51
|
Saleembhasha A, Mishra S. Novel molecules lncRNAs, tRFs and circRNAs deciphered from next-generation sequencing/RNA sequencing: computational databases and tools. Brief Funct Genomics 2019. [PMID: 28637169 DOI: 10.1093/bfgp/elx013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Powerful next-generation sequencing (NGS) technologies, more specifically RNA sequencing (RNA-seq), have been pivotal toward the detection and analysis and hypotheses generation of novel biomolecules, long noncoding RNAs (lncRNAs), tRNA-derived fragments (tRFs) and circular RNAs (circRNAs). Experimental validation of the occurrence of these biomolecules inside the cell has been reported. Their differential expression and functionally important role in several cancers types as well as other diseases such as Alzheimer's and cardiovascular diseases have garnered interest toward further studies in this research arena. In this review, starting from a brief relevant introduction to NGS and RNA-seq and the expression and role of lncRNAs, tRFs and circRNAs in cancer, we have comprehensively analyzed the current landscape of databases developed and computational software used for analyses and visualization for this emerging and highly interesting field of these novel biomolecules. Our review will help the end users and research investigators gain information on the existing databases and tools as well as an understanding of the specific features which these offer. This will be useful for the researchers in their proper usage thereby guiding them toward novel hypotheses generation and saving time and costs involved in extensive experimental processes in these three different novel functional RNAs.
Collapse
|
52
|
Wang Y, Liu Z, Lian B, Liu L, Xie L. Integrative Analysis of Dysfunctional Modules Driven by Genomic Alterations at System Level Across 11 Cancer Types. Comb Chem High Throughput Screen 2019; 21:771-783. [DOI: 10.2174/1386207322666190122110726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/15/2018] [Accepted: 11/09/2018] [Indexed: 01/05/2023]
Abstract
Aim and Objective:
Integrating multi-omics data to identify driver genes and key
biological functions for tumorigenesis remains a major challenge.
Method:
A new computational pipeline was developed to identify the Driver Mutation-Differential
Co-Expression (DM-DCE) modules based on dysfunctional networks across 11 TCGA cancers.
Results:
Functional analyses provided insight into the properties of various cancers, and found
common cellular signals / pathways of cancers. Furthermore, the corresponding network analysis
identified conservations or interactions across different types of cancers, thus the crosstalk between
the key signaling pathways, immunity and cancers was found. Clinical analysis also identified key
prognostic / survival patterns.
Conclusion:
Taken together, our study sheds light on both cancer-specific and cross-cancer
characteristics systematically.
Collapse
Affiliation(s)
- Yin Wang
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai 201203, China
| | - Zhenhao Liu
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai 201203, China
| | - Baofeng Lian
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai 201203, China
| | - Lei Liu
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai 201203, China
| | - Lu Xie
- Shanghai Center for Bioinformation Technology, Shanghai Academy of Science and Technology, Shanghai 201203, China
| |
Collapse
|
53
|
Orlando E, Aebersold DM, Medová M, Zimmer Y. Oncogene addiction as a foundation of targeted cancer therapy: The paradigm of the MET receptor tyrosine kinase. Cancer Lett 2019; 443:189-202. [DOI: 10.1016/j.canlet.2018.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/20/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022]
|
54
|
Orlando E, Aebersold R. On the contribution of mass spectrometry-based platforms to the field of personalized oncology. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2018.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
55
|
Krivtsova O, Makarova A, Lazarevich N. Aberrant expression of alternative isoforms of transcription factors in hepatocellular carcinoma. World J Hepatol 2018; 10:645-661. [PMID: 30386458 PMCID: PMC6206146 DOI: 10.4254/wjh.v10.i10.645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/08/2018] [Accepted: 06/28/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies worldwide and the second leading cause of death among all cancer types. Deregulation of the networks of tissue-specific transcription factors (TFs) observed in HCC leads to profound changes in the hepatic transcriptional program that facilitates tumor progression. In addition, recent reports suggest that substantial aberrations in the production of TF isoforms occur in HCC. In vitro experiments have identified distinct isoform-specific regulatory functions and related biological effects of liver-specific TFs that are implicated in carcinogenesis, which may be relevant for tumor progression and clinical outcome. This study reviews available data on the expression of isoforms of liver-specific and ubiquitous TFs in the liver and HCC and their effects, including HNF4α, C/EBPs, p73 and TCF7L2, and indicates that assessment of the ratio of isoforms and targeting specific TF variants may be beneficial for the prognosis and treatment of HCC.
Collapse
Affiliation(s)
- Olga Krivtsova
- Federal State Budgetary Institution, “N. N. Blokhin Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Russian
- M. V. Lomonosov Moscow State University, Moscow 119991, Russian
| | - Anna Makarova
- Federal State Budgetary Institution, “N. N. Blokhin Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Russian
| | - Natalia Lazarevich
- Federal State Budgetary Institution, “N. N. Blokhin Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation, Moscow 115478, Russian
- M. V. Lomonosov Moscow State University, Moscow 119991, Russian
| |
Collapse
|
56
|
Transcriptome profiling of the interconnection of pathways involved in malignant transformation and response to hypoxia. Oncotarget 2018; 9:19730-19744. [PMID: 29731978 PMCID: PMC5929421 DOI: 10.18632/oncotarget.24808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 02/24/2018] [Indexed: 11/25/2022] Open
Abstract
In tumor tissues, hypoxia is a commonly observed feature resulting from rapidly proliferating cancer cells outgrowing their surrounding vasculature network. Transformed cancer cells are known to exhibit phenotypic alterations, enabling continuous proliferation despite a limited oxygen supply. The four-step isogenic BJ cell model enables studies of defined steps of tumorigenesis: the normal, immortalized, transformed, and metastasizing stages. By transcriptome profiling under atmospheric and moderate hypoxic (3% O2) conditions, we observed that despite being highly similar, the four cell lines of the BJ model responded strikingly different to hypoxia. Besides corroborating many of the known responses to hypoxia, we demonstrate that the transcriptome adaptation to moderate hypoxia resembles the process of malignant transformation. The transformed cells displayed a distinct capability of metabolic switching, reflected in reversed gene expression patterns for several genes involved in oxidative phosphorylation and glycolytic pathways. By profiling the stage-specific responses to hypoxia, we identified ASS1 as a potential prognostic marker in hypoxic tumors. This study demonstrates the usefulness of the BJ cell model for highlighting the interconnection of pathways involved in malignant transformation and hypoxic response.
Collapse
|
57
|
Hong Y, Park C, Kim N, Cho J, Moon SU, Kim J, Jeong E, Yoon S. QSurface: fast identification of surface expression markers in cancers. BMC SYSTEMS BIOLOGY 2018; 12:17. [PMID: 29560830 PMCID: PMC5861488 DOI: 10.1186/s12918-018-0541-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Cell surface proteins have provided useful targets and biomarkers for advanced cancer therapies. The recent clinical success of antibody-drug conjugates (ADCs) highlights the importance of finding selective surface antigens for given cancer subtypes. We thus attempted to develop stand-alone software for the analysis of the cell surface transcriptome of patient cancer samples and to prioritize lineage- and/or mutation-specific over-expression markers in cancer cells. Results A total of 519 genes were selected as surface proteins, and their expression was profiled in 14 cancer subtypes using patient sample transcriptome data. Lineage/mutation-oriented analysis was used to identify subtype-specific surface markers with statistical confidence. Experimental validation confirmed the unique over-expression of predicted surface markers (MUC4, MSLN, and SLC7A11) in lung cancer cells at the protein level. The differential cell surface gene expression of cell lines may differ from that of tissue samples due to the absence of the tumor microenvironment. Conclusions In the present study, advanced 3D models of lung cell lines successfully reproduced the predicted patterns, demonstrating the physiological relevance of cell line-based 3D models in validating surface markers from patient tumor data. Also QSurface software is freely available at http://compbio.sookmyung.ac.kr/~qsurface. Electronic supplementary material The online version of this article (10.1186/s12918-018-0541-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Nayoung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Juyeon Cho
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Sung Ung Moon
- Center for Advanced Bioinformatics & Systems medicine, Department of Biological Sciences, Sookmyung Women's University, Hyochangwon-gil 52, Yongsan-gu, Seoul, 140-742, Republic of Korea
| | - Jongmin Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 140-742, Republic of Korea
| | - Euna Jeong
- Center for Advanced Bioinformatics & Systems medicine, Department of Biological Sciences, Sookmyung Women's University, Hyochangwon-gil 52, Yongsan-gu, Seoul, 140-742, Republic of Korea.
| | - Sukjoon Yoon
- Center for Advanced Bioinformatics & Systems medicine, Department of Biological Sciences, Sookmyung Women's University, Hyochangwon-gil 52, Yongsan-gu, Seoul, 140-742, Republic of Korea. .,Department of Biological Sciences, Sookmyung Women's University, Seoul, 140-742, Republic of Korea.
| |
Collapse
|
58
|
Kuo CHS, Liu CY, Pavlidis S, Lo YL, Wang YW, Chen CH, Ko HW, Chung FT, Lin TY, Wang TY, Lee KY, Guo YK, Wang TH, Yang CT. Unique Immune Gene Expression Patterns in Bronchoalveolar Lavage and Tumor Adjacent Non-Neoplastic Lung Tissue in Non-Small Cell Lung Cancer. Front Immunol 2018; 9:232. [PMID: 29483918 PMCID: PMC5816075 DOI: 10.3389/fimmu.2018.00232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/26/2018] [Indexed: 12/13/2022] Open
Abstract
Background The immune cells in the local environments surrounding non-small cell lung cancer (NSCLC) implicate the balance of pro- and antitumor immunity; however, their transcriptomic profiles remain poorly understood. Methods A transcriptomic microarray study of bronchoalveolar lavage (BAL) cells harvested from tumor-bearing lung segments was performed in a discovery group. The findings were validated (1) in published microarray datasets, (2) in an independent group by RT-qPCR, and (3) in non-diseased and tumor adjacent non-neoplastic lung tissue by immunohistochemistry and in BAL cell lysates by immunoblotting. Result The differential expression of 129 genes was identified in the discovery group. These genes revealed functional enrichment in Fc gamma receptor-dependent phagocytosis and circulating immunoglobulin complex among others. Microarray datasets analysis (n = 607) showed that gene expression of BAL cells of tumor-bearing lung segment was also the unique transcriptomic profile of tumor adjacent non-neoplastic lung of early stage NSCLC and a significantly gradient increase of immunoglobulin genes’ expression for non-diseased lungs, tumor adjacent non-neoplastic lungs, and tumors was identified (ANOVA, p < 2 × 10−16). A 53-gene signature was determined with significant correlation with inhibitory checkpoint PDCD1 (r = 0.59, p = 0.0078) among others, where the nine top genes including IGJ and IGKC were RT-qPCR validated with high diagnostic performance (AUC: 0.920, 95% CI: 0.831–0.985, p = 2.98 × 10−7). Increased staining and expression of IGKC revealed by immunohistochemistry and immunoblotting in tumor adjacent non-neoplastic lung tissues (Wilcoxon signed-rank test, p < 0.001) and in BAL cell lysates (p < 0.01) of NSCLC, respectively, were noted. Conclusion The BAL cells of tumor-bearing lung segments and tumor adjacent non-neoplastic lung tissues present a unique gene expression characterized by IGKC in relation to inhibitory checkpoints. Further study of humoral immune responses to NSCLC is warranted.
Collapse
Affiliation(s)
- Chih-Hsi Scott Kuo
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Computing, Imperial College London, Data Science Institute, London, United Kingdom
| | - Chien-Ying Liu
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Stelios Pavlidis
- Department of Computing, Imperial College London, Data Science Institute, London, United Kingdom
| | - Yu-Lun Lo
- Division of Airway Diseases, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Yen-Wen Wang
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chih-Hung Chen
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - How-Wen Ko
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Fu-Tsai Chung
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Tin-Yu Lin
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Tsai-Yu Wang
- Division of Airway Diseases, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Kang-Yun Lee
- Division of Thoracic Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| | - Yi-Ke Guo
- Department of Computing, Imperial College London, Data Science Institute, London, United Kingdom
| | - Tzu-Hao Wang
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Cheng-Ta Yang
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
59
|
Innovative methods for biomarker discovery in the evaluation and development of cancer precision therapies. Cancer Metastasis Rev 2018; 37:125-145. [PMID: 29392535 DOI: 10.1007/s10555-017-9710-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The discovery of biomarkers able to detect cancer at an early stage, to evaluate its aggressiveness, and to predict the response to therapy remains a major challenge in clinical oncology and precision medicine. In this review, we summarize recent achievements in the discovery and development of cancer biomarkers. We also highlight emerging innovative methods in biomarker discovery and provide insights into the challenges faced in their evaluation and validation.
Collapse
|
60
|
Tso FY, Kossenkov AV, Lidenge SJ, Ngalamika O, Ngowi JR, Mwaiselage J, Wickramasinghe J, Kwon EH, West JT, Lieberman PM, Wood C. RNA-Seq of Kaposi's sarcoma reveals alterations in glucose and lipid metabolism. PLoS Pathog 2018; 14:e1006844. [PMID: 29352292 PMCID: PMC5792027 DOI: 10.1371/journal.ppat.1006844] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/31/2018] [Accepted: 12/27/2017] [Indexed: 12/31/2022] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi’s sarcoma (KS). It is endemic in a number of sub-Saharan African countries with infection rate of >50%. The high prevalence of HIV-1 coupled with late presentation of advanced cancer staging make KS the leading cancer in the region with poor prognosis and high mortality. Disease markers and cellular functions associated with KS tumorigenesis remain ill-defined. Several studies have attempted to investigate changes of the gene profile with in vitro infection of monoculture models, which are not likely to reflect the cellular complexity of the in vivo lesion environment. Our approach is to characterize and compare the gene expression profile in KS lesions versus non-cancer tissues from the same individual. Such comparisons could identify pathways critical for KS formation and maintenance. This is the first study that utilized high throughput RNA-seq to characterize the viral and cellular transcriptome in tumor and non-cancer biopsies of African epidemic KS patients. These patients were treated anti-retroviral therapy with undetectable HIV-1 plasma viral load. We found remarkable variability in the viral transcriptome among these patients, with viral latency and immune modulation genes most abundantly expressed. The presence of KSHV also significantly affected the cellular transcriptome profile. Specifically, genes involved in lipid and glucose metabolism disorder pathways were substantially affected. Moreover, infiltration of immune cells into the tumor did not prevent KS formation, suggesting some functional deficits of these cells. Lastly, we found only minimal overlaps between our in vivo cellular transcriptome dataset with those from in vitro studies, reflecting the limitation of in vitro models in representing tumor lesions. These findings could lead to the identification of diagnostic and therapeutic markers for KS, and will provide bases for further mechanistic studies on the functions of both viral and cellular genes that are involved. Kaposi’s sarcoma-associated herpesvirus (KSHV) is endemic in sub-Saharan Africa and cause Kaposi’s sarcoma (KS). KS is one of the most common cancer among HIV-1 patients in this region. Despite anti-retroviral treatment, prognosis for KS is poor with high mortality often due to presentation of late cancer stage. In order to identify biomarkers or therapeutic targets against KS, a better understanding of the viral and cellular genes expression/transcriptome in the tumor will be necessary. We used RNA-seq, a highly efficient method to sequence transcriptome, to characterize and compare the viral and cellular transcriptome in tumor and non-cancer tissues from KS patients. We found that viral genes involved in latency and immune modulation are most commonly expressed among KS patients. Additionally, cellular genes involved in lipid and glucose metabolism disorder pathways are significantly affected by the presence of KSHV. Despite the detection of immune cells in the tumor, it did not prevent the tumor progression, suggesting some level of immune cell dysfunctions in KS patients. Lastly, we found limited overlap of our data, derived from actual KS biopsy, with other cell culture models, suggesting that the complexity of tumor is difficult to be reflected in cell line models.
Collapse
Affiliation(s)
- For Yue Tso
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | | | - Salum J. Lidenge
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Owen Ngalamika
- Dermatology and Venereology section, University Teaching Hospitals, University of Zambia School of Medicine, Lusaka, Zambia
| | - John R. Ngowi
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Julius Mwaiselage
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Eun Hee Kwon
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - John T. West
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Paul M. Lieberman
- Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Charles Wood
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
61
|
Chemotherapy-induced uridine diphosphate release promotes breast cancer metastasis through P2Y6 activation. Oncotarget 2018; 7:29036-50. [PMID: 27074554 PMCID: PMC5045376 DOI: 10.18632/oncotarget.8664] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/28/2016] [Indexed: 11/28/2022] Open
Abstract
Although purinergic signaling is important in regulation of immune responses, the therapeutic potential of it in the tumor microenvironment is little defined. In this study, we demonstrate that UDP/P2Y6 signaling facilitates breast cancer metastasis both in vitro and in vivo. We found that P2Y6 is not only aberrantly expressed and mutated in most tumor types, but also highly correlated with poor prognosis in breast cancer patients. Furthermore, the migration and invasion of breast cancer cells was obviously increased by UDP and blocked by P2Y6 specific inhibitor MRS2578 and P2Y6 shRNA. Similar results was also found in breast cancer cell metastasis mouse model. Interestingly, the endogenous agonist UDP was released significantly by doxorubicin treated cells. In addition, the expression and enzyme activity of MMP-9 were both promoted by UDP and inhibited by MRS2578 or P2Y6 shRNA. Furthermore, UDP-induced cell invasion was blocked by an MMP-9 inhibitor. Mechanistically, the MAPKs and NF-κB signaling pathways, known to be involved in regulation of MMP-9 expression, were both activated by UDP. Taken together, our study reveals a relationship between extracellular danger signals and breast cancer metastasis, which suggests the potential therapeutic significance of UDP/P2Y6 signaling in cancer therapy.
Collapse
|
62
|
Uhlen M, Zhang C, Lee S, Sjöstedt E, Fagerberg L, Bidkhori G, Benfeitas R, Arif M, Liu Z, Edfors F, Sanli K, von Feilitzen K, Oksvold P, Lundberg E, Hober S, Nilsson P, Mattsson J, Schwenk JM, Brunnström H, Glimelius B, Sjöblom T, Edqvist PH, Djureinovic D, Micke P, Lindskog C, Mardinoglu A, Ponten F. A pathology atlas of the human cancer transcriptome. Science 2017; 357:357/6352/eaan2507. [PMID: 28818916 DOI: 10.1126/science.aan2507] [Citation(s) in RCA: 2156] [Impact Index Per Article: 308.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/02/2017] [Accepted: 07/14/2017] [Indexed: 12/11/2022]
Abstract
Cancer is one of the leading causes of death, and there is great interest in understanding the underlying molecular mechanisms involved in the pathogenesis and progression of individual tumors. We used systems-level approaches to analyze the genome-wide transcriptome of the protein-coding genes of 17 major cancer types with respect to clinical outcome. A general pattern emerged: Shorter patient survival was associated with up-regulation of genes involved in cell growth and with down-regulation of genes involved in cellular differentiation. Using genome-scale metabolic models, we show that cancer patients have widespread metabolic heterogeneity, highlighting the need for precise and personalized medicine for cancer treatment. All data are presented in an interactive open-access database (www.proteinatlas.org/pathology) to allow genome-wide exploration of the impact of individual proteins on clinical outcomes.
Collapse
Affiliation(s)
- Mathias Uhlen
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden. .,Center for Biosustainability, Danish Technical University, Copenhagen, Denmark.,School of Biotechnology, AlbaNova University Center, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Sunjae Lee
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Evelina Sjöstedt
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden.,Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Gholamreza Bidkhori
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Rui Benfeitas
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Muhammad Arif
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Zhengtao Liu
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Kemal Sanli
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Kalle von Feilitzen
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Per Oksvold
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Emma Lundberg
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Sophia Hober
- School of Biotechnology, AlbaNova University Center, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Peter Nilsson
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Johanna Mattsson
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jochen M Schwenk
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Hans Brunnström
- Division of Pathology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Bengt Glimelius
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Tobias Sjöblom
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Per-Henrik Edqvist
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Dijana Djureinovic
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Cecilia Lindskog
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden.,School of Biotechnology, AlbaNova University Center, KTH-Royal Institute of Technology, Stockholm, Sweden.,Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
| | - Fredrik Ponten
- Department of Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
63
|
Yang S, Sui J, Liang G. Diagnosis value of aberrantly expressed microRNA profiles in lung squamous cell carcinoma: a study based on the Cancer Genome Atlas. PeerJ 2017; 5:e4101. [PMID: 29204322 PMCID: PMC5712466 DOI: 10.7717/peerj.4101] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/06/2017] [Indexed: 12/22/2022] Open
Abstract
Background Lung cancer is considered as one of the most frequent and deadly cancers with high mortality all around the world. It is critical to find new biomarkers for early diagnosis of lung cancer, especially lung squamous cell carcinoma (LUSC). The Cancer Genome Atlas (TCGA) is a database which provides both cancer and clinical information. This study is a comprehensive analysis of a novel diagnostic biomarker for LUSC, based on TCGA. Methods and Results The present study investigated LUSC-specific key microRNAs (miRNAs) from large-scale samples in TCGA. According to exclusion criteria and inclusion criteria, the expression profiles of miRNAs with related clinical information of 332 LUSC patients were obtained. Most aberrantly expressed miRNAs were identified between tumor and normal samples. Forty-two LUSC-specific intersection miRNAs (fold change >2, p < 0.05) were obtained by an integrative computational method, among them six miRNAs were found to be aberrantly expressed concerning characteristics of patients (gender, lymphatic metastasis, patient outcome assessment) through Student t-test. Five miRNAs correlated with overall survival (log-rank p < 0.05) were obtained through the univariate Cox proportional hazards regression model and Mantel–Haenszel test. Then, five miRNAs were randomly selected to validate the expression in 47 LUSC patient tissues using quantitative real-time polymerase chain reaction. The results showed that the test findings were consistent with the TCGA findings. Also, the diagnostic value of the specific key miRNAs was determined by areas under receiver operating characteristic curves. Finally, 577 interaction mRNAs as the targets of 42 LUSC-specific intersection miRNAs were selected for further bioinformatics analysis. Conclusion This study indicates that this novel microRNA expression signature may be a useful biomarker of the diagnosis for LUSC patients, based on bioinformatics analysis.
Collapse
Affiliation(s)
- Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Jing Sui
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
64
|
Suman S, Mishra A. Network analysis revealed aurora kinase dysregulation in five gynecological types of cancer. Oncol Lett 2017; 15:1125-1132. [PMID: 29391900 DOI: 10.3892/ol.2017.7368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/13/2017] [Indexed: 01/04/2023] Open
Abstract
Gene markers are crucial for cancer prognosis and treatment. Previous studies have placed greater emphasis on individual diagnostic genes, thereby ignoring systemic-level attributes across diseases. Female-specific cells namely, breast, endometrium, cervical, ovarian and vulvar cells are highly susceptible to cancer. To date, a limited number of molecular studies have been performed that evaluate common biological processes across gynecological types of cancer. Differentially expressed genes in breast, cervical, endometrial, vulvar and ovarian cancer were utilized to construct protein-protein interaction networks, and to identify a common module across the five cancer types. A single common module with 8 nodes and 26 edges was mined among the five cancer systems. In total, four hub genes were present across the five cancer gene sets. Genes in the common module were enriched for the common pathways and associated diseases. The aurora kinase pathway was revealed to be conserved across the five cancer types surveyed. The present study, therefore, revealed that the aurora kinase pathway has a crucial function in the pathogenesis of the five aforementioned gynecological types of cancer through cross-tumor conservation.
Collapse
Affiliation(s)
- Shikha Suman
- Division of Applied Sciences, Indian Institute of Information Technology, Allahabad, Uttar Pradesh 211012, India
| | - Ashutosh Mishra
- Division of Applied Sciences, Indian Institute of Information Technology, Allahabad, Uttar Pradesh 211012, India
| |
Collapse
|
65
|
Durand S, Trillet K, Uguen A, Saint-Pierre A, Le Jossic-Corcos C, Corcos L. A transcriptome-based protein network that identifies new therapeutic targets in colorectal cancer. BMC Genomics 2017; 18:758. [PMID: 28962550 PMCID: PMC5622428 DOI: 10.1186/s12864-017-4139-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 09/13/2017] [Indexed: 01/22/2023] Open
Abstract
Background Colon cancer occurrence is increasing worldwide, making it the third most frequent cancer. Although many therapeutic options are available and quite efficient at the early stages, survival is strongly decreased when the disease has spread to other organs. The identification of molecular markers of colon cancer is likely to help understanding its course and, eventually, to uncover novel genes to be targeted by drugs. In this study, we compared gene expression in a set of 95 human colon cancer samples to that in 19 normal colon mucosae, focusing on 401 genes from 5 selected pathways (Apoptosis, Cancer, Cholesterol metabolism and lipoprotein signaling, Drug metabolism, Wnt/beta-catenin). Deregulation of mRNA levels largely matched that of proteins, leading us to build in silico protein networks, starting from mRNA levels, to identify key proteins central to network activity. Results Among the analyzed genes, 10.5% (42) had no reported link with colon cancer, including the SFRP1, IGF1 and ADH1B (down), and MYC and IL8 (up), whose encoded proteins were most interacting with other proteins from the same or even distinct networks. Analyzing all pathways globally led us to uncover novel functional links between a priori unrelated or rather remotely connected pathways, such as the Drug metabolism and the Cancer pathways or, even more strikingly, between the Cholesterol metabolism and lipoprotein signaling and the Cancer pathways. In addition, we analyzed the responsiveness of some of the deregulated genes essential to network activities, to chemotherapeutic agents used alone or in presence of Lovastatin, a lipid-lowering drug. Some of these treatments could oppose the deregulations occurring in cancer samples, including those of the CHECK2, CYP51A1, HMGCS1, ITGA2, NME1 or VEGFA genes. Conclusions Our network-based approach allowed discovering genes not previously known to play regulatory roles in colon cancer. Our results also showed that selected drug treatments might revert the cancer-specific deregulation of genes playing prominent roles within the networks operating to maintain colon homeostasis. Among those genes, some could constitute novel testable targets to eliminate colon cancer cells, either directly or, potentially, through the use of lipid-lowering drugs such as statins, in association with selected anticancer drugs. Electronic supplementary material The online version of this article (10.1186/s12864-017-4139-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stéphanie Durand
- INSERM 1078 Unit, "Cancérologie appliquée et épissage alternatif" team, Brest Institute of Health, Agronomy and Material (IBSAM), Faculty of medicine, University of Western Brittany (UBO), 22 avenue Camille Desmoulins, F-29200, Brest, France
| | - Killian Trillet
- INSERM 1078 Unit, "Cancérologie appliquée et épissage alternatif" team, Brest Institute of Health, Agronomy and Material (IBSAM), Faculty of medicine, University of Western Brittany (UBO), 22 avenue Camille Desmoulins, F-29200, Brest, France
| | - Arnaud Uguen
- INSERM 1078 Unit, "Cancérologie appliquée et épissage alternatif" team, Brest Institute of Health, Agronomy and Material (IBSAM), Faculty of medicine, University of Western Brittany (UBO), 22 avenue Camille Desmoulins, F-29200, Brest, France.,Department of Pathology, Brest University Hospital, F-29200, Brest, France
| | - Aude Saint-Pierre
- INSERM 1078 Unit, "Epidemiology, genetic Epidemiology and population genetics" team, 46 rue Félix Le Dantec, F-29200, Brest, France
| | - Catherine Le Jossic-Corcos
- INSERM 1078 Unit, "Cancérologie appliquée et épissage alternatif" team, Brest Institute of Health, Agronomy and Material (IBSAM), Faculty of medicine, University of Western Brittany (UBO), 22 avenue Camille Desmoulins, F-29200, Brest, France
| | - Laurent Corcos
- INSERM 1078 Unit, "Cancérologie appliquée et épissage alternatif" team, Brest Institute of Health, Agronomy and Material (IBSAM), Faculty of medicine, University of Western Brittany (UBO), 22 avenue Camille Desmoulins, F-29200, Brest, France. .,INSERM 1078 Unit, "Cancérologie appliquée et épissage alternatif" laboratory, University of Western Brittany (UBO), Faculty of medicine, 22, rue Camille Desmoulins, 29200, Brest, France.
| |
Collapse
|
66
|
Li J, Zheng S, Kang H, Hou Z, Qian Q. Identifying Scientific Project-generated Data Citation from Full-text Articles: An Investigation of TCGA Data Citation. JOURNAL OF DATA AND INFORMATION SCIENCE 2017. [DOI: 10.20309/jdis.201612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Abstract
Purpose
In the open science era, it is typical to share project-generated scientific data by depositing it in an open and accessible database. Moreover, scientific publications are preserved in a digital library archive. It is challenging to identify the data usage that is mentioned in literature and associate it with its source. Here, we investigated the data usage of a government-funded cancer genomics project, The Cancer Genome Atlas (TCGA), via a full-text literature analysis.
Design/methodology/approach
We focused on identifying articles using the TCGA dataset and constructing linkages between the articles and the specific TCGA dataset. First, we collected 5,372 TCGA-related articles from PubMed Central (PMC). Second, we constructed a benchmark set with 25 full-text articles that truly used the TCGA data in their studies, and we summarized the key features of the benchmark set. Third, the key features were applied to the remaining PMC full-text articles that were collected from PMC.
Findings
The amount of publications that use TCGA data has increased significantly since 2011, although the TCGA project was launched in 2005. Additionally, we found that the critical areas of focus in the studies that use the TCGA data were glioblastoma multiforme, lung cancer, and breast cancer; meanwhile, data from the RNA-sequencing (RNA-seq) platform is the most preferable for use.
Research limitations
The current workflow to identify articles that truly used TCGA data is labor-intensive. An automatic method is expected to improve the performance.
Practical implications
This study will help cancer genomics researchers determine the latest advancements in cancer molecular therapy, and it will promote data sharing and data-intensive scientific discovery.
Originality/value
Few studies have been conducted to investigate data usage by government-funded projects/programs since their launch. In this preliminary study, we extracted articles that use TCGA data from PMC, and we created a link between the full-text articles and the source data.
Collapse
Affiliation(s)
- Jiao Li
- Institute of Medical Information and Library , Chinese Academy of Medical Sciences , Beijing 100020 , China
| | - Si Zheng
- Institute of Medical Information and Library , Chinese Academy of Medical Sciences , Beijing 100020 , China
| | - Hongyu Kang
- Institute of Medical Information and Library , Chinese Academy of Medical Sciences , Beijing 100020 , China
| | - Zhen Hou
- Institute of Medical Information and Library , Chinese Academy of Medical Sciences , Beijing 100020 , China
| | - Qing Qian
- Institute of Medical Information and Library , Chinese Academy of Medical Sciences , Beijing 100020 , China
| |
Collapse
|
67
|
Aziz MA, Yousef Z, Saleh AM, Mohammad S, Al Knawy B. Towards personalized medicine of colorectal cancer. Crit Rev Oncol Hematol 2017; 118:70-78. [PMID: 28917272 DOI: 10.1016/j.critrevonc.2017.08.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 04/18/2017] [Accepted: 08/21/2017] [Indexed: 02/07/2023] Open
Abstract
Efforts in colorectal cancer (CRC) research aim to improve early detection and treatment for metastatic stages which could translate into better prognosis of this disease. One of the major challenges that hinder these efforts is the heterogeneous nature of CRC and involvement of diverse molecular pathways. New large-scale 'omics' technologies are making it possible to generate, analyze and interpret biological data from molecular determinants of CRC. The developments of sophisticated computational analyses would allow information from different omics platforms to be integrated, thus providing new insights into the biology of CRC. Together, these technological advances and an improved mechanistic understanding might allow CRC to be clinically managed at the level of the individual patient. This review provides an account of the current challenges in CRC management and an insight into how new technologies could allow the development of personalized medicine for CRC.
Collapse
Affiliation(s)
- Mohammad Azhar Aziz
- King Abdullah International Medical Research Center [KAIMRC], King Saud Bin Abdulaziz University for Health Sciences, Colorectal Cancer Research Program, National Guard Health Affairs, P.O. Box 22490, Riyadh 11426, Saudi Arabia.
| | - Zeyad Yousef
- King Abdullah International Medical Research Center [KAIMRC], King Saud Bin Abdulaziz University for Health Sciences, Department of Surgery, National Guard Health Affairs, P.O. Box 22490, Riyadh 11426, Saudi Arabia.
| | - Ayman M Saleh
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, National Guard Health Affairs, Mail Code 6610, P. O. Box 9515 Jeddah 21423, Saudi Arabia; King Abdullah International Medical Research Center [KAIMRC], King Abdulaziz Medical City, National Guard Health Affairs, P. O. Box 9515, Jeddah 21423, Saudi Arabia.
| | - Sameer Mohammad
- King Abdullah International Medical Research Center [KAIMRC], King Saud Bin Abdulaziz University for Health Sciences, Department of Experimental Medicine, National Guard Health Affairs, P.O. Box 22490, Riyadh 11426, Saudi Arabia.
| | - Bandar Al Knawy
- King Abdullah International Medical Research Center [KAIMRC], King Saud Bin Abdulaziz University for Health Sciences, Office of the Chief Executive Officer, National Guard Health Affairs, P.O. Box 22490, Riyadh 11426, Saudi Arabia.
| |
Collapse
|
68
|
Behjati S, Haniffa M. Genetics: Taking single-cell transcriptomics to the bedside. Nat Rev Clin Oncol 2017; 14:590-592. [PMID: 28762386 DOI: 10.1038/nrclinonc.2017.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sam Behjati
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK; and at the Department of Paediatrics, University of Cambridge, Cambridge, Hills Road, CB2 0QQ, UK
| | - Muzlifah Haniffa
- Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK; and at the Department of Dermatology and Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Queen Victoria Road, Newcastle upon Tyne NE2 4LP, UK
| |
Collapse
|
69
|
Benfeitas R, Uhlen M, Nielsen J, Mardinoglu A. New Challenges to Study Heterogeneity in Cancer Redox Metabolism. Front Cell Dev Biol 2017; 5:65. [PMID: 28744456 PMCID: PMC5504267 DOI: 10.3389/fcell.2017.00065] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/26/2017] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are important pathophysiological molecules involved in vital cellular processes. They are extremely harmful at high concentrations because they promote the generation of radicals and the oxidation of lipids, proteins, and nucleic acids, which can result in apoptosis. An imbalance of ROS and a disturbance of redox homeostasis are now recognized as a hallmark of complex diseases. Considering that ROS levels are significantly increased in cancer cells due to mitochondrial dysfunction, ROS metabolism has been targeted for the development of efficient treatment strategies, and antioxidants are used as potential chemotherapeutic drugs. However, initial ROS-focused clinical trials in which antioxidants were supplemented to patients provided inconsistent results, i.e., improved treatment or increased malignancy. These different outcomes may result from the highly heterogeneous redox responses of tumors in different patients. Hence, population-based treatment strategies are unsuitable and patient-tailored therapeutic approaches are required for the effective treatment of patients. Moreover, due to the crosstalk between ROS, reducing equivalents [e.g., NAD(P)H] and central metabolism, which is heterogeneous in cancer, finding the best therapeutic target requires the consideration of system-wide approaches that are capable of capturing the complex alterations observed in all of the associated pathways. Systems biology and engineering approaches may be employed to overcome these challenges, together with tools developed in personalized medicine. However, ROS- and redox-based therapies have yet to be addressed by these methodologies in the context of disease treatment. Here, we review the role of ROS and their coupled redox partners in tumorigenesis. Specifically, we highlight some of the challenges in understanding the role of hydrogen peroxide (H2O2), one of the most important ROS in pathophysiology in the progression of cancer. We also discuss its interplay with antioxidant defenses, such as the coupled peroxiredoxin/thioredoxin and glutathione/glutathione peroxidase systems, and its reducing equivalent metabolism. Finally, we highlight the need for system-level and patient-tailored approaches to clarify the roles of these systems and identify therapeutic targets through the use of the tools developed in personalized medicine.
Collapse
Affiliation(s)
- Rui Benfeitas
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden
| | - Jens Nielsen
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden.,Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH Royal Institute of TechnologyStockholm, Sweden.,Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| |
Collapse
|
70
|
Pepke S, Ver Steeg G. Comprehensive discovery of subsample gene expression components by information explanation: therapeutic implications in cancer. BMC Med Genomics 2017; 10:12. [PMID: 28292312 PMCID: PMC5351169 DOI: 10.1186/s12920-017-0245-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 02/08/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND De novo inference of clinically relevant gene function relationships from tumor RNA-seq remains a challenging task. Current methods typically either partition patient samples into a few subtypes or rely upon analysis of pairwise gene correlations that will miss some groups in noisy data. Leveraging higher dimensional information can be expected to increase the power to discern targetable pathways, but this is commonly thought to be an intractable computational problem. METHODS In this work we adapt a recently developed machine learning algorithm for sensitive detection of complex gene relationships. The algorithm, CorEx, efficiently optimizes over multivariate mutual information and can be iteratively applied to generate a hierarchy of relatively independent latent factors. The learned latent factors are used to stratify patients for survival analysis with respect to both single factors and combinations. These analyses are performed and interpreted in the context of biological function annotations and protein network interactions that might be utilized to match patients to multiple therapies. RESULTS Analysis of ovarian tumor RNA-seq samples demonstrates the algorithm's power to infer well over one hundred biologically interpretable gene cohorts, several times more than standard methods such as hierarchical clustering and k-means. The CorEx factor hierarchy is also informative, with related but distinct gene clusters grouped by upper nodes. Some latent factors correlate with patient survival, including one for a pathway connected with the epithelial-mesenchymal transition in breast cancer that is regulated by a microRNA that modulates epigenetics. Further, combinations of factors lead to a synergistic survival advantage in some cases. CONCLUSIONS In contrast to studies that attempt to partition patients into a small number of subtypes (typically 4 or fewer) for treatment purposes, our approach utilizes subgroup information for combinatoric transcriptional phenotyping. Considering only the 66 gene expression groups that are found to both have significant Gene Ontology enrichment and are small enough to indicate specific drug targets implies a computational phenotype for ovarian cancer that allows for 366 possible patient profiles, enabling truly personalized treatment. The findings here demonstrate a new technique that sheds light on the complexity of gene expression dependencies in tumors and could eventually enable the use of patient RNA-seq profiles for selection of personalized and effective cancer treatments.
Collapse
Affiliation(s)
| | - Greg Ver Steeg
- Information Sciences Institute, University of Southern California, Marina Del Rey, USA
| |
Collapse
|
71
|
Chien MN, Yang PS, Lee JJ, Wang TY, Hsu YC, Cheng SP. Recurrence-associated genes in papillary thyroid cancer: An analysis of data from The Cancer Genome Atlas. Surgery 2017; 161:1642-1650. [PMID: 28237646 DOI: 10.1016/j.surg.2016.12.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/18/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Recurrence of papillary thyroid cancer is not uncommon, but incorporating clinicopathologic parameters to predict recurrence is suboptimal. The aim of this study was to identify systemically recurrence-associated genes using The Cancer Genome Atlas RNA sequencing database. METHODS A total of 504 patients with transcriptome sequencing data of the primary neoplasm were included in this study. High and low levels of expression of each gene were defined by median splits. Differences in recurrence-free survival were compared using Kaplan-Meier curves and log-rank tests. Recurrence-associated genes were subjected to functional enrichment analyses with Kyoto Encyclopedia of Genes and Genomes annotation databases and Ingenuity Pathway Analysis. RESULTS We found that 1,807 genes were associated with recurrence-free survival. There were 676 genes of which high expression was associated with a greater risk of recurrence. These genes were enriched in pathways involved in cell cycle regulation and DNA repair. Among 1,131 genes of which low expression was associated with recurrence, Kyoto Encyclopedia of Genes and Genomes-annotated functions were metabolism, calcium signaling, glycan biosynthesis, and the Notch signaling pathway. Canonical pathways identified by Ingenuity Pathway Analysis included RXR function, nitric oxide signaling, interleukin-8 signaling, and nutrient sensing. In addition, low expression of the majority of thyroid differentiation genes was associated with a significantly less recurrence-free survival. CONCLUSION Upregulation of cell cycle-regulating and DNA repair genes appears to have a negative impact on recurrence-free survival in patients with papillary thyroid cancer. Furthermore, recurrence is associated with thyroid dedifferentiation.
Collapse
Affiliation(s)
- Ming-Nan Chien
- Division of Endocrinology and Metabolism, Department of Internal Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Po-Sheng Yang
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan; Graduate Institute of Medical Sciences and Department of Pharmacology, Taipei Medical University, Taipei, Taiwan
| | - Jie-Jen Lee
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan; Graduate Institute of Medical Sciences and Department of Pharmacology, Taipei Medical University, Taipei, Taiwan
| | - Tao-Yeuan Wang
- Department of Pathology, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan; Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Shih-Ping Cheng
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan; Graduate Institute of Medical Sciences and Department of Pharmacology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
72
|
Yang K, Xia B, Wang W, Cheng J, Yin M, Xie H, Li J, Ma L, Yang C, Li A, Fan X, Dhillon HS, Hou Y, Lou G, Li K. A Comprehensive Analysis of Metabolomics and Transcriptomics in Cervical Cancer. Sci Rep 2017; 7:43353. [PMID: 28225065 PMCID: PMC5320559 DOI: 10.1038/srep43353] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/24/2017] [Indexed: 12/20/2022] Open
Abstract
Cervical cancer (CC) still remains a common and deadly malignancy among females in developing countries. More accurate and reliable diagnostic methods/biomarkers should be discovered. In this study, we performed a comprehensive analysis of metabolomics (285 samples) and transcriptomics (52 samples) on the potential diagnostic implication and metabolic characteristic description in cervical cancer. Sixty-two metabolites were different between CC and normal controls (NOR), in which 5 metabolites (bilirubin, LysoPC(17:0), n-oleoyl threonine, 12-hydroxydodecanoic acid and tetracosahexaenoic acid) were selected as candidate biomarkers for CC. The AUC value, sensitivity (SE), and specificity (SP) of these 5 biomarkers were 0.99, 0.98 and 0.99, respectively. We further analysed the genes in 7 significantly enriched pathways, of which 117 genes, that were expressed differentially, were mainly involved in catalytic activity. Finally, a fully connected network of metabolites and genes in these pathways was built, which can increase the credibility of our selected metabolites. In conclusion, our biomarkers from metabolomics could set a path for CC diagnosis and screening. Our results also showed that variables of both transcriptomics and metabolomics were associated with CC.
Collapse
Affiliation(s)
- Kai Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, P.R. China
| | - Bairong Xia
- Department of Gynecology Oncology, the Tumor Hospital, Harbin Medical University, Harbin, 150086, P.R. China
| | - Wenjie Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, P.R. China
| | - Jinlong Cheng
- Department of Gynecology Oncology, the Tumor Hospital, Harbin Medical University, Harbin, 150086, P.R. China
| | - Mingzhu Yin
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation; Translational Research Department of Complex Prescription of TCM, Pharmaceutical University, 639 Longmian Road, Nanjing 211198, P.R. China
| | - Hongyu Xie
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, P.R. China
| | - Junnan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, P.R. China
| | - Libing Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, P.R. China
| | - Chunyan Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, P.R. China
| | - Ang Li
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, P.R. China
| | - Xin Fan
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | | | - Yan Hou
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, P.R. China.,Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, Harbin, 150086, P.R. China
| | - Ge Lou
- Department of Gynecology Oncology, the Tumor Hospital, Harbin Medical University, Harbin, 150086, P.R. China
| | - Kang Li
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, P.R. China
| |
Collapse
|
73
|
Identification of Methylation-Driven, Differentially Expressed STXBP6 as a Novel Biomarker in Lung Adenocarcinoma. Sci Rep 2017; 7:42573. [PMID: 28198450 PMCID: PMC5309775 DOI: 10.1038/srep42573] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 01/12/2017] [Indexed: 02/06/2023] Open
Abstract
DNA methylation is an essential epigenetic marker associated with the silencing of gene expression. Although various genome-wide studies revealed aberrantly methylated gene targets as molecular biomarkers for early detection, the survival rate of lung cancer patients is still poor. In order to identify methylation-driven biomarkers, genome-wide changes in DNA methylation and differential expression in 32 pairs of lung adenocarcinoma and adjacent normal lung tissue in non-smoking women were examined. This concurrent analysis identified 21 negatively correlated probes (r ≤ −0.5), corresponding to 17 genes. Examining the endogenous expression in lung cancer cell lines, five of the genes were found to be significantly down-regulated. Furthermore, in tumor cells alone, 5-aza-2′-deoxycytidine treatment increased the expression levels of STXBP6 in a dose dependent manner and pyrosequencing showed higher percentage of methylation in STXBP6 promoter. Functional analysis revealed that overexpressed STXBP6 in A549 and H1299 cells significantly decreased cell proliferation, colony formation, and migration, and increased apoptosis. Finally, significantly lower survival rates (P < 0.05) were observed when expression levels of STXBP6 were low. Our results provide a basis for the genetic etiology of lung adenocarcinoma by demonstrating the possible role of hypermethylation of STXBP6 in poor clinical outcomes in lung cancer patients.
Collapse
|
74
|
Palmirotta R, Quaresmini D, Lovero D, Silvestris F. ALK gene alterations in cancer: biological aspects and therapeutic implications. Pharmacogenomics 2017; 18:277-292. [PMID: 28112990 DOI: 10.2217/pgs-2016-0166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
ALK was first reported in 1994 as a translocation in anaplastic large cell lymphoma and then described with different abnormalities in a number of tumors. Recently, a shortly accumulated biomedical research clarified the numerous biological processes underlying its ability to support cancer development, growth and progression. Advent of precision medicine has finally provided unexpected advances, leading to the development of ALK-targeting inhibitors with superior efficacy as compared with standard chemotherapy regimens, as well as the identification of resistance mechanisms and the creation of ‘next-generation’ treatments. This review summarizes the current understanding of ALK-driven cancers from the oncogenesis and mutation frequency by The Cancer Genome Atlas database through the diagnostic approach, to an updated portrait of available tyrosine kinase inhibitors, considering their effectiveness in cancer treatment, the molecular reasons of therapeutic failure, and the actual and future ways to overcome resistances.
Collapse
Affiliation(s)
- Raffaele Palmirotta
- Department of Biomedical Sciences & Human Oncology, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Davide Quaresmini
- Department of Biomedical Sciences & Human Oncology, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Domenica Lovero
- Department of Biomedical Sciences & Human Oncology, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Franco Silvestris
- Department of Biomedical Sciences & Human Oncology, University of Bari ‘Aldo Moro’, Bari, Italy
| |
Collapse
|
75
|
Wouters J, Kalender Atak Z, Aerts S. Decoding transcriptional states in cancer. Curr Opin Genet Dev 2017; 43:82-92. [PMID: 28129557 DOI: 10.1016/j.gde.2017.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/05/2017] [Accepted: 01/09/2017] [Indexed: 12/27/2022]
Abstract
Gene regulatory networks determine cellular identity. In cancer, aberrations of gene networks are caused by driver mutations that often affect transcription factors and chromatin modifiers. Nevertheless, gene transcription in cancer follows the same cis-regulatory rules as normal cells, and cancer cells have served as convenient model systems to study transcriptional regulation. Tumours often show regulatory heterogeneity, with subpopulations of cells in different transcriptional states, which has important therapeutic implications. Here, we review recent experimental and computational techniques to reverse engineer cancer gene networks using transcriptome and epigenome data. New algorithms, data integration strategies, and increasing amounts of single cell genomics data provide exciting opportunities to model dynamic regulatory states at unprecedented resolution.
Collapse
Affiliation(s)
- Jasper Wouters
- Laboratory of Computational Biology, VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Human Genetics, KU Leuven (University of Leuven), Leuven, Belgium
| | - Zeynep Kalender Atak
- Laboratory of Computational Biology, VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Human Genetics, KU Leuven (University of Leuven), Leuven, Belgium
| | - Stein Aerts
- Laboratory of Computational Biology, VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Human Genetics, KU Leuven (University of Leuven), Leuven, Belgium.
| |
Collapse
|
76
|
Transcriptome analysis of non-small cell lung cancer and genetically matched adjacent normal tissues identifies novel prognostic marker genes. Genes Genomics 2016. [DOI: 10.1007/s13258-016-0492-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
77
|
Wu X, Li G. Prevalent Accumulation of Non-Optimal Codons through Somatic Mutations in Human Cancers. PLoS One 2016; 11:e0160463. [PMID: 27513638 PMCID: PMC4981346 DOI: 10.1371/journal.pone.0160463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/19/2016] [Indexed: 11/27/2022] Open
Abstract
Cancer is characterized by uncontrolled cell growth, and the cause of different cancers is generally attributed to checkpoint dysregulation of cell proliferation and apoptosis. Recent studies have shown that non-optimal codons were preferentially adopted by genes to generate cell cycle-dependent oscillations in protein levels. This raises the intriguing question of how dynamic changes of codon usage modulate the cancer genome to cope with a non-controlled proliferative cell cycle. In this study, we comprehensively analyzed the somatic mutations of codons in human cancers, and found that non-optimal codons tended to be accumulated through both synonymous and non-synonymous mutations compared with other types of genomic substitution. We further demonstrated that non-optimal codons were prevalently accumulated across different types of cancers, amino acids, and chromosomes, and genes with accumulation of non-optimal codons tended to be involved in protein interaction/signaling networks and encoded important enzymes in metabolic networks that played roles in cancer-related pathways. This study provides insights into the dynamics of codons in the cancer genome and demonstrates that accumulation of non-optimal codons may be an adaptive strategy for cancerous cells to win the competition with normal cells. This deeper interpretation of the patterns and the functional characterization of somatic mutations of codons will help to broaden the current understanding of the molecular basis of cancers.
Collapse
Affiliation(s)
- Xudong Wu
- Laboratory of Molecular Modeling and Design, State key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Rd., Dalian 116023, PR China
| | - Guohui Li
- Laboratory of Molecular Modeling and Design, State key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Rd., Dalian 116023, PR China
- * E-mail:
| |
Collapse
|
78
|
Rana S, Elci SG, Mout R, Singla AK, Yazdani M, Bender M, Bajaj A, Saha K, Bunz UHF, Jirik FR, Rotello VM. Ratiometric Array of Conjugated Polymers-Fluorescent Protein Provides a Robust Mammalian Cell Sensor. J Am Chem Soc 2016; 138:4522-9. [PMID: 26967961 PMCID: PMC5846335 DOI: 10.1021/jacs.6b00067] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Supramolecular complexes of a family of positively charged conjugated polymers (CPs) and green fluorescent protein (GFP) create a fluorescence resonance energy transfer (FRET)-based ratiometric biosensor array. Selective multivalent interactions of the CPs with mammalian cell surfaces caused differential change in FRET signals, providing a fingerprint signature for each cell type. The resulting fluorescence signatures allowed the identification of 16 different cell types and discrimination between healthy, cancerous, and metastatic cells, with the same genetic background. While the CP-GFP sensor array completely differentiated between the cell types, only partial classification was achieved for the CPs alone, validating the effectiveness of the ratiometric sensor. The utility of the biosensor was further demonstrated in the detection of blinded unknown samples, where 121 of 128 samples were correctly identified. Notably, this selectivity-based sensor stratified diverse cell types in minutes, using only 2000 cells, without requiring specific biomarkers or cell labeling.
Collapse
Affiliation(s)
- Subinoy Rana
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
- Department of Materials, Imperial College London, London SW7 2AZ, United Kingdom
| | - S. Gokhan Elci
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Rubul Mout
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Arvind K. Singla
- Department of Biochemistry and Molecular Biology, The McCaig Institute for Bone and Joint Health, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mahdieh Yazdani
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Markus Bender
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld 270, 69120 Heidelberg, FRG
| | - Avinash Bajaj
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, 180 Udyog Vihar, Phase I, Gurgaon-122016, Haryana, India
| | - Krishnendu Saha
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Uwe H. F. Bunz
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld 270, 69120 Heidelberg, FRG
| | - Frank R. Jirik
- Department of Biochemistry and Molecular Biology, The McCaig Institute for Bone and Joint Health, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| |
Collapse
|
79
|
Park SJ, Saito-Adachi M, Komiyama Y, Nakai K. Advances, practice, and clinical perspectives in high-throughput sequencing. Oral Dis 2016; 22:353-64. [DOI: 10.1111/odi.12403] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 01/06/2023]
Affiliation(s)
- S-J Park
- Human Genome Center; The Institute of Medical Science; The University of Tokyo; Tokyo Japan
| | - M Saito-Adachi
- Division of Cancer Genomics; National Cancer Center Research Institute; Tokyo Japan
| | - Y Komiyama
- Human Genome Center; The Institute of Medical Science; The University of Tokyo; Tokyo Japan
| | - K Nakai
- Human Genome Center; The Institute of Medical Science; The University of Tokyo; Tokyo Japan
| |
Collapse
|
80
|
Pappa KI, Polyzos A, Jacob-Hirsch J, Amariglio N, Vlachos GD, Loutradis D, Anagnou NP. Profiling of Discrete Gynecological Cancers Reveals Novel Transcriptional Modules and Common Features Shared by Other Cancer Types and Embryonic Stem Cells. PLoS One 2015; 10:e0142229. [PMID: 26559525 PMCID: PMC4641642 DOI: 10.1371/journal.pone.0142229] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/18/2015] [Indexed: 01/07/2023] Open
Abstract
Studies on individual types of gynecological cancers (GCs), utilizing novel expression technologies, have revealed specific pathogenetic patterns and gene markers for cervical (CC), endometrial (EC) and vulvar cancer (VC). Although the clinical phenotypes of the three types of gynecological cancers are discrete, the fact they originate from a common embryological origin, has led to the hypothesis that they might share common features reflecting regression to early embryogenesis. To address this question, we performed a comprehensive comparative analysis of their profiles. Our data identified both common features (pathways and networks) and novel distinct modules controlling the same deregulated biological processes in all three types. Specifically, four novel transcriptional modules were discovered regulating cell cycle and apoptosis. Integration and comparison of our data with other databases, led to the identification of common features among cancer types, embryonic stem (ES) cells and the newly discovered cell population of squamocolumnar (SC) junction of the cervix, considered to host the early cancer events. Conclusively, these data lead us to propose the presence of common features among gynecological cancers, other types of cancers, ES cells and the pre-malignant SC junction cells, where the novel E2F/NFY and MAX/CEBP modules play an important role for the pathogenesis of gynecological carcinomas.
Collapse
Affiliation(s)
- Kalliopi I. Pappa
- First Department of Obstetrics and Gynecology, University of Athens School of Medicine, Alexandra Hospital, Athens, Greece
- Laboratory of Cell and Gene Therapy, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Alexander Polyzos
- Basic Research Centre, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Jasmine Jacob-Hirsch
- Cancer Research Center, The Chaim Sheba Medical Center, Tel-Hashomer and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ninette Amariglio
- Cancer Research Center, The Chaim Sheba Medical Center, Tel-Hashomer and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - George D. Vlachos
- First Department of Obstetrics and Gynecology, University of Athens School of Medicine, Alexandra Hospital, Athens, Greece
| | - Dimitrios Loutradis
- First Department of Obstetrics and Gynecology, University of Athens School of Medicine, Alexandra Hospital, Athens, Greece
| | - Nicholas P. Anagnou
- Laboratory of Cell and Gene Therapy, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Laboratory of Biology, University of Athens School of Medicine, Athens, Greece
- * E-mail:
| |
Collapse
|