F. S. Collins, G. M. Gray, and J. R. Bucher, Toxicology. Transforming environmental health protection. Science, vol.319, 2008.

J. Rockström, W. Steffen, K. Noone, A. Persson, F. S. Chapin et al., A safe operating space for humanity, Nature, vol.461, pp.472-475, 2009.

, A provokative review proposing finite planetary boundaries, including chemical pollution, that must not be transgressed by human activities if we are to avoid unacceptable changes to the environment

E. F. Nuwaysir, M. Bittner, J. Trent, J. C. Barrett, and C. A. Afshari, Microarrays and toxicology: the advent of toxicogenomics, Mol Carcinog, vol.24, pp.153-159, 1999.

, Applications of Toxicogenomic Technologies to Predictive Toxicology and Risk Assessment, 2007.

W. M. Haschek, C. G. Rosseaux, and M. A. Wallig, Haschek and Rousseaux's handbook of toxicological pathology, vol.3, 2013.

. I. **6, N. Moffat, S. Chepelev, J. Labib, B. Bourdon-lacombe et al., Comparison of toxicogenomics and traditional approaches to inform mode of action and points of departure in human health risk assessment of benzo[a]pyrene in drinking water, Seminal publication introducing transcriptomic as a starting point for benchmark dose analysis in human health risk assessment, vol.45, 2015.

V. Makarov and A. Gorlin, Computational method for discovery of biomarker signatures from large, complex data sets, Comput Biol Chem, vol.76, pp.161-168, 2018.

F. A. Tilton, S. C. Tilton, T. K. Bammler, R. P. Beyer, P. L. Stapleton et al., Transcriptional impact of organophosphate and metal mixtures on olfaction: copper dominates the chlorpyrifos-induced response in adult zebrafish, Aquat Toxicol, vol.102, pp.205-215, 2011.

C. M. Hutchins, D. F. Simon, W. Zerges, and K. J. Wilkinson, Transcriptomic signatures in Chlamydomonas reinhardtii as Cd biomarkers in metal mixtures, Aquat Toxicol, vol.100, pp.120-127, 2010.

R. Altenburger, S. Scholz, M. Schmitt-jansen, W. Busch, and B. I. Escher, Mixture toxicity revisited from a toxicogenomic perspective, Environ Sci Technol, vol.46, 2012.

T. Svingen and A. M. Vinggaard, The risk of chemical cocktail effects and how to deal with the issue, J Epidemiol Community Health, vol.70, pp.322-323, 2016.

A. Kortenkamp and M. Faust, Regulate to reduce chemical mixture risk, Science, vol.361, pp.224-226, 2018.

Y. Song, J. Asselman, K. A. De-schamphelaere, B. Salbu, and K. E. Tollefsen, Deciphering the Combined Effects of Environmental Stressors on Gene Transcription: A Conceptual Approach, Environ Sci Technol, vol.52, pp.5479-5489, 2016.

D. Sims, I. Sudbery, N. E. Ilott, A. Heger, and C. P. Ponting, Sequencing depth and coverage: key considerations in genomic analyses, Nat Rev Genet, vol.15, pp.121-132, 2014.

F. Izadi, H. N. Zarrini, G. Kiani, and N. B. Jelodar, A comparative analytical assay of gene regulatory networks inferred using microarray and RNA-seq datasets, Bioinformation, vol.12, pp.340-346, 2016.

L. Chen, F. Sun, X. Yang, Y. Jin, M. Shi et al., Correlation between RNA-Seq and microarrays results using TCGA data, Gene, vol.628, pp.200-204, 2017.

C. Wang, B. Gong, P. R. Bushel, J. Thierry-mieg, D. Thierry-mieg et al., The concordance between RNA-seq and microarray data depends on chemical treatment and transcript abundance, Nat Biotechnol, vol.32, 2014.

S. Linnarsson and S. A. Teichmann, Single-cell genomics: coming of age, Genome Biol, vol.17, p.97, 2016.

S. Audic and J. M. Claverie, The significance of digital gene expression profiles

, Genome Res, vol.7, pp.986-995, 1997.

M. Soumillon, D. Cacchiarelli, S. Semrau, A. Van-oudenaarden, and T. S. Mikkelsen, Characterization of directed differentiation by high-throughput single-cell RNA-Seq. bioRxiv, 2014.

A. S. Nikitina, V. V. Babenko, K. A. Babalyan, A. O. Vasiliev, A. V. Govorov et al., Primary candidate rna biomarker screening by RNA-seq for prostate cancer diagnostics

, Biomed Khim, vol.61, 2015.

V. Costa, M. Aprile, R. Esposito, and A. Ciccodicola, RNA-Seq and human complex diseases: recent accomplishments and future perspectives, Eur J Hum Genet, vol.21, pp.134-142, 2013.

T. Bischler, H. S. Tan, K. Nieselt, and C. M. Sharma, Differential RNA-seq (dRNA-seq) for annotation of transcriptional start sites and small RNAs in Helicobacter pylori, Methods, vol.86, pp.89-101, 2015.

R. Yin, L. Gu, M. Li, C. Jiang, T. Cao et al., Gene expression profiling analysis of bisphenol A-induced perturbation in biological processes in ER-negative HEK293 cells, PLoS One, vol.9, 2014.

T. Urushidani, Prediction of hepatotoxicity based on the toxicogenomics database, in Hepatoxicity: from genomics to in vitro and in vivo models, 2008.

J. Quackenbush, Microarray data normalization and transformation, Nat Genet, vol.32

. Suppl, , pp.496-501, 2002.

N. Mah, A. Thelin, T. Lu, S. Nikolaus, T. Kühbacher et al., A comparison of oligonucleotide and cDNA-based microarray systems, Physiol Genomics, vol.16, 2004.

. G. **29 and . Miller, Improving reproducibility in toxicology, Toxicol Sci, vol.139, 2014.

, A thoughtful editorial arguing for data-driven rather than agenda-driven science in toxicology, with a keen focus on minimising biases in experimental design and data interpretations

C. A. Poland, M. R. Miller, R. Duffin, and F. Cassee, The elephant in the room: reproducibility in toxicology, Part Fibre Toxicol, vol.11, p.42, 2014.

F. Niu, D. C. Wang, J. Lu, W. Wu, and X. Wang, Potentials of single-cell biology in identification and validation of disease biomarkers, J Cell Mol Med, vol.20, pp.1789-1795, 2016.

.. P. *32, T. Dalerba, D. Kalisky, P. S. Sahoo, M. E. Rajendran et al., Single-cell dissection of transcriptional heterogeneity in human colon tumors, Nat Biotechnol, vol.29, 2011.

R. A. Gibbs, G. M. Weinstock, M. L. Metzker, D. M. Muzny, E. J. Sodergren et al., Genome sequence of the Brown Norway rat yields insights into mammalian evolution, Nature, vol.428, pp.493-521, 2004.

W. Mattes, K. Davis, E. Fabian, J. Greenhaw, M. Herold et al., Detection of hepatotoxicity potential with metabolite profiling (metabolomics) of rat plasma, Toxicol Lett, vol.230, pp.467-478, 2014.

L. Luo, S. Schomaker, C. Houle, J. Aubrecht, and J. L. Colangelo, Evaluation of serum bile acid profiles as biomarkers of liver injury in rodents, Toxicol Sci, vol.137, pp.12-25, 2014.

J. H. Shannahan, M. C. Schladweiler, J. H. Richards, A. D. Ledbetter, A. J. Ghio et al., Pulmonary oxidative stress, inflammation, and dysregulated iron homeostasis in rat models of cardiovascular disease, J Toxicol Environ Health A, vol.73, pp.641-656, 2010.

T. A. Clayton, J. C. Lindon, O. Cloarec, H. Antti, C. Charuel et al., Pharmaco-metabonomic phenotyping and personalized drug treatment, Nature, vol.440, 2006.

T. Uehara, A. Ono, T. Maruyama, I. Kato, H. Yamada et al., The Japanese toxicogenomics project: application of toxicogenomics, Mol Nutr Food Res, vol.54, pp.218-227, 2010.

M. S. Rye, M. F. Bhutta, M. T. Cheeseman, D. Burgner, J. M. Blackwell et al., Unraveling the genetics of otitis media: from mouse to human and back again, Mamm Genome, vol.22, pp.66-82, 2011.

E. M. Blais, K. D. Rawls, B. V. Dougherty, Z. I. Li, G. L. Kolling et al., Reconciled rat and human metabolic networks for comparative toxicogenomics and biomarker predictions, Nat Commun, vol.8, 2017.

. A. **41, C. J. Davis, R. J. Grondin, D. Johnson, B. L. Sciaky et al., The Comparative Toxicogenomics Database: update 2017, Nucleic Acids Res, vol.45, 2017.

, Guidance on information requirements and chemical safety assessment, European Chemicals Agency, vol.8, 2012.

K. De-raat and . Science, Understanding the importance of assessment factors in finding safe human exposure levels, ECHA Newsletter, vol.1, 2014.

K. Raja, M. Patrick, Y. Gao, D. Madu, Y. Yang et al., A review of recent advancement in integrating omics data with literature mining towards biomedical discoveries, Int J Genomics, vol.2017, 2017.

M. D. Wilkinson, M. Dumontier, I. J. Aalbersberg, G. Appleton, M. Axton et al., The FAIR Guiding Principles for scientific data management and stewardship, Sci Data, vol.3, 2016.

T. Barrett, S. E. Wilhite, P. Ledoux, C. Evangelista, I. F. Kim et al., NCBI GEO: archive for functional genomics data sets--update, Nucleaic Acids Res, vol.41, pp.991-995, 2013.

N. Kolesnikov, E. Hastings, M. Keays, O. Melnichuk, Y. A. Tang et al., ArrayExpress update--simplifying data submissions, Nucleaic Acids Res, vol.43, pp.1113-1116, 2015.

A. P. Davis, C. J. Grondin, K. Lennon-hopkins, C. Saraceni-richards, D. Sciaky et al., The Comparative Toxicogenomics Database's 10th year anniversary: update 2015, Nucleaic Acids Res, vol.43, 2015.

D. M. Hendrickx, H. J. Aerts, F. Caiment, D. Clark, T. M. Ebbels et al., diXa: a data infrastructure for chemical safety assessment, Bioinformatics, vol.3, pp.1505-1507, 2015.

C. Hardt, M. E. Beber, A. Rasche, A. Kamburov, D. G. Hebels et al., ToxDB: pathway-level interpretation of drug-treatment data, Database (Oxford), vol.2016, 2016.

I. A. Lea, H. Gong, A. Paleja, A. Rashid, and J. Fostel, CEBS: a comprehensive annotated database of toxicological data, Nucleaic Acids Res, vol.45, 2017.

H. G. Roider, N. Pavlova, I. Kirov, S. Slavov, Z. Uzunov et al., Drug2Gene: an exhaustive resource to explore effectively the drug-target relation network, BMC Bioinformatics, vol.15, p.68, 2014.

A. M. Richard, R. S. Judson, K. A. Houck, C. M. Grulke, P. Volarath et al., ToxCast Chemical Landscape: Paving the Road to 21st Century Toxicology, Chem Res Toxicol, vol.29, pp.1225-1251, 2016.

G. W. Miller, Data sharing in toxicology: beyond show and tell, Toxicol Sci, vol.143, pp.3-5, 2015.

T. A. Darde, P. Gaudriault, R. Beranger, C. Lancien, A. Caillarec-joly et al., TOXsIgN: a cross-species repository for toxicogenomic signatures, Bioinformatics, vol.34, pp.2116-2122, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01863047

.. Y. *56, N. Igarashi, T. Nakatsu, A. Yamashita, Y. Ono et al., This paper presents Open TG-GATES, a collaborative project between the National Institute of Biomedical Innovation (NIB), the NIES and around 15 pharmaceutical companies that sought to study 150 chemicals and their transcriptional responses in rat tissues, Nucleaic Acids Res, vol.43, pp.1025-1044, 2006.

, Set up by the National Institute of Environmental Sciences (NIES, USA), Drugmatrix aimed at studying transcriptional responses in rats of 376 compounds in five different tissues at several doses and multiple exposure times

C. W. Tung, C. C. Wang, S. S. Wang, and P. Lin, ChemDIS-Mixture: an online tool for analyzing potential interaction effects of chemical mixtures, Sci Rep, vol.8, p.10047, 2018.

J. Nyström-persson, Y. Natsume-kitatani, Y. Igarashi, D. Satoh, and K. Mizuguchi, Interactive Toxicogenomics: Gene set discovery, clustering and analysis in Toxygates, Sci Rep, vol.7, p.1390, 2017.

J. Kringelum, S. K. Kjaerulff, S. Brunak, O. Lund, T. I. Oprea et al., ChemProt-3.0: a global chemical biology diseases mapping. Database (Oxford), p.2016, 2016.

L. Xing, L. Wu, Y. Liu, N. Ai, X. Lu et al., LTMap: a web server for assessing the potential liver toxicity by genome-wide transcriptional expression data, J Appl Toxicol, vol.34, pp.805-809, 2014.

M. Römer, L. Backert, J. Eichner, and A. Zell, ToxDBScan: Large-scale similarity screening of toxicological databases for drug candidates, Int J Mol Sci, vol.15, 2014.

J. Lamb, E. D. Crawford, D. Peck, J. W. Modell, I. C. Blat et al., The Connectivity Map: using geneexpression signatures to connect small molecules, genes, and disease, Science, vol.313, 2006.

C. Liu, J. Su, F. Yang, K. Wei, J. Ma et al., Compound signature detection on LINCS L1000 big data. Molecular bioSystems, vol.11, 2015.

J. Setoain, M. Franch, M. Martinez, D. Tabas-madrid, C. O. Sorzano et al., NFFinder: an online bioinformatics tool for searching similar transcriptomics experiments in the context of drug repositioning, Nucleaic Acids Res, vol.43, pp.193-199, 2015.