a multistep molecular network process, which is lethal for more than 90% of the cancer patients. Understanding the regulatory functions of metastasis-inducing molecules is in high demand for improved therapeutic cancer approaches. Thus, we studied the post-transcriptional regulation of the crucial carcinogenic and metastasis-mediating molecule metastasis associated in colon cancer 1 (MACC1). In silico analysis revealed MACC1 as a potential target of miR-218, a tumor suppressor miRNA. Expression of these two molecules inversely correlated in colorectal cancer (CRC) cell lines. In a cohort of CRC patient tissues (n = 59), miR-218 is significantly downregulated and MACC1 is upregulated compared with normal mucosa. Luciferase reporter assays with a construct of the MACC1-3′-UTR harboring either the wild type or the mutated miR-218 seed sequence confirmed the specificity of the targeting. miR-218 inhibited significantly MACC1 protein expression, and consistently, MACC1-mediated migration, invasion and colony formation in CRC cells. Anti-miR-218 enhanced the MACC1-mediated migration, invasion and colony formation. Similar findings were observed in the gastric cancer cell line MKN-45. Further, we performed methylation-specific PCR of the SLIT2 and SLIT3 promoter, where miR-218 is encoded in intronic regions. The SLIT2 and SLIT3 promoters are hypermethylated in CRC cell lines. miR-218 and SLIT2 expressions correlated positively. Methyltransferase inhibitor 5-Azacytidine induced miR-218 expression and inhibited the expression of its target MACC1. We also determined that MACC1 has alternative polyadenylation (APA) sites, which results in different lengths of 3′-UTR variants in a CRC cell line. Taken together, miR-218 is post-transcriptionally inhibiting the MACC1 expression and its metastasis-inducing abilities.
Collapse
|
111
|
Zhang J, Chen D, Liang S, Wang J, Liu C, Nie C, Shan Z, Wang L, Fan Q, Wang F. miR-106b promotes cell invasion and metastasis via PTEN mediated EMT in ESCC. Oncol Lett 2018; 15:4619-4626. [PMID: 29552108 DOI: 10.3892/ol.2018.7861] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 11/22/2017] [Indexed: 02/06/2023] Open
Abstract
MicroRNA (miR)-106b serves an essential function in a variety of human cancer types, particularly in the process of invasion and metastasis. However, the function and mechanism of miR-106b in the invasion and metastasis of esophageal squamous cell carcinoma (ESCC) has remained elusive. In the present study, it was demonstrated that miR-106b was upregulated in ESCC tissues and cell lines. Furthermore, miR-106b expression in ESCC tissues was positively associated with lymphatic metastasis. Inhibition of miR-106b in EC-1 and EC9706 cells decreased not only the invasion and metastasis ability but also the proliferation ability of EC-1 and EC9706 cells. In addition, miR-106b had the ability to induce epithelial-to-mesenchymal transition (EMT) in EC-1 and EC9706 cells. In terms of the underlying mechanism, it was revealed that miR-106b promoted the invasion, metastasis and proliferation ability of EC-1 and EC9706 cells by directly targeting phosphatase and tension homolog (PTEN). Furthermore, miR-106b induced EMT in EC-1 and EC9706 cells by suppressing the expression of PTEN. In summary, the present study revealed that miR-106b contributed to invasion and metastasis in ESCC by regulating PTEN mediated EMT. Downregulation of miR-106b may be a novel strategy for preventing tumor invasion and metastasis.
Collapse
Affiliation(s)
- Jianxiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Danjie Chen
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shuying Liang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Can Liu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Caiping Nie
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhengzheng Shan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Liuxing Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qinxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
112
|
Ma G, Jing C, Li L, Huang F, Ding F, Wang B, Lin D, Luo A, Liu Z. MicroRNA-92b represses invasion-metastasis cascade of esophageal squamous cell carcinoma. Oncotarget 2018; 7:20209-22. [PMID: 26934001 PMCID: PMC4991448 DOI: 10.18632/oncotarget.7747] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/16/2016] [Indexed: 12/17/2022] Open
Abstract
Invasion and metastasis are major contributors to cancer-caused death in patients suffered from esophageal squamous cell carcinoma (ESCC). To explore the microRNAs involved in regulating invasion-metastasis cascade of ESCC, we established two pairs of sublines (30-U/D and 180-U/D) with distinct motility capacity from two ESCC cell lines (KYSE30 and KYSE180). Screening of the differentially expressed microRNAs identified that microRNA-92b-3p (miR-92b) could dramatically inhibit invasion and metastasis of ESCC cells in vitro and in vivo. Subsequent studies showed that miR-92b exerted its inhibitory function through suppressing the expression of integrin αV (ITGAV), which further reduced phosphrylated FAK and impaired Rac1 activation. Moreover, higher expression of miR-92b in ESCC tissues correlated inversely with lymph node metastasis and indicated better prognosis. Together, these results for the first time describe how miR-92b suppresses the motility of ESCC cells and provide a promise for diagnosis or therapy of ESCC invasion and metastasis.
Collapse
Affiliation(s)
- Gang Ma
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Chao Jing
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Lin Li
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Furong Huang
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Fang Ding
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Baona Wang
- Department of Anesthesiology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Dongmei Lin
- Department of Pathology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Aiping Luo
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Zhihua Liu
- The State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| |
Collapse
|
113
|
MicroRNA-26b inhibits tumor metastasis by targeting the KPNA2/c-jun pathway in human gastric cancer. Oncotarget 2018; 7:39511-39526. [PMID: 27078844 PMCID: PMC5129949 DOI: 10.18632/oncotarget.8629] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 03/12/2016] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNA) play an important role in carcinogenesis. Previously, we identified miR-26b as a significantly downregulated miRNA in gastric cancer (GC) tissues (n = 106) based on differential quantitative RT-PCR (RT-qPCR) miRNA expression profiles. In the current study, we aimed to clarify the potential role of miR-26b and related target genes in GC progression. Downregulation of miR-26b was associated with advanced tumor-node-metastasis stage (TNM stage) and poor 5-year survival rate. Forced expression of miR-26b led to inhibition of GC cell migration and invasion in vitro and lung metastasis formation in vivo. Conversely, depletion of miR-26b had stimulatory effects. Additionally, miR-26b affected GC cell behavior through negative regulation of the metastasis promoter, karyopherin alpha 2 (KPNA2). Ectopic expression of miR-26b induced a reduction in KPNA2 protein levels, confirmed by luciferase assay data showing that miR-26b directly binds to the 3' untranslated regions (UTR) of KPNA2 mRNA. Furthermore, miR-26b and KPNA2 mRNA/protein expression patterns were inversely correlated in GC tissues. Cag A of Helicobacter pylori (Hp) enhanced miR-26b levels through regulation of the KPNA2/c-jun pathway. Taken together, our data indicate that miR-26b plays an anti-metastatic role and is downregulated in GC tissues via the KPNA2/c-jun pathway. Based on the study findings, we propose that miR-26b overexpression or KPNA2/c-jun suppression may have therapeutic potential in inhibiting GC metastasis.
Collapse
|
114
|
Zhu K, Ding H, Wang W, Liao Z, Fu Z, Hong Y, Zhou Y, Zhang CY, Chen X. Tumor-suppressive miR-218-5p inhibits cancer cell proliferation and migration via EGFR in non-small cell lung cancer. Oncotarget 2018; 7:28075-85. [PMID: 27057632 PMCID: PMC5053710 DOI: 10.18632/oncotarget.8576] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/28/2016] [Indexed: 11/25/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related death worldwide, and non-small cell lung cancer (NSCLC) accounts for approximately 80% of lung cancer cases. Recently, microRNAs (miRNAs) have been consistently demonstrated to be involved in NSCLC and to act as either tumor oncogenes or tumor suppressors. In this study, we identified a specific binding site for miR-218-5p in the 3'-untranslated region of the epidermal growth factor receptor (EGFR). We further experimentally validated miR-218-5p as a direct regulator of EGFR. We also identified an inverse correlation between miR-218-5p and EGFR protein levels in NSCLC tissue samples. Moreover, we demonstrated that miR-218-5p plays a critical role in suppressing the proliferation and migration of lung cancer cells probably by binding to EGFR. Finally, we examined the function of miR-218-5p in vivo and revealed that miR-218-5p exerts an anti-tumor effect by negatively regulating EGFR in a xenograft mouse model. Taken together, the results of this study highlight an important role for miR-218-5p in the regulation of EGFR in NSCLC and may open new avenues for future lung cancer therapies.
Collapse
Affiliation(s)
- Kegan Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, NJU Advanced Institute for Life Sciences (NAILS), Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, China
| | - Hanying Ding
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, NJU Advanced Institute for Life Sciences (NAILS), Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, China
| | - Wengong Wang
- Department of Thoracic and Cardiovascular Surgery, Department of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Zhicong Liao
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, NJU Advanced Institute for Life Sciences (NAILS), Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, China
| | - Zheng Fu
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, NJU Advanced Institute for Life Sciences (NAILS), Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, China
| | - Yeting Hong
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, NJU Advanced Institute for Life Sciences (NAILS), Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, China
| | - Yong Zhou
- Department of Thoracic and Cardiovascular Surgery, Department of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Chen-Yu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, NJU Advanced Institute for Life Sciences (NAILS), Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, China
| | - Xi Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Collaborative Innovation Center of Chemistry for Life Sciences, NJU Advanced Institute for Life Sciences (NAILS), Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, China
| |
Collapse
|
115
|
Wu C, Zheng X, Li X, Fesler A, Hu W, Chen L, Xu B, Wang Q, Tong A, Burke S, Ju J, Jiang J. Reduction of gastric cancer proliferation and invasion by miR-15a mediated suppression of Bmi-1 translation. Oncotarget 2018; 7:14522-36. [PMID: 26894855 PMCID: PMC4924733 DOI: 10.18632/oncotarget.7392] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 01/12/2016] [Indexed: 12/14/2022] Open
Abstract
B-cell specific moloney leukemia virus insertion site 1 (Bmi-1) gene plays important roles in gastric cancer, but the epigenetic regulatory mechanism by microRNA (miRNA) and the functional significance of Bmi-1 inhibition in gastric cancer remains elusive. In this study, we systematically investigated the functional roles of miRNA mediated Bmi-1 suppression in gastric cancer. Our results show that the expression of miR-15a is significantly reduced in gastric cancer and the protein expression levels of Bmi-1 are inversely correlated with miR-15a (P = 0.034) in gastric cancer patient samples. Functional studies revealed that ectopic expression of miR-15a decreased Bmi-1 in gastric cancer cell lines with reduced proliferation and tumor invasion. High levels of Bmi-1 in gastric cancer patients are significantly associated with better overall survival (P = 0.024) based on the Kaplan-Meier survival analysis.
Collapse
Affiliation(s)
- Changping Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China
| | - Xiao Zheng
- Department of Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.,Translational Research Laboratory, Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Xiaodong Li
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.,Translational Research Laboratory, Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Andrew Fesler
- Translational Research Laboratory, Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Wenwei Hu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China
| | - Lujun Chen
- Department of Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China
| | - Bin Xu
- Department of Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China
| | - Qi Wang
- Department of Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China
| | | | - Stephanie Burke
- Translational Research Laboratory, Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Jingfang Ju
- Translational Research Laboratory, Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Jingting Jiang
- Department of Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China
| |
Collapse
|
116
|
De A, Powers B, De A, Zhou J, Sharma S, Van Veldhuizen P, Bansal A, Sharma R, Sharma M. Emblica officinalis extract downregulates pro-angiogenic molecules via upregulation of cellular and exosomal miR-375 in human ovarian cancer cells. Oncotarget 2017; 7:31484-500. [PMID: 27129171 PMCID: PMC5058772 DOI: 10.18632/oncotarget.8966] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/31/2016] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer (OC) is highly resistant to current treatment strategies based on a combination of surgery, chemotherapy and radiation therapy. We have recently demonstrated the anti-neoplastic effect of Amla extract (Emblica officinalis, AE) on OC cells in vitro and in vivo. We hypothesized that AE attenuates growth of OC through microRNA (miR)-regulated mechanism(s). The inhibitory effect of AE on proliferation, migration and invasiveness (P≤0.001) of SKOV3 cells and >90% attenuation of tumor growth in a xenograft mouse model suggested multiple targets. RT-qPCR analysis of microRNAs associated with OC showed a >2,000-fold increase in the expression of miR-375 in AE-treated SKOV3 cells that was blocked by an exogenous miR-375 inhibitor (P≤0.001). AE also decreased the gene and protein expression of IGF1R, a target of miR-375 (P≤0.001), and SNAIL1 (P≤0.002), an EMT-associated transcription factor that represses E-cadherin expression (P≤0.003). AE increased E-cadherin expression (P≤0.001). Treatment of SKOV3 cells with AE resulted in increased miR-375 in exosomes in the medium (P≤0.01). Finally, AE significantly decreased the expression of IGF1R and SNAIL1 proteins during attenuation of SKOV3-derived xenograft tumor. Together, these results show that AE modulates cancer cells and the tumor microenvironment via activation of miR-375 and by targeting IGF1R and SNAIL1 in OC cells.
Collapse
Affiliation(s)
- Alok De
- Kansas City VA Medical Center and Midwest Biomedical Research Foundation, Kansas City, MO 64128, United States
| | - Benjamin Powers
- Kansas City VA Medical Center and Midwest Biomedical Research Foundation, Kansas City, MO 64128, United States
| | - Archana De
- Kansas City VA Medical Center and Midwest Biomedical Research Foundation, Kansas City, MO 64128, United States
| | - Jianping Zhou
- Kansas City VA Medical Center and Midwest Biomedical Research Foundation, Kansas City, MO 64128, United States
| | - Siddarth Sharma
- Kansas City VA Medical Center and Midwest Biomedical Research Foundation, Kansas City, MO 64128, United States
| | - Peter Van Veldhuizen
- Kansas City VA Medical Center and Midwest Biomedical Research Foundation, Kansas City, MO 64128, United States
| | - Ajay Bansal
- Kansas City VA Medical Center and Midwest Biomedical Research Foundation, Kansas City, MO 64128, United States
| | - Ramratan Sharma
- Kansas City VA Medical Center and Midwest Biomedical Research Foundation, Kansas City, MO 64128, United States
| | - Mukut Sharma
- Kansas City VA Medical Center and Midwest Biomedical Research Foundation, Kansas City, MO 64128, United States
| |
Collapse
|
117
|
Lou W, Liu J, Gao Y, Zhong G, Chen D, Shen J, Bao C, Xu L, Pan J, Cheng J, Ding B, Fan W. MicroRNAs in cancer metastasis and angiogenesis. Oncotarget 2017; 8:115787-115802. [PMID: 29383201 PMCID: PMC5777813 DOI: 10.18632/oncotarget.23115] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/17/2017] [Indexed: 12/15/2022] Open
Abstract
Cancer metastasis is a malignant process by which tumor cells migrate from their primary site of origin to other organs. It is the main cause of poor prognosis in cancer patients. Angiogenesis is the process of generating new blood capillaries from pre-existing vasculature. It plays a vital role in primary tumor growth and distant metastasis. MicroRNAs are small non-coding RNAs involved in regulating normal physiological processes as well as cancer pathogenesis. They suppress gene expression by specifically binding to the 3′-untranslated region (3′-UTR) of their target genes. They can thus act as oncogenes or tumor suppressors depending on the function of their target genes. MicroRNAs have shown great promise for use in anti-metastatic cancer therapy. In this article, we review the roles of various miRNAs in cancer angiogenesis and metastasis and highlight their potential for use in future therapies against metastatic cancer.
Collapse
Affiliation(s)
- Weiyang Lou
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Jingxing Liu
- Department of Intensive Care Unit, Changxing People's Hospital of Zhejiang, Zhejiang Province, Huzhou 313100, China
| | - Yanjia Gao
- Department of Anesthesiology, International Hospital of Zhejiang University, Shulan (Hangzhou) Hospital, Zhejiang Province, Hangzhou 310003, China
| | - Guansheng Zhong
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Danni Chen
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Jiaying Shen
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Chang Bao
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Liang Xu
- Clinical Research Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang Province, Hangzhou 310003, China
| | - Jie Pan
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Junchi Cheng
- Department of Chemotherapy, Zhejiang Cancer Hospital, Zhejiang Province, Hangzhou 310003, China
| | - Bisha Ding
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China
| | - Weimin Fan
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310003, China.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
118
|
Li YJ, Zhang W, Xia H, Zhang BS, Chen P, Zhao YL, Li J. miR-218 suppresses epithelial-to-mesenchymal transition by targeting Robo1 and Ecop in lung adenocarcinoma cells. Future Oncol 2017; 13:2571-2582. [PMID: 28936884 DOI: 10.2217/fon-2017-0398] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Although, miR-218 has been implicated in epithelial-to-mesenchymal transition process, the detailed mechanisms of miR-218 involvement in epithelial-to-mesenchymal transition in human lung adenocarcinoma cell are still unclear. MATERIALS & METHODS miR-218 function assays and its target gene analysis were performed. RESULTS miR-218 suppresses human lung adenocarcinoma cell migration and invasion and inhibits its target gene, Ecop and Robo1 expression, which subsequently suppresses NF-κB activity and its downstream targets. CONCLUSION miR-218 inhibits human lung adenocarcinoma cell migration and invasion via the suppression of Ecop and Robo1 expression, thus suggesting that miR-218 could serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Ying-Jie Li
- Department of Cardio-thoracic Surgery, First Affiliated Hospital of Chinese PLA General Hospital, Beijing, 100048, China
| | - Wen Zhang
- Department of Cardio-thoracic Surgery, First Affiliated Hospital of Chinese PLA General Hospital, Beijing, 100048, China
| | - Hui Xia
- Department of Cardio-thoracic Surgery, First Affiliated Hospital of Chinese PLA General Hospital, Beijing, 100048, China
| | - Bao-Shi Zhang
- Department of Cardio-thoracic Surgery, First Affiliated Hospital of Chinese PLA General Hospital, Beijing, 100048, China
| | - Ping Chen
- Department of Cardio-thoracic Surgery, First Affiliated Hospital of Chinese PLA General Hospital, Beijing, 100048, China
| | - Yun-Long Zhao
- Department of Cardio-thoracic Surgery, First Affiliated Hospital of Chinese PLA General Hospital, Beijing, 100048, China
| | - Jie Li
- Thoracic Surgeon, Department of Thoracic Surgery, The Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
119
|
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: 111] [Impact Index Per Article: 13.9] [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
|
120
|
Chuanyin L, Xiaona W, Zhiling Y, Yu Z, Shuyuan L, Jie Y, Chao H, Li S, Hongying Y, Yufeng Y. The association between polymorphisms in microRNA genes and cervical cancer in a Chinese Han population. Oncotarget 2017; 8:87914-87927. [PMID: 29152130 PMCID: PMC5675682 DOI: 10.18632/oncotarget.21235] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/27/2017] [Indexed: 01/09/2023] Open
Abstract
Several studies have confirmed the crucial roles of microRNAs (miRNAs) in cancer occurrence. In addition, single nucleotide polymorphisms (SNPs) in miRNA genes have been associated with various cancers. The aim of the present study was to investigate the association of SNPs in miRNA genes with cervical intraepithelial neoplasia (CIN) and cervical cancer in a Chinese Han population. We searched SNPs in nineteen miRNAs by sequencing healthy individuals (n=50). Then, a total of 400 patients with CIN, 609 patients with cervical cancer and 583 healthy individuals were recruited to genotype the SNPs using a Taqman assay. The results showed that only five of the nineteen miRNAs had SNPs (rs11134527 in pri-miR-218-2; rs74693964 in pri-miR-145; rs6062251 in pri-miR-133a2; rs531564 in pri-miR-124-1; and rs1834306 in pri-miR-100) in this Chinese Han population. The frequency of the rs11134527A allele was significantly higher in the control group than in CIN and cervical cancer groups (P=0.011 and 0.035, respectively). The frequency of the rs531564G allele was higher in the CIN and control groups than in the cervical cancer group (P=0.019 and 0.017, respectively). These results indicated that rs11134527 in pri-miR-218-2 and rs531564 in pri-miR-124-1 could be associated with CIN and cervical cancer in the Chinese Han population.
Collapse
Affiliation(s)
- Li Chuanyin
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Wang Xiaona
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Yan Zhiling
- Department of Gynaecologic Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China
| | - Zhang Yu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Liu Shuyuan
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Yang Jie
- Wenzhou Medical University, Wenzhou 325035, China
| | - Hong Chao
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Shi Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Yang Hongying
- Department of Gynaecologic Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Kunming 650118, China
| | - Yao Yufeng
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| |
Collapse
|
121
|
Peng Z, Pan L, Niu Z, Li W, Dang X, Wan L, Zhang R, Yang S. Identification of microRNAs as potential biomarkers for lung adenocarcinoma using integrating genomics analysis. Oncotarget 2017; 8:64143-64156. [PMID: 28969058 PMCID: PMC5609990 DOI: 10.18632/oncotarget.19358] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/05/2017] [Indexed: 02/05/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common histological subtype of non-small cell lung cancer, but novel biomarkers for early diagnosis are lacking. Extensive effort has been exerted to identify miRNA biomarkers in LUAD. Unfortunately, high inter-lab variability and small sample sizes have produced inconsistent conclusions in this field. To resolve the above-mentioned limitations, we performed a comprehensive analysis based on LUAD miRNome profiling studies using the robust rank aggregation (RRA) method. Moreover, miRNA-gene interaction network, pathway enrichment analysis and Kaplan-Meier survival curves were used to investigate the clinical values and biological functions of the identified miRNAs. A total of six common differentially expressed miRNAs (DEMs) were identified in LUAD. An independent cohort further confirmed that four miRNAs (miR-21-5p, miR-210-3p, miR-182-5p and miR-183-5p) were up-regulated and two miRNAs (miR-126-3p and miR-218-5p) were down-regulated in LUAD tissues. Pathway enrichment analysis also suggested that the above-listed six DEMs may affect LUAD progression via the estrogen signaling pathway. Survival analysis based on the TCGA dataset revealed the potential prognostic values of six DEMs in patients with LUAD (P-value<0.01). In conclusion, we identified a panel of six miRNAs from LUAD using miRNome profiling studies. Our results provide evidence for the use of these six DEMs as novel diagnostic and prognostic biomarkers for LUAD patients.
Collapse
Affiliation(s)
- Zhuo Peng
- Department of Emergency Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Longfei Pan
- Department of Emergency Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Zequn Niu
- Department of Emergency Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Wei Li
- Department of Respiratory Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Xiaoyan Dang
- Department of Emergency Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Lin Wan
- Department of Emergency Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Rui Zhang
- Department of Emergency Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Shuanying Yang
- Department of Respiratory Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| |
Collapse
|
122
|
Ahmadinejad F, Mowla SJ, Honardoost MA, Arjenaki MG, Moazeni-Bistgani M, Kheiri S, Teimori H. Lower expression of miR-218 in human breast cancer is associated with lymph node metastases, higher grades, and poorer prognosis. Tumour Biol 2017; 39:1010428317698362. [DOI: 10.1177/1010428317698362] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is considered as the most prevalent malignancy in women worldwide. Despite emergence of several prognosticators for better management of patients, there are still limitations for their clinical application due to the complexity of breast tumors, and therefore, new biomarkers for better prognosis of clinical outcomes would be of the great essence. MicroRNAs are highly conserved small non-coding regulatory RNAs involved in post-transcriptional regulating of gene expression during different cellular mechanisms. Accumulating studies suggest that miR-218 plays a multifunctional role in various cancer types and different stages. Here, to address prognostic significance of miR-218 in breast cancer, we investigate the expression profile of miR-218 and B-cell-specific Moloney murine leukemia virus integration site 1 ( BMI1) gene, as one of the putative targets of miR-218, in 33 paired breast tumors and their adjacent normal tissues with respect to the clinicopathological features of patients using quantitative real-time polymerase chain reaction. The correlation of both miR-218 and BMI1 gene expression with overall survival of breast cancer patients was also examined recruiting OncoLNC data portal. Finally, to better understand biological function of miR-218 in breast cancer, we performed in silico Gene Ontology and signaling pathway enrichment analysis on miR-218 targetome. According to our data, significant elevation of the expression of miR-218 and downregulation of BMI1 were observed in clinical breast cancer specimens compared with normal tissues ( p < 0.0001). The lower expression of miR-218 was associated with lymph node metastases, higher grades, and poorer prognosis (logrank p = 0.00988), whereas no significant difference in overall survival was observed between patients with higher and lower expression of BMI1 (logrank p = 0.254). These findings suggest that miR-218 expression profiling might be clinically applicable as a prognostic biomarker in breast cancer. In addition, our in silico enrichment analyses revealed that the association of miR-218 expression with breast cancer prognosis might be through its involvement in endocytosis and gap junction biological pathways.
Collapse
Affiliation(s)
- Fereshteh Ahmadinejad
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad-Amin Honardoost
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of Science, University of Isfahan, Isfahan, Iran
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, A*STAR (Agency for Science, Technology and Research), Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mostafa Gholami Arjenaki
- Clinical Biochemistry Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Soleyman Kheiri
- Department of Epidemiology and Biostatistics, School of Health, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Teimori
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
123
|
Yang Y, Ding L, Hu Q, Xia J, Sun J, Wang X, Xiong H, Gurbani D, Li L, Liu Y, Liu A. MicroRNA-218 functions as a tumor suppressor in lung cancer by targeting IL-6/STAT3 and negatively correlates with poor prognosis. Mol Cancer 2017; 16:141. [PMID: 28830450 PMCID: PMC5567631 DOI: 10.1186/s12943-017-0710-z] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 08/07/2017] [Indexed: 12/14/2022] Open
Abstract
Background Aberrant expression of microRNAs in different human cancer types has been widely reported. MiR-218 acts as a tumor suppressor in diverse human cancer types impacting regulation of multiple genes in oncogenic pathways. Here, we evaluated the expression and function of miR-218 in human lung cancer and ALDH positive lung cancer cells to understand the potential mechanisms responsible for disease pathology. Also, the association between its host genes and the target genes could be useful towards the better understanding of prognosis in clinical settings. Methods Publicly-available data from The Cancer Genome Atlas (TCGA) was mined to compare the levels of miR-218 and its host gene SLIT2/3 between lung cancer tissues and normal lung tissues. Transfection of miR-218 to investigate its function in lung cancer cells was done and in vivo effects were determined using miR-218 expressing lentiviruses. Aldefluor assay and Flow cytometry was used to quantify and enrich ALDH positive lung cancer cells. Levels of miR-218, IL-6R, JAK3 and phosphorylated STAT3 were compared in ALDH1A1 positive and ALDH1A1 negative cells. Overexpression of miR-218 in ALDH positive cells was carried to test the survival by tumorsphere culture. Finally, utilizing TCGA data we studied the association of target genes of miR-218 with the prognosis of lung cancer. Results We observed that the expression of miR-218 was significantly down-regulated in lung cancer tissues compared to normal lung tissues. Overexpression of miR-218 decreased cell proliferation, invasion, colony formation, and tumor sphere formation in vitro and repressed tumor growth in vivo. We further found that miR-218 negatively regulated IL-6 receptor and JAK3 gene expression by directly targeting the 3′-UTR of their mRNAs. In addition, the levels of both miR-218 host genes and the components of IL-6/STAT3 pathway correlated with prognosis of lung cancer patients. Conclusions MiR-218 acts as a tumor suppressor in lung cancer via IL-6/STAT3 signaling pathway regulation. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0710-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Yang
- Experimental Medicine Center of Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Lili Ding
- Department of Pediatrics of Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, People's Republic of China
| | - Qun Hu
- Department of Pediatrics of Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, People's Republic of China
| | - Jia Xia
- Department of Pediatrics of Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, People's Republic of China
| | - Junjie Sun
- Department of Pediatrics of Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, People's Republic of China
| | - Xudong Wang
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hua Xiong
- Department of Oncology of Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Deepak Gurbani
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lianbo Li
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Yan Liu
- Department of Medicine, Division of Epidemiology, Vanderbilt University, Nashville, TN, 37232, USA.,Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Aiguo Liu
- Department of Pediatrics of Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
124
|
Zhang Y, Guan DH, Bi RX, Xie J, Yang CH, Jiang YH. Prognostic value of microRNAs in gastric cancer: a meta-analysis. Oncotarget 2017; 8:55489-55510. [PMID: 28903436 PMCID: PMC5589675 DOI: 10.18632/oncotarget.18590] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/08/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Previous articles have reported that expression levels of microRNAs (miRNAs) are associated with survival time of patients with gastric cancer (GC). A systematic review and meta-analysis was performed to study the outcome of it. DESIGN Meta-analysis. METHODS English studies estimating expression levels of miRNAs with any of survival curves in GC were identified up till March 19, 2017 through performing online searches in PubMed, EMBASE, Web of Science and Cochrane Database of Systematic Reviews by two authors independently. The pooled hazard ratios (HR) with 95% confidence intervals (CI) were used to estimate the correlation between miRNA expression and overall survival (OS). RESULTS Sixty-nine relevant articles about 26 miRNAs with 6148 patients were ultimately included. GC patients with high expression of miR-20b (HR=2.38, 95%CI=1.16-4.87), 21 (HR=1.77, 95%CI=1.01-3.08), 106b (HR=1.84, 95%CI=1.15-2.94), 196a (HR=2.66, 95%CI=1.94-3.63), 196b (HR=1.67, 95%CI=1.38-2.02), 214 (HR=1.84, 95%CI=1.27-2.67) or low expression of miR-125a (HR=2.06, 95%CI=1.26-3.37), 137 (HR=3.21, 95%CI=1.68-6.13), 141 (HR=2.47, 95%CI=1.34-4.56), 145 (HR=1.62, 95%CI=1.07-2.46), 146a (HR=2.60, 95%CI=1.63-4.13), 206 (HR=2.85, 95%CI=1.73-4.70), 218 (HR=2.61, 95%CI=1.74-3.92), 451 (HR=1.73, 95%CI=1.19-2.52), 486-5p (HR=2.45, 95%CI=1.65-3.65), 506 (HR=2.07, 95%CI=1.33-3.23) have significantly poor OS (P<0.05). CONCLUSIONS In summary, miR-20b, 21, 106b, 125a, 137, 141, 145, 146a, 196a, 196b, 206, 214, 218, 451, 486-5p and 506 demonstrate significantly prognostic value. Among them, miR-20b, 125a, 137, 141, 146a, 196a, 206, 218, 486-5p and 506 are strong biomarkers of prognosis in GC.
Collapse
Affiliation(s)
- Yue Zhang
- 1 First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, People's Republic of China
| | - Dong-Hui Guan
- 2 Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Rong-Xiu Bi
- 2 Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Jin Xie
- 2 Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Chuan-Hua Yang
- 3 Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| | - Yue-Hua Jiang
- 4 Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People's Republic of China
| |
Collapse
|
125
|
Zhuang Z, Hu F, Hu J, Wang C, Hou J, Yu Z, Wang TT, Liu X, Huang H. MicroRNA-218 promotes cisplatin resistance in oral cancer via the PPP2R5A/Wnt signaling pathway. Oncol Rep 2017; 38:2051-2061. [PMID: 28849187 PMCID: PMC5652945 DOI: 10.3892/or.2017.5899] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 04/20/2017] [Indexed: 12/14/2022] Open
Abstract
Accumulating data suggest that microRNAs (miRNAs) play a pivotal role in the regulation of tumor cell sensitivity to chemotherapeutic agents. Although the roles of a few miRNAs have been identified in cisplatin resistance, little is known in regards to the concerted contribution of miRNA‑mediated biological networks. In the present study, we demonstrated that microRNA-218 (miR-218) was significantly upregulated in cisplatin-resistant oral cancer cells. The results of cell viability and apoptosis assay showed that ectopic expression of miR-218 induced cell survival and resistance to cisplatin, whereas suppression of miR-218 caused apoptosis and enhanced sensitivity to cisplatin. Moreover, we identified PPP2R5A as a new direct target of miR-218 by using the dual luciferase reporter assay. Overexpression of miR-218 led to inhibition of PPP2R5A expression, whereas knockdown of miR-218 increased PPP2R5A levels. Introduction of PPP2R5A abrogated miR‑218-mediated cell survival and drug resistance. Furthermore, suppression of miR-218 or PPP2R5A significantly promoted or reduced cisplatin-induced apoptosis, respectively. Finally, PPP2R5A overexpression or β-catenin knockdown inhibited miR-218-mediated Wnt activation and partially restored cell sensitivity. Our data revealed a molecular link between miR-218 and PPP2R5A/Wnt signaling and implicates miR-218 as a potential target for oral cancer therapy.
Collapse
Affiliation(s)
- Zehang Zhuang
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
| | - Fengchun Hu
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
| | - Jing Hu
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
| | - Cheng Wang
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
| | - Jinsong Hou
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Tony T Wang
- Center for Infectious Diseases, SRI International, Harrisonburg, VA 22802, USA
| | - Xiqiang Liu
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
| | - Hongzhang Huang
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
126
|
Ye G, Huang K, Yu J, Zhao L, Zhu X, Yang Q, Li W, Jiang Y, Zhuang B, Liu H, Shen Z, Wang D, Yan L, Zhang L, Zhou H, Hu Y, Deng H, Liu H, Li G, Qi X. MicroRNA-647 Targets SRF-MYH9 Axis to Suppress Invasion and Metastasis of Gastric Cancer. Theranostics 2017; 7:3338-3353. [PMID: 28900514 PMCID: PMC5595136 DOI: 10.7150/thno.20512] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/29/2017] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs (miRNAs) play important roles in regulating tumour development and progression. Here we show that miR-647 is repressed in gastric cancer (GC), and associated with GC metastasis. Moreover, we identify that miR-647 can suppress GC cell migration and invasion in vitro. Mechanistically, we confirm miR-647 directly binds to the 3' untranslated regions of SRF mRNA, and SRF binds to the CArG box located at the MYH9 promoter. CCG-1423, an inhibitor of RhoA/SRF-mediated gene transcription, inhibits the expression of MYH9, especially in SRF downregulated cells. Overexpression of miR-647 inhibits MGC 80-3 cells' metastasis in orthotropic GC models, but increasing SRF expression in these cells reverses this change. Importantly, we found the synergistic inhibition effect of CCG-1423 and agomir-647, an engineered miRNA mimic, on cancer metastasis in orthotropic GC models. Our study demonstrates that miR-647 functions as a tumor metastasis suppressor in GC by targeting SRF/MYH9 axis.
Collapse
Affiliation(s)
- Gengtai Ye
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Kunzhai Huang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Liying Zhao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Xianjun Zhu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Qingbin Yang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Wende Li
- Guangdong Key Laboratory of Laboratory Animal, Guangdong Laboratory Animal Monitoring Institute, Guangzhou 510663, China
| | - Yuming Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Baoxiong Zhuang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Hao Liu
- Leder Human Biology and Translational Medicine, Biology and Biomedical Sciences, Division of Medical Sciences, Harvard Medical School, Boston, MA 02115
| | - Zhiyong Shen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Da Wang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Li Yan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Lei Zhang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Haipeng Zhou
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Haijun Deng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Xiaolong Qi
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| |
Collapse
|
127
|
Pu M, Li C, Qi X, Chen J, Wang Y, Gao L, Miao L, Ren J. MiR-1254 suppresses HO-1 expression through seed region-dependent silencing and non-seed interaction with TFAP2A transcript to attenuate NSCLC growth. PLoS Genet 2017; 13:e1006896. [PMID: 28749936 PMCID: PMC5549757 DOI: 10.1371/journal.pgen.1006896] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/08/2017] [Accepted: 06/26/2017] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs, which direct post-transcriptional gene silencing (PTGS) and function in a vast range of biological events including cancer development. Most miRNAs pair to the target sites through seed region near the 5’ end, leading to mRNA cleavage and/or translation repression. Here, we demonstrated a miRNA-induced dual regulation of heme oxygenase-1 (HO-1) via seed region and non-seed region, consequently inhibited tumor growth of NSCLC. We identified miR-1254 as a negative regulator inhibiting HO-1 translation by directly targeting HO-1 3’UTR via its seed region, and suppressing HO-1 transcription via non-seed region-dependent inhibition of transcriptional factor AP-2 alpha (TFAP2A), a transcriptional activator of HO-1. MiR-1254 induced cell apoptosis and cell cycle arrest in human non-small cell lung carcinoma (NSCLC) cells by inhibiting the expression of HO-1, consequently suppressed NSCLC cell growth. Consistently with the in vitro studies, mouse xenograft studies validated that miR-1254 suppressed NSCLC tumor growth in vivo. Moreover, we found that HO-1 expression was inversely correlated with miR-1254 level in human NSCLC tumor samples and cell lines. Overall, these findings identify the dual inhibition of HO-1 by miR-1254 as a novel functional mechanism of miRNA, which results in a more effective inhibition of oncogenic mRNA, and leads to a tumor suppressive effect. It is generally accepted that miRNAs bind to 3`UTR of target mRNAs and direct post-transcriptional gene silencing (PTGS) via its seed sequence. Here we report a new dual regulatory mechanism of miRNA. We described that miR-1254 repressed HO-1 at post-transcriptional level by directly targeting HO-1 3’UTR via its seed sequence and also inhibited HO-1 transcription by suppressing the transcriptional factor AP-2 alpha (TFAP2A) via its non-seed sequence. MiR-1254 induced cell apoptosis and cell cycle arrest in human non-small cell lung carcinoma (NSCLC) cells by inhibiting the expression of HO-1, consequently suppressed NSCLC cell growth. Moreover, in vivo mouse xenograft studies also supported the inhibitory effect of miR-1254 on NSCLC growth. These findings identify the dual regulation of miR-1254 on HO-1 as a novel functional mechanism of miRNA, which results in a more effective inhibition on the oncogenic mRNA, and leads to a suppressive effect on NSCLC growth, thus significantly advance our understanding of miRNA-directed gene regulation.
Collapse
Affiliation(s)
- Mengfan Pu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chenggang Li
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinming Qi
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jing Chen
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yizheng Wang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lulu Gao
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lingling Miao
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- * E-mail: (LM); (JR)
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- * E-mail: (LM); (JR)
| |
Collapse
|
128
|
陈 平, 赵 云, 李 英. [MiR-218 Inhibits Migration and Invasion of Lung Cancer Cell
by Regulating Robo1 Expression]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 20:452-458. [PMID: 28738960 PMCID: PMC5972947 DOI: 10.3779/j.issn.1009-3419.2017.07.03] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 05/19/2017] [Accepted: 05/20/2017] [Indexed: 11/05/2022]
Abstract
BACKGROUND To explore the function and the potential molecular mechanism of miR-218 in lung cancer cell. METHODS The expression of miR-218 mRNA was determined by real-time PCR in lung cancer tissues, adjacent tissues and lung cancer cells. Transwell assay was used to detect the migration and invasion of A549 cell after transfected with Anti-miR-218 or negative control and HC4006 cell after transfected with miR-218 mimics and miR-218 negative control. Targetscan and MiRanda were used to calculate the potential targets of miR-218 and Luciferase reporter assay was performed to identify that the Robo1 was one target genes of miR-218. Transwell assay was used to detect whether miR-218 regulated the invasion of lung cancer cell transfected with anti-miR-218 or negative control via Robo1. RESULTS The expression of miR-218 in the lung cancer tissues was significantly lower than that in the adjacent tissues (P<0.05). Inhibition of miR-218 improved the migration and invasion of A549 cell. Overexpression of miR-218 suppressed the migration and invasion of HCC4006 cell. The co-transfection of anti-miR-218 or miR-218 mimics and the Robo1 3'UTR increased or reduced the luciferase activity of Robo1 compared with the control group (P<0.05). Inhibition of miR-218 and Robo1 recovered the invaded cells of A549. Overexpression of miR-218 and inhibition of Robo1 reduced the number of the invased cells of HCC4006. These results suggested that miR-218 banded Robo1 directly and inhibited lung cancer cell invasion by targeting Robo1. CONCLUSIONS MiR-218 inhibited the migration and invasion of lung cancer cells through regulating Robo1 expression.
.
Collapse
Affiliation(s)
- 平 陈
- />100048 北京,解放军总医院第一附属医院First Affiliated Hospital of PLA General Hospital, Beijing 100048, China
| | - 云龙 赵
- />100048 北京,解放军总医院第一附属医院First Affiliated Hospital of PLA General Hospital, Beijing 100048, China
| | - 英杰 李
- />100048 北京,解放军总医院第一附属医院First Affiliated Hospital of PLA General Hospital, Beijing 100048, China
| |
Collapse
|
129
|
Zhang Y, Hu H. Long non-coding RNA CCAT1/miR-218/ZFX axis modulates the progression of laryngeal squamous cell cancer. Tumour Biol 2017. [PMID: 28631575 DOI: 10.1177/1010428317699417] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Long non-coding RNAs have been proved to be closely associated with different cancers. This study was designed to elucidate the function and mechanisms of colon cancer-associated transcript-1 in the progression of human laryngeal squamous cell cancer. Expressions of colon cancer-associated transcript-1, microRNA-218, and zinc finger protein, X-linked messenger RNA were measured using quantitative real-time polymerase chain reaction, and the expression level of zinc finger protein, X-linked protein was detected using western blot. Proliferation and invasion of laryngeal squamous cell cancer cell lines were detected by Cell Counting Kit-8 assay and Transwell invasion assay, respectively. Luciferase assay was used to confirm whether microRNA-218 is a target of colon cancer-associated transcript-1 and whether microRNA-218 directly binds to 3'-untranslated region of zinc finger protein, X-linked messenger RNA. Effect of colon cancer-associated transcript-1 on tumor growth was observed through xenograft mice models in vivo. The results showed that expressions of colon cancer-associated transcript-1 and zinc finger protein, X-linked were significantly higher while microRNA-218 expression was significantly lower in the laryngeal squamous cell cancer tissues than those in the adjacent normal tissues. MicroRNA-218 overexpression or zinc finger protein, X-linked silencing significantly suppressed proliferation and invasion of laryngeal squamous cell cancer cells. Moreover, knockdown of colon cancer-associated transcript-1 significantly inhibited proliferation and invasion of laryngeal squamous cell cancer cells, which were reversed by microRNA-218 downregulation or zinc finger protein, X-linked upregulation. Finally, colon cancer-associated transcript-1 silencing inhibited xenograft tumor growth of laryngeal squamous cell cancer in vivo. In conclusion, colon cancer-associated transcript-1 knockdown inhibits proliferation and invasion of laryngeal squamous cell cancer cells through enhancing zinc finger protein, X-linked by sponging microRNA-218, elucidating a novel colon cancer-associated transcript-1-microRNA-218-zinc finger protein, X-linked regulatory axis in laryngeal squamous cell cancer and providing a promising therapeutic target for laryngeal squamous cell cancer patients.
Collapse
Affiliation(s)
- Yaming Zhang
- Department of Otolaryngology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Haili Hu
- Department of Otolaryngology, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
130
|
Wang T, Liu Y, Zhao M. Mutational analysis of driver genes with tumor suppressive and oncogenic roles in gastric cancer. PeerJ 2017; 5:e3585. [PMID: 28729958 PMCID: PMC5516769 DOI: 10.7717/peerj.3585] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/26/2017] [Indexed: 12/18/2022] Open
Abstract
Gastric cancer (GC) is a complex disease with heterogeneous genetic mechanisms. Genomic mutational profiling of gastric cancer not only expands our knowledge about cancer progression at a fundamental genetic level, but also could provide guidance on new treatment decisions, currently based on tumor histology. The fact that precise medicine-based treatment is successful in a subset of tumors indicates the need for better identification of clinically related molecular tumor phenotypes, especially with regard to those driver mutations on tumor suppressor genes (TSGs) and oncogenes (ONGs). We surveyed 313 TSGs and 160 ONGs associated with 48 protein coding and 19 miRNA genes with both TSG and ONG roles. Using public cancer mutational profiles, we confirmed the dual roles of CDKN1A and CDKN1B. In addition to the widely recognized alterations, we identified another 82 frequently mutated genes in public gastric cancer cohort. In summary, these driver mutation profiles of individual GC will form the basis of personalized treatment of gastric cancer, leading to substantial therapeutic improvements.
Collapse
Affiliation(s)
- Tianfang Wang
- School of Science and Engineering, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydore DC, Australia
| | - Yining Liu
- The School of Public Health, Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, China
| | - Min Zhao
- School of Science and Engineering, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydore DC, Australia
| |
Collapse
|
131
|
Li P, Zhang X, Wang L, Du L, Yang Y, Liu T, Li C, Wang C. lncRNA HOTAIR Contributes to 5FU Resistance through Suppressing miR-218 and Activating NF-κB/TS Signaling in Colorectal Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:356-369. [PMID: 28918035 PMCID: PMC5537205 DOI: 10.1016/j.omtn.2017.07.007] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 12/30/2022]
Abstract
One major reason for the failure of advanced colorectal cancer (CRC) treatment is the occurrence of chemoresistance to fluoropyrimidine (FU)-based chemotherapy. Long non-coding RNA HOTAIR has been considered as a pro-oncogene in multiple cancers. However, the precise functional mechanism of HOTAIR in chemoresistance is not well known. In this study, we investigated the biological and clinical role of HOTAIR in 5FU resistance in CRC. Our results showed that HOTAIR negatively regulated miR-218 expression in CRC through an EZH2-targeting miR-218-2 promoter regulatory axis. HOTAIR knockdown dramatically inhibited cell viability and induced G1-phase arrest by promoting miR-218 expression. VOPP1 was shown to be a functional target of miR-218, and the main downstream signaling, NF-κB, was inactivated by HOTAIR through the suppression of miR-218 expression. Additionally, HOTAIR knockdown partially reversed 5FU resistance through promoting miR-218 and inactivating NF-κB signaling. Furthermore, HOTAIR restrained 5FU-induced cytotoxicity on CRC cells through promotion of thymidylate synthase expression. More importantly, high HOTAIR expression was associated with poor response to 5FU treatment. In conclusion, we demonstrated that HOTAIR contributes to 5FU resistance through suppressing miR-218 and activating NF-κB signaling in CRC. Thus, HOTAIR may serve as a promising therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Peilong Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, 250033 Shandong Province, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, 250012 Shandong Province, China
| | - Lili Wang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, 250012 Shandong Province, China
| | - Lutao Du
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, 250012 Shandong Province, China
| | - Yongmei Yang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, 250012 Shandong Province, China
| | - Tong Liu
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, 250033 Shandong Province, China
| | - Chen Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, 250033 Shandong Province, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, 250033 Shandong Province, China.
| |
Collapse
|
132
|
Nie Y, Wu K, Yu J, Liang Q, Cai X, Shang Y, Zhou J, Pan K, Sun L, Fang J, Yuan Y, You W, Fan D. A global burden of gastric cancer: the major impact of China. Expert Rev Gastroenterol Hepatol 2017; 11:651-661. [PMID: 28351219 DOI: 10.1080/17474124.2017.1312342] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gastric cancer (GC) is a highly aggressive cancer and a major cause of cancer-related deaths worldwide. Approximately half of the world's GC cases and deaths occur in china. GC presents challenges in early diagnosis and effective therapy due to a lack of understanding of the underlying molecular biology. The primary goals of this review are to outline current GC research in china and describe future trends in this field. Areas covered: This review mainly focuses on a series of GC-related advances China has achieved. Considerable progress has been made in understanding the role of H. pylori in GC by a series of population-based studies in well-established high-risk areas; A few germline and somatic alterations have been identified by 'omics' studies; Studies on the mechanisms of malignant phenotypes have helped us to form an in-depth understanding of GC and advance drug discovery. Moreover, identification of potential biomarkers and targeted therapies have facilitated the diagnosis and treatment of GC. However, many challenges remain. Expert commentary: To combat GC, sufficient funding is important. More attention should be paid on early diagnosis and the discovery of novel efficient biomarkers and the development of biomarker-based or targeted therapeutics in GC.
Collapse
Affiliation(s)
- Yongzhan Nie
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Kaichun Wu
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Jun Yu
- b Department of Medicine and Therapeutics and Institute of Digestive Disease , Chinese University of Hong Kong , Hong Kong , China
| | - Qiaoyi Liang
- b Department of Medicine and Therapeutics and Institute of Digestive Disease , Chinese University of Hong Kong , Hong Kong , China
| | - Xiqiang Cai
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Yulong Shang
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Jinfeng Zhou
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| | - Kaifeng Pan
- c Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University School of Oncology , Peking University Cancer Hospital & Institute , Beijing , China
| | - Liping Sun
- d Tumor Etiology and Screening, Department of Cancer Institute and General Surgery , The First Affiliated Hospital of China Medical University , Shenyang , China
| | - Jingyuan Fang
- e Renji Hospital , Shanghai Jiao-Tong University School of Medicine , Shanghai , China
| | - Yuan Yuan
- d Tumor Etiology and Screening, Department of Cancer Institute and General Surgery , The First Affiliated Hospital of China Medical University , Shenyang , China
| | - Weicheng You
- c Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University School of Oncology , Peking University Cancer Hospital & Institute , Beijing , China
| | - Daiming Fan
- a State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases , Fourth Military Medical University , Xi'an , China
| |
Collapse
|
133
|
Ge F, Wang C, Wang W, Liu W, Wu B. MicroRNA-31 inhibits tumor invasion and metastasis by targeting RhoA in human gastric cancer. Oncol Rep 2017; 38:1133-1139. [PMID: 28656284 DOI: 10.3892/or.2017.5758] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 05/18/2017] [Indexed: 11/05/2022] Open
Abstract
Previous studies have shown that microRNA-31 (miR-31) functions as a tumor-suppressor in various types of cancer. In the present study we found that miR-31 was significantly downregulated in gastric cancer (GC) as determined by microRNA (miRNA) array screening analysis. Although aberrant expression of miR-31 has been found in different types of cancer, its pathophysiological effect and role in tumorigenesis still remain to be elucidated. In the present study, we detected miR-31 expression in both metastatic GC cell lines and tissues that are potentially highly metastatic by real-time polymerase chain reaction (PCR). Transwell and scratch healing assays were conducted to examine whether the ectopic expression of miR-31 could modify the invasion and migration abilities of GC cells in vitro. We found that miR-31 inhibited GC metastasis in a nude mouse xenograft model of GC. Luciferase reporter assays demonstrated that miR-31 could directly bind to the 3' untranslated region of RhoA and downregulate the expression of RhoA. Significant downregulation of miR-31 in 78 GC tissues and four GC cell lines was examined by real-time reverse transcription-PCR. Moreover, the decreased expression of miR-31 was demonstrated to be associated with lymph node metastasis, poor pT and pN stage, and invasion ability into lymphatic vessels as determined by the Mann-Whitney U test. We also found that miR-31 could inhibit cell invasion and migration abilities in vitro using the Transwell and scratch healing assays in BGC-823, SGC-7901, MGC-803 as well as AGS cells. Experiments in a nude mouse model revealed that miR-31 suppressed tumorigenicity in vivo. The luciferase activity assay and western blotting indicated that RhoA was the potential target of miR-31 in GC cells. Collectively, our results provide important evidence that the downregulation of miR-31 inhibited the invasion and migration abilities of GC cells through direct targeting of the tumor metastasis‑associated gene, RhoA. These findings suggest that miR-31 may be a promising therapeutic candidate in the treatment of GC metastasis.
Collapse
Affiliation(s)
- Fulin Ge
- Department of Gastroenterology, Division of Southern Building, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Changzheng Wang
- Department of Gastroenterology, Division of Southern Building, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Weihua Wang
- Department of Gastroenterology, Division of Southern Building, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Wenhui Liu
- Department of Gastroenterology, Division of Southern Building, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Benyan Wu
- Department of Gastroenterology, Division of Southern Building, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
134
|
Deng M, Zeng C, Lu X, He X, Zhang R, Qiu Q, Zheng G, Jia X, Liu H, He Z. miR-218 suppresses gastric cancer cell cycle progression through the CDK6/Cyclin D1/E2F1 axis in a feedback loop. Cancer Lett 2017. [PMID: 28634044 DOI: 10.1016/j.canlet.2017.06.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Studies in several cancers have suggested that miR-218 has anti-tumor activities, but its function is yet to be elucidated. In this study, we investigated the regulation and function of miR-218 (miR-218-5p) in the cell cycle progression of gastric cancer (GC). We found that miR-218 could suppress proliferation of gastric cancer cells, induce cell cycle arrest at the G1 phase and inhibit tumor growth and metastasis in vivo. We also demonstrated that miR-218 specifically targeted the 3'-UTR regions of CDK6 and cyclin D1 and inhibited the expression of these molecules, which in turn repressed the pRb/E2F1 signaling pathway. Overexpression of CDK6 and Cyclin D1 reversed miR-218-mediated inhibition of pRB/E2F1 signaling and attenuated the miR-218-induced cell cycle arrest. More importantly, miR-218 expression was significantly reduced and inversely correlated with the levels of CDK6 and Cyclin D1 in gastric cancer tissues. Decreased miR-218 expression was also correlated with advanced clinical stage, lymph node metastasis, and poor prognosis in gastric cancer patients. Furthermore, we showed that miR-218 expression was directly activated by E2F1 through the transactivation of miR-218 host genes, SLIT2 and SLIT3, revealing a negative feedback regulation of miR-218 expression. Taken together, our results describe a regulatory loop miR-218-CDK6/CyclinD1-E2F1 whose disruption may contribute to cell cycle progression in gastric cancer and indicate the potential application of miR-218 in cancer therapy.
Collapse
Affiliation(s)
- Min Deng
- Cancer Hospital and Cancer Research Institute, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Chao Zeng
- Department of Pathology, Guangdong Medical University, Dongguan, Guangdong Province, China
| | - Xihong Lu
- Guangzhou Eighth People's Hospital, Guangzhou, Guangdong Province, China
| | - Xiusheng He
- Cancer Research Institute, University of South China, Hengyang, Hunan Province, China
| | - Ruixin Zhang
- Cancer Hospital and Cancer Research Institute, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Qinwei Qiu
- Cancer Hospital and Cancer Research Institute, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Guopei Zheng
- Cancer Hospital and Cancer Research Institute, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Xiaoting Jia
- Cancer Hospital and Cancer Research Institute, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Hao Liu
- Cancer Hospital and Cancer Research Institute, Guangzhou Medical University, Guangzhou, Guangdong Province, China.
| | - Zhimin He
- Cancer Hospital and Cancer Research Institute, Guangzhou Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
135
|
Zhang X, Dong J, He Y, Zhao M, Liu Z, Wang N, Jiang M, Zhang Z, Liu G, Liu H, Nie Y, Fan D, Tie J. miR-218 inhibited tumor angiogenesis by targeting ROBO1 in gastric cancer. Gene 2017; 615:42-49. [PMID: 28323002 DOI: 10.1016/j.gene.2017.03.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/22/2017] [Accepted: 03/16/2017] [Indexed: 12/20/2022]
Abstract
Aberrant expression of miRNAs is involved in several carcinogenic processes, including tumor growth, metastasis and angiogenesis. The aim of this study was to determine the role of miR-218 in gastric cancer angiogenesis. In situ hybridization was performed on a set of tissue microarray samples to assess the difference in miR-218 expression in vessels between tumor tissues and normal gastric mucosa. In vitro, ectopic expression of miR-218 disturbed the tubular structure and inhibited the migration of endothelial cells. Motility and tube formation were rescued when miR-218 was downregulated. Moreover, miR-218 suppressed endothelial cell sprouting in a fibrin bead sprouting assay. Subsequently, we identified ROBO1 as a target of miR-218 in endothelial cells and determined it was responsible for the effect of miR-218 on tumor angiogenesis. In vivo, local injection of mature miR-218 in xenografted tumors disrupted the vessel plexus and thus inhibited tumor growth. Taken together, our study demonstrated an anti-angiogenic role of miR-218 in gastric cancer and indicated that delivery of miR-218 may be a potential therapeutic strategy to inhibit tumor angiogenesis.
Collapse
Affiliation(s)
- Xiangyuan Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jiaqiang Dong
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yan He
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ming Zhao
- Yan'an University, Yan'an 716000, China
| | - Zhen Liu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Na Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Mingzuo Jiang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhe Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Gang Liu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Haiming Liu
- College of Computer Science and Technology, Jilin University, Changchun 120012, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Jun Tie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
136
|
Li J, Li T, Lu Y, Shen G, Guo H, Wu J, Lei C, Du F, Zhou F, Zhao X, Nie Y, Fan D. MiR-2392 suppresses metastasis and epithelial-mesenchymal transition by targeting MAML3 and WHSC1 in gastric cancer. FASEB J 2017; 31:3774-3786. [PMID: 28512191 DOI: 10.1096/fj.201601140rr] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 04/24/2017] [Indexed: 01/06/2023]
Abstract
MicroRNAs have emerged as essential regulators of various cellular processes. We identified the role and underlying mechanisms of miR-2392 in gastric cancer (GC) metastasis. MiR-2392 was down-regulated in GC cell lines and tissues, and overexpression of miR-2392 significantly inhibited GC invasion and metastasis in vitro and in vivo We identified MAML3 and WHSC1 as novel targets of miR-2392, and knockdown of MAML3 and WHSC1 had the same antimetastatic effect as that of miR-2392 in GC cells. These effects were clinically relevant, as low miR-2392 expression was correlated with high MAML3 and WHSC1 expression and poor survival in patients with GC. Furthermore, forced expression of miR-2392 substantially suppressed Slug and Twist1, transcriptional repressors of E-cadherin, by targeting MAML3 and WHSC1, respectively, resulting in inhibition of the epithelial-mesenchymal transition. These findings indicate that the miR-2392-MAML3/WHSC1-Slug/Twist1 regulatory axis plays a critical role in GC metastasis. Restoration of miR-2392 may be a therapeutic approach for blocking GC metastasis.-Li, J., Li, T., Lu, Y., Shen, G., Guo, H., Wu, J., Lei, C., Du, F., Zhou, F., Zhao, X., Nie, Y., Fan, D. MiR-2392 suppresses metastasis and epithelial-mesenchymal transition by targeting MAML3 and WHSC1 in gastric cancer.
Collapse
Affiliation(s)
- Jinjing Li
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Tingyu Li
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yuanyuan Lu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Gaofei Shen
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Hao Guo
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Jian Wu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Chao Lei
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Feng Du
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Fenli Zhou
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Xiaodi Zhao
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
137
|
Xiang Z, Zhou ZJ, Xia GK, Zhang XH, Wei ZW, Zhu JT, Yu J, Chen W, He Y, Schwarz RE, Brekken RA, Awasthi N, Zhang CH. A positive crosstalk between CXCR4 and CXCR2 promotes gastric cancer metastasis. Oncogene 2017; 36:5122-5133. [PMID: 28481874 DOI: 10.1038/onc.2017.108] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 02/16/2017] [Accepted: 03/14/2017] [Indexed: 02/07/2023]
Abstract
The molecular mechanism underlying gastric cancer (GC) invasion and metastasis is still poorly understood. In this study, we tried to investigate the roles of CXCR4 and CXCR2 signalings in gastric cancer metastasis. A highly invasive gastric cancer cell model was established. Chemokines receptors were profiled to search for the accountable ones. Then the underlying molecular mechanism was investigated using both in vitro and in vivo techniques, and the clinical relevance of CXCR4 and CXCR2 expression was studied in gastric cancer samples. CXCR4 and CXCR2 were highly expressed in a high invasive gastric cancer cell model and in gastric cancer tissues. Overexpression of CXCR4 and CXCR2 was associated with more advanced tumor stage and poorer survival for GC patients. CXCR4 and CXCR2 expression strongly correlated with each other in the way that CXCR2 expression changed accordingly with the activity of CXCR4 signaling and CXCR4 expression also changed in agreement with CXCR2 activity. Further studies demonstrated CXCR4 and CXCR2 can both activated NF-κB and STAT3 signaling, while NF-κBp65 can then transcriptionally activate CXCR4 and STAT3 can activate CXCR2 expression. This crosstalk between CXCR4 and CXCR2 contributed to EMT, migration and invasion of gastric cancer. Finally, Co-inhibition of CXCR4 and CXCR2 is more effective in reducing gastric cancer metastasis. Our results demonstrated that CXCR4 and CXCR2 cross-activate each other to promote the metastasis of gastric cancer.
Collapse
Affiliation(s)
- Z Xiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Z-J Zhou
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - G-K Xia
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - X-H Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Z-W Wei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - J-T Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - J Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - W Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Y He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Gastrointestinal Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - R E Schwarz
- Indiana University School of Medicine, South Bend, and IU Health Goshen Center for Cancer Care, Goshen, IN, USA
| | - R A Brekken
- Division of Surgical Oncology, Department of Surgery, and the Hamon Center for Therapeutic Oncology Research, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - N Awasthi
- Indiana University School of Medicine, South Bend, and IU Health Goshen Center for Cancer Care, Goshen, IN, USA
| | - C-H Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Gastric Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
138
|
Expression of miRNA-21, miRNA-107, miRNA-137 and miRNA-29b in meningioma. Clin Neurol Neurosurg 2017; 156:66-70. [PMID: 28349893 DOI: 10.1016/j.clineuro.2017.03.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/01/2017] [Accepted: 03/18/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Meningiomas are among the most common intracranial tumors, accounting for 30% of all tumors of the central nervous system. Recent studies analyzing microRNA (miRNA) profiles and functions in cancer have provided valuable information about the molecular pathogenesis of several tumor types, including glioblastoma multiforme (GBM), hepatocellular carcinoma, and breast, lung, colon, and prostate cancer. miRNAs are a family of small, endogenous, noncoding RNAs of 18-25 nucleotides. In this study, we carried out a genome-wide array screen comparing miRNA-21, miRNA-107, miRNA-137 and miRNA-29b expression in meningiomas. PATIENTS AND METHODS A total of 50 meningioma patients (16 men and 34 women) aged between 32 and 80 years were included. The study was conducted at Istanbul Research and Training Hospital Neurosurgery Clinic. RESULTS Our results have shown a significant increase in miRNA-21 expression with increasing histopathologic grade, while there was a significant reduction in miRNA-107 expression with the increasing histopathological grade. miRNA-137 and miRNA-29b expression did not differ significantly according to histopathologic grade. CONCLUSION The subject of our study, i.e. the association between miRNA expression and meningioma, is continuously gaining more importance in the wider context of the recent developments in genetic treatments.
Collapse
|
139
|
Wang Y, Xu P, Yao J, Yang R, Shi Z, Zhu X, Feng X, Gao S. MicroRNA-216b is Down-Regulated in Human Gastric Adenocarcinoma and Inhibits Proliferation and Cell Cycle Progression by Targeting Oncogene HDAC8. Target Oncol 2017; 11:197-207. [PMID: 26408293 DOI: 10.1007/s11523-015-0390-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE Accumulating evidence indicates that micro (mi)RNAs play a critical role in carcinogenesis and cancer progression; however, their role in the tumorigenesis of gastric adenocarcinoma remains unclear so the present study investigated this in gastric cancer (GC) tissues and cell lines. METHODS Human GC specimens (n = 57) and patient-paired non-cancerous specimens were obtained from patients at the First Affiliated Hospital, Henan University of Science and Technology. The AGS and GC9811 gastric cancer cell lines were also used. Expression levels of miR-216b and HDAC8 were examined by quantitative real-time PCR and the expression of HDAC8 was also examined by Western blotting and immunohistochemistry assay. The cell cycle progression was determined by FACS. MiR-216b inhibitor, mimics, and siRNA-HDAC8 transfections were performed to study the loss and gain of function. RESULTS We reported a significantly decreased expression of miR-216b in GC clinical specimens compared with paired non-cancerous tissues. We also observed a significant down-regulation of miR-216b expression in GC cell lines AGS and GC9811 (p < 0.0001). The introduction of miR-216b suppressed GC cell proliferation and cell cycle progression by targeting HDAC8, an oncogene shown to promote malignant tumor development with a potential miR-216b binding site in its 3' untranslated region. HDAC8 expression was shown to be significantly increased in AGS and GC9811 cell lines (p < 0.0001) and GC tissues compared with controls. Moreover, HDAC8 inhibition suppressed cell cycle progression compared with control groups (22 % ± 1.6 % vs 34 % ± 2.1), indicating that HDAC8 may function as an oncogene in the development of GC. Furthermore, HDAC8 expression was negatively correlated (p < 0.0001), while miR-216b expression was positively correlated with the clinical outcome of GC patients (p < 0.0001). DISCUSSION Our data suggest that miR-216b functions as a tumor suppressor in human GC by, at least partially, targeting HDAC8.
Collapse
Affiliation(s)
- Ying Wang
- Oncology Department of the First Affiliated Hospital of Henan University of Science and Technology, No. 24 Jinghua Road, Luoyang, Henan, 471003, China.
| | - Po Xu
- Urology Surgery Department of the First Affiliated Hospital of Henan University of Science and Technology, No. 24 Jinghua Road, Luoyang, Henan, 471003, China
| | - Jun Yao
- Oncology Department of the First Affiliated Hospital of Henan University of Science and Technology, No. 24 Jinghua Road, Luoyang, Henan, 471003, China
| | - Ruina Yang
- Oncology Department of the First Affiliated Hospital of Henan University of Science and Technology, No. 24 Jinghua Road, Luoyang, Henan, 471003, China
| | - Zhenguo Shi
- Urology Surgery Department of the First Affiliated Hospital of Henan University of Science and Technology, No. 24 Jinghua Road, Luoyang, Henan, 471003, China
| | - Xiaojuan Zhu
- Oncology Department of the First Affiliated Hospital of Henan University of Science and Technology, No. 24 Jinghua Road, Luoyang, Henan, 471003, China
| | - Xiaoshan Feng
- Oncology Department of the First Affiliated Hospital of Henan University of Science and Technology, No. 24 Jinghua Road, Luoyang, Henan, 471003, China.
| | - Shegan Gao
- Oncology Department of the First Affiliated Hospital of Henan University of Science and Technology, No. 24 Jinghua Road, Luoyang, Henan, 471003, China.
| |
Collapse
|
140
|
Predictive Value of MiR-219-1, MiR-938, MiR-34b/c, and MiR-218 Polymorphisms for Gastric Cancer Susceptibility and Prognosis. DISEASE MARKERS 2017; 2017:4731891. [PMID: 28298809 PMCID: PMC5337385 DOI: 10.1155/2017/4731891] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/28/2016] [Accepted: 01/16/2017] [Indexed: 12/15/2022]
Abstract
Gastric cancer (GC) is one of the most prominent global cancer-related health threats. Genes play a key role in the precise mechanisms of gastric cancer. SNPs in mi-RNAs could affect mRNA expression and then affect the risk and prognosis of GC. Firstly, we have decided to perform a case-control study which included 897 GC patients and 992 controls to evaluate the association of miR-219-1 rs213210, miR-938 rs2505901, miR-34b/c rs4938723, and miR-218 rs11134527 polymorphisms with gastric cancer susceptibility. Secondly, among the 897 GC patients above, 755 cases underwent a radical operation, without distant metastasis and with negative surgical margins included in the survival analysis to evaluate the association of the four SNPs above with gastric cancer prognosis. The C/T or C/C genotypes of rs213210 were related to a lower GC risk (OR = 0.76, 95% CI: 0.62–0.93, P = 0.009) compared to the T/T genotype. Rs11134527 in miR-218 was associated with GC survival, and the G/A and G/G genotypes of rs11134527 resulted in a decreased risk of death when compared with the A/A genotype (HR = 0.75, 95% CI: 0.61–0.95, P = 0.016). This study found that miR-219-1 rs213210 polymorphism was associated with GC susceptibility and rs11134527 in miR-218 was positively correlated with GC prognosis.
Collapse
|
141
|
Zhu W, Shao Y, Peng Y. MicroRNA-218 inhibits tumor growth and increases chemosensitivity to CDDP treatment by targeting BCAT1 in prostate cancer. Mol Carcinog 2017; 56:1570-1577. [PMID: 28052414 DOI: 10.1002/mc.22612] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 11/16/2016] [Accepted: 12/31/2016] [Indexed: 12/16/2022]
Abstract
MicroRNAs have been reported to be associated with chemosensitivity of several types of cancers. However, the underlying molecular mechanisms are poorly understood. In this study, we explored miR-218 increased the chemosensitivity to cis-diaminedichloroplatinum treatment of prostate cancer. We found that the expression level of miR-218 was down-regulated in the human prostate cancer specimens. Moreover, overexpression of miR-218 inhibited cell viability, migration, and invasion in PC3 and DU145 cells. Furthermore, we demonstrated that the tumor suppressive role of miR-218 was mediated by negatively regulating branched-chain amino acid transaminase 1 (BCAT1) protein expression. Importantly, overexpression of BCAT1 decreased the chemosensitivity to CDDP treatment of PC3 and DU145 cells. Our study is the first to identify the positive role of miR-218 in chemosensitivity, which will facilitate the development of novel therapeutic strategies for prostate cancer in the future.
Collapse
Affiliation(s)
- Wenjing Zhu
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqun Shao
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Peng
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
142
|
Hu L, Ai J, Long H, Liu W, Wang X, Zuo Y, Li Y, Wu Q, Deng Y. Integrative microRNA and gene profiling data analysis reveals novel biomarkers and mechanisms for lung cancer. Oncotarget 2017; 7:8441-54. [PMID: 26870998 PMCID: PMC4890978 DOI: 10.18632/oncotarget.7264] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 01/13/2016] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Studies on the accuracy of microRNAs (miRNAs) in diagnosing non-small cell lung cancer (NSCLC) have still controversial. Therefore, we conduct to systematically identify miRNAs related to NSCLC, and their target genes expression changes using microarray data sets. METHODS We screened out five miRNAs and six genes microarray data sets that contained miRNAs and genes expression in NSCLC from Gene Expression Omnibus. RESULTS Our analysis results indicated that fourteen miRNAs were significantly dysregulated in NSCLC. Five of them were up-regulated (miR-9, miR-708, miR-296-3p, miR-892b, miR-140-5P) while nine were down-regulated (miR-584, miR-218, miR-30b, miR-522, miR486-5P, miR-34c-3p, miR-34b, miR-516b, miR-592). The integrating diagnosis sensitivity (SE) and specificity (SP) were 82.6% and 89.9%, respectively. We also found that 4 target genes (p < 0.05, fold change > 2.0) were significant correlation with the 14 discovered miRNAs, and the classifiers we built from one training set predicted the validation set with higher accuracy (SE = 0.987, SP = 0.824). CONCLUSIONS Our results demonstrate that integrating miRNAs and target genes are valuable for identifying promising biomarkers, and provided a new insight on underlying mechanism of NSCLC. Further, our well-designed validation studies surely warrant the investigation of the role of target genes related to these 14 miRNAs in the prediction and development of NSCLC.
Collapse
Affiliation(s)
- Ling Hu
- Department of Anesthesiology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China.,Department of Internal Medicine and Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Junmei Ai
- Department of Internal Medicine and Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Hui Long
- Department of Gastroenterology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Weijun Liu
- Department of Orthopedics, Pu Ai Hospital, Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomei Wang
- Department of Biological Science and Technology, Wuhan Bioengineering Institute, Wuhan, China
| | - Yi Zuo
- Department of Orthopedic, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Yan Li
- Department of Internal Medicine and Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Qingming Wu
- Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Youping Deng
- Medical College, Wuhan University of Science and Technology, Wuhan, China.,Department of Internal Medicine and Biochemistry, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
143
|
Azarnezhad A, Mehdipour P. Cancer Genetics at a Glance: The Comprehensive Insights. CANCER GENETICS AND PSYCHOTHERAPY 2017:79-389. [DOI: 10.1007/978-3-319-64550-6_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
144
|
Establishment of a gastric cancer subline with high metastatic potential using a novel microfluidic system. Sci Rep 2016; 6:38376. [PMID: 27917905 PMCID: PMC5137147 DOI: 10.1038/srep38376] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 11/08/2016] [Indexed: 12/11/2022] Open
Abstract
Metastasis is an important hallmark of malignant tumors. In this study, we developed a microfluidic system to screen highly metastatic sublines via differential resolution of cell invasiveness. The system was composed of a PDMS-glass device connected with a syringe pump and a Petri dish. To facilitate the selection process, a long-term cell invasion driving force based on a chemotactic factor gradient was created using the Petri dish-based liquid supply pattern, and the invasive cells were collected for round-by-round selection via an open region in the chip. Using the system, we established an SGC-7901/B2 subline from the human gastric cancer SGC-7901 cell line by only two rounds of selection. In vitro assays showed that the SGC-7901/B2 cells were superior to the parental cells in proliferation and invasiveness. Furthermore, an in vivo tumorigenicity assay demonstrated that compared with the parental cells, the subline had stronger spontaneous metastatic and proliferative capability, which led to a shorter survival duration. Additionally, the protein expression differences including E-cadherin and Smad3 between the subline and parental cells were revealed. In conclusion, this microfluidic system is a highly effective tool for selecting highly metastatic sublines, and SGC-7901/B2 cells could serve as a potential model for tumor metastasis research.
Collapse
|
145
|
Tie J, Zhang X, Fan D. Epigenetic roles in the malignant transformation of gastric mucosal cells. Cell Mol Life Sci 2016; 73:4599-4610. [PMID: 27464701 PMCID: PMC5097112 DOI: 10.1007/s00018-016-2308-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/10/2016] [Accepted: 07/08/2016] [Indexed: 12/14/2022]
Abstract
Gastric carcinogenesis occurs when gastric epithelial cells transition through the initial, immortal, premalignant, and malignant stages of transformation. Epigenetic regulations contribute to this multistep process. Due to the critical role of epigenetic modifications , these changes are highly likely to be of clinical use in the future as new biomarkers and therapeutic targets for the early detection and treatment of cancers. Here, we summarize the recent findings on how epigenetic modifications, including DNA methylation, histone modifications, and non-coding RNAs, regulate gastric carcinogenesis, and we discuss potential new strategies for the diagnosis and treatments of gastric cancer. The strategies may be helpful in the further understanding of epigenetic regulation in human diseases.
Collapse
Affiliation(s)
- Jun Tie
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, No. 127, West Chang-Le Road, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Xiangyuan Zhang
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, No. 127, West Chang-Le Road, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, No. 127, West Chang-Le Road, Xi'an, Shaanxi, 710032, People's Republic of China.
| |
Collapse
|
146
|
hsa-miR-376c-3p Regulates Gastric Tumor Growth Both In Vitro and In Vivo. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9604257. [PMID: 27965982 PMCID: PMC5124681 DOI: 10.1155/2016/9604257] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/10/2016] [Indexed: 11/18/2022]
Abstract
Background. In recent studies, aberrant expression of various microRNAs (miRNAs) is reported to be associated with gastric cancer metastasis. Method. Overexpression construct and inhibitor of hsa-miR-376c-3p were expressed in human gastric adenocarcinoma cell line SGC-7901. The expression level of tumor related genes was detected by qPCR, western blot, and immunostaining. Cell apoptosis was determined by flow cytometry. Xenograft of SGC-7901 cells was used to elucidate the function of hsa-miR-376c-3p in gastric tumor growth in vivo. Result. Expression of hsa-miR-376c-3p was detected in SGC-7901 cells. Downregulation of hsa-miR-376c-3p increased the expression level of BCL-2 and decreased the expression of smad4 and BAD. On the contrary, overexpression of hsa-miR-376c-3p increased the expression of BAD and smad4, while it led to the decreasing expression level of BCL-2. Overexpression of hsa-miR-376c-3p also promoted cell apoptosis in vitro and inhibited gastric tumor growth in vivo. Furthermore, the expression of BCL-2 was higher and expression of smad4 and BAD was lower in tumor tissue than the tissue adjacent to tumor from gastric cancer patients. Conclusion. This study demonstrated that hsa-miR-376c-3p plays an important role in the inhibition of gastric tumor growth and tumor related gene expression both in vitro and in vivo.
Collapse
|
147
|
Tang S, Wang D, Zhang Q, Li L. miR-218 suppresses gastric cancer cell proliferation and invasion via regulation of angiopoietin-2. Exp Ther Med 2016; 12:3837-3842. [PMID: 28105117 PMCID: PMC5228446 DOI: 10.3892/etm.2016.3893] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 09/01/2016] [Indexed: 12/14/2022] Open
Abstract
Novel targeted therapies need to be developed for gastric cancer, the third most common cancer type and the second most common cause of cancer-related mortality in China. Previous studies indicate that angiopoietin (Ang)-2 serves a role in the proliferation, migration, invasion and adhesion of malignant cells. The present study identified, using functional studies, that exogenous expression of miR-218 increased migration of NCI-87 and HGC-27 gastric cancer cells, which coincided with a reduction in the expression of Ang-2. In addition, intratumoral delivery of miR-218 inhibited proliferation and angiogenesis of gastric cancer cells in vivo, with a corresponding decreased in Ang-2 expression. These results indicate that miR-218 serves an important role in gastric cancer tumorigenesis through regulating the expression of Ang-2. Therefore, components of miR-218/Ang-2 signaling could provide novel therapeutic targets for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Sifeng Tang
- Department of Gastrointestinal Surgery, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China; Department of General Surgery, Laiwu People's Hospital, Laiwu, Shandong 271100, P.R. China
| | - Deyou Wang
- Department of General Surgery, Laiwu People's Hospital, Laiwu, Shandong 271100, P.R. China
| | - Qiwen Zhang
- Department of General Surgery, Laiwu People's Hospital, Laiwu, Shandong 271100, P.R. China
| | - Leping Li
- Department of Gastrointestinal Surgery, School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China; Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
148
|
MicroRNA-218 inhibits the proliferation, migration, and invasion and promotes apoptosis of gastric cancer cells by targeting LASP1. Tumour Biol 2016; 37:15241-15252. [DOI: 10.1007/s13277-016-5388-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/09/2016] [Indexed: 12/20/2022] Open
|
149
|
da Silva Oliveira KC, Thomaz Araújo TM, Albuquerque CI, Barata GA, Gigek CO, Leal MF, Wisnieski F, Rodrigues Mello Junior FA, Khayat AS, de Assumpção PP, Rodriguez Burbano RM, Smith MC, Calcagno DQ. Role of miRNAs and their potential to be useful as diagnostic and prognostic biomarkers in gastric cancer. World J Gastroenterol 2016; 22:7951-7962. [PMID: 27672290 PMCID: PMC5028809 DOI: 10.3748/wjg.v22.i35.7951] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 06/14/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023] Open
Abstract
Alterations in epigenetic control of gene expression play an important role in many diseases, including gastric cancer. Many studies have identified a large number of upregulated oncogenic miRNAs and downregulated tumour-suppressor miRNAs in this type of cancer. In this review, we provide an overview of the role of miRNAs, pointing to their potential to be useful as diagnostic and/or prognostic biomarkers in gastric cancer. Moreover, we discuss the influence of polymorphisms and epigenetic modifications on miRNA activity.
Collapse
|
150
|
Wang F, Wang J, Yang X, Chen D, Wang L. MiR-424-5p participates in esophageal squamous cell carcinoma invasion and metastasis via SMAD7 pathway mediated EMT. Diagn Pathol 2016; 11:88. [PMID: 27628042 PMCID: PMC5024440 DOI: 10.1186/s13000-016-0536-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 09/02/2016] [Indexed: 12/13/2022] Open
Abstract
Backgrounds ESCC is a life-threatening disease due to invasion and metastasis in the early stage. Great efforts had been made to detect the molecular mechanisms which led to the invasion and metastasis in ESCC. Recent evidence had suggested that deregulation of miR-424-5p took an important role in cancers. However, its role and functional mechanism in ESCC had seldom been elucidated. Methods The expression levels of miR-424-5p were detected in ESCC tissues and cell lines by real-time PCR methods. Then, the invasion, metastasis and proliferation ability of ESCC cell lines transfected with miR-424-5p mimics were analyzed separately by transwell invasion assay, wound healing assay and cell proliferation assay. Finally, the target gene of miR-424-5p was studied and verified by luciferase activity assay. And the role of miR-424-5p in EMT was also investigated by real-time PCR and western blot assay. Results We showed that the expression levels of miR-424-5p were decreased both in ESCC tissues and cell lines. Furthermore, the expression levels of miR-424-5p were negatively linked to lymph node metastasis in ESCC tissues. Restoration of miR-424-5p in EC-1 cells by using miR-424-5p mimics could decrease the invasion, metastasis and proliferation of EC-1 cells, indicating its role in inhibition on the invasion and metastasis ability of ESCC cells and tissues. In addition, we demonstrated that SMAD7 was a specific target gene for miR-424-5p by luciferase activity assay and miR-424-5p could not only negatively regulate SMAD7 expression but also participate in EMT via SMAD7, because overexpression of SMAD7 could partly enhance the miR-424-5p anti-EMT function. Conclusions Our results described that miR-424-5p -SMAD7 pathway contributed to ESCC invasion and metastasis and up-regulation of miR-424-5p perhaps provided a strategy for preventing tumor invasion, metastasis.
Collapse
Affiliation(s)
- Feng Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Jun Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Xuan Yang
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Danjie Chen
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
| | - Liuxing Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
| |
Collapse
|