1
|
Liu Q, Zhang X, Song Y, Si J, Li Z, Dong Q. Construction and analysis of a reliable five-gene prognostic signature for colon adenocarcinoma associated with the wild-type allelic state of the COL6A6 gene. Transl Cancer Res 2024; 13:2475-2496. [PMID: 38881933 PMCID: PMC11170513 DOI: 10.21037/tcr-23-463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 11/29/2023] [Indexed: 06/18/2024]
Abstract
Background Tumors emerge by acquiring a number of mutations over time. The first mutation provides a selective growth advantage compared to adjacent epithelial cells, allowing the cell to create a clone that can outgrow the cells that surround it. Subsequent mutations determine the risk of the tumor progressing to metastatic cancer. Some secondary mutations may inhibit the aggressiveness of the tumor while still increasing the survival of the clone. Meaningful mutations in genes may provide a strong molecular foundation for developing novel therapeutic strategies for cancer. Methods The somatic mutation and prognosis in colon adenocarcinoma (COAD) were analyzed. The copy number variation (CNV) and differentially expressed genes (DEGs) between the collagen type VI alpha 6 chain (COL6A6) mutation (COL6A6-MUT) and the COL6A6 wild-type (COL6A6-WT) subgroups were evaluated. The independent prognostic signatures based on COL6A6-allelic state were determined to construct a Cox model. The biological characteristics and the immune microenvironment between the two risk groups were compared. Results COL6A6 was found to be highly mutated in COAD at a frequency of 9%. Patients with COL6A6-MUT had a good overall survival (OS) compared to those with COL6A6-WT, who had a different CNV pattern. Significant differences in gene expression were established for 593 genes between the COL6A6-MUT and COL6A6-WT samples. Among them, MUC16, ASNSP1, PRR18, PEG10, and RPL26P8 were determined to be independent prognostic factors. The internally validated prognostic risk model, constructed using these five genes, demonstrated its value by revealing a significant difference in patient prognosis between the high-risk and low-risk groups. Specifically, patients in the high-risk group exhibited a considerably worse prognosis than did those in the low-risk group. The high-risk group had a significantly higher proportion of patients over 60 years of age and patients in stage III. Moreover, the tumor immune dysfunction and exclusion (TIDE) score and the expression of human leukocyte antigen (HLA) family genes were all higher in the high-risk group than that in the low-risk group. Conclusions The allelic state of COL6A6 and the five associated DEGs were identified as novel biomarkers for the diagnosis and prognosis of COAD and may be therapeutic targets in COAD.
Collapse
Affiliation(s)
- Qun Liu
- Second Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Xiaohua Zhang
- Gastroenterology Center, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, China
| | - Yan Song
- Outpatient Department, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, China
| | - Junli Si
- Second Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Zhaoshui Li
- Qingdao University, Qingdao Medical College, Qingdao, China
| | - Quanjiang Dong
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| |
Collapse
|
2
|
Wang Y, Wan X, Du S. Integrated analysis revealing a novel stemness-metabolism-related gene signature for predicting prognosis and immunotherapy response in hepatocellular carcinoma. Front Immunol 2023; 14:1100100. [PMID: 37622118 PMCID: PMC10445950 DOI: 10.3389/fimmu.2023.1100100] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 07/10/2023] [Indexed: 08/26/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant lethal tumor and both cancer stem cells (CSCs) and metabolism reprogramming have been proven to play indispensable roles in HCC. This study aimed to reveal the connection between metabolism reprogramming and the stemness characteristics of HCC, established a new gene signature related to stemness and metabolism and utilized it to assess HCC prognosis and immunotherapy response. The clinical information and gene expression profiles (GEPs) of 478 HCC patients came from the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA). The one-class logistic regression (OCLR) algorithm was employed to calculate the messenger ribonucleic acid expression-based stemness index (mRNAsi), a new stemness index quantifying stemness features. Differentially expressed analyses were done between high- and low-mRNAsi groups and 74 differentially expressed metabolism-related genes (DEMRGs) were identified with the help of metabolism-related gene sets from Molecular Signatures Database (MSigDB). After integrated analysis, a risk score model based on the three most efficient prognostic DEMRGs, including Recombinant Phosphofructokinase Platelet (PFKP), phosphodiesterase 2A (PDE2A) and UDP-glucuronosyltransferase 1A5 (UGT1A5) was constructed and HCC patients were divided into high-risk and low-risk groups. Significant differences were found in pathway enrichment, immune cell infiltration patterns, and gene alterations between the two groups. High-risk group patients tended to have worse clinical outcomes and were more likely to respond to immunotherapy. A stemness-metabolism-related model composed of gender, age, the risk score model and tumor-node-metastasis (TNM) staging was generated and showed great discrimination and strong ability in predicting HCC prognosis and immunotherapy response.
Collapse
Affiliation(s)
| | | | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| |
Collapse
|
3
|
Fan P, Wang J, Li R, Chang K, Liu L, Wang Y, Wang Z, Zhang B, Ji C, Zhang J, Chen S, Ling R. Development and validation of an endoplasmic reticulum stress-related molecular prognostic model for breast cancer. Front Oncol 2023; 13:1178595. [PMID: 37313465 PMCID: PMC10258344 DOI: 10.3389/fonc.2023.1178595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/15/2023] [Indexed: 06/15/2023] Open
Abstract
Background Breast cancer is the most frequently diagnosed cancer and a leading cause of cancer-related death in women. Endoplasmic reticulum stress (ERS) plays a crucial role in the pathogenesis of several malignancies. However, the prognostic value of ERS-related genes in breast cancer has not been thoroughly investigated. Methods We downloaded and analyzed expression profiling data for breast invasive carcinoma samples in The Cancer Genome Atlas-Breast Invasive Carcinoma (TCGA-BRCA) and identified 23 ERS-related genes differentially expressed between the normal breast tissue and primary breast tumor tissues. We constructed and validated risk models using external test datasets. We assessed the differences in sensitivity to common antitumor drugs between high- and low-scoring groups using the Genomics of Drug Sensitivity in Cancer (GDSC) database, evaluated the sensitivity of patients in high- and low-scoring groups to immunotherapy using the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm, and assessed immune and stromal cell infiltration in the tumor microenvironment (TME) using the Estimation of Stromal and Immune cells in Malignant Tumor tissues using Expression data (ESTIMATE) algorithm. We also analyzed the expression of independent factors in the prognostic model using the Western-blot analysis for correlation in relation to breast cancer. Results Using multivariate Cox analysis, FBXO6, PMAIP1, ERP27, and CHAC1 were identified as independent prognostic factors in patients with breast cancer. The risk score in our model was defined as the endoplasmic reticulum score (ERScore). ERScore had high predictive power for overall survival in patients with breast cancer. The high-ERScore group exhibited a worse prognosis, lower drug sensitivity, and lower immunotherapy response and immune infiltration than did the low-ERScore group. Conclusions based on ERScore were consistent with Western-blot results. Conclusion We constructed and validated for the first time an endoplasmic reticulum stress-related molecular prognostic model for breast cancer with reliable predictive properties and good sensitivity, as an important addition to the prognostic prediction model for breast cancer.
Collapse
Affiliation(s)
- Pengyu Fan
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jiajia Wang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi’an, China
| | - Ruolei Li
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Kexin Chang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Liuyin Liu
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yaping Wang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhe Wang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Bo Zhang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Cheng Ji
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jian Zhang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi’an, China
| | - Suning Chen
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Rui Ling
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
4
|
Yu Y, Liu Y, Li Y, Yang X, Han M, Fan Q. Construction of a CCL20-centered circadian-signature based prognostic model in cervical cancer. Cancer Cell Int 2023; 23:92. [PMID: 37183243 PMCID: PMC10184429 DOI: 10.1186/s12935-023-02926-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
BACKGROUND Rather low vaccination rates for Human papillomavirus (HPV) and pre-existing cervical cancer patients with limited therapeutic strategies ask for more precise prognostic model development. On the other side, the clinical significance of circadian clock signatures in cervical cancer lacks investigation. METHODS Subtypes classification based upon eight circadian clock core genes were implemented in TCGA-CESC through k-means clustering methods. Afterwards, KEGG, GO and GSEA analysis were conducted upon differentially expressed genes (DEGs) between high and low-risk groups, and tumor microenvironment (TME) investigation by CIBERSORT and ESTIMATE. Furthermore, a prognostic model was developed by cox and lasso regression methods, and verified in GSE44001 by time-dependent receiver-operating characteristic curve (ROC) analysis. Lastly, FISH and IHC were used for validation of CCL20 expression in patients' specimens and U14 subcutaneous tumor models were built for TME composition. RESULTS We successfully classified cervical patients into high-risk and low-risk groups based upon circadian-oscillation-signatures. Afterwards, we built a prognostic risk model composed of GJB2, CCL20 and KRT24 with excellent predictive value on patients' overall survival (OS). We then proposed metabolism unbalance, especially for glycolysis, and immune related pathways to be major enriched signatures between the high-risk and low-risk groups. Then, we proposed an 'immune-desert'-like suppressive myeloid cells infiltration pattern in high-risk group TME and verified its resistance to immunotherapies. Finally, CCL20 was proved positively correlated with real-world patients' stages and induced significant less CD8+ T cells and more M2 macrophages infiltration in mouse model. CONCLUSIONS We unraveled a prognostic risk model based upon circadian oscillation and verified its solidity. Specifically, we unveiled distinct TME immune signatures in high-risk groups.
Collapse
Affiliation(s)
- Yuchong Yu
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty of Gynecologic Oncology, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases Affifiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Liu
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty of Gynecologic Oncology, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases Affifiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Li
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty of Gynecologic Oncology, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases Affifiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoming Yang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty of Gynecologic Oncology, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases Affifiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mi Han
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Municipal Key Clinical Specialty of Gynecologic Oncology, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases Affifiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qiong Fan
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Municipal Key Clinical Specialty of Gynecologic Oncology, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases Affifiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Lu X, Ying Y, Zhang W, Zhang J, Li R, Wang W. Prognosis, immune microenvironment, and personalized treatment prediction in Rho GTPase-activating protein 4-mutant cervical cancer: Computer strategies for precision oncology. Life Sci 2023; 315:121360. [PMID: 36608869 DOI: 10.1016/j.lfs.2022.121360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023]
Abstract
AIMS Cervical cancer with different mutations is associated with specific genomic differences. We developed a new mutation prediction model of the ARHGAP4 gene for cervical cancer. MAIN METHODS We conducted a panoramic analysis of CESC mutations based on The Cancer Genome Atlas-Cervical Squamous Cell Carcinoma and Endocervical Adenocarcinoma (TCGA-CESC) database. We made copy number variation analysis and correlation analysis of somatic mutations and tumor mutation load fraction. Then we established a prediction model of ARHGAP4 mutation, screened related genes based on the risk scores, calculated the correlation between the risk score and immune microenvironment, and analyzed drug sensitivity. KEY FINDINGS The prediction model of ARHGAP4 mutation based on mRNA expression is closely related to the survival rate of cervical cancer patients and to the effect of immunotherapy. The prediction model is also related to the infiltration of immune cells and human leukocyte antigen family expression in the immune microenvironment. After computational analysis, three drugs (cytarabine, docetaxel, imatinib) were identified as potential agents for the ARHGAP4 mutation high-risk group, and two drugs (erlotinib, methotrexate) were shown to have therapeutic significance for patients in the low-risk group. The expression of ARHGAP4 was higher in cervical cancer tissues. The proliferation ability of HeLa and SiHa cells decreased after ARHGAP4 knockdown. SIGNIFICANCE This study provides not only a new approach for the prediction of the response of the cervical cancer patients to targeted drug therapy but also a new strategy for combining risk stratification with precision treatment.
Collapse
Affiliation(s)
- Xiaoqin Lu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Zhengzhou University, 2nd, Jingba Road, Zhengzhou, Henan Province 450053, China
| | - Yanqi Ying
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Zhengzhou University, 2nd, Jingba Road, Zhengzhou, Henan Province 450053, China
| | - Wenyi Zhang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Zhengzhou University, 2nd, Jingba Road, Zhengzhou, Henan Province 450053, China
| | - Jingyan Zhang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Zhengzhou University, 2nd, Jingba Road, Zhengzhou, Henan Province 450053, China
| | - Rui Li
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Zhengzhou University, 2nd, Jingba Road, Zhengzhou, Henan Province 450053, China
| | - Wuliang Wang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Zhengzhou University, 2nd, Jingba Road, Zhengzhou, Henan Province 450053, China.
| |
Collapse
|
6
|
Hao S, Huang M, Xu X, Wang X, Huo L, Wang L, Gu J. MDN1 Mutation Is Associated With High Tumor Mutation Burden and Unfavorable Prognosis in Breast Cancer. Front Genet 2022; 13:857836. [PMID: 35386280 PMCID: PMC8978890 DOI: 10.3389/fgene.2022.857836] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/28/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Breast cancer (BRCA) is the most common cancer worldwide and a serious threat to human health. MDN1 mutations have been observed in several cancers. However, the associations of MDN1 mutation with tumor mutation burden (TMB) and prognosis of BRCA have not been investigated. Methods: Genomic, transcriptomic, and clinical data of 973 patients with BRCA from The Cancer Genome Atlas (TCGA) database were analyzed. The clinical attributes of BRCA based on the MDN1 mutation status were assessed by comparing TMB and tumor infiltrating immune cells. Gene ontology analysis and gene set enrichment analysis (GSEA) were conducted to identify the key signaling pathways associated with MDN1 mutation. Moreover, univariate and multivariate Cox regression analyses were performed to assess the association between prognostic factors and survival outcomes. Finally, nomograms were used to determine the predictive value of MDN1 mutation on clinical outcomes in patients with BRCA. Results: MDN1 was found to have a relatively high mutation rate (2.77%). Compared to the MDN1 wild-type patients, the TMB value was significantly higher in MDN1 mutant patients (p < 0.001). Prognostic analysis revealed that MDN1 mutant patients had a worse survival probability than MDN1 wild-type patients (hazard ratio = 2.91; 95% CI:1.07–7.92; p = 0.036). GSEA revealed samples with MDN1 mutation enriched in retinol metabolism, drug metabolism cytochrome P450, glucuronidation, miscellaneous transport, and binding event pathways. Conclusion: MDN1 mutation was found to be associated with high TMB and inferior prognosis, suggesting that MDN1 mutation may play a potential role in prognosis prediction and immunotherapy guidance in BRCA.
Collapse
Affiliation(s)
- Shuai Hao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Miao Huang
- Nursing School, Chongqing Medical University, Chongqing, China
| | - Xiaofan Xu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xulin Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Liqun Huo
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lu Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jun Gu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
7
|
Engineering for an HPV 9-valent vaccine candidate using genomic constitutive over-expression and low lipopolysaccharide levels in Escherichia coli cells. Microb Cell Fact 2021; 20:227. [PMID: 34930257 PMCID: PMC8686089 DOI: 10.1186/s12934-021-01719-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Background The various advantages associated with the growth properties of Escherichia coli have justified their use in the production of genetically engineered vaccines. However, endotoxin contamination, plasmid vector instability, and the requirement for antibiotic supplementation are frequent bottlenecks in the successful production of recombinant proteins that are safe for industrial-scaled applications. To overcome these drawbacks, we focused on interrupting the expression of several key genes involved in the synthesis of lipopolysaccharide (LPS), an endotoxin frequently responsible for toxicity in recombinant proteins, to eliminate endotoxin contamination and produce better recombinant proteins with E. coli. Results Of 8 potential target genes associated with LPS synthesis, we successfully constructed 7 LPS biosynthesis-defective recombinant strains to reduce the production of LPS. The endotoxin residue in the protein products from these modified E. coli strains were about two orders of magnitude lower than that produced by the wild-type strain. Further, we found that 6 loci—lpxM, lpxP, lpxL, eptA, gutQ and kdsD—were suitable for chromosomal integrated expression of HPV L1 protein. We found that a single copy of the expression cassette conferred stable expression during long-term antibiotic-free cultivation as compared with the more variable protein production from plasmid-based expression. In large-scale fermentation, we found that recombinant strains bearing 3 to 5 copies of the expression cassette had 1.5- to 2-fold higher overall expression along with lower endotoxin levels as compared with the parental ER2566 strain. Finally, we engineered and constructed 9 recombinant E. coli strains for the later production of an HPV 9-valent capsid protein with desirable purity, VLP morphology, and antigenicity. Conclusions Reengineering the LPS synthesis loci in the E. coli ER2566 strain through chromosomal integration of expression cassettes has potential uses for the production of a 9-valent HPV vaccine candidate, with markedly reduced residual endotoxin levels. Our results offer a new strategy for recombinant E. coli strain construction, engineering, and the development of suitable recombinant protein drugs. Supplementary Information The online version contains supplementary material available at 10.1186/s12934-021-01719-8.
Collapse
|
8
|
Maura F, Bolli N, Angelopoulos N, Dawson KJ, Leongamornlert D, Martincorena I, Mitchell TJ, Fullam A, Gonzalez S, Szalat R, Abascal F, Rodriguez-Martin B, Samur MK, Glodzik D, Roncador M, Fulciniti M, Tai YT, Minvielle S, Magrangeas F, Moreau P, Corradini P, Anderson KC, Tubio JMC, Wedge DC, Gerstung M, Avet-Loiseau H, Munshi N, Campbell PJ. Genomic landscape and chronological reconstruction of driver events in multiple myeloma. Nat Commun 2019; 10:3835. [PMID: 31444325 PMCID: PMC6707220 DOI: 10.1038/s41467-019-11680-1] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/23/2019] [Indexed: 01/11/2023] Open
Abstract
The multiple myeloma (MM) genome is heterogeneous and evolves through preclinical and post-diagnosis phases. Here we report a catalog and hierarchy of driver lesions using sequences from 67 MM genomes serially collected from 30 patients together with public exome datasets. Bayesian clustering defines at least 7 genomic subgroups with distinct sets of co-operating events. Focusing on whole genome sequencing data, complex structural events emerge as major drivers, including chromothripsis and a novel replication-based mechanism of templated insertions, which typically occur early. Hyperdiploidy also occurs early, with individual trisomies often acquired in different chronological windows during evolution, and with a preferred order of acquisition. Conversely, positively selected point mutations, whole genome duplication and chromoplexy events occur in later disease phases. Thus, initiating driver events, drawn from a limited repertoire of structural and numerical chromosomal changes, shape preferred trajectories of evolution that are biologically relevant but heterogeneous across patients.
Collapse
Affiliation(s)
- Francesco Maura
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
- Department of Medical Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Niccoló Bolli
- Department of Medical Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nicos Angelopoulos
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
- School of Computer Science and Electronic Engineering, University of Essex, Colchester, UK
| | - Kevin J Dawson
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Daniel Leongamornlert
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Inigo Martincorena
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Thomas J Mitchell
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Anthony Fullam
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Santiago Gonzalez
- European Bioinformatics Institute, European Molecular Biology Laboratory (EMBL-EBI), Hinxton, UK
| | - Raphael Szalat
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Federico Abascal
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Bernardo Rodriguez-Martin
- CIMUS - Molecular Medicine and Chronic Diseases Research Centre, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Mehmet Kemal Samur
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Dominik Glodzik
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
- Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marco Roncador
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Mariateresa Fulciniti
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yu Tzu Tai
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Stephane Minvielle
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Florence Magrangeas
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Philippe Moreau
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Paolo Corradini
- Department of Medical Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jose M C Tubio
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
- CIMUS - Molecular Medicine and Chronic Diseases Research Centre, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - David C Wedge
- University of Oxford, Big Data Institute, Oxford, UK
| | - Moritz Gerstung
- European Bioinformatics Institute, European Molecular Biology Laboratory (EMBL-EBI), Hinxton, UK
| | | | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Veterans Administration Boston Healthcare System, West Roxbury, MA, USA.
| | - Peter J Campbell
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK.
| |
Collapse
|
9
|
Gendoo DMA, Zon M, Sandhu V, Manem VSK, Ratanasirigulchai N, Chen GM, Waldron L, Haibe-Kains B. MetaGxData: Clinically Annotated Breast, Ovarian and Pancreatic Cancer Datasets and their Use in Generating a Multi-Cancer Gene Signature. Sci Rep 2019; 9:8770. [PMID: 31217513 PMCID: PMC6584731 DOI: 10.1038/s41598-019-45165-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/31/2019] [Indexed: 12/13/2022] Open
Abstract
A wealth of transcriptomic and clinical data on solid tumours are under-utilized due to unharmonized data storage and format. We have developed the MetaGxData package compendium, which includes manually-curated and standardized clinical, pathological, survival, and treatment metadata across breast, ovarian, and pancreatic cancer data. MetaGxData is the largest compendium of curated transcriptomic data for these cancer types to date, spanning 86 datasets and encompassing 15,249 samples. Open access to standardized metadata across cancer types promotes use of their transcriptomic and clinical data in a variety of cross-tumour analyses, including identification of common biomarkers, and assessing the validity of prognostic signatures. Here, we demonstrate that MetaGxData is a flexible framework that facilitates meta-analyses by using it to identify common prognostic genes in ovarian and breast cancer. Furthermore, we use the data compendium to create the first gene signature that is prognostic in a meta-analysis across 3 cancer types. These findings demonstrate the potential of MetaGxData to serve as an important resource in oncology research, and provide a foundation for future development of cancer-specific compendia.
Collapse
Affiliation(s)
- Deena M A Gendoo
- Centre for Computational Biology, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom.
| | - Michael Zon
- Princess Margaret Cancer Center, University Health Network, Toronto, M5G 2C1, Canada.,Department of Biomedical Engineering, McMaster University, Toronto, L8S 4L8, Canada
| | - Vandana Sandhu
- Princess Margaret Cancer Center, University Health Network, Toronto, M5G 2C1, Canada
| | - Venkata S K Manem
- Princess Margaret Cancer Center, University Health Network, Toronto, M5G 2C1, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, M5S 3H7, Canada.,Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, G1V 4G5, Canada
| | | | - Gregory M Chen
- Princess Margaret Cancer Center, University Health Network, Toronto, M5G 2C1, Canada
| | - Levi Waldron
- Graduate School of Public Health and Health Policy, Institute of Implementation Science in Population Health, City University of New York School, New York, 11101, USA.
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Center, University Health Network, Toronto, M5G 2C1, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, M5S 3H7, Canada. .,Department of Computer Science, University of Toronto, Toronto, M5T 3A1, Canada. .,Ontario Institute of Cancer Research, Toronto, M5G 0A3, Canada. .,Vector Institute, Toronto, M5G 1M1, Canada.
| |
Collapse
|
10
|
Pelekanou V, Anastasiou E, Bakogeorgou E, Notas G, Kampa M, Garcia-Milian R, Lavredaki K, Moustou E, Chinari G, Arapantoni P, O'Grady A, Georgoulias V, Tsapis A, Stathopoulos EN, Castanas E. Estrogen receptor-alpha isoforms are the main estrogen receptors expressed in non-small cell lung carcinoma. Steroids 2019; 142:65-76. [PMID: 29454903 DOI: 10.1016/j.steroids.2018.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 11/21/2017] [Accepted: 01/18/2018] [Indexed: 01/10/2023]
Abstract
The expression profile of estrogen receptors (ER) in Non-Small Cell Lung Carcinoma (NSCLC) remains contradictory. Here we investigated protein and transcriptome expression of ERα wild type and variants. Tissue Micro-Arrays of 200 cases of NSCLC (paired tumor/non-tumor) were assayed by immunohistochemistry using a panel of ERα antibodies targeting different epitopes (HC20, 6F11, 1D5, ERα36 and ERα17p). ERβ epitopes were also examined for comparison. In parallel we conducted a probe-set mapping (Affymetrix HGU133 plus 2 chip) meta-analysis of 12 NSCLC tumor public transcriptomic studies (1418 cases) and 39 NSCLC cell lines. Finally, we have investigated early transcriptional effects of 17β-estradiol, 17β-estradiol-BSA, tamoxifen and their combination in two NSCLC cell lines (A549, H520). ERα transcript and protein detection in NSCLC specimens and cell lines suggests that extranuclear ERα variants, like ERα36, prevail, while wild-type ERα66 is minimally expressed. In non-tumor lung, the wild-type ERα66 is quasi-absent. The combined evaluation of ERα isoform staining intensity and subcellular localization with sex, can discriminate NSCLC subtypes and normal lung. Overall ERα transcription decreases in NSCLC. ERα expression is sex-related in non-tumor tissue, but in NSCLC it is exclusively correlating with tumor histologic subtype. ERα isoform protein expression is higher than ERβ. ERα isoforms are functional and display specific early transcriptional effects following steroid treatment. In conclusion, our data show a wide extranuclear ERα-variant expression in normal lung and NSCLC that is not reported by routine pathology ER evaluation criteria, limited in the nuclear wild type receptor.
Collapse
Affiliation(s)
- Vasiliki Pelekanou
- Laboratory of Pathology, School of Medicine, University of Crete, Heraklion, Greece; Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion Greece; Department of Pathology, Yale School of Medicine, New Haven, CT, United States.
| | - Eleftheria Anastasiou
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion Greece
| | - Efstathia Bakogeorgou
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion Greece
| | - George Notas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion Greece
| | - Marilena Kampa
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion Greece
| | | | - Katerina Lavredaki
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion Greece
| | - Eleni Moustou
- Laboratory of Pathology, School of Medicine, University of Crete, Heraklion, Greece
| | | | | | - Anthony O'Grady
- Molecular Histopathology Laboratory, Dept. of Pathology, Royal College of Surgeons of Ireland (RCSI), Education & Research Centre, Dublin, Ireland; Beaumont Hospital, Dublin, Ireland
| | | | - Andreas Tsapis
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion Greece; INSERM U976, Hôpital Saint Louis, Paris, France; Université Paris Diderot, Paris, France
| | | | - Elias Castanas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion Greece
| |
Collapse
|
11
|
Lim SB, Tan SJ, Lim WT, Lim CT. A merged lung cancer transcriptome dataset for clinical predictive modeling. Sci Data 2018; 5:180136. [PMID: 30040079 PMCID: PMC6057440 DOI: 10.1038/sdata.2018.136] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/26/2018] [Indexed: 12/01/2022] Open
Abstract
The Gene Expression Omnibus (GEO) database is an excellent public source of whole transcriptomic profiles of multiple cancers. The main challenge is the limited accessibility of such large-scale genomic data to people without a background in bioinformatics or computer science. This presents difficulties in data analysis, sharing and visualization. Here, we present an integrated bioinformatics pipeline and a normalized dataset that has been preprocessed using a robust statistical methodology; allowing others to perform large-scale meta-analysis, without having to conduct time-consuming data mining and statistical correction. Comprising 1,118 patient-derived samples, the normalized dataset includes primary non-small cell lung cancer (NSCLC) tumors and paired normal lung tissues from ten independent GEO datasets, facilitating differential expression analysis. The data has been merged, normalized, batch effect-corrected and filtered for genes with low variance via multiple open source R packages integrated into our workflow. Overall this dataset (with associated clinical metadata) better represents the diseased population and serves as a powerful tool for early predictive biomarker discovery.
Collapse
Affiliation(s)
- Su Bin Lim
- NUS Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, #05-01, 28 Medical Drive, Singapore 117456, Singapore
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore 117583, Singapore
| | - Swee Jin Tan
- Sysmex Asia Pacific Pte Ltd, 9 Tampines Grande, #06-18, Singapore 528735, Singapore
| | - Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore, 169610 Singapore
- Office of Clinical Sciences, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Institute of Molecular and Cell Biology, A*Star, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Chwee Teck Lim
- NUS Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, #05-01, 28 Medical Drive, Singapore 117456, Singapore
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore 117583, Singapore
- Mechanobiology Institute, National University of Singapore, #10-01, 5A Engineering Drive 1, Singapore 117411, Singapore
- Biomedical Institute for Global Health Research and Technology, National University of Singapore, #14-01, MD6, 14 Medical Drive, Singapore 117599, Singapore
| |
Collapse
|
12
|
Cieślik M, Chinnaiyan AM. Cancer transcriptome profiling at the juncture of clinical translation. Nat Rev Genet 2017; 19:93-109. [PMID: 29279605 DOI: 10.1038/nrg.2017.96] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Methodological breakthroughs over the past four decades have repeatedly revolutionized transcriptome profiling. Using RNA sequencing (RNA-seq), it has now become possible to sequence and quantify the transcriptional outputs of individual cells or thousands of samples. These transcriptomes provide a link between cellular phenotypes and their molecular underpinnings, such as mutations. In the context of cancer, this link represents an opportunity to dissect the complexity and heterogeneity of tumours and to discover new biomarkers or therapeutic strategies. Here, we review the rationale, methodology and translational impact of transcriptome profiling in cancer.
Collapse
Affiliation(s)
- Marcin Cieślik
- Michigan Center for Translational Pathology, University of Michigan.,Department of Pathology, University of Michigan
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan.,Department of Pathology, University of Michigan.,Comprehensive Cancer Center, University of Michigan.,Department of Urology, University of Michigan.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
13
|
Duyck K, DuTell V, Ma L, Paulson A, Yu CR. Pronounced strain-specific chemosensory receptor gene expression in the mouse vomeronasal organ. BMC Genomics 2017; 18:965. [PMID: 29233099 PMCID: PMC5727874 DOI: 10.1186/s12864-017-4364-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/01/2017] [Indexed: 01/07/2023] Open
Abstract
Background The chemosensory system plays an important role in orchestrating sexual behaviors in mammals. Pheromones trigger sexually dimorphic behaviors and different mouse strains exhibit differential responses to pheromone stimuli. It has been speculated that differential gene expression in the sensory organs that detect pheromones may underlie sexually-dimorphic and strain-specific responses to pheromone cues. Results We have performed transcriptome analyses of the mouse vomeronasal organ, a sensory organ recognizing pheromones and interspecies cues. We find little evidence of sexual dimorphism in gene expression except for Xist, an essential gene for X-linked gene inactivation. Variations in gene expression are found mainly among strains, with genes from immune response and chemosensory receptor classes dominating the list. Differentially expressed genes are concentrated in genomic hotspots enriched in these families of genes. Some chemosensory receptors show exclusive patterns of expression in different strains. We find high levels of single nucleotide polymorphism in chemosensory receptor pseudogenes, some of which lead to functionalized receptors. Moreover, we identify a number of differentially expressed long noncoding RNA species showing strong correlation or anti-correlation with chemoreceptor genes. Conclusions Our analyses provide little evidence supporting sexually dimorphic gene expression in the vomeronasal organ that may underlie dimorphic pheromone responses. In contrast, we find pronounced variations in the expression of immune response related genes, vomeronasal and G-protein coupled receptor genes among different mouse strains. These findings raised the possibility that diverse strains of mouse perceive pheromone cues differently and behavioral difference among strains in response to pheromone may first arise from differential detection of pheromones. On the other hand, sexually dimorphic responses to pheromones more likely originate from dimorphic neural circuits in the brain than from differential detection. Moreover, noncoding RNA may offer a potential regulatory mechanism controlling the differential expression patterns. Electronic supplementary material The online version of this article (10.1186/s12864-017-4364-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kyle Duyck
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO, 64110, USA
| | - Vasha DuTell
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO, 64110, USA.,Redwood Center for Theoretical Neuroscience, University of California, 567 Evans Hall, Berkeley, 94720, USA
| | - Limei Ma
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO, 64110, USA
| | - Ariel Paulson
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO, 64110, USA
| | - C Ron Yu
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO, 64110, USA. .,Department of Anatomy and Cell Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| |
Collapse
|
14
|
Lim SB, Tan SJ, Lim WT, Lim CT. An extracellular matrix-related prognostic and predictive indicator for early-stage non-small cell lung cancer. Nat Commun 2017; 8:1734. [PMID: 29170406 PMCID: PMC5700969 DOI: 10.1038/s41467-017-01430-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/18/2017] [Indexed: 12/17/2022] Open
Abstract
The prognosis and prediction of adjuvant chemotherapy (ACT) response in early-stage non-small cell lung cancer (NSCLC) patients remain poor in this era of personalized medicine. We hypothesize that extracellular matrix (ECM)-associated components could be potential markers for better diagnosis and prognosis due to their differential expression in 1,943 primary NSCLC tumors as compared to 303 normal lung tissues. Here we develop a 29-gene ECM-related prognostic and predictive indicator (EPPI). We validate a robust performance of the EPPI risk scoring system in multiple independent data sets, comprising a total of 2,071 early-stage NSCLC tumors. Patients are stratified according to the universal cutoff score based on the EPPI when applied in the clinical setting; the low-risk group has significantly better survival outcome. The functional EPPI gene set represents a potential genomic tool to improve patient selection in early-stage NSCLC to further derive the best benefits of ACT and prevent unnecessary treatment or ACT-associated morbidity. Prognosis and prediction of adjuvant chemotherapy response in non-small cell lung cancer can have significant clinical impact. Here, the authors show that differential expression of a 29 extracellular matrix gene indicator, EPPI, can predict patient outcome.
Collapse
Affiliation(s)
- Su Bin Lim
- NUS Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, #05-01, 28 Medical Drive, Singapore, 117456, Singapore.,Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore, 117583, Singapore
| | - Swee Jin Tan
- Clearbridge Accelerator Pte Ltd, Singapore, 118257, Singapore.,Sysmex Asia Pacific Pte Ltd, 9 Tampines Grande, #06-18, Singapore, 528735, Singapore
| | - Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore, 169610, Singapore.,Office of Clinical Sciences, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.,Institute of Molecular and Cell Biology, A*Star, 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Chwee Teck Lim
- NUS Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, #05-01, 28 Medical Drive, Singapore, 117456, Singapore. .,Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore, 117583, Singapore. .,Mechanobiology Institute, National University of Singapore, #10-01, 5A Engineering Drive 1, Singapore, 117411, Singapore. .,Biomedical Institute for Global Health Research and Technology, National University of Singapore, #14-01, MD6, 14 Medical Drive, Singapore, 117599, Singapore.
| |
Collapse
|
15
|
Li Y, Xia Y, Han M, Chen G, Zhang D, Thasler WE, Protzer U, Ning Q. IFN-α-mediated Base Excision Repair Pathway Correlates with Antiviral Response Against Hepatitis B Virus Infection. Sci Rep 2017; 7:12715. [PMID: 28983111 PMCID: PMC5629255 DOI: 10.1038/s41598-017-13082-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/18/2017] [Indexed: 01/05/2023] Open
Abstract
Previous studies identified APOBEC deaminases as enzymes targeting hepatitis B virus (HBV) DNA in the nucleus thus affecting its persistence. Interferon (IFN)-α treated chimpanzees and hepatitis C patients showed elevated APOBEC expression. We thus hypothesized that the responses to IFN-α treatment of chronic hepatitis B (CHB) patients is influenced by IFN-induced base excision repair (BER). CHB-treatment naïve patients, patients treated with PEGylated IFN-α, and patients with sequential treatment of Entecavior and PEGylated IFN-α were recruited. Blood and liver biopsy samples were collected before treatment and at treatment endpoint. BER genes were assessed by quantitative RT-PCR. BER gene expression levels and IFN treatment responses were correlated in patient liver biopsies. APOBEC3A, -B, -C, -D/E, and-G mRNA levels were up-regulated in IFN-treated patients. APOBEC3A expression was significantly higher in IFN-responders than in non-responders. BER genes NEIL3 was down-regulated in IFN-treated patients. APOBEC3 and BER gene expression at treatment endpoints partially correlated with the corresponding absolute DNA level or degree of HBsAg and HBV DNA decline. Our study suggests that the expression of APOBEC3A positively correlates with IFN-treatment responses in CHB patients, while NEIL3 shows negative correlation. These genes may involve to IFN mediated viral suppression and serve as biomarkers for CHB disease management.
Collapse
Affiliation(s)
- Yong Li
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuchen Xia
- Institute of Virology, Technical University of Munich / Helmholtz Zentrum München, 81675, Munich, Germany.,Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, 20892, Bethesda, Maryland, USA
| | - Meifang Han
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guang Chen
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dake Zhang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wolfgang E Thasler
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Grosshadern Hospital, Ludwig Maximilians University, 81377, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich / Helmholtz Zentrum München, 81675, Munich, Germany.,German Center for Infection research (DZIF), Munich, Germany
| | - Qin Ning
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
16
|
Hardonnière K, Fernier M, Gallais I, Mograbi B, Podechard N, Le Ferrec E, Grova N, Appenzeller B, Burel A, Chevanne M, Sergent O, Huc L, Bortoli S, Lagadic-Gossmann D. Role for the ATPase inhibitory factor 1 in the environmental carcinogen-induced Warburg phenotype. Sci Rep 2017; 7:195. [PMID: 28298645 PMCID: PMC5428028 DOI: 10.1038/s41598-017-00269-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/14/2017] [Indexed: 11/28/2022] Open
Abstract
Most tumors undergo metabolic reprogramming towards glycolysis, the so-called Warburg effect, to support growth and survival. Overexpression of IF1, the physiological inhibitor of the F0F1ATPase, has been related to this phenomenon and appears to be a relevant marker in cancer. Environmental contributions to cancer development are now widely accepted but little is known about the underlying intracellular mechanisms. Among the environmental pollutants humans are commonly exposed to, benzo[a]pyrene (B[a]P), the prototype molecule of polycyclic aromatic hydrocarbons (PAHs), is a well-known human carcinogen. Besides apoptotic signals, B[a]P can also induce survival signals in liver cells, both likely involved in cancer promotion. Our previous works showed that B[a]P elicited a Warburg-like effect, thus favoring cell survival. The present study aimed at further elucidating the molecular mechanisms involved in the B[a]P-induced metabolic reprogramming, by testing the possible involvement of IF1. We presently demonstrate, both in vitro and in vivo, that PAHs, especially B[a]P, strongly increase IF1 expression. Such an increase, which might rely on β2-adrenergic receptor activation, notably participates to the B[a]P-induced glycolytic shift and cell survival in liver cells. By identifying IF1 as a target of PAHs, this study provides new insights about how environmental factors may contribute to related carcinogenesis.
Collapse
Affiliation(s)
- Kévin Hardonnière
- Inserm U1085, Institut de Recherche en Santé, Environnement, Travail, Rennes, France
- Université de Rennes 1, Biosit UMS3080, 35043, Rennes Cédex, France
| | - Morgane Fernier
- Inserm U1085, Institut de Recherche en Santé, Environnement, Travail, Rennes, France
- Université de Rennes 1, Biosit UMS3080, 35043, Rennes Cédex, France
| | - Isabelle Gallais
- Inserm U1085, Institut de Recherche en Santé, Environnement, Travail, Rennes, France
- Université de Rennes 1, Biosit UMS3080, 35043, Rennes Cédex, France
| | - Baharia Mograbi
- Institute of Research on Cancer and Ageing of Nice (IRCAN), INSERM U1081, CNRS UMR7284, Université de Nice-Sophia Antipolis, Faculté de Médecine, Centre Antoine Lacassagne, Nice, F-06107, France
| | - Normand Podechard
- Inserm U1085, Institut de Recherche en Santé, Environnement, Travail, Rennes, France
- Université de Rennes 1, Biosit UMS3080, 35043, Rennes Cédex, France
| | - Eric Le Ferrec
- Inserm U1085, Institut de Recherche en Santé, Environnement, Travail, Rennes, France
- Université de Rennes 1, Biosit UMS3080, 35043, Rennes Cédex, France
| | - Nathalie Grova
- HBRU, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg
| | - Brice Appenzeller
- HBRU, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg
| | - Agnès Burel
- Université de Rennes 1, Biosit UMS3080, 35043, Rennes Cédex, France
| | - Martine Chevanne
- Inserm U1085, Institut de Recherche en Santé, Environnement, Travail, Rennes, France
- Université de Rennes 1, Biosit UMS3080, 35043, Rennes Cédex, France
| | - Odile Sergent
- Inserm U1085, Institut de Recherche en Santé, Environnement, Travail, Rennes, France
- Université de Rennes 1, Biosit UMS3080, 35043, Rennes Cédex, France
| | | | - Sylvie Bortoli
- INSERM UMR-S 1124, Université Paris Descartes, Centre Universitaire des Saint-Pères, Paris, France
| | - Dominique Lagadic-Gossmann
- Inserm U1085, Institut de Recherche en Santé, Environnement, Travail, Rennes, France.
- Université de Rennes 1, Biosit UMS3080, 35043, Rennes Cédex, France.
| |
Collapse
|
17
|
Expanding the Immunology Toolbox: Embracing Public-Data Reuse and Crowdsourcing. Immunity 2016; 45:1191-1204. [DOI: 10.1016/j.immuni.2016.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 12/15/2022]
|
18
|
Nandal UK, van Kampen AHC, Moerland PD. compendiumdb: an R package for retrieval and storage of functional genomics data. Bioinformatics 2016; 32:2856-7. [DOI: 10.1093/bioinformatics/btw335] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 05/23/2016] [Indexed: 01/28/2023] Open
|
19
|
Corpas M, Valdivia-Granda W, Torres N, Greshake B, Coletta A, Knaus A, Harrison AP, Cariaso M, Moran F, Nielsen F, Swan D, Weiss Solís DY, Krawitz P, Schacherer F, Schols P, Yang H, Borry P, Glusman G, Robinson PN. Crowdsourced direct-to-consumer genomic analysis of a family quartet. BMC Genomics 2015; 16:910. [PMID: 26547235 PMCID: PMC4636840 DOI: 10.1186/s12864-015-1973-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 10/03/2015] [Indexed: 12/22/2022] Open
Abstract
Background We describe the pioneering experience of a Spanish family pursuing the goal of understanding their own personal genetic data to the fullest possible extent using Direct to Consumer (DTC) tests. With full informed consent from the Corpas family, all genotype, exome and metagenome data from members of this family, are publicly available under a public domain Creative Commons 0 (CC0) license waiver. All scientists or companies analysing these data (“the Corpasome”) were invited to return results to the family. Methods We released 5 genotypes, 4 exomes, 1 metagenome from the Corpas family via a blog and figshare under a public domain license, inviting scientists to join the crowdsourcing efforts to analyse the genomes in return for coauthorship or acknowldgement in derived papers. Resulting analysis data were compiled via social media and direct email. Results Here we present the results of our investigations, combining the crowdsourced contributions and our own efforts. Four companies offering annotations for genomic variants were applied to four family exomes: BIOBASE, Ingenuity, Diploid, and GeneTalk. Starting from a common VCF file and after selecting for significant results from company reports, we find no overlap among described annotations. We additionally report on a gut microbiome analysis of a member of the Corpas family. Conclusions This study presents an analysis of a diverse set of tools and methods offered by four DTC companies. The striking discordance of the results mirrors previous findings with respect to DTC analysis of SNP chip data, and highlights the difficulties of using DTC data for preventive medical care. To our knowledge, the data and analysis results from our crowdsourced study represent the most comprehensive exome and analysis for a family quartet using solely DTC data generation to date. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1973-7) contains supplementary material, which is available to authorised users.
Collapse
Affiliation(s)
- Manuel Corpas
- The Genome Analysis Centre, Norwich Research Park, Norwich, UK.
| | | | - Nazareth Torres
- Universidad de Navarra, Grupo de Fisiología del Estrés en Plantas (Dpto. de Biología Ambiental), Pamplona, Spain.
| | - Bastian Greshake
- Department for Applied Bioinformatics, Institute for Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany.
| | - Alain Coletta
- InSilico Genomics S.A., Avenue Adolphe Buyl, 87, Building C, 5th floor, B-1050, Brussels, Belgium. .,Institut de Recherches Interdisciplinaires et de Developpements en Intelligence Artificielle, the Computer and Decision Engineering Department, Universite Libre de Bruxelles, 87 av. Adolphe Buyl, Bruxelles, 1050, Belguim.
| | - Alexej Knaus
- Institute for Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany.
| | - Andrew P Harrison
- Department of Mathematical Sciences, University of Essex, Wivenhoe Park, Colchester, Essex, CO4 3SQ, UK.
| | - Mike Cariaso
- SNPedia, River Road Bio, LLC, 9812 Falls Road #114-237, Potomac, Maryland, MD, 20854, USA.
| | - Federico Moran
- Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| | - Fiona Nielsen
- DNADigest, Windsor House, Station Court, Station Road, Great Shelford, Cambridge, CB22 5NE, UK.
| | - Daniel Swan
- The Genome Analysis Centre, Norwich Research Park, Norwich, UK. .,Oxford Gene Technology, Woodstock Road, Begbroke, Oxfordshire, OX5 1PF, UK.
| | - David Y Weiss Solís
- InSilico Genomics S.A., Avenue Adolphe Buyl, 87, Building C, 5th floor, B-1050, Brussels, Belgium. .,Institut de Recherches Interdisciplinaires et de Developpements en Intelligence Artificielle, the Computer and Decision Engineering Department, Universite Libre de Bruxelles, 87 av. Adolphe Buyl, Bruxelles, 1050, Belguim.
| | - Peter Krawitz
- Institute for Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany. .,GeneTalk, Finckensteinallee 84, 12205, Berlin, Germany.
| | - Frank Schacherer
- BIOBASE, Halchtersche Strasse 33, D-38304, Wolfenbuettel, Germany.
| | - Peter Schols
- Diploid Genomics, Middelweg 129, 3001, Leuven, Belgium.
| | - Huangming Yang
- BGI, Main Building, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China.
| | - Pascal Borry
- Department of Public Health and Primary Care, Catholic University of Leuven, Leuven, Belgium.
| | - Gustavo Glusman
- The Institute for Systems Biology, 401 Terry Ave N, Seattle, WA, 98109, USA.
| | - Peter N Robinson
- Institute for Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany. .,Berlin Brandenburg Center for Regenerative Therapies, Charité Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
20
|
Bagewadi S, Adhikari S, Dhrangadhariya A, Irin AK, Ebeling C, Namasivayam AA, Page M, Hofmann-Apitius M, Senger P. NeuroTransDB: highly curated and structured transcriptomic metadata for neurodegenerative diseases. Database (Oxford) 2015; 2015:bav099. [PMID: 26475471 PMCID: PMC4608514 DOI: 10.1093/database/bav099] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 09/07/2015] [Accepted: 09/10/2015] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are chronic debilitating conditions, characterized by progressive loss of neurons that represent a significant health care burden as the global elderly population continues to grow. Over the past decade, high-throughput technologies such as the Affymetrix GeneChip microarrays have provided new perspectives into the pathomechanisms underlying neurodegeneration. Public transcriptomic data repositories, namely Gene Expression Omnibus and curated ArrayExpress, enable researchers to conduct integrative meta-analysis; increasing the power to detect differentially regulated genes in disease and explore patterns of gene dysregulation across biologically related studies. The reliability of retrospective, large-scale integrative analyses depends on an appropriate combination of related datasets, in turn requiring detailed meta-annotations capturing the experimental setup. In most cases, we observe huge variation in compliance to defined standards for submitted metadata in public databases. Much of the information to complete, or refine meta-annotations are distributed in the associated publications. For example, tissue preparation or comorbidity information is frequently described in an article's supplementary tables. Several value-added databases have employed additional manual efforts to overcome this limitation. However, none of these databases explicate annotations that distinguish human and animal models in neurodegeneration context. Therefore, adopting a more specific disease focus, in combination with dedicated disease ontologies, will better empower the selection of comparable studies with refined annotations to address the research question at hand. In this article, we describe the detailed development of NeuroTransDB, a manually curated database containing metadata annotations for neurodegenerative studies. The database contains more than 20 dimensions of metadata annotations within 31 mouse, 5 rat and 45 human studies, defined in collaboration with domain disease experts. We elucidate the step-by-step guidelines used to critically prioritize studies from public archives and their metadata curation and discuss the key challenges encountered. Curated metadata for Alzheimer's disease gene expression studies are available for download. Database URL: www.scai.fraunhofer.de/NeuroTransDB.html.
Collapse
Affiliation(s)
- Shweta Bagewadi
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754 Sankt Augustin, Germany, Rheinische Friedrich-Wilhelms-Universitaet Bonn, Bonn-Aachen International Center for Information Technology, 53113, Bonn, Germany,
| | - Subash Adhikari
- Department of Chemistry, South University of Science and Technology of China, No 1088, Xueyuan Road, Xili, Shenzhen, China
| | - Anjani Dhrangadhariya
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754 Sankt Augustin, Germany, Rheinische Friedrich-Wilhelms-Universitaet Bonn, Bonn-Aachen International Center for Information Technology, 53113, Bonn, Germany
| | - Afroza Khanam Irin
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754 Sankt Augustin, Germany, Rheinische Friedrich-Wilhelms-Universitaet Bonn, Bonn-Aachen International Center for Information Technology, 53113, Bonn, Germany
| | - Christian Ebeling
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754 Sankt Augustin, Germany
| | - Aishwarya Alex Namasivayam
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7, avenue des Hauts-Fourneaux, L-4362 Esch-sur-Alzette, Luxembourg and
| | - Matthew Page
- Translational Bioinformatics, UCB Pharma, 216 Bath Rd, Slough SL1 3WE, United Kingdom
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754 Sankt Augustin, Germany, Rheinische Friedrich-Wilhelms-Universitaet Bonn, Bonn-Aachen International Center for Information Technology, 53113, Bonn, Germany
| | - Philipp Senger
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754 Sankt Augustin, Germany,
| |
Collapse
|
21
|
Guo Z, Tzvetkova B, Bassik JM, Bodziak T, Wojnar BM, Qiao W, Obaida MA, Nelson SB, Hu BH, Yu P. RNASeqMetaDB: a database and web server for navigating metadata of publicly available mouse RNA-Seq datasets. Bioinformatics 2015; 31:4038-40. [PMID: 26323714 DOI: 10.1093/bioinformatics/btv503] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 08/23/2015] [Indexed: 01/24/2023] Open
Abstract
UNLABELLED Gene targeting is a protocol for introducing a mutation to a specific gene in an organism. Because of the importance of in vivo assessment of gene function and modeling of human diseases, this technique has been widely adopted to generate a large number of mutant mouse models. Due to the recent breakthroughs in high-throughput sequencing technologies, RNA-Seq experiments have been performed on many of these mouse models, leading to hundreds of publicly available datasets. To facilitate the reuse of these datasets, we collected the associated metadata and organized them in a database called RNASeqMetaDB. The metadata were manually curated to ensure annotation consistency. We developed a web server to allow easy database navigation and data querying. Users can search the database using multiple parameters like genes, diseases, tissue types, keywords and associated publications in order to find datasets that match their interests. Summary statistics of the metadata are also presented on the web server showing interesting global patterns of RNA-Seq studies. AVAILABILITY AND IMPLEMENTATION Freely available on the web at http://rnaseqmetadb.ece.tamu.edu.
Collapse
Affiliation(s)
- Zhengyu Guo
- Department of Electrical and Computer Engineering & TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Boriana Tzvetkova
- Department of Biology & Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA and
| | - Jennifer M Bassik
- Department of Communicative Disorders and Sciences & Center for Hearing and Deafness, University at Buffalo, Buffalo, NY 14214, USA
| | - Tara Bodziak
- Department of Communicative Disorders and Sciences & Center for Hearing and Deafness, University at Buffalo, Buffalo, NY 14214, USA
| | - Brianna M Wojnar
- Department of Communicative Disorders and Sciences & Center for Hearing and Deafness, University at Buffalo, Buffalo, NY 14214, USA
| | - Wei Qiao
- Department of Electrical and Computer Engineering &
| | - Md A Obaida
- Department of Electrical and Computer Engineering &
| | - Sacha B Nelson
- Department of Biology & Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, USA and
| | - Bo Hua Hu
- Department of Communicative Disorders and Sciences & Center for Hearing and Deafness, University at Buffalo, Buffalo, NY 14214, USA
| | - Peng Yu
- Department of Electrical and Computer Engineering & TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
22
|
Frazee AC, Pertea G, Jaffe AE, Langmead B, Salzberg SL, Leek JT. Ballgown bridges the gap between transcriptome assembly and expression analysis. Nat Biotechnol 2015; 33:243-6. [PMID: 25748911 PMCID: PMC4792117 DOI: 10.1038/nbt.3172] [Citation(s) in RCA: 517] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Alyssa C Frazee
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Geo Pertea
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew E Jaffe
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland, USA
| | - Ben Langmead
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland, USA
| | - Steven L Salzberg
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jeffrey T Leek
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Bostan H, Chiusano ML. NexGenEx-Tom: a gene expression platform to investigate the functionalities of the tomato genome. BMC PLANT BIOLOGY 2015; 15:48. [PMID: 25849067 PMCID: PMC4340097 DOI: 10.1186/s12870-014-0412-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 12/30/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND Next Generation Sequencing technologies (NGS) unexpectedly pushed forward the capability of solving genome organization and of widely depicting gene expression. However, although the flourishing of tools to process the NGS data, versatile and user-friendly computational environments for integrative and comparative analyses of the results from the increasing amount of collections are still required. DESCRIPTION Here we present the architecture and the facilities of NexGenEx-, a web based platform that offers processed NGS transcriptome collections and enables immediate analyses of the results. The platform allows gene expression investigations, profiling and comparisons, and exploits different resources. CONCLUSION In the current version, NexGenEx-Tom includes processed and normalized NGS expression data from three collections covering several tissue/stages from different genotypes. Beyond providing a user-friendly interface, the platform was designed with the aim to easily be expanded to include other NGS based transcriptome collections. It can also integrate different genome releases, possibly from different cultivars or genotypes, but even from different species. The platform is proposed as an example effort in tomato, and is described as a profitable approach for the exploitation of these challenging and precious datasets.
Collapse
Affiliation(s)
- Hamed Bostan
- Department of Agricultural Sciences, University of Naples “Federico II”, via Università 100, 80055 Portici, Italy
| | - Maria Luisa Chiusano
- Department of Agricultural Sciences, University of Naples “Federico II”, via Università 100, 80055 Portici, Italy
| |
Collapse
|
24
|
Weiler S, Ademokun JA, Norton JD. ID helix-loop-helix proteins as determinants of cell survival in B-cell chronic lymphocytic leukemia cells in vitro. Mol Cancer 2015; 14:30. [PMID: 25644253 PMCID: PMC4320821 DOI: 10.1186/s12943-014-0286-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 12/30/2014] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Members of the inhibitor of DNA-binding (ID) family of helix-loop-helix proteins have been causally implicated in the pathogenesis of several types of B-cell lineage malignancy, either on the basis of mutation or by altered expression. B-cell chronic lymphocytic leukemia encompasses a heterogeneous group of disorders and is the commonest leukaemia type in the Western world. In this study, we have investigated the pathobiological functions of the ID2 and ID3 proteins in this disease with an emphasis on their role in regulating leukemic cell death/survival. METHODS Bioinformatics analysis of microarray gene expression data was used to investigate expression of ID2/ID3 in leukemic versus normal B cells, their association with clinical course of disease and molecular sub-type and to reconstruct a gene regulatory network using the 'maximum information coefficient' (MIC) for target gene inference. In vitro cultured primary leukemia cells, either in isolation or co-cultured with accessory vascular endothelial cells, were used to investigate ID2/ID3 protein expression by western blotting and to assess the cytotoxic response of different drugs (fludarabine, chlorambucil, ethacrynic acid) by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. ID2/ID3 protein levels in primary leukemia cells and in MEC1 cells were manipulated by transduction with siRNA reagents. RESULTS Datamining showed that the expression profiles of ID2 and ID3 are associated with distinct pathobiological features of disease and implicated both genes in regulating cell death/survival by targeting multiple non-overlapping sets of apoptosis effecter genes. Consistent with microarray data, the overall pattern of ID2/ID3 protein expression in relation to cell death/survival responses of primary leukemia cells was suggestive of a pro-survival function for both ID proteins. This was confirmed by siRNA knock-down experiments in MEC1 cells and in primary leukemia cells, but with variability in the dependence of leukemic cells from different patients on ID protein expression for cell survival. Vascular endothelial cells rescued leukemia cells from spontaneous and cytotoxic drug-induced cell death at least in part, via an ID protein-coupled redox-dependent mechanism. CONCLUSIONS Our study provides evidence for a pro-survival function of the ID2/ID3 proteins in chronic lymphocytic leukemia cells and also highlights these proteins as potential determinants of the pathobiology of this disorder.
Collapse
Affiliation(s)
- Sarah Weiler
- School of Biological Sciences, University of Essex, Colchester, Essex, CO4 3SQ, UK.
| | - Jolaolu A Ademokun
- Department of Haematology, Ipswich Hospital NHS Trust, Heath Road, Ipswich, Suffolk, IP4 5PD, UK.
| | - John D Norton
- School of Biological Sciences, University of Essex, Colchester, Essex, CO4 3SQ, UK.
| |
Collapse
|
25
|
Beck TN, Chikwem AJ, Solanki NR, Golemis EA. Bioinformatic approaches to augment study of epithelial-to-mesenchymal transition in lung cancer. Physiol Genomics 2014; 46:699-724. [PMID: 25096367 PMCID: PMC4187119 DOI: 10.1152/physiolgenomics.00062.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/04/2014] [Indexed: 12/22/2022] Open
Abstract
Bioinformatic approaches are intended to provide systems level insight into the complex biological processes that underlie serious diseases such as cancer. In this review we describe current bioinformatic resources, and illustrate how they have been used to study a clinically important example: epithelial-to-mesenchymal transition (EMT) in lung cancer. Lung cancer is the leading cause of cancer-related deaths and is often diagnosed at advanced stages, leading to limited therapeutic success. While EMT is essential during development and wound healing, pathological reactivation of this program by cancer cells contributes to metastasis and drug resistance, both major causes of death from lung cancer. Challenges of studying EMT include its transient nature, its molecular and phenotypic heterogeneity, and the complicated networks of rewired signaling cascades. Given the biology of lung cancer and the role of EMT, it is critical to better align the two in order to advance the impact of precision oncology. This task relies heavily on the application of bioinformatic resources. Besides summarizing recent work in this area, we use four EMT-associated genes, TGF-β (TGFB1), NEDD9/HEF1, β-catenin (CTNNB1) and E-cadherin (CDH1), as exemplars to demonstrate the current capacities and limitations of probing bioinformatic resources to inform hypothesis-driven studies with therapeutic goals.
Collapse
Affiliation(s)
- Tim N Beck
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Program in Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, Pennsylvania; and
| | - Adaeze J Chikwem
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Temple University School of Medicine, Philadelphia, Pennsylvania; and
| | - Nehal R Solanki
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Program in Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Erica A Golemis
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania; Temple University School of Medicine, Philadelphia, Pennsylvania; and Program in Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, Pennsylvania; and
| |
Collapse
|
26
|
Badgeley MA, Sealfon SC, Chikina MD. Hybrid Bayesian-rank integration approach improves the predictive power of genomic dataset aggregation. ACTA ACUST UNITED AC 2014; 31:209-15. [PMID: 25266226 DOI: 10.1093/bioinformatics/btu518] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
MOTIVATION Modern molecular technologies allow the collection of large amounts of high-throughput data on the functional attributes of genes. Often multiple technologies and study designs are used to address the same biological question such as which genes are overexpressed in a specific disease state. Consequently, there is considerable interest in methods that can integrate across datasets to present a unified set of predictions. RESULTS An important aspect of data integration is being able to account for the fact that datasets may differ in how accurately they capture the biological signal of interest. While many methods to address this problem exist, they always rely either on dataset internal statistics, which reflect data structure and not necessarily biological relevance, or external gold standards, which may not always be available. We present a new rank aggregation method for data integration that requires neither external standards nor internal statistics but relies on Bayesian reasoning to assess dataset relevance. We demonstrate that our method outperforms established techniques and significantly improves the predictive power of rank-based aggregations. We show that our method, which does not require an external gold standard, provides reliable estimates of dataset relevance and allows the same set of data to be integrated differently depending on the specific signal of interest. AVAILABILITY The method is implemented in R and is freely available at http://www.pitt.edu/~mchikina/BIRRA/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Marcus A Badgeley
- Department of Neurology, Mount Sinai School of Medicine, New York, NY 10029 and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Stuart C Sealfon
- Department of Neurology, Mount Sinai School of Medicine, New York, NY 10029 and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Maria D Chikina
- Department of Neurology, Mount Sinai School of Medicine, New York, NY 10029 and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
27
|
iRegulon: from a gene list to a gene regulatory network using large motif and track collections. PLoS Comput Biol 2014; 10:e1003731. [PMID: 25058159 PMCID: PMC4109854 DOI: 10.1371/journal.pcbi.1003731] [Citation(s) in RCA: 618] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/27/2014] [Indexed: 01/17/2023] Open
Abstract
Identifying master regulators of biological processes and mapping their downstream gene networks are key challenges in systems biology. We developed a computational method, called iRegulon, to reverse-engineer the transcriptional regulatory network underlying a co-expressed gene set using cis-regulatory sequence analysis. iRegulon implements a genome-wide ranking-and-recovery approach to detect enriched transcription factor motifs and their optimal sets of direct targets. We increase the accuracy of network inference by using very large motif collections of up to ten thousand position weight matrices collected from various species, and linking these to candidate human TFs via a motif2TF procedure. We validate iRegulon on gene sets derived from ENCODE ChIP-seq data with increasing levels of noise, and we compare iRegulon with existing motif discovery methods. Next, we use iRegulon on more challenging types of gene lists, including microRNA target sets, protein-protein interaction networks, and genetic perturbation data. In particular, we over-activate p53 in breast cancer cells, followed by RNA-seq and ChIP-seq, and could identify an extensive up-regulated network controlled directly by p53. Similarly we map a repressive network with no indication of direct p53 regulation but rather an indirect effect via E2F and NFY. Finally, we generalize our computational framework to include regulatory tracks such as ChIP-seq data and show how motif and track discovery can be combined to map functional regulatory interactions among co-expressed genes. iRegulon is available as a Cytoscape plugin from http://iregulon.aertslab.org. Gene regulatory networks control developmental, homeostatic, and disease processes by governing precise levels and spatio-temporal patterns of gene expression. Determining their topology can provide mechanistic insight into these processes. Gene regulatory networks consist of interactions between transcription factors and their direct target genes. Each regulatory interaction represents the binding of the transcription factor to a specific DNA binding site near its target gene. Here we present a computational method, called iRegulon, to identify master regulators and direct target genes in a human gene signature, i.e. a set of co-expressed genes. iRegulon relies on the analysis of the regulatory sequences around each gene in the gene set to detect enriched TF motifs or ChIP-seq peaks, using databases of nearly 10.000 TF motifs and 1000 ChIP-seq data sets or “tracks”. Next, it associates enriched motifs and tracks with candidate transcription factors and determines the optimal subset of direct target genes. We validate iRegulon on ENCODE data, and use it in combination with RNA-seq and ChIP-seq data to map a p53 downstream network with new predicted co-factors and targets. iRegulon is available as a Cytoscape plugin, supporting human, mouse, and Drosophila genes, and provides access to hundreds of cancer-related TF-target subnetworks or “regulons”.
Collapse
|
28
|
Glycerol-3-phosphate acyltranferase-2 behaves as a cancer testis gene and promotes growth and tumorigenicity of the breast cancer MDA-MB-231 cell line. PLoS One 2014; 9:e100896. [PMID: 24967918 PMCID: PMC4072688 DOI: 10.1371/journal.pone.0100896] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 06/01/2014] [Indexed: 02/06/2023] Open
Abstract
The de novo synthesis of glycerolipids in mammalian cells begins with the acylation of glycerol-3-phosphate, catalyzed by glycerol-3-phosphate acyltransferase (GPAT). GPAT2 is a mitochondrial isoform primarily expressed in testis under physiological conditions. Because it is aberrantly expressed in multiple myeloma, it has been proposed as a novel cancer testis gene. Using a bioinformatics approach, we found that GPAT2 is highly expressed in melanoma, lung, prostate and breast cancer, and we validated GPAT2 expression at the protein level in breast cancer by immunohistochemistry. In this case GPAT2 expression correlated with a higher histological grade. 5-Aza-2′ deoxycytidine treatment of human cells lines induced GPAT2 expression suggesting epigenetic regulation of gene expression. In order to evaluate the contribution of GPAT2 to the tumor phenotype, we silenced its expression in MDA-MB-231 cells. GPAT2 knockdown diminished cell proliferation, anchorage independent growth, migration and tumorigenicity, and increased staurosporine-induced apoptosis. In contrast, GPAT2 over-expression increased cell proliferation rate and resistance to staurosporine-induced apoptosis. To understand the functional role of GPAT2, we performed a co-expression analysis in mouse and human testis and found a significant association with semantic terms involved in cell cycle, DNA integrity maintenance, piRNA biogenesis and epigenetic regulation. Overall, these results indicate the GPAT2 would be directly associated with the control of cell proliferation. In conclusion, we confirm GPAT2 as a cancer testis gene and that its expression contributes to the tumor phenotype of MDA-MB-231 cells.
Collapse
|
29
|
SOX2 controls tumour initiation and cancer stem-cell functions in squamous-cell carcinoma. Nature 2014; 511:246-50. [PMID: 24909994 DOI: 10.1038/nature13305] [Citation(s) in RCA: 499] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 03/31/2014] [Indexed: 12/15/2022]
Abstract
Cancer stem cells (CSCs) have been reported in various cancers, including in skin squamous-cell carcinoma (SCC). The molecular mechanisms regulating tumour initiation and stemness are still poorly characterized. Here we find that Sox2, a transcription factor expressed in various types of embryonic and adult stem cells, was the most upregulated transcription factor in the CSCs of squamous skin tumours in mice. SOX2 is absent in normal epidermis but begins to be expressed in the vast majority of mouse and human pre-neoplastic skin tumours, and continues to be expressed in a heterogeneous manner in invasive mouse and human SCCs. In contrast to other SCCs, in which SOX2 is frequently genetically amplified, the expression of SOX2 in mouse and human skin SCCs is transcriptionally regulated. Conditional deletion of Sox2 in the mouse epidermis markedly decreases skin tumour formation after chemical-induced carcinogenesis. Using green fluorescent protein (GFP) as a reporter of Sox2 transcriptional expression (SOX2-GFP knock-in mice), we showed that SOX2-expressing cells in invasive SCC are greatly enriched in tumour-propagating cells, which further increase upon serial transplantations. Lineage ablation of SOX2-expressing cells within primary benign and malignant SCCs leads to tumour regression, consistent with the critical role of SOX2-expressing cells in tumour maintenance. Conditional Sox2 deletion in pre-existing skin papilloma and SCC leads to tumour regression and decreases the ability of cancer cells to be propagated upon transplantation into immunodeficient mice, supporting the essential role of SOX2 in regulating CSC functions. Transcriptional profiling of SOX2-GFP-expressing CSCs and of tumour epithelial cells upon Sox2 deletion uncovered a gene network regulated by SOX2 in primary tumour cells in vivo. Chromatin immunoprecipitation identified several direct SOX2 target genes controlling tumour stemness, survival, proliferation, adhesion, invasion and paraneoplastic syndrome. We demonstrate that SOX2, by marking and regulating the functions of skin tumour-initiating cells and CSCs, establishes a continuum between tumour initiation and progression in primary skin tumours.
Collapse
|
30
|
McNamara KM, Yoda T, Nurani AM, Shibahara Y, Miki Y, Wang L, Nakamura Y, Suzuki K, Yang Y, Abe E, Hirakawa H, Suzuki T, Nemoto N, Miyashita M, Tamaki K, Ishida T, Brown KA, Ohuchi N, Sasano H. Androgenic pathways in the progression of triple-negative breast carcinoma: a comparison between aggressive and non-aggressive subtypes. Breast Cancer Res Treat 2014; 145:281-93. [PMID: 24715382 DOI: 10.1007/s10549-014-2942-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/27/2014] [Indexed: 12/21/2022]
Abstract
One of the active intracellular pathways/networks in triple-negative breast carcinoma (TNBC) is that of the androgen receptor (AR). In this study, we examined AR and androgen-metabolising enzyme immunoreactivity in subcategories of TNBC to further elucidate the roles of androgenic pathways in TNBC. We utilised formalin-fixed paraffin-embedded breast cancer samples from ductal carcinoma in situ (DCIS) and invasive ductal carcinoma patient cohorts. We then used immunohistochemistry to classify these samples into basal-like and non-basal samples and to assess interactions between AR, androgen-metabolising enzymes and proliferation. To further substantiate our hypothesis and provide mechanistic insights, we also looked at the expression and regulation of these factors in publically available microarray data and in a panel of TNBC AR-positive cell lines. DCIS was associated with higher levels of AR and enzymes (p < 0.02), although a similar difference was not noticed in basal and non-basal samples. AR and enzymes were correlated in all states. In TNBC cell lines (MDA-MD-453, MFM-223 and SUM185-PE), we found that DHT treatment up-regulated 5αR1 and 17βHSD5 suggesting a mechanistic explanation for the correlations observed in the histological samples. Publicly available microarray data in TNBC cases suggested similar patterns to those observed in histological samples. In the majority of settings, including publically available microarray data, an inverse association between AR and proliferation was detected. These findings suggest that decreases in AR and androgen-metabolising enzymes may be involved in the increased biological aggressiveness in TNBC development.
Collapse
MESH Headings
- 3-Hydroxysteroid Dehydrogenases/genetics
- 3-Hydroxysteroid Dehydrogenases/metabolism
- Aldo-Keto Reductase Family 1 Member C3
- Androgens/metabolism
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/drug therapy
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cell Line, Tumor
- Cell Proliferation
- Cholestenone 5 alpha-Reductase/genetics
- Cholestenone 5 alpha-Reductase/metabolism
- Dihydrotestosterone/pharmacology
- ErbB Receptors/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hydroxyprostaglandin Dehydrogenases/genetics
- Hydroxyprostaglandin Dehydrogenases/metabolism
- Keratin-5/metabolism
- Keratin-6/metabolism
- Receptors, Androgen/metabolism
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
Collapse
Affiliation(s)
- Keely M McNamara
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Comparison of merging and meta-analysis as alternative approaches for integrative gene expression analysis. ISRN BIOINFORMATICS 2014; 2014:345106. [PMID: 25937953 PMCID: PMC4393058 DOI: 10.1155/2014/345106] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 09/24/2013] [Indexed: 01/19/2023]
Abstract
An increasing amount of microarray gene expression data sets is available through public repositories. Their huge potential in making new findings is yet to be unlocked by making them available for large-scale analysis. In order to do so it is essential that independent studies designed for similar biological problems can be integrated, so that new insights can be obtained. These insights would remain undiscovered when analyzing the individual data sets because it is well known that the small number of biological samples used per experiment is a bottleneck in genomic analysis. By increasing the number of samples the statistical power is increased and more general and reliable conclusions can be drawn. In this work, two different approaches for conducting large-scale analysis of microarray gene expression data-meta-analysis and data merging-are compared in the context of the identification of cancer-related biomarkers, by analyzing six independent lung cancer studies. Within this study, we investigate the hypothesis that analyzing large cohorts of samples resulting in merging independent data sets designed to study the same biological problem results in lower false discovery rates than analyzing the same data sets within a more conservative meta-analysis approach.
Collapse
|
32
|
Ferguson BW, Gao X, Zelazowski MJ, Lee J, Jeter CR, Abba MC, Aldaz CM. The cancer gene WWOX behaves as an inhibitor of SMAD3 transcriptional activity via direct binding. BMC Cancer 2013; 13:593. [PMID: 24330518 PMCID: PMC3871008 DOI: 10.1186/1471-2407-13-593] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/06/2013] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The WW domain containing protein WWOX has been postulated to behave as a tumor suppressor in breast and other cancers. Expression of this protein is lost in over 70% of ER negative tumors. This prompted us to investigate the phenotypic and gene expression effects of loss of WWOX expression in breast cells. METHODS Gene expression microarrays and standard in vitro assays were performed on stably silenced WWOX (shRNA) normal breast cells. Bioinformatic analyses were used to identify gene networks and transcriptional regulators affected by WWOX silencing. Co-immunoprecipitations and GST-pulldowns were used to demonstrate a direct interaction between WWOX and SMAD3. Reporter assays, ChIP, confocal microscopy and in silico analyses were employed to determine the effect of WWOX silencing on TGFβ-signaling. RESULTS WWOX silencing affected cell proliferation, motility, attachment and deregulated expression of genes involved in cell cycle, motility and DNA damage. Interestingly, we detected an enrichment of targets activated by the SMAD3 transcription factor, including significant upregulation of ANGPTL4, FST, PTHLH and SERPINE1 transcripts. Importantly, we demonstrate that the WWOX protein physically interacts with SMAD3 via WW domain 1. Furthermore, WWOX expression dramatically decreases SMAD3 occupancy at the ANGPTL4 and SERPINE1 promoters and significantly quenches activation of a TGFβ responsive reporter. Additionally, WWOX expression leads to redistribution of SMAD3 from the nuclear to the cytoplasmic compartment. Since the TGFβ target ANGPTL4 plays a key role in lung metastasis development, we performed a meta-analysis of ANGPTL4 expression relative to WWOX in microarray datasets from breast carcinomas. We observed a significant inverse correlation between WWOX and ANGPTL4. Furthermore, the WWOX(lo)/ANGPTL4(hi) cluster of breast tumors is enriched in triple-negative and basal-like sub-types. Tumors with this gene expression signature could represent candidates for anti-TGFβ targeted therapies. CONCLUSIONS We show for the first time that WWOX modulates SMAD3 signaling in breast cells via direct WW-domain mediated binding and potential cytoplasmic sequestration of SMAD3 protein. Since loss of WWOX expression increases with breast cancer progression and it behaves as an inhibitor of SMAD3 transcriptional activity these observations may help explain, at least in part, the paradoxical pro-tumorigenic effects of TGFβ signaling in advanced breast cancer.
Collapse
Affiliation(s)
- Brent W Ferguson
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Xinsheng Gao
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Maciej J Zelazowski
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Jaeho Lee
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Collene R Jeter
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Martin C Abba
- CINIBA, Facultad de Medicina, Universidad Nacional de La Plata, La Plata, Argentina
| | - C Marcelo Aldaz
- Department of Molecular Carcinogenesis, Science Park, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| |
Collapse
|
33
|
McCall MN, Jaffee HA, Zelisko SJ, Sinha N, Hooiveld G, Irizarry RA, Zilliox MJ. The Gene Expression Barcode 3.0: improved data processing and mining tools. Nucleic Acids Res 2013; 42:D938-43. [PMID: 24271388 PMCID: PMC3965035 DOI: 10.1093/nar/gkt1204] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The Gene Expression Barcode project, http://barcode.luhs.org, seeks to determine the genes expressed for every tissue and cell type in humans and mice. Understanding the absolute expression of genes across tissues and cell types has applications in basic cell biology, hypothesis generation for gene function and clinical predictions using gene expression signatures. In its current version, this project uses the abundant publicly available microarray data sets combined with a suite of single-array preprocessing, quality control and analysis methods. In this article, we present the improvements that have been made since the previous version of the Gene Expression Barcode in 2011. These include a variety of new data mining tools and summaries, estimated transcriptomes and curated annotations.
Collapse
Affiliation(s)
- Matthew N. McCall
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA, Department of Biostatistics, Johns Hopkins University, Baltimore, MD 21205, USA, Informatics and Systems Development, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA, Nutrition, Metabolism & Genomics Group, Division of Human Nutrition, Wageningen University, The Netherlands, Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA and Center for Biomedical Informatics, Department of Pharmacology and Molecular Therapeutics, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA
| | - Harris A. Jaffee
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA, Department of Biostatistics, Johns Hopkins University, Baltimore, MD 21205, USA, Informatics and Systems Development, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA, Nutrition, Metabolism & Genomics Group, Division of Human Nutrition, Wageningen University, The Netherlands, Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA and Center for Biomedical Informatics, Department of Pharmacology and Molecular Therapeutics, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA
| | - Susan J. Zelisko
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA, Department of Biostatistics, Johns Hopkins University, Baltimore, MD 21205, USA, Informatics and Systems Development, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA, Nutrition, Metabolism & Genomics Group, Division of Human Nutrition, Wageningen University, The Netherlands, Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA and Center for Biomedical Informatics, Department of Pharmacology and Molecular Therapeutics, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA
| | - Neeraj Sinha
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA, Department of Biostatistics, Johns Hopkins University, Baltimore, MD 21205, USA, Informatics and Systems Development, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA, Nutrition, Metabolism & Genomics Group, Division of Human Nutrition, Wageningen University, The Netherlands, Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA and Center for Biomedical Informatics, Department of Pharmacology and Molecular Therapeutics, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA
| | - Guido Hooiveld
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA, Department of Biostatistics, Johns Hopkins University, Baltimore, MD 21205, USA, Informatics and Systems Development, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA, Nutrition, Metabolism & Genomics Group, Division of Human Nutrition, Wageningen University, The Netherlands, Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA and Center for Biomedical Informatics, Department of Pharmacology and Molecular Therapeutics, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA
| | - Rafael A. Irizarry
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA, Department of Biostatistics, Johns Hopkins University, Baltimore, MD 21205, USA, Informatics and Systems Development, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA, Nutrition, Metabolism & Genomics Group, Division of Human Nutrition, Wageningen University, The Netherlands, Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA and Center for Biomedical Informatics, Department of Pharmacology and Molecular Therapeutics, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA
| | - Michael J. Zilliox
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA, Department of Biostatistics, Johns Hopkins University, Baltimore, MD 21205, USA, Informatics and Systems Development, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA, Nutrition, Metabolism & Genomics Group, Division of Human Nutrition, Wageningen University, The Netherlands, Department of Biostatistics and Computational Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA and Center for Biomedical Informatics, Department of Pharmacology and Molecular Therapeutics, Loyola University Stritch School of Medicine, Maywood, IL 60153, USA
- *To whom correspondence should be addressed. Tel: +1 708 216 4520; Fax: +1 708 216 5881;
| |
Collapse
|
34
|
Simonis N, Migeotte I, Lambert N, Perazzolo C, de Silva DC, Dimitrov B, Heinrichs C, Janssens S, Kerr B, Mortier G, Van Vliet G, Lepage P, Casimir G, Abramowicz M, Smits G, Vilain C. FGFR1 mutations cause Hartsfield syndrome, the unique association of holoprosencephaly and ectrodactyly. J Med Genet 2013; 50:585-92. [PMID: 23812909 PMCID: PMC3756455 DOI: 10.1136/jmedgenet-2013-101603] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background Harstfield syndrome is the rare and unique association of holoprosencephaly (HPE) and ectrodactyly, with or without cleft lip and palate, and variable additional features. All the reported cases occurred sporadically. Although several causal genes of HPE and ectrodactyly have been identified, the genetic cause of Hartsfield syndrome remains unknown. We hypothesised that a single key developmental gene may underlie the co-occurrence of HPE and ectrodactyly. Methods We used whole exome sequencing in four isolated cases including one case-parents trio, and direct Sanger sequencing of three additional cases, to investigate the causative variants in Hartsfield syndrome. Results We identified a novel FGFR1 mutation in six out of seven patients. Affected residues are highly conserved and are located in the extracellular binding domain of the receptor (two homozygous mutations) or the intracellular tyrosine kinase domain (four heterozygous de novo variants). Strikingly, among the six novel mutations, three are located in close proximity to the ATP's phosphates or the coordinating magnesium, with one position required for kinase activity, and three are adjacent to known mutations involved in Kallmann syndrome plus other developmental anomalies. Conclusions Dominant or recessive FGFR1 mutations are responsible for Hartsfield syndrome, consistent with the known roles of FGFR1 in vertebrate ontogeny and conditional Fgfr1-deficient mice. Our study shows that, in humans, lack of accurate FGFR1 activation can disrupt both brain and hand/foot midline development, and that FGFR1 loss-of-function mutations are responsible for a wider spectrum of clinical anomalies than previously thought, ranging in severity from seemingly isolated hypogonadotropic hypogonadism, through Kallmann syndrome with or without additional features, to Hartsfield syndrome at its most severe end.
Collapse
Affiliation(s)
- Nicolas Simonis
- Laboratoire de Bioinformatique des Génomes et des Réseaux (BiGRe), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Lazar C, Taminau J, Meganck S, Steenhoff D, Coletta A, Solís DYW, Molter C, Duque R, Bersini H, Nowé A. GENESHIFT: a nonparametric approach for integrating microarray gene expression data based on the inner product as a distance measure between the distributions of genes. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2013; 10:383-392. [PMID: 23929862 DOI: 10.1109/tcbb.2013.12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The potential of microarray gene expression (MAGE) data is only partially explored due to the limited number of samples in individual studies. This limitation can be surmounted by merging or integrating data sets originating from independent MAGE experiments, which are designed to study the same biological problem. However, this process is hindered by batch effects that are study-dependent and result in random data distortion; therefore numerical transformations are needed to render the integration of different data sets accurate and meaningful. Our contribution in this paper is two-fold. First we propose GENESHIFT, a new nonparametric batch effect removal method based on two key elements from statistics: empirical density estimation and the inner product as a distance measure between two probability density functions; second we introduce a new validation index of batch effect removal methods based on the observation that samples from two independent studies drawn from a same population should exhibit similar probability density functions. We evaluated and compared the GENESHIFT method with four other state-of-the-art methods for batch effect removal: Batch-mean centering, empirical Bayes or COMBAT, distance-weighted discrimination, and cross-platform normalization. Several validation indices providing complementary information about the efficiency of batch effect removal methods have been employed in our validation framework. The results show that none of the methods clearly outperforms the others. More than that, most of the methods used for comparison perform very well with respect to some validation indices while performing very poor with respect to others. GENESHIFT exhibits robust performances and its average rank is the highest among the average ranks of all methods used for comparison.
Collapse
|
36
|
Taminau J, Meganck S, Lazar C, Steenhoff D, Coletta A, Molter C, Duque R, de Schaetzen V, Weiss Solís DY, Bersini H, Nowé A. Unlocking the potential of publicly available microarray data using inSilicoDb and inSilicoMerging R/Bioconductor packages. BMC Bioinformatics 2012; 13:335. [PMID: 23259851 PMCID: PMC3568420 DOI: 10.1186/1471-2105-13-335] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 12/18/2012] [Indexed: 12/20/2022] Open
Abstract
Background With an abundant amount of microarray gene expression data sets available through public repositories, new possibilities lie in combining multiple existing data sets. In this new context, analysis itself is no longer the problem, but retrieving and consistently integrating all this data before delivering it to the wide variety of existing analysis tools becomes the new bottleneck. Results We present the newly released inSilicoMerging R/Bioconductor package which, together with the earlier released inSilicoDb R/Bioconductor package, allows consistent retrieval, integration and analysis of publicly available microarray gene expression data sets. Inside the inSilicoMerging package a set of five visual and six quantitative validation measures are available as well. Conclusions By providing (i) access to uniformly curated and preprocessed data, (ii) a collection of techniques to remove the batch effects between data sets from different sources, and (iii) several validation tools enabling the inspection of the integration process, these packages enable researchers to fully explore the potential of combining gene expression data for downstream analysis. The power of using both packages is demonstrated by programmatically retrieving and integrating gene expression studies from the InSilico DB repository [https://insilicodb.org/app/].
Collapse
Affiliation(s)
- Jonatan Taminau
- AI (CoMo), Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|