151
|
Gartlgruber M, Sharma AK, Quintero A, Dreidax D, Jansky S, Park YG, Kreth S, Meder J, Doncevic D, Saary P, Toprak UH, Ishaque N, Afanasyeva E, Wecht E, Koster J, Versteeg R, Grünewald TGP, Jones DTW, Pfister SM, Henrich KO, van Nes J, Herrmann C, Westermann F. Super enhancers define regulatory subtypes and cell identity in neuroblastoma. NATURE CANCER 2021; 2:114-128. [PMID: 35121888 DOI: 10.1038/s43018-020-00145-w] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
Half of the children diagnosed with neuroblastoma (NB) have high-risk disease, disproportionately contributing to overall childhood cancer-related deaths. In addition to recurrent gene mutations, there is increasing evidence supporting the role of epigenetic deregulation in disease pathogenesis. Yet, comprehensive cis-regulatory network descriptions from NB are lacking. Here, using genome-wide H3K27ac profiles across 60 NBs, covering the different clinical and molecular subtypes, we identified four major super-enhancer-driven epigenetic subtypes and their underlying master regulatory networks. Three of these subtypes recapitulated known clinical groups; namely, MYCN-amplified, MYCN non-amplified high-risk and MYCN non-amplified low-risk NBs. The fourth subtype, exhibiting mesenchymal characteristics, shared cellular identity with multipotent Schwann cell precursors, was induced by RAS activation and was enriched in relapsed disease. Notably, CCND1, an essential gene in NB, was regulated by both mesenchymal and adrenergic regulatory networks converging on distinct super-enhancer modules. Overall, this study reveals subtype-specific super-enhancer regulation in NBs.
Collapse
Affiliation(s)
- Moritz Gartlgruber
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Ashwini Kumar Sharma
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
- Health Data Science Unit, Medical Faculty Heidelberg and BioQuant, Heidelberg, Germany
| | - Andrés Quintero
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
- Health Data Science Unit, Medical Faculty Heidelberg and BioQuant, Heidelberg, Germany
| | - Daniel Dreidax
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Selina Jansky
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Young-Gyu Park
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Sina Kreth
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Johanna Meder
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Daria Doncevic
- Health Data Science Unit, Medical Faculty Heidelberg and BioQuant, Heidelberg, Germany
| | - Paul Saary
- Health Data Science Unit, Medical Faculty Heidelberg and BioQuant, Heidelberg, Germany
| | - Umut H Toprak
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Naveed Ishaque
- Center for Digital Health, Berlin Institute of Health and Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Elena Afanasyeva
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Elisa Wecht
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Jan Koster
- Department of Oncogenomics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rogier Versteeg
- Department of Oncogenomics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Thomas G P Grünewald
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Translational Pediatric Sarcoma Research, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Pediatric Glioma Research Group, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital and Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Kai-Oliver Henrich
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany
| | - Johan van Nes
- Department of Oncogenomics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Carl Herrmann
- Health Data Science Unit, Medical Faculty Heidelberg and BioQuant, Heidelberg, Germany.
| | - Frank Westermann
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Division of Neuroblastoma Genomics, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
152
|
Mercatelli D, Lopez-Garcia G, Giorgi FM. corto: a lightweight R package for gene network inference and master regulator analysis. Bioinformatics 2020; 36:3916-3917. [PMID: 32232425 DOI: 10.1093/bioinformatics/btaa223] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/26/2020] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Gene network inference and master regulator analysis (MRA) have been widely adopted to define specific transcriptional perturbations from gene expression signatures. Several tools exist to perform such analyses but most require a computer cluster or large amounts of RAM to be executed. RESULTS We developed corto, a fast and lightweight R package to infer gene networks and perform MRA from gene expression data, with optional corrections for copy-number variations and able to run on signatures generated from RNA-Seq or ATAC-Seq data. We extensively benchmarked it to infer context-specific gene networks in 39 human tumor and 27 normal tissue datasets. AVAILABILITY AND IMPLEMENTATION Cross-platform and multi-threaded R package on CRAN (stable version) https://cran.r-project.org/package=corto and Github (development release) https://github.com/federicogiorgi/corto. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Gonzalo Lopez-Garcia
- Genetics and Genomics Science Department, Ichan School of Medicine at Mount Sinai, New York City, NY 10029-5674, USA
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| |
Collapse
|
153
|
Rosenberger G, Heusel M, Bludau I, Collins BC, Martelli C, Williams EG, Xue P, Liu Y, Aebersold R, Califano A. SECAT: Quantifying Protein Complex Dynamics across Cell States by Network-Centric Analysis of SEC-SWATH-MS Profiles. Cell Syst 2020; 11:589-607.e8. [PMID: 33333029 PMCID: PMC8034988 DOI: 10.1016/j.cels.2020.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/25/2020] [Accepted: 11/17/2020] [Indexed: 12/29/2022]
Abstract
Protein-protein interactions (PPIs) play critical functional and regulatory roles in cellular processes. They are essential for macromolecular complex formation, which in turn constitutes the basis for protein interaction networks that determine the functional state of a cell. We and others have previously shown that chromatographic fractionation of native protein complexes in combination with bottom-up mass spectrometric analysis of consecutive fractions supports the multiplexed characterization and detection of state-specific changes of protein complexes. In this study, we extend co-fractionation and mass spectrometric data analysis to perform quantitative, network-based studies of proteome organization, via the size-exclusion chromatography algorithmic toolkit (SECAT). This framework explicitly accounts for the dynamic nature and rewiring of protein complexes across multiple cell states and samples, thus, elucidating molecular mechanisms that are differentially implemented across different experimental settings. Systematic analysis of multiple datasets shows that SECAT represents a highly scalable and effective methodology to assess condition/state-specific protein-network state. A record of this paper's transparent peer review process is included in the Supplemental Information.
Collapse
Affiliation(s)
| | - Moritz Heusel
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Isabell Bludau
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Ben C Collins
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Claudia Martelli
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Evan G Williams
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Peng Xue
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland; Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yansheng Liu
- Yale Cancer Biology Institute, Yale University, West Haven CT, USA; Department of Pharmacology, Yale University School of Medicine, New Haven CT, USA
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland; Faculty of Science, University of Zürich, Zürich, Switzerland.
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York NY, USA; Department of Biomedical Informatics, Columbia University, New York NY, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York NY, USA; J.P. Sulzberger Columbia Genome Center, Columbia University, New York NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York NY, USA; Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York NY, USA.
| |
Collapse
|
154
|
Non-Coding RNAs as Prognostic Biomarkers: A miRNA Signature Specific for Aggressive Early-Stage Lung Adenocarcinomas. Noncoding RNA 2020; 6:ncrna6040048. [PMID: 33333738 PMCID: PMC7768474 DOI: 10.3390/ncrna6040048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023] Open
Abstract
Lung cancer burden can be reduced by adopting primary and secondary prevention strategies such as anti-smoking campaigns and low-dose CT screening for high risk subjects (aged >50 and smokers >30 packs/year). Recent CT screening trials demonstrated a stage-shift towards earlier stage lung cancer and reduction of mortality (~20%). However, a sizable fraction of patients (30–50%) with early stage disease still experience relapse and an adverse prognosis. Thus, the identification of effective prognostic biomarkers in stage I lung cancer is nowadays paramount. Here, we applied a multi-tiered approach relying on coupled RNA-seq and miRNA-seq data analysis of a large cohort of lung cancer patients (TCGA-LUAD, n = 510), which enabled us to identify prognostic miRNA signatures in stage I lung adenocarcinoma. Such signatures showed high accuracy (AUC ranging between 0.79 and 0.85) in scoring aggressive disease. Importantly, using a network-based approach we rewired miRNA-mRNA regulatory networks, identifying a minimal signature of 7 miRNAs, which was validated in a cohort of FFPE lung adenocarcinoma samples (CSS, n = 44) and controls a variety of genes overlapping with cancer relevant pathways. Our results further demonstrate the reliability of miRNA-based biomarkers for lung cancer prognostication and make a step forward to the application of miRNA biomarkers in the clinical routine.
Collapse
|
155
|
Simpson CM, Gnad F. Applying graph database technology for analyzing perturbed co-expression networks in cancer. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2020; 2020:6029398. [PMID: 33306799 PMCID: PMC7731929 DOI: 10.1093/database/baaa110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/20/2020] [Accepted: 11/30/2020] [Indexed: 11/13/2022]
Abstract
Graph representations provide an elegant solution to capture and analyze complex molecular mechanisms in the cell. Co-expression networks are undirected graph representations of transcriptional co-behavior indicating (co-)regulations, functional modules or even physical interactions between the corresponding gene products. The growing avalanche of available RNA sequencing (RNAseq) data fuels the construction of such networks, which are usually stored in relational databases like most other biological data. Inferring linkage by recursive multiple-join statements, however, is computationally expensive and complex to design in relational databases. In contrast, graph databases store and represent complex interconnected data as nodes, edges and properties, making it fast and intuitive to query and analyze relationships. While graph-based database technologies are on their way from a fringe domain to going mainstream, there are only a few studies reporting their application to biological data. We used the graph database management system Neo4j to store and analyze co-expression networks derived from RNAseq data from The Cancer Genome Atlas. Comparing co-expression in tumors versus healthy tissues in six cancer types revealed significant perturbation tracing back to erroneous or rewired gene regulation. Applying centrality, community detection and pathfinding graph algorithms uncovered the destruction or creation of central nodes, modules and relationships in co-expression networks of tumors. Given the speed, accuracy and straightforwardness of managing these densely connected networks, we conclude that graph databases are ready for entering the arena of biological data.
Collapse
Affiliation(s)
- Claire M Simpson
- Department of Bioinformatics and Data Science, Cell Signaling Technology Inc., 3 Trask Lane, Danvers, MA 01923, USA
| | - Florian Gnad
- Department of Bioinformatics and Data Science, Cell Signaling Technology Inc., 3 Trask Lane, Danvers, MA 01923, USA
| |
Collapse
|
156
|
Andey T, Attah MM, Akwaaba-Reynolds NA, Cheema S, Parvin-Nejad S, Acquaah-Mensah GK. Enhanced immortalization, HUWE1 mutations and other biological drivers of breast invasive carcinoma in Black/African American patients. Gene 2020; 5:100030. [PMID: 32550556 PMCID: PMC7286073 DOI: 10.1016/j.gene.2020.100030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 03/08/2020] [Indexed: 02/07/2023]
Abstract
Black/African-American (B/AA) breast cancer patients tend to have more aggressive tumor biology compared to White/Caucasians. In this study, a variety of breast tumor molecular expression profiles of patients derived from the two racial groupings were investigated. Breast invasive carcinoma sample data (RNASeq version 2, Reverse Phase Protein Array, mutation, and miRSeq data) from the Cancer Genome Atlas were examined. The results affirm that B/AA patients are more likely than Caucasian patients to harbor the aggressive basal-like or the poor prognosis-associated HER2-enriched molecular subtypes of breast cancer. There is also a higher incidence of the triple-negative breast cancer (TNBC) among B/AA patients than the general population, a fact reflected in the mutation patterns of genes such as PIK3CA and TP53. Furthermore, an immortalization signature gene set, is enriched in samples from B/AA patients. Among stage III patients, TERT, DRAP1, and PQBP1, all members of the immortalization gene signature set, are among master-regulators with increased activity in B/AA patients. Master-regulators driving differences in expression profiles between the two groups include immortalization markers, senescence markers, and immune response and redox gene products. Differences in expression, between B/AA and Caucasian patients, of RB1, hsa-let-7a, E2F1, c-MYC, TERT, and other biomolecules appear to cooperate to enhance entry into the S-phase of the cell cycle in B/AA patients. Higher expression of miR-221, an oncomiR that facilitates entry into the cell cycle S-phase, is regulated by c-MYC, which is expressed more in breast cancer samples from B/AA patients. Furthermore, the cell migration- and invasion-promoting miRNA, miR-135b, has increased relative expression in B/AA patients. Knock down of the immortalization marker TERT inhibited triple-negative breast cancer cell lines (MDA-MB-231 and MDA-MB-468) cell viability and decreased expression of TERT, MYC and WNT11. For those patients with available survival data, prognosis of stage II patients 50 years of age or younger at diagnosis, was distinctly poorer in B/AA patients. Also associated with this subset of B/AA patients are missense mutations in HUWE1 and PTEN expression loss. Relative to Caucasian non-responders to endocrine therapy, B/AA non-responders show suppressed expression of a signature gene set on which biological processes including signaling by interleukins, circadian clock, regulation of lipid metabolism by PPARα, FOXO-mediated transcription, and regulation of TP53 degradation are over-represented. Thus, we identify molecular expression patterns suggesting diminished response to oxidative stress, changes in regulation of tumor suppressors/facilitators, and enhanced immortalization in B/AA patients are likely important in defining the more aggressive molecular tumor phenotype reported in B/AA patients.
Collapse
Key Words
- ARACNe, Algorithm for the Reconstruction of Accurate Cellular Networks
- African
- B/AA, Black/African-American breast cancer patients
- B/AA50, Black/African-American stage II breast invasive carcinoma patients diagnosed at age 50 years or younger
- BrCA, breast invasive carcinoma
- Breast invasive carcinoma
- DE, differential expression
- DM, differential mutation
- EMT, Epithelial-Mesenchymal Transition
- GSEA, Gene Set Enrichment Analysis
- Immortalization
- Molecular subtype
- RMA, robust multi-array average
- RPPA, Reverse Phase Protein Array
- Race
- TCGA, the Cancer Genome Atlas
- TNBC, triple-negative breast cancer
- TRN, Transcriptional Regulatory Network
- Triple-negative breast cancer
- VIPER, Virtual Inference of Protein activity by Enriched Regulon Analysis
- W50, White stage II breast invasive carcinoma patients diagnosed at age 50 years or younger
Collapse
Affiliation(s)
- Terrick Andey
- Department of Pharmaceutical Sciences, School of Pharmacy, MCPHS University, 19 Foster St, Worcester, MA 01608, USA
| | | | - Nana Adwoa Akwaaba-Reynolds
- Department of Pharmaceutical Sciences, School of Pharmacy, MCPHS University, 19 Foster St, Worcester, MA 01608, USA
| | - Sana Cheema
- Department of Pharmaceutical Sciences, School of Pharmacy, MCPHS University, 19 Foster St, Worcester, MA 01608, USA
| | - Sara Parvin-Nejad
- Department of Pharmaceutical Sciences, School of Pharmacy, MCPHS University, 19 Foster St, Worcester, MA 01608, USA
| | - George K. Acquaah-Mensah
- Department of Pharmaceutical Sciences, School of Pharmacy, MCPHS University, 19 Foster St, Worcester, MA 01608, USA
| |
Collapse
|
157
|
Arumugam K, Shin W, Schiavone V, Vlahos L, Tu X, Carnevali D, Kesner J, Paull EO, Romo N, Subramaniam P, Worley J, Tan X, Califano A, Cosma MP. The Master Regulator Protein BAZ2B Can Reprogram Human Hematopoietic Lineage-Committed Progenitors into a Multipotent State. Cell Rep 2020; 33:108474. [PMID: 33296649 DOI: 10.1016/j.celrep.2020.108474] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/25/2020] [Accepted: 11/12/2020] [Indexed: 01/03/2023] Open
Abstract
Bi-species, fusion-mediated, somatic cell reprogramming allows precise, organism-specific tracking of unknown lineage drivers. The fusion of Tcf7l1-/- murine embryonic stem cells with EBV-transformed human B cell lymphocytes, leads to the generation of bi-species heterokaryons. Human mRNA transcript profiling at multiple time points permits the tracking of the reprogramming of B cell nuclei to a multipotent state. Interrogation of a human B cell regulatory network with gene expression signatures identifies 8 candidate master regulator proteins. Of these 8 candidates, ectopic expression of BAZ2B, from the bromodomain family, efficiently reprograms hematopoietic committed progenitors into a multipotent state and significantly enhances their long-term clonogenicity, stemness, and engraftment in immunocompromised mice. Unbiased systems biology approaches let us identify the early driving events of human B cell reprogramming.
Collapse
Affiliation(s)
- Karthik Arumugam
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - William Shin
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Valentina Schiavone
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Lukas Vlahos
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Xiaochuan Tu
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Davide Carnevali
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Jordan Kesner
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Evan O Paull
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Neus Romo
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Prem Subramaniam
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Jeremy Worley
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Xiangtian Tan
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, J.P. Sulzberger Columbia Genome Center, Department of Biomedical Informatics, Department of Biochemistry and Molecular Biophysics, Department of Medicine, Columbia University, New York, NY, USA.
| | - Maria Pia Cosma
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Dr. Aiguader 88, 08003 Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain; Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China; CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
| |
Collapse
|
158
|
Zhao L, Yao C, Xing X, Jing T, Li P, Zhu Z, Yang C, Zhai J, Tian R, Chen H, Luo J, Liu N, Deng Z, Lin X, Li N, Fang J, Sun J, Wang C, Zhou Z, Li Z. Single-cell analysis of developing and azoospermia human testicles reveals central role of Sertoli cells. Nat Commun 2020; 11:5683. [PMID: 33173058 PMCID: PMC7655944 DOI: 10.1038/s41467-020-19414-4] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 10/09/2020] [Indexed: 12/21/2022] Open
Abstract
Clinical efficacy of treatments against non-obstructive azoospermia (NOA), which affects 1% of men, are currently limited by the incomplete understanding of NOA pathogenesis and normal spermatogenic microenvironment. Here, we profile >80,000 human testicular single-cell transcriptomes from 10 healthy donors spanning the range from infant to adult and 7 NOA patients. We show that Sertoli cells, which form the scaffold in the testicular microenvironment, are severely damaged in NOA patients and identify the roadmap of Sertoli cell maturation. Notably, Sertoli cells of patients with congenital causes (Klinefelter syndrome and Y chromosome microdeletions) are mature, but exhibit abnormal immune responses, while the cells in idiopathic NOA (iNOA) are physiologically immature. Furthermore, we find that inhibition of Wnt signaling promotes the maturation of Sertoli cells from iNOA patients, allowing these cells to regain their ability to support germ cell survival. We provide a novel perspective on the development of diagnostic methods and therapeutic targets for NOA. Non-obstructive azoospermia affects 1% of men. Here, authors perform single-cell transcriptomic analysis of human testicular cells from healthy donors and non-obstructive azoospermia patients and find that inhibition of Wnt signaling promotes the maturation of Sertoli cells from patients.
Collapse
Affiliation(s)
- LiangYu Zhao
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - ChenCheng Yao
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - XiaoYu Xing
- Department of Urology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200120, China
| | - Tao Jing
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.,Department of Andrology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Peng Li
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - ZiJue Zhu
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Chao Yang
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jing Zhai
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - RuHui Tian
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - HuiXing Chen
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - JiaQiang Luo
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - NaChuan Liu
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - ZhiWen Deng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - XiaoHan Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Na Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jing Fang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.,Shanghai Advanced Research Institute, Stem Cell and Reproductive Biology Laboratory, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Jie Sun
- Department of Urology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200120, China.
| | - ChenChen Wang
- Shanghai Advanced Research Institute, Stem Cell and Reproductive Biology Laboratory, Chinese Academy of Sciences, Shanghai, 201210, China.
| | - Zhi Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Zheng Li
- Department of Andrology, the Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
159
|
Abstract
OBJECTIVES Numerous software has been developed to infer the gene regulatory network, a long-standing key topic in biology and computational biology. Yet the slowness and inaccuracy inherited in current software hamper their applications to the increasing massive data. Here, we develop a software, FINET (Fast Inferring NETwork), to infer a network with high accuracy and rapidity from big data. RESULTS The high accuracy results from integrating algorithms with stability-selection, elastic-net, and parameter optimization. Tested by a known biological network, FINET infers interactions with over 94% precision. The high speed comes from partnering parallel computations implemented with Julia, a new compiled language that runs much faster than existing languages used in the current software, such as R, Python, and MATLAB. Regardless of FINET's implementations with Julia, users with no background in the language or computer science can easily operate it, with only a user-friendly single command line. In addition, FINET can infer other networks such as chemical networks and social networks. Overall, FINET provides a confident way to efficiently and accurately infer any type of network for any scale of data.
Collapse
Affiliation(s)
- Anyou Wang
- The Institute for Integrative Genome Biology, University of California at Riverside, Riverside, CA, 92521, USA.
| | - Rong Hai
- The Institute for Integrative Genome Biology, University of California at Riverside, Riverside, CA, 92521, USA. .,Department of Microbiology and Plant Pathology, University of California at Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
160
|
Shi M, Tan S, Xie XP, Li A, Yang W, Zhu T, Wang HQ. Globally learning gene regulatory networks based on hidden atomic regulators from transcriptomic big data. BMC Genomics 2020; 21:711. [PMID: 33054712 PMCID: PMC7559338 DOI: 10.1186/s12864-020-07079-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/18/2020] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Genes are regulated by various types of regulators and most of them are still unknown or unobserved. Current gene regulatory networks (GRNs) reverse engineering methods often neglect the unknown regulators and infer regulatory relationships in a local and sub-optimal manner. RESULTS This paper proposes a global GRNs inference framework based on dictionary learning, named dlGRN. The method intends to learn atomic regulators (ARs) from gene expression data using a modified dictionary learning (DL) algorithm, which reflects the whole gene regulatory system, and predicts the regulation between a known regulator and a target gene in a global regression way. The modified DL algorithm fits the scale-free property of biological network, rendering dlGRN intrinsically discern direct and indirect regulations. CONCLUSIONS Extensive experimental results on simulation and real-world data demonstrate the effectiveness and efficiency of dlGRN in reverse engineering GRNs. A novel predicted transcription regulation between a TF TFAP2C and an oncogene EGFR was experimentally verified in lung cancer cells. Furthermore, the real application reveals the prevalence of DNA methylation regulation in gene regulatory system. dlGRN can be a standalone tool for GRN inference for its globalization and robustness.
Collapse
Affiliation(s)
- Ming Shi
- MICB Laboratory, Institute of Intelligent Machines, Hefei Institutes of Physical Science, CAS, 350 Shushanghu Road, Hefei, Anhui, 230031, P. R. China
- Current Address: MOE Key Laboratory of Bioinformatics, Division of Bioinformatics and Center for Synthetic and Systems Biology, TNLIST, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Sheng Tan
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
| | - Xin-Ping Xie
- School of Mathematics and Physics, Anhui Jianzhu University, 856 Jinzhai Road, Hefei, Anhui, 230022, P. R. China
| | - Ao Li
- School of Information Science and Technology, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
| | - Wulin Yang
- Cancer hospital & Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, CAS, 350 Shushanghu Road, Hefei, Anhui, 230031, P. R. China
| | - Tao Zhu
- Current Address: MOE Key Laboratory of Bioinformatics, Division of Bioinformatics and Center for Synthetic and Systems Biology, TNLIST, Department of Automation, Tsinghua University, Beijing, 100084, China.
| | - Hong-Qiang Wang
- MICB Laboratory, Institute of Intelligent Machines, Hefei Institutes of Physical Science, CAS, 350 Shushanghu Road, Hefei, Anhui, 230031, P. R. China.
- Cancer hospital & Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, CAS, 350 Shushanghu Road, Hefei, Anhui, 230031, P. R. China.
| |
Collapse
|
161
|
Yu B, Chen L, Zhang W, Li Y, Zhang Y, Gao Y, Teng X, Zou L, Wang Q, Jia H, Liu X, Zheng H, Hou P, Yu H, Sun Y, Zhang Z, Zhang P, Zhang L. TOP2A and CENPF are synergistic master regulators activated in cervical cancer. BMC Med Genomics 2020; 13:145. [PMID: 33023625 PMCID: PMC7541258 DOI: 10.1186/s12920-020-00800-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Background Identification of master regulators (MRs) using transcriptome data in cervical cancer (CC) could help us to develop biomarkers and find novel drug targets to fight this disease. Methods We performed differential expression (DE) analyses of public microarray and RNA-seq transcriptome data of CC and normal cervical tissues (N). Virtual Inference of Protein activity by Enriched Regulon analysis (VIPER) was used to convert the DE outcomes to differential activity (DA) signature for MRs. Synergy analysis was conducted to study synergistic effect of MR-pairs. TCGA and microarray data were used to test the association of expression of a MR and a clinical feature or a molecular feature (e.g. somatic mutations). Various bioinformatic tools/websites (DAVID, GEPIA2, Oncomine, cBioPortal) were used to analyze the expression of the top MRs and their regulons. Results Ten DE and 10 DA signatures were generated for CC. Two MRs, DNA topoisomerase II alpha (TOP2A) and centromere protein F (CENPF) were found to be up-regulated, activated and synergistic in CC compared to N across the 10 datasets. The two MRs activate a common set of genes (regulons) with functions in cell cycle, chromosome, DNA damage etc. Higher expression of CENPF was associated with metastasis. High expression of both MRs is associated with somatic mutation of a set of genes including tumor suppressors (TP53, MSH2, RB1) and genes involved in cancer pathways, cell cycle, DNA damage and repair. The magnitude of up-regulation and the absolute expression level of both MRs in CC are significantly higher compared to many other cancer types. Conclusion TOP2A and CENPF are a synergistic pair of MRs that are overexpressed and activated in CC. Their high expression is correlated with some prognosis features (e.g. metastasis) and molecular features (e.g. somatic mutations) and distinctly high in CC vs. many other cancer types. They may be good biomarkers and anticancer drug targets for CC.
Collapse
Affiliation(s)
- Beiwei Yu
- Department of Laboratory, Hangzhou Jianggan District People's Hospital, Hangzhou, Zhejiang, China
| | - Long Chen
- Department of Gynecology, Xiao shan Hospital, Hangzhou, Zhejiang, China
| | - Weina Zhang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Yue Li
- Department of Laboratory, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Yibiao Zhang
- Department of Laboratory, Zhejiang Jinhua Guangfu Hospital, Jinhua, Zhejiang, China
| | - Yuan Gao
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Xianlin Teng
- Department of Laboratory, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Libo Zou
- Medicine Reproductive Centre, Jinhua People's Hospital, Jinhua, Zhejiang, China
| | - Qian Wang
- Tianjia Genomes Tech CO., LTD., Hefei, Anhui, China
| | - Hongtao Jia
- Tianjia Genomes Tech CO., LTD., Hefei, Anhui, China
| | - Xiangtao Liu
- Tianjia Genomes Tech CO., LTD., Hefei, Anhui, China
| | - Hui Zheng
- Tianjia Genomes Tech CO., LTD., Hefei, Anhui, China
| | - Ping Hou
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Hongyan Yu
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Ying Sun
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Zhiqin Zhang
- Department of Functional Discipline, School of medicine Jinhua, Jinhua, Zhejiang, China
| | - Ping Zhang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Liqin Zhang
- Department of Laboratory, Hangzhou Jianggan District People's Hospital, Hangzhou, Zhejiang, China. .,Department of Laboratory, Jinhua People's Hospital, Jinhua, Zhejiang, China.
| |
Collapse
|
162
|
Wu J, Xiao Y, Sun J, Sun H, Chen H, Zhu Y, Fu H, Yu C, E W, Lai S, Ma L, Li J, Fei L, Jiang M, Wang J, Ye F, Wang R, Zhou Z, Zhang G, Zhang T, Ding Q, Wang Z, Hao S, Liu L, Zheng W, He J, Huang W, Wang Y, Xie J, Li T, Cheng T, Han X, Huang H, Guo G. A single-cell survey of cellular hierarchy in acute myeloid leukemia. J Hematol Oncol 2020; 13:128. [PMID: 32977829 PMCID: PMC7517826 DOI: 10.1186/s13045-020-00941-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
Background Acute myeloid leukemia (AML) is a fatal hematopoietic malignancy and has a prognosis that varies with its genetic complexity. However, there has been no appropriate integrative analysis on the hierarchy of different AML subtypes. Methods Using Microwell-seq, a high-throughput single-cell mRNA sequencing platform, we analyzed the cellular hierarchy of bone marrow samples from 40 patients and 3 healthy donors. We also used single-cell single-molecule real-time (SMRT) sequencing to investigate the clonal heterogeneity of AML cells. Results From the integrative analysis of 191727 AML cells, we established a single-cell AML landscape and identified an AML progenitor cell cluster with novel AML markers. Patients with ribosomal protein high progenitor cells had a low remission rate. We deduced two types of AML with diverse clinical outcomes. We traced mitochondrial mutations in the AML landscape by combining Microwell-seq with SMRT sequencing. We propose the existence of a phenotypic “cancer attractor” that might help to define a common phenotype for AML progenitor cells. Finally, we explored the potential drug targets by making comparisons between the AML landscape and the Human Cell Landscape. Conclusions We identified a key AML progenitor cell cluster. A high ribosomal protein gene level indicates the poor prognosis. We deduced two types of AML and explored the potential drug targets. Our results suggest the existence of a cancer attractor.
Collapse
Affiliation(s)
- Junqing Wu
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Yanyu Xiao
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Jie Sun
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Huiyu Sun
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Haide Chen
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Yuanyuan Zhu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Huarui Fu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chengxuan Yu
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Weigao E
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Shujing Lai
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Lifeng Ma
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Jiaqi Li
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Lijiang Fei
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Mengmeng Jiang
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Jingjing Wang
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Fang Ye
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Renying Wang
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Ziming Zhou
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Guodong Zhang
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Tingyue Zhang
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Qiong Ding
- Wuhan Biobank Co., LTD, Wuhan, 430075, China
| | - Zou Wang
- Wuhan Biobank Co., LTD, Wuhan, 430075, China
| | - Sheng Hao
- Wuhan Biobank Co., LTD, Wuhan, 430075, China
| | - Lizhen Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Weiyan Zheng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jingsong He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Weijia Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yungui Wang
- Institute of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jin Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
| | - Tiefeng Li
- Institute of Applied Mechanics, Zhejiang University, Hangzhou, 310027, China
| | - Tao Cheng
- Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300000, China.,Alliance for Atlas of Blood Cells, Tianjin, China
| | - Xiaoping Han
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China. .,Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 311121, China.
| | - He Huang
- Institute of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China. .,Alliance for Atlas of Blood Cells, Tianjin, China. .,Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 311121, China.
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Institute of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China. .,Alliance for Atlas of Blood Cells, Tianjin, China. .,Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 311121, China.
| |
Collapse
|
163
|
Single-cell transcriptomic atlas of the human endometrium during the menstrual cycle. Nat Med 2020; 26:1644-1653. [PMID: 32929266 DOI: 10.1038/s41591-020-1040-z] [Citation(s) in RCA: 309] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 07/29/2020] [Indexed: 12/20/2022]
Abstract
In a human menstrual cycle the endometrium undergoes remodeling, shedding and regeneration, all of which are driven by substantial gene expression changes in the underlying cellular hierarchy. Despite its importance in human fertility and regenerative biology, our understanding of this unique type of tissue homeostasis remains rudimentary. We characterized the transcriptomic transformation of human endometrium at single-cell resolution across the menstrual cycle, resolving cellular heterogeneity in multiple dimensions. We profiled the behavior of seven endometrial cell types, including a previously uncharacterized ciliated cell type, during four major phases of endometrial transformation, and found characteristic signatures for each cell type and phase. We discovered that the human window of implantation opens with an abrupt and discontinuous transcriptomic activation in the epithelia, accompanied with a widespread decidualization feature in the stromal fibroblasts. Our study provides a high-resolution molecular and cellular characterization of human endometrial transformation across the menstrual cycle, providing insights into this essential physiological process.
Collapse
|
164
|
Liu Y, Shi N, Regev A, He S, Hemann MT. Integrated regulatory models for inference of subtype-specific susceptibilities in glioblastoma. Mol Syst Biol 2020; 16:e9506. [PMID: 32974985 PMCID: PMC7516378 DOI: 10.15252/msb.20209506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a highly malignant form of cancer that lacks effective treatment options or well-defined strategies for personalized cancer therapy. The disease has been stratified into distinct molecular subtypes; however, the underlying regulatory circuitry that gives rise to such heterogeneity and its implications for therapy remain unclear. We developed a modular computational pipeline, Integrative Modeling of Transcription Regulatory Interactions for Systematic Inference of Susceptibility in Cancer (inTRINSiC), to dissect subtype-specific regulatory programs and predict genetic dependencies in individual patient tumors. Using a multilayer network consisting of 518 transcription factors (TFs), 10,733 target genes, and a signaling layer of 3,132 proteins, we were able to accurately identify differential regulatory activity of TFs that shape subtype-specific expression landscapes. Our models also allowed inference of mechanisms for altered TF behavior in different GBM subtypes. Most importantly, we were able to use the multilayer models to perform an in silico perturbation analysis to infer differential genetic vulnerabilities across GBM subtypes and pinpoint the MYB family member MYBL2 as a drug target specific for the Proneural subtype.
Collapse
Affiliation(s)
- Yunpeng Liu
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMAUSA
- MIT Koch Institute for Integrative Cancer ResearchCambridgeMAUSA
- Broad Institute of MIT and HarvardCambridgeMAUSA
| | - Ning Shi
- School of Computer ScienceUniversity of BirminghamBirminghamUK
| | - Aviv Regev
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMAUSA
- MIT Koch Institute for Integrative Cancer ResearchCambridgeMAUSA
- Broad Institute of MIT and HarvardCambridgeMAUSA
| | - Shan He
- School of Computer ScienceUniversity of BirminghamBirminghamUK
| | - Michael T Hemann
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMAUSA
- MIT Koch Institute for Integrative Cancer ResearchCambridgeMAUSA
- Broad Institute of MIT and HarvardCambridgeMAUSA
| |
Collapse
|
165
|
Clarke R, Kraikivski P, Jones BC, Sevigny CM, Sengupta S, Wang Y. A systems biology approach to discovering pathway signaling dysregulation in metastasis. Cancer Metastasis Rev 2020; 39:903-918. [PMID: 32776157 PMCID: PMC7487029 DOI: 10.1007/s10555-020-09921-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
Abstract
Total metastatic burden is the primary cause of death for many cancer patients. While the process of metastasis has been studied widely, much remains to be understood. Moreover, few agents have been developed that specifically target the major steps of the metastatic cascade. Many individual genes and pathways have been implicated in metastasis but a holistic view of how these interact and cooperate to regulate and execute the process remains somewhat rudimentary. It is unclear whether all of the signaling features that regulate and execute metastasis are yet fully understood. Novel features of a complex system such as metastasis can often be discovered by taking a systems-based approach. We introduce the concepts of systems modeling and define some of the central challenges facing the application of a multidisciplinary systems-based approach to understanding metastasis and finding actionable targets therein. These challenges include appreciating the unique properties of the high-dimensional omics data often used for modeling, limitations in knowledge of the system (metastasis), tumor heterogeneity and sampling bias, and some of the issues key to understanding critical features of molecular signaling in the context of metastasis. We also provide a brief introduction to integrative modeling that focuses on both the nodes and edges of molecular signaling networks. Finally, we offer some observations on future directions as they relate to developing a systems-based model of the metastatic cascade.
Collapse
Affiliation(s)
- Robert Clarke
- Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA.
- Hormel Institute and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Austin, MN, 55912, USA.
| | - Pavel Kraikivski
- Academy of Integrated Science, Division of Systems Biology, Virginia Polytechnic and State University, Blacksburg, VA, 24061, USA
| | - Brandon C Jones
- Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA
| | - Catherine M Sevigny
- Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA
| | - Surojeet Sengupta
- Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Rd NW, Washington, DC, 20057, USA
| | - Yue Wang
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| |
Collapse
|
166
|
Fagnan A, Bagger FO, Piqué-Borràs MR, Ignacimouttou C, Caulier A, Lopez CK, Robert E, Uzan B, Gelsi-Boyer V, Aid Z, Thirant C, Moll U, Tauchmann S, Kurtovic-Kozaric A, Maciejewski J, Dierks C, Spinelli O, Salmoiraghi S, Pabst T, Shimoda K, Deleuze V, Lapillonne H, Sweeney C, De Mas V, Leite B, Kadri Z, Malinge S, de Botton S, Micol JB, Kile B, Carmichael CL, Iacobucci I, Mullighan CG, Carroll M, Valent P, Bernard OA, Delabesse E, Vyas P, Birnbaum D, Anguita E, Garçon L, Soler E, Schwaller J, Mercher T. Human erythroleukemia genetics and transcriptomes identify master transcription factors as functional disease drivers. Blood 2020; 136:698-714. [PMID: 32350520 PMCID: PMC8215330 DOI: 10.1182/blood.2019003062] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
Acute erythroleukemia (AEL or acute myeloid leukemia [AML]-M6) is a rare but aggressive hematologic malignancy. Previous studies showed that AEL leukemic cells often carry complex karyotypes and mutations in known AML-associated oncogenes. To better define the underlying molecular mechanisms driving the erythroid phenotype, we studied a series of 33 AEL samples representing 3 genetic AEL subgroups including TP53-mutated, epigenetic regulator-mutated (eg, DNMT3A, TET2, or IDH2), and undefined cases with low mutational burden. We established an erythroid vs myeloid transcriptome-based space in which, independently of the molecular subgroup, the majority of the AEL samples exhibited a unique mapping different from both non-M6 AML and myelodysplastic syndrome samples. Notably, >25% of AEL patients, including in the genetically undefined subgroup, showed aberrant expression of key transcriptional regulators, including SKI, ERG, and ETO2. Ectopic expression of these factors in murine erythroid progenitors blocked in vitro erythroid differentiation and led to immortalization associated with decreased chromatin accessibility at GATA1-binding sites and functional interference with GATA1 activity. In vivo models showed development of lethal erythroid, mixed erythroid/myeloid, or other malignancies depending on the cell population in which AEL-associated alterations were expressed. Collectively, our data indicate that AEL is a molecularly heterogeneous disease with an erythroid identity that results in part from the aberrant activity of key erythroid transcription factors in hematopoietic stem or progenitor cells.
Collapse
Affiliation(s)
- Alexandre Fagnan
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Frederik Otzen Bagger
- University Children's Hospital Beider Basel (UKBB), Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Center for Genomic Medicine, Copenhagen University Hospital, Copenhagen, Denmark
- Swiss Institute of Bioinformatics, Basel, Basel, Switzerland
| | - Maria-Riera Piqué-Borràs
- University Children's Hospital Beider Basel (UKBB), Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Cathy Ignacimouttou
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Alexis Caulier
- Equipe d'Accueil (EA) 4666, Hématopoïèse et Immunologie (HEMATIM), Université de Picardie Jules Verne (UPJV), Amiens, France
- Service Hématologie Biologique, Centre Hospitalier Universitaire (CHU) Amiens, Amiens, France
| | - Cécile K Lopez
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Elie Robert
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Benjamin Uzan
- Unité Mixte de Recherche 967 (UMR 967), INSERM-Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA)/Direction de la Recherche Fondamentale (DRF)/Institut de Biologie François Jacob (IBFJ)/Institut de Radiobiologie Cellulaire et Moléculaire (IRCM)/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL)-Université Paris-Diderot-Université Paris-Sud, Fontenay-aux-Roses, France
| | - Véronique Gelsi-Boyer
- U1068 and
- UMR7258, Centre de Recherche en Cancérologie de Marseille, Centre National de la Recherche Scientifique (CNRS)/INSERM/Institut Paoli Calmettes/Aix-Marseille Université, Marseille, France
| | - Zakia Aid
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Cécile Thirant
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Ute Moll
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
- Department of Pathology, Stony Brook University, Stony Brook, NY
| | - Samantha Tauchmann
- University Children's Hospital Beider Basel (UKBB), Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Amina Kurtovic-Kozaric
- Clinical Center of the University of Sarajevo, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Jaroslaw Maciejewski
- Department of Translational Hematology and Oncologic Research, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | - Christine Dierks
- Hämatologie, Onkologie und Stammzelltransplantation, Klinik für Innere Medizin I, Freiburg, Germany
| | - Orietta Spinelli
- UOC Ematologia, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Silvia Salmoiraghi
- UOC Ematologia, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII Hospital, Bergamo, Italy
- FROM Research Foundation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Thomas Pabst
- Department of Oncology, Inselspital, University Hospital Bern/University of Bern, Bern, Switzerland
| | - Kazuya Shimoda
- Gastroenterology and Hematology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Virginie Deleuze
- IGMM, University of Montpellier, CNRS, Montpellier, France
- Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | - Hélène Lapillonne
- Centre de Recherche Saint Antoine (CRSA)-Unité INSERM, Sorbonne Université/Assistance Publique-Hôpitaux de Paris (AP-HP)/Hôpital Trousseau, Paris, France
| | - Connor Sweeney
- Medical Research Council Molecular Haematology Unit (MRC MHU), Biomedical Research Centre (BRC) Hematology Theme, Oxford Biomedical Research Centre, Oxford Centre for Haematology, Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Véronique De Mas
- Team 16, Hematology Laboratory, Center of Research of Cancerology of Toulouse, U1037, INSERM/Institut Universitaire du Cancer de Toulouse (IUCT) Oncopole, Toulouse, France
| | - Betty Leite
- Genomic Platform, Unité Mixte de Service - Analyse Moléculaire, Modélisation et Imagerie de la maladie Cancéreuse (UMS AMMICA), Gustave Roussy/Université Paris-Saclay, Villejuif, France
| | - Zahra Kadri
- Division of Innovative Therapies, UMR-1184, Immunologie des Maladies Virales, Auto-immunes, Hématologiques et Bactériennes (IMVA-HB) and Infectious Disease Models and Innovative Therapies (IDMIT) Center, CEA/INSERM/Paris-Saclay University, Fontenay-aux-Roses, France
| | - Sébastien Malinge
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
- Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, Australia
| | - Stéphane de Botton
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Jean-Baptiste Micol
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
| | - Benjamin Kile
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | | | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
- Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN
| | - Martin Carroll
- Division of Hematology and Oncology, University of Pennsylvania, PA
| | - Peter Valent
- Division of Hematology and Hemostaseology, Department of Internal Medicine I and
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Olivier A Bernard
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Eric Delabesse
- Team 16, Hematology Laboratory, Center of Research of Cancerology of Toulouse, U1037, INSERM/Institut Universitaire du Cancer de Toulouse (IUCT) Oncopole, Toulouse, France
| | - Paresh Vyas
- Medical Research Council Molecular Haematology Unit (MRC MHU), Biomedical Research Centre (BRC) Hematology Theme, Oxford Biomedical Research Centre, Oxford Centre for Haematology, Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Daniel Birnbaum
- U1068 and
- UMR7258, Centre de Recherche en Cancérologie de Marseille, Centre National de la Recherche Scientifique (CNRS)/INSERM/Institut Paoli Calmettes/Aix-Marseille Université, Marseille, France
| | - Eduardo Anguita
- Hematology Department
- Instituto de Medicina de Laboratorio (IML), and
- Instituto de Investigación Sanitaria San Carlos, (IdISSC), Hospital Clínico San Carlos (HCSC), Madrid, Spain; and
- Department of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Loïc Garçon
- Equipe d'Accueil (EA) 4666, Hématopoïèse et Immunologie (HEMATIM), Université de Picardie Jules Verne (UPJV), Amiens, France
- Service Hématologie Biologique, Centre Hospitalier Universitaire (CHU) Amiens, Amiens, France
| | - Eric Soler
- IGMM, University of Montpellier, CNRS, Montpellier, France
- Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | - Juerg Schwaller
- University Children's Hospital Beider Basel (UKBB), Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Thomas Mercher
- Unité 1170 (U1170), INSERM, Gustave Roussy, Université Paris Diderot, Villejuif, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| |
Collapse
|
167
|
Martinez-Gutierrez AD, Cantú de León D, Millan-Catalan O, Coronel-Hernandez J, Campos-Parra AD, Porras-Reyes F, Exayana-Alderete A, López-Camarillo C, Jacobo-Herrera NJ, Ramos-Payan R, Pérez-Plasencia C. Identification of miRNA Master Regulators in Breast Cancer. Cells 2020; 9:1610. [PMID: 32635183 PMCID: PMC7407970 DOI: 10.3390/cells9071610] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/18/2020] [Accepted: 06/25/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the neoplasm with the highest number of deaths in women. Although the molecular mechanisms associated with the development of this tumor have been widely described, metastatic disease has a high mortality rate. In recent years, several studies show that microRNAs or miRNAs regulate complex processes in different biological systems including cancer. In the present work, we describe a group of 61 miRNAs consistently over-expressed in breast cancer (BC) samples that regulate the breast cancer transcriptome. By means of data mining from TCGA, miRNA and mRNA sequencing data corresponding to 1091 BC patients and 110 normal adjacent tissues were downloaded and a miRNA-mRNA network was inferred. Calculations of their oncogenic activity demonstrated that they were involved in the regulation of classical cancer pathways such as cell cycle, PI3K-AKT, DNA repair, and k-Ras signaling. Using univariate and multivariate analysis, we found that five of these miRNAs could be used as biomarkers for the prognosis of overall survival. Furthermore, we confirmed the over-expression of two of them in 56 locally advanced BC samples obtained from the histopathological archive of the National Cancer Institute of Mexico, showing concordance with our previous bioinformatic analysis.
Collapse
Affiliation(s)
- Antonio Daniel Martinez-Gutierrez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX 14080, Mexico; (A.D.M.-G.); (D.C.d.L.); (O.M.-C.); (J.C.-H.); (A.D.C.-P.)
| | - David Cantú de León
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX 14080, Mexico; (A.D.M.-G.); (D.C.d.L.); (O.M.-C.); (J.C.-H.); (A.D.C.-P.)
| | - Oliver Millan-Catalan
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX 14080, Mexico; (A.D.M.-G.); (D.C.d.L.); (O.M.-C.); (J.C.-H.); (A.D.C.-P.)
| | - Jossimar Coronel-Hernandez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX 14080, Mexico; (A.D.M.-G.); (D.C.d.L.); (O.M.-C.); (J.C.-H.); (A.D.C.-P.)
| | - Alma D. Campos-Parra
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX 14080, Mexico; (A.D.M.-G.); (D.C.d.L.); (O.M.-C.); (J.C.-H.); (A.D.C.-P.)
| | - Fany Porras-Reyes
- Servicio de Anatomía Patológica, Instituto Nacional de Cancerología, Tlalpan, CDMX 14080, Mexico;
| | | | - César López-Camarillo
- Posgrado en Ciencias Biomédicas, Universidad Autónoma de la Ciudad de México, CDMX 03100, Mexico;
| | | | - Rosalio Ramos-Payan
- Faculty of Biology, Autonomous University of Sinaloa, Culiacán 80007. Sin, Mexico;
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX 14080, Mexico; (A.D.M.-G.); (D.C.d.L.); (O.M.-C.); (J.C.-H.); (A.D.C.-P.)
- Laboratorio de Genómica, Unidad de Biomedicina, FES-IZTACALA, UNAM, Tlalnepantla 54090, Mexico
| |
Collapse
|
168
|
Khatamian A, Paull EO, Califano A, Yu J. SJARACNe: a scalable software tool for gene network reverse engineering from big data. Bioinformatics 2020; 35:2165-2166. [PMID: 30388204 PMCID: PMC6581437 DOI: 10.1093/bioinformatics/bty907] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 12/31/2022] Open
Abstract
SUMMARY Over the last two decades, we have observed an exponential increase in the number of generated array or sequencing-based transcriptomic profiles. Reverse engineering of biological networks from high-throughput gene expression profiles has been one of the grand challenges in systems biology. The Algorithm for the Reconstruction of Accurate Cellular Networks (ARACNe) represents one of the most effective and widely-used tools to address this challenge. However, existing ARACNe implementations do not efficiently process big input data with thousands of samples. Here we present an improved implementation of the algorithm, SJARACNe, to solve this big data problem, based on sophisticated software engineering. The new scalable SJARACNe package achieves a dramatic improvement in computational performance in both time and memory usage and implements new features while preserving the network inference accuracy of the original algorithm. Given that large-sampled transcriptomic data is increasingly available and ARACNe is extremely demanding for network reconstruction, the scalable SJARACNe will allow even researchers with modest computational resources to efficiently construct complex regulatory and signaling networks from thousands of gene expression profiles. AVAILABILITY AND IMPLEMENTATION SJARACNe is implemented in C++ (computational core) and Python (pipelining scripting wrapper, ≥3.6.1). It is freely available at https://github.com/jyyulab/SJARACNe. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Alireza Khatamian
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Evan O Paull
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
169
|
Chanda P, Costa E, Hu J, Sukumar S, Van Hemert J, Walia R. Information Theory in Computational Biology: Where We Stand Today. ENTROPY (BASEL, SWITZERLAND) 2020; 22:E627. [PMID: 33286399 PMCID: PMC7517167 DOI: 10.3390/e22060627] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/31/2020] [Accepted: 06/03/2020] [Indexed: 12/30/2022]
Abstract
"A Mathematical Theory of Communication" was published in 1948 by Claude Shannon to address the problems in the field of data compression and communication over (noisy) communication channels. Since then, the concepts and ideas developed in Shannon's work have formed the basis of information theory, a cornerstone of statistical learning and inference, and has been playing a key role in disciplines such as physics and thermodynamics, probability and statistics, computational sciences and biological sciences. In this article we review the basic information theory based concepts and describe their key applications in multiple major areas of research in computational biology-gene expression and transcriptomics, alignment-free sequence comparison, sequencing and error correction, genome-wide disease-gene association mapping, metabolic networks and metabolomics, and protein sequence, structure and interaction analysis.
Collapse
Affiliation(s)
- Pritam Chanda
- Corteva Agriscience™, Indianapolis, IN 46268, USA
- Computer and Information Science, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | - Eduardo Costa
- Corteva Agriscience™, Mogi Mirim, Sao Paulo 13801-540, Brazil
| | - Jie Hu
- Corteva Agriscience™, Indianapolis, IN 46268, USA
| | | | | | - Rasna Walia
- Corteva Agriscience™, Johnston, IA 50131, USA
| |
Collapse
|
170
|
Díaz-Valenzuela E, Sawers RH, Cibrián-Jaramillo A. Cis- and Trans-Regulatory Variations in the Domestication of the Chili Pepper Fruit. Mol Biol Evol 2020; 37:1593-1603. [PMID: 32031611 PMCID: PMC7253206 DOI: 10.1093/molbev/msaa027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The process of domestication requires the rapid transformation of the wild morphology into the cultivated forms that humans select for. This process often takes place through changes in the regulation of genes, yet, there is no definite pattern on the role of cis- and trans-acting regulatory variations in the domestication of the fruit among crops. Using allele-specific expression and network analyses, we characterized the regulatory patterns and the inheritance of gene expression in wild and cultivated accessions of chili pepper, a crop with remarkable fruit morphological variation. We propose that gene expression differences associated to the cultivated form are best explained by cis-regulatory hubs acting through trans-regulatory cascades. We show that in cultivated chili, the expression of genes associated with fruit morphology is partially recessive with respect to those in the wild relative, consistent with the hybrid fruit phenotype. Decreased expression of fruit maturation and growth genes in cultivated chili suggest that selection for loss-of-function took place in its domestication. Trans-regulatory changes underlie the majority of the genes showing regulatory divergence and had larger effect sizes on gene expression than cis-regulatory variants. Network analysis of selected cis-regulated genes, including ARP9 and MED25, indicated their interaction with many transcription factors involved in organ growth and fruit ripening. Differentially expressed genes linked to cis-regulatory variants and their interactions with downstream trans-acting genes have the potential to drive the morphological differences observed between wild and cultivated fruits and provide an attractive mechanism of morphological transformation during the domestication of the chili pepper.
Collapse
Affiliation(s)
- Erik Díaz-Valenzuela
- Ecological and Evolutionary Genomics Laboratory, Unidad de Genomica Avanzada (Langebio), Irapuato, Guanajuato, México
| | - Ruairidh H Sawers
- Department of Plant Science, The Pennsylvania State University, University Park State College, University Park, PA
| | - Angélica Cibrián-Jaramillo
- Ecological and Evolutionary Genomics Laboratory, Unidad de Genomica Avanzada (Langebio), Irapuato, Guanajuato, México
| |
Collapse
|
171
|
Holding AN, Cook HV, Markowetz F. Data generation and network reconstruction strategies for single cell transcriptomic profiles of CRISPR-mediated gene perturbations. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2020; 1863:194441. [PMID: 31756390 DOI: 10.1016/j.bbagrm.2019.194441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 02/05/2023]
Abstract
Recent advances in single-cell RNA-sequencing (scRNA-seq) in combination with CRISPR/Cas9 technologies have enabled the development of methods for large-scale perturbation studies with transcriptional readouts. These methods are highly scalable and have the potential to provide a wealth of information on the biological networks that underlie cellular response. Here we discuss how to overcome several key challenges to generate and analyse data for the confident reconstruction of models of the underlying cellular network. Some challenges are generic, and apply to analysing any single-cell transcriptomic data, while others are specific to combined single-cell CRISPR/Cas9 data, in particular barcode swapping, knockdown efficiency, multiplicity of infection and potential confounding factors. We also provide a curated collection of published data sets to aid the development of analysis strategies. Finally, we discuss several network reconstruction approaches, including co-expression networks and Bayesian networks, as well as their limitations, and highlight the potential of Nested Effects Models for network reconstruction from scRNA-seq data. This article is part of a Special Issue entitled: Transcriptional Profiles and Regulatory Gene Networks edited by Dr. Dr. Federico Manuel Giorgi and Dr. Shaun Mahony.
Collapse
Affiliation(s)
- Andrew N Holding
- Department of Biology, University of York, York, UK; York Biomedical Research Institute, University of York, York, UK; CRUK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, UK; The Alan Turing Institute, 96 Euston Road, Kings Cross, London, UK
| | - Helen V Cook
- Department of Biology, University of York, York, UK
| | | |
Collapse
|
172
|
Sauta E, Demartini A, Vitali F, Riva A, Bellazzi R. A Bayesian data fusion based approach for learning genome-wide transcriptional regulatory networks. BMC Bioinformatics 2020; 21:219. [PMID: 32471360 PMCID: PMC7257163 DOI: 10.1186/s12859-020-3510-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/22/2020] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Reverse engineering of transcriptional regulatory networks (TRN) from genomics data has always represented a computational challenge in System Biology. The major issue is modeling the complex crosstalk among transcription factors (TFs) and their target genes, with a method able to handle both the high number of interacting variables and the noise in the available heterogeneous experimental sources of information. RESULTS In this work, we propose a data fusion approach that exploits the integration of complementary omics-data as prior knowledge within a Bayesian framework, in order to learn and model large-scale transcriptional networks. We develop a hybrid structure-learning algorithm able to jointly combine TFs ChIP-Sequencing data and gene expression compendia to reconstruct TRNs in a genome-wide perspective. Applying our method to high-throughput data, we verified its ability to deal with the complexity of a genomic TRN, providing a snapshot of the synergistic TFs regulatory activity. Given the noisy nature of data-driven prior knowledge, which potentially contains incorrect information, we also tested the method's robustness to false priors on a benchmark dataset, comparing the proposed approach to other regulatory network reconstruction algorithms. We demonstrated the effectiveness of our framework by evaluating structural commonalities of our learned genomic network with other existing networks inferred by different DNA binding information-based methods. CONCLUSIONS This Bayesian omics-data fusion based methodology allows to gain a genome-wide picture of the transcriptional interplay, helping to unravel key hierarchical transcriptional interactions, which could be subsequently investigated, and it represents a promising learning approach suitable for multi-layered genomic data integration, given its robustness to noisy sources and its tailored framework for handling high dimensional data.
Collapse
Affiliation(s)
- Elisabetta Sauta
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Via Ferrata 5, 27100, Pavia, Italy.
| | - Andrea Demartini
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Via Ferrata 5, 27100, Pavia, Italy
| | - Francesca Vitali
- Center for Biomedical Informatics and Biostatistics, Dept. of Medicine, The University of Arizona Health Sciences, 1230 Cherry Ave, Tucson, AZ, 85719, USA
| | - Alberto Riva
- Bioinformatics Core, Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL, 32610, USA
| | - Riccardo Bellazzi
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Via Ferrata 5, 27100, Pavia, Italy
| |
Collapse
|
173
|
Espinoza JL, Shah N, Singh S, Nelson KE, Dupont CL. Applications of weighted association networks applied to compositional data in biology. Environ Microbiol 2020; 22:3020-3038. [PMID: 32436334 DOI: 10.1111/1462-2920.15091] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022]
Abstract
Next-generation sequencing technologies have generated, and continue to produce, an increasingly large corpus of biological data. The data generated are inherently compositional as they convey only relative information dependent upon the capacity of the instrument, experimental design and technical bias. There is considerable information to be gained through network analysis by studying the interactions between components within a system. Network theory methods using compositional data are powerful approaches for quantifying relationships between biological components and their relevance to phenotype, environmental conditions or other external variables. However, many of the statistical assumptions used for network analysis are not designed for compositional data and can bias downstream results. In this mini-review, we illustrate the utility of network theory in biological systems and investigate modern techniques while introducing researchers to frameworks for implementation. We overview (1) compositional data analysis, (2) data transformations and (3) network theory along with insight on a battery of network types including static-, temporal-, sample-specific- and differential-networks. The intention of this mini-review is not to provide a comprehensive overview of network methods, rather to introduce microbiology researchers to (semi)-unsupervised data-driven approaches for inferring latent structures that may give insight into biological phenomena or abstract mechanics of complex systems.
Collapse
Affiliation(s)
- Josh L Espinoza
- J. Craig Venter Institute, La Jolla, USA.,Applied Sciences, Durban University of Technology, Durban, South Africa
| | | | - Suren Singh
- Applied Sciences, Durban University of Technology, Durban, South Africa
| | - Karen E Nelson
- J. Craig Venter Institute, La Jolla, USA.,Applied Sciences, Durban University of Technology, Durban, South Africa.,J. Craig Venter Institute, Rockville, USA
| | | |
Collapse
|
174
|
Single-Cell Transcriptome Analysis Maps the Developmental Track of the Human Heart. Cell Rep 2020; 26:1934-1950.e5. [PMID: 30759401 DOI: 10.1016/j.celrep.2019.01.079] [Citation(s) in RCA: 320] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/14/2018] [Accepted: 01/22/2019] [Indexed: 02/06/2023] Open
Abstract
The heart is the central organ of the circulatory system, and its proper development is vital for maintaining human life. Here, we used single-cell RNA sequencing to profile the gene expression landscapes of ∼4,000 cardiac cells from human embryos and identified four major types of cells: cardiomyocytes (CMs), cardiac fibroblasts, endothelial cells (ECs), and valvar interstitial cells (VICs). Atrial and ventricular CMs acquired distinct features early in heart development. Furthermore, both CMs and fibroblasts show stepwise changes in gene expression. As development proceeds, VICs may be involved in the remodeling phase, and ECs display location-specific characteristics. Finally, we compared gene expression profiles between humans and mice and identified a series of unique features of human heart development. Our study lays the groundwork for elucidating the mechanisms of in vivo human cardiac development and provides potential clues to understand cardiac regeneration.
Collapse
|
175
|
Liu P, Luo J, Zheng Q, Chen Q, Zhai N, Xu S, Xu Y, Jin L, Xu G, Lu X, Xu G, Wang G, Shao J, Xu H, Cao P, Zhou H, Wang X. Integrating transcriptome and metabolome reveals molecular networks involved in genetic and environmental variation in tobacco. DNA Res 2020; 27:dsaa006. [PMID: 32324848 PMCID: PMC7320822 DOI: 10.1093/dnares/dsaa006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/16/2020] [Indexed: 12/23/2022] Open
Abstract
Tobacco (Nicotiana tabacum) is one of the most widely cultivated commercial non-food crops with significant social and economic impacts. Here we profiled transcriptome and metabolome from 54 tobacco samples (2-3 replicates; n = 151 in total) collected from three varieties (i.e. genetic factor), three locations (i.e. environmental factor), and six developmental stages (i.e. developmental process). We identified 3,405 differentially expressed (DE) genes (DEGs) and 371 DE metabolites, respectively. We used quantitative real-time PCR to validate 20 DEGs, and confirmed 18/20 (90%) DEGs between three locations and 16/20 (80%) with the same trend across developmental stages. We then constructed nine co-expression gene modules and four co-expression metabolite modules , and defined seven de novo regulatory networks, including nicotine- and carotenoid-related regulatory networks. A novel two-way Pearson correlation approach was further proposed to integrate co-expression gene and metabolite modules to identify joint gene-metabolite relations. Finally, we further integrated DE and network results to prioritize genes by its functional importance and identified a top-ranked novel gene, LOC107773232, as a potential regulator involved in the carotenoid metabolism pathway. Thus, the results and systems-biology approaches provide a new avenue to understand the molecular mechanisms underlying complex genetic and environmental perturbations in tobacco.
Collapse
Affiliation(s)
- Pingping Liu
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Jie Luo
- Central Laboratory of Zhejiang Academy of Agricultural Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Qingxia Zheng
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Qiansi Chen
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Niu Zhai
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Shengchun Xu
- Central Laboratory of Zhejiang Academy of Agricultural Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Yalong Xu
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Lifeng Jin
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Guoyun Xu
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Xin Lu
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Guowang Xu
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Gangjun Wang
- Central Laboratory of Zhejiang Academy of Agricultural Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Jianfeng Shao
- Central Laboratory of Zhejiang Academy of Agricultural Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Hai‐Ming Xu
- Institute of Crop Science and Institute of Bioinformatics, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Peijian Cao
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Huina Zhou
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Xusheng Wang
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA
| |
Collapse
|
176
|
Abstract
A key goal of cancer systems biology is to use big data to elucidate the molecular networks by which cancer develops. However, to date there has been no systematic evaluation of how far these efforts have progressed. In this Analysis, we survey six major systems biology approaches for mapping and modelling cancer pathways with attention to how well their resulting network maps cover and enhance current knowledge. Our sample of 2,070 systems biology maps captures all literature-curated cancer pathways with significant enrichment, although the strong tendency is for these maps to recover isolated mechanisms rather than entire integrated processes. Systems biology maps also identify previously underappreciated functions, such as a potential role for human papillomavirus-induced chromosomal alterations in ovarian tumorigenesis, and they add new genes to known cancer pathways, such as those related to metabolism, Hippo signalling and immunity. Notably, we find that many cancer networks have been provided only in journal figures and not for programmatic access, underscoring the need to deposit network maps in community databases to ensure they can be readily accessed. Finally, few of these findings have yet been clinically translated, leaving ample opportunity for future translational studies. Periodic surveys of cancer pathway maps, such as the one reported here, are critical to assess progress in the field and identify underserved areas of methodology and cancer biology.
Collapse
Affiliation(s)
- Brent M Kuenzi
- Division of Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
177
|
Ung M, Schaafsma E, Mattox D, Wang GL, Cheng C. Pan-cancer systematic identification of lncRNAs associated with cancer prognosis. PeerJ 2020; 8:e8797. [PMID: 32231885 PMCID: PMC7100599 DOI: 10.7717/peerj.8797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/25/2020] [Indexed: 01/24/2023] Open
Abstract
Background The "dark matter" of the genome harbors several non-coding RNA species including Long non-coding RNAs (lncRNAs), which have been implicated in neoplasia but remain understudied. RNA-seq has provided deep insights into the nature of lncRNAs in cancer but current RNA-seq data are rarely accompanied by longitudinal patient survival information. In contrast, a plethora of microarray studies have collected these clinical metadata that can be leveraged to identify novel associations between gene expression and clinical phenotypes. Methods In this study, we developed an analysis framework that computationally integrates RNA-seq and microarray data to systematically screen 9,463 lncRNAs for association with mortality risk across 20 cancer types. Results In total, we identified a comprehensive list of associations between lncRNAs and patient survival and demonstrate that these prognostic lncRNAs are under selective pressure and may be functional. Our results provide valuable insights that facilitate further exploration of lncRNAs and their potential as cancer biomarkers and drug targets.
Collapse
Affiliation(s)
- Matthew Ung
- Department of Molecular and Systems Biology, Dartmouth College, Hanover, NH, USA
| | - Evelien Schaafsma
- Department of Molecular and Systems Biology, Dartmouth College, Hanover, NH, USA
| | - Daniel Mattox
- Department of Computer Science, Dartmouth College, Hanover, NH, USA
| | - George L Wang
- Department of Molecular and Systems Biology, Dartmouth College, Hanover, NH, USA
| | - Chao Cheng
- Department of Molecular and Systems Biology, Dartmouth College, Hanover, NH, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA.,Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
178
|
Fu Y, Wu Z, Guo Z, Chen L, Ma Y, Wang Z, Xiao W, Wang Y. Systems-level analysis identifies key regulators driving epileptogenesis in temporal lobe epilepsy. Genomics 2020; 112:1768-1780. [DOI: 10.1016/j.ygeno.2019.09.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/31/2019] [Accepted: 09/25/2019] [Indexed: 01/05/2023]
|
179
|
Evaluating genetic causes of azoospermia: What can we learn from a complex cellular structure and single-cell transcriptomics of the human testis? Hum Genet 2020; 140:183-201. [PMID: 31950241 DOI: 10.1007/s00439-020-02116-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/06/2020] [Indexed: 12/13/2022]
Abstract
Azoospermia is a condition defined as the absence of spermatozoa in the ejaculate, but the testicular phenotype of men with azoospermia may be very variable, ranging from full spermatogenesis, through arrested maturation of germ cells at different stages, to completely degenerated tissue with ghost tubules. Hence, information regarding the cell-type-specific expression patterns is needed to prioritise potential pathogenic variants that contribute to the pathogenesis of azoospermia. Thanks to technological advances within next-generation sequencing, it is now possible to obtain detailed cell-type-specific expression patterns in the testis by single-cell RNA sequencing. However, to interpret single-cell RNA sequencing data properly, substantial knowledge of the highly sophisticated data processing and visualisation methods is needed. Here we review the complex cellular structure of the human testis in different types of azoospermia and outline how known genetic alterations affect the pathology of the testis. We combined the currently available single-cell RNA sequencing datasets originating from the human testis into one dataset covering 62,751 testicular cells, each with a median of 2637 transcripts quantified. We show what effects the most common data-processing steps have, and how different visualisation methods can be used. Furthermore, we calculated expression patterns in pseudotime, and show how splicing rates can be used to determine the velocity of differentiation during spermatogenesis. With the combined dataset we show expression patterns and network analysis of genes known to be involved in the pathogenesis of azoospermia. Finally, we provide the combined dataset as an interactive online resource where expression of genes and different visualisation methods can be explored ( https://testis.cells.ucsc.edu/ ).
Collapse
|
180
|
Middelhoff M, Nienhüser H, Valenti G, Maurer HC, Hayakawa Y, Takahashi R, Kim W, Jiang Z, Malagola E, Cuti K, Tailor Y, Zamechek LB, Renz BW, Quante M, Yan KS, Wang TC. Prox1-positive cells monitor and sustain the murine intestinal epithelial cholinergic niche. Nat Commun 2020; 11:111. [PMID: 31913277 PMCID: PMC6949263 DOI: 10.1038/s41467-019-13850-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 11/26/2019] [Indexed: 02/08/2023] Open
Abstract
The enteric neurotransmitter acetylcholine governs important intestinal epithelial secretory and immune functions through its actions on epithelial muscarinic Gq-coupled receptors such as M3R. Its role in the regulation of intestinal stem cell function and differentiation, however, has not been clarified. Here, we find that nonselective muscarinic receptor antagonism in mice as well as epithelial-specific ablation of M3R induces a selective expansion of DCLK1-positive tuft cells, suggesting a model of feedback inhibition. Cholinergic blockade reduces Lgr5-positive intestinal stem cell tracing and cell number. In contrast, Prox1-positive endocrine cells appear as primary sensors of cholinergic blockade inducing the expansion of tuft cells, which adopt an enteroendocrine phenotype and contribute to increased mucosal levels of acetylcholine. This compensatory mechanism is lost with acute irradiation injury, resulting in a paucity of tuft cells and acetylcholine production. Thus, enteroendocrine tuft cells appear essential to maintain epithelial homeostasis following modifications of the cholinergic intestinal niche.
Collapse
Affiliation(s)
- Moritz Middelhoff
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Henrik Nienhüser
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Giovanni Valenti
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - H Carlo Maurer
- Klinikum rechts der Isar, II. Medizinische Klinik, Technische Universität München, 81675, Munich, Germany
| | - Yoku Hayakawa
- Graduate School of Medicine, Department of Gastroenterology, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Ryota Takahashi
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Woosook Kim
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Zhengyu Jiang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Krystle Cuti
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Yagnesh Tailor
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Leah B Zamechek
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Bernhard W Renz
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Ludwig-Maximilians-Universität München, 81377, Munich, Germany
| | - Michael Quante
- Klinikum rechts der Isar, II. Medizinische Klinik, Technische Universität München, 81675, Munich, Germany
| | - Kelley S Yan
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
181
|
Poos AM, Kordaß T, Kolte A, Ast V, Oswald M, Rippe K, König R. Modelling TERT regulation across 19 different cancer types based on the MIPRIP 2.0 gene regulatory network approach. BMC Bioinformatics 2019; 20:737. [PMID: 31888467 PMCID: PMC6937852 DOI: 10.1186/s12859-019-3323-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 12/16/2019] [Indexed: 01/15/2023] Open
Abstract
Background Reactivation of the telomerase reverse transcriptase gene TERT is a central feature for unlimited proliferation of the majority of cancers. However, the underlying regulatory processes are only partly understood. Results We assembled regulator binding information from serveral sources to construct a generic human and mouse gene regulatory network. Advancing our “Mixed Integer linear Programming based Regulatory Interaction Predictor” (MIPRIP) approach, we identified the most common and cancer-type specific regulators of TERT across 19 different human cancers. The results were validated by using the well-known TERT regulation by the ETS1 transcription factor in a subset of melanomas with mutations in the TERT promoter. Our improved MIPRIP2 R-package and the associated generic regulatory networks are freely available at https://github.com/KoenigLabNM/MIPRIP. Conclusion MIPRIP 2.0 identified common as well as tumor type specific regulators of TERT. The software can be easily applied to transcriptome datasets to predict gene regulation for any gene and disease/condition under investigation.
Collapse
Affiliation(s)
- Alexandra M Poos
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany.,Division of Chromatin Networks, German Cancer Research Center (DKFZ) and Bioquant, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Theresa Kordaß
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Amol Kolte
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Volker Ast
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Marcus Oswald
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Karsten Rippe
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and Bioquant, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
| | - Rainer König
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany.
| |
Collapse
|
182
|
A genetically defined disease model reveals that urothelial cells can initiate divergent bladder cancer phenotypes. Proc Natl Acad Sci U S A 2019; 117:563-572. [PMID: 31871155 PMCID: PMC6955337 DOI: 10.1073/pnas.1915770117] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Small cell carcinoma of the bladder (SCCB) is a lethal variant of bladder cancer with no effective treatment. A lack of available preclinical models and clinical cohorts impedes our understanding of its molecular pathogenesis. In this study, we provided a tumor model as functional evidence showing that SCCB and other bladder cancer phenotypes can be derived from normal human urothelial cells. We further demonstrated that SCCB has a distinct transcriptome and identified SCCB-associated cell surface proteins (CSPs) that can be further evaluated as potential therapeutic targets. We show that our model shares CSP profile with clinical SCCB samples. Our findings create a foundation to understand the molecular underpinnings of SCCB and provide tools for developing therapeutic strategies. Small cell carcinoma of the bladder (SCCB) is a rare and lethal phenotype of bladder cancer. The pathogenesis and molecular features are unknown. Here, we established a genetically engineered SCCB model and a cohort of patient SCCB and urothelial carcinoma samples to characterize molecular similarities and differences between bladder cancer phenotypes. We demonstrate that SCCB shares a urothelial origin with other bladder cancer phenotypes by showing that urothelial cells driven by a set of defined oncogenic factors give rise to a mixture of tumor phenotypes, including small cell carcinoma, urothelial carcinoma, and squamous cell carcinoma. Tumor-derived single-cell clones also give rise to both SCCB and urothelial carcinoma in xenografts. Despite this shared urothelial origin, clinical SCCB samples have a distinct transcriptional profile and a unique transcriptional regulatory network. Using the transcriptional profile from our cohort, we identified cell surface proteins (CSPs) associated with the SCCB phenotype. We found that the majority of SCCB samples have PD-L1 expression in both tumor cells and tumor-infiltrating lymphocytes, suggesting that immune checkpoint inhibitors could be a treatment option for SCCB. We further demonstrate that our genetically engineered tumor model is a representative tool for investigating CSPs in SCCB by showing that it shares a similar a CSP profile with clinical samples and expresses SCCB–up-regulated CSPs at both the mRNA and protein levels. Our findings reveal distinct molecular features of SCCB and provide a transcriptional dataset and a preclinical model for further investigating SCCB biology.
Collapse
|
183
|
Lopez CK, Noguera E, Stavropoulou V, Robert E, Aid Z, Ballerini P, Bilhou-Nabera C, Lapillonne H, Boudia F, Thirant C, Fagnan A, Arcangeli ML, Kinston SJ, Diop M, Job B, Lecluse Y, Brunet E, Babin L, Villeval JL, Delabesse E, Peters AHFM, Vainchenker W, Gaudry M, Masetti R, Locatelli F, Malinge S, Nerlov C, Droin N, Lobry C, Godin I, Bernard OA, Göttgens B, Petit A, Pflumio F, Schwaller J, Mercher T. Ontogenic Changes in Hematopoietic Hierarchy Determine Pediatric Specificity and Disease Phenotype in Fusion Oncogene-Driven Myeloid Leukemia. Cancer Discov 2019; 9:1736-1753. [PMID: 31662298 DOI: 10.1158/2159-8290.cd-18-1463] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 08/05/2019] [Accepted: 09/23/2019] [Indexed: 01/18/2023]
Abstract
Fusion oncogenes are prevalent in several pediatric cancers, yet little is known about the specific associations between age and phenotype. We observed that fusion oncogenes, such as ETO2-GLIS2, are associated with acute megakaryoblastic or other myeloid leukemia subtypes in an age-dependent manner. Analysis of a novel inducible transgenic mouse model showed that ETO2-GLIS2 expression in fetal hematopoietic stem cells induced rapid megakaryoblastic leukemia whereas expression in adult bone marrow hematopoietic stem cells resulted in a shift toward myeloid transformation with a strikingly delayed in vivo leukemogenic potential. Chromatin accessibility and single-cell transcriptome analyses indicate ontogeny-dependent intrinsic and ETO2-GLIS2-induced differences in the activities of key transcription factors, including ERG, SPI1, GATA1, and CEBPA. Importantly, switching off the fusion oncogene restored terminal differentiation of the leukemic blasts. Together, these data show that aggressiveness and phenotypes in pediatric acute myeloid leukemia result from an ontogeny-related differential susceptibility to transformation by fusion oncogenes. SIGNIFICANCE: This work demonstrates that the clinical phenotype of pediatric acute myeloid leukemia is determined by ontogeny-dependent susceptibility for transformation by oncogenic fusion genes. The phenotype is maintained by potentially reversible alteration of key transcription factors, indicating that targeting of the fusions may overcome the differentiation blockage and revert the leukemic state.See related commentary by Cruz Hernandez and Vyas, p. 1653.This article is highlighted in the In This Issue feature, p. 1631.
Collapse
Affiliation(s)
- Cécile K Lopez
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Esteve Noguera
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Vaia Stavropoulou
- University Children's Hospital Beider Basel (UKBB) and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Elie Robert
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Zakia Aid
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | | | | | | | - Fabien Boudia
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
- Université Paris Diderot, Paris, France
| | - Cécile Thirant
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Alexandre Fagnan
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
- Université Paris Diderot, Paris, France
| | - Marie-Laure Arcangeli
- Unité Mixte de Recherche 967 INSERM, CEA/DRF/IBFJ/IRCM/LSHL, Université Paris-Diderot-Université Paris-Sud, Equipe labellisée Association Recherche Contre le Cancer, Fontenay-aux-roses, France
| | - Sarah J Kinston
- Wellcome and MRC Cambridge Stem Cell Institute and the Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | - Erika Brunet
- Genome Dynamics in the Immune System Laboratory, Institut Imagine, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Loélia Babin
- Genome Dynamics in the Immune System Laboratory, Institut Imagine, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Jean Luc Villeval
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Eric Delabesse
- INSERM U1037, Team 16, Center of Research of Cancerology of Toulouse, Hematology Laboratory, IUCT-Oncopole, France
| | - Antoine H F M Peters
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- Faculty of Sciences, University of Basel, Basel, Switzerland
| | - William Vainchenker
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Muriel Gaudry
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Riccardo Masetti
- Department of Pediatrics, "Lalla Seràgnoli," Hematology-Oncology Unit, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Franco Locatelli
- Department of Pediatrics, Sapienza, University of Rome, Rome, Italy
- Hematology-Oncology-IRCCS Ospedale Bambino Gesù, Rome, Italy
| | - Sébastien Malinge
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Claus Nerlov
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | | | - Isabelle Godin
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Olivier A Bernard
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Berthold Göttgens
- Wellcome and MRC Cambridge Stem Cell Institute and the Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | | - Françoise Pflumio
- Unité Mixte de Recherche 967 INSERM, CEA/DRF/IBFJ/IRCM/LSHL, Université Paris-Diderot-Université Paris-Sud, Equipe labellisée Association Recherche Contre le Cancer, Fontenay-aux-roses, France
| | - Juerg Schwaller
- University Children's Hospital Beider Basel (UKBB) and Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Thomas Mercher
- INSERM U1170, Gustave Roussy, Villejuif, France.
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
- Université Paris Diderot, Paris, France
| |
Collapse
|
184
|
Chromatin regulators mediate anthracycline sensitivity in breast cancer. Nat Med 2019; 25:1721-1727. [PMID: 31700186 PMCID: PMC7220800 DOI: 10.1038/s41591-019-0638-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 09/12/2019] [Indexed: 12/26/2022]
Abstract
Anthracyclines are a highly effective component of curative breast cancer chemotherapy, but are associated with significant morbidity1,2. Since anthracyclines work in part via inhibition of topoisomerase-II (TOP2) on accessible DNA3,4, we hypothesized that chromatin regulatory genes (CRGs) that mediate DNA accessibility might predict anthracycline response. We elucidate the role of CRGs in anthracycline sensitivity in breast cancer through integrative analysis of patient and cell line data. We identify a consensus set of 38 CRGs associated with anthracycline response across ten cell line datasets. Evaluating the interaction between expression and treatment in predicting survival in a metacohort of 1006 early-stage breast cancer patients, we identify 54 CRGs whose expression levels dictate anthracycline benefit across the clinical subgroups, 12 of which overlapped with those identified in vitro. CRGs that promote DNA accessibility, including Trithorax complex members, were associated with anthracycline sensitivity when highly expressed, whereas CRGs that reduce accessibility such as Polycomb complex proteins, were associated with decreased anthracycline sensitivity. We show that KDM4B modulates TOP2 accessibility to chromatin, elucidating a mechanism of TOP2 inhibitor sensitivity. These findings indicate that CRGs mediate anthracycline benefit by modulating DNA accessibility with implications for breast cancer patient stratification and treatment decision making.
Collapse
|
185
|
Mercatelli D, Scalambra L, Triboli L, Ray F, Giorgi FM. Gene regulatory network inference resources: A practical overview. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1863:194430. [PMID: 31678629 DOI: 10.1016/j.bbagrm.2019.194430] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023]
Abstract
Transcriptional regulation is a fundamental molecular mechanism involved in almost every aspect of life, from homeostasis to development, from metabolism to behavior, from reaction to stimuli to disease progression. In recent years, the concept of Gene Regulatory Networks (GRNs) has grown popular as an effective applied biology approach for describing the complex and highly dynamic set of transcriptional interactions, due to its easy-to-interpret features. Since cataloguing, predicting and understanding every GRN connection in all species and cellular contexts remains a great challenge for biology, researchers have developed numerous tools and methods to infer regulatory processes. In this review, we catalogue these methods in six major areas, based on the dominant underlying information leveraged to infer GRNs: Coexpression, Sequence Motifs, Chromatin Immunoprecipitation (ChIP), Orthology, Literature and Protein-Protein Interaction (PPI) specifically focused on transcriptional complexes. The methods described here cover a wide range of user-friendliness: from web tools that require no prior computational expertise to command line programs and algorithms for large scale GRN inferences. Each method for GRN inference described herein effectively illustrates a type of transcriptional relationship, with many methods being complementary to others. While a truly holistic approach for inferring and displaying GRNs remains one of the greatest challenges in the field of systems biology, we believe that the integration of multiple methods described herein provides an effective means with which experimental and computational biologists alike may obtain the most complete pictures of transcriptional relationships. This article is part of a Special Issue entitled: Transcriptional Profiles and Regulatory Gene Networks edited by Dr. Federico Manuel Giorgi and Dr. Shaun Mahony.
Collapse
Affiliation(s)
- Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Laura Scalambra
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Luca Triboli
- Centre for Integrative Biology (CIBIO), University of Trento, Italy
| | - Forest Ray
- Department of Systems Biology, Columbia University Medical Center, New York, NY, United States
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.
| |
Collapse
|
186
|
Schubert M, Colomé-Tatché M, Foijer F. Gene networks in cancer are biased by aneuploidies and sample impurities. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1863:194444. [PMID: 31654805 DOI: 10.1016/j.bbagrm.2019.194444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/05/2019] [Accepted: 10/14/2019] [Indexed: 12/14/2022]
Abstract
Gene regulatory network inference is a standard technique for obtaining structured regulatory information from, for instance, gene expression measurements. Methods performing this task have been extensively evaluated on synthetic, and to a lesser extent real data sets. In contrast to these test evaluations, applications to gene expression data of human cancers are often limited by fewer samples and more potential regulatory links, and are biased by copy number aberrations as well as cell mixtures and sample impurities. Here, we take networks inferred from TCGA cohorts as an example to show that (1) transcription factor annotations are essential to obtain reliable networks, and (2) even for state of the art methods, we expect that between 20 and 80% of edges are caused by copy number changes and cell mixtures rather than transcription factor regulation.
Collapse
Affiliation(s)
- Michael Schubert
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, the Netherlands; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany.
| | - Maria Colomé-Tatché
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, the Netherlands; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, the Netherlands.
| |
Collapse
|
187
|
Tan QW, Mutwil M. Inferring biosynthetic and gene regulatory networks from Artemisia annua RNA sequencing data on a credit card-sized ARM computer. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1863:194429. [PMID: 31634636 DOI: 10.1016/j.bbagrm.2019.194429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 02/05/2023]
Abstract
Prediction of gene function and gene regulatory networks is one of the most active topics in bioinformatics. The accumulation of publicly available gene expression data for hundreds of plant species, together with advances in bioinformatical methods and affordable computing, sets ingenuity as one of the major bottlenecks in understanding gene function and regulation. Here, we show how a credit card-sized computer retailing for <50 USD can be used to rapidly predict gene function and infer regulatory networks from RNA sequencing data. To achieve this, we constructed a bioinformatical pipeline that downloads and allows quality-control of RNA sequencing data; and generates a gene co-expression network that can reveal enzymes and transcription factors participating and controlling a given biosynthetic pathway. We exemplify this by first identifying genes and transcription factors involved in the biosynthesis of secondary cell wall in the plant Artemisia annua, the main natural source of the anti-malarial drug artemisinin. Networks were then used to dissect the artemisinin biosynthesis pathway, which suggest potential transcription factors regulating artemisinin biosynthesis. We provide the source code of our pipeline (https://github.com/mutwil/LSTrAP-Lite) and envision that the ubiquity of affordable computing, availability of biological data and increased bioinformatical training of biologists will transform the field of bioinformatics. This article is part of a Special Issue entitled: Transcriptional Profiles and Regulatory Gene Networks edited by Dr. Dr. Federico Manuel Giorgi and Dr. Shaun Mahony.
Collapse
Affiliation(s)
- Qiao Wen Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Marek Mutwil
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
188
|
Circulatory factors associated with function and prognosis in patients with severe heart failure. Clin Res Cardiol 2019; 109:655-672. [PMID: 31562542 PMCID: PMC7239817 DOI: 10.1007/s00392-019-01554-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 09/13/2019] [Indexed: 02/07/2023]
Abstract
Background Multiple circulatory factors are increased in heart failure (HF). Many have been linked to cardiac and/or skeletal muscle tissue processes, which in turn might influence physical activity and/or capacity during HF. This study aimed to provide a better understanding of the mechanisms linking HF with the loss of peripheral function. Methods and results Physical capacity measured by maximum oxygen uptake, myocardial function (measured by echocardiography), physical activity (measured by accelerometry), and mortality data was collected for patients with severe symptomatic heart failure an ejection fraction < 35% (n = 66) and controls (n = 28). Plasma circulatory factors were quantified using a multiplex immunoassay. Multivariate (orthogonal projections to latent structures discriminant analysis) and univariate analyses identified many factors that differed significantly between HF and control subjects, mainly involving biological functions related to cell growth and cell adhesion, extracellular matrix organization, angiogenesis, and inflammation. Then, using principal component analysis, links between circulatory factors and physical capacity, daily physical activity, and myocardial function were identified. A subset of ten biomarkers differentially expressed in patients with HF vs controls covaried with physical capacity, daily physical activity, and myocardial function; eight of these also carried prognostic value. These included established plasma biomarkers of HF, such as NT-proBNP and ST2 along with recently identified factors such as GDF15, IGFBP7, and TfR, as well as a new factor, galectin-4. Conclusions These findings reinforce the importance of systemic circulatory factors linked to hemodynamic stress responses and inflammation in the pathogenesis and progress of HF disease. They also support established biomarkers for HF and suggest new plausible markers. Graphic abstract ![]()
Electronic supplementary material The online version of this article (10.1007/s00392-019-01554-3) contains supplementary material, which is available to authorized users.
Collapse
|
189
|
Ahsen ME, Chun Y, Grishin A, Grishina G, Stolovitzky G, Pandey G, Bunyavanich S. NeTFactor, a framework for identifying transcriptional regulators of gene expression-based biomarkers. Sci Rep 2019; 9:12970. [PMID: 31506535 PMCID: PMC6737052 DOI: 10.1038/s41598-019-49498-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 08/27/2019] [Indexed: 12/21/2022] Open
Abstract
Biological and regulatory mechanisms underlying many multi-gene expression-based disease biomarkers are often not readily evident. We describe an innovative framework, NeTFactor, that combines network analyses with gene expression data to identify transcription factors (TFs) that significantly and maximally regulate such a biomarker. NeTFactor uses a computationally-inferred context-specific gene regulatory network and applies topological, statistical, and optimization methods to identify regulator TFs. Application of NeTFactor to a multi-gene expression-based asthma biomarker identified ETS translocation variant 4 (ETV4) and peroxisome proliferator-activated receptor gamma (PPARG) as the biomarker's most significant TF regulators. siRNA-based knock down of these TFs in an airway epithelial cell line model demonstrated significant reduction of cytokine expression relevant to asthma, validating NeTFactor's top-scoring findings. While PPARG has been associated with airway inflammation, ETV4 has not yet been implicated in asthma, thus indicating the possibility of novel, disease-relevant discovery by NeTFactor. We also show that NeTFactor's results are robust when the gene regulatory network and biomarker are derived from independent data. Additionally, our application of NeTFactor to a different disease biomarker identified TF regulators of interest. These results illustrate that the application of NeTFactor to multi-gene expression-based biomarkers could yield valuable insights into regulatory mechanisms and biological processes underlying disease.
Collapse
Affiliation(s)
- Mehmet Eren Ahsen
- Icahn Institute for Genomics and Multiscale Biology and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yoojin Chun
- Icahn Institute for Genomics and Multiscale Biology and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Grishin
- Division of Allergy & Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Galina Grishina
- Division of Allergy & Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gustavo Stolovitzky
- Icahn Institute for Genomics and Multiscale Biology and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- IBM T.J. Watson Research Center, Yorktown Heights, New York, NY, USA
| | - Gaurav Pandey
- Icahn Institute for Genomics and Multiscale Biology and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Supinda Bunyavanich
- Icahn Institute for Genomics and Multiscale Biology and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Division of Allergy & Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
190
|
Zhang W, Zhang F, Zhang J, Wang N. Hierarchical parameter estimation of GRN based on topological analysis. IET Syst Biol 2019; 12:294-303. [PMID: 30472694 DOI: 10.1049/iet-syb.2018.5015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Reverse engineering of gene regulatory network (GRN) is an important and challenging task in systems biology. Existing parameter estimation approaches that compute model parameters with the same importance are usually computationally expensive or infeasible, especially in dealing with complex biological networks.In order to improve the efficiency of computational modeling, the paper applies a hierarchical estimation methodology in computational modeling of GRN based on topological analysis. This paper divides nodes in a network into various priority levels using the graph-based measure and genetic algorithm. The nodes in the first level, that correspond to root strongly connected components(SCC) in the digraph of GRN, are given top priority in parameter estimation. The estimated parameters of vertices in the previous priority level ARE used to infer the parameters for nodes in the next priority level. The proposed hierarchical estimation methodology obtains lower error indexes while consuming less computational resources compared with single estimation methodology. Experimental outcomes with insilico networks and a realistic network show that gene networks are decomposed into no more than four levels, which is consistent with the properties of inherent modularity for GRN. In addition, the proposed hierarchical parameter estimation achieves a balance between computational efficiency and accuracy.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Control Science and Engineering, Zhejiang University, Zheda Road 38, Hangzhou, People's Republic of China
| | - Feng Zhang
- Department of Control Science and Engineering, Zhejiang University, Zheda Road 38, Hangzhou, People's Republic of China
| | - Jianming Zhang
- Department of Control Science and Engineering, Zhejiang University, Zheda Road 38, Hangzhou, People's Republic of China.
| | - Ning Wang
- Department of Control Science and Engineering, Zhejiang University, Zheda Road 38, Hangzhou, People's Republic of China
| |
Collapse
|
191
|
Non-coding RNA regulatory networks. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1863:194417. [PMID: 31493559 DOI: 10.1016/j.bbagrm.2019.194417] [Citation(s) in RCA: 308] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/13/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023]
Abstract
It is well established that the vast majority of human RNA transcripts do not encode for proteins and that non-coding RNAs regulate cell physiology and shape cellular functions. A subset of them is involved in gene regulation at different levels, from epigenetic gene silencing to post-transcriptional regulation of mRNA stability. Notably, the aberrant expression of many non-coding RNAs has been associated with aggressive pathologies. Rapid advances in network biology indicates that the robustness of cellular processes is the result of specific properties of biological networks such as scale-free degree distribution and hierarchical modularity, suggesting that regulatory network analyses could provide new insights on gene regulation and dysfunction mechanisms. In this study we present an overview of public repositories where non-coding RNA-regulatory interactions are collected and annotated, we discuss unresolved questions for data integration and we recall existing resources to build and analyse networks.
Collapse
|
192
|
Laaniste L, Srivastava PK, Stylianou J, Syed N, Cases-Cunillera S, Shkura K, Zeng Q, Rackham OJL, Langley SR, Delahaye-Duriez A, O'Neill K, Williams M, Becker A, Roncaroli F, Petretto E, Johnson MR. Integrated systems-genetic analyses reveal a network target for delaying glioma progression. Ann Clin Transl Neurol 2019; 6:1616-1638. [PMID: 31420939 PMCID: PMC6764637 DOI: 10.1002/acn3.50850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/12/2022] Open
Abstract
Objective To identify a convergent, multitarget proliferation characteristic for astrocytoma transformation that could be targeted for therapy discovery. Methods Using an integrated functional genomics approach, we prioritized networks associated with astrocytoma progression using the following criteria: differential co‐expression between grade II and grade III IDH1‐mutated and 1p/19q euploid astrocytomas, preferential enrichment for genetic risk to cancer, association with patient survival and sample‐level genomic features. Drugs targeting the identified multitarget network characteristic for astrocytoma transformation were computationally predicted using drug transcriptional perturbation data and validated using primary human astrocytoma cells. Results A single network, M2, consisting of 177 genes, was associated with glioma progression on the basis of the above criteria. Functionally, M2 encoded physically interacting proteins regulating cell cycle processes and analysis of genome‐wide gene‐regulatory interactions using mutual information and DNA–protein interactions revealed the known regulators of cell cycle processes FoxM1, B‐Myb, and E2F2 as key regulators of M2. These results suggest functional disruption of M2 via gene mutation or altered expression as a convergent pathway regulating astrocytoma transformation. By considering M2 as a multitarget drug target regulating astrocytoma transformation, we identified several drugs that are predicted to restore M2 expression in anaplastic astrocytoma toward its low‐grade profile and of these, we validated the known antiproliferative drug resveratrol as down‐regulating multiple nodes of M2 including at nanomolar concentrations achievable in human cerebrospinal fluid by oral dosing. Interpretation Our results identify M2 as a multitarget network characteristic for astrocytoma progression and encourage M2‐based drug screening to identify new compounds for preventing glioma transformation.
Collapse
Affiliation(s)
- Liisi Laaniste
- Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK
| | | | - Julianna Stylianou
- John Fulcher Neuro-oncology Laboratory, Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK
| | - Nelofer Syed
- John Fulcher Neuro-oncology Laboratory, Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK
| | | | - Kirill Shkura
- Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK
| | - Qingyu Zeng
- John Fulcher Neuro-oncology Laboratory, Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK
| | | | - Sarah R Langley
- Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK.,Duke-NUS Medical School, Singapore
| | - Andree Delahaye-Duriez
- Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK.,PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, France
| | - Kevin O'Neill
- Department of Neurosurgery, Imperial College Healthcare NHS Trust, London, UK
| | - Matthew Williams
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Albert Becker
- Department of Neuropathology, University of Bonn Medical Centre, Bonn, Germany
| | - Federico Roncaroli
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Enrico Petretto
- Duke-NUS Medical School, Singapore.,MRC London Institute of Medical Sciences (LMS), Imperial College London, London, UK
| | - Michael R Johnson
- Division of Brain Sciences, Imperial College Faculty of Medicine, London, UK
| |
Collapse
|
193
|
Ru B, Sun J, Tong Y, Wong CN, Chandra A, Tang ATS, Chow LKY, Wun WL, Levitskaya Z, Zhang J. CR2Cancer: a database for chromatin regulators in human cancer. Nucleic Acids Res 2019; 46:D918-D924. [PMID: 29036683 PMCID: PMC5753221 DOI: 10.1093/nar/gkx877] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 09/28/2017] [Indexed: 12/19/2022] Open
Abstract
Chromatin regulators (CRs) can dynamically modulate chromatin architecture to epigenetically regulate gene expression in response to intrinsic and extrinsic signalling cues. Somatic alterations or misexpression of CRs might reprogram the epigenomic landscape of chromatin, which in turn lead to a wide range of common diseases, notably cancer. Here, we present CR2Cancer, a comprehensive annotation and visualization database for CRs in human cancer constructed by high throughput data analysis and literature mining. We collected and integrated genomic, transcriptomic, proteomic, clinical and functional information for over 400 CRs across multiple cancer types. We also built diverse types of CR-associated relations, including cancer type dependent (CR-target and miRNA-CR) and independent (protein-protein interaction and drug-target) ones. Furthermore, we manually curated around 6000 items of aberrant molecular alterations and interactions of CRs in cancer development from 5007 publications. CR2Cancer provides a user-friendly web interface to conveniently browse, search and download data of interest. We believe that this database would become a valuable resource for cancer epigenetics investigation and potential clinical application. CR2Cancer is freely available at http://cis.hku.hk/CR2Cancer.
Collapse
Affiliation(s)
- Beibei Ru
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Jianlong Sun
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Yin Tong
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Ching Ngar Wong
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Aditi Chandra
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Acacia Tsz So Tang
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Larry Ka Yue Chow
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Wai Lam Wun
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Zarina Levitskaya
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Jiangwen Zhang
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
194
|
Mercatelli D, Ray F, Giorgi FM. Pan-Cancer and Single-Cell Modeling of Genomic Alterations Through Gene Expression. Front Genet 2019; 10:671. [PMID: 31379928 PMCID: PMC6657420 DOI: 10.3389/fgene.2019.00671] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/27/2019] [Indexed: 12/27/2022] Open
Abstract
Cancer is a disease often characterized by the presence of multiple genomic alterations, which trigger altered transcriptional patterns and gene expression, which in turn sustain the processes of tumorigenesis, tumor progression, and tumor maintenance. The links between genomic alterations and gene expression profiles can be utilized as the basis to build specific molecular tumorigenic relationships. In this study, we perform pan-cancer predictions of the presence of single somatic mutations and copy number variations using machine learning approaches on gene expression profiles. We show that gene expression can be used to predict genomic alterations in every tumor type, where some alterations are more predictable than others. We propose gene aggregation as a tool to improve the accuracy of alteration prediction models from gene expression profiles. Ultimately, we show how this principle can be beneficial in intrinsically noisy datasets, such as those based on single-cell sequencing.
Collapse
Affiliation(s)
- Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Forest Ray
- Department of Systems Biology, Columbia University Medical Center, New York, NY, United States
| | - Federico M. Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
195
|
Balanis NG, Sheu KM, Esedebe FN, Patel SJ, Smith BA, Park JW, Alhani S, Gomperts BN, Huang J, Witte ON, Graeber TG. Pan-cancer Convergence to a Small-Cell Neuroendocrine Phenotype that Shares Susceptibilities with Hematological Malignancies. Cancer Cell 2019; 36:17-34.e7. [PMID: 31287989 PMCID: PMC6703903 DOI: 10.1016/j.ccell.2019.06.005] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/04/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023]
Abstract
Small-cell neuroendocrine cancers (SCNCs) are an aggressive cancer subtype. Transdifferentiation toward an SCN phenotype has been reported as a resistance route in response to targeted therapies. Here, we identified a convergence to an SCN state that is widespread across epithelial cancers and is associated with poor prognosis. More broadly, non-SCN metastases have higher expression of SCN-associated transcription factors than non-SCN primary tumors. Drug sensitivity and gene dependency screens demonstrate that these convergent SCNCs have shared vulnerabilities. These common vulnerabilities are found across unannotated SCN-like epithelial cases, small-round-blue cell tumors, and unexpectedly in hematological malignancies. The SCN convergent phenotype and common sensitivity profiles with hematological cancers can guide treatment options beyond tissue-specific targeted therapies.
Collapse
Affiliation(s)
- Nikolas G Balanis
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA
| | - Katherine M Sheu
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA
| | - Favour N Esedebe
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA
| | - Saahil J Patel
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA
| | - Bryan A Smith
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
| | - Jung Wook Park
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
| | - Salwan Alhani
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA
| | - Brigitte N Gomperts
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA; UCLA Children's Discovery and Innovation Institute, Mattel Children's Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC 27708, USA
| | - Owen N Witte
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA.
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA 90095, USA; California NanoSystems Institute, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
196
|
Zhernovkov V, Santra T, Cassidy H, Rukhlenko O, Matallanas D, Krstic A, Kolch W, Lobaskin V, Kholodenko BN. An integrative computational approach for a prioritization of key transcription regulators associated with nanomaterial-induced toxicity. Toxicol Sci 2019; 171:303-314. [PMID: 31271423 DOI: 10.1093/toxsci/kfz151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/19/2022] Open
Abstract
A rapid increase of new nanomaterial products poses new challenges for their risk assessment. Current traditional methods for estimating potential adverse health effect of nanomaterials (NMs) are complex, time consuming and expensive. In order to develop new prediction tests for nanotoxicity evaluation, a systems biology approach and data from high-throughput omics experiments can be used. We present a computational approach that combines reverse engineering techniques, network analysis and pathway enrichment analysis for inferring the transcriptional regulation landscape and its functional interpretation. To illustrate this approach, we used published transcriptomic data derived from mice lung tissue exposed to carbon nanotubes (NM-401 and NRCWE-26). Because fibrosis is the most common adverse effect of these NMs, we included in our analysis the data for bleomycin (BLM) treatment, which is a well-known fibrosis inducer. We inferred gene regulatory networks for each NM and BLM to capture functional hierarchical regulatory structures between genes and their regulators. Despite the different nature of the lung injury caused by nanoparticles and BLM, we identified several conserved core regulators for all agents. We reason that these regulators can be considered as early predictors of toxic responses after NMs exposure. This integrative approach, which refines traditional methods of transcriptomic analysis, can be useful for prioritization of potential core regulators and generation of new hypothesis about mechanisms of nanoparticles toxicity.
Collapse
Affiliation(s)
- Vadim Zhernovkov
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Tapesh Santra
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Hilary Cassidy
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Oleksii Rukhlenko
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Aleksandar Krstic
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Ireland
| | | | - Boris N Kholodenko
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.,School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Ireland.,Department of Pharmacology, Yale University School of Medicine, New Haven CT, USA
| |
Collapse
|
197
|
Keenan AB, Torre D, Lachmann A, Leong AK, Wojciechowicz ML, Utti V, Jagodnik KM, Kropiwnicki E, Wang Z, Ma’ayan A. ChEA3: transcription factor enrichment analysis by orthogonal omics integration. Nucleic Acids Res 2019; 47:W212-W224. [PMID: 31114921 PMCID: PMC6602523 DOI: 10.1093/nar/gkz446] [Citation(s) in RCA: 616] [Impact Index Per Article: 102.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 01/12/2023] Open
Abstract
Identifying the transcription factors (TFs) responsible for observed changes in gene expression is an important step in understanding gene regulatory networks. ChIP-X Enrichment Analysis 3 (ChEA3) is a transcription factor enrichment analysis tool that ranks TFs associated with user-submitted gene sets. The ChEA3 background database contains a collection of gene set libraries generated from multiple sources including TF-gene co-expression from RNA-seq studies, TF-target associations from ChIP-seq experiments, and TF-gene co-occurrence computed from crowd-submitted gene lists. Enrichment results from these distinct sources are integrated to generate a composite rank that improves the prediction of the correct upstream TF compared to ranks produced by individual libraries. We compare ChEA3 with existing TF prediction tools and show that ChEA3 performs better. By integrating the ChEA3 libraries, we illuminate general transcription factor properties such as whether the TF behaves as an activator or a repressor. The ChEA3 web-server is available from https://amp.pharm.mssm.edu/ChEA3.
Collapse
Affiliation(s)
- Alexandra B Keenan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Denis Torre
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Alexander Lachmann
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Ariel K Leong
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Megan L Wojciechowicz
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Vivian Utti
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Kathleen M Jagodnik
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Eryk Kropiwnicki
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Zichen Wang
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Avi Ma’ayan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| |
Collapse
|
198
|
Lu X, Tian J, Wen H, Jiang M, Liu W, Wu F, Yu L, Zhong S. Microcystin-LR-regulated transcriptome dynamics in ZFL cells. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 212:222-232. [PMID: 31136897 DOI: 10.1016/j.aquatox.2019.04.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 06/09/2023]
Abstract
Microcystin-LR (MC-LR) is a highly toxic hepatotoxin that poses great hazards to aquatic organisms and even human health. The zebrafish liver cell line (ZFL) is a valuable model for investigating toxicity and metabolism of toxicants. However, the toxicity of MC-LR and its effects on gene transcription of ZFL cells remains to be characterized. In this study, we determined the toxicity of MC-LR for ZFL cells and investigated the effects of MC-LR on cellular transcriptome dynamics. The EC50 of MC-LR for ZFL cells was 80.123 μg/mL. The ZFL cells were exposed to 10 μg/mL MC-LR for 0, 1, 3, 6, 12 or 24 h, and RNA-sequencing was performed to analyze gene transcription. A total of 10,209 genes were found to be regulated by MC-LR. The numbers of up- and down-regulated genes at different time points ranged from 2179 to 3202 and from 1501 to 2597, respectively. Furthermore, 1543 genes underwent differential splicing (AS) upon MC-LR exposure, of which 620 were not identified as differentially expressed gene (DEG). The effects of MC-LR on cellular functions were highly time-dependent. MAPK (mitogen-activated protein kinase) and FoxO (forkhead box O) signaling pathways were the most prominent pathways activated by MC-LR. Steroid biosynthesis and terpenoid backbone biosynthesis were the most enriched for the down-regulated genes. A gene regulatory network was constructed from the expression profile datasets and the candidate master transcription factors were identified. Our results shed light on the molecular mechanisms of MC-LR cellular toxicity and the transcriptome landscapes of ZFL cells upon MC-LR toxicity.
Collapse
Affiliation(s)
- Xing Lu
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China.
| | - Juan Tian
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China.
| | - Hua Wen
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China.
| | - Ming Jiang
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China.
| | - Wei Liu
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China.
| | - Fan Wu
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China.
| | - Lijuan Yu
- Key Laboratory of Freshwater Biodiversity Conservation and Utilization of Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, Hubei, China.
| | - Shan Zhong
- Department of Genetics, Wuhan University, Wuhan 430071, Hubei, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, Hubei, China; Hubei Province Key Laboratory of Allergy and Immunology, Wuhan 430071, Hubei, China.
| |
Collapse
|
199
|
Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, Teinor JA, Belleau P, Biffi G, Lucito MS, Sivajothi S, Armstrong TD, Engle DD, Yu KH, Hao Y, Wolfgang CL, Park Y, Preall J, Jaffee EM, Califano A, Robson P, Tuveson DA. Cross-Species Single-Cell Analysis of Pancreatic Ductal Adenocarcinoma Reveals Antigen-Presenting Cancer-Associated Fibroblasts. Cancer Discov 2019; 9:1102-1123. [PMID: 31197017 DOI: 10.1158/2159-8290.cd-19-0094] [Citation(s) in RCA: 1307] [Impact Index Per Article: 217.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/18/2019] [Accepted: 05/30/2019] [Indexed: 12/21/2022]
Abstract
Cancer-associated fibroblasts (CAF) are major players in the progression and drug resistance of pancreatic ductal adenocarcinoma (PDAC). CAFs constitute a diverse cell population consisting of several recently described subtypes, although the extent of CAF heterogeneity has remained undefined. Here we use single-cell RNA sequencing to thoroughly characterize the neoplastic and tumor microenvironment content of human and mouse PDAC tumors. We corroborate the presence of myofibroblastic CAFs and inflammatory CAFs and define their unique gene signatures in vivo. Moreover, we describe a new population of CAFs that express MHC class II and CD74, but do not express classic costimulatory molecules. We term this cell population "antigen-presenting CAFs" and find that they activate CD4+ T cells in an antigen-specific fashion in a model system, confirming their putative immune-modulatory capacity. Our cross-species analysis paves the way for investigating distinct functions of CAF subtypes in PDAC immunity and progression. SIGNIFICANCE: Appreciating the full spectrum of fibroblast heterogeneity in pancreatic ductal adenocarcinoma is crucial to developing therapies that specifically target tumor-promoting CAFs. This work identifies MHC class II-expressing CAFs with a capacity to present antigens to CD4+ T cells, and potentially to modulate the immune response in pancreatic tumors.See related commentary by Belle and DeNardo, p. 1001.This article is highlighted in the In This Issue feature, p. 983.
Collapse
Affiliation(s)
- Ela Elyada
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Mohan Bolisetty
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.,Bristol-Myers Squibb, Pennington, New Jersey
| | - Pasquale Laise
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York
| | - William F Flynn
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Elise T Courtois
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Richard A Burkhart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Jonathan A Teinor
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Pascal Belleau
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Giulia Biffi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | - Matthew S Lucito
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | | | - Todd D Armstrong
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Dannielle D Engle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York.,Salk institute for Biological Studies, La Jolla, California
| | - Kenneth H Yu
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuan Hao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Christopher L Wolfgang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| | | | - Elizabeth M Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Andrea Califano
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York.,J.P. Sulzberger Columbia Genome Center, Columbia University, New York, New York.,Department of Biomedical Informatics, Columbia University, New York, New York.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut. .,Department of Genetics and Genome Sciences, Institute for Systems Genomics, University of Connecticut, Farmington, Connecticut
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York. .,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York
| |
Collapse
|
200
|
Hawe JS, Theis FJ, Heinig M. Inferring Interaction Networks From Multi-Omics Data. Front Genet 2019; 10:535. [PMID: 31249591 PMCID: PMC6582773 DOI: 10.3389/fgene.2019.00535] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/16/2019] [Indexed: 01/24/2023] Open
Abstract
A major goal in systems biology is a comprehensive description of the entirety of all complex interactions between different types of biomolecules-also referred to as the interactome-and how these interactions give rise to higher, cellular and organism level functions or diseases. Numerous efforts have been undertaken to define such interactomes experimentally, for example yeast-two-hybrid based protein-protein interaction networks or ChIP-seq based protein-DNA interactions for individual proteins. To complement these direct measurements, genome-scale quantitative multi-omics data (transcriptomics, proteomics, metabolomics, etc.) enable researchers to predict novel functional interactions between molecular species. Moreover, these data allow to distinguish relevant functional from non-functional interactions in specific biological contexts. However, integration of multi-omics data is not straight forward due to their heterogeneity. Numerous methods for the inference of interaction networks from homogeneous functional data exist, but with the advent of large-scale paired multi-omics data a new class of methods for inferring comprehensive networks across different molecular species began to emerge. Here we review state-of-the-art techniques for inferring the topology of interaction networks from functional multi-omics data, encompassing graphical models with multiple node types and quantitative-trait-loci (QTL) based approaches. In addition, we will discuss Bayesian aspects of network inference, which allow for leveraging already established biological information such as known protein-protein or protein-DNA interactions, to guide the inference process.
Collapse
Affiliation(s)
- Johann S. Hawe
- Institute of Computational Biology, HelmholtzZentrum München, Munich, Germany
- Department of Informatics, Technische Universität München, Munich, Germany
| | - Fabian J. Theis
- Institute of Computational Biology, HelmholtzZentrum München, Munich, Germany
- Department of Mathematics, Technische Universität München, Munich, Germany
| | - Matthias Heinig
- Institute of Computational Biology, HelmholtzZentrum München, Munich, Germany
- Department of Informatics, Technische Universität München, Munich, Germany
| |
Collapse
|