101
|
Prati B, da Silva Abjaude W, Termini L, Morale M, Herbster S, Longatto-Filho A, Nunes RAL, Córdoba Camacho LC, Rabelo-Santos SH, Zeferino LC, Aguayo F, Boccardo E. Three Prime Repair Exonuclease 1 (TREX1) expression correlates with cervical cancer cells growth in vitro and disease progression in vivo. Sci Rep 2019; 9:351. [PMID: 30674977 PMCID: PMC6344518 DOI: 10.1038/s41598-018-37064-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 10/17/2018] [Indexed: 12/17/2022] Open
Abstract
Alterations in specific DNA damage repair mechanisms in the presence of human papillomavirus (HPV) infection have been described in different experimental models. However, the global effect of HPV on the expression of genes involved in these pathways has not been analyzed in detail. In the present study, we compared the expression profile of 135 genes involved in DNA damage repair among primary human keratinocytes (PHK), HPV-positive (SiHa and HeLa) and HPV-negative (C33A) cervical cancer derived cell lines. We identified 9 genes which expression pattern distinguishes HPV-positive tumor cell lines from C33A. Moreover, we observed that Three Prime Repair Exonuclease 1 (TREX1) expression is upregulated exclusively in HPV-transformed cell lines and PHK expressing HPV16 E6 and E7 oncogenes. We demonstrated that TREX1 silencing greatly affects tumor cells clonogenic and anchorage independent growth potential. We showed that this effect is associated with p53 upregulation, accumulation of subG1 cells, and requires the expression of E7 from high-risk HPV types. Finally, we observed an increase in TREX1 levels in precancerous lesions, squamous carcinomas and adenocarcinomas clinical samples. Altogether, our results indicate that TREX1 upregulation is important for cervical tumor cells growth and may contribute with tumor establishment and progression.
Collapse
Affiliation(s)
- Bruna Prati
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo (USP), Av. Prof. Lineu Prestes 1374, 05508-900, São Paulo, SP, Brazil
| | - Walason da Silva Abjaude
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo (USP), Av. Prof. Lineu Prestes 1374, 05508-900, São Paulo, SP, Brazil
| | - Lara Termini
- Centro de Investigação Translacional em Oncologia (LIM24), Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
| | - Mirian Morale
- Department of Biochemistry, Institute of Chemistry, USP, São Paulo, Brazil
| | - Suellen Herbster
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo (USP), Av. Prof. Lineu Prestes 1374, 05508-900, São Paulo, SP, Brazil
| | - Adhemar Longatto-Filho
- Laboratory of Medical Investigation (LIM 14), Department of Pathology, School of Medicine, USP, Av. Dr. Arnaldo 455, São Paulo, 01246-903, Brazil.,Life and Health Sciences Research Institute, School of Health Sciences, ICVS/3B's - PT Government Associate Laboratory, University of Minho, Braga, Guimarães, Portugal.,Molecular Oncology Research Center, Barretos Cancer Hospital, Pio XII Foundation, Barretos, Rua Antenor Duarte Villela, 1331, Barretos, 14784-400, Brazil
| | - Rafaella Almeida Lima Nunes
- Centro de Investigação Translacional em Oncologia (LIM24), Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
| | - Lizeth Carolina Córdoba Camacho
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo (USP), Av. Prof. Lineu Prestes 1374, 05508-900, São Paulo, SP, Brazil.,Laboratório de Oncologia Experimental, Departamento de Radiologia, Faculdade de Medicina, USP, São Paulo, SP, Brazil.,Centro de Investigação Translacional em Oncologia, ICESP, São Paulo, SP, Brazil
| | | | - Luiz Carlos Zeferino
- School of Medical Sciences, State University of Campinas (UNICAMP), Rua Alexander Fleming 101, 13083-881, Campinas, SP, Brazil
| | - Francisco Aguayo
- Basic and Clinical Oncology Department, Faculty of Medicine, University of Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrique Boccardo
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo (USP), Av. Prof. Lineu Prestes 1374, 05508-900, São Paulo, SP, Brazil.
| |
Collapse
|
102
|
Zou Z, Zheng B, Li J, Lv X, Zhang H, Yu F, Kong L, Li Y, Yu M, Fang L, Liang B. TPX2 level correlates with cholangiocarcinoma cell proliferation, apoptosis, and EMT. Biomed Pharmacother 2018; 107:1286-1293. [DOI: 10.1016/j.biopha.2018.08.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 12/27/2022] Open
|
103
|
Application of induced pluripotent stem cell transplants: Autologous or allogeneic? Life Sci 2018; 212:145-149. [DOI: 10.1016/j.lfs.2018.09.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 09/30/2018] [Indexed: 12/14/2022]
|
104
|
Wang J, Wang Y, Shen F, Xu Y, Zhang Y, Zou X, Zhou J, Chen Y. Maternal embryonic leucine zipper kinase: A novel biomarker and a potential therapeutic target of cervical cancer. Cancer Med 2018; 7:5665-5678. [PMID: 30334367 PMCID: PMC6246930 DOI: 10.1002/cam4.1816] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/12/2018] [Accepted: 08/31/2018] [Indexed: 12/30/2022] Open
Abstract
Maternal embryo leucine zipper kinase (MELK) is highly expressed in a variety of malignant tumors and involved in cell cycle regulation, cell proliferation, apoptosis, tumor formation etc However, the biological effects of MELK in cervical cancer are still uninvestigated. This study aimed to explore the expression of MELK in cervical cancer, as well as its effects on the proliferation, apoptosis, DNA damage repair on cervical cancer cell line in vitro and to provide novel ideas for further improving the clinical efficacy of cervical cancer. Immunohistochemistry, Western blot, RT‐qPCR, CCK8, and immunofluorescence techniques were used to detect the expression of MELK in cervical cancer tissues, paracancerous tissues, and cervical cancer cell lines. Several cervical cancer cell lines were treated with MELK knockdown by siRNA and MELK selective inhibitor OTSSP167. The effects on proliferation, apoptosis, and colony formation capacity, and tumor cell DNA damage repair‐related factor were detected in cell lines. Our data showed that the high expression rate of MELK in cervical cancer patients was 56.92%. MELK expression in cervical cancer samples was significantly higher than that in paraneoplastic tissues. Highly expressed MELK correlated with the cervical histopathological grading and greatly increased with the cervical histopathological grading, from normal cervix and cervical intraepithelial neoplasia to cervical cancer. Moreover, the abnormal expression of MELK was related to cervical cancer metastasis at early stage. The knockdown of MELK with siRNA and OTSSP167 had strong inhibition effects on the proliferation, apoptosis, and colony formation of cervical cancer cells. MELK knockdown could also aggravate the DNA damage of cervical cancer cells possibly by homologous recombination repair pathway. Therefore, MELK may be a predicting marker of poor prognosis of cervical cancer and may also be a new therapeutic target for cervical cancer, providing ideas for improving the therapeutic effect of cervical cancer.
Collapse
Affiliation(s)
- Juan Wang
- Department of Obstetrics and Gynecology, The First Hospital Affiliated Soochow University, Suzhou, China
| | - Yamei Wang
- Department of Obstetrics and Gynecology, The First Hospital Affiliated Soochow University, Suzhou, China
| | - Fangrong Shen
- Department of Obstetrics and Gynecology, The First Hospital Affiliated Soochow University, Suzhou, China
| | - Yanting Xu
- Department of Obstetrics and Gynecology, The First Hospital Affiliated Soochow University, Suzhou, China
| | - Yinghui Zhang
- Department of Obstetrics and Gynecology, The First Hospital Affiliated Soochow University, Suzhou, China
| | - Xinwei Zou
- Department of Obstetrics and Gynecology, The First Hospital Affiliated Soochow University, Suzhou, China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, The First Hospital Affiliated Soochow University, Suzhou, China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Hospital Affiliated Soochow University, Suzhou, China
| |
Collapse
|
105
|
Tang X, Xu Y, Lu L, Jiao Y, Liu J, Wang L, Zhao H. Identification of key candidate genes and small molecule drugs in cervical cancer by bioinformatics strategy. Cancer Manag Res 2018; 10:3533-3549. [PMID: 30271202 PMCID: PMC6145638 DOI: 10.2147/cmar.s171661] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose Cervical cancer (CC) is one of the most common malignant tumors among women. The present study aimed at integrating two expression profile datasets to identify critical genes and potential drugs in CC. Materials and methods Expression profiles, GSE7803 and GSE9750, were integrated using bioinformatics methods, including differentially expressed genes analysis, Kyoto Encyclopedia of Genes and Genomes pathway analysis, and protein–protein interaction (PPI) network construction. Subsequently, survival analysis was performed among the key genes using Gene Expression Profiling Interactive Analysis websites. Connectivity Map (CMap) was used to query potential drugs for CC. Results A total of 145 upregulated genes and 135 downregulated genes in CC were identified. The functional changes of these differentially expressed genes related to CC were mainly associated with cell cycle, DNA replication, p53 signaling pathway, and oocyte meiosis. A PPI network was identified by STRING with 220 nodes and 2,111 edges. Thirteen key genes were identified as the intersecting genes of the enrichment pathways and the top 20 nodes in PPI network. Survival analysis revealed that high mRNA expression of MCM2, PCNA, and RFC4 was significantly associated with longer overall survival, and the survival was significantly better in the low-expression RRM2 group. Moreover, CMap predicted nine small molecules as possible adjuvant drugs to treat CC. Conclusion Our study found key dysregulated genes involved in CC and potential drugs to combat it, which might provide insights into CC pathogenesis and might shed light on potential CC treatments.
Collapse
Affiliation(s)
- Xin Tang
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Yicong Xu
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China, .,Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Kunming, China,
| | - Lin Lu
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China, .,Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Kunming, China,
| | - Yang Jiao
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China, .,Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Kunming, China,
| | - Jianjun Liu
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China, .,Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Kunming, China,
| | - Linlin Wang
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China, .,Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Kunming, China,
| | - Hongbo Zhao
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China, .,Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Kunming, China,
| |
Collapse
|
106
|
Wang S, Chen X. Identification of potential biomarkers in cervical cancer with combined public mRNA and miRNA expression microarray data analysis. Oncol Lett 2018; 16:5200-5208. [PMID: 30250588 PMCID: PMC6144068 DOI: 10.3892/ol.2018.9323] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 07/23/2018] [Indexed: 12/31/2022] Open
Abstract
Cervical cancer is the fourth most prevalent malignancy in females worldwide. Early diagnosis is key to improving survival rates. Molecular biomarkers are an important method for diagnosing a number of types of cancer, including cervical cancer. The present study utilized public data from three mRNA microarray datasets and one microRNA dataset to analyze the key genes involved in cervical cancer. The mRNA and microRNA expression profile datasets (GSE9750, GSE46857, GSE67522 and GSE30656) were downloaded from the Gene Expression Omnibus database (GEO). Differentially expressed genes (DEGs) and microRNAs (DEMs) were screened using the online tool GEO2R. By using the DEGs consistent across the three mRNA datasets, a functional and pathway enrichment analysis was performed using the Database for Annotation, Visualization and Integrated Discovery. A protein-protein interaction (PPI) network was constructed and module analysis performed using the Search Tool for the Retrieval of Interacting Genes. Validated target genes of the DEMs were identified using the miRecords website. Using the identified target genes of the DEMs, a survival analysis was performed using the OncoLnc online tool. A total of 73 DEGs and 19 DEMs were screened from the microarray expression profile datasets. ‘Integrin-mediated’, ‘proteolysis’ and ‘phosphoinositide 3 kinase-protein kinase 3’ signaling pathways were the most enriched in the DEGs. Three of the DEGs, including Ras homolog family member B (RhoB), stathmin 1 (STMN1) and cyclin D1 (CCNB1) were validated DEM target genes. The OncoLnc survival analysis identified that RhoB was associated with a significantly longer overall survival, whereas STMN1 was associated with a significantly reduced overall survival time in patients with cervical cancer. Finally, data from The Cancer Genome Atlas revealed an association between the mRNA expression levels of RhoB and STMN1, and the overall survival time for patients with cervical cancer. In conclusion, RhoB and STMN1 were identified as key genes that may provide potential targets for cervical cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Sizhe Wang
- Department of Women Health Care, Beijing Chaoyang District Maternal and Child Health Care Hospital, Beijing 100000, P.R. China
| | - Xiaojin Chen
- Department of Women Health Care, Beijing Chaoyang District Maternal and Child Health Care Hospital, Beijing 100000, P.R. China
| |
Collapse
|
107
|
Sun B, Lin G, Ji D, Li S, Chi G, Jin X. Dysfunction of Sister Chromatids Separation Promotes Progression of Hepatocellular Carcinoma According to Analysis of Gene Expression Profiling. Front Physiol 2018; 9:1019. [PMID: 30100882 PMCID: PMC6072861 DOI: 10.3389/fphys.2018.01019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022] Open
Abstract
Despite studying the various molecular mechanisms of hepatocellular carcinoma (HCC), effective drugs and biomarkers in HCC therapy are still scarce. The present study was designed to investigate dysregulated pathways, novel biomarkers and therapeutic targets for HCC. The gene expression dataset of GSE14520, which included 362 tumor and their paired non-tumor tissues of HCC, was extracted for processing by the Robust multi-array average (RMA) algorithm in the R environment. SAM methods were leveraged to identify differentially expressed genes (DEGs). Functional analysis of DEGs was performed using DAVID. The GeneMania and Cytohubba were used to construct the PPI network. To avoid individual bias, GSEA and survival analysis were employed to verify the results. The results of these analyses indicated that separation of sister chromatids was the most aberrant phase in the progression of HCC, and the most frequently involved genes, EZH2, GINS1, TPX2, CENPF, and BUB1B, require further study to be used as drug targets or biomarkers in diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Baozhen Sun
- Department of Hepatopancreatobiliary, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Guibo Lin
- First Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Degang Ji
- Department of Hepatopancreatobiliary, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shuo Li
- Department of Hepatopancreatobiliary, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Guonan Chi
- First Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xingyi Jin
- First Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
108
|
Tan MS, Chang SW, Cheah PL, Yap HJ. Integrative machine learning analysis of multiple gene expression profiles in cervical cancer. PeerJ 2018; 6:e5285. [PMID: 30065881 PMCID: PMC6064203 DOI: 10.7717/peerj.5285] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/02/2018] [Indexed: 01/06/2023] Open
Abstract
Although most of the cervical cancer cases are reported to be closely related to the Human Papillomavirus (HPV) infection, there is a need to study genes that stand up differentially in the final actualization of cervical cancers following HPV infection. In this study, we proposed an integrative machine learning approach to analyse multiple gene expression profiles in cervical cancer in order to identify a set of genetic markers that are associated with and may eventually aid in the diagnosis or prognosis of cervical cancers. The proposed integrative analysis is composed of three steps: namely, (i) gene expression analysis of individual dataset; (ii) meta-analysis of multiple datasets; and (iii) feature selection and machine learning analysis. As a result, 21 gene expressions were identified through the integrative machine learning analysis which including seven supervised and one unsupervised methods. A functional analysis with GSEA (Gene Set Enrichment Analysis) was performed on the selected 21-gene expression set and showed significant enrichment in a nine-potential gene expression signature, namely PEG3, SPON1, BTD and RPLP2 (upregulated genes) and PRDX3, COPB2, LSM3, SLC5A3 and AS1B (downregulated genes).
Collapse
Affiliation(s)
- Mei Sze Tan
- Bioinformatics Programme, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Siow-Wee Chang
- Bioinformatics Programme, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Phaik Leng Cheah
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Hwa Jen Yap
- Department of Mechanical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
109
|
Kori M, Yalcin Arga K. Potential biomarkers and therapeutic targets in cervical cancer: Insights from the meta-analysis of transcriptomics data within network biomedicine perspective. PLoS One 2018; 13:e0200717. [PMID: 30020984 PMCID: PMC6051662 DOI: 10.1371/journal.pone.0200717] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 07/02/2018] [Indexed: 12/13/2022] Open
Abstract
The malignant neoplasm of the cervix, cervical cancer, has effects on the reproductive tract. Although infection with oncogenic human papillomavirus is essential for cervical cancer development, it alone is insufficient to explain the development of cervical cancer. Therefore, other risk factors such as host genetic factors should be identified, and their importance in cervical cancer induction should be determined. Although gene expression profiling studies in the last decade have made significant molecular findings about cervical cancer, adequate screening and effective treatment strategies have yet to be achieved. In the current study, meta-analysis was performed on cervical cancer-associated transcriptome data and reporter biomolecules were identified at RNA (mRNA, miRNA), protein (receptor, transcription factor, etc.), and metabolite levels by the integration of gene expression profiles with genome-scale biomolecular networks. This approach revealed already-known biomarkers, tumor suppressors and oncogenes in cervical cancer as well as various receptors (e.g. ephrin receptors EPHA4, EPHA5, and EPHB2; endothelin receptors EDNRA and EDNRB; nuclear receptors NCOA3, NR2C1, and NR2C2), miRNAs (e.g., miR-192-5p, miR-193b-3p, and miR-215-5p), transcription factors (particularly E2F4, ETS1, and CUTL1), other proteins (e.g., KAT2B, PARP1, CDK1, GSK3B, WNK1, and CRYAB), and metabolites (particularly, arachidonic acids) as novel biomarker candidates and potential therapeutic targets. The differential expression profiles of all reporter biomolecules were cross-validated in independent RNA-Seq and miRNA-Seq datasets, and the prognostic power of several reporter biomolecules, including KAT2B, PCNA, CD86, miR-192-5p and miR-215-5p was also demonstrated. In this study, we reported valuable data for further experimental and clinical efforts, because the proposed biomolecules have significant potential as systems biomarkers for screening or therapeutic purposes in cervical carcinoma.
Collapse
Affiliation(s)
- Medi Kori
- Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Turkey
| | - Kazim Yalcin Arga
- Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Turkey
| |
Collapse
|
110
|
SOX17 restrains proliferation and tumor formation by down-regulating activity of the Wnt/β-catenin signaling pathway via trans-suppressing β-catenin in cervical cancer. Cell Death Dis 2018; 9:741. [PMID: 29970906 PMCID: PMC6030085 DOI: 10.1038/s41419-018-0782-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 01/17/2023]
Abstract
The SRY-box containing gene 17 (SOX17) is considered as a regulator in stemness maintenance and a suppressor in some malignant tumors. However, the biological function and molecular mechanism of SOX17 in the process of initiation and progression of cervical cancer remain obscure. In this study, immunohistochemistry showed that the expression of SOX17 was high in the normal cervix, moderate in the high-grade squamous intraepithelial lesion, and low in the cervical cancer. SOX17 inhibited the proliferation and viability of cervical cancer cells in vitro as well as tumor formation in vivo. Additionally, SOX17 induced the cell cycle arrest at the transition from the G0/G1 phase to the S phase. The TOP/ FOP-Flash reporter assay and Western blotting showed SOX17 inhibited the activity of the Wnt/β-catenin signaling pathway in cervical cancer. Further, firefly luciferase reporter assay and quantitative chromatin immunoprecipitation (qChIP) assays confirmed that SOX17 trans-suppressed the expression of β-catenin by directly binding to the specific region of the β-catenin promoter. Together, our data demonstrated that SOX17 restrained the proliferation and tumor formation by down-regulating the activity of the Wnt/β-catenin signaling pathway via trans-suppression of β-catenin in cervical cancer.
Collapse
|
111
|
Ding F, Zhang S, Gao S, Shang J, Li Y, Cui N, Zhao Q. MiR-137 functions as a tumor suppressor in pancreatic cancer by targeting MRGBP. J Cell Biochem 2018; 119:4799-4807. [PMID: 29331027 DOI: 10.1002/jcb.26676] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/11/2018] [Indexed: 12/29/2022]
Abstract
miRNAs are small noncoding RNAs that act as critical epigenetic regulators in tumor carcinogenesis. In this study, our data showed that miR-137 was significantly downregulated in 58 pairs of human pancreatic cancer (PanCa) tissues and PanCa cell lines. Furthermore, the deregulated miR-137 was correlated with increased tumor size, higher TNM stage, and worse prognosis in pancreatic cancer. Functional studies demonstrated that overexpression of miR-137 dramatically suppressed cell proliferation and induced cell apoptosis in vitro. Meanwhile, upregulated miR-137 remarkably inhibited migration and invasion of pancreatic cancer cells. Further studies indicated that MRGBP was identified as the direct downstream target gene of miR-137. In addition, MRGBP expression is significantly downregulated in miR-137-transfected cells. Our previous study revealed that silencing of MRGBP suppressed the growth of PanCa cells in vitro and in vivo and also promoted apoptosis, and inhibited migration and invasion of PanCa cells, which are consistent with the effects of miR-137 overexpression. Taken together, our findings suggest that miR-137 may function as a novel tumor promoter through directly targeting MRGBP in PanCa.
Collapse
Affiliation(s)
- Feng Ding
- Department of Gastroenterology/Hepatology, ZhongNan Hospital of Wuhan University, Wuhan, China
- The Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan, China
| | - Shuang Zhang
- Laboratory of Clinical Immunology, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoyang Gao
- Department of Pathology, Hubei Cancer Hospital, Wuhan, China
| | - Jian Shang
- Department of Gastroenterology/Hepatology, ZhongNan Hospital of Wuhan University, Wuhan, China
- The Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan, China
| | - Yanxia Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ning Cui
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiu Zhao
- Department of Gastroenterology/Hepatology, ZhongNan Hospital of Wuhan University, Wuhan, China
- The Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan, China
| |
Collapse
|
112
|
Qiao L, Zheng J, Tian Y, Zhang Q, Wang X, Chen JJ, Zhang W. Regulator of chromatin condensation 1 abrogates the G1 cell cycle checkpoint via Cdk1 in human papillomavirus E7-expressing epithelium and cervical cancer cells. Cell Death Dis 2018; 9:583. [PMID: 29789527 PMCID: PMC5964113 DOI: 10.1038/s41419-018-0584-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/31/2018] [Accepted: 04/06/2018] [Indexed: 01/01/2023]
Abstract
Regulator of chromatin condensation 1 (RCC1) is a major guanine-nucleotide exchange factor for Ran GTPase and plays key roles in nucleo-cytoplasmic transport, mitosis, and nuclear envelope assembly. RCC1 is known to be a critical cell cycle regulator whose loss causes G1 phase arrest, but the molecular basis for this regulation is poorly understood. Furthermore, little is known about the relationship between RCC1 and carcinomas. Human papillomavirus (HPV) infection is highly associated with the development of cervical cancer. The expression and function of RCC1 in HPV-related cervical cancer and cell cycle regulation have not yet been explored. In this study, we first observed that RCC1 immunostaining was mildly increased in cervical cancer tissues and significantly upregulated in HPV E7-expressing cells; this localization was primarily nuclear. We showed that the transcription factor c-Jun transcriptionally upregulates RCC1 via a direct interaction with the RCC1 promoter. Moreover, siRNA-mediated knockdown of RCC1 inhibited G1/S cell cycle progression and DNA synthesis, while overexpression of RCC1 abrogated the G1 checkpoint. RCC1 knockdown downregulated the protein levels of the transcription factor E2F1, especially nuclear E2F1, by promoting its degradation in HPV E7-expressing cells. Overexpression of E2F1 rescued RCC1 knockdown-mediated inhibition of G1/S progression. Additionally, we showed that cyclin-dependent kinase 1 (Cdk1), a known target of E2F1, is involved in G1 checkpoint regulation, as Cdk1 knockdown hindered G1/S progression, while Cdk1 overexpression rescued RCC1 knockdown-mediated effect on G1 cell cycle progression. Furthermore, RCC1 knockdown reduced HPV E7 protein levels, which may in turn downregulate E2F1. Our study explores the function of RCC1 in G1/S cell cycle progression and suggests that RCC1 may be involved in HPV E7-mediated genomic instability.
Collapse
Affiliation(s)
- Lijun Qiao
- Cancer Research Center and Department of Microbiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Jingyi Zheng
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Yonghao Tian
- Department of Orthopedic Surgery, Qilu Hospital Affiliated Shandong University, Jinan, Shandong, China
| | - Qishu Zhang
- Cancer Research Center and Department of Microbiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xiao Wang
- Institute of Pathobiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Jason J Chen
- Cancer Research Center and Department of Microbiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Weifang Zhang
- Department of Microbiology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
113
|
Hu J, Liu J, Chen A, Lyu J, Ai G, Zeng Q, Sun Y, Chen C, Wang J, Qiu J, Wu Y, Cheng J, Shi X, Song L. Ino80 promotes cervical cancer tumorigenesis by activating Nanog expression. Oncotarget 2018; 7:72250-72262. [PMID: 27750218 PMCID: PMC5342159 DOI: 10.18632/oncotarget.12667] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/10/2016] [Indexed: 02/03/2023] Open
Abstract
Ino80 ATPase is an integral component of the INO80 ATP-dependent chromatin-remodeling complex, which regulates transcription, DNA repair and replication. We found that Ino80 was highly expressed in cervical cancer cell lines and tumor samples. Ino80 knockdown inhibited cervical cancer cell proliferation, induced G0/G1 phase cell cycle arrest in vitro and suppressed tumor growth in vivo. However, Ino80 knockdown did not affect cell apoptosis, migration or invasion in vitro. Ino80 overexpression promoted proliferation in the H8 immortalized cervical epithelial cell line, which has low endogenous Ino80 expression as compared to cervical cancer cell lines. Ino80 bound to the Nanog transcription start site (TSS) and enhanced its expression in cervical cancer cells. Nanog overexpression in Ino80 knockdown cell lines promoted cell proliferation. This study demonstrated for the first time that Ino80 was upregulated in cervical cancer and promoted cell proliferation and tumorigenesis. Our findings suggest that Ino80 may be a potential therapeutic target for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Jing Hu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Liu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Aozheng Chen
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia Lyu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guihai Ai
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiongjing Zeng
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Sun
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunxia Chen
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinbo Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin Qiu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Wu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiajing Cheng
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiujuan Shi
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liwen Song
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
114
|
SRSF10-mediated IL1RAP alternative splicing regulates cervical cancer oncogenesis via mIL1RAP-NF-κB-CD47 axis. Oncogene 2018; 37:2394-2409. [PMID: 29429992 PMCID: PMC5931977 DOI: 10.1038/s41388-017-0119-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/06/2017] [Accepted: 12/14/2017] [Indexed: 02/05/2023]
Abstract
High-risk human papillomavirus oncoproteins E6 and E7 are the major etiological factors of cervical cancer but are insufficient for malignant transformation of cervical cancer. Dysregulated alternative splicing, mainly ascribed to aberrant splicing factor levels and activities, contributes to most cancer hallmarks. However, do E6 and E7 regulate the expression of splicing factors? Does alternative splicing acts as an “accomplice” of E6E7 to promote cervical cancer progression? Here, we identified that the splicing factor SRSF10, which promotes tumorigenesis of cervix, was upregulated by E6E7 via E2F1 transcriptional activation. SRSF10 modulates the alternate terminator of interleukin-1 receptor accessory protein exon 13 to increase production of the membrane form of interleukin-1 receptor accessory protein. SRSF10-mediated mIL1RAP upregulates the expression of the “don’t eat me” signal CD47 to inhibit macrophage phagocytosis by promoting nuclear factor-κB activation, which is pivotal in inflammatory, immune, and tumorigenesis processes. Altogether, these data reveal a close relationship among HPV infection, alternative splicing and tumor immune evasion, and also suggests that the SRSF10-mIL1RAP-CD47 axis could be an attractive therapeutic target for the treatment of cervical cancer.
Collapse
|
115
|
Tian R, Li X, Gao Y, Li Y, Yang P, Wang K. Identification and validation of the role of matrix metalloproteinase-1 in cervical cancer. Int J Oncol 2018; 52:1198-1208. [PMID: 29436615 PMCID: PMC5843389 DOI: 10.3892/ijo.2018.4267] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022] Open
Abstract
Lymph node (LN) metastasis at an early stage of cervical cancer is often an indicator of poor prognosis and is critical for subsequent adjuvant therapy. The current study aimed to identify aberrant gene signatures and biomarkers of metastasis for patients with cervical cancer. RNA-sequencing data of 132 LN negative (N0) and 60 LN positive (N1) cervical cancer samples obtained from The Cancer Genome Atlas database were analyzed. Differentially expressed genes were identified using R packages 'edgeR' and 'limma'. Kyoto Encyclopedia of Genes and Genomes pathway enrichment and Gene Set Enrichment Analysis (GSEA) were conducted. The GSE9750 dataset obtained from Gene Expression Omnibus was analyzed to identify genes that are persistently aberrantly expressed during the development of cervical cancer. The peroxisome proliferator-activated receptor (PPAR) signaling pathway was screened out to be significant during LN metastasis. In the two analyzed datasets, 11 genes were aberrantly expressed, while matrix metalloproteinase 1 (MMP1) was the only gene that was persistently overexpressed. Cell viability, wound healing and Transwell assays were performed to evaluate the effects of MMP1 knockdown in cervical cancer cell lines, and the expression of epithelial mesenchymal transition (EMT) markers was detected. Finally, the clinical significance of MMP1 was investigated. The current study identified that MMP1 was overexpressed and the PPAR signaling pathway was associated LN metastasis in patients with cervical cancer. Following knockdown of MMP1, the proliferation, migration and invasion of cervical cancer cell lines were weakened, the expression of epithelial marker E-cadherin was increased, and the expression of metastasis-associated gene vimentin was decreased. MMP1 was an independent prognostic factor for cervical cancer. The current study indicated that MMP1 has a key role in the regulation of cervical tumor growth and LN metastasis via EMT to a certain extent. The results suggest that MMP1 may be a biomarker for LN metastasis of cervical cancer, and further validation should be performed.
Collapse
Affiliation(s)
- Run Tian
- Department of Orthopedics, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xiaofang Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yan'e Gao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yue Li
- Department of Orthopedics, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Pei Yang
- Department of Orthopedics, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Kunzheng Wang
- Department of Orthopedics, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
116
|
Wu DM, Shi J, Liu T, Deng SH, Han R, Xu Y. Integrated analysis reveals down-regulation of SPARCL1 is correlated with cervical cancer development and progression. Cancer Biomark 2018; 21:355-365. [DOI: 10.3233/cbm-170501] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
117
|
da Silva-Junior IA, Dalmaso B, Herbster S, Lepique AP, Jancar S. Platelet-Activating Factor Receptor Ligands Protect Tumor Cells from Radiation-Induced Cell Death. Front Oncol 2018; 8:10. [PMID: 29459885 PMCID: PMC5807395 DOI: 10.3389/fonc.2018.00010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/15/2018] [Indexed: 12/17/2022] Open
Abstract
Irradiation generates oxidized phospholipids that activate platelet-activating factor receptor (PAFR) associated with pro-tumorigenic effects. Here, we investigated the involvement of PAFR in tumor cell survival after irradiation. Cervical cancer samples presented higher levels of PAF-receptor gene (PTAFR) when compared with normal cervical tissue. In cervical cancer patients submitted to radiotherapy (RT), the expression of PTAFR was significantly increased. Cervical cancer-derived cell lines (C33, SiHa, and HeLa) and squamous carcinoma cell lines (SCC90 and SCC78) express higher levels of PAFR mRNA and protein than immortalized keratinocytes. Gamma radiation increased PAFR expression and induced PAFR ligands and prostaglandin E2 (PGE2) in these tumor cells. The blocking of PAFR with the antagonist CV3938 before irradiation inhibited PGE2 and increased tumor cells death. Similarly, human carcinoma cells transfected with PAFR (KBP) were more resistant to radiation compared to those lacking the receptor (KBM). PGE2 production by irradiated KBP cells was also inhibited by CV3988. These results show that irradiation of carcinoma cells generates PAFR ligands that protect tumor cells from death and suggests that the combination of RT with a PAFR antagonist could be a promising strategy for cancer treatment.
Collapse
Affiliation(s)
| | - Barbara Dalmaso
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Suellen Herbster
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Paula Lepique
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Sonia Jancar
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
118
|
Liu Q, Wang J, Tang M, Chen L, Qi X, Li J, Yu J, Qiu H, Wang Y. The overexpression of PXN promotes tumor progression and leads to radioresistance in cervical cancer. Future Oncol 2018; 14:241-253. [PMID: 29318915 DOI: 10.2217/fon-2017-0474] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM We aim to investigate the functions of PXN in cervical cancer. MATERIALS & METHODS PXN protein was investigated by immunohistochemistry in a panel of cervical cancer. A series of in vitro and in vivo assays were used to explore the efficacy of PXN. RESULTS PXN was significantly upregulated in cervical cancer, which associated with tumor stage, poor differentiation, lymphovascular space invasion and lymphatic metastasis. Knockdown of PXN notably impaired cellular growth and colony formation by suppressing Bcl-2 and inducing marked apoptosis. Moreover, PXN led to resistance to radiation, and downregulation of PXN resensitized C33A cells to radiation. CONCLUSION PXN was frequently upregulated and acted as an oncogene via regulating Bcl-2 in cervical cancer, which supports PXN as a potent therapeutic target.
Collapse
Affiliation(s)
- Qiuli Liu
- Department of Obstetrics & Gynecology, The Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| | - Jing Wang
- Department of Obstetrics & Gynecology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai 264000, China
| | - Mei Tang
- Department of Obstetrics & Gynecology, The Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| | - Ling Chen
- Department of Obstetrics & Gynecology, The Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| | - Xiaowei Qi
- Department of Pathology, The Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| | - Juan Li
- Department of Obstetrics and Gynecology, Wuxi Hospital For Maternal & Child Health Care, Wuxi 214062, China
| | - Jinjin Yu
- Department of Obstetrics & Gynecology, The Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| | - Haifeng Qiu
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Yuan Wang
- Department of Obstetrics & Gynecology, The Affiliated Hospital of Jiangnan University, Wuxi 214062, China
| |
Collapse
|
119
|
Cantini L, Calzone L, Martignetti L, Rydenfelt M, Blüthgen N, Barillot E, Zinovyev A. Classification of gene signatures for their information value and functional redundancy. NPJ Syst Biol Appl 2017; 4:2. [PMID: 29263798 PMCID: PMC5736638 DOI: 10.1038/s41540-017-0038-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 11/15/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022] Open
Abstract
Gene signatures are more and more used to interpret results of omics data analyses but suffer from compositional (large overlap) and functional (correlated read-outs) redundancy. Moreover, many gene signatures rarely come out as significant in statistical tests. Based on pan-cancer data analysis, we construct a restricted set of 962 signatures defined as informative and demonstrate that they have a higher probability to appear enriched in comparative cancer studies. We show that the majority of informative signatures conserve their weights for the genes composing the signature (eigengenes) from one cancer type to another. We finally construct InfoSigMap, an interactive online map of these signatures and their cross-correlations. This map highlights the structure of compositional and functional redundancies between informative signatures, and it charts the territories of biological functions. InfoSigMap can be used to visualize the results of omics data analyses and suggests a rearrangement of existing gene sets. An informative collection of gene signatures for transcriptomic data analysis is constructed. The number of transcriptomic signatures grows fast and their collections are highly redundant that hampers omics data analyses interpretation. A computational biology team from Institut Curie led by Andrei Zinovyev selected a collection of 962 gene signatures shown to be informative for cancer studies and reflecting mechanisms of cancer progression. The signatures were filtered from a large compendium without requiring any manual curation by experts through a large-scale unbiased analysis of pancancer data. They have much higher chance to obtain significant enrichment scores in a comparative trancriptomic study. The authors integrated the 962 signatures into InfoSigMap, a new data visualization resource for the interpretation of the results of omics data analyses, which facilitates getting an insight into the mechanisms driving cancer.
Collapse
Affiliation(s)
- Laura Cantini
- Institut Curie, PSL Research University, INSERM U900, Mines ParisTech, 26, rue d'Ulm, F-75248 Paris, France
| | - Laurence Calzone
- Institut Curie, PSL Research University, INSERM U900, Mines ParisTech, 26, rue d'Ulm, F-75248 Paris, France
| | - Loredana Martignetti
- Institut Curie, PSL Research University, INSERM U900, Mines ParisTech, 26, rue d'Ulm, F-75248 Paris, France
| | - Mattias Rydenfelt
- Institute of Pathology, Charite Universitätsmedizin Berlin, Chariteplatz 1, 10117 Berlin, Germany.,IRI Life Sciences and Institute for Theoretical Biology, Humboldt University, Philippstr. 13, Haus 18, 10115 Berlin, Germany
| | - Nils Blüthgen
- Institute of Pathology, Charite Universitätsmedizin Berlin, Chariteplatz 1, 10117 Berlin, Germany.,IRI Life Sciences and Institute for Theoretical Biology, Humboldt University, Philippstr. 13, Haus 18, 10115 Berlin, Germany
| | - Emmanuel Barillot
- Institut Curie, PSL Research University, INSERM U900, Mines ParisTech, 26, rue d'Ulm, F-75248 Paris, France
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, INSERM U900, Mines ParisTech, 26, rue d'Ulm, F-75248 Paris, France
| |
Collapse
|
120
|
Bergqvist C, Kadara H, Hamie L, Nemer G, Safi R, Karouni M, Marrouche N, Abbas O, Hasbani DJ, Kibbi AG, Nassar D, Shimomura Y, Kurban M. SLURP-1is mutated in Mal de Meleda, a potential molecular signature for melanoma and a putative squamous lineage tumor suppressor gene. Int J Dermatol 2017; 57:162-170. [DOI: 10.1111/ijd.13850] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 11/02/2017] [Indexed: 02/01/2023]
Affiliation(s)
| | - Humam Kadara
- Department of Biochemistry and Molecular Genetics; American University of Beirut; Beirut Lebanon
| | - Lamiaa Hamie
- Department of Internal Medicine; American University of Beirut; Beirut Lebanon
| | - Georges Nemer
- Department of Biochemistry and Molecular Genetics; American University of Beirut; Beirut Lebanon
| | - Remi Safi
- Department of Dermatology; American University of Beirut; Beirut Lebanon
| | - Mirna Karouni
- Department of Dermatology; American University of Beirut; Beirut Lebanon
| | - Nadine Marrouche
- Department of Dermatology; Norfolk and Norwich University; Norwich UK
| | - Ossama Abbas
- Department of Dermatology; American University of Beirut; Beirut Lebanon
| | | | - Abdul G. Kibbi
- Department of Dermatology; American University of Beirut; Beirut Lebanon
| | - Dany Nassar
- Department of Dermatology; American University of Beirut; Beirut Lebanon
| | - Yutaka Shimomura
- Division of Dermatology; Niigata University Graduate School of Medical and Dental Sciences; Niigata Japan
| | - Mazen Kurban
- Department of Dermatology; American University of Beirut; Beirut Lebanon
- Department of Biochemistry and Molecular Genetics; American University of Beirut; Beirut Lebanon
- Department of Dermatology; Columbia University; New York NY USA
| |
Collapse
|
121
|
Long NP, Jung KH, Yoon SJ, Anh NH, Nghi TD, Kang YP, Yan HH, Min JE, Hong SS, Kwon SW. Systematic assessment of cervical cancer initiation and progression uncovers genetic panels for deep learning-based early diagnosis and proposes novel diagnostic and prognostic biomarkers. Oncotarget 2017; 8:109436-109456. [PMID: 29312619 PMCID: PMC5752532 DOI: 10.18632/oncotarget.22689] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/27/2017] [Indexed: 12/18/2022] Open
Abstract
Although many outstanding achievements in the management of cervical cancer (CxCa) have obtained, it still imposes a major burden which has prompted scientists to discover and validate new CxCa biomarkers to improve the diagnostic and prognostic assessment of CxCa. In this study, eight different gene expression data sets containing 202 cancer, 115 cervical intraepithelial neoplasia (CIN), and 105 normal samples were utilized for an integrative systems biology assessment in a multi-stage carcinogenesis manner. Deep learning-based diagnostic models were established based on the genetic panels of intrinsic genes of cervical carcinogenesis as well as on the unbiased variable selection approach. Survival analysis was also conducted to explore the potential biomarker candidates for prognostic assessment. Our results showed that cell cycle, RNA transport, mRNA surveillance, and one carbon pool by folate were the key regulatory mechanisms involved in the initiation, progression, and metastasis of CxCa. Various genetic panels combined with machine learning algorithms successfully differentiated CxCa from CIN and normalcy in cross-study normalized data sets. In particular, the 168-gene deep learning model for the differentiation of cancer from normalcy achieved an externally validated accuracy of 97.96% (99.01% sensitivity and 95.65% specificity). Survival analysis revealed that ZNF281 and EPHB6 were the two most promising prognostic genetic markers for CxCa among others. Our findings open new opportunities to enhance current understanding of the characteristics of CxCa pathobiology. In addition, the combination of transcriptomics-based signatures and deep learning classification may become an important approach to improve CxCa diagnosis and management in clinical practice.
Collapse
Affiliation(s)
| | - Kyung Hee Jung
- Department of Drug Development, College of Medicine, Inha University, Incheon 22212, Korea
| | - Sang Jun Yoon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Nguyen Hoang Anh
- School of Medicine, Vietnam National University, Ho Chi Minh 70000, Vietnam
| | - Tran Diem Nghi
- School of Medicine, Vietnam National University, Ho Chi Minh 70000, Vietnam
| | - Yun Pyo Kang
- College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Hong Hua Yan
- Department of Drug Development, College of Medicine, Inha University, Incheon 22212, Korea
| | - Jung Eun Min
- College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Soon-Sun Hong
- Department of Drug Development, College of Medicine, Inha University, Incheon 22212, Korea
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
122
|
Luo L, McGarvey P, Madhavan S, Kumar R, Gusev Y, Upadhyay G. Distinct lymphocyte antigens 6 (Ly6) family members Ly6D, Ly6E, Ly6K and Ly6H drive tumorigenesis and clinical outcome. Oncotarget 2017; 7:11165-93. [PMID: 26862846 PMCID: PMC4905465 DOI: 10.18632/oncotarget.7163] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/23/2016] [Indexed: 12/21/2022] Open
Abstract
Stem cell antigen-1 (Sca-1) is used to isolate and characterize tumor initiating cell populations from tumors of various murine models [1]. Sca-1 induced disruption of TGF-β signaling is required in vivo tumorigenesis in breast cancer models [2, 3-5]. The role of human Ly6 gene family is only beginning to be appreciated in recent literature [6-9]. To study the significance of Ly6 gene family members, we have visualized one hundred thirty gene expression omnibus (GEO) dataset using Oncomine (Invitrogen) and Georgetown Database of Cancer (G-DOC). This analysis showed that four different members Ly6D, Ly6E, Ly6H or Ly6K have increased gene expressed in bladder, brain and CNS, breast, colorectal, cervical, ovarian, lung, head and neck, pancreatic and prostate cancer than their normal counter part tissues. Increased expression of Ly6D, Ly6E, Ly6H or Ly6K was observed in sub-set of cancer type. The increased expression of Ly6D, Ly6E, Ly6H and Ly6K was found to be associated with poor outcome in ovarian, colorectal, gastric, breast, lung, bladder or brain and CNS as observed by KM plotter and PROGgeneV2 platform. The remarkable findings of increased expression of Ly6 family members and its positive correlation with poor outcome on patient survival in multiple cancer type indicate that Ly6 family members Ly6D, Ly6E, Ly6K and Ly6H will be an important targets in clinical practice as marker of poor prognosis and for developing novel therapeutics in multiple cancer type.
Collapse
Affiliation(s)
- Linlin Luo
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, District of Columbia 20007, United States of America.,Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20007, United States of America
| | - Peter McGarvey
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, District of Columbia 20007, United States of America.,Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20007, United States of America
| | - Subha Madhavan
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, District of Columbia 20007, United States of America.,Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20007, United States of America
| | - Rakesh Kumar
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, District of Columbia 20037, United States of America
| | - Yuriy Gusev
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, District of Columbia 20007, United States of America.,Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20007, United States of America
| | - Geeta Upadhyay
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, District of Columbia 20007, United States of America.,Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20007, United States of America
| |
Collapse
|
123
|
Yoshihara M, Hayashizaki Y, Murakawa Y. Genomic Instability of iPSCs: Challenges Towards Their Clinical Applications. Stem Cell Rev Rep 2017; 13:7-16. [PMID: 27592701 PMCID: PMC5346115 DOI: 10.1007/s12015-016-9680-6] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are a type of pluripotent stem cells generated directly from mature cells through the introduction of key transcription factors. iPSCs can be propagated and differentiated into many cell types in the human body, holding enormous potential in the field of regenerative medicine. However, genomic instability of iPSCs has been reported with the advent of high-throughput technologies such as next-generation sequencing. The presence of genetic variations in iPSCs has raised serious safety concerns, hampering the advancement of iPSC-based novel therapies. Here we summarize our current knowledge on genomic instability of iPSCs, with a particular focus on types of genetic variations and their origins. Importantly, it remains elusive whether genetic variations in iPSCs can be an actual risk factor for adverse effects including malignant outgrowth. Furthermore, we discuss novel approaches to generate iPSCs with fewer genetic variations. Lastly, we outline the safety issues and monitoring strategies of iPSCs in clinical settings.
Collapse
Affiliation(s)
- Masahito Yoshihara
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa, Japan.,Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | - Yasuhiro Murakawa
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa, Japan. .,RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Saitama, Japan.
| |
Collapse
|
124
|
Wei Y, Dong J, Li F, Wei Z, Tian Y. Knockdown of SLC39A7 suppresses cell proliferation, migration and invasion in cervical cancer. EXCLI JOURNAL 2017; 16:1165-1176. [PMID: 29285013 PMCID: PMC5735344 DOI: 10.17179/excli2017-690] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/16/2017] [Indexed: 11/10/2022]
Abstract
Cervical cancer is the fourth leading cause of malignancy related mortality in women worldwide. SLC39A7 (ZIP7) is a zinc transporter that plays a key role in intestinal epithelial self-renewal. However, whether or not SLC39A7 is involved in human cervical cancer remains unclear. In this study, we investigated the effects of SLC39A7 in cervical cancer in vitro and elucidate related underlying mechanisms. Using Oncomine data analysis, we first found SLC39A7 is commonly upregulated in cervical cancer tissues in comparison with corresponding normal controls. The in vitro experiments indicated that silencing of SLC39A7 expression resulted in decreased cell proliferation, increased cell apoptosis, and attenuated migratory and invasive ability using CCK-8, colony formation, flow cytometry, transwell assays, respectively in cervical cancer cell lines, HeLa and ME-180 cells. In molecular levels, Western blot further demonstrated that silencing of SLC39A7 significantly upregulated the expression of Bax and E-cadherin, downregulated the expression of Bcl-2 and MMP-2 in both HeLa and ME-180 cells. These findings provide evidence that SLC39A7 plays a positive role in the progression of cervical cancer and its knockdown might be as a potential therapeutic target for cervical cancer treatment.
Collapse
Affiliation(s)
- Yongqing Wei
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong University, Jinan 250012, China
| | - Jie Dong
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong University, Jinan 250012, China
| | - Fuli Li
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong University, Jinan 250012, China
| | - Zhuqing Wei
- Department of Stomatology, Chengyang People's Hospital, Qingdao 266109, China
| | - Yuling Tian
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong University, Jinan 250012, China
| |
Collapse
|
125
|
High-Risk Alphapapillomavirus Oncogenes Impair the Homologous Recombination Pathway. J Virol 2017; 91:JVI.01084-17. [PMID: 28768872 DOI: 10.1128/jvi.01084-17] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/28/2017] [Indexed: 01/25/2023] Open
Abstract
Persistent high-risk genus human Alphapapillomavirus (HPV) infections cause nearly every cervical carcinoma and a subset of tumors in the oropharyngeal tract. During the decades required for HPV-associated tumorigenesis, the cellular genome becomes significantly destabilized. Our analysis of cervical tumors from four separate data sets found a significant upregulation of the homologous-recombination (HR) pathway genes. The increased abundance of HR proteins can be replicated in primary cells by expression of the two HPV oncogenes (E6 and E7) required for HPV-associated transformation. HPV E6 and E7 also enhanced the ability of HR proteins to form repair foci, and yet both E6 and E7 reduce the ability of the HR pathway to complete double-strand break (DSB) repair by about 50%. The HPV oncogenes hinder HR by allowing the process to begin at points in the cell cycle when the lack of a sister chromatid to serve as a homologous template prevents completion of the repair. Further, HPV E6 attenuates repair by causing RAD51 to be mislocalized away from both transient and persistent DSBs, whereas HPV E7 is only capable of impairing RAD51 localization to transient lesions. Finally, we show that the inability to robustly repair DSBs causes some of these lesions to be more persistent, a phenotype that correlates with increased integration of episomal DNA. Together, these data support our hypothesis that HPV oncogenes contribute to the genomic instability observed in HPV-associated malignancies by attenuating the repair of damaged DNA.IMPORTANCE This study expands the understanding of HPV biology, establishing a direct role for both HPV E6 and E7 in the destabilization of the host genome by blocking the homologous repair of DSBs. To our knowledge, this is the first time that both viral oncogenes were shown to disrupt this DSB repair pathway. We show that HPV E6 and E7 allow HR to initiate at an inappropriate part of the cell cycle. The mislocalization of RAD51 away from DSBs in cells expressing HPV E6 and E7 hinders HR through a distinct mechanism. These observations have broad implications. The impairment of HR by HPV oncogenes may be targeted for treatment of HPV+ malignancies. Further, this attenuation of repair suggests HPV oncogenes may contribute to tumorigenesis by promoting the integration of the HPV genome, a common feature of HPV-transformed cells. Our data support this idea since HPV E6 stimulates the integration of episomes.
Collapse
|
126
|
Marzook H, Deivendran S, George B, Reshmi G, Santhoshkumar TR, Kumar R, Pillai MR. Cytoplasmic translocation of MTA1 coregulator promotes de-repression of SGK1 transcription in hypoxic cancer cells. Oncogene 2017; 36:5263-5273. [PMID: 28504714 DOI: 10.1038/onc.2017.19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 12/16/2016] [Accepted: 01/05/2017] [Indexed: 12/17/2022]
Abstract
Chromatin remodeling factor metastatic tumor protein 1 (MTA1), one of the most upregulated oncogene in human cancer, has an important role in gene expression, cell survival and promoting hypoxic response. Successful cancer progression is dependent on the ability of cells to utilize its survival pathways for adapting to hypoxic microenvironment. Although MTA1 is a stress-responsive gene, but whether hypoxia modulates its function and its role in engaging other core stress-responsive survival pathway(s) remains unknown. Here we have discovered that MTA1 is a novel corepressor of serum and glucocorticoid-inducible kinase 1 (SGK1). Surprisingly, this regulatory corepressive function of MTA1 is lost under hypoxia, allowing upregulation of SGK1 expression and engaging the MTA1-SGK1 axis for the benefit of the cell survival. The underlying mechanism of the noticed stimulation of SGK1 expression by hypoxia includes de-repression of SGK1 transcription because of hypoxia-triggered nucleus-to-cytoplasmic translocation of MTA1. In addition, the newly recognized cytoplasmic translocation of MTA1 was dependent on the chaperoning function of heat shock protein 90 (HSP90) and co-accompanied by the formation of MTA1, HSP90 and HIF1α complex under hypoxic condition but not under normoxic condition. Hypoxia-triggered redistribution of MTA1, SGK1 upregulation and cell survival functions were compromised by a pharmacological SGK1 inhibitor. In summary, for the first time, we report MTA1 regulation of SGK1 expression, hypoxia-dependent MTA1 translocation to the cytoplasm and de-repression of SGK1 transcription. These findings illustrate how cancer cells utilize a chromatin remodeling factor to engage a core survival pathway to support its cancerous phenotypes, and reveal new facets of MTA1-SGK1 axis by a physiologic signal in cancer progression.
Collapse
Affiliation(s)
- H Marzook
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - S Deivendran
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - B George
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - G Reshmi
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - T R Santhoshkumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - R Kumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| | - M R Pillai
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
127
|
Turinetto V, Orlando L, Giachino C. Induced Pluripotent Stem Cells: Advances in the Quest for Genetic Stability during Reprogramming Process. Int J Mol Sci 2017; 18:E1952. [PMID: 28902128 PMCID: PMC5618601 DOI: 10.3390/ijms18091952] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/01/2017] [Accepted: 09/08/2017] [Indexed: 12/19/2022] Open
Abstract
Evaluation of the extent and nature of induced pluripotent stem cell (iPSC) genetic instability is important for both basic research and future clinical use. As previously demonstrated regarding embryonic stem cells, such DNA aberrations might affect the differentiation capacity of the cells and increase their tumorigenicity. Here, we first focus on the contribution of multiple DNA damage response pathways during cellular reprogramming. We then discuss the origin and mechanisms responsible for the modification of genetic material in iPSCs (pre-existing variations in somatic cells, mutations induced by reprogramming factors, and mutations induced by culture expansion) and deepen the possible functional consequences of genetic variations in these cells. Lastly, we present some recent improvements of iPSC generation methods aimed at obtaining cells with fewer genetic variations.
Collapse
Affiliation(s)
- Valentina Turinetto
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy.
| | - Luca Orlando
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada.
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy.
| |
Collapse
|
128
|
Liao SC, Li JX, Yu L, Sun SR. Protein tyrosine phosphatase 1B expression contributes to the development of breast cancer. J Zhejiang Univ Sci B 2017; 18:334-342. [PMID: 28378571 DOI: 10.1631/jzus.b1600184] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The protein tyrosine phosphatase 1B (PTP1B) is an important regulator of metabolism. The relationship between PTP1B and tumors is quite complex. The purpose of this study is to explore the expression pattern and role of PTP1B in breast cancer. The expression of PTP1B was detected in 67 samples of breast cancer tissue by Western blot. Cell growth assay, Transwell migration assay, and Scratch motility assay were used to examine the proliferation and migration of MCF-7 with and without PTP1B. The total levels and phosphorylated levels of signal transduction and activator of transcription 3 (STAT3) and the expression of C-C motif chemokine ligand 5 (CCL5) were also examined by Western blot. PTP1B was overexpressed in over 70% of breast cancer tissues, correlating with patients with estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and human epidermal growth factor receptor 2 (HER2)-positive tumors. The data also showed that both tumor size and lymph node metastasis were significantly higher in patients with a higher level of PTP1B. The proliferation and migration of MCF-7 cells were found to be inhibited after knocking down the gene of PTP1B. Our data also showed that PTP1B could up-regulate the dephosphorylated level of STAT3, which could increase the expression of CCL5. These phenomena indicated that PTP1B may play a crucial role in the development of breast cancer.
Collapse
Affiliation(s)
- Shi-Chong Liao
- Department of Thyroid and Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jin-Xin Li
- Department of Teaching Administration, Wuhan University School of Medicine, Wuhan 430071, China
| | - Li Yu
- Intensive Care Unit, the Central Hospital of Wuhan, Wuhan 430014, China
| | - Sheng-Rong Sun
- Department of Thyroid and Breast Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
129
|
Uchiyama N, Dougan DR, Lawson JD, Kimura H, Matsumoto SI, Tanaka Y, Kawamoto T. Identification of AHCY inhibitors using novel high-throughput mass spectrometry. Biochem Biophys Res Commun 2017; 491:1-7. [DOI: 10.1016/j.bbrc.2017.05.107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 05/19/2017] [Indexed: 10/19/2022]
|
130
|
Li D, Lin C, Chen M, Li N, Du Y, Su C, Yang C, Gong N, Wu H, Wu R, Jain A, Zhang Y, Li X. Comprehensive bioinformatics analysis of the characterization and determination underlying mechanisms of over-expression and co-expression of genes residing on 20q in colorectal cancer. Oncotarget 2017; 8:78642-78659. [PMID: 29108255 PMCID: PMC5667988 DOI: 10.18632/oncotarget.20204] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/19/2017] [Indexed: 12/20/2022] Open
Abstract
The Long arm of chromosome 20 (20q) is closely related to the development of colorectal cancer, so identifying the expression profile of genes on 20q through a comprehensive overview is indispensable. In this article, preliminar experimental data, several available databases and bioinformatics tools such as the Cancer Genome Atlas, the Encyclopedia of DNA Elements, the JASPAR database and starBase were combined to analyze the correlation between genes and chromosomal aberrations, microRNA and transcription factors, as well as to explore the expression feature and potential regulative mechanism. The results showed that the most frequently unregulated genes in colorectal cancer arelocated on chromosome 20q, present a significant CNA–mRNA correlation.Furthermore, the genes with mRNA overexpression showed co-expression features and tended to be clustered within the same genomic neighborhoods. Then, several genes were selected to carry out further analysis and demonstrated that shared transcription factors, a conserved bidirectional promoter, and competition for a limited pool of microRNAin the 3’UTR of mRNA may be the underlying mechanisms behind the co-expression of physically adjacent genes.Finally, the databases, Lentivirus shRNA, and qPCR were used to find that these adjacent genes with co-expression cooperatively participated in the same biological pathways associated with the pathogenesis and development of colorectal cancer.
Collapse
Affiliation(s)
- Daojiang Li
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Changwei Lin
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Miao Chen
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Nanpeng Li
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Yuheng Du
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Chen Su
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Chunxing Yang
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Ni Gong
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Hao Wu
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Runliu Wu
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Arad Jain
- College of Arts and Science, University of Virginia, Charlottesville, Virginia 22904, The United States of America
| | - Yi Zhang
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China.,Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Xiaorong Li
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China.,Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
131
|
Ding F, Zhang S, Gao S, Shang J, Li Y, Cui N, Zhao Q. MRGBP as a potential biomarker for the malignancy of pancreatic ductal adenocarcinoma. Oncotarget 2017; 8:64224-64236. [PMID: 28969065 PMCID: PMC5609997 DOI: 10.18632/oncotarget.19451] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/12/2017] [Indexed: 12/30/2022] Open
Abstract
MORF4-related gene-binding protein (MRGBP), which is also known as chromosome 20 open reading frame 20 (C20orf20), is commonly highly expressed in several types of malignant tumors and tumor progression. However, the expression pattern and underlying mechanism of MRGBP in pancreatic ductal adenocarcinoma (PDAC) remain unknown. In the study, we found that MRGBP was frequently upregulated in PDAC tissues and cell lines. In addition, the upregulation of MRGBP was positively associated with TNM stage, T classification, and poor prognosis. Knockdown of MRGBP in the PDAC cell lines ASPC-1 and Mia PaCa-2 by transiently transfected with small interfering RNA (siRNA) drastically attenuated the proliferation, migration, and invasion of those cells, whereas ectopic MRGBP overexpression in BxPC-3 cells produced exactly the opposite effect. Furthermore, we also found that overexpression of MRGBP remarkably led to cell morphological changes and induced an increased expression of mesenchymal marker Vimentin, whereas a decreased expression of epithelial marker E-cadherin. Taken together, this study indicates that MRGBP acts as a tumor oncogene in PDAC and is a promising target of carcinogenesis.
Collapse
Affiliation(s)
- Feng Ding
- Department of Gastroenterology/Hepatology, ZhongNan Hospital of Wuhan University, Wuhan 430071, China.,The Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Shuang Zhang
- Laboratory of Clinical Immunology, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shaoyang Gao
- Department of Pathology, Hubei Cancer Hospital, Wuhan 430079, China
| | - Jian Shang
- Department of Gastroenterology/Hepatology, ZhongNan Hospital of Wuhan University, Wuhan 430071, China.,The Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan 430071, China
| | - Yanxia Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ning Cui
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qiu Zhao
- Department of Gastroenterology/Hepatology, ZhongNan Hospital of Wuhan University, Wuhan 430071, China.,The Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan 430071, China
| |
Collapse
|
132
|
Gov E, Arga KY. Differential co-expression analysis reveals a novel prognostic gene module in ovarian cancer. Sci Rep 2017; 7:4996. [PMID: 28694494 PMCID: PMC5504034 DOI: 10.1038/s41598-017-05298-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 05/26/2017] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is one of the most significant disease among gynecological disorders that women suffered from over the centuries. However, disease-specific and effective biomarkers were still not available, since studies have focused on individual genes associated with ovarian cancer, ignoring the interactions and associations among the gene products. Here, ovarian cancer differential co-expression networks were reconstructed via meta-analysis of gene expression data and co-expressed gene modules were identified in epithelial cells from ovarian tumor and healthy ovarian surface epithelial samples to propose ovarian cancer associated genes and their interactions. We propose a novel, highly interconnected, differentially co-expressed, and co-regulated gene module in ovarian cancer consisting of 84 prognostic genes. Furthermore, the specificity of the module to ovarian cancer was shown through analyses of datasets in nine other cancers. These observations underscore the importance of transcriptome based systems biomarkers research in deciphering the elusive pathophysiology of ovarian cancer, and here, we present reciprocal interplay between candidate ovarian cancer genes and their transcriptional regulatory dynamics. The corresponding gene module might provide new insights on ovarian cancer prognosis and treatment strategies that continue to place a significant burden on global health.
Collapse
Affiliation(s)
- Esra Gov
- Department of Bioengineering, Marmara University, 34722, Goztepe, Istanbul, Turkey
| | - Kazim Yalcin Arga
- Department of Bioengineering, Marmara University, 34722, Goztepe, Istanbul, Turkey.
| |
Collapse
|
133
|
Niu G, Wang D, Pei Y, Sun L. Systematic identification of key genes and pathways in the development of invasive cervical cancer. Gene 2017; 618:28-41. [DOI: 10.1016/j.gene.2017.03.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/13/2017] [Accepted: 03/16/2017] [Indexed: 11/30/2022]
|
134
|
Mu Y, Zou H, Chen B, Fan Y, Luo S. FAM83D knockdown regulates proliferation, migration and invasion of colorectal cancer through inhibiting FBXW7/Notch-1 signalling pathway. Biomed Pharmacother 2017; 90:548-554. [DOI: 10.1016/j.biopha.2017.03.073] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 03/08/2017] [Accepted: 03/25/2017] [Indexed: 01/02/2023] Open
|
135
|
Identification of novel cancer therapeutic targets using a designed and pooled shRNA library screen. Sci Rep 2017; 7:43023. [PMID: 28223711 PMCID: PMC5320502 DOI: 10.1038/srep43023] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/17/2017] [Indexed: 01/08/2023] Open
Abstract
Targeted cancer therapeutics aim to exploit tumor-specific, genetic vulnerabilities specifically affecting neoplastic cells without similarly affecting normal cells. Here we performed sequencing-based screening of an shRNA library on a panel of cancer cells of different origins as well as normal cells. The shRNA library was designed to target a subset of genes previously identified using a whole genome screening approach. This focused shRNA library was infected into cells followed by analysis of enrichment and depletion of the shRNAs over the course of cell proliferation. We developed a bootstrap likelihood ratio test for the interpretation of the effects of multiple shRNAs over multiple cell line passages. Our analysis identified 44 genes whose depletion preferentially inhibited the growth of cancer cells. Among these genes ribosomal protein RPL35A, putative RNA helicase DDX24, and coatomer complex I (COPI) subunit ARCN1 most significantly inhibited growth of multiple cancer cell lines without affecting normal cell growth and survival. Further investigation revealed that the growth inhibition caused by DDX24 depletion is independent of p53 status underlining its value as a drug target. Overall, our study establishes a new approach for the analysis of proliferation-based shRNA selection strategies and identifies new targets for the development of cancer therapeutics.
Collapse
|
136
|
Shen HQ, Xiao YX, She ZY, Tan FQ, Yang WX. A novel role of KIF3b in the seminoma cell cycle. Exp Cell Res 2017; 352:95-103. [PMID: 28161539 DOI: 10.1016/j.yexcr.2017.01.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 01/29/2017] [Accepted: 01/31/2017] [Indexed: 11/16/2022]
Abstract
KIF3b is a protein of the kinesin-2 family which plays an important role in intraflagellar transport. Testis cancer is a common cancer among young men. Its diagnostic rate is increasing and over half of the cases are seminomas. Many aspects of the mechanism and gene expression background of this cancer remain unclear. Using western-blotting and semi-quantitative PCR we found high protein levels of KIF3b enrichment in seminoma tissue despite the mRNA levels remaining equivalent to that of normal testicular tissues. The distribution of KIF3b was mainly in cells with division potential. Wound-healing assays and cell counting kit assays showed that the knockdown of KIF3b significantly suppressed cell migration ability, viability and number in HeLa cells. Immunofluorescence images during the cell cycle revealed that KIF3b tended to gather at the spindles and was enriched at the central spindle. This indicated that KIF3b may also have direct impacts upon spindle formation and cytokinesis. By counting the numbers of nuclei, spindles and cells, we found that the rates of multipolar division and multi-nucleation were raised in KIF3b-knockdown cells. In this way we demonstrate that KIF3b functions importantly in mitosis and may be essential to seminoma cell division and proliferation as well as being necessary for normal cell division.
Collapse
Affiliation(s)
- Hao-Qing Shen
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China
| | - Yu-Xi Xiao
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China
| | - Zhen-Yu She
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China
| | - Fu-Qing Tan
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou 310058, China.
| |
Collapse
|
137
|
Zhang F, Lin JD, Zuo XY, Zhuang YX, Hong CQ, Zhang GJ, Cui XJ, Cui YK. Elevated transcriptional levels of aldolase A (ALDOA) associates with cell cycle-related genes in patients with NSCLC and several solid tumors. BioData Min 2017; 10:6. [PMID: 28191039 PMCID: PMC5297095 DOI: 10.1186/s13040-016-0122-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 12/27/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Aldolase A (ALDOA) is one of the glycolytic enzymes primarily found in the developing embryo and adult muscle. Recently, a new role of ALDOA in several cancers has been proposed. However, the underlying mechanism remains obscure and inconsistent. In this study, we tried to investigate ALDOA-associated (AA) genes using available microarray datasets to help elucidating the role of ALDOA in cancer. RESULTS In the dataset of patients with non-small-cell lung cancer (NSCLC, E-GEOD-19188), 3448 differentially expressed genes (DEGs) including ALDOA were identified, in which 710 AA genes were found to be positively associated with ALDOA. Then according to correlation coefficients between each pair of AA genes, ALDOA-associated gene co-expression network (GCN) was constructed including 182 nodes and 1619 edges. 11 clusters out of GCN were detected by ClusterOne plugin in Cytoscape, and only 3 of them have more than three nodes. These three clusters were functionally enriched. A great number of genes (43/79, 54.4%) in the biggest cluster (Cluster 1) primarily involved in biological process like cell cycle process (Pa = 6.76E-26), mitotic cell cycle (Pa = 4.09E-19), DNA repair (Pa = 1.13E-04), M phase of meiotic cell cycle (Pa = 0.006), positive regulation of ubiquitin-protein ligase activity during mitotic cell cycle (Pa = 0.014). AA genes with highest degree and betweenness were considered as hub genes of GCN, namely CDC20, MELK, PTTG1, CCNB2, CDC45, CCNB1, TK1 and PSMB2, which could distinguish cancer from normal controls with ALDOA. Their positive association with ALDOA remained after removing the effect of HK2 and PKM, the two rate limiting enzymes in glycolysis. Further, knocking down ALDOA blocked breast cancer cells in the G0/G1 phase under minimized glycolysis. All suggested that ALDOA might affect cell cycle progression independent of glycolysis. RT-qPCR detection confirmed the relationship of ALDOA with CDC45 and CCNB2 in breast tumors. High expression of the hub genes indicated poor outcome in NSCLC. ALDOA could improve their predictive power. CONCLUSIONS ALDOA could contribute to the progress of cancer, at least partially through its association with genes relevant to cell cycle independent of glycolysis. AA genes plus ALDOA represent a potential new signature for development and prognosis in several cancers.
Collapse
Affiliation(s)
- Fan Zhang
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, 515041 China
| | - Jie-Diao Lin
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, 515041 China
| | - Xiao-Yu Zuo
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060 China
| | - Yi-Xuan Zhuang
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, 515041 China
| | - Chao-Qun Hong
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, 515041 China
| | - Guo-Jun Zhang
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, 515041 China
| | - Xiao-Jiang Cui
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Yu-Kun Cui
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, 515041 China
| |
Collapse
|
138
|
Chakravarthy A, Henderson S, Thirdborough SM, Ottensmeier CH, Su X, Lechner M, Feber A, Thomas GJ, Fenton TR. Human Papillomavirus Drives Tumor Development Throughout the Head and Neck: Improved Prognosis Is Associated With an Immune Response Largely Restricted to the Oropharynx. J Clin Oncol 2016; 34:4132-4141. [PMID: 27863190 PMCID: PMC5477823 DOI: 10.1200/jco.2016.68.2955] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose In squamous cell carcinomas of the head and neck (HNSCC), the increasing incidence of oropharyngeal squamous cell carcinomas (OPSCCs) is attributable to human papillomavirus (HPV) infection. Despite commonly presenting at late stage, HPV-driven OPSCCs are associated with improved prognosis compared with HPV-negative disease. HPV DNA is also detectable in nonoropharyngeal (non-OPSCC), but its pathogenic role and clinical significance are unclear. The objectives of this study were to determine whether HPV plays a causal role in non-OPSCC and to investigate whether HPV confers a survival benefit in these tumors. Methods Meta-analysis was used to build a cross-tissue gene-expression signature for HPV-driven cancer. Classifiers trained by machine-learning approaches were used to predict the HPV status of 520 HNSCCs profiled by The Cancer Genome Atlas project. DNA methylation data were similarly used to classify 464 HNSCCs and these analyses were integrated with genomic, histopathology, and survival data to permit a comprehensive comparison of HPV transcript-positive OPSCC and non-OPSCC. Results HPV-driven tumors accounted for 4.1% of non-OPSCCs. Regardless of anatomic site, HPV+ HNSCCs shared highly similar gene expression and DNA methylation profiles; nonkeratinizing, basaloid histopathological features; and lack of TP53 or CDKN2A alterations. Improved overall survival, however, was largely restricted to HPV-driven OPSCCs, which were associated with increased levels of tumor-infiltrating lymphocytes compared with HPV-driven non-OPSCCs. Conclusion Our analysis identified a causal role for HPV in transcript-positive non-OPSCCs throughout the head and neck. Notably, however, HPV-driven non-OPSCCs display a distinct immune microenvironment and clinical behavior compared with HPV-driven OPSCCs.
Collapse
Affiliation(s)
- Ankur Chakravarthy
- Ankur Chakravarthy, Stephen Henderson, Matt Lechner, Andrew Feber, and Tim R. Fenton, UCL Cancer Institute, University College London, London; Stephen M. Thirdborough, Christian H. Ottensmeier, and Gareth J. Thomas, University of Southampton, Southampton, United Kingdom; Xiaoping Su, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stephen Henderson
- Ankur Chakravarthy, Stephen Henderson, Matt Lechner, Andrew Feber, and Tim R. Fenton, UCL Cancer Institute, University College London, London; Stephen M. Thirdborough, Christian H. Ottensmeier, and Gareth J. Thomas, University of Southampton, Southampton, United Kingdom; Xiaoping Su, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stephen M. Thirdborough
- Ankur Chakravarthy, Stephen Henderson, Matt Lechner, Andrew Feber, and Tim R. Fenton, UCL Cancer Institute, University College London, London; Stephen M. Thirdborough, Christian H. Ottensmeier, and Gareth J. Thomas, University of Southampton, Southampton, United Kingdom; Xiaoping Su, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Christian H. Ottensmeier
- Ankur Chakravarthy, Stephen Henderson, Matt Lechner, Andrew Feber, and Tim R. Fenton, UCL Cancer Institute, University College London, London; Stephen M. Thirdborough, Christian H. Ottensmeier, and Gareth J. Thomas, University of Southampton, Southampton, United Kingdom; Xiaoping Su, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiaoping Su
- Ankur Chakravarthy, Stephen Henderson, Matt Lechner, Andrew Feber, and Tim R. Fenton, UCL Cancer Institute, University College London, London; Stephen M. Thirdborough, Christian H. Ottensmeier, and Gareth J. Thomas, University of Southampton, Southampton, United Kingdom; Xiaoping Su, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Matt Lechner
- Ankur Chakravarthy, Stephen Henderson, Matt Lechner, Andrew Feber, and Tim R. Fenton, UCL Cancer Institute, University College London, London; Stephen M. Thirdborough, Christian H. Ottensmeier, and Gareth J. Thomas, University of Southampton, Southampton, United Kingdom; Xiaoping Su, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Andrew Feber
- Ankur Chakravarthy, Stephen Henderson, Matt Lechner, Andrew Feber, and Tim R. Fenton, UCL Cancer Institute, University College London, London; Stephen M. Thirdborough, Christian H. Ottensmeier, and Gareth J. Thomas, University of Southampton, Southampton, United Kingdom; Xiaoping Su, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gareth J. Thomas
- Ankur Chakravarthy, Stephen Henderson, Matt Lechner, Andrew Feber, and Tim R. Fenton, UCL Cancer Institute, University College London, London; Stephen M. Thirdborough, Christian H. Ottensmeier, and Gareth J. Thomas, University of Southampton, Southampton, United Kingdom; Xiaoping Su, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tim R. Fenton
- Ankur Chakravarthy, Stephen Henderson, Matt Lechner, Andrew Feber, and Tim R. Fenton, UCL Cancer Institute, University College London, London; Stephen M. Thirdborough, Christian H. Ottensmeier, and Gareth J. Thomas, University of Southampton, Southampton, United Kingdom; Xiaoping Su, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
139
|
The microRNA-635 suppresses tumorigenesis in non-small cell lung cancer. Biomed Pharmacother 2016; 84:1274-1281. [DOI: 10.1016/j.biopha.2016.10.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/08/2016] [Accepted: 10/13/2016] [Indexed: 02/07/2023] Open
|
140
|
Bernhart SH, Kretzmer H, Holdt LM, Jühling F, Ammerpohl O, Bergmann AK, Northoff BH, Doose G, Siebert R, Stadler PF, Hoffmann S. Changes of bivalent chromatin coincide with increased expression of developmental genes in cancer. Sci Rep 2016; 6:37393. [PMID: 27876760 PMCID: PMC5120258 DOI: 10.1038/srep37393] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/27/2016] [Indexed: 02/08/2023] Open
Abstract
Bivalent (poised or paused) chromatin comprises activating and repressing histone modifications at the same location. This combination of epigenetic marks at promoter or enhancer regions keeps genes expressed at low levels but poised for rapid activation. Typically, DNA at bivalent promoters is only lowly methylated in normal cells, but frequently shows elevated methylation levels in cancer samples. Here, we developed a universal classifier built from chromatin data that can identify cancer samples solely from hypermethylation of bivalent chromatin. Tested on over 7,000 DNA methylation data sets from several cancer types, it reaches an AUC of 0.92. Although higher levels of DNA methylation are often associated with transcriptional silencing, counter-intuitive positive statistical dependencies between DNA methylation and expression levels have been recently reported for two cancer types. Here, we re-analyze combined expression and DNA methylation data sets, comprising over 5,000 samples, and demonstrate that the conjunction of hypermethylation of bivalent chromatin and up-regulation of the corresponding genes is a general phenomenon in cancer. This up-regulation affects many developmental genes and transcription factors, including dozens of homeobox genes and other genes implicated in cancer. Thus, we reason that the disturbance of bivalent chromatin may be intimately linked to tumorigenesis.
Collapse
Affiliation(s)
- Stephan H Bernhart
- Leipzig University, Chair of Bioinformatics, Leipzig, 04107, Germany.,Leipzig University, Transcriptome Bioinformatics Group - Interdisciplinary Center for Bioinformatics, Leipzig, 04107, Germany
| | - Helene Kretzmer
- Leipzig University, Chair of Bioinformatics, Leipzig, 04107, Germany.,Leipzig University, Transcriptome Bioinformatics Group - Interdisciplinary Center for Bioinformatics, Leipzig, 04107, Germany
| | - Lesca M Holdt
- Ludwig-Maximilians-University, Institute of Laboratory Medicine, Munich, 81377, Germany
| | - Frank Jühling
- Leipzig University, Chair of Bioinformatics, Leipzig, 04107, Germany.,Leipzig University, Transcriptome Bioinformatics Group - Interdisciplinary Center for Bioinformatics, Leipzig, 04107, Germany.,Inserm, U1110 - Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, 67000, France.,Université de Strasbourg, Strasbourg, 67000, France
| | - Ole Ammerpohl
- Christian Albrechts University &University Hospital Schleswig-Holstein - Campus Kiel, Institute of Human Genetics, Kiel, 24105, Germany
| | - Anke K Bergmann
- Christian Albrechts University &University Hospital Schleswig-Holstein - Campus Kiel, Institute of Human Genetics, Kiel, 24105, Germany.,Christian Albrechts University Kiel &University Hospital Schleswig-Holstein - Campus Kiel, Department of Pediatrics, Kiel, 24105, Germany
| | - Bernd H Northoff
- Ludwig-Maximilians-University, Institute of Laboratory Medicine, Munich, 81377, Germany
| | - Gero Doose
- Leipzig University, Chair of Bioinformatics, Leipzig, 04107, Germany.,Leipzig University, Transcriptome Bioinformatics Group - Interdisciplinary Center for Bioinformatics, Leipzig, 04107, Germany
| | - Reiner Siebert
- Christian Albrechts University &University Hospital Schleswig-Holstein - Campus Kiel, Institute of Human Genetics, Kiel, 24105, Germany.,Ulm University &Ulm University Medical Center, Institute for Human Genetics, Ulm, 89081, Germany
| | - Peter F Stadler
- Leipzig University, Chair of Bioinformatics, Leipzig, 04107, Germany.,Leipzig University, Transcriptome Bioinformatics Group - Interdisciplinary Center for Bioinformatics, Leipzig, 04107, Germany.,Leipzig University, LIFE - Leipzig Research Center for Civilization Diseases, Leipzig, 04107, Germany.,University of Vienna, Department of Theoretical Chemistry, Vienna, 1090, Austria.,Max-Planck-Institute for Mathematics in Sciences, Leipzig, 04103, Germany.,Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Steve Hoffmann
- Leipzig University, Chair of Bioinformatics, Leipzig, 04107, Germany.,Leipzig University, Transcriptome Bioinformatics Group - Interdisciplinary Center for Bioinformatics, Leipzig, 04107, Germany.,Leipzig University, LIFE - Leipzig Research Center for Civilization Diseases, Leipzig, 04107, Germany
| |
Collapse
|
141
|
Thomas LD, Vyshenska D, Shulzhenko N, Yambartsev A, Morgun A. Differentially correlated genes in co-expression networks control phenotype transitions. F1000Res 2016; 5:2740. [PMID: 28163897 PMCID: PMC5247791 DOI: 10.12688/f1000research.9708.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2016] [Indexed: 01/06/2023] Open
Abstract
Background: Co-expression networks are a tool widely used for analysis of “Big Data” in biology that can range from transcriptomes to proteomes, metabolomes and more recently even microbiomes. Several methods were proposed to answer biological questions interrogating these networks. Differential co-expression analysis is a recent approach that measures how gene interactions change when a biological system transitions from one state to another. Although the importance of differentially co-expressed genes to identify dysregulated pathways has been noted, their role in gene regulation is not well studied. Herein we investigated differentially co-expressed genes in a relatively simple mono-causal process (B lymphocyte deficiency) and in a complex multi-causal system (cervical cancer). Methods: Co-expression networks of B cell deficiency (Control and BcKO) were reconstructed using Pearson correlation coefficient for two
mus musculus datasets: B10.A strain (12 normal, 12 BcKO) and BALB/c strain (10 normal, 10 BcKO). Co-expression networks of cervical cancer (normal and cancer) were reconstructed using local partial correlation method for five datasets (total of 64 normal, 148 cancer). Differentially correlated pairs were identified along with the location of their genes in BcKO and in cancer networks. Minimum Shortest Path and Bi-partite Betweenness Centrality where statistically evaluated for differentially co-expressed genes in corresponding networks. Results: We show that in B cell deficiency the differentially co-expressed genes are highly enriched with immunoglobulin genes (causal genes). In cancer we found that differentially co-expressed genes act as “bottlenecks” rather than causal drivers with most flows that come from the key driver genes to the peripheral genes passing through differentially co-expressed genes. Using
in vitro knockdown experiments for two out of 14 differentially co-expressed genes found in cervical cancer (FGFR2 and CACYBP), we showed that they play regulatory roles in cancer cell growth. Conclusion: Identifying differentially co-expressed genes in co-expression networks is an important tool in detecting regulatory genes involved in alterations of phenotype.
Collapse
Affiliation(s)
- Lina D Thomas
- Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Anatoly Yambartsev
- Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, Brazil
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, USA
| |
Collapse
|
142
|
Kaveh F, Baumbusch LO, Nebdal D, Børresen-Dale AL, Lingjærde OC, Edvardsen H, Kristensen VN, Solvang HK. A systematic comparison of copy number alterations in four types of female cancer. BMC Cancer 2016; 16:913. [PMID: 27876019 PMCID: PMC5120489 DOI: 10.1186/s12885-016-2899-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/30/2016] [Indexed: 01/06/2023] Open
Abstract
Background Detection and localization of genomic alterations and breakpoints are crucial in cancer research. The purpose of this study was to investigate, in a methodological and biological perspective, different female, hormone-dependent cancers to identify common and diverse DNA aberrations, genes, and pathways. Methods In this work, we analyzed tissue samples from patients with breast (n = 112), ovarian (n = 74), endometrial (n = 84), or cervical (n = 76) cancer. To identify genomic aberrations, the Circular Binary Segmentation (CBS) and Piecewise Constant Fitting (PCF) algorithms were used and segmentation thresholds optimized. The Genomic Identification of Significant Targets in Cancer (GISTIC) algorithm was applied to the segmented data to identify significantly altered regions and the associated genes were analyzed by Ingenuity Pathway Analysis (IPA) to detect over-represented pathways and functions within the identified gene sets. Results and Discussion Analyses of high-resolution copy number alterations in four different female cancer types are presented. For appropriately adjusted segmentation parameters the two segmentation algorithms CBS and PCF performed similarly. We identified one region at 8q24.3 with focal aberrations that was altered at significant frequency across all four cancer types. Considering both, broad regions and focal peaks, three additional regions with gains at significant frequency were revealed at 1p21.1, 8p22, and 13q21.33, respectively. Several of these events involve known cancer-related genes, like PPP2R2A, PSCA, PTP4A3, and PTK2. In the female reproductive system (ovarian, endometrial, and cervix [OEC]), we discovered three common events: copy number gains at 5p15.33 and 15q11.2, further a copy number loss at 8p21.2. Interestingly, as many as 75% of the aberrations (75% amplifications and 86% deletions) identified by GISTIC were specific for just one cancer type and represented distinct molecular pathways. Conclusions Our results disclose that some prominent copy number changes are shared in the four examined female, hormone-dependent cancer whereas others are definitive to specific cancer types. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2899-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fatemeh Kaveh
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Medical Genetics Department, Oslo University Hospital Ullevål, Oslo, Norway.,Department of Pediatric Research, Division of Pediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars O Baumbusch
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Department of Pediatric Research, Division of Pediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Daniel Nebdal
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Ole Christian Lingjærde
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Department of Computer Science, University of Oslo, Oslo, Norway
| | - Hege Edvardsen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Vessela N Kristensen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway. .,Department of Clinical Molecular Biology (EpiGen), Medical Division, Akershus University Hospital, Lørenskog, Norway.
| | - Hiroko K Solvang
- Marine Mammals Research Group, Institute of Marine Research, Bergen, Norway
| |
Collapse
|
143
|
Gu JJ, Zhang JH, Chen HJ, Wang SS. TPX2 promotes glioma cell proliferation and invasion via activation of the AKT signaling pathway. Oncol Lett 2016; 12:5015-5022. [PMID: 28105208 PMCID: PMC5228448 DOI: 10.3892/ol.2016.5371] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 10/14/2016] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and most malignant type of primary adult brain cancer. The most common phenotype associated with GBM is cellular invasion; however, the molecular mechanisms governing this process are poorly understood. Targeting protein for Xenopus kinesin-like protein 2 (TPX2) is a nuclear protein with roles in cellular proliferation and mitotic spindle assembly. TPX2 is overexpressed in various malignancies, including human malignant astrocytoma. Despite this finding, the exact role of TPX2 in human glioma is not well defined. The present study reports the elevated expression of TPX2 in a number of glioma cell lines. TPX2 overexpression promoted cellular proliferation, decreased the percentage of cells in G0/G1 phase, and increased invasion of both U251 and U87 cells. Overexpression of TPX2 also significantly enhanced the phosphorylation of AKT, decreased the expression of p21, and increased the expression of cyclin D1 and matrix metallopeptidase (MMP)-9. In both U251 and U87 cells, knockdown of TPX2 resulted in phenotypes that are in direct contrast to those observed following TPX2 overexpression. Specifically, TPX2 knockdown inhibited cell proliferation, increased the percentage of cells in G0/G1 phase, inhibited invasion, decreased AKT phosphorylation, decreased the expression of MMP-9 and cyclin D1, and increased p21 expression. The AKT inhibitor IV in large part phenocopied the effect of TPX2 knockdown. The present data suggest that TPX2 promotes glioma cell proliferation and invasion via AKT signaling.
Collapse
Affiliation(s)
- Jian-Jun Gu
- Department of Neurosurgery, Fuzhou General Hospital, Xiamen University Medical College, Fuzhou, Fujian 350025, P.R. China; Department of Neurosurgery, Taizhou People's Hospital Affiliated to Medical College of Nantong University, Taizhou, Jiangsu 225300, P.R. China
| | - Jian-He Zhang
- Department of Neurosurgery, Fuzhou General Hospital, Xiamen University Medical College, Fuzhou, Fujian 350025, P.R. China
| | - Hong-Jie Chen
- Department of Neurosurgery, Fuzhou General Hospital, Xiamen University Medical College, Fuzhou, Fujian 350025, P.R. China
| | - Shou-Sen Wang
- Department of Neurosurgery, Fuzhou General Hospital, Xiamen University Medical College, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
144
|
Alekhina O, Marchese A. β-Arrestin1 and Signal-transducing Adaptor Molecule 1 (STAM1) Cooperate to Promote Focal Adhesion Kinase Autophosphorylation and Chemotaxis via the Chemokine Receptor CXCR4. J Biol Chem 2016; 291:26083-26097. [PMID: 27789711 DOI: 10.1074/jbc.m116.757138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/26/2016] [Indexed: 01/14/2023] Open
Abstract
The chemokine receptor CXCR4 and its chemokine ligand CXCL12 mediate directed cell migration during organogenesis, immune responses, and metastatic disease. However, the mechanisms governing CXCL12/CXCR4-dependent chemotaxis remain poorly understood. Here, we show that the β-arrestin1·signal-transducing adaptor molecule 1 (STAM1) complex, initially identified to govern lysosomal trafficking of CXCR4, also mediates CXCR4-dependent chemotaxis. Expression of minigene fragments from β-arrestin1 or STAM1, known to disrupt the β-arrestin1·STAM1 complex, and RNAi against β-arrestin1 or STAM1, attenuates CXCL12-induced chemotaxis. The β-arrestin1·STAM1 complex is necessary for promoting autophosphorylation of focal adhesion kinase (FAK). FAK is necessary for CXCL12-induced chemotaxis and associates with and localizes with β-arrestin1 and STAM1 in a CXCL12-dependent manner. Our data reveal previously unknown roles in CXCR4-dependent chemotaxis for β-arrestin1 and STAM1, which we propose act in concert to regulate FAK signaling. The β-arrestin1·STAM1 complex is a promising target for blocking CXCR4-promoted FAK autophosphorylation and chemotaxis.
Collapse
Affiliation(s)
- Olga Alekhina
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Adriano Marchese
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| |
Collapse
|
145
|
Lin H, Ma Y, Wei Y, Shang H. Genome-wide analysis of aberrant gene expression and methylation profiles reveals susceptibility genes and underlying mechanism of cervical cancer. Eur J Obstet Gynecol Reprod Biol 2016; 207:147-152. [PMID: 27863272 DOI: 10.1016/j.ejogrb.2016.10.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/09/2016] [Accepted: 10/18/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND This study aimed to explore the molecular mechanism of cervical cancer (CC) by integrated bioinformatic analyses of gene expression and methylation profiles. METHODS The gene expression and methylation microarrays in CC samples and normal controls were respectively downloaded from the GEO database. After screening the differentially expressed genes (DEGs) with Limma package and the CC-related methylation sites with CpGassoc package in R language, DEGs with CC-related methylation sites were identified from the intersection of the above two groups of results with 50kb upstream and downstream of a gene as the gene region. Then GO enrichment was performed by GenCLIP2.0 software. Sequentially, analysis of metabolic sub-pathways with pathogenic risk was predicted by iSubpathwayMiner package in R language. RESULTS A total of 1357 DEGs including 721 up-regulated and 636 down-regulated, as well as 666 CC-related methylation sites were screened out. After being analyzed, 26 DEGs with 35 CC-related methylation sites were identified. EDN3 and EDNRB were significantly involved in a function cluster in GO terms of vein smooth muscle contraction, vascular smooth muscle contraction and phasic smooth muscle contraction. LHX2 and PAX6 were significantly involved in a function cluster in GO terms of telencephalon regionalization and forebrain regionalization. ACOX3, CYP39A1 and DPYS were significantly enriched in 25 sub-pathways of 6 major pathways. CONCLUSIONS EDN3 and EDNRB might play important roles in the molecular mechanism of CC, and LHX2, ACOX3, CYP39A1 and DPYS might be susceptibility genes and potential risk markers in CC.
Collapse
Affiliation(s)
- Hongmei Lin
- Department of Gynaecology and Obstetrics, Shandong University Affiliated Jinan Center Hospital, Jinan 250013, Shandong, China
| | - Yifei Ma
- Department of Gynaecology and Obstetrics, Shandong University Affiliated Jinan Center Hospital, Jinan 250013, Shandong, China.
| | - Yongqing Wei
- Department of Gynaecology and Obstetrics, Shandong University Affiliated Jinan Center Hospital, Jinan 250013, Shandong, China
| | - Hui Shang
- Department of Gynaecology and Obstetrics, Shandong University Affiliated Jinan Center Hospital, Jinan 250013, Shandong, China
| |
Collapse
|
146
|
Li J, Lu J, He Y, Wu Y, Wu Y, Song X, Jiang Y, Tang M, Weng X, Yi W, Luo X, Sun L, Bode AM, Cao Y. A new functional IDH2 genetic variant is associated with the risk of lung cancer. Mol Carcinog 2016; 56:1082-1087. [PMID: 27649069 DOI: 10.1002/mc.22573] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 09/06/2016] [Accepted: 09/16/2016] [Indexed: 12/20/2022]
Abstract
Recently, mutations in isocitrate dehydrogenase 1/2 (IDH1/2) were discovered in 70% of low-grade glioma and secondary glioblastoma multiforme. The discovery of an oncogenic function and the identification of onco-metabolites of IDH1/2 support new roles for metabolism in cancer. For example, some evidence indicates that IDH2 might also exhibit oncogenic functions by promoting cellular metabolism and cancer cell growth. We examined the association between IDH2 rs11540478 and lung cancer risk in 262 lung cancer patient cases and 602 healthy control subjects and also investigated the biological function of rs11540478 in vivo. We found that a higher risk was observed in lung cancer patient carriers of rs11540478 TT and CT compared with CC carriers (OR = 1.44; 95%CI = 1.04-2.00; P = 0.03). The frequency of IDH2 rs11540478 TT and CT carriers was decreased in healthy individuals between the ages of 50-77 compared to those aged 30-49 (OR = 0.67; 95%CI = 0.47-0.96; P = 0.03). Functional analysis showed the effect of rs11540478 on IDH2 expression and lung cancer cell viability, with higher IDH2 expression and cell viability among T allele compared with C allele. IDH2 mRNA was higher in peripheral blood lymphocytes from lung cancer patients compared to healthy subjects. Herein, for the first time we identified IDH2 rs11540478 as a new susceptibility locus for lung cancer. The effect of rs11540478 on mRNA expression of IDH2 and lung cancer cell viability might provide new insight for the genetic basis of lung cancer. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jiangjiang Li
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Jingchen Lu
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China.,Department of Medical Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Ya He
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Yong Wu
- Department of Medicine Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yuan Wu
- Department of Medicine Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xin Song
- Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, China
| | - Yuee Jiang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Min Tang
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Xinxian Weng
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Wei Yi
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Lunquan Sun
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| |
Collapse
|
147
|
Roychowdhury A, Samadder S, Das P, Mandloi S, Addya S, Chakraborty C, Basu PS, Mondal R, Roy A, Chakrabarti S, Roychoudhury S, Panda CK. Integrative genomic and network analysis identified novel genes associated with the development of advanced cervical squamous cell carcinoma. Biochim Biophys Acta Gen Subj 2016; 1861:2899-2911. [PMID: 27641506 DOI: 10.1016/j.bbagen.2016.09.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 08/17/2016] [Accepted: 09/12/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND CSCC is one of the most common cancer affecting women globally. Though it is caused by the infection of hrHPV but long latency period for malignant outcome in only a subset of hrHPV infected women indicates involvement of additional alterations, primarily CNVs. Here, we showed how CNVs played a crucial role in development of advanced tumors (stage III/IV) in Indian patients. METHODS Initially, high-resolution CGH-SNP microarray analysis pointed out frequent CNVs followed by significantly altered genes. After comparison with TCGA dataset, expressions of the genes were checked in three CSCC datasets to identify key genes followed by Ingenuity® Pathway analysis. Then node effect property analysis was applied on the constructed PPI network to rank the key proteins. Finally, validations in independent samples were performed. RESULTS For the first time, frequent chromosomal amplifications at 3q13.13-3q29, 1p36.11-1p31.1, 1q21.1-1q44 and 5p15.33-5p12 followed by common deletions at 11q14.1-11q25, 2q34-2q37.3, 4p16.3-4p12 and 13q13.3-13q14.3 were identified in Indian CSCC patients. Integrative analysis found 78 key genes including several novel ones, which were mostly associated with 'Cancer' and may regulate DNA repair and metabolic pathways. Analysis showed PARP1 and ATR were among the top ranking protein interactors. CONCLUSIONS Frequent amplification and over-expression of ATR and PARP1 were further confirmed in cervical lesions, indicating their association with poor prognosis of advanced CSCC patients. GENERAL SIGNIFICANCE Our novel approach identified precise CNVs along with several novel genes within these loci and showed that PARP1 and ATR, having biologically significant interactions, may be involved in development of advanced CSCC.
Collapse
Affiliation(s)
- Anirban Roychowdhury
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Sudip Samadder
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India
| | - Pijush Das
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sapan Mandloi
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sankar Addya
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Partha Sarathi Basu
- India Screening Group (SCR), Early Detection and Prevention Section (EDP), International Agency for Research on Cancer (IARC), World Health Organization (WHO), Lyon, France
| | - Ranajit Mondal
- Department of Gynaecology Oncology, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anup Roy
- North Bengal Medical College and Hospital, West Bengal, India
| | - Saikat Chakrabarti
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | | | - Chinmay Kumar Panda
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, India.
| |
Collapse
|
148
|
Treekitkarnmongkol W, Katayama H, Kai K, Sasai K, Jones JC, Wang J, Shen L, Sahin AA, Gagea M, Ueno NT, Creighton CJ, Sen S. Aurora kinase-A overexpression in mouse mammary epithelium induces mammary adenocarcinomas harboring genetic alterations shared with human breast cancer. Carcinogenesis 2016; 37:1180-1189. [PMID: 27624071 PMCID: PMC5137261 DOI: 10.1093/carcin/bgw097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 08/24/2016] [Accepted: 08/30/2016] [Indexed: 12/20/2022] Open
Abstract
Recent data from The Cancer Genome Atlas analysis have revealed that Aurora kinase A (AURKA) amplification and overexpression characterize a distinct subset of human tumors across multiple cancer types. Although elevated expression of AURKA has been shown to induce oncogenic phenotypes in cells in vitro, findings from transgenic mouse models of Aurora-A overexpression in mammary glands have been distinct depending on the models generated. In the present study, we report that prolonged overexpression of AURKA transgene in mammary epithelium driven by ovine β-lactoglobulin promoter, activated through multiple pregnancy and lactation cycles, results in the development of mammary adenocarcinomas with alterations in cancer-relevant genes and epithelial-to-mesenchymal transition. The tumor incidence was 38.9% (7/18) in Aurora-A transgenic mice at 16 months of age following 4-5 pregnancy cycles. Aurora-A overexpression in the tumor tissues accompanied activation of Akt, elevation of Cyclin D1, Tpx2 and Plk1 along with downregulation of ERα and p53 proteins, albeit at varying levels. Microarray comparative genomic hybridization (CGH) analyses of transgenic mouse mammary adenocarcinomas revealed copy gain of Glp1r and losses of Ercc5, Pten and Tcf7l2 loci. Review of human breast tumor transcriptomic data sets showed association of these genes at varying levels with Aurora-A gain of function alterations. Whole exome sequencing of the mouse tumors also identified gene mutations detected in Aurora-A overexpressing human breast cancers. Our findings demonstrate that prolonged overexpression of Aurora-A can be a driver somatic genetic event in mammary adenocarcinomas associated with deregulated tumor-relevant pathways in the Aurora-A subset of human breast cancer.
Collapse
Affiliation(s)
| | - Hiroshi Katayama
- Department of Translational Molecular Pathology.,Present address: Department of Molecular Oncology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikatacho, Kitaku, Okayama 700-8558, Japan and
| | | | - Kaori Sasai
- Department of Translational Molecular Pathology.,Present address: Department of Molecular Oncology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikatacho, Kitaku, Okayama 700-8558, Japan and
| | - Jennifer Carter Jones
- Department of Translational Molecular Pathology.,Genomics Field Application, Agilent Technologies, Santa Clara, CA 95051, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology
| | - Li Shen
- Department of Bioinformatics and Computational Biology
| | | | - Mihai Gagea
- Department of Veterinary Medicine and Surgery
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA and
| | - Chad J Creighton
- Department of Bioinformatics and Computational Biology.,Department of Medicine, Dan L Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Subrata Sen
- Department of Translational Molecular Pathology,
| |
Collapse
|
149
|
Systems-level effects of ectopic galectin-7 reconstitution in cervical cancer and its microenvironment. BMC Cancer 2016; 16:680. [PMID: 27558259 PMCID: PMC4997669 DOI: 10.1186/s12885-016-2700-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
Background Galectin-7 (Gal-7) is negatively regulated in cervical cancer, and appears to be a link between the apoptotic response triggered by cancer and the anti-tumoral activity of the immune system. Our understanding of how cervical cancer cells and their molecular networks adapt in response to the expression of Gal-7 remains limited. Methods Meta-analysis of Gal-7 expression was conducted in three cervical cancer cohort studies and TCGA. In silico prediction and bisulfite sequencing were performed to inquire epigenetic alterations. To study the effect of Gal-7 on cervical cancer, we ectopically re-expressed it in the HeLa and SiHa cervical cancer cell lines, and analyzed their transcriptome and SILAC-based proteome. We also examined the tumor and microenvironment host cell transcriptomes after xenotransplantation into immunocompromised mice. Differences between samples were assessed with the Kruskall-Wallis, Dunn’s Multiple Comparison and T tests. Kaplan–Meier and log-rank tests were used to determine overall survival. Results Gal-7 was constantly downregulated in our meta-analysis (p < 0.0001). Tumors with combined high Gal-7 and low galectin-1 expression (p = 0.0001) presented significantly better prognoses (p = 0.005). In silico and bisulfite sequencing assays showed de novo methylation in the Gal-7 promoter and first intron. Cells re-expressing Gal-7 showed a high apoptosis ratio (p < 0.05) and their xenografts displayed strong growth retardation (p < 0.001). Multiple gene modules and transcriptional regulators were modulated in response to Gal-7 reconstitution, both in cervical cancer cells and their microenvironments (FDR < 0.05 %). Most of these genes and modules were associated with tissue morphogenesis, metabolism, transport, chemokine activity, and immune response. These functional modules could exert the same effects in vitro and in vivo, even despite different compositions between HeLa and SiHa samples. Conclusions Gal-7 re-expression affects the regulation of molecular networks in cervical cancer that are involved in diverse cancer hallmarks, such as metabolism, growth control, invasion and evasion of apoptosis. The effect of Gal-7 extends to the microenvironment, where networks involved in its configuration and in immune surveillance are particularly affected. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2700-8) contains supplementary material, which is available to authorized users.
Collapse
|
150
|
Lando M, Fjeldbo CS, Wilting SM, C Snoek B, Aarnes EK, Forsberg MF, Kristensen GB, Steenbergen RD, Lyng H. Interplay between promoter methylation and chromosomal loss in gene silencing at 3p11-p14 in cervical cancer. Epigenetics 2016; 10:970-80. [PMID: 26291246 DOI: 10.1080/15592294.2015.1085140] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Loss of 3p11-p14 is a frequent event in epithelial cancer and a candidate prognostic biomarker in cervical cancer. In addition to loss, promoter methylation can participate in gene silencing and promote tumor aggressiveness. We have performed a complete mapping of promoter methylation at 3p11-p14 in two independent cohorts of cervical cancer patients (n = 149, n = 121), using Illumina 450K methylation arrays. The aim was to investigate whether hyperm-ethylation was frequent and could contribute to gene silencing and disease aggressiveness either alone or combined with loss. By comparing the methylation level of individual CpG sites with corresponding data of normal cervical tissue, 26 out of 41 genes were found to be hypermethylated in both cohorts. The frequency of patients with hypermethylation of these genes was found to be higher at tumor stages of 3 and 4 than in stage 1 tumors. Seventeen of the 26 genes were transcriptionally downregulated in cancer compared to normal tissue, whereof 6 genes showed a significant correlation between methylation and expression. Integrated analysis of methylation, gene dosage, and expression of the 26 hypermethylated genes identified 3 regulation patterns encompassing 8 hypermethylated genes; a methylation driven pattern (C3orf14, GPR27, ZNF717), a gene dosage driven pattern (THOC7, PSMD6), and a combined methylation and gene dosage driven pattern (FHIT, ADAMTS9, LRIG1). In survival analysis, patients with both hypermethylation and loss of LRIG1 had a worse outcome compared to those harboring only hypermethylation or none of the events. C3orf14 emerged as a novel methylation regulated suppressor gene, for which knockdown was found to promote invasive growth in human papilloma virus (HPV)-transformed keratinocytes. In conclusion, hypermethylation at 3p11-p14 is common in cervical cancer and may exert a selection pressure during carcinogenesis alone or combined with loss. Information on both events could lead to improved prognostic markers.
Collapse
Affiliation(s)
- Malin Lando
- a Department of Radiation Biology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway
| | - Christina S Fjeldbo
- a Department of Radiation Biology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway
| | - Saskia M Wilting
- b Department of Pathology ; VU University Medical Center ; Amsterdam , the Netherlands
| | - Barbara C Snoek
- b Department of Pathology ; VU University Medical Center ; Amsterdam , the Netherlands
| | - Eva-Katrine Aarnes
- a Department of Radiation Biology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway
| | - Malin F Forsberg
- a Department of Radiation Biology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway
| | - Gunnar B Kristensen
- c Department of Gynecologic Oncology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway.,d Institute for Cancer Genetics and Informatics; Oslo University Hospital ; Oslo , Norway.,e Faculty of Medicine; University of Oslo ; Oslo , Norway
| | - Renske Dm Steenbergen
- b Department of Pathology ; VU University Medical Center ; Amsterdam , the Netherlands
| | - Heidi Lyng
- a Department of Radiation Biology ; Norwegian Radium Hospital; Oslo University Hospital ; Oslo , Norway
| |
Collapse
|