701
|
Liu WR, Tian MX, Yang LX, Lin YL, Jin L, Ding ZB, Shen YH, Peng YF, Gao DM, Zhou J, Qiu SJ, Dai Z, He R, Fan J, Shi YH. PKM2 promotes metastasis by recruiting myeloid-derived suppressor cells and indicates poor prognosis for hepatocellular carcinoma. Oncotarget 2015; 6:846-61. [PMID: 25514599 PMCID: PMC4359260 DOI: 10.18632/oncotarget.2749] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 11/11/2014] [Indexed: 12/21/2022] Open
Abstract
Pyruvate kinase M2 (PKM2) is a member of the pyruvate kinase family. Recent work has defined the "non-metabolic" functions of PKM2. However, the role of PKM2 in HCC remains unclear. To investigate the role of PKM2 in tumor growth, invasion and the prognosis of hepatocellular carcinoma (HCC), PKM2 expression was measured in HCC cell lines and tissues using qRT-PCR, western blot, and immunofluorescence assays. In in vitro experiments, PKM2 was knocked down using a short hairpin RNA lentivirus vector, and tumor cell behavior and the downstream signaling pathways and chemokine were analyzed. For the analysis of in vivo tumor growth, intratumoral and peritumoral lymphocyte infiltration were examined in nude mice. The prognostic value of PKM2 was analyzed by immunohistochemistry in two cohorts including 721 HCC patients. Together, our data obtained from cell lines, tumorigenicity studies, and primary HCC samples illustrate an oncogenic role for PKM2 in tumors. Moreover, PKM2 may serve as a novel prognostic indicator for HCC patients after curative resection, targeted therapy aimed at PKM2 may represent an effective treatment approach for HCC.
Collapse
Affiliation(s)
- Wei-Ren Liu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| | - Meng-Xin Tian
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| | - Liu-Xiao Yang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| | - Yu-Li Lin
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Lei Jin
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| | - Zhen-Bin Ding
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| | - Ying-Hao Shen
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| | - Yuan-Fei Peng
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| | - Dong-Mei Gao
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| | - Jian Zhou
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Shuang-Jian Qiu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| | - Zhi Dai
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| | - Rui He
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Jia Fan
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Ying-Hong Shi
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, People's Republic of China
| |
Collapse
|
702
|
Integrative genomics identifies YY1AP1 as an oncogenic driver in EpCAM(+) AFP(+) hepatocellular carcinoma. Oncogene 2015; 34:5095-104. [PMID: 25597408 PMCID: PMC4506915 DOI: 10.1038/onc.2014.438] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 11/26/2014] [Accepted: 12/05/2014] [Indexed: 12/12/2022]
Abstract
Identification of key drivers and new therapeutic targets is important given the poor prognosis for hepatocellular carcinoma (HCC) patients, particularly those ineligible for surgical resection or liver transplant. However, the approach to identify such driver genes is facing significant challenges due to the genomically heterogenous nature of HCC. Here, we tested whether the integrative genomic profiling of a well-defined HCC subset that is classified by an extreme EpCAM+ AFP+ gene expression signature and associated with poor prognosis, all attributes of a stem cell-like phenotype, could uncover survival-related driver genes in HCC. Following transcriptomic analysis of the well-defined HCC cases, a Gene Set Enrichment Analysis (GSEA) coupled with genomic copy number alteration assessment revealed that YY1-associated protein 1 (YY1AP1) is a critical oncoprotein specifically activated in EpCAM+ AFP+ HCC. YY1AP1 silencing eliminates oncogene addiction by altering the chromatin landscape and triggering massive apoptosis in vitro and tumor suppression in vivo. YY1AP1 expression promotes HCC proliferation and is required for the maintenance of stem cell features. We revealed that YY1AP1 cooperates with YY1 to alter the chromatin landscape and activate transcription of stemness regulators. Thus, YY1AP1 may serve as a key molecular target for EpCAM+ AFP+ HCC subtype. Our results demonstrate the feasibility and power of a new strategy by utilizing well-defined patient samples and integrative genomics to uncover critical pathways linked to HCC subtypes with prognostic impact.
Collapse
|
703
|
Roessler S, Lin G, Forgues M, Budhu A, Hoover S, Simpson RM, Wu X, He P, Qin LX, Tang ZY, Ye QH, Wang XW. Integrative genomic and transcriptomic characterization of matched primary and metastatic liver and colorectal carcinoma. Int J Biol Sci 2015; 11:88-98. [PMID: 25552933 PMCID: PMC4278258 DOI: 10.7150/ijbs.10583] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 10/13/2014] [Indexed: 12/13/2022] Open
Abstract
Metastasis is the main cause of cancer mortality but its process remains poorly understood and thus hampers more effective treatment and improved cancer prognosis. To search for metastasis driver genes responsible for tumor spread, we integrated genomic and transcriptomic profiles of 61 matched primary tumors and distant metastases of liver or colorectal carcinoma isolated by laser-capture microdissection and assayed by array-based technologies. We found that primary tumor lesions and their matched distant metastases were largely similar at the genomic and transcriptomic levels, but substantial differences could be found between primary tumors with or without accompanying metastases. Interestingly, metastasis genes were principally tumor type and organ site-specific. Despite distinct pathway enrichment, different metastasis gene sets shared common prognostic capacity and were predictive of hepatocellular carcinoma survival in an independent cohort. Thus, the metastatic propensity is inherent to the primary tumor and the lack of general metastasis genes necessitates the development of specific treatment modalities.
Collapse
Affiliation(s)
- Stephanie Roessler
- 1. Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Guoling Lin
- 1. Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD, USA; ; 2. Liver Cancer Institute, Fudan University, Shanghai, China
| | - Marshonna Forgues
- 1. Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Anuradha Budhu
- 1. Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Shelley Hoover
- 3. Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - R Mark Simpson
- 3. Laboratory of Cancer Biology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Xiaolin Wu
- 4. Laboratory of Molecular Technology, NCI-Frederick, SAIC-Frederick, Frederick, MD 21701, USA
| | - Ping He
- 5. Division of Hematology, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA
| | - Lun-Xiu Qin
- 2. Liver Cancer Institute, Fudan University, Shanghai, China
| | - Zhao-You Tang
- 2. Liver Cancer Institute, Fudan University, Shanghai, China
| | - Qing-Hai Ye
- 2. Liver Cancer Institute, Fudan University, Shanghai, China
| | - Xin Wei Wang
- 1. Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
704
|
Tummala KS, Gomes AL, Yilmaz M, Graña O, Bakiri L, Ruppen I, Ximénez-Embún P, Sheshappanavar V, Rodriguez-Justo M, Pisano DG, Wagner EF, Djouder N. Inhibition of de novo NAD(+) synthesis by oncogenic URI causes liver tumorigenesis through DNA damage. Cancer Cell 2014; 26:826-839. [PMID: 25453901 DOI: 10.1016/j.ccell.2014.10.002] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 07/23/2014] [Accepted: 10/02/2014] [Indexed: 02/07/2023]
Abstract
Molecular mechanisms responsible for hepatocellular carcinoma (HCC) remain largely unknown. Using genetically engineered mouse models, we show that hepatocyte-specific expression of unconventional prefoldin RPB5 interactor (URI) leads to a multistep process of HCC development, whereas its genetic reduction in hepatocytes protects against diethylnitrosamine (DEN)-induced HCC. URI inhibits aryl hydrocarbon (AhR)- and estrogen receptor (ER)-mediated transcription of enzymes implicated in L-tryptophan/kynurenine/nicotinamide adenine dinucleotide (NAD(+)) metabolism, thereby causing DNA damage at early stages of tumorigenesis. Restoring NAD(+) pools with nicotinamide riboside (NR) prevents DNA damage and tumor formation. Consistently, URI expression in human HCC is associated with poor survival and correlates negatively with L-tryptophan catabolism pathway. Our results suggest that boosting NAD(+) can be prophylactic or therapeutic in HCC.
Collapse
Affiliation(s)
- Krishna S Tummala
- Growth Factors, Nutrients and Cancer Group, BBVA Foundation-Cancer Cell Biology Programme, Spanish National Cancer Research Centre, CNIO, 28029 Madrid, Spain
| | - Ana L Gomes
- Growth Factors, Nutrients and Cancer Group, BBVA Foundation-Cancer Cell Biology Programme, Spanish National Cancer Research Centre, CNIO, 28029 Madrid, Spain
| | - Mahmut Yilmaz
- Growth Factors, Nutrients and Cancer Group, BBVA Foundation-Cancer Cell Biology Programme, Spanish National Cancer Research Centre, CNIO, 28029 Madrid, Spain
| | - Osvaldo Graña
- Bioinformatics Unit, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre, CNIO, 28029 Madrid, Spain
| | - Latifa Bakiri
- Genes, Development, and Disease Group, BBVA Foundation-Cancer Cell Biology Programme, Spanish National Cancer Research Centre, CNIO, 28029 Madrid, Spain
| | - Isabel Ruppen
- Proteomics Core Unit, ProteoRed ISCIII, Biotechnology Programme, Spanish National Cancer Research Centre, CNIO, 28029 Madrid, Spain
| | - Pilar Ximénez-Embún
- Proteomics Core Unit, ProteoRed ISCIII, Biotechnology Programme, Spanish National Cancer Research Centre, CNIO, 28029 Madrid, Spain
| | | | - Manuel Rodriguez-Justo
- Department of Cellular Pathology, University College London NHS Trust, London NW1 2BU, UK
| | - David G Pisano
- Bioinformatics Unit, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre, CNIO, 28029 Madrid, Spain
| | - Erwin F Wagner
- Genes, Development, and Disease Group, BBVA Foundation-Cancer Cell Biology Programme, Spanish National Cancer Research Centre, CNIO, 28029 Madrid, Spain
| | - Nabil Djouder
- Growth Factors, Nutrients and Cancer Group, BBVA Foundation-Cancer Cell Biology Programme, Spanish National Cancer Research Centre, CNIO, 28029 Madrid, Spain.
| |
Collapse
|
705
|
Wilson GK, Tennant DA, McKeating JA. Hypoxia inducible factors in liver disease and hepatocellular carcinoma: current understanding and future directions. J Hepatol 2014; 61:1397-406. [PMID: 25157983 DOI: 10.1016/j.jhep.2014.08.025] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 08/07/2014] [Accepted: 08/17/2014] [Indexed: 02/07/2023]
Abstract
Hypoxia inducible transcription factors (HIFs) activate diverse pathways that regulate cellular metabolism, angiogenesis, proliferation, and migration, enabling a cell to respond to a low oxygen or hypoxic environment. HIFs are regulated by oxygen-dependent and independent signals including: mitochondrial dysfunction, reactive oxygen species, endoplasmic reticular stress, and viral infection. HIFs have been reported to play a role in the pathogenesis of liver disease of diverse aetiologies. This review explores the impact of HIFs on hepatocellular biology and inflammatory responses, highlighting the therapeutic potential of targeting HIFs for an array of liver pathologies.
Collapse
Affiliation(s)
- Garrick K Wilson
- Viral Hepatitis Research Group, Centre for Human Virology, University of Birmingham, Birmingham, UK
| | - Daniel A Tennant
- School of Cancer Sciences, University of Birmingham, Birmingham, UK
| | - Jane A McKeating
- Viral Hepatitis Research Group, Centre for Human Virology, University of Birmingham, Birmingham, UK; NIHR Liver Biomedical Research Unit, University of Birmingham, Birmingham, UK.
| |
Collapse
|
706
|
Gozdecka M, Lyons S, Kondo S, Taylor J, Li Y, Walczynski J, Thiel G, Breitwieser W, Jones N. JNK suppresses tumor formation via a gene-expression program mediated by ATF2. Cell Rep 2014; 9:1361-74. [PMID: 25456131 DOI: 10.1016/j.celrep.2014.10.043] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 07/16/2014] [Accepted: 10/14/2014] [Indexed: 02/09/2023] Open
Abstract
JNK and p38 phosphorylate a diverse set of substrates and, consequently, can act in a context-dependent manner to either promote or inhibit tumor growth. Elucidating the functions of specific substrates of JNK and p38 is therefore critical for our understanding of these kinases in cancer. ATF2 is a phosphorylation-dependent transcription factor and substrate of both JNK and p38. Here, we show ATF2 suppresses tumor formation in an orthotopic model of liver cancer and cellular transformation in vitro. Furthermore, we find that suppression of tumorigenesis by JNK requires ATF2. We identify a transcriptional program activated by JNK via ATF2 and provide examples of JNK- and ATF2-dependent genes that block cellular transformation. Significantly, we also show that ATF2-dependent gene expression is frequently downregulated in human cancers, indicating that amelioration of JNK-ATF2-mediated suppression may be a common event during tumor development.
Collapse
Affiliation(s)
- Malgorzata Gozdecka
- Department of Cell Regulation, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester M20 4BX, UK; Haematological Cancer Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Stephen Lyons
- Department of Cell Regulation, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester M20 4BX, UK
| | - Saki Kondo
- Department of Cell Regulation, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester M20 4BX, UK; Laboratory of Molecular Genetics, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Janet Taylor
- Central Manchester NHS Trust and University of Manchester, the Nowgen Centre, 29 Grafton Street, Manchester M13 9WU, UK; Applied Computational Biology and Bioinformatics Group, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester M20 4BX, UK
| | - Yaoyong Li
- Applied Computational Biology and Bioinformatics Group, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester M20 4BX, UK
| | - Jacek Walczynski
- Department of Cell Regulation, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester M20 4BX, UK
| | - Gerald Thiel
- Department of Medical Biochemistry and Molecular Biology, University of Saarland Medical Center, Building 44, 66421 Homburg, Germany
| | - Wolfgang Breitwieser
- Department of Cell Regulation, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester M20 4BX, UK
| | - Nic Jones
- Department of Cell Regulation, CRUK Manchester Institute, Paterson Building, University of Manchester, Manchester M20 4BX, UK.
| |
Collapse
|
707
|
Hirata H, Sugimachi K, Takahashi Y, Ueda M, Sakimura S, Uchi R, Kurashige J, Takano Y, Nanbara S, Komatsu H, Saito T, Shinden Y, Iguchi T, Eguchi H, Atsumi K, Sakamoto K, Doi T, Hirakawa M, Honda H, Mimori K. Downregulation of PRRX1 Confers Cancer Stem Cell-Like Properties and Predicts Poor Prognosis in Hepatocellular Carcinoma. Ann Surg Oncol 2014; 22 Suppl 3:S1402-9. [PMID: 25404478 DOI: 10.1245/s10434-014-4242-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Downregulation of paired related homeobox 1 (PRRX1) is associated with the acquisition of cancer stem cell (CSC)-like properties and poor prognosis in cancers. The purpose of this study is to clarify the role of PRRX1 expression in predicting prognosis and mediating CSC-like properties in hepatocellular carcinoma (HCC). METHODS The association between PRRX1 expression and overall survival (OS) of patients with HCC was analyzed in three independent datasets: 62 resected primary cases, 242 cases from GSE14520, and 162 cases from The Cancer Genome Atlas (TCGA). A cell line expressing PRRX1 (HuH7) was established for the functional analyses. The ability to form spheres, the expression levels of the hepatic CSC surface markers (CD13, CD133, and EpCAM), in vitro chemosensitivity to 5-fluorouracil (FU), and radiosensitivity were evaluated. RESULTS Univariate and multivariate analyses showed that the 5-year OS of the low PRRX1 expression group was significantly poorer than that of the high PRRX1 expression group (P = 0.024 and P = 0.045, respectively). Consistent with this, the low PRRX1 expression group in GSE14520 and TCGA datasets showed significantly shorter OS (P = 0.027 and P = 0.010, respectively). Gene set enrichment analysis on GSE14520 and TCGA datasets indicated that downregulation of PRRX1 was correlated with the stemness signature. The number of spheres and the expression levels of CSC markers were significantly decreased when PRRX1 was expressed. Moreover, PRRX1 impaired resistance to 5-FU and radiation. CONCLUSIONS Downregulation of PRRX1 expression contributes to the poor prognosis of patients with HCC through acquisition of CSC-like properties.
Collapse
Affiliation(s)
- Hidenari Hirata
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.,Department of Radiology, Kyushu University Beppu Hospital, Beppu, Japan
| | - Keishi Sugimachi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yusuke Takahashi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Masami Ueda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Shotaro Sakimura
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Ryutaro Uchi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Junji Kurashige
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yuki Takano
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Sho Nanbara
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Hisateru Komatsu
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Tomoko Saito
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yoshiaki Shinden
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Tomohiro Iguchi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Hidetoshi Eguchi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Kazushige Atsumi
- Department of Radiology, Kyushu University Beppu Hospital, Beppu, Japan
| | - Katsumi Sakamoto
- Department of Radiology, Kyushu University Beppu Hospital, Beppu, Japan
| | - Toshiro Doi
- Department of Orthopedic Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Masakazu Hirakawa
- Department of Radiology, Kyushu University Beppu Hospital, Beppu, Japan
| | - Hiroshi Honda
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.
| |
Collapse
|
708
|
Zhou X, Zhu HQ, Lu J. Regulation of gene expression in HBV- and HCV-related hepatocellular carcinoma: integrated GWRS and GWGS analyses. Int J Clin Exp Med 2014; 7:4038-4050. [PMID: 25550913 PMCID: PMC4276171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/08/2014] [Indexed: 06/04/2023]
Abstract
OBJECTIVES To explore the molecular mechanism of hepatitis B virus-related and hepatitis C virus-related hepatocellular carcinoma, samples from hepatitis B virus and hepatitis C virus infected patients and the normal were compared, respectively. METHODS In both experiments, genes with high value were selected based on a genome-wide relative significance and genome-wide global significance model. Co-expression network of the selected genes was constructed, and transcription factors in the network were identified. Molecular complex detection algorithm was used to obtain sub-networks. RESULTS Based on the new model, the top 300 genes were selected. Co-expression network was constructed and transcription factors were identified. We obtained two common genes FCN2 and CXCL14, and two common transcription factors RFX5 and EZH2. In hepatitis B virus experiment, cluster 1 and 3 had the higher value. In cluster 1, ten of the 17 genes and one transcription factor were all reported associated with hepatocellular carcinoma. In cluster 3, transcription factor ESR1 was reported related with hepatocellular carcinoma. In hepatitis C virus experiment, the value of cluster 3 and 4 was higher. In cluster 3, nine genes were reported to play a key role in hepatocellular carcinoma. In cluster 4, there were 5 genes in the 34 genes. To compare the relevance of a node in holding together communicating nodes, centralities based analysis was performed and we obtained some genes with high stress value. CONCLUSION The analysis above helped us to understand the pathogenesis of hepatitis B virus and hepatitis C virus associated hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xu Zhou
- Department of General Surgery, Provincial Hospital Affiliated to Shandong University Ji'nan 250014, China
| | - Hua-Qiang Zhu
- Department of General Surgery, Provincial Hospital Affiliated to Shandong University Ji'nan 250014, China
| | - Jun Lu
- Department of General Surgery, Provincial Hospital Affiliated to Shandong University Ji'nan 250014, China
| |
Collapse
|
709
|
Ding ZY, Jin GN, Wang W, Chen WX, Wu YH, Ai X, Chen L, Zhang WG, Liang HF, Laurence A, Zhang MZ, Datta PK, Zhang B, Chen XP. Reduced expression of transcriptional intermediary factor 1 gamma promotes metastasis and indicates poor prognosis of hepatocellular carcinoma. Hepatology 2014; 60:1620-1636. [PMID: 24954480 DOI: 10.1002/hep.27273] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 06/17/2014] [Indexed: 12/20/2022]
Abstract
UNLABELLED Transcriptional intermediary factor 1 gamma (TIF1γ) may play either a potential tumor-suppressor or -promoter role in cancer. Here we report on a critical role of TIF1γ in the progression of hepatocellular carcinoma (HCC). Reduced expression of TIF1γ was detected in HCC, especially in advanced HCC tissues, compared to adjacent noncancerous tissues. HCC patients with low TIF1γ expression had shorter overall survival times and higher recurrence rates than those with high TIF1γ expression. Reduced TIF1γ expression was an independent and significant risk factor for recurrence and survival after curative resection. In HCC cells, TIF1γ played a dual role: It promoted tumor growth in early-stage HCC, but not in advanced-stage HCC, whereas it inhibited invasion and metastasis in both early- and advanced-stage HCC. Mechanistically, we confirmed that TIF1γ inhibited transforming growth factor-β/ Drosophila mothers against decapentaplegic protein (TGF-β/Smad) signaling through monoubiquitination of Smad4 and suppressed the formation of Smad2/3/4 complex in HCC cells. TGF-β-inducing cytostasis and metastasis were both inhibited by TIF1γ in HCC. We further proved that TIF1γ suppressed cyotstasis-related TGF-β/Smad downstream c-myc down-regulation, as well as p21/cip1 and p15/ink4b up-regulation in early-stage HCC. Meanwhile, TGF-β inducible epithelial-mesenchymal transition and TGF-β/Smad downstream metastatic cascades, including phosphatase and tensin homolog deleted on chromosome ten down-regulation, chemokine (CXC motif) receptor 4 and matrix metalloproteinase 1 induction, and epidermal growth factor receptor- and protein kinase B-signaling transactivation, were inhibited by TIF1γ. In addition, we found that the down-regulation of TIF1γ in HCC was caused by hypermethylation of CpG islands in the TIF1γ promoter, and demonstrated that the combination of TIF1γ and phosphorylated Smad2 was a more powerful predictor of poor prognosis. CONCLUSION TIF1γ regulates tumor growth and metastasis through inhibition of TGF-β/Smad signaling and may serve as a novel prognostic biomarker in HCC.
Collapse
Affiliation(s)
- Ze-yang Ding
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
710
|
Ueno A, Masugi Y, Yamazaki K, Komuta M, Effendi K, Tanami Y, Tsujikawa H, Tanimoto A, Okuda S, Itano O, Kitagawa Y, Kuribayashi S, Sakamoto M. OATP1B3 expression is strongly associated with Wnt/β-catenin signalling and represents the transporter of gadoxetic acid in hepatocellular carcinoma. J Hepatol 2014; 61:1080-7. [PMID: 24946283 DOI: 10.1016/j.jhep.2014.06.008] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 05/31/2014] [Accepted: 06/06/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS In the current era of emerging molecular targeted drugs, it is necessary to identify before treatment the specific subclass to which a tumour belongs. Gadoxetic acid is a liver-specific contrast agent that is preferentially taken up by hepatocytes. Therefore, gadoxetic acid-enhanced magnetic resonance imaging (EOB-MRI) should provide precise molecular information about hepatocellular carcinomas (HCCs). The aim of this study was to investigate the transporters of gadoxetic acid in HCC comprehensively and to analyse the molecular regulatory mechanism of such transporters. METHODS Expression levels of transporters, transcriptional factors and Wnt target genes in clinical samples were examined by quantitative real-time reverse transcription polymerase chain reaction and immunohistochemistry. LiCl treatment of the HCC cell line KYN-2 was conducted in vitro to assess the effects of Wnt signalling activity. RESULTS Comprehensive analyses of transporter mRNAs and protein expressions revealed that the organic anion transporting polypeptide 1B3 (OATP1B3) had the strongest correlation with tumour enhancement in hepatobiliary-phase images of EOB-MRI. Association analysis with OATP1B3 expression revealed significant correlation with the expression of Wnt/β-catenin target genes. Further, LiCl treatment induced OATP1B3 mRNA expression in KYN-2 cells, indicating a strong association between OATP1B3 expression and Wnt/β-catenin signalling. The sensitivity and specificity to predict Wnt/β-catenin-activated HCC using tumour enhancement in EOB-MRI were 78.9% and 81.7%, respectively. CONCLUSIONS OATP1B3 was confirmed as the most important transporter mediating HCC enhancement in EOB-MRI. OATP1B3 expression showed a strong association with the expression of Wnt/β-catenin target genes, therefore, OATP1B3-upregulated HCC likely represents a specific subclass of Wnt/β-catenin-activated HCC.
Collapse
Affiliation(s)
- Akihisa Ueno
- Department of Pathology, School of Medicine, Keio University, Tokyo, Japan; Department of Diagnostic Radiology, School of Medicine, Keio University, Tokyo, Japan
| | - Yohei Masugi
- Department of Pathology, School of Medicine, Keio University, Tokyo, Japan
| | - Ken Yamazaki
- Department of Pathology, School of Medicine, Keio University, Tokyo, Japan
| | - Mina Komuta
- Department of Pathology, School of Medicine, Keio University, Tokyo, Japan; Department of Translational Cell & Tissue Research, University Hospitals Leuven, Leuven, Belgium
| | - Kathryn Effendi
- Department of Pathology, School of Medicine, Keio University, Tokyo, Japan
| | - Yutaka Tanami
- Department of Diagnostic Radiology, School of Medicine, Keio University, Tokyo, Japan
| | - Hanako Tsujikawa
- Department of Pathology, School of Medicine, Keio University, Tokyo, Japan
| | - Akihiro Tanimoto
- Department of Diagnostic Radiology, School of Medicine, Keio University, Tokyo, Japan
| | - Shigeo Okuda
- Department of Diagnostic Radiology, School of Medicine, Keio University, Tokyo, Japan
| | - Osamu Itano
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Sachio Kuribayashi
- Department of Diagnostic Radiology, School of Medicine, Keio University, Tokyo, Japan
| | - Michiie Sakamoto
- Department of Pathology, School of Medicine, Keio University, Tokyo, Japan.
| |
Collapse
|
711
|
Ko JH, Gu W, Lim I, Zhou T, Bang H. Expression profiling of mitochondrial voltage-dependent anion channel-1 associated genes predicts recurrence-free survival in human carcinomas. PLoS One 2014; 9:e110094. [PMID: 25333947 PMCID: PMC4198298 DOI: 10.1371/journal.pone.0110094] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 09/08/2014] [Indexed: 01/17/2023] Open
Abstract
Background Mitochondrial voltage-dependent anion channels (VDACs) play a key role in mitochondria-mediated apoptosis. Both in vivo and in vitro evidences indicate that VDACs are actively involved in tumor progression. Specifically, VDAC-1, one member of the VDAC family, was thought to be a potential anti-cancer therapeutic target. Our previous study demonstrated that the human gene VDAC1 (encoding the VDAC-1 isoform) was significantly up-regulated in lung tumor tissue compared with normal tissue. Also, we found a significant positive correlation between the gene expression of VDAC1 and histological grade in breast cancer. However, the prognostic power of VDAC1 and its associated genes in human cancers is largely unknown. Methods We systematically analyzed the expression pattern of VDAC1 and its interacting genes in breast, colon, liver, lung, pancreatic, and thyroid cancers. The genes differentially expressed between normal and tumor tissues in human carcinomas were identified. Results The expression level of VDAC1 was uniformly up-regulated in tumor tissue compared with normal tissue in breast, colon, liver, lung, pancreatic, and thyroid cancers. Forty-four VDAC1 interacting genes were identified as being commonly differentially expressed between normal and tumor tissues in human carcinomas. We designated VDAC1 and the 44 dysregulated interacting genes as the VDAC1 associated gene signature (VAG). We demonstrate that the VAG signature is a robust prognostic biomarker to predict recurrence-free survival in breast, colon, and lung cancers, and is independent of standard clinical and pathological prognostic factors. Conclusions VAG represents a promising prognostic biomarker in human cancers, which may enhance prediction accuracy in identifying patients at higher risk for recurrence. Future therapies aimed specifically at VDAC1 associated genes may lead to novel agents in the treatment of cancer.
Collapse
Affiliation(s)
- Jae-Hong Ko
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, South Korea
| | - Wanjun Gu
- Research Center for Learning Sciences, Southeast University, Nanjing, Jiangsu, China
| | - Inja Lim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, South Korea
| | - Tong Zhou
- Department of Medicine, University of Arizona, Tucson, Arizona, United States of America
- * E-mail: (TZ); (HB)
| | - Hyoweon Bang
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, South Korea
- * E-mail: (TZ); (HB)
| |
Collapse
|
712
|
Malz M, Bovet M, Samarin J, Rabenhorst U, Sticht C, Bissinger M, Roessler S, Bermejo JL, Renner M, Calvisi DF, Singer S, Ganzinger M, Weber A, Gretz N, Zörnig M, Schirmacher P, Breuhahn K. Overexpression of far upstream element (FUSE) binding protein (FBP)-interacting repressor (FIR) supports growth of hepatocellular carcinoma. Hepatology 2014; 60:1241-50. [PMID: 24824848 DOI: 10.1002/hep.27218] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 05/09/2014] [Indexed: 12/24/2022]
Abstract
UNLABELLED The far upstream element binding protein (FBP) and the FBP-interacting repressor (FIR) represent molecular tools for transcriptional fine tuning of target genes. Strong overexpression of FBP in human hepatocellular carcinoma (HCC) supports tumor growth and correlates with poor patient prognosis. However, the role of the transcriptional repressor FIR in hepatocarcinogenesis remains poorly delineated. We show that overexpression of FIR correlates with tumor dedifferentiation and tumor cell proliferation in about 60% of primary HCCs. Elevated FIR levels are associated with genomic gains of the FIR gene locus at chromosome 8q24.3 in human HCC specimens. In vitro, nuclear enrichment of FIR supports HCC cell proliferation and migration. Expression profiling of HCC cells after small interfering RNA (siRNA)-mediated silencing of FIR identified the transcription factor DP-1 (TFDP1) as a transcriptional target of FIR. Surprisingly, FIR stimulates the expression of FBP in a TFDP1/E2F1-dependent manner. FIR splice variants lacking or containing exon 2 and/or exon 5 are expressed in the majority of HCCs but not in normal hepatocytes. Specific inhibition of FIR isoforms with and without exon 2 revealed that both groups of FIR splice variants facilitate tumor-supporting effects. This finding was confirmed in xenograft transplantation experiments with lentiviral-infected short hairpin RNA (shRNA) targeting all FIR variants as well as FIR with and without exon 2. CONCLUSION High-level nuclear FIR does not facilitate repressor properties but supports tumor growth in HCC cells. Thus, the pharmacological inhibition of FIR might represent a promising therapeutic strategy for HCC patients with elevated FIR expression.
Collapse
Affiliation(s)
- Mona Malz
- Institute of Pathology, University Hospital Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
713
|
Integration of genomic information in the clinical management of HCC. Best Pract Res Clin Gastroenterol 2014; 28:831-42. [PMID: 25260311 DOI: 10.1016/j.bpg.2014.08.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 07/11/2014] [Accepted: 08/15/2014] [Indexed: 02/06/2023]
Abstract
Molecular profiling of hepatocellular carcinoma (HCC) is enabling the advancement of novel approaches to disease diagnosis and management. Accurate prognosis prediction in HCC is specially critical. Clinical staging systems for HCC support clinical decision-making (e.g., BCLC algorithm) might be complemented by molecular-based information in the near future. Molecular signatures derived from tumour and non-tumour samples are associated with patient recurrence an outcome. Single nucleotide polymorphisms have been linked with HCC development. Next generation sequencing studies have brought to light the genomic diversity of this disease. Gens recurrently altered in HCC and susceptible to be targeted belong to signalling pathways including telomere maintenance, cell cycle, chromatin remodelling, Wnt/beta-catenin, RAS/RAF/MAPK and PI3K/AKT/mTOR pathways. Oncogenic loops are unknown but might include some of the already discovered aberrations. Despite the intratumoral heterogeneity observed in HCC tumours, studies including large number of samples can identify key genetic drivers and contribute to the development of novel treatments and a personalized medicine.
Collapse
|
714
|
Ganne-Carrié N, Nault JC, Ziol M, N'Kontchou G, Nahon P, Grando V, Bourcier V, Barge S, Beaugrand M, Trinchet JC, Seror O. Predicting recurrence following radiofrequency percutaneous ablation for hepatocellular carcinoma. Hepat Oncol 2014; 1:395-408. [PMID: 30190975 PMCID: PMC6095149 DOI: 10.2217/hep.14.22] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Within 5 years after percutaneous ablation of hepatocellular carcinoma, roughly 70% of patients experience tumor recurrence. Relapses beyond curative options affected patients' survival. Ablation shares with resection common predictive factors of recurrence as size of the tumor, multinodularity and presence of vascular invasion. High serum α-fetoprotein level and markers of severity of underlying liver disease have also been found to be associated with recurrence and even survival. However, predictive values for recurrence of technical factors, histopathological and molecular tumors' features have been rarely studied. Few comparative studies have shown that ablation techniques impact recurrence rates. Moreover, although ablation does not allow analysis of the whole tumor, some reports suggest that biopsies allow histopathological and even molecular testing of the risk of recurrence.
Collapse
Affiliation(s)
- Nathalie Ganne-Carrié
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Pôle d'Activités Cancérologiques Spécialisées, Service d'Hépatologie, F-93143 Bondy, France
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, 27 rue Juliette Dodu, F-75010 Paris, France
| | - Jean-Charles Nault
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Pôle d'Activités Cancérologiques Spécialisées, Service d'Hépatologie, F-93143 Bondy, France
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, 27 rue Juliette Dodu, F-75010 Paris, France
| | - Marianne Ziol
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, 27 rue Juliette Dodu, F-75010 Paris, France
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Centre de Ressources Biologiques, F-93143 Bondy, France
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Laboratoire d'Anatomie Pathologique, F-93143 Bondy, France
| | - Gisèle N'Kontchou
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Pôle d'Activités Cancérologiques Spécialisées, Service d'Hépatologie, F-93143 Bondy, France
| | - Pierre Nahon
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Pôle d'Activités Cancérologiques Spécialisées, Service d'Hépatologie, F-93143 Bondy, France
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, 27 rue Juliette Dodu, F-75010 Paris, France
| | - Véronique Grando
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Pôle d'Activités Cancérologiques Spécialisées, Service d'Hépatologie, F-93143 Bondy, France
| | - Valérie Bourcier
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Pôle d'Activités Cancérologiques Spécialisées, Service d'Hépatologie, F-93143 Bondy, France
| | - Sandrine Barge
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Pôle d'Activités Cancérologiques Spécialisées, Service d'Hépatologie, F-93143 Bondy, France
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France
| | - Michel Beaugrand
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Pôle d'Activités Cancérologiques Spécialisées, Service d'Hépatologie, F-93143 Bondy, France
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France
| | - Jean-Claude Trinchet
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Pôle d'Activités Cancérologiques Spécialisées, Service d'Hépatologie, F-93143 Bondy, France
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, 27 rue Juliette Dodu, F-75010 Paris, France
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Centre de Ressources Biologiques, F-93143 Bondy, France
| | - Olivier Seror
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, F-93000 Bobigny, France
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, 27 rue Juliette Dodu, F-75010 Paris, France
- APHP, Hôpitaux Universitaires Paris – Seine-Saint Denis, Site Jean Verdier, Département d'imagerie Médicale, 93143 Bondy, France
| |
Collapse
|
715
|
Kim H, Park YN. Role of biopsy sampling for diagnosis of early and progressed hepatocellular carcinoma. Best Pract Res Clin Gastroenterol 2014; 28:813-29. [PMID: 25260310 DOI: 10.1016/j.bpg.2014.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/15/2014] [Indexed: 01/31/2023]
Abstract
The current guidelines for the diagnosis of hepatocellular carcinoma (HCC) recommend liver biopsy for hepatic nodules which do not demonstrate the typical features of HCC on imaging. Thus, while not all HCCs are biopsied for histological confirmation, the nodules that pathologists now encounter on biopsy specimens are frequently well-differentiated early HCCs. This paper reviews the pathological features of HCC and its precursor lesions on liver biopsy specimens, with special emphasis on the differential diagnosis between well-differentiated HCCs and high-grade dysplastic nodules, and discusses the different roles of liver biopsy in diagnosis and management of early and progressed HCC. The potential role of liver biopsy for the development of molecular markers to predict prognosis and response to targeted therapy is also discussed.
Collapse
Affiliation(s)
- Haeryoung Kim
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi-do, Republic of Korea.
| | - Young Nyun Park
- Department of Pathology, Brain Korea 21 PLUS Project for Medical Science, and Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
716
|
Itzel T, Scholz P, Maass T, Krupp M, Marquardt JU, Strand S, Becker D, Staib F, Binder H, Roessler S, Wang XW, Thorgeirsson S, Müller M, Galle PR, Teufel A. Translating bioinformatics in oncology: guilt-by-profiling analysis and identification of KIF18B and CDCA3 as novel driver genes in carcinogenesis. ACTA ACUST UNITED AC 2014; 31:216-24. [PMID: 25236463 DOI: 10.1093/bioinformatics/btu586] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
MOTIVATION Co-regulated genes are not identified in traditional microarray analyses, but may theoretically be closely functionally linked [guilt-by-association (GBA), guilt-by-profiling]. Thus, bioinformatics procedures for guilt-by-profiling/association analysis have yet to be applied to large-scale cancer biology. We analyzed 2158 full cancer transcriptomes from 163 diverse cancer entities in regard of their similarity of gene expression, using Pearson's correlation coefficient (CC). Subsequently, 428 highly co-regulated genes (|CC| ≥ 0.8) were clustered unsupervised to obtain small co-regulated networks. A major subnetwork containing 61 closely co-regulated genes showed highly significant enrichment of cancer bio-functions. All genes except kinesin family member 18B (KIF18B) and cell division cycle associated 3 (CDCA3) were of confirmed relevance for tumor biology. Therefore, we independently analyzed their differential regulation in multiple tumors and found severe deregulation in liver, breast, lung, ovarian and kidney cancers, thus proving our GBA hypothesis. Overexpression of KIF18B and CDCA3 in hepatoma cells and subsequent microarray analysis revealed significant deregulation of central cell cycle regulatory genes. Consistently, RT-PCR and proliferation assay confirmed the role of both genes in cell cycle progression. Finally, the prognostic significance of the identified KIF18B- and CDCA3-dependent predictors (P = 0.01, P = 0.04) was demonstrated in three independent HCC cohorts and several other tumors. In summary, we proved the efficacy of large-scale guilt-by-profiling/association strategies in oncology. We identified two novel oncogenes and functionally characterized them. The strong prognostic importance of downstream predictors for HCC and many other tumors indicates the clinical relevance of our findings. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Timo Itzel
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Peter Scholz
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Thorsten Maass
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Markus Krupp
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Jens U Marquardt
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Susanne Strand
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Diana Becker
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Frank Staib
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Harald Binder
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Stephanie Roessler
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Xin Wei Wang
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Snorri Thorgeirsson
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Martina Müller
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Peter R Galle
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| | - Andreas Teufel
- Department of Medicine I, University of Regensburg, 93053, Regensburg, Department of Medicine I, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, 55131, Mainz, Department of Pathology, University of Heidelberg, 69120, Germany and Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD, USA
| |
Collapse
|
717
|
Parpart S, Roessler S, Dong F, Rao V, Takai A, Ji J, Qin L, Ye Q, Jia H, Tang Z, Wang XW. Modulation of miR-29 expression by α-fetoprotein is linked to the hepatocellular carcinoma epigenome. Hepatology 2014; 60:872-83. [PMID: 24798303 PMCID: PMC4146718 DOI: 10.1002/hep.27200] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 04/29/2014] [Indexed: 01/09/2023]
Abstract
UNLABELLED Globally, hepatocellular carcinoma (HCC) accounts for 70%-85% of primary liver cancers and ranks as the second leading cause of male cancer death. Serum alpha-fetoprotein (AFP), normally highly expressed in the liver only during fetal development, is reactivated in 60% of HCC tumors and associated with poor patient outcome. We hypothesize that AFP+ and AFP- tumors differ biologically. Multivariable analysis in 237 HCC cases demonstrates that AFP level predicts poor survival independent of tumor stage (P<0.043). Using microarray-based global microRNA (miRNA) profiling, we found that miRNA-29 (miR-29) family members were the most significantly (P<0.001) down-regulated miRNAs in AFP+ tumors. Consistent with miR-29's role in targeting DNA methyltransferase 3A (DNMT3A), a key enzyme regulating DNA methylation, we found a significant inverse correlation (P<0.001) between miR-29 and DNMT3A gene expression, suggesting that they might be functionally antagonistic. Moreover, global DNA methylation profiling reveals that AFP+ and AFP- HCC tumors have distinct global DNA methylation patterns and that increased DNA methylation is associated with AFP+ HCC. Experimentally, we found that AFP expression in AFP- HCC cells induces cell proliferation, migration, and invasion. Overexpression of AFP, or conditioned media from AFP+ cells, inhibits miR-29a expression and induces DNMT3A expression in AFP- HCC cells. AFP also inhibited transcription of the miR-29a/b-1 locus, and this effect is mediated through c-MYC binding to the transcript of miR-29a/b-1. Furthermore, AFP expression promotes tumor growth of AFP- HCC cells in nude mice. CONCLUSION Tumor biology differs considerably between AFP+ HCC and AFP- HCC; AFP is a functional antagonist of miR-29, which may contribute to global epigenetic alterations and poor prognosis in HCC.
Collapse
Affiliation(s)
- Sonya Parpart
- Laboratory of Human Carcinogenesis, NCI, Bethesda, MD,Tumor Biology Department, Georgetown University, Washington, DC
| | | | - Fei Dong
- Laboratory of Human Carcinogenesis, NCI, Bethesda, MD
| | - Vinay Rao
- Laboratory of Human Carcinogenesis, NCI, Bethesda, MD
| | - Atsushi Takai
- Laboratory of Human Carcinogenesis, NCI, Bethesda, MD
| | - Junfang Ji
- Laboratory of Human Carcinogenesis, NCI, Bethesda, MD
| | - Lun–Xiu Qin
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing–Hai Ye
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hu–Liang Jia
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhao–You Tang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, NCI, Bethesda, MD,Correspondence: Xin Wei Wang, National Cancer Institute, 37 Convent Drive, Bethesda, Maryland 20892; ; Phone: 301-496-2099; Fax: 301-496-0497
| |
Collapse
|
718
|
Zheng Y, Gery S, Sun H, Shacham S, Kauffman M, Koeffler HP. KPT-330 inhibitor of XPO1-mediated nuclear export has anti-proliferative activity in hepatocellular carcinoma. Cancer Chemother Pharmacol 2014; 74:487-95. [PMID: 25030088 PMCID: PMC4146741 DOI: 10.1007/s00280-014-2495-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 05/22/2014] [Indexed: 12/14/2022]
Abstract
PURPOSE Exportin-1 (XPO1, CRM1) mediates the nuclear export of several key growth regulatory and tumor suppressor proteins. Cancer cells often overexpress XPO1 resulting in cytoplasmic mislocalization and aberrant activity of its target proteins. Orally bioavailable selective inhibitors of nuclear export (SINE) that irreversibly bind to and inhibit the function of XPO1 have been recently developed. The aim of this study was to investigate the efficacy of the clinical staged, orally available, SINE compound, KPT-330 in hepatocellular carcinoma (HCC). METHODS In silico, meta-analysis showed that XPO1 is overexpressed in HCC. Six HCC cell lines were treated with KPT-330, and cell proliferation and expression of cell growth regulators were examined by cell proliferation assays and Western blot analysis, respectively. The in vivo anti-cancer activity of KPT-330 was examined in a HCC xenograft murine model. RESULTS KPT-330 reduced the viability of HCC cell lines in vitro and this anti-proliferative effect was associated with cell cycle arrest and induction of apoptosis. The expression of the pro-apoptotic protein PUMA was markedly up-regulated by KPT-330. In addition, SINE treatment increased the expression of the tumor suppressor proteins p53 and p27, while it reduced the expression of HCC promoting proteins, c-Myc and c-Met. XPO1 levels itself were also down-regulated following KPT-330 treatment. Finally, a HCC xenograft murine model showed that treatment of mice with oral KPT-330 significantly inhibited tumor growth with little evidence of toxicity. CONCLUSION Our results suggest that SINE compounds, such as KPT-330, are promising novel drugs for the targeted therapy of HCC.
Collapse
Affiliation(s)
- Yun Zheng
- Cedars-Sinai Medical Center, UCLA School of Medicine Los Angeles, CA
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology, Collaborative Innovation Center for Cancer Medicine China
| | - Sigal Gery
- Cedars-Sinai Medical Center, UCLA School of Medicine Los Angeles, CA
| | - Haibo Sun
- Cedars-Sinai Medical Center, UCLA School of Medicine Los Angeles, CA
| | | | | | - H. Phillip Koeffler
- Cedars-Sinai Medical Center, UCLA School of Medicine Los Angeles, CA
- NCIS, CSI at National University of Singapore, Singapore
| |
Collapse
|
719
|
Huang D, Li T, Li X, Zhang L, Sun L, He X, Zhong X, Jia D, Song L, Semenza G, Gao P, Zhang H. HIF-1-Mediated Suppression of Acyl-CoA Dehydrogenases and Fatty Acid Oxidation Is Critical for Cancer Progression. Cell Rep 2014; 8:1930-1942. [DOI: 10.1016/j.celrep.2014.08.028] [Citation(s) in RCA: 271] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/16/2014] [Accepted: 08/13/2014] [Indexed: 02/05/2023] Open
|
720
|
Budhu A, Terunuma A, Zhang G, Hussain SP, Ambs S, Wang XW. Metabolic profiles are principally different between cancers of the liver, pancreas and breast. Int J Biol Sci 2014; 10:966-72. [PMID: 25210494 PMCID: PMC4159687 DOI: 10.7150/ijbs.9810] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/14/2014] [Indexed: 02/04/2023] Open
Abstract
Molecular profiling of primary tumors may facilitate the classification of patients with cancer into more homogenous biological groups to aid clinical management. Metabolomic profiling has been shown to be a powerful tool in characterizing the biological mechanisms underlying a disease but has not been evaluated for its ability to classify cancers by their tissue of origin. Thus, we assessed metabolomic profiling as a novel tool for multiclass cancer characterization. Global metabolic profiling was employed to identify metabolites in paired tumor and non-tumor liver (n=60), breast (n=130) and pancreatic (n=76) tissue specimens. Unsupervised principal component analysis showed that metabolites are principally unique to each tissue and cancer type. Such a difference can also be observed even among early stage cancers, suggesting a significant and unique alteration of global metabolic pathways associated with each cancer type. Our global high-throughput metabolomic profiling study shows that specific biochemical alterations distinguish liver, pancreatic and breast cancer and could be applied as cancer classification tools to differentiate tumors based on tissue of origin.
Collapse
Affiliation(s)
| | | | - Geng Zhang
- 3. Pancreatic Cancer Unit, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - S Perwez Hussain
- 3. Pancreatic Cancer Unit, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | | |
Collapse
|
721
|
Ding QS, Sun RZ, Wang XC, Shen SQ. Analysis of clinical significance of expression of 6-phosphofructo-2-kinase in hepatocellular carcinoma utilizing GEO datasets. Shijie Huaren Xiaohua Zazhi 2014; 22:3675-3680. [DOI: 10.11569/wcjd.v22.i24.3675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of 6-phosphofructo-2-kinase (PFKFB3) in multiple tumors, to clarify the relationship between PFKFB3 expression and clinicopathological characteristics of hepatocellular carcinoma, and to evaluate the possibility of using PFKFB3 as a prognosis marker in hepatocellular carcinoma.
METHODS: GEO datasets were collected and expression profile and clinical information were analyzed. Gene set enrichment analysis (GSEA) was conducted to explore the gene sets enriched in samples with high PFKFB3 expression.
RESULTS: The expression of PFKFB3 was up-regulated in high grade tumors (P < 0.05). PFKFB3 expression was significantly associated with age, alanine transaminase (ALT), tumor volume, the number of nodules, T stage, Barcelona Clinic Liver Cancer (BCLC) stage and Cancer Liver Italian Program (CLIP) stage. Higher expression of PFKFB3 indicated poor prognosis in hepatocellular carcinoma. GSEA indicated that PFKFB3 regulates gene sets associated with anti-apoptosis, negative regulation of programmed cell death, and positive regulation of cell proliferation and cellular metabolic process.
CONCLUSION: PFKFB3 is highly expressed in multiple tumors and functions as a potential marker and target for diagnosis and treatment of hepatocellular carcinoma.
Collapse
|
722
|
Li J, Gao JZ, Du JL, Huang ZX, Wei LX. Increased CDC20 expression is associated with development and progression of hepatocellular carcinoma. Int J Oncol 2014; 45:1547-55. [PMID: 25069850 DOI: 10.3892/ijo.2014.2559] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/22/2014] [Indexed: 11/05/2022] Open
Abstract
Cell division cycle 20 (CDC20) encodes a regulatory protein interacting with the anaphase-promoting complex/cyclosome (APC/C) in the cell cycle and plays important roles in tumorigenesis and progression of multiple tumors. The present study aimed to investigate the clinical significance of CDC20 in hepatocellular carcinoma (HCC) and the role of CDC20 in the progression of HCC. By bioinformatics analysis, CDC20 was found to be the major node in HCC molecular interaction networks. Quantitative PCR and western blot analyses were applied to examine CDC20 expression in 16 paired primary HCC tissues. Immunohistochemistry (IHC) was performed to examine CDC20 protein expression in 132 matched paraffin-embedded HCC tissues and to analyze the relationship between CDC20 staining and clinical characteristics. Small interfering RNA (siRNA) targeting CDC20 was synthesized and transfected into HepG2 cells to investigate the role of CDC20 in cell growth and the cell cycle. Results show that CDC20 expression was upregulated in HCC tissues compared to adjacent non-tumor liver tissues. In the 132 matched HCC tissues, high expression levels of CDC20 were detected in 68.18% HCC samples, and overexpression of CDC20 was positively correlated with gender (P=0.013), tumor differentiation (P=0.000), TNM stage (P=0.012), P53 and Ki-67 expression (P=0.023 and P=0.007, respectively). Cells transfected with CDC20 siRNA showed a decrease in cell proliferation and increase in the number of cells in G2/M-phase. In conclusion, increased expression of CDC20 was demonstrated to be associated with the development and progression of HCC, and may be regarded as a promising therapeutic target for HCC.
Collapse
Affiliation(s)
- Jia Li
- Department of Pathology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jian-Zhi Gao
- Department of Pathology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jing-Li Du
- Department of Pathology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Zhong-Xi Huang
- Cancer Institute, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Li-Xin Wei
- Department of Pathology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
723
|
Wong YH, Chen RH, Chen BS. Core and specific network markers of carcinogenesis from multiple cancer samples. J Theor Biol 2014; 362:17-34. [PMID: 25016045 DOI: 10.1016/j.jtbi.2014.05.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 05/19/2014] [Accepted: 05/28/2014] [Indexed: 01/07/2023]
Abstract
Cancer is the leading cause of death worldwide and is generally caused by mutations in multiple proteins or the dysregulation of pathways. Understanding the causes and the underlying carcinogenic mechanisms can help fight this disease. In this study, a systems biology approach was used to construct the protein-protein interaction (PPI) networks of four cancers and the non-cancers by their corresponding microarray data, PPI modeling and database-mining. By comparing PPI networks between cancer and non-cancer samples to find significant proteins with large PPI changes during carcinogenesis process, core and specific network markers were identified by the intersection and difference of significant proteins, respectively, with carcinogenesis relevance values (CRVs) for each cancer. A total of 28 significant proteins were identified as core network markers in the carcinogenesis of four types of cancer, two of which are novel cancer-related proteins (e.g., UBC and PSMA3). Moreover, seven crucial common pathways were found among these cancers based on their core network markers, and some specific pathways were particularly prominent based on the specific network markers of different cancers (e.g., the RIG-I-like receptor pathway in bladder cancer, the proteasome pathway and TCR pathway in liver cancer, and the HR pathway in lung cancer). Additional validation of these network markers using the literature and new tested datasets could strengthen our findings and confirm the proposed method. From these core and specific network markers, we could not only gain an insight into crucial common and specific pathways in the carcinogenesis, but also obtain a high promising PPI target for cancer therapy.
Collapse
Affiliation(s)
- Yung-Hao Wong
- Lab of Control and Systems Biology, Department of Electrical Engineering National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Ru-Hong Chen
- Lab of Control and Systems Biology, Department of Electrical Engineering National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Bor-Sen Chen
- Lab of Control and Systems Biology, Department of Electrical Engineering National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
724
|
Malouf GG, Job S, Paradis V, Fabre M, Brugières L, Saintigny P, Vescovo L, Belghiti J, Branchereau S, Faivre S, de Reyniès A, Raymond E. Transcriptional profiling of pure fibrolamellar hepatocellular carcinoma reveals an endocrine signature. Hepatology 2014; 59:2228-37. [PMID: 24443104 DOI: 10.1002/hep.27018] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 01/14/2014] [Indexed: 12/16/2022]
Abstract
UNLABELLED Fibrolamellar hepatocellular carcinoma (FLC) is a rare subtype of liver cancer occurring mostly in children and young adults. We have shown that FLC comprises two separate entities: pure (p-FLC) and mixed-FLC (m-FLC), differing in clinical presentation and course. We show that p-FLCs have a distinct gene expression signature different from that of m-FLCs, which have a signature similar to that of classical hepatocellular carcinomas. We found p-FLC profiles to be unique among 263 profiles related to diverse tumoral and nontumoral liver samples. We identified two distinct molecular subgroups of p-FLCs with different outcomes. Pathway analysis of p-FLCs revealed ERBB2 overexpression and an up-regulation of glycolysis, possibly leading to compensatory mitochondrial hyperplasia and oncocytic differentiation. Four of the sixteen genes most significantly overexpressed in p-FLCs were neuroendocrine genes: prohormone convertase 1 (PCSK1); neurotensin; delta/notch-like EGF repeat containing; and calcitonin. PCSK1 overexpression was validated by immunohistochemistry, yielding specific, diffuse staining of the protein throughout the cytoplasm, possibly corresponding to a functional form of this convertase. CONCLUSION p-FLCs have a unique transcriptomic signature characterized by the strong expression of specific neuroendocrine genes, suggesting that these tumors may have a cellular origin different from that of HCC. Our data have implications for the use of genomic profiling for diagnosis and selection of targeted therapies in patients with p-FLC.
Collapse
Affiliation(s)
- Gabriel G Malouf
- Department of Medical Oncology, Beaujon University Hospital and INSERM U728, Clichy, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
725
|
Colecchia A, Schiumerini R, Cucchetti A, Cescon M, Taddia M, Marasco G, Festi D. Prognostic factors for hepatocellular carcinoma recurrence. World J Gastroenterol 2014; 20:5935-5950. [PMID: 24876717 PMCID: PMC4033434 DOI: 10.3748/wjg.v20.i20.5935] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/14/2013] [Accepted: 04/29/2014] [Indexed: 02/06/2023] Open
Abstract
The recurrence of hepatocellular carcinoma, the sixth most common neoplasm and the third leading cause of cancer-related mortality worldwide, represents an important clinical problem, since it may occur after both surgical and medical treatment. The recurrence rate involves 2 phases: an early phase and a late phase. The early phase usually occurs within 2 years after resection; it is mainly related to local invasion and intrahepatic metastases and, therefore, to the intrinsic biology of the tumor. On the other hand, the late phase occurs more than 2 years after surgery and is mainly related to de novo tumor formation as a consequence of the carcinogenic cirrhotic environment. Since recent studies have reported that early and late recurrences may have different risk factors, it is clinically important to recognize these factors in the individual patient as soon as possible. The aim of this review was, therefore, to identify predicting factors for the recurrence of hepatocellular carcinoma, by means of invasive and non-invasive methods, according to the different therapeutic strategies available. In particular the role of emerging techniques (e.g., transient elastography) and biological features of hepatocellular carcinoma in predicting recurrence have been discussed. In particular, invasive methods were differentiated from non-invasive ones for research purposes, taking into consideration the emerging role of the genetic signature of hepatocellular carcinoma in order to better allocate treatment strategies and surveillance follow-up in patients with this type of tumor.
Collapse
|
726
|
Lin CW, Sun MS, Wu HC. Podocalyxin-like 1 is associated with tumor aggressiveness and metastatic gene expression in human oral squamous cell carcinoma. Int J Oncol 2014; 45:710-8. [PMID: 24821609 DOI: 10.3892/ijo.2014.2427] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 03/07/2014] [Indexed: 11/05/2022] Open
Abstract
Metastasis-mediated death remains a major challenge in cancer treatment due to the lack of identifiable biomarkers for early diagnosis. Identifying tumor-specific biomarkers is critical for the development of diagnostic and therapeutic tools. In the present study, we found that podocalyxin-like 1 (PODXL), a cell surface glycoprotein, was overexpressed in cancer tissues and was upregulated in lymph node metastatic tumor cells. The expression of PODXL was associated with the migratory ability of human oral squamous cell carcinoma (OSCC). Knockdown of PODXL by small hairpin RNA in the SAS OSCC cell line reduced tumor migration and invasion, and inhibited cell proliferation and colony formation. Suppression of PODXL resulted in downregulation of focal adhesion kinase (FAK) and paxillin phosphorylation. PODXL silencing inhibited filopodia formation, and suppressed F-actin and cortactin colocalization. In addition, PODXL expression was associated with the DNA methylation status, and treatment with the DNA methyltransferase inhibitor 5-aza-deoxycytidine increased the PODXL transcriptional level. Moreover, DNA microarray analysis data revealed that suppression of PODXL significantly affected subsets of genes associated with extracellular matrix organization, the epithelial-mesenchymal transition, and the expression of metastasis-related cytokines. Collectively, these data showed that the overexpression of PODXL may be associated with tumor aggressiveness and that PODXL could be a diagnostic biomarker for metastatic OSCC.
Collapse
Affiliation(s)
- Cheng-Wei Lin
- Department of Biochemistry, School of Medicine, Taipei Medical University, Taipei 115, Taiwan, R.O.C
| | - Min-Siou Sun
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan, R.O.C
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan, R.O.C
| |
Collapse
|
727
|
Liu C, Xia Y, Jiang W, Liu Y, Yu L. Low expression of GABARAPL1 is associated with a poor outcome for patients with hepatocellular carcinoma. Oncol Rep 2014; 31:2043-8. [PMID: 24647565 DOI: 10.3892/or.2014.3096] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 12/27/2013] [Indexed: 12/19/2022] Open
Abstract
Autophagy is an evolutionarily conserved cellular process that degrades cytoplasmic materials through the lysosomal pathway. The deregulation of autophagy is associated with several diseases, particularly cancer. Hepatocellular carcinoma (HCC) is one of the most aggressive cancers with a poor prognosis. The expression of autophagy-related genes in HCC and their relationships with HCC are largely unknown. In the present study, we analyzed the expression of autophagy-related genes based on the Oncomine database and quantitative PCR of HCC and adjacent liver tissues. We found that the mRNA and protein expression of GABARAPL1 was significantly decreased in HCC tissues compared with their adjacent liver tissues. In HCC cancer cell lines, overexpression of GABARAPL1 inhibited cell growth, while knockdown of GABARAPL1 expression via siRNA promoted cell growth. In addition, we found a significant correlation of low GABARAPL1 expression with poor differentiation of HCC cells (P=0.018), and with the absence of tumor capsules (P=0.047). Kaplan-Meier survival analysis showed a significant association between low GABARAPL1 expression and poor prognosis of HCC patients (P=0.0094). Our data showed for the first time that GABARAPL1 expression is associated with poor prognosis of HCC patients.
Collapse
Affiliation(s)
- Chao Liu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China
| | - Yan Xia
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China
| | - Wei Jiang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China
| | - Yinkun Liu
- Liver Cancer Institute, Fudan University, Shanghai 200032, P.R. China
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, P.R. China
| |
Collapse
|
728
|
TACC3 deregulates the DNA damage response and confers sensitivity to radiation and PARP inhibition. Oncogene 2014; 34:1667-78. [PMID: 24769898 DOI: 10.1038/onc.2014.105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 02/28/2014] [Accepted: 03/12/2014] [Indexed: 12/21/2022]
Abstract
Deregulation of the transforming acidic coiled-coil protein 3 (TACC3), an important factor in the centrosome-microtubule system, has been linked to a variety of human cancer types. We have recently reported on the oncogenic potential of TACC3; however, the molecular mechanisms by which TACC3 mediates oncogenic function remain to be elucidated. In this study, we show that high levels of TACC3 lead to the accumulation of DNA double-strand breaks (DSBs) and disrupt the normal cellular response to DNA damage, at least in part, by negatively regulating the expression of ataxia telangiectasia mutated (ATM) and the subsequent DNA damage response (DDR) signaling cascade. Cells expressing high levels of TACC3 display defective checkpoints and DSB-mediated homologous recombination (HR) and non-homologous end joining (NHEJ) repair systems, leading to genomic instability. Importantly, high levels of TACC3 confer cellular sensitization to radiation and poly(ADP-ribose) polymerase (PARP) inhibition. Overall, our findings provide critical information regarding the mechanisms by which TACC3 contributes to genomic instability, potentially leading to cancer development, and suggest a novel prognostic, diagnostic and therapeutic strategy for the treatment of cancer types expressing high levels of TACC3.
Collapse
|
729
|
Roessler S, Budhu A, Wang XW. Deciphering cancer heterogeneity: the biological space. Front Cell Dev Biol 2014; 2:12. [PMID: 25364720 PMCID: PMC4207029 DOI: 10.3389/fcell.2014.00012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 03/17/2014] [Indexed: 01/15/2023] Open
Abstract
Most lethal solid tumors including hepatocellular carcinoma (HCC) are considered incurable due to extensive heterogeneity in clinical presentation and tumor biology. Tumor heterogeneity may result from different cells of origin, patient ethnicity, etiology, underlying disease, and diversity of genomic and epigenomic changes which drive tumor development. Cancer genomic heterogeneity thereby impedes treatment options and poses a significant challenge to cancer management. Studies of the HCC genome have revealed that although various genomic signatures identified in different HCC subgroups share a common prognosis, each carries unique molecular changes which are linked to different sets of cancer hallmarks whose misregulation has been proposed by Hanahan and Weinberg to be essential for tumorigenesis. We hypothesize that these specific sets of cancer hallmarks collectively occupy different tumor biological space representing the misregulation of different biological processes. In principle, a combination of different cancer hallmarks can result in new convergent molecular networks that are unique to each tumor subgroup and represent ideal druggable targets. Due to the ability of the tumor to adapt to external factors such as treatment or changes in the tumor microenvironment, the tumor biological space is elastic. Our ability to identify distinct groups of cancer patients with similar tumor biology who are most likely to respond to a specific therapy would have a significant impact on improving patient outcome. It is currently a challenge to identify a particular hallmark or a newly emerged convergent molecular network for a particular tumor. Thus, it is anticipated that the integration of multiple levels of data such as genomic mutations, somatic copy number aberration, gene expression, proteomics, and metabolomics, may help us grasp the tumor biological space occupied by each individual, leading to improved therapeutic intervention and outcome.
Collapse
Affiliation(s)
- Stephanie Roessler
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute Bethesda, MD, USA
| | - Anuradha Budhu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute Bethesda, MD, USA
| | - Xin W Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute Bethesda, MD, USA
| |
Collapse
|
730
|
Lee SC, Tan HT, Chung MCM. Prognostic biomarkers for prediction of recurrence of hepatocellular carcinoma: Current status and future prospects. World J Gastroenterol 2014; 20:3112-3124. [PMID: 24696598 PMCID: PMC3964383 DOI: 10.3748/wjg.v20.i12.3112] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 12/03/2013] [Accepted: 01/02/2014] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide, with region specific etiologies. Despite improvements made in the diagnosis of HCC, the prognosis of HCC patients remains poor due to the high recurrence rate of HCC. There is an urgent need for development of prognostic biomarkers to predict the risk of recurrence in HCC patients after “curative” treatment. Such stratification may aid in patient management and development of personalized medicine for HCC treatment. Omics based studies facilitate the study of global changes in biomolecules in a disease in a high throughput manner, and hence are well poised to understand the complex changes which led to HCC recurrence. The quantitative nature of data obtained from omics based studies allow for development of prognostic biomarkers based on changes in gene, protein and metabolite expression. In this review, we surveyed the application of transcriptomics, proteomics and metabolomics in the study of HCC recurrence. We summarised the data in the literature from these three fields of studies that claimed to be prognostic for HCC recurrence. We critiqued on the limitations of each area of research and the challenges faced in translating the research results for clinical application in predicting HCC recurrence.
Collapse
|
731
|
Zheng W, Li Z, Nguyen AT, Li C, Emelyanov A, Gong Z. Xmrk, kras and myc transgenic zebrafish liver cancer models share molecular signatures with subsets of human hepatocellular carcinoma. PLoS One 2014; 9:e91179. [PMID: 24633177 PMCID: PMC3954698 DOI: 10.1371/journal.pone.0091179] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 02/09/2014] [Indexed: 12/11/2022] Open
Abstract
Previously three oncogene transgenic zebrafish lines with inducible expression of xmrk, kras or Myc in the liver have been generated and these transgenic lines develop oncogene-addicted liver tumors upon chemical induction. In the current study, comparative transcriptomic approaches were used to examine the correlation of the three induced transgenic liver cancers with human liver cancers. RNA profiles from the three zebrafish tumors indicated relatively small overlaps of significantly deregulated genes and biological pathways. Nevertheless, the three transgenic tumor signatures all showed significant correlation with advanced or very advanced human hepatocellular carcinoma (HCC). Interestingly, molecular signature from each oncogene-induced zebrafish liver tumor correlated with only a small subset of human HCC samples (24–29%) and there were conserved up-regulated pathways between the zebrafish and correlated human HCC subgroup. The three zebrafish liver cancer models together represented nearly half (47.2%) of human HCCs while some human HCCs showed significant correlation with more than one signature defined from the three oncogene-addicted zebrafish tumors. In contrast, commonly deregulated genes (21 up and 16 down) in the three zebrafish tumor models generally showed accordant deregulation in the majority of human HCCs, suggesting that these genes might be more consistently deregulated in a broad range of human HCCs with different molecular mechanisms and thus serve as common diagnosis markers and therapeutic targets. Thus, these transgenic zebrafish models with well-defined oncogene-induced tumors are valuable tools for molecular classification of human HCCs and for understanding of molecular drivers in hepatocarcinogenesis in each human HCC subgroup.
Collapse
Affiliation(s)
- Weiling Zheng
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Zhen Li
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Anh Tuan Nguyen
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Caixia Li
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | | | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
732
|
Jia RJ, Cao L, Zhang L, Jing W, Chen R, Zhu MH, Guo SW, Wu GB, Fan XY, Wang H, Zhang YY, Zhou XY, Zhao J, Guo YJ. Enhanced myeloid differentiation factor 88 promotes tumor metastasis via induction of epithelial-mesenchymal transition in human hepatocellular carcinoma. Cell Death Dis 2014; 5:e1103. [PMID: 24603331 PMCID: PMC3973199 DOI: 10.1038/cddis.2014.71] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/27/2014] [Accepted: 01/28/2014] [Indexed: 02/07/2023]
Abstract
Metastasis is the leading cause of death in patients with hepatocellular carcinoma (HCC) after curative resection. Therefore, it is critical to understand the mechanisms underlying tumor metastasis in HCC. We have previously shown that elevated expression of myeloid differentiation factor 88 (MyD88) may promote tumor growth and metastasis in HCC. In this study, we reported that enhanced expression of MyD88 promoted epithelial–mesenchymal transition (EMT) properties and tumor-initiating capabilities in HCC cells. MyD88 was found to be able to interact with p85, a regulatory subunit of phosphoinositide 3-kinase (PI3-K), independent of TLR/IL-1R-mediated response and caused PI3-K/v-akt murine thymoma viral oncogene homolog (Akt) activation, which resulted in subsequent phosphorylation of glycogen synthase kinase-3β and stabilization of Snail, a critical EMT mediator. Consistently, we observed a significant correlation between MyD88 expression and p-Akt levels in a cohort of HCC patients, and found that the combination of these two parameters have better prognostic value for HCC patients. Taken together, these results suggest that elevated MyD88 may facilitate HCC metastasis by promoting EMT properties and tumor-initiating capabilities via PI3–K/Akt pathway.
Collapse
Affiliation(s)
- R-J Jia
- 1] International Joint Cancer Institute, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China [2] School of Pharmacy, Liaocheng University, 1 Hunan Road, Liaocheng 252059, People's Republic of China
| | - L Cao
- 1] International Joint Cancer Institute, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China [2] School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai 200025, People's Republic of China
| | - L Zhang
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China
| | - W Jing
- Changhai Hospital, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - R Chen
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China
| | - M-H Zhu
- Changhai Hospital, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - S-W Guo
- Changhai Hospital, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - G-B Wu
- Guangxi Cancer Hospital, Guangxi Medical University, Nanning 530021, People's Republic of China
| | - X-Y Fan
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China
| | - H Wang
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China
| | - Y-Y Zhang
- School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Road, Shanghai 200025, People's Republic of China
| | - X-Y Zhou
- Changhai Hospital, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - J Zhao
- 1] International Joint Cancer Institute, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China [2] School of Pharmacy, Liaocheng University, 1 Hunan Road, Liaocheng 252059, People's Republic of China [3] PLA General Hospital Cancer Center, PLA postgraduate School of Medicine, 28 Fuxing Road, Beijing 100853, People's Republic of China
| | - Y-J Guo
- 1] International Joint Cancer Institute, The Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, People's Republic of China [2] School of Pharmacy, Liaocheng University, 1 Hunan Road, Liaocheng 252059, People's Republic of China [3] PLA General Hospital Cancer Center, PLA postgraduate School of Medicine, 28 Fuxing Road, Beijing 100853, People's Republic of China [4] National Engineering Research Center of Antibody Medicine and State Key Laboratory of Antibody Medicine and Targeting Therapy, 99 Libing Road, Shanghai 201203, People's Republic of China
| |
Collapse
|
733
|
Qu Y, Li J, Cai Q, Liu B. Hec1/Ndc80 is overexpressed in human gastric cancer and regulates cell growth. J Gastroenterol 2014; 49:408-18. [PMID: 23591767 DOI: 10.1007/s00535-013-0809-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 03/29/2013] [Indexed: 02/04/2023]
Abstract
BACKGROUND Chromosomal instability caused by abnormal cell division is a major cause of heterogeneity which evokes highly complex and malignant features of gastric cancer. Hec1/Ndc80 is critical in regulating proper cell division at the G2/M phase. The aim of our study is to investigate the in vitro and in vivo effects of Hec1 on gastric cancer cell growth. METHODS The mRNA levels of Hec1 in human normal and cancer tissues were analyzed using the Oncomine database. Hec1 mRNA and protein levels in human gastric cancer tissues were analyzed by quantitative realtime-PCR and immunohistochemical staining, respectively. The effects of Hec1 on cell growth were explored by Hec1 knockdown and Hec1 overexpression. Apoptosis and cell cycle distributions were analyzed by flow cytometry. In vivo tumorigenicity was performed by engrafting tumor cells into nude mice. RESULTS Hec1 mRNA and protein were broadly overexpressed in many human cancers including gastric cancer. Hec1 knockdown dramatically suppressed gastric cancer cell growth in vitro and in vivo, induced apoptosis, and arrested cell division at the G2/M phase. On the contrary, Hec1 overexpression moderately promoted gastric cancer cell growth in vivo. Hec1 overexpression induced asymmetrical chromosome alignments, abnormal cell division, and thus rendered chromosomal instability. CONCLUSIONS Hec1 is critical in maintaining the in vitro and in vivo growth of gastric cancer cells. Elevated Hec1 levels may occur at the early stage of gastric tumorigenesis.
Collapse
Affiliation(s)
- Ying Qu
- Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Department of Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025, Shanghai, People's Republic of China
| | | | | | | |
Collapse
|
734
|
Nault JC. Molecular Determinants of Prognosis in Hepatocellular Carcinoma. J Clin Transl Hepatol 2014; 2:31-6. [PMID: 26357615 PMCID: PMC4521254 DOI: 10.14218/jcth.2013.00029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/25/2014] [Accepted: 01/26/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma is one of the leading causes of death by cancer worldwide. Prognosis of hepatocellular carcinoma is determined by characteristics of the tumor and the surrounding cirrhotic liver. Several molecular signatures reflecting tumor biology and derived from tumor analyses predict early tumor recurrence and survival. In contrast, molecular signatures from cirrhotic non-tumor samples are enriched in immunity/inflammation related genes and could predict late tumor recurrence. Moreover, combination of clinical, pathological, and molecular features may refine prognosis prediction in these patients. Finally, molecular signatures from both tumor and non-tumor tissues will be helpful in the future to guide treatments in different clinical settings.
Collapse
Affiliation(s)
- Jean-Charles Nault
- Inserm, UMR-1162, Génomique fonctionnelle des Tumeurs solides, IUH, Paris, France
- Université Paris Descartes, Labex Immuno-Oncology, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
- Service d’Hépatologie, Hôpital Jean Verdier, AP-HP, Bondy, and Université Paris 13, Bobigny, France
| |
Collapse
|
735
|
Tcherkezian J, Cargnello M, Romeo Y, Huttlin EL, Lavoie G, Gygi SP, Roux PP. Proteomic analysis of cap-dependent translation identifies LARP1 as a key regulator of 5'TOP mRNA translation. Genes Dev 2014; 28:357-71. [PMID: 24532714 PMCID: PMC3937514 DOI: 10.1101/gad.231407.113] [Citation(s) in RCA: 200] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 01/06/2014] [Indexed: 12/20/2022]
Abstract
The mammalian target of rapamycin (mTOR) promotes cell growth and proliferation by promoting mRNA translation and increasing the protein synthetic capacity of the cell. Although mTOR globally promotes translation by regulating the mRNA 5' cap-binding protein eIF4E (eukaryotic initiation factor 4E), it also preferentially regulates the translation of certain classes of mRNA via unclear mechanisms. To help fill this gap in knowledge, we performed a quantitative proteomic screen to identify proteins that associate with the mRNA 5' cap in an mTOR-dependent manner. Using this approach, we identified many potential regulatory factors, including the putative RNA-binding protein LARP1 (La-related protein 1). Our results indicate that LARP1 associates with actively translating ribosomes via PABP and that LARP1 stimulates the translation of mRNAs containing a 5' terminal oligopyrimidine (TOP) motif, encoding for components of the translational machinery. We found that LARP1 associates with the mTOR complex 1 (mTORC1) and is required for global protein synthesis as well as cell growth and proliferation. Together, these data reveal important molecular mechanisms involved in TOP mRNA translation and implicate LARP1 as an important regulator of cell growth and proliferation.
Collapse
Affiliation(s)
- Joseph Tcherkezian
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Marie Cargnello
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Yves Romeo
- Centre National de la Recherche Scientifique, Laboratoire de Biologie Moléculaire Eucaryote, 31062 Toulouse, Cedex 04, France
- Université de Toulouse, Université Paul Sabatier (UPS), F-31000 Toulouse, France
| | - Edward L. Huttlin
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Taplin Biological Mass Spectrometry Facility, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Genevieve Lavoie
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Taplin Biological Mass Spectrometry Facility, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Philippe P. Roux
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec H3C 3J7, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
736
|
Sequential transcriptome analysis of human liver cancer indicates late stage acquisition of malignant traits. J Hepatol 2014; 60:346-353. [PMID: 24512821 PMCID: PMC3943679 DOI: 10.1016/j.jhep.2013.10.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 09/30/2013] [Accepted: 10/09/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Human hepatocarcinogenesis is as a multi-step process starting from dysplastic lesions to early carcinomas (eHCC) that ultimately progress to HCC (pHCC). However, the sequential molecular alterations driving malignant transformation of the pre-neoplastic lesions are not clearly defined. This lack of information represents a major challenge in the clinical management of patients at risk. METHODS We applied next-generation transcriptome sequencing to tumor-free surrounding liver (n = 7), low- (n = 4) and high-grade (n = 9) dysplastic lesions, eHCC (n = 5) and pHCC (n = 3) from 8 HCC patients with hepatitis B infection. Integrative analyses of genetic and transcriptomic changes were performed to characterize the genomic alterations during hepatocarcinogenesis. RESULTS We report that changes in transcriptomes of early lesions including eHCC were modest and surprisingly homogenous. Extensive genetic alterations and subsequent activation of prognostic adverse signaling pathways occurred only late during hepatocarcinogenesis and were centered on TGFβ, WNT, NOTCH, and EMT-related genes highlighting the molecular diversity of pHCC. We further identify IGFALS as a key genetic determinant preferentially down-regulated in pHCC. CONCLUSIONS Our results define new hallmarks in molecular stratification and therapy options for patients at risk for HCC, and merit larger prospective investigations to develop a modified clinical-decision making algorithm based on the individualized next-generation sequencing analyses.
Collapse
|
737
|
Wei L, Lian B, Zhang Y, Li W, Gu J, He X, Xie L. Application of microRNA and mRNA expression profiling on prognostic biomarker discovery for hepatocellular carcinoma. BMC Genomics 2014; 15 Suppl 1:S13. [PMID: 24564407 PMCID: PMC4046763 DOI: 10.1186/1471-2164-15-s1-s13] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most highly malignant and lethal cancers of the world. Its pathogenesis has been reported to be multi-factorial, and the molecular carcinogenesis of HCC can not be attributed to just a few individual genes. Based on the microRNA and mRNA expression profiling of normal liver tissues, pericancerous hepatocellular tissues and hepatocellular carcinoma tissues, we attempted to find prognosis related gene sets for HCC patients. Results We identified differentially expressed genes (DEG) from three comparisons: Cancer/Normal, Cancer/Pericancerous and Pericancerous/Normal. GSEA (gene set enrichment analysis) were performed. Based on the enriched gene sets of GO terms, pathways and transcription factor targets, it was found that the genome instability and cell proliferation increased while the metabolism and differentiation decreased in HCC tissues. The expression profile of DEGs in each enriched gene set was used to correlate to the postoperative survival time of HCC patients. Nine gene sets were found to prognostic correlation. Furthermore, after substituting DEG-targeting-microRNA for DEG members of each gene set, two gene sets with the microRNA expression profiles were obtained that had prognostic potential. Conclusions The malignancy of HCC could be represented by gene sets, and pericancerous liver exhibits important characteristics of liver cancer. The expression level of gene sets not only in HCC but also in the pericancerous liver showed potential for prognosis implying an option for HCC prognosis at an early stage. Additionally, the gene-targeting-microRNA expression profiles also showed prognostic potential, demonstrating that the multi-factorial molecular pathogenesis of HCC is contributed by various genes and microRNAs. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-S1-S13) contains supplementary material, which is available to authorized users.
Collapse
|
738
|
Bard-Chapeau EA, Nguyen AT, Rust AG, Sayadi A, Lee P, Chua BQ, New LS, de Jong J, Ward JM, Chin CK, Chew V, Toh HC, Abastado JP, Benoukraf T, Soong R, Bard FA, Dupuy AJ, Johnson RL, Radda GK, Chan EC, Wessels LF, Adams DJ, Jenkins NA, Copeland NG. Transposon mutagenesis identifies genes driving hepatocellular carcinoma in a chronic hepatitis B mouse model. Nat Genet 2014; 46:24-32. [PMID: 24316982 PMCID: PMC4131144 DOI: 10.1038/ng.2847] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 11/08/2013] [Indexed: 12/15/2022]
Abstract
The most common risk factor for developing hepatocellular carcinoma (HCC) is chronic infection with hepatitis B virus (HBV). To better understand the evolutionary forces driving HCC, we performed a near-saturating transposon mutagenesis screen in a mouse HBV model of HCC. This screen identified 21 candidate early stage drivers and a very large number (2,860) of candidate later stage drivers that were enriched for genes that are mutated, deregulated or functioning in signaling pathways important for human HCC, with a striking 1,199 genes being linked to cellular metabolic processes. Our study provides a comprehensive overview of the genetic landscape of HCC.
Collapse
Affiliation(s)
- Emilie A Bard-Chapeau
- Institute Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| | - Anh-Tuan Nguyen
- Institute Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| | - Alistair G Rust
- Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Ahmed Sayadi
- Institute Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| | - Philip Lee
- Clinical Imaging Research Centre, National University of Singapore, Centre for Translation Medicine, 14 Medical Drive, #B1-01, 117599, Singapore
| | - Belinda Q Chua
- Institute Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| | - Lee-Sun New
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, 117543, Singapore
| | - Johann de Jong
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam
| | - Jerrold M Ward
- Institute Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| | - Christopher Ky Chin
- Institute Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| | - Valerie Chew
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| | | | - Jean-Pierre Abastado
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| | - Touati Benoukraf
- Cancer Science Institute of Singapore, National University of Singapore, Department of Pathology, National University Health System, Singapore
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Department of Pathology, National University Health System, Singapore
| | - Frederic A Bard
- Institute Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| | - Adam J Dupuy
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242
| | - Randy L Johnson
- Department of Biochemistry and Molecular Biology, University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - George K Radda
- Clinical Imaging Research Centre, National University of Singapore, Centre for Translation Medicine, 14 Medical Drive, #B1-01, 117599, Singapore
| | - Eric Cy Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, 117543, Singapore
| | - Lodewyk Fa Wessels
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Nancy A Jenkins
- Institute Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| | - Neal G Copeland
- Institute Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore
| |
Collapse
|
739
|
Abstract
In the past decade, considerable effort has been made in elucidating the mechanism underlying the high level of aerobic glycolysis in cancer cells. While some recent studies have attempted to address this issue, the potential role of microRNAs in this process has not been explored until recently. These studies have demonstrated involvement of just five deregulated miRNAs in glucose metabolism in hepatocarcinogenesis. This review discusses the metabolic significance of these miRNAs in hepatoceullular carcinoma, their targets in glycolysis, gluconeogenesis, and pentose phosphate pathways, and provides an insight into the therapeutic potential of targeting specific miRNAs.
Collapse
Affiliation(s)
- Ryan K. Reyes
- *Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- †Department of Molecular and Cellular Biochemistry, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Tasneem Motiwala
- †Department of Molecular and Cellular Biochemistry, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
- ‡Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Samson T. Jacob
- †Department of Molecular and Cellular Biochemistry, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
- ‡Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
740
|
Jia D, Jing Y, Zhang Z, Liu L, Ding J, Zhao F, Ge C, Wang Q, Chen T, Yao M, Li J, Gu J, He X. Amplification of MPZL1/PZR promotes tumor cell migration through Src-mediated phosphorylation of cortactin in hepatocellular carcinoma. Cell Res 2013; 24:204-17. [PMID: 24296779 DOI: 10.1038/cr.2013.158] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 09/10/2013] [Accepted: 09/25/2013] [Indexed: 12/15/2022] Open
Abstract
We have previously identified 1 241 regions of somatic copy number alterations (CNAs) in hepatocellular carcinoma (HCC). In the present study, we found that a novel recurrent focal amplicon, 1q24.1-24.2, targets the MPZL1 gene in HCC. Notably, there is a positive correlation between the expression levels of MPZL1 and intrahepatic metastasis of the HCC specimens. MPZL1 can significantly enhance the migratory and metastatic potential of the HCC cells. Moreover, we found that one of the mechanisms by which MPZL1 promotes HCC cell migration is by inducing the phosphorylation and activation of the pro-metastatic protein, cortactin. Additionally, we found that Src kinase mediates the phosphorylation and activation of cortactin induced by MPZL1 overexpression. Taken together, these findings suggest that MPZL1 is a novel pro-metastatic gene targeted by a recurrent region of copy number amplification at 1q24.1-24.2 in HCC.
Collapse
Affiliation(s)
- Deshui Jia
- 1] State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China [2] Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ying Jing
- 1] State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China [2] Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhenfeng Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Li Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Jie Ding
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Fangyu Zhao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Chao Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Qifeng Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Taoyang Chen
- Qidong Liver Cancer Institute, Qidong, Jiangsu 226200, China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Jinjun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Jianren Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Xianghuo He
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| |
Collapse
|
741
|
Tennstedt P, Bölch C, Strobel G, Minner S, Burkhardt L, Grob T, Masser S, Sauter G, Schlomm T, Simon R. Patterns of TPD52 overexpression in multiple human solid tumor types analyzed by quantitative PCR. Int J Oncol 2013; 44:609-15. [PMID: 24317684 DOI: 10.3892/ijo.2013.2200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 10/29/2013] [Indexed: 11/06/2022] Open
Abstract
Tumor protein D52 (TPD52) is located at chromosome 8q21, a region that is frequently gained or amplified in multiple human cancer types. TPD52 has been suggested as a potential target for new anticancer therapies. In order to analyze TPD52 expression in the most prevalent human cancer types, we employed quantitative PCR to measure TPD52 mRNA levels in formalin-fixed tissue samples from more than 900 cancer tissues obtained from 29 different human cancer types. TPD52 was expressed at varying levels in all tested normal tissues, including skin, lymph node, lung, oral mucosa, breast, endometrium, ovary, vulva, myometrium, liver, pancreas, stomach, kidney, prostate, testis, urinary bladder, thyroid gland, brain, muscle and fat tissue. TPD52 was upregulated in 18/29 (62%) tested cancer types. Strongest expression was found in non-seminoma (56-fold overexpression compared to corresponding normal tissue), seminoma (42-fold), ductal (28-fold) and lobular breast cancer (14-fold). In these tumor types, TPD52 upregulation was found in the vast majority (>80%) of tested samples. Downregulation was found in 11 (38%) tumor types, most strongly in papillary renal cell cancer (-8-fold), leiomyosarcoma (-6-fold), clear cell renal cell cancer (-5-fold), liposarcoma (-5-fold) and lung cancer (-4-fold). These results demonstrate that TPD52 is frequently and strongly upregulated in many human cancer types, which may represent candidate tumor types for potential anti-TPD52 therapies.
Collapse
Affiliation(s)
- Pierre Tennstedt
- Martini-Clinic, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Charlotte Bölch
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gundula Strobel
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lia Burkhardt
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Grob
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sawinee Masser
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schlomm
- Martini-Clinic, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
742
|
iSubgraph: integrative genomics for subgroup discovery in hepatocellular carcinoma using graph mining and mixture models. PLoS One 2013; 8:e78624. [PMID: 24223834 PMCID: PMC3817163 DOI: 10.1371/journal.pone.0078624] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/13/2013] [Indexed: 11/24/2022] Open
Abstract
The high tumor heterogeneity makes it very challenging to identify key tumorigenic pathways as therapeutic targets. The integration of multiple omics data is a promising approach to identify driving regulatory networks in patient subgroups. Here, we propose a novel conceptual framework to discover patterns of miRNA-gene networks, observed frequently up- or down-regulated in a group of patients and to use such networks for patient stratification in hepatocellular carcinoma (HCC). We developed an integrative subgraph mining approach, called iSubgraph, and identified altered regulatory networks frequently observed in HCC patients. The miRNA and gene expression profiles were jointly analyzed in a graph structure. We defined a method to transform microarray data into graph representation that encodes miRNA and gene expression levels and the interactions between them as well. The iSubgraph algorithm was capable to detect cooperative regulation of miRNAs and genes even if it occurred only in some patients. Next, the miRNA-mRNA modules were used in an unsupervised class prediction model to discover HCC subgroups via patient clustering by mixture models. The robustness analysis of the mixture model showed that the class predictions are highly stable. Moreover, the Kaplan-Meier survival analysis revealed that the HCC subgroups identified by the algorithm have different survival characteristics. The pathway analyses of the miRNA-mRNA co-modules identified by the algorithm demonstrate key roles of Myc, E2F1, let-7, TGFB1, TNF and EGFR in HCC subgroups. Thus, our method can integrate various omics data derived from different platforms and with different dynamic scales to better define molecular tumor subtypes. iSubgraph is available as MATLAB code at http://www.cs.umd.edu/~ozdemir/isubgraph/.
Collapse
|
743
|
Xie B, Wang D, Duan Y, Yu J, Lei H. Functional networking of human divergently paired genes (DPGs). PLoS One 2013; 8:e78896. [PMID: 24205343 PMCID: PMC3815023 DOI: 10.1371/journal.pone.0078896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 09/17/2013] [Indexed: 11/18/2022] Open
Abstract
Divergently paired genes (DPGs), also known as bidirectional (head-to-head positioned) genes, are conserved across species and lineages, and thus deemed to be exceptional in genomic organization and functional regulation. Despite previous investigations on the features of their conservation and gene organization, the functional relationship among DPGs in a given species and lineage has not been thoroughly clarified. Here we report a network-based comprehensive analysis on human DPGs and our results indicate that the two members of the DPGs tend to participate in different biological processes while enforcing related functions as modules. Comparing to randomly paired genes as a control, the DPG pairs have a tendency to be clustered in similar “cellular components” and involved in similar “molecular functions”. The functional network bridged by DPGs consists of three major modules. The largest module includes many house-keeping genes involved in core cellular activities. This module also shows low variation in expression in both CNS (central nervous system) and non-CNS tissues. Based on analyses of disease transcriptome data, we further suggest that this particular module may play crucial roles in HIV infection and its disease mechanism.
Collapse
Affiliation(s)
- Bin Xie
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Dapeng Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yong Duan
- UC Davis Genome Center and Department of Biomedical Engineering, Davis, California, United States of America
| | - Jun Yu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- * E-mail: (JY); (HL)
| | - Hongxing Lei
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- UC Davis Genome Center and Department of Biomedical Engineering, Davis, California, United States of America
- * E-mail: (JY); (HL)
| |
Collapse
|
744
|
Dai Y, Cros MP, Pontoizeau C, Elena-Hermann B, Bonn GK, Hainaut P. Downregulation of transcription factor E4F1 in hepatocarcinoma cells: HBV-dependent effects on autophagy, proliferation and metabolism. Carcinogenesis 2013; 35:635-50. [PMID: 24163401 DOI: 10.1093/carcin/bgt353] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The multifunctional E4F1 protein is a cellular target of the E1A adenoviral oncoprotein. Interaction between E4F1 and the hepatitis B virus (HBV) protein HBx has been demonstrated in vitro. In this study, RNA interference has been used to downregulate E4F1 in the hepatocellular carcinoma (HCC) cell line HepG2 (HBV negative) and its derivative, HBV expressing HepG2/2.2.15. Reduction of E4F1 levels induced hepatocyte vacuolation (formation of large cytoplasmic vesicles), increased autophagy and caused mitochondrial defects and metabolism changes in HepG2/2.2.15, but not in HepG2. Moreover, downregulation of E4F1 reduced DNA synthesis with partial cell cycle arrest in G1 in both cell types and this effect was more marked in HepG2/2.2.15 than in HepG2. These effects were partially prevented by RNA interference directed to either HBx or to p53. Coprecipitation and western blot experiments detected complexes between E4F1 and HBx in several HCC cell lines. Although a review of mutation and gene expression public databases did not support that E4F1 is specifically altered in liver cancer, our results suggest that E4F1 may neutralize the capacity of HBx to activate a p53-dependent, metabolic and growth arrest phenotype in liver cells, thus possibly contributing to the viability and proliferation of HBV-infected cells.
Collapse
Affiliation(s)
- Yayun Dai
- Molecular Carcinogenesis, International Agency for Research on Cancer, 150 Cours Albert Thomas, Lyon 69372, France
| | | | | | | | | | | |
Collapse
|
745
|
Huang KC, Yang KC, Lin H, Tsao Tsun-Hui T, Lee WK, Lee SA, Kao CY. Analysis of schizophrenia and hepatocellular carcinoma genetic network with corresponding modularity and pathways: novel insights to the immune system. BMC Genomics 2013; 14 Suppl 5:S10. [PMID: 24564241 PMCID: PMC3852078 DOI: 10.1186/1471-2164-14-s5-s10] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Schizophrenic patients show lower incidences of cancer, implicating schizophrenia may be a protective factor against cancer. To study the genetic correlation between the two diseases, a specific PPI network was constructed with candidate genes of both schizophrenia and hepatocellular carcinoma. The network, designated schizophrenia-hepatocellular carcinoma network (SHCN), was analysed and cliques were identified as potential functional modules or complexes. The findings were compared with information from pathway databases such as KEGG, Reactome, PID and ConsensusPathDB. Results The functions of mediator genes from SHCN show immune system and cell cycle regulation have important roles in the eitology mechanism of schizophrenia. For example, the over-expressing schizophrenia candidate genes, SIRPB1, SYK and LCK, are responsible for signal transduction in cytokine production; immune responses involving IL-2 and TREM-1/DAP12 pathways are relevant for the etiology mechanism of schizophrenia. Novel treatments were proposed by searching the target genes of FDA approved drugs with genes in potential protein complexes and pathways. It was found that Vitamin A, retinoid acid and a few other immune response agents modulated by RARA and LCK genes may be potential treatments for both schizophrenia and hepatocellular carcinoma. Conclusions This is the first study showing specific mediator genes in the SHCN which may suppress tumors. We also show that the schizophrenic protein interactions and modulation with cancer implicates the importance of immune system for etiology of schizophrenia.
Collapse
|
746
|
Petrochilos D, Shojaie A, Gennari J, Abernethy N. Using random walks to identify cancer-associated modules in expression data. BioData Min 2013; 6:17. [PMID: 24128261 PMCID: PMC4015830 DOI: 10.1186/1756-0381-6-17] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 09/24/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The etiology of cancer involves a complex series of genetic and environmental conditions. To better represent and study the intricate genetics of cancer onset and progression, we construct a network of biological interactions to search for groups of genes that compose cancer-related modules. Three cancer expression datasets are investigated to prioritize genes and interactions associated with cancer outcomes. Using a graph-based approach to search for communities of phenotype-related genes in microarray data, we find modules of genes associated with cancer phenotypes in a weighted interaction network. RESULTS We implement Walktrap, a random-walk-based community detection algorithm, to identify biological modules predisposing to tumor growth in 22 hepatocellular carcinoma samples (GSE14520), adenoma development in 32 colorectal cancer samples (GSE8671), and prognosis in 198 breast cancer patients (GSE7390). For each study, we find the best scoring partitions under a maximum cluster size of 200 nodes. Significant modules highlight groups of genes that are functionally related to cancer and show promise as therapeutic targets; these include interactions among transcription factors (SPIB, RPS6KA2 and RPS6KA6), cell-cycle regulatory genes (BRSK1, WEE1 and CDC25C), modulators of the cell-cycle and proliferation (CBLC and IRS2) and genes that regulate and participate in the map-kinase pathway (MAPK9, DUSP1, DUSP9, RIPK2). To assess the performance of Walktrap to find genomic modules (Walktrap-GM), we evaluate our results against other tools recently developed to discover disease modules in biological networks. Compared with other highly cited module-finding tools, jActiveModules and Matisse, Walktrap-GM shows strong performance in the discovery of modules enriched with known cancer genes. CONCLUSIONS These results demonstrate that the Walktrap-GM algorithm identifies modules significantly enriched with cancer genes, their joint effects and promising candidate genes. The approach performs well when evaluated against similar tools and smaller overall module size allows for more specific functional annotation and facilitates the interpretation of these modules.
Collapse
Affiliation(s)
- Deanna Petrochilos
- Biomedical and Health Informatics, Dept of Biomedical Informatics and Medical Education, University of Washington, Box 357240, 1959 NE Pacific Street, HSB I-264, Seattle, WA 98195-7240, USA.
| | | | | | | |
Collapse
|
747
|
Gao J, Chen H, Yu Y, Song J, Song H, Su X, Li W, Tong X, Qian W, Wang H, Dai J, Guo Y. Inhibition of hepatocellular carcinoma growth using immunoliposomes for co-delivery of adriamycin and ribonucleotide reductase M2 siRNA. Biomaterials 2013; 34:10084-98. [PMID: 24060417 DOI: 10.1016/j.biomaterials.2013.08.088] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/30/2013] [Indexed: 12/18/2022]
Abstract
The chemotherapy combined with gene therapy has received great attention. We developed targeted LPD (liposome-polycation-DNA complex) conjugated with anti-EGFR (epidermal growth factor receptor) Fab' co-delivering adriamycin (ADR) and ribonucleotide reductase M2 (RRM2) siRNA (ADR-RRM2-TLPD), to achieve combined therapeutic effects in human hepatocellular carcinoma (HCC) overexpressing EGFR. The antitumor activity and mechanisms of ADR-RRM2-TLPD were investigated. The results showed that RRM2 expression was higher in HCC than in non-HCC tissue, and RRM2 siRNA inhibited HCC cell proliferation, suggesting that RRM2 is a candidate target for HCC therapy. ADR-RRM2-TLPD delivered ADR and RRM2 siRNA to EGFR overexpressing HCC cells specifically and efficiently both in vitro and in vivo, resulting in enhanced therapeutic effects (cytotoxicity, apoptosis and senescence-inducing activity) compared with single-drug loaded or non-targeted controls, including ADR-NC-TLPD (targeted LPD co-delivering ADR and negative control siRNA), RRM2-TLPD (targeted LPD delivering RRM2 siRNA) and ADR-RRM2-NTLPD (non-targeted LPD co-delivering ADR and RRM2 siRNA). Mechanism studies showed that p21 is involved in the combined therapeutic effect of ADR-RRM2-TLPD. The average weight of the orthotopic HCC in mice treated with ADR-RRM2-TLPD was significantly lighter than that of mice treated with other controls. Thus, ADR-RRM2-TLPD represents a potential strategy for combined therapy of HCC overexpressing EGFR.
Collapse
Affiliation(s)
- Jie Gao
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China; National Engineering Research Center for Antibody Medicine & Shanghai Key Laboratory of Cell Engineering and Antibody, 399 Libing Road, Shanghai 201203, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
748
|
O'Shea JM, Ayer DE. Coordination of nutrient availability and utilization by MAX- and MLX-centered transcription networks. Cold Spring Harb Perspect Med 2013; 3:a014258. [PMID: 24003245 DOI: 10.1101/cshperspect.a014258] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cell growth and division require the biosynthesis of macromolecule components and cofactors (e.g., nucleotides, lipids, amino acids, and nicotinamide adenine dinucleotide phosphate [NADPH]). Normally, macromolecular biosynthesis is under tight regulatory control, yet these anabolic pathways are often dysregulated in cancer. The resulting metabolic reprogramming of cancer cells is thought to support their high rates of growth and division. The mechanisms that underlie the metabolic changes in cancer are at least partially understood, providing a rationale for their targeting with known or novel therapeutics. This review is focused on how cells sense and respond transcriptionally to essential nutrients, including glucose and glutamine, and how MAX- and MLX-centered transcription networks contribute to metabolic homeostasis in normal and neoplastic cells.
Collapse
Affiliation(s)
- John M O'Shea
- Huntsman Cancer Institute, Department of Oncological Science, University of Utah, Salt Lake City, Utah 84112-5550
| | | |
Collapse
|
749
|
Li Y, He L, Zeng N, Sahu D, Cadenas E, Shearn C, Li W, Stiles BL. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) signaling regulates mitochondrial biogenesis and respiration via estrogen-related receptor α (ERRα). J Biol Chem 2013; 288:25007-25024. [PMID: 23836899 PMCID: PMC3757167 DOI: 10.1074/jbc.m113.450353] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 06/13/2013] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial abnormalities are associated with cancer development, yet how oncogenic signals affect mitochondrial functions has not been fully understood. In this study, we investigate the relationship between mitochondrial alterations and PI3K/protein kinase B (AKT) signaling activation using hepatocytes and liver tissues as our experimental models. We show here that liver-specific deletion of Pten, which leads to activation of PI3K/AKT, is associated with elevated oxidative stress, increased mitochondrial mass, and augmented respiration accompanied by enhanced glycolysis. Consistent with these observations, estrogen-related receptor α (ERRα), an orphan nuclear receptor known for its role in mitochondrial biogenesis, is up-regulated in the absence of phosphatase and tensin homolog deleted on chromosome 10 (PTEN). Our pharmacological and genetic studies show that PI3K/AKT activity regulates the expression of ERRα and mitochondrial biogenesis/respiration. Furthermore, cAMP-response element-binding protein, as a downstream target of AKT, plays a role in the regulation of ERRα, independent of PKA signaling. ERRα regulates reactive oxygen species production, and ERRα knockdown attenuates proliferation and colony-forming potential in Pten-null hepatocytes. Finally, analysis of clinical datasets from liver tissues showed a negative correlation between expressions of ERRα and PTEN in patients with liver cancer. Therefore, this study has established a previously unrecognized link between a growth signal and mitochondrial metabolism.
Collapse
Affiliation(s)
- Yang Li
- From Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089
| | - Lina He
- From Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089
| | - Ni Zeng
- From Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089
| | - Divya Sahu
- Department of Dermatology, Norris Comprehensive Cancer Center, Keck Medical Center, University of Southern California, Los Angeles, California 90033, and
| | - Enrique Cadenas
- From Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089,; Biochemistry, Keck School of Medicine, and
| | - Colin Shearn
- Pharmaceutical Sciences, School of Pharmacy, University of Colorado, Aurora, Colorado 80045
| | - Wei Li
- Department of Dermatology, Norris Comprehensive Cancer Center, Keck Medical Center, University of Southern California, Los Angeles, California 90033, and
| | - Bangyan L Stiles
- From Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089,; the Departments of Pathology and.
| |
Collapse
|
750
|
Schirmacher P, Calvisi DF. Molecular diagnostic algorithms in hepatocellular carcinoma: dead-end street or light at the end of the tunnel? Gastroenterology 2013; 145:49-53. [PMID: 23727497 DOI: 10.1053/j.gastro.2013.05.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Peter Schirmacher
- Pathologisches Institut, Universität Heidelberg, Heidelberg, Germany.
| | - Diego F Calvisi
- Institut für Pathologie, Universitätsmedizin Greifswald, Greifswald, Germany.
| |
Collapse
|