1
|
Durinck S, Spellman PT, Birney E, Huber W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat Protoc 2009; 4:1184-91. [PMID: 19617889 PMCID: PMC3159387 DOI: 10.1038/nprot.2009.97] [Citation(s) in RCA: 2590] [Impact Index Per Article: 161.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genomic experiments produce multiple views of biological systems, among them are DNA sequence and copy number variation, and mRNA and protein abundance. Understanding these systems needs integrated bioinformatic analysis. Public databases such as Ensembl provide relationships and mappings between the relevant sets of probe and target molecules. However, the relationships can be biologically complex and the content of the databases is dynamic. We demonstrate how to use the computational environment R to integrate and jointly analyze experimental datasets, employing BioMart web services to provide the molecule mappings. We also discuss typical problems that are encountered in making gene-to-transcript-to-protein mappings. The approach provides a flexible, programmable and reproducible basis for state-of-the-art bioinformatic data integration.
Collapse
|
Research Support, N.I.H., Extramural |
16 |
2590 |
2
|
Battye TGG, Kontogiannis L, Johnson O, Powell HR, Leslie AGW. iMOSFLM: a new graphical interface for diffraction-image processing with MOSFLM. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2011; 67:271-81. [PMID: 21460445 PMCID: PMC3069742 DOI: 10.1107/s0907444910048675] [Citation(s) in RCA: 2495] [Impact Index Per Article: 178.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 11/22/2010] [Indexed: 11/12/2022]
Abstract
iMOSFLM is a graphical user interface to the diffraction data-integration program MOSFLM. It is designed to simplify data processing by dividing the process into a series of steps, which are normally carried out sequentially. Each step has its own display pane, allowing control over parameters that influence that step and providing graphical feedback to the user. Suitable values for integration parameters are set automatically, but additional menus provide a detailed level of control for experienced users. The image display and the interfaces to the different tasks (indexing, strategy calculation, cell refinement, integration and history) are described. The most important parameters for each step and the best way of assessing success or failure are discussed.
Collapse
|
research-article |
14 |
2495 |
3
|
The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 2014; 507:315-22. [PMID: 24476821 PMCID: PMC3962515 DOI: 10.1038/nature12965] [Citation(s) in RCA: 2307] [Impact Index Per Article: 209.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 12/19/2013] [Indexed: 12/11/2022]
Abstract
Urothelial carcinoma of the bladder is a common malignancy that causes approximately 150,000 deaths per year worldwide. So far, no molecularly targeted agents have been approved for treatment of the disease. As part of The Cancer Genome Atlas project, we report here an integrated analysis of 131 urothelial carcinomas to provide a comprehensive landscape of molecular alterations. There were statistically significant recurrent mutations in 32 genes, including multiple genes involved in cell-cycle regulation, chromatin regulation, and kinase signalling pathways, as well as 9 genes not previously reported as significantly mutated in any cancer. RNA sequencing revealed four expression subtypes, two of which (papillary-like and basal/squamous-like) were also evident in microRNA sequencing and protein data. Whole-genome and RNA sequencing identified recurrent in-frame activating FGFR3-TACC3 fusions and expression or integration of several viruses (including HPV16) that are associated with gene inactivation. Our analyses identified potential therapeutic targets in 69% of the tumours, including 42% with targets in the phosphatidylinositol-3-OH kinase/AKT/mTOR pathway and 45% with targets (including ERBB2) in the RTK/MAPK pathway. Chromatin regulatory genes were more frequently mutated in urothelial carcinoma than in any other common cancer studied so far, indicating the future possibility of targeted therapy for chromatin abnormalities.
Collapse
|
Research Support, N.I.H., Extramural |
11 |
2307 |
4
|
Moore JE, Purcaro MJ, Pratt HE, Epstein CB, Shoresh N, Adrian J, Kawli T, Davis CA, Dobin A, Kaul R, Halow J, Van Nostrand EL, Freese P, Gorkin DU, Shen Y, He Y, Mackiewicz M, Pauli-Behn F, Williams BA, Mortazavi A, Keller CA, Zhang XO, Elhajjajy SI, Huey J, Dickel DE, Snetkova V, Wei X, Wang X, Rivera-Mulia JC, Rozowsky J, Zhang J, Chhetri SB, Zhang J, Victorsen A, White KP, Visel A, Yeo GW, Burge CB, Lécuyer E, Gilbert DM, Dekker J, Rinn J, Mendenhall EM, Ecker JR, Kellis M, Klein RJ, Noble WS, Kundaje A, Guigó R, Farnham PJ, Cherry JM, Myers RM, Ren B, Graveley BR, Gerstein MB, Pennacchio LA, Snyder MP, Bernstein BE, Wold B, Hardison RC, Gingeras TR, Stamatoyannopoulos JA, Weng Z. Expanded encyclopaedias of DNA elements in the human and mouse genomes. Nature 2020; 583:699-710. [PMID: 32728249 PMCID: PMC7410828 DOI: 10.1038/s41586-020-2493-4] [Citation(s) in RCA: 1327] [Impact Index Per Article: 265.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
The human and mouse genomes contain instructions that specify RNAs and proteins and govern the timing, magnitude, and cellular context of their production. To better delineate these elements, phase III of the Encyclopedia of DNA Elements (ENCODE) Project has expanded analysis of the cell and tissue repertoires of RNA transcription, chromatin structure and modification, DNA methylation, chromatin looping, and occupancy by transcription factors and RNA-binding proteins. Here we summarize these efforts, which have produced 5,992 new experimental datasets, including systematic determinations across mouse fetal development. All data are available through the ENCODE data portal (https://www.encodeproject.org), including phase II ENCODE1 and Roadmap Epigenomics2 data. We have developed a registry of 926,535 human and 339,815 mouse candidate cis-regulatory elements, covering 7.9 and 3.4% of their respective genomes, by integrating selected datatypes associated with gene regulation, and constructed a web-based server (SCREEN; http://screen.encodeproject.org) to provide flexible, user-defined access to this resource. Collectively, the ENCODE data and registry provide an expansive resource for the scientific community to build a better understanding of the organization and function of the human and mouse genomes.
Collapse
|
Research Support, N.I.H., Extramural |
5 |
1327 |
5
|
Arkin AP, Cottingham RW, Henry CS, Harris NL, Stevens RL, Maslov S, Dehal P, Ware D, Perez F, Canon S, Sneddon MW, Henderson ML, Riehl WJ, Murphy-Olson D, Chan SY, Kamimura RT, Kumari S, Drake MM, Brettin TS, Glass EM, Chivian D, Gunter D, Weston DJ, Allen BH, Baumohl J, Best AA, Bowen B, Brenner SE, Bun CC, Chandonia JM, Chia JM, Colasanti R, Conrad N, Davis JJ, Davison BH, DeJongh M, Devoid S, Dietrich E, Dubchak I, Edirisinghe JN, Fang G, Faria JP, Frybarger PM, Gerlach W, Gerstein M, Greiner A, Gurtowski J, Haun HL, He F, Jain R, Joachimiak MP, Keegan KP, Kondo S, Kumar V, Land ML, Meyer F, Mills M, Novichkov PS, Oh T, Olsen GJ, Olson R, Parrello B, Pasternak S, Pearson E, Poon SS, Price GA, Ramakrishnan S, Ranjan P, Ronald PC, Schatz MC, Seaver SMD, Shukla M, Sutormin RA, Syed MH, Thomason J, Tintle NL, Wang D, Xia F, Yoo H, Yoo S, Yu D. KBase: The United States Department of Energy Systems Biology Knowledgebase. Nat Biotechnol 2018; 36:566-569. [PMID: 29979655 PMCID: PMC6870991 DOI: 10.1038/nbt.4163] [Citation(s) in RCA: 893] [Impact Index Per Article: 127.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
letter |
7 |
893 |
6
|
Welch JD, Kozareva V, Ferreira A, Vanderburg C, Martin C, Macosko EZ. Single-Cell Multi-omic Integration Compares and Contrasts Features of Brain Cell Identity. Cell 2019; 177:1873-1887.e17. [PMID: 31178122 PMCID: PMC6716797 DOI: 10.1016/j.cell.2019.05.006] [Citation(s) in RCA: 721] [Impact Index Per Article: 120.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/21/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023]
Abstract
Defining cell types requires integrating diverse single-cell measurements from multiple experiments and biological contexts. To flexibly model single-cell datasets, we developed LIGER, an algorithm that delineates shared and dataset-specific features of cell identity. We applied it to four diverse and challenging analyses of human and mouse brain cells. First, we defined region-specific and sexually dimorphic gene expression in the mouse bed nucleus of the stria terminalis. Second, we analyzed expression in the human substantia nigra, comparing cell states in specific donors and relating cell types to those in the mouse. Third, we integrated in situ and single-cell expression data to spatially locate fine subtypes of cells present in the mouse frontal cortex. Finally, we jointly defined mouse cortical cell types using single-cell RNA-seq and DNA methylation profiles, revealing putative mechanisms of cell-type-specific epigenomic regulation. Integrative analyses using LIGER promise to accelerate investigations of cell-type definition, gene regulation, and disease states.
Collapse
|
Comparative Study |
6 |
721 |
7
|
Heirendt L, Arreckx S, Pfau T, Mendoza SN, Richelle A, Heinken A, Haraldsdóttir HS, Wachowiak J, Keating SM, Vlasov V, Magnusdóttir S, Ng CY, Preciat G, Žagare A, Chan SHJ, Aurich MK, Clancy CM, Modamio J, Sauls JT, Noronha A, Bordbar A, Cousins B, El Assal DC, Valcarcel LV, Apaolaza I, Ghaderi S, Ahookhosh M, Ben Guebila M, Kostromins A, Sompairac N, Le HM, Ma D, Sun Y, Wang L, Yurkovich JT, Oliveira MAP, Vuong PT, El Assal LP, Kuperstein I, Zinovyev A, Hinton HS, Bryant WA, Aragón Artacho FJ, Planes FJ, Stalidzans E, Maass A, Vempala S, Hucka M, Saunders MA, Maranas CD, Lewis NE, Sauter T, Palsson BØ, Thiele I, Fleming RMT. Creation and analysis of biochemical constraint-based models using the COBRA Toolbox v.3.0. Nat Protoc 2019; 14:639-702. [PMID: 30787451 PMCID: PMC6635304 DOI: 10.1038/s41596-018-0098-2] [Citation(s) in RCA: 695] [Impact Index Per Article: 115.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Constraint-based reconstruction and analysis (COBRA) provides a molecular mechanistic framework for integrative analysis of experimental molecular systems biology data and quantitative prediction of physicochemically and biochemically feasible phenotypic states. The COBRA Toolbox is a comprehensive desktop software suite of interoperable COBRA methods. It has found widespread application in biology, biomedicine, and biotechnology because its functions can be flexibly combined to implement tailored COBRA protocols for any biochemical network. This protocol is an update to the COBRA Toolbox v.1.0 and v.2.0. Version 3.0 includes new methods for quality-controlled reconstruction, modeling, topological analysis, strain and experimental design, and network visualization, as well as network integration of chemoinformatic, metabolomic, transcriptomic, proteomic, and thermochemical data. New multi-lingual code integration also enables an expansion in COBRA application scope via high-precision, high-performance, and nonlinear numerical optimization solvers for multi-scale, multi-cellular, and reaction kinetic modeling, respectively. This protocol provides an overview of all these new features and can be adapted to generate and analyze constraint-based models in a wide variety of scenarios. The COBRA Toolbox v.3.0 provides an unparalleled depth of COBRA methods.
Collapse
|
research-article |
6 |
695 |
8
|
Subramanian I, Verma S, Kumar S, Jere A, Anamika K. Multi-omics Data Integration, Interpretation, and Its Application. Bioinform Biol Insights 2020; 14:1177932219899051. [PMID: 32076369 PMCID: PMC7003173 DOI: 10.1177/1177932219899051] [Citation(s) in RCA: 683] [Impact Index Per Article: 136.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 11/09/2019] [Indexed: 12/22/2022] Open
Abstract
To study complex biological processes holistically, it is imperative to take an integrative approach that combines multi-omics data to highlight the interrelationships of the involved biomolecules and their functions. With the advent of high-throughput techniques and availability of multi-omics data generated from a large set of samples, several promising tools and methods have been developed for data integration and interpretation. In this review, we collected the tools and methods that adopt integrative approach to analyze multiple omics data and summarized their ability to address applications such as disease subtyping, biomarker prediction, and deriving insights into the data. We provide the methodology, use-cases, and limitations of these tools; brief account of multi-omics data repositories and visualization portals; and challenges associated with multi-omics data integration.
Collapse
|
Review |
5 |
683 |
9
|
Argelaguet R, Velten B, Arnol D, Dietrich S, Zenz T, Marioni JC, Buettner F, Huber W, Stegle O. Multi-Omics Factor Analysis-a framework for unsupervised integration of multi-omics data sets. Mol Syst Biol 2018; 14:e8124. [PMID: 29925568 PMCID: PMC6010767 DOI: 10.15252/msb.20178124] [Citation(s) in RCA: 619] [Impact Index Per Article: 88.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 12/19/2022] Open
Abstract
Multi-omics studies promise the improved characterization of biological processes across molecular layers. However, methods for the unsupervised integration of the resulting heterogeneous data sets are lacking. We present Multi-Omics Factor Analysis (MOFA), a computational method for discovering the principal sources of variation in multi-omics data sets. MOFA infers a set of (hidden) factors that capture biological and technical sources of variability. It disentangles axes of heterogeneity that are shared across multiple modalities and those specific to individual data modalities. The learnt factors enable a variety of downstream analyses, including identification of sample subgroups, data imputation and the detection of outlier samples. We applied MOFA to a cohort of 200 patient samples of chronic lymphocytic leukaemia, profiled for somatic mutations, RNA expression, DNA methylation and ex vivo drug responses. MOFA identified major dimensions of disease heterogeneity, including immunoglobulin heavy-chain variable region status, trisomy of chromosome 12 and previously underappreciated drivers, such as response to oxidative stress. In a second application, we used MOFA to analyse single-cell multi-omics data, identifying coordinated transcriptional and epigenetic changes along cell differentiation.
Collapse
|
Validation Study |
7 |
619 |
10
|
Kattge J, Bönisch G, Díaz S, Lavorel S, Prentice IC, Leadley P, Tautenhahn S, Werner GDA, Aakala T, Abedi M, Acosta ATR, Adamidis GC, Adamson K, Aiba M, Albert CH, Alcántara JM, Alcázar C C, Aleixo I, Ali H, Amiaud B, Ammer C, Amoroso MM, Anand M, Anderson C, Anten N, Antos J, Apgaua DMG, Ashman TL, Asmara DH, Asner GP, Aspinwall M, Atkin O, Aubin I, Baastrup-Spohr L, Bahalkeh K, Bahn M, Baker T, Baker WJ, Bakker JP, Baldocchi D, Baltzer J, Banerjee A, Baranger A, Barlow J, Barneche DR, Baruch Z, Bastianelli D, Battles J, Bauerle W, Bauters M, Bazzato E, Beckmann M, Beeckman H, Beierkuhnlein C, Bekker R, Belfry G, Belluau M, Beloiu M, Benavides R, Benomar L, Berdugo-Lattke ML, Berenguer E, Bergamin R, Bergmann J, Bergmann Carlucci M, Berner L, Bernhardt-Römermann M, Bigler C, Bjorkman AD, Blackman C, Blanco C, Blonder B, Blumenthal D, Bocanegra-González KT, Boeckx P, Bohlman S, Böhning-Gaese K, Boisvert-Marsh L, Bond W, Bond-Lamberty B, Boom A, Boonman CCF, Bordin K, Boughton EH, Boukili V, Bowman DMJS, Bravo S, Brendel MR, Broadley MR, Brown KA, Bruelheide H, Brumnich F, Bruun HH, Bruy D, Buchanan SW, Bucher SF, Buchmann N, Buitenwerf R, Bunker DE, Bürger J, et alKattge J, Bönisch G, Díaz S, Lavorel S, Prentice IC, Leadley P, Tautenhahn S, Werner GDA, Aakala T, Abedi M, Acosta ATR, Adamidis GC, Adamson K, Aiba M, Albert CH, Alcántara JM, Alcázar C C, Aleixo I, Ali H, Amiaud B, Ammer C, Amoroso MM, Anand M, Anderson C, Anten N, Antos J, Apgaua DMG, Ashman TL, Asmara DH, Asner GP, Aspinwall M, Atkin O, Aubin I, Baastrup-Spohr L, Bahalkeh K, Bahn M, Baker T, Baker WJ, Bakker JP, Baldocchi D, Baltzer J, Banerjee A, Baranger A, Barlow J, Barneche DR, Baruch Z, Bastianelli D, Battles J, Bauerle W, Bauters M, Bazzato E, Beckmann M, Beeckman H, Beierkuhnlein C, Bekker R, Belfry G, Belluau M, Beloiu M, Benavides R, Benomar L, Berdugo-Lattke ML, Berenguer E, Bergamin R, Bergmann J, Bergmann Carlucci M, Berner L, Bernhardt-Römermann M, Bigler C, Bjorkman AD, Blackman C, Blanco C, Blonder B, Blumenthal D, Bocanegra-González KT, Boeckx P, Bohlman S, Böhning-Gaese K, Boisvert-Marsh L, Bond W, Bond-Lamberty B, Boom A, Boonman CCF, Bordin K, Boughton EH, Boukili V, Bowman DMJS, Bravo S, Brendel MR, Broadley MR, Brown KA, Bruelheide H, Brumnich F, Bruun HH, Bruy D, Buchanan SW, Bucher SF, Buchmann N, Buitenwerf R, Bunker DE, Bürger J, Burrascano S, Burslem DFRP, Butterfield BJ, Byun C, Marques M, Scalon MC, Caccianiga M, Cadotte M, Cailleret M, Camac J, Camarero JJ, Campany C, Campetella G, Campos JA, Cano-Arboleda L, Canullo R, Carbognani M, Carvalho F, Casanoves F, Castagneyrol B, Catford JA, Cavender-Bares J, Cerabolini BEL, Cervellini M, Chacón-Madrigal E, Chapin K, Chapin FS, Chelli S, Chen SC, Chen A, Cherubini P, Chianucci F, Choat B, Chung KS, Chytrý M, Ciccarelli D, Coll L, Collins CG, Conti L, Coomes D, Cornelissen JHC, Cornwell WK, Corona P, Coyea M, Craine J, Craven D, Cromsigt JPGM, Csecserits A, Cufar K, Cuntz M, da Silva AC, Dahlin KM, Dainese M, Dalke I, Dalle Fratte M, Dang-Le AT, Danihelka J, Dannoura M, Dawson S, de Beer AJ, De Frutos A, De Long JR, Dechant B, Delagrange S, Delpierre N, Derroire G, Dias AS, Diaz-Toribio MH, Dimitrakopoulos PG, Dobrowolski M, Doktor D, Dřevojan P, Dong N, Dransfield J, Dressler S, Duarte L, Ducouret E, Dullinger S, Durka W, Duursma R, Dymova O, E-Vojtkó A, Eckstein RL, Ejtehadi H, Elser J, Emilio T, Engemann K, Erfanian MB, Erfmeier A, Esquivel-Muelbert A, Esser G, Estiarte M, Domingues TF, Fagan WF, Fagúndez J, Falster DS, Fan Y, Fang J, Farris E, Fazlioglu F, Feng Y, Fernandez-Mendez F, Ferrara C, Ferreira J, Fidelis A, Finegan B, Firn J, Flowers TJ, Flynn DFB, Fontana V, Forey E, Forgiarini C, François L, Frangipani M, Frank D, Frenette-Dussault C, Freschet GT, Fry EL, Fyllas NM, Mazzochini GG, Gachet S, Gallagher R, Ganade G, Ganga F, García-Palacios P, Gargaglione V, Garnier E, Garrido JL, de Gasper AL, Gea-Izquierdo G, Gibson D, Gillison AN, Giroldo A, Glasenhardt MC, Gleason S, Gliesch M, Goldberg E, Göldel B, Gonzalez-Akre E, Gonzalez-Andujar JL, González-Melo A, González-Robles A, Graae BJ, Granda E, Graves S, Green WA, Gregor T, Gross N, Guerin GR, Günther A, Gutiérrez AG, Haddock L, Haines A, Hall J, Hambuckers A, Han W, Harrison SP, Hattingh W, Hawes JE, He T, He P, Heberling JM, Helm A, Hempel S, Hentschel J, Hérault B, Hereş AM, Herz K, Heuertz M, Hickler T, Hietz P, Higuchi P, Hipp AL, Hirons A, Hock M, Hogan JA, Holl K, Honnay O, Hornstein D, Hou E, Hough-Snee N, Hovstad KA, Ichie T, Igić B, Illa E, Isaac M, Ishihara M, Ivanov L, Ivanova L, Iversen CM, Izquierdo J, Jackson RB, Jackson B, Jactel H, Jagodzinski AM, Jandt U, Jansen S, Jenkins T, Jentsch A, Jespersen JRP, Jiang GF, Johansen JL, Johnson D, Jokela EJ, Joly CA, Jordan GJ, Joseph GS, Junaedi D, Junker RR, Justes E, Kabzems R, Kane J, Kaplan Z, Kattenborn T, Kavelenova L, Kearsley E, Kempel A, Kenzo T, Kerkhoff A, Khalil MI, Kinlock NL, Kissling WD, Kitajima K, Kitzberger T, Kjøller R, Klein T, Kleyer M, Klimešová J, Klipel J, Kloeppel B, Klotz S, Knops JMH, Kohyama T, Koike F, Kollmann J, Komac B, Komatsu K, König C, Kraft NJB, Kramer K, Kreft H, Kühn I, Kumarathunge D, Kuppler J, Kurokawa H, Kurosawa Y, Kuyah S, Laclau JP, Lafleur B, Lallai E, Lamb E, Lamprecht A, Larkin DJ, Laughlin D, Le Bagousse-Pinguet Y, le Maire G, le Roux PC, le Roux E, Lee T, Lens F, Lewis SL, Lhotsky B, Li Y, Li X, Lichstein JW, Liebergesell M, Lim JY, Lin YS, Linares JC, Liu C, Liu D, Liu U, Livingstone S, Llusià J, Lohbeck M, López-García Á, Lopez-Gonzalez G, Lososová Z, Louault F, Lukács BA, Lukeš P, Luo Y, Lussu M, Ma S, Maciel Rabelo Pereira C, Mack M, Maire V, Mäkelä A, Mäkinen H, Malhado ACM, Mallik A, Manning P, Manzoni S, Marchetti Z, Marchino L, Marcilio-Silva V, Marcon E, Marignani M, Markesteijn L, Martin A, Martínez-Garza C, Martínez-Vilalta J, Mašková T, Mason K, Mason N, Massad TJ, Masse J, Mayrose I, McCarthy J, McCormack ML, McCulloh K, McFadden IR, McGill BJ, McPartland MY, Medeiros JS, Medlyn B, Meerts P, Mehrabi Z, Meir P, Melo FPL, Mencuccini M, Meredieu C, Messier J, Mészáros I, Metsaranta J, Michaletz ST, Michelaki C, Migalina S, Milla R, Miller JED, Minden V, Ming R, Mokany K, Moles AT, Molnár A, Molofsky J, Molz M, Montgomery RA, Monty A, Moravcová L, Moreno-Martínez A, Moretti M, Mori AS, Mori S, Morris D, Morrison J, Mucina L, Mueller S, Muir CD, Müller SC, Munoz F, Myers-Smith IH, Myster RW, Nagano M, Naidu S, Narayanan A, Natesan B, Negoita L, Nelson AS, Neuschulz EL, Ni J, Niedrist G, Nieto J, Niinemets Ü, Nolan R, Nottebrock H, Nouvellon Y, Novakovskiy A, Nystuen KO, O'Grady A, O'Hara K, O'Reilly-Nugent A, Oakley S, Oberhuber W, Ohtsuka T, Oliveira R, Öllerer K, Olson ME, Onipchenko V, Onoda Y, Onstein RE, Ordonez JC, Osada N, Ostonen I, Ottaviani G, Otto S, Overbeck GE, Ozinga WA, Pahl AT, Paine CET, Pakeman RJ, Papageorgiou AC, Parfionova E, Pärtel M, Patacca M, Paula S, Paule J, Pauli H, Pausas JG, Peco B, Penuelas J, Perea A, Peri PL, Petisco-Souza AC, Petraglia A, Petritan AM, Phillips OL, Pierce S, Pillar VD, Pisek J, Pomogaybin A, Poorter H, Portsmuth A, Poschlod P, Potvin C, Pounds D, Powell AS, Power SA, Prinzing A, Puglielli G, Pyšek P, Raevel V, Rammig A, Ransijn J, Ray CA, Reich PB, Reichstein M, Reid DEB, Réjou-Méchain M, de Dios VR, Ribeiro S, Richardson S, Riibak K, Rillig MC, Riviera F, Robert EMR, Roberts S, Robroek B, Roddy A, Rodrigues AV, Rogers A, Rollinson E, Rolo V, Römermann C, Ronzhina D, Roscher C, Rosell JA, Rosenfield MF, Rossi C, Roy DB, Royer-Tardif S, Rüger N, Ruiz-Peinado R, Rumpf SB, Rusch GM, Ryo M, Sack L, Saldaña A, Salgado-Negret B, Salguero-Gomez R, Santa-Regina I, Santacruz-García AC, Santos J, Sardans J, Schamp B, Scherer-Lorenzen M, Schleuning M, Schmid B, Schmidt M, Schmitt S, Schneider JV, Schowanek SD, Schrader J, Schrodt F, Schuldt B, Schurr F, Selaya Garvizu G, Semchenko M, Seymour C, Sfair JC, Sharpe JM, Sheppard CS, Sheremetiev S, Shiodera S, Shipley B, Shovon TA, Siebenkäs A, Sierra C, Silva V, Silva M, Sitzia T, Sjöman H, Slot M, Smith NG, Sodhi D, Soltis P, Soltis D, Somers B, Sonnier G, Sørensen MV, Sosinski EE, Soudzilovskaia NA, Souza AF, Spasojevic M, Sperandii MG, Stan AB, Stegen J, Steinbauer K, Stephan JG, Sterck F, Stojanovic DB, Strydom T, Suarez ML, Svenning JC, Svitková I, Svitok M, Svoboda M, Swaine E, Swenson N, Tabarelli M, Takagi K, Tappeiner U, Tarifa R, Tauugourdeau S, Tavsanoglu C, Te Beest M, Tedersoo L, Thiffault N, Thom D, Thomas E, Thompson K, Thornton PE, Thuiller W, Tichý L, Tissue D, Tjoelker MG, Tng DYP, Tobias J, Török P, Tarin T, Torres-Ruiz JM, Tóthmérész B, Treurnicht M, Trivellone V, Trolliet F, Trotsiuk V, Tsakalos JL, Tsiripidis I, Tysklind N, Umehara T, Usoltsev V, Vadeboncoeur M, Vaezi J, Valladares F, Vamosi J, van Bodegom PM, van Breugel M, Van Cleemput E, van de Weg M, van der Merwe S, van der Plas F, van der Sande MT, van Kleunen M, Van Meerbeek K, Vanderwel M, Vanselow KA, Vårhammar A, Varone L, Vasquez Valderrama MY, Vassilev K, Vellend M, Veneklaas EJ, Verbeeck H, Verheyen K, Vibrans A, Vieira I, Villacís J, Violle C, Vivek P, Wagner K, Waldram M, Waldron A, Walker AP, Waller M, Walther G, Wang H, Wang F, Wang W, Watkins H, Watkins J, Weber U, Weedon JT, Wei L, Weigelt P, Weiher E, Wells AW, Wellstein C, Wenk E, Westoby M, Westwood A, White PJ, Whitten M, Williams M, Winkler DE, Winter K, Womack C, Wright IJ, Wright SJ, Wright J, Pinho BX, Ximenes F, Yamada T, Yamaji K, Yanai R, Yankov N, Yguel B, Zanini KJ, Zanne AE, Zelený D, Zhao YP, Zheng J, Zheng J, Ziemińska K, Zirbel CR, Zizka G, Zo-Bi IC, Zotz G, Wirth C. TRY plant trait database - enhanced coverage and open access. GLOBAL CHANGE BIOLOGY 2020; 26:119-188. [PMID: 31891233 DOI: 10.1111/gcb.14904] [Show More Authors] [Citation(s) in RCA: 610] [Impact Index Per Article: 122.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 09/12/2019] [Indexed: 05/17/2023]
Abstract
Plant traits-the morphological, anatomical, physiological, biochemical and phenological characteristics of plants-determine how plants respond to environmental factors, affect other trophic levels, and influence ecosystem properties and their benefits and detriments to people. Plant trait data thus represent the basis for a vast area of research spanning from evolutionary biology, community and functional ecology, to biodiversity conservation, ecosystem and landscape management, restoration, biogeography and earth system modelling. Since its foundation in 2007, the TRY database of plant traits has grown continuously. It now provides unprecedented data coverage under an open access data policy and is the main plant trait database used by the research community worldwide. Increasingly, the TRY database also supports new frontiers of trait-based plant research, including the identification of data gaps and the subsequent mobilization or measurement of new data. To support this development, in this article we evaluate the extent of the trait data compiled in TRY and analyse emerging patterns of data coverage and representativeness. Best species coverage is achieved for categorical traits-almost complete coverage for 'plant growth form'. However, most traits relevant for ecology and vegetation modelling are characterized by continuous intraspecific variation and trait-environmental relationships. These traits have to be measured on individual plants in their respective environment. Despite unprecedented data coverage, we observe a humbling lack of completeness and representativeness of these continuous traits in many aspects. We, therefore, conclude that reducing data gaps and biases in the TRY database remains a key challenge and requires a coordinated approach to data mobilization and trait measurements. This can only be achieved in collaboration with other initiatives.
Collapse
|
|
5 |
610 |
11
|
Luecken MD, Büttner M, Chaichoompu K, Danese A, Interlandi M, Mueller MF, Strobl DC, Zappia L, Dugas M, Colomé-Tatché M, Theis FJ. Benchmarking atlas-level data integration in single-cell genomics. Nat Methods 2022; 19:41-50. [PMID: 34949812 PMCID: PMC8748196 DOI: 10.1038/s41592-021-01336-8] [Citation(s) in RCA: 501] [Impact Index Per Article: 167.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/01/2021] [Indexed: 12/12/2022]
Abstract
Single-cell atlases often include samples that span locations, laboratories and conditions, leading to complex, nested batch effects in data. Thus, joint analysis of atlas datasets requires reliable data integration. To guide integration method choice, we benchmarked 68 method and preprocessing combinations on 85 batches of gene expression, chromatin accessibility and simulation data from 23 publications, altogether representing >1.2 million cells distributed in 13 atlas-level integration tasks. We evaluated methods according to scalability, usability and their ability to remove batch effects while retaining biological variation using 14 evaluation metrics. We show that highly variable gene selection improves the performance of data integration methods, whereas scaling pushes methods to prioritize batch removal over conservation of biological variation. Overall, scANVI, Scanorama, scVI and scGen perform well, particularly on complex integration tasks, while single-cell ATAC-sequencing integration performance is strongly affected by choice of feature space. Our freely available Python module and benchmarking pipeline can identify optimal data integration methods for new data, benchmark new methods and improve method development.
Collapse
|
research-article |
3 |
501 |
12
|
Gonzalez-Perez A, Perez-Llamas C, Deu-Pons J, Tamborero D, Schroeder MP, Jene-Sanz A, Santos A, Lopez-Bigas N. IntOGen-mutations identifies cancer drivers across tumor types. Nat Methods 2013; 10:1081-2. [PMID: 24037244 PMCID: PMC5758042 DOI: 10.1038/nmeth.2642] [Citation(s) in RCA: 426] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/12/2013] [Indexed: 01/08/2023]
Abstract
The IntOGen-mutations platform (http://www.intogen.org/mutations/) summarizes somatic mutations, genes and pathways involved in tumorigenesis. It identifies and visualizes cancer drivers, analyzing 4,623 exomes from 13 cancer sites. It provides support to cancer researchers, aids the identification of drivers across tumor cohorts and helps rank mutations for better clinical decision-making.
Collapse
|
research-article |
12 |
426 |
13
|
Zhang GQ, Cui L, Mueller R, Tao S, Kim M, Rueschman M, Mariani S, Mobley D, Redline S. The National Sleep Research Resource: towards a sleep data commons. J Am Med Inform Assoc 2018; 25:1351-1358. [PMID: 29860441 PMCID: PMC6188513 DOI: 10.1093/jamia/ocy064] [Citation(s) in RCA: 392] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 04/05/2018] [Accepted: 04/26/2018] [Indexed: 11/12/2022] Open
Abstract
Objective The gold standard for diagnosing sleep disorders is polysomnography, which generates extensive data about biophysical changes occurring during sleep. We developed the National Sleep Research Resource (NSRR), a comprehensive system for sharing sleep data. The NSRR embodies elements of a data commons aimed at accelerating research to address critical questions about the impact of sleep disorders on important health outcomes. Approach We used a metadata-guided approach, with a set of common sleep-specific terms enforcing uniform semantic interpretation of data elements across three main components: (1) annotated datasets; (2) user interfaces for accessing data; and (3) computational tools for the analysis of polysomnography recordings. We incorporated the process for managing dataset-specific data use agreements, evidence of Institutional Review Board review, and the corresponding access control in the NSRR web portal. The metadata-guided approach facilitates structural and semantic interoperability, ultimately leading to enhanced data reusability and scientific rigor. Results The authors curated and deposited retrospective data from 10 large, NIH-funded sleep cohort studies, including several from the Trans-Omics for Precision Medicine (TOPMed) program, into the NSRR. The NSRR currently contains data on 26 808 subjects and 31 166 signal files in European Data Format. Launched in April 2014, over 3000 registered users have downloaded over 130 terabytes of data. Conclusions The NSRR offers a use case and an example for creating a full-fledged data commons. It provides a single point of access to analysis-ready physiological signals from polysomnography obtained from multiple sources, and a wide variety of clinical data to facilitate sleep research.
Collapse
|
Research Support, N.I.H., Extramural |
7 |
392 |
14
|
Pinu FR, Beale DJ, Paten AM, Kouremenos K, Swarup S, Schirra HJ, Wishart D. Systems Biology and Multi-Omics Integration: Viewpoints from the Metabolomics Research Community. Metabolites 2019; 9:E76. [PMID: 31003499 PMCID: PMC6523452 DOI: 10.3390/metabo9040076] [Citation(s) in RCA: 347] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 02/07/2023] Open
Abstract
The use of multiple omics techniques (i.e., genomics, transcriptomics, proteomics, and metabolomics) is becoming increasingly popular in all facets of life science. Omics techniques provide a more holistic molecular perspective of studied biological systems compared to traditional approaches. However, due to their inherent data differences, integrating multiple omics platforms remains an ongoing challenge for many researchers. As metabolites represent the downstream products of multiple interactions between genes, transcripts, and proteins, metabolomics, the tools and approaches routinely used in this field could assist with the integration of these complex multi-omics data sets. The question is, how? Here we provide some answers (in terms of methods, software tools and databases) along with a variety of recommendations and a list of continuing challenges as identified during a peer session on multi-omics integration that was held at the recent 'Australian and New Zealand Metabolomics Conference' (ANZMET 2018) in Auckland, New Zealand (Sept. 2018). We envisage that this document will serve as a guide to metabolomics researchers and other members of the community wishing to perform multi-omics studies. We also believe that these ideas may allow the full promise of integrated multi-omics research and, ultimately, of systems biology to be realized.
Collapse
|
other |
6 |
347 |
15
|
Madhavan S, Zenklusen JC, Kotliarov Y, Sahni H, Fine HA, Buetow K. Rembrandt: helping personalized medicine become a reality through integrative translational research. Mol Cancer Res 2009; 7:157-67. [PMID: 19208739 PMCID: PMC2645472 DOI: 10.1158/1541-7786.mcr-08-0435] [Citation(s) in RCA: 340] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Finding better therapies for the treatment of brain tumors is hampered by the lack of consistently obtained molecular data in a large sample set and the ability to integrate biomedical data from disparate sources enabling translation of therapies from bench to bedside. Hence, a critical factor in the advancement of biomedical research and clinical translation is the ease with which data can be integrated, redistributed, and analyzed both within and across functional domains. Novel biomedical informatics infrastructure and tools are essential for developing individualized patient treatment based on the specific genomic signatures in each patient's tumor. Here, we present Repository of Molecular Brain Neoplasia Data (Rembrandt), a cancer clinical genomics database and a Web-based data mining and analysis platform aimed at facilitating discovery by connecting the dots between clinical information and genomic characterization data. To date, Rembrandt contains data generated through the Glioma Molecular Diagnostic Initiative from 874 glioma specimens comprising approximately 566 gene expression arrays, 834 copy number arrays, and 13,472 clinical phenotype data points. Data can be queried and visualized for a selected gene across all data platforms or for multiple genes in a selected platform. Additionally, gene sets can be limited to clinically important annotations including secreted, kinase, membrane, and known gene-anomaly pairs to facilitate the discovery of novel biomarkers and therapeutic targets. We believe that Rembrandt represents a prototype of how high-throughput genomic and clinical data can be integrated in a way that will allow expeditious and efficient translation of laboratory discoveries to the clinic.
Collapse
|
Research Support, N.I.H., Intramural |
16 |
340 |
16
|
Rozenblatt-Rosen O, Regev A, Oberdoerffer P, Nawy T, Hupalowska A, Rood JE, Ashenberg O, Cerami E, Coffey RJ, Demir E, Ding L, Esplin ED, Ford JM, Goecks J, Ghosh S, Gray JW, Guinney J, Hanlon SE, Hughes SK, Hwang ES, Iacobuzio-Donahue CA, Jané-Valbuena J, Johnson BE, Lau KS, Lively T, Mazzilli SA, Pe'er D, Santagata S, Shalek AK, Schapiro D, Snyder MP, Sorger PK, Spira AE, Srivastava S, Tan K, West RB, Williams EH. The Human Tumor Atlas Network: Charting Tumor Transitions across Space and Time at Single-Cell Resolution. Cell 2020; 181:236-249. [PMID: 32302568 PMCID: PMC7376497 DOI: 10.1016/j.cell.2020.03.053] [Citation(s) in RCA: 335] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 12/22/2022]
Abstract
Crucial transitions in cancer-including tumor initiation, local expansion, metastasis, and therapeutic resistance-involve complex interactions between cells within the dynamic tumor ecosystem. Transformative single-cell genomics technologies and spatial multiplex in situ methods now provide an opportunity to interrogate this complexity at unprecedented resolution. The Human Tumor Atlas Network (HTAN), part of the National Cancer Institute (NCI) Cancer Moonshot Initiative, will establish a clinical, experimental, computational, and organizational framework to generate informative and accessible three-dimensional atlases of cancer transitions for a diverse set of tumor types. This effort complements both ongoing efforts to map healthy organs and previous large-scale cancer genomics approaches focused on bulk sequencing at a single point in time. Generating single-cell, multiparametric, longitudinal atlases and integrating them with clinical outcomes should help identify novel predictive biomarkers and features as well as therapeutically relevant cell types, cell states, and cellular interactions across transitions. The resulting tumor atlases should have a profound impact on our understanding of cancer biology and have the potential to improve cancer detection, prevention, and therapeutic discovery for better precision-medicine treatments of cancer patients and those at risk for cancer.
Collapse
|
Research Support, N.I.H., Extramural |
5 |
335 |
17
|
Kuppe C, Ramirez Flores RO, Li Z, Hayat S, Levinson RT, Liao X, Hannani MT, Tanevski J, Wünnemann F, Nagai JS, Halder M, Schumacher D, Menzel S, Schäfer G, Hoeft K, Cheng M, Ziegler S, Zhang X, Peisker F, Kaesler N, Saritas T, Xu Y, Kassner A, Gummert J, Morshuis M, Amrute J, Veltrop RJA, Boor P, Klingel K, Van Laake LW, Vink A, Hoogenboezem RM, Bindels EMJ, Schurgers L, Sattler S, Schapiro D, Schneider RK, Lavine K, Milting H, Costa IG, Saez-Rodriguez J, Kramann R. Spatial multi-omic map of human myocardial infarction. Nature 2022; 608:766-777. [PMID: 35948637 PMCID: PMC9364862 DOI: 10.1038/s41586-022-05060-x] [Citation(s) in RCA: 319] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 06/29/2022] [Indexed: 02/01/2023]
Abstract
Myocardial infarction is a leading cause of death worldwide1. Although advances have been made in acute treatment, an incomplete understanding of remodelling processes has limited the effectiveness of therapies to reduce late-stage mortality2. Here we generate an integrative high-resolution map of human cardiac remodelling after myocardial infarction using single-cell gene expression, chromatin accessibility and spatial transcriptomic profiling of multiple physiological zones at distinct time points in myocardium from patients with myocardial infarction and controls. Multi-modal data integration enabled us to evaluate cardiac cell-type compositions at increased resolution, yielding insights into changes of the cardiac transcriptome and epigenome through the identification of distinct tissue structures of injury, repair and remodelling. We identified and validated disease-specific cardiac cell states of major cell types and analysed them in their spatial context, evaluating their dependency on other cell types. Our data elucidate the molecular principles of human myocardial tissue organization, recapitulating a gradual cardiomyocyte and myeloid continuum following ischaemic injury. In sum, our study provides an integrative molecular map of human myocardial infarction, represents an essential reference for the field and paves the way for advanced mechanistic and therapeutic studies of cardiac disease.
Collapse
|
research-article |
3 |
319 |
18
|
Wang LG, Lam TTY, Xu S, Dai Z, Zhou L, Feng T, Guo P, Dunn CW, Jones BR, Bradley T, Zhu H, Guan Y, Jiang Y, Yu G. Treeio: An R Package for Phylogenetic Tree Input and Output with Richly Annotated and Associated Data. Mol Biol Evol 2020; 37:599-603. [PMID: 31633786 DOI: 10.1093/molbev/msz240] [Citation(s) in RCA: 308] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Phylogenetic trees and data are often stored in incompatible and inconsistent formats. The outputs of software tools that contain trees with analysis findings are often not compatible with each other, making it hard to integrate the results of different analyses in a comparative study. The treeio package is designed to connect phylogenetic tree input and output. It supports extracting phylogenetic trees as well as the outputs of commonly used analytical software. It can link external data to phylogenies and merge tree data obtained from different sources, enabling analyses of phylogeny-associated data from different disciplines in an evolutionary context. Treeio also supports export of a phylogenetic tree with heterogeneous-associated data to a single tree file, including BEAST compatible NEXUS and jtree formats; these facilitate data sharing as well as file format conversion for downstream analysis. The treeio package is designed to work with the tidytree and ggtree packages. Tree data can be processed using the tidy interface with tidytree and visualized by ggtree. The treeio package is released within the Bioconductor and rOpenSci projects. It is available at https://www.bioconductor.org/packages/treeio/.
Collapse
|
Research Support, Non-U.S. Gov't |
5 |
308 |
19
|
PCAWG Transcriptome Core Group CalabreseClaudia2DavidsonNatalie R.34567DemircioğluDeniz89FonsecaNuno A.2HeYao10KahlesAndré3467LehmannKjong-Van3467LiuFenglin10ShiraishiYuichi11SouletteCameron M.12UrbanLara2, Calabrese C, Davidson NR, Demircioğlu D, Fonseca NA, He Y, Kahles A, Lehmann KV, Liu F, Shiraishi Y, Soulette CM, Urban L, Greger L, Li S, Liu D, Perry MD, Xiang Q, Zhang F, Zhang J, Bailey P, Erkek S, Hoadley KA, Hou Y, Huska MR, Kilpinen H, Korbel JO, Marin MG, Markowski J, Nandi T, Pan-Hammarström Q, Pedamallu CS, Siebert R, Stark SG, Su H, Tan P, Waszak SM, Yung C, Zhu S, Awadalla P, Creighton CJ, Meyerson M, Ouellette BFF, Wu K, Yang H, PCAWG Transcriptome Working Group FonsecaNuno A.2KahlesAndré3467LehmannKjong-Van3467UrbanLara2SouletteCameron M.12ShiraishiYuichi11LiuFenglin10HeYao10DemircioğluDeniz89DavidsonNatalie R.34567CalabreseClaudia2ZhangJunjun15PerryMarc D.1516XiangQian15GregerLiliana2LiSiliang1314LiuDongbing1314StarkStefan G.3467ZhangFan10AminSamirkumar B.37BaileyPeter17ChateignerAurélien15Cortés-CirianoIsidro293839CraftBrian12ErkekSerap18Frenkel-MorgensternMilana40GoldmanMary12HoadleyKatherine A.19HouYong1314HuskaMatthew R.20KhuranaEkta5KilpinenHelena21KorbelJan O.18LamazeFabien C.15LiChang1314LiXiaobo1314LiXinyue13LiuXingmin1314MarinMaximillian G.12MarkowskiJulia20NandiTannistha9NielsenMorten M.41OjesinaAkinyemi I.23284243Pan-HammarströmQiang1322ParkPeter J.2938PedamalluChandra Sekhar232829PedersenJakob S.41SiebertReiner24SuHong1314TanPatrick925TehBin Tean31WangJian13WaszakSebastian M.18XiongHeng1314YakneenSergei18YeChen1314YungChristina15ZhangXiuqing13ZhengLiangtao10ZhuJingchun12ZhuShida1314AwadallaPhilip1526CreightonChad J.27MeyersonMatthew232829OuelletteB. F. Francis30WuKui1314YangHuanming13GökeJonathan931SchwarzRoland F.2203233StegleOliver21833ZhangZemin10BrazmaAlvis2RätschGunnar34567BrooksAngela N.122328, Brazma A, Brooks AN, Göke J, Rätsch G, Schwarz RF, Stegle O, Zhang Z, PCAWG Consortium. Genomic basis for RNA alterations in cancer. Nature 2020; 578:129-136. [PMID: 32025019 PMCID: PMC7054216 DOI: 10.1038/s41586-020-1970-0] [Citation(s) in RCA: 294] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 12/11/2019] [Indexed: 01/27/2023]
Abstract
Transcript alterations often result from somatic changes in cancer genomes1. Various forms of RNA alterations have been described in cancer, including overexpression2, altered splicing3 and gene fusions4; however, it is difficult to attribute these to underlying genomic changes owing to heterogeneity among patients and tumour types, and the relatively small cohorts of patients for whom samples have been analysed by both transcriptome and whole-genome sequencing. Here we present, to our knowledge, the most comprehensive catalogue of cancer-associated gene alterations to date, obtained by characterizing tumour transcriptomes from 1,188 donors of the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA)5. Using matched whole-genome sequencing data, we associated several categories of RNA alterations with germline and somatic DNA alterations, and identified probable genetic mechanisms. Somatic copy-number alterations were the major drivers of variations in total gene and allele-specific expression. We identified 649 associations of somatic single-nucleotide variants with gene expression in cis, of which 68.4% involved associations with flanking non-coding regions of the gene. We found 1,900 splicing alterations associated with somatic mutations, including the formation of exons within introns in proximity to Alu elements. In addition, 82% of gene fusions were associated with structural variants, including 75 of a new class, termed 'bridged' fusions, in which a third genomic location bridges two genes. We observed transcriptomic alteration signatures that differ between cancer types and have associations with variations in DNA mutational signatures. This compendium of RNA alterations in the genomic context provides a rich resource for identifying genes and mechanisms that are functionally implicated in cancer.
Collapse
|
Dataset |
5 |
294 |
20
|
Keenan AB, Jenkins SL, Jagodnik KM, Koplev S, He E, Torre D, Wang Z, Dohlman AB, Silverstein MC, Lachmann A, Kuleshov MV, Ma'ayan A, Stathias V, Terryn R, Cooper D, Forlin M, Koleti A, Vidovic D, Chung C, Schürer SC, Vasiliauskas J, Pilarczyk M, Shamsaei B, Fazel M, Ren Y, Niu W, Clark NA, White S, Mahi N, Zhang L, Kouril M, Reichard JF, Sivaganesan S, Medvedovic M, Meller J, Koch RJ, Birtwistle MR, Iyengar R, Sobie EA, Azeloglu EU, Kaye J, Osterloh J, Haston K, Kalra J, Finkbiener S, Li J, Milani P, Adam M, Escalante-Chong R, Sachs K, Lenail A, Ramamoorthy D, Fraenkel E, Daigle G, Hussain U, Coye A, Rothstein J, Sareen D, Ornelas L, Banuelos M, Mandefro B, Ho R, Svendsen CN, Lim RG, Stocksdale J, Casale MS, Thompson TG, Wu J, Thompson LM, Dardov V, Venkatraman V, Matlock A, Van Eyk JE, Jaffe JD, Papanastasiou M, Subramanian A, Golub TR, Erickson SD, Fallahi-Sichani M, Hafner M, Gray NS, Lin JR, Mills CE, Muhlich JL, Niepel M, Shamu CE, Williams EH, Wrobel D, Sorger PK, Heiser LM, Gray JW, Korkola JE, Mills GB, LaBarge M, Feiler HS, Dane MA, Bucher E, Nederlof M, Sudar D, Gross S, et alKeenan AB, Jenkins SL, Jagodnik KM, Koplev S, He E, Torre D, Wang Z, Dohlman AB, Silverstein MC, Lachmann A, Kuleshov MV, Ma'ayan A, Stathias V, Terryn R, Cooper D, Forlin M, Koleti A, Vidovic D, Chung C, Schürer SC, Vasiliauskas J, Pilarczyk M, Shamsaei B, Fazel M, Ren Y, Niu W, Clark NA, White S, Mahi N, Zhang L, Kouril M, Reichard JF, Sivaganesan S, Medvedovic M, Meller J, Koch RJ, Birtwistle MR, Iyengar R, Sobie EA, Azeloglu EU, Kaye J, Osterloh J, Haston K, Kalra J, Finkbiener S, Li J, Milani P, Adam M, Escalante-Chong R, Sachs K, Lenail A, Ramamoorthy D, Fraenkel E, Daigle G, Hussain U, Coye A, Rothstein J, Sareen D, Ornelas L, Banuelos M, Mandefro B, Ho R, Svendsen CN, Lim RG, Stocksdale J, Casale MS, Thompson TG, Wu J, Thompson LM, Dardov V, Venkatraman V, Matlock A, Van Eyk JE, Jaffe JD, Papanastasiou M, Subramanian A, Golub TR, Erickson SD, Fallahi-Sichani M, Hafner M, Gray NS, Lin JR, Mills CE, Muhlich JL, Niepel M, Shamu CE, Williams EH, Wrobel D, Sorger PK, Heiser LM, Gray JW, Korkola JE, Mills GB, LaBarge M, Feiler HS, Dane MA, Bucher E, Nederlof M, Sudar D, Gross S, Kilburn DF, Smith R, Devlin K, Margolis R, Derr L, Lee A, Pillai A. The Library of Integrated Network-Based Cellular Signatures NIH Program: System-Level Cataloging of Human Cells Response to Perturbations. Cell Syst 2018; 6:13-24. [PMID: 29199020 PMCID: PMC5799026 DOI: 10.1016/j.cels.2017.11.001] [Show More Authors] [Citation(s) in RCA: 283] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/13/2017] [Accepted: 11/01/2017] [Indexed: 12/19/2022]
Abstract
The Library of Integrated Network-Based Cellular Signatures (LINCS) is an NIH Common Fund program that catalogs how human cells globally respond to chemical, genetic, and disease perturbations. Resources generated by LINCS include experimental and computational methods, visualization tools, molecular and imaging data, and signatures. By assembling an integrated picture of the range of responses of human cells exposed to many perturbations, the LINCS program aims to better understand human disease and to advance the development of new therapies. Perturbations under study include drugs, genetic perturbations, tissue micro-environments, antibodies, and disease-causing mutations. Responses to perturbations are measured by transcript profiling, mass spectrometry, cell imaging, and biochemical methods, among other assays. The LINCS program focuses on cellular physiology shared among tissues and cell types relevant to an array of diseases, including cancer, heart disease, and neurodegenerative disorders. This Perspective describes LINCS technologies, datasets, tools, and approaches to data accessibility and reusability.
Collapse
|
Research Support, N.I.H., Extramural |
7 |
283 |
21
|
Sammut SJ, Crispin-Ortuzar M, Chin SF, Provenzano E, Bardwell HA, Ma W, Cope W, Dariush A, Dawson SJ, Abraham JE, Dunn J, Hiller L, Thomas J, Cameron DA, Bartlett JMS, Hayward L, Pharoah PD, Markowetz F, Rueda OM, Earl HM, Caldas C. Multi-omic machine learning predictor of breast cancer therapy response. Nature 2022; 601:623-629. [PMID: 34875674 PMCID: PMC8791834 DOI: 10.1038/s41586-021-04278-5] [Citation(s) in RCA: 280] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/23/2021] [Indexed: 11/09/2022]
Abstract
Breast cancers are complex ecosystems of malignant cells and the tumour microenvironment1. The composition of these tumour ecosystems and interactions within them contribute to responses to cytotoxic therapy2. Efforts to build response predictors have not incorporated this knowledge. We collected clinical, digital pathology, genomic and transcriptomic profiles of pre-treatment biopsies of breast tumours from 168 patients treated with chemotherapy with or without HER2 (encoded by ERBB2)-targeted therapy before surgery. Pathology end points (complete response or residual disease) at surgery3 were then correlated with multi-omic features in these diagnostic biopsies. Here we show that response to treatment is modulated by the pre-treated tumour ecosystem, and its multi-omics landscape can be integrated in predictive models using machine learning. The degree of residual disease following therapy is monotonically associated with pre-therapy features, including tumour mutational and copy number landscapes, tumour proliferation, immune infiltration and T cell dysfunction and exclusion. Combining these features into a multi-omic machine learning model predicted a pathological complete response in an external validation cohort (75 patients) with an area under the curve of 0.87. In conclusion, response to therapy is determined by the baseline characteristics of the totality of the tumour ecosystem captured through data integration and machine learning. This approach could be used to develop predictors for other cancers.
Collapse
|
research-article |
3 |
280 |
22
|
Sikkema L, Ramírez-Suástegui C, Strobl DC, Gillett TE, Zappia L, Madissoon E, Markov NS, Zaragosi LE, Ji Y, Ansari M, Arguel MJ, Apperloo L, Banchero M, Bécavin C, Berg M, Chichelnitskiy E, Chung MI, Collin A, Gay ACA, Gote-Schniering J, Hooshiar Kashani B, Inecik K, Jain M, Kapellos TS, Kole TM, Leroy S, Mayr CH, Oliver AJ, von Papen M, Peter L, Taylor CJ, Walzthoeni T, Xu C, Bui LT, De Donno C, Dony L, Faiz A, Guo M, Gutierrez AJ, Heumos L, Huang N, Ibarra IL, Jackson ND, Kadur Lakshminarasimha Murthy P, Lotfollahi M, Tabib T, Talavera-López C, Travaglini KJ, Wilbrey-Clark A, Worlock KB, Yoshida M, van den Berge M, Bossé Y, Desai TJ, Eickelberg O, Kaminski N, Krasnow MA, Lafyatis R, Nikolic MZ, Powell JE, Rajagopal J, Rojas M, Rozenblatt-Rosen O, Seibold MA, Sheppard D, Shepherd DP, Sin DD, Timens W, Tsankov AM, Whitsett J, Xu Y, Banovich NE, Barbry P, Duong TE, Falk CS, Meyer KB, Kropski JA, Pe'er D, Schiller HB, Tata PR, Schultze JL, Teichmann SA, Misharin AV, Nawijn MC, Luecken MD, Theis FJ. An integrated cell atlas of the lung in health and disease. Nat Med 2023; 29:1563-1577. [PMID: 37291214 PMCID: PMC10287567 DOI: 10.1038/s41591-023-02327-2] [Citation(s) in RCA: 277] [Impact Index Per Article: 138.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 03/30/2023] [Indexed: 06/10/2023]
Abstract
Single-cell technologies have transformed our understanding of human tissues. Yet, studies typically capture only a limited number of donors and disagree on cell type definitions. Integrating many single-cell datasets can address these limitations of individual studies and capture the variability present in the population. Here we present the integrated Human Lung Cell Atlas (HLCA), combining 49 datasets of the human respiratory system into a single atlas spanning over 2.4 million cells from 486 individuals. The HLCA presents a consensus cell type re-annotation with matching marker genes, including annotations of rare and previously undescribed cell types. Leveraging the number and diversity of individuals in the HLCA, we identify gene modules that are associated with demographic covariates such as age, sex and body mass index, as well as gene modules changing expression along the proximal-to-distal axis of the bronchial tree. Mapping new data to the HLCA enables rapid data annotation and interpretation. Using the HLCA as a reference for the study of disease, we identify shared cell states across multiple lung diseases, including SPP1+ profibrotic monocyte-derived macrophages in COVID-19, pulmonary fibrosis and lung carcinoma. Overall, the HLCA serves as an example for the development and use of large-scale, cross-dataset organ atlases within the Human Cell Atlas.
Collapse
Grants
- P50 AR080612 NIAMS NIH HHS
- R01 HL153375 NHLBI NIH HHS
- R01 HL127349 NHLBI NIH HHS
- U54 HL165443 NHLBI NIH HHS
- P01 HL107202 NHLBI NIH HHS
- U01 HL148856 NHLBI NIH HHS
- R21 HL156124 NHLBI NIH HHS
- U54 AG075931 NIA NIH HHS
- Wellcome Trust
- R01 HL146557 NHLBI NIH HHS
- R01 HL123766 NHLBI NIH HHS
- U01 HL148861 NHLBI NIH HHS
- R01 HL141852 NHLBI NIH HHS
- R01 ES034350 NIEHS NIH HHS
- UL1 TR001863 NCATS NIH HHS
- R01 HL126176 NHLBI NIH HHS
- R21 HL161760 NHLBI NIH HHS
- R01 HL145372 NHLBI NIH HHS
- P01 AG049665 NIA NIH HHS
- K12 HD105271 NICHD NIH HHS
- U19 AI135964 NIAID NIH HHS
- P30 CA008748 NCI NIH HHS
- R01 HL142568 NHLBI NIH HHS
- R01 HL153312 NHLBI NIH HHS
- U54 AG079754 NIA NIH HHS
- R56 HL157632 NHLBI NIH HHS
- R01 HL158139 NHLBI NIH HHS
- R01 HL135156 NHLBI NIH HHS
- R01 HL153045 NHLBI NIH HHS
- U54 HL145608 NHLBI NIH HHS
- P50 AR060780 NIAMS NIH HHS
- R01 HL128439 NHLBI NIH HHS
- R01 HL146519 NHLBI NIH HHS
- R01 HL117004 NHLBI NIH HHS
- R01 HL068702 NHLBI NIH HHS
- U01 HL145567 NHLBI NIH HHS
- P01 HL132821 NHLBI NIH HHS
- MR/R015635/1 Medical Research Council
- R01 MD010443 NIMHD NIH HHS
- Chan Zuckerberg Initiative, LLC Seed Network grant (CZF2019-002438) “Lung Cell Atlas 1.0” NIH 1U54HL145608-01 CZIF2022-007488 from the Chan Zuckerberg Initiative Foundation CZIF2022-007488 from the Chan Zuckerberg Initiative Foundation
- ESPOD fellowship of EMBL-EBI and Sanger Institute
- 3IA Cote d’Azur PhD program
- The Ministry of Economic Affairs and Climate Policy by means of the PPP
- EC | Horizon 2020 Framework Programme (EU Framework Programme for Research and Innovation H2020)
- Joachim Herz Stiftung (Joachim Herz Foundation)
- P50 AR060780-06A1
- University College London, Birkbeck MRC Doctoral Training Programme
- Jikei University School of Medicine (Jikei University)
- 5R01HL14254903, 4UH3CA25513503
- R01HL127349, R01HL141852, U01HL145567 and CZI
- MRC Clinician Scientist Fellowship (MR/W00111X/1)
- Chan Zuckerberg Initiative, LLC Seed Network grant (CZF2019-002438) “Lung Cell Atlas 1.0” 2R01HL068702
- R01 HL135156, R01 MD010443, R01 HL128439, P01 HL132821, P01 HL107202, R01 HL117004, and DOD Grant W81WH-16-2-0018
- HL142568 and HL14507 from the NHLBI
- Chan Zuckerberg Initiative, LLC Seed Network grant (CZF2019-002438) “Lung Cell Atlas 1.0”, 2R01HL068702
- Wellcome (WT211276/Z/18/Z) Sanger core grant WT206194 CZIF2022-007488 from the Chan Zuckerberg Initiative Foundation
- R21HL156124, R56HL157632, and R21HL161760
- CZI, 5U01HL148856
- CZI, 5U01HL148856, R01 HL153045
- U.S. Department of Defense (United States Department of Defense)
- The National Institute of Health R01HL145372
- Fondation pour la Recherche Médicale (Foundation for Medical Research in France)
- Conseil Départemental des Alpes Maritimes
- Inserm Cross-cutting Scientific Program HuDeCA 2018, ANR SAHARRA (ANR-19-CE14–0027), ANR-19-P3IA-0002–3IA, the National Infrastructure France Génomique (ANR-10-INBS-09-03), PPIA 4D-OMICS (21-ESRE-0052), and the Chan Zuckerberg Initiative, LLC Seed Network grant (CZF2019-002438) “Lung Cell Atlas 1.0”.
- Wellcome Trust (Wellcome)
- Sanger core grant WT206194 Chan Zuckerberg Initiative, LLC Seed Network grant (CZF2019-002438) “Lung Cell Atlas 1.0” CZIF2022-007488 from the Chan Zuckerberg Initiative Foundation
- Doris Duke Charitable Foundation (DDCF)
- The National Institute of Health R01HL145372 Department of Defense W81XWH-19-1-0416
- The National Institute of Health R01HL146557 and R01HL153375 and funds from Chan Zuckerberg Initiative - Human Lung Cell Atlas-pilot award
- 1U54HL145608-01
- CZI Deep Visual Proteomics
- 1U54HL145608-01, U01HL148861-03
- 1) the Chan Zuckerberg Initiative, LLC Seed Network grant CZF2019-002438 “Lung Cell Atlas 1.0”; 2) R01 HL153312; 3) U19 AI135964; 4) P01 AG049665
- Netherlands Lung Foundation project nos. 5.1.14.020 and 4.1.18.226, LLC Seed Network grant CZF2019-002438 “Lung Cell Atlas 1.0”
- grant number 2019-002438 from the Chan Zuckerberg Foundation, by the Helmholtz Association’s Initiative and Networking Fund through Helmholtz AI [ZT-I-PF-5-01] and by the Bavarian Ministry of Science and the Arts in the framework of the Bavarian Research Association “ForInter” (Interaction of human brain cells)
- 1 U01 HL14555-01, R01 HL123766-04
- NIH U54 AG075931, 5R01 HL146519
Collapse
|
research-article |
2 |
277 |
23
|
Reif DM, Martin MT, Tan SW, Houck KA, Judson RS, Richard AM, Knudsen TB, Dix DJ, Kavlock RJ. Endocrine profiling and prioritization of environmental chemicals using ToxCast data. ENVIRONMENTAL HEALTH PERSPECTIVES 2010; 118:1714-20. [PMID: 20826373 PMCID: PMC3002190 DOI: 10.1289/ehp.1002180] [Citation(s) in RCA: 271] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 09/08/2010] [Indexed: 05/19/2023]
Abstract
BACKGROUND The prioritization of chemicals for toxicity testing is a primary goal of the U.S. Environmental Protection Agency (EPA) ToxCast™ program. Phase I of ToxCast used a battery of 467 in vitro, high-throughput screening assays to assess 309 environmental chemicals. One important mode of action leading to toxicity is endocrine disruption, and the U.S. EPA's Endocrine Disruptor Screening Program (EDSP) has been charged with screening pesticide chemicals and environmental contaminants for their potential to affect the endocrine systems of humans and wildlife. OBJECTIVE The goal of this study was to develop a flexible method to facilitate the rational prioritization of chemicals for further evaluation and demonstrate its application as a candidate decision-support tool for EDSP. METHODS Focusing on estrogen, androgen, and thyroid pathways, we defined putative endocrine profiles and derived a relative rank or score for the entire ToxCast library of 309 unique chemicals. Effects on other nuclear receptors and xenobiotic metabolizing enzymes were also considered, as were pertinent chemical descriptors and pathways relevant to endocrine-mediated signaling. RESULTS Combining multiple data sources into an overall, weight-of-evidence Toxicological Priority Index (ToxPi) score for prioritizing further chemical testing resulted in more robust conclusions than any single data source taken alone. CONCLUSIONS Incorporating data from in vitro assays, chemical descriptors, and biological pathways in this prioritization schema provided a flexible, comprehensive visualization and ranking of each chemical's potential endocrine activity. Importantly, ToxPi profiles provide a transparent visualization of the relative contribution of all information sources to an overall priority ranking. The method developed here is readily adaptable to diverse chemical prioritization tasks.
Collapse
|
research-article |
15 |
271 |
24
|
Tobias JA, Sheard C, Pigot AL, Devenish AJM, Yang J, Sayol F, Neate-Clegg MHC, Alioravainen N, Weeks TL, Barber RA, Walkden PA, MacGregor HEA, Jones SEI, Vincent C, Phillips AG, Marples NM, Montaño-Centellas FA, Leandro-Silva V, Claramunt S, Darski B, Freeman BG, Bregman TP, Cooney CR, Hughes EC, Capp EJR, Varley ZK, Friedman NR, Korntheuer H, Corrales-Vargas A, Trisos CH, Weeks BC, Hanz DM, Töpfer T, Bravo GA, Remeš V, Nowak L, Carneiro LS, Moncada R AJ, Matysioková B, Baldassarre DT, Martínez-Salinas A, Wolfe JD, Chapman PM, Daly BG, Sorensen MC, Neu A, Ford MA, Mayhew RJ, Fabio Silveira L, Kelly DJ, Annorbah NND, Pollock HS, Grabowska-Zhang AM, McEntee JP, Carlos T Gonzalez J, Meneses CG, Muñoz MC, Powell LL, Jamie GA, Matthews TJ, Johnson O, Brito GRR, Zyskowski K, Crates R, Harvey MG, Jurado Zevallos M, Hosner PA, Bradfer-Lawrence T, Maley JM, Stiles FG, Lima HS, Provost KL, Chibesa M, Mashao M, Howard JT, Mlamba E, Chua MAH, Li B, Gómez MI, García NC, Päckert M, Fuchs J, Ali JR, Derryberry EP, Carlson ML, Urriza RC, Brzeski KE, Prawiradilaga DM, Rayner MJ, Miller ET, Bowie RCK, Lafontaine RM, Scofield RP, Lou Y, Somarathna L, Lepage D, Illif M, Neuschulz EL, Templin M, Dehling DM, et alTobias JA, Sheard C, Pigot AL, Devenish AJM, Yang J, Sayol F, Neate-Clegg MHC, Alioravainen N, Weeks TL, Barber RA, Walkden PA, MacGregor HEA, Jones SEI, Vincent C, Phillips AG, Marples NM, Montaño-Centellas FA, Leandro-Silva V, Claramunt S, Darski B, Freeman BG, Bregman TP, Cooney CR, Hughes EC, Capp EJR, Varley ZK, Friedman NR, Korntheuer H, Corrales-Vargas A, Trisos CH, Weeks BC, Hanz DM, Töpfer T, Bravo GA, Remeš V, Nowak L, Carneiro LS, Moncada R AJ, Matysioková B, Baldassarre DT, Martínez-Salinas A, Wolfe JD, Chapman PM, Daly BG, Sorensen MC, Neu A, Ford MA, Mayhew RJ, Fabio Silveira L, Kelly DJ, Annorbah NND, Pollock HS, Grabowska-Zhang AM, McEntee JP, Carlos T Gonzalez J, Meneses CG, Muñoz MC, Powell LL, Jamie GA, Matthews TJ, Johnson O, Brito GRR, Zyskowski K, Crates R, Harvey MG, Jurado Zevallos M, Hosner PA, Bradfer-Lawrence T, Maley JM, Stiles FG, Lima HS, Provost KL, Chibesa M, Mashao M, Howard JT, Mlamba E, Chua MAH, Li B, Gómez MI, García NC, Päckert M, Fuchs J, Ali JR, Derryberry EP, Carlson ML, Urriza RC, Brzeski KE, Prawiradilaga DM, Rayner MJ, Miller ET, Bowie RCK, Lafontaine RM, Scofield RP, Lou Y, Somarathna L, Lepage D, Illif M, Neuschulz EL, Templin M, Dehling DM, Cooper JC, Pauwels OSG, Analuddin K, Fjeldså J, Seddon N, Sweet PR, DeClerck FAJ, Naka LN, Brawn JD, Aleixo A, Böhning-Gaese K, Rahbek C, Fritz SA, Thomas GH, Schleuning M. AVONET: morphological, ecological and geographical data for all birds. Ecol Lett 2022; 25:581-597. [PMID: 35199922 DOI: 10.1111/ele.13898] [Show More Authors] [Citation(s) in RCA: 261] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/10/2021] [Accepted: 09/10/2021] [Indexed: 01/02/2023]
Abstract
Functional traits offer a rich quantitative framework for developing and testing theories in evolutionary biology, ecology and ecosystem science. However, the potential of functional traits to drive theoretical advances and refine models of global change can only be fully realised when species-level information is complete. Here we present the AVONET dataset containing comprehensive functional trait data for all birds, including six ecological variables, 11 continuous morphological traits, and information on range size and location. Raw morphological measurements are presented from 90,020 individuals of 11,009 extant bird species sampled from 181 countries. These data are also summarised as species averages in three taxonomic formats, allowing integration with a global phylogeny, geographical range maps, IUCN Red List data and the eBird citizen science database. The AVONET dataset provides the most detailed picture of continuous trait variation for any major radiation of organisms, offering a global template for testing hypotheses and exploring the evolutionary origins, structure and functioning of biodiversity.
Collapse
|
Letter |
3 |
261 |
25
|
Pearce OMT, Delaine-Smith RM, Maniati E, Nichols S, Wang J, Böhm S, Rajeeve V, Ullah D, Chakravarty P, Jones RR, Montfort A, Dowe T, Gribben J, Jones JL, Kocher HM, Serody JS, Vincent BG, Connelly J, Brenton JD, Chelala C, Cutillas PR, Lockley M, Bessant C, Knight MM, Balkwill FR. Deconstruction of a Metastatic Tumor Microenvironment Reveals a Common Matrix Response in Human Cancers. Cancer Discov 2018; 8:304-319. [PMID: 29196464 PMCID: PMC5837004 DOI: 10.1158/2159-8290.cd-17-0284] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/08/2017] [Accepted: 11/28/2017] [Indexed: 12/21/2022]
Abstract
We have profiled, for the first time, an evolving human metastatic microenvironment by measuring gene expression, matrisome proteomics, cytokine and chemokine levels, cellularity, extracellular matrix organization, and biomechanical properties, all on the same sample. Using biopsies of high-grade serous ovarian cancer metastases that ranged from minimal to extensive disease, we show how nonmalignant cell densities and cytokine networks evolve with disease progression. Multivariate integration of the different components allowed us to define, for the first time, gene and protein profiles that predict extent of disease and tissue stiffness, while also revealing the complexity and dynamic nature of matrisome remodeling during development of metastases. Although we studied a single metastatic site from one human malignancy, a pattern of expression of 22 matrisome genes distinguished patients with a shorter overall survival in ovarian and 12 other primary solid cancers, suggesting that there may be a common matrix response to human cancer.Significance: Conducting multilevel analysis with data integration on biopsies with a range of disease involvement identifies important features of the evolving tumor microenvironment. The data suggest that despite the large spectrum of genomic alterations, some human malignancies may have a common and potentially targetable matrix response that influences the course of disease. Cancer Discov; 8(3); 304-19. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 253.
Collapse
|
research-article |
7 |
251 |