T. Gao and . Substances, EPA Has Increased Efforts to Assess and Control Chemicals but Could Strengthen Its Approach, Governement Accountability Office, 2013.

I. Navarro, A. De-la-torre, P. Sanz, C. Fernández, G. Carbonell et al., Environmental risk assessment of perfluoroalkyl substances and halogenated flame retardants released from biosolids-amended soils, Chemosphere, vol.210, pp.147-155, 2018.

B. M. Sharma, J. Be?anová, M. Scheringer, A. Sharma, G. K. Bharat et al., Health and ecological risk assessment of emerging contaminants (pharmaceuticals, personal care products, and artificial sweeteners) in surface and groundwater (drinking water) in the Ganges River Basin, Sci. Total Environ, vol.646, pp.1459-1467, 2019.

O. H. Force, One Health: A New Professional Imperative, A.V.M. Association, 2008.

R. Truhaut, Ecotoxicology: objectives, principles and perspectives, Ecotoxicol. Environ. Saf, vol.1, issue.2, pp.151-173, 1977.

R. T. Corlett, The anthropocene concept in ecology and conservation, Trends Ecol. Evol, vol.30, issue.1, pp.36-41, 2015.

A. Gredelj, A. Barausse, L. Grechi, and L. Palmeri, Deriving predicted no-effect concentrations (PNECs) for emerging contaminants in the river Po, Italy, using three approaches: assessment factor, species sensitivity distribution and AQUATOX ecosystem modelling, Environ. Int, vol.119, pp.66-78, 2018.

E. D. Goldberg, V. T. Bowen, J. W. Farrington, G. Harvey, J. H. Martin et al., The mussel watch, Environ. Conserv, vol.5, issue.2, pp.101-125, 1978.

J. Armengaud, J. Trapp, O. Pible, O. Geffard, A. Chaumot et al., Nonmodel organisms, a species endangered by proteogenomics, J. Proteomics, vol.105, pp.5-18, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02600746

A. Bjørnstad, B. K. Larsen, A. Skadsheim, M. B. Jones, and O. K. Andersen, The potential of ecotoxicoproteomics in environmental monitoring: Biomarker profiling in mussel plasma using proteinchip array technology, J. Toxicol. Environ. Health A, vol.69, issue.1-2, pp.77-96, 2006.

T. Monsinjon and T. Knigge, Proteomic applications in ecotoxicology, Proteomics, vol.7, pp.2997-3009, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02022074

M. F. Lemos, A. M. Soares, A. C. Correia, and A. C. Esteves, Proteins in ecotoxicology -how, why and why not?, Proteomics, vol.10, pp.873-887, 2010.

L. Tomanek, Environmental proteomics: changes in the proteome of marine organisms in response to environmental stress, pollutants, infection, symbiosis, and development, Annu. Rev. Mar. Sci, vol.3, pp.373-399, 2011.

B. C. Sanchez, K. Ralston-hooper, and M. S. Sepulveda, Review of recent proteomic applications in aquatic toxicology, Environ. Toxicol. Chem, vol.30, issue.2, pp.274-282, 2011.

C. J. Martyniuk, S. Alvarez, and N. D. Denslow, DIGE and iTRAQ as biomarker discovery tools in aquatic toxicology, Ecotoxicol. Environ. Saf, vol.76, pp.3-10, 2012.

F. Silvestre, V. Gillardin, and J. Dorts, Proteomics to assess the role of phenotypic plasticity in aquatic organisms exposed to pollution and global warming, Integr. Comp. Biol, vol.52, issue.5, pp.681-694, 2012.

J. Trapp, J. Armengaud, A. Salvador, A. Chaumot, and O. Geffard, Next-generation proteomics: toward customized biomarkers for environmental biomonitoring, Environ. Sci. Technol, vol.48, issue.23, pp.13560-13572, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01230133

R. D. Handy and M. H. Depledge, Physiological responses: their measurement and use as environmental biomarkers in ecotoxicology, Ecotoxicology, vol.8, issue.5, pp.329-349, 1999.

J. Hellou, Behavioural ecotoxicology, an "early warning" signal to assess environmental quality, Environ. Sci. Pollut. Res, vol.18, issue.1, pp.1-11, 2011.

F. Gagné, Biochemical Ecotoxicology: Principles and Methods, 2014.

F. Sánchez-bayo and K. Goka, Evaluation of suitable endpoints for assessing the impacts of toxicants at the community level, Ecotoxicology, vol.21, issue.3, pp.667-680, 2012.

D. Moreels, P. Lodewijks, H. Zegers, E. Rurangwa, N. Vromant et al., Effect of short-term exposure to methyl-tert-butyl ether and tert-butyl alcohol on the hatch rate and development of the African catfish, Clarias gariepinus, Environ. Toxicol. Chem, vol.25, issue.2, pp.514-519, 2006.

M. Boudreau, S. C. Courtenay, D. L. Maclatchy, C. H. Bérubé, L. M. Hewitt et al., Morphological abnormalities during early-life development of the estuarine mummichog, Fundulus heteroclitus, as an indicator of androgenic and antiandrogenic endocrine disruption, Aquat. Toxicol, vol.71, issue.4, pp.357-369, 2005.

J. C. Dewitt, D. S. Millsap, R. L. Yeager, S. S. Heise, D. W. Sparks et al., External heart deformities in passerine birds exposed to environmental mixtures of polychlorinated biphenyls during development, Environ. Toxicol. Chem, vol.25, issue.2, pp.541-551, 2006.

R. Coulaud, O. Geffard, B. Xuereb, E. Lacaze, H. Quéau et al., situ feeding assay with Gammarus fossarum (Crustacea): Modelling the influence of confounding factors to improve water quality biomonitoring, vol.45, pp.6417-6429, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01920247

M. M. Watts, D. Pascoe, and K. Carroll, Survival and precopulatory behaviour of Gammarus pulex (L.) exposed to two xenoestrogens, Water Res, vol.35, issue.10, pp.2347-2352, 2001.

T. B. Hayes, V. Khoury, A. Narayan, M. Nazir, A. Park et al., Atrazine induces complete feminization and chemical castration in male African clawed frogs (< em > Xenopus laevis < /em >), Proc. Natl. Acad. Sci, vol.107, issue.10, pp.4612-4617, 2010.

A. Jemec, D. Drobne, T. Ti?ler, and K. Sep?i?, Biochemical biomarkers in environmental studies-lessons learnt from enzymes catalase, glutathione S-transferase and cholinesterase in two crustacean species, Environ. Sci. Pollut. Res, vol.17, issue.3, pp.571-581, 2010.

P. A. Bahamonde, A. Feswick, M. A. Isaacs, K. R. Munkittrick, and C. J. Martyniuk, Defining the role of omics in assessing ecosystem health: Perspectives from the Canadian environmental monitoring program, Environ. Toxicol. Chem, vol.35, issue.1, pp.20-35, 2016.

X. Zhang, P. Xia, P. Wang, J. Yang, and D. J. Baird, Omics advances in ecotoxicology, Environ. Sci. Technol, vol.52, issue.7, pp.3842-3851, 2018.

T. S. Galloway, R. J. Brown, M. A. Browne, A. Dissanayake, D. Lowe et al., A multibiomarker approach to environmental assessment, Environ. Sci. Technol, vol.38, issue.6, pp.1723-1731, 2004.

M. C. Celander, J. V. Goldstone, N. D. Denslow, T. Iguchi, P. Kille et al., Species extrapolation for the 21st century, Environ. Toxicol. Chem, vol.30, issue.1, pp.52-63, 2011.

S. Jobling and C. R. Tyler, Endocrine disruption in wild freshwater fish, Pure Appl. Chem, p.2219, 2003.

J. E. Wright and G. E. Spates, A new approach in integrated control: insect juvenile hormone plus a hymenopteran parasite against the stable fly, Science, vol.178, issue.4067, p.1292, 1972.

C. J. Martyniuk, K. J. Kroll, N. J. Doperalski, D. S. Barber, and N. D. Denslow, Environmentally relevant exposure to 17?-ethinylestradiol affects the telencephalic proteome of male fathead minnows, Aquat. Toxicol, vol.98, issue.4, pp.344-353, 2010.

K. A. Kidd, P. J. Blanchfield, K. H. Mills, V. P. Palace, R. E. Evans et al., Collapse of a fish population after exposure to a synthetic estrogen, Proc. Natl. Acad. Sci. U. S. A, vol.104, issue.21, pp.8897-8901, 2007.

M. Konus, C. Koy, S. Mikkat, M. Kreutzer, R. Zimmermann et al., Molecular adaptations of Helicoverpa armigera midgut tissue under pyrethroid insecticide stress characterized by differential proteome analysis and enzyme activity assays, Compar. Biochem. Physiol. D, vol.8, issue.2, pp.152-162, 2013.

C. Guarino, B. Conte, V. Spada, S. Arena, R. Sciarrillo et al., Proteomic analysis of eucalyptus leaves unveils putative mechanisms involved in the plant response to a real condition of soil contamination by multiple heavy metals in the presence or absence of mycorrhizal/rhizobacterial additives, Environ. Sci. Technol, vol.48, issue.19, pp.11487-11496, 2014.

J. Wang, Y. Wei, D. Wang, L. L. Chan, and J. Dai, Proteomic study of the effects of complex environmental stresses in the livers of goldfish (Carassius auratus) that inhabit Gaobeidian Lake, Ecotoxicology, vol.17, issue.3, pp.213-220, 2008.

J. L. Shepard, B. Olsson, M. Tedengren, and B. P. Bradley, Protein expression signatures identified in Mytilus edulis exposed to PCBs, copper and salinity stress, Mar. Environ. Res, vol.50, issue.1, pp.337-340, 2000.

J. Mi, A. Orbea, N. Syme, M. Ahmed, M. P. Cajaraville et al., Peroxisomal proteomics, a new tool for risk assessment of peroxisome proliferating pollutants in the marine environment, Proteomics, vol.5, issue.15, pp.3954-3965, 2005.

I. Apraiz, M. P. Cajaraville, and S. , Peroxisomal proteomics: Biomonitoring in mussels after the Prestige's oil spill, Mar. Pollut. Bull, vol.58, issue.12, pp.1815-1826, 2009.

E. Gianazza, R. Wait, A. Sozzi, S. Regondi, D. Saco et al., Growth and protein profile changes in Lepidium sativum L. plantlets exposed to cadmium, Environ. Exp. Botany, vol.59, issue.2, pp.179-187, 2007.

T. Karuppanapandian, S. J. Rhee, E. J. Kim, B. K. Han, O. A. Hoekenga et al., Proteomic analysis of differentially expressed proteins in the roots of Columbia-0 and Landsberg erecta ecotypes of Arabidopsis thaliana in response to aluminum toxicity, Can. J. Plant Sci, vol.92, issue.7, pp.1267-1282, 2012.

N. Ahsan, S. H. Lee, D. G. Lee, H. Lee, S. W. Lee et al., Physiological and protein profiles alternation of germinating rice seedlings exposed to acute cadmium toxicity, Comptes Rendus Biol, vol.330, issue.10, pp.735-746, 2007.

C. S. Liao, J. H. Yen, and Y. S. Wang, Growth inhibition in Chinese cabbage (Brassica rapa var. chinensis) growth exposed to di-n-butyl phthalate, J. Hazard. Mater, vol.163, issue.2-3, pp.625-631, 2009.

L. Fan, A. Wang, and Y. Wu, Comparative proteomic identification of the hemocyte response to cold stress in white shrimp, Litopenaeus vannamei, J. Proteomics, vol.80, pp.196-206, 2013.

C. Ji, H. Wu, L. Wei, J. Zhao, H. Lu et al., Proteomic and metabolomic analysis of earthworm Eisenia fetida exposed to different concentrations of 2,2?,4,4?-tetrabromodiphenyl ether, J. Proteomics, vol.91, pp.405-416, 2013.

M. A. Pierrard, P. Kestemont, N. T. Phuong, M. P. Tran, E. Delaive et al., Proteomic analysis of blood cells in fish exposed to chemotherapeutics: evidence for long term effects, J. Proteomics, vol.75, issue.8, pp.2454-2467, 2012.

Q. H. Zhang, L. Huang, Y. Zhang, C. H. Ke, and H. Q. Huang, Proteomic approach for identifying gonad differential proteins in the oyster (Crassostrea angulata) following food-chain contamination with HgCl < inf > 2 < /inf >, J. Proteom, vol.94, pp.37-53, 2013.

R. Guo, X. Ding, W. Xiong, X. Zhong, W. Liang et al., Earthworms as agents for ecotoxicity in roxarsone-contaminated soil ecosystem: a modeling study of ultrastructure and proteomics, Environ. Sci. Pollut. Res, vol.22, issue.16, pp.12435-12449, 2015.

M. F. Lemos, A. Esteves, B. Samyn, I. Timperman, J. Van-beeumen et al., Protein differential expression induced by endocrine disrupting compounds in a terrestrial isopod, Chemosphere, vol.79, issue.5, pp.570-576, 2010.

K. J. Ralston-hooper, M. E. Turner, E. J. Soderblom, D. Villeneuve, G. T. Ankley et al., Application of a label-free, gel-free quantitative proteomics method for ecotoxicological studies of small fish species, Environ. Sci. Technol, vol.47, issue.2, pp.1091-1100, 2013.

C. J. Martyniuk and S. Alvarez, Proteome analysis of the fathead minnow (Pimephales promelas) reproductive testes, J. Proteomics, vol.79, pp.28-42, 2013.

M. Borgatta, C. Hernandez, L. A. Decosterd, N. Chèvre, and P. Waridel, Shotgun ecotoxicoproteomics of Daphnia pulex: biochemical effects of the anticancer drug tamoxifen, J. Proteome Res, vol.14, issue.1, pp.279-291, 2015.

J. Trapp, C. Almunia, J. Gaillard, O. Pible, A. Chaumot et al., Proteogenomic insights into the core-proteome of female reproductive tissues from crustacean amphipods, J. Proteomics, vol.135, pp.51-61, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02604682

O. Pible and J. Armengaud, Improving the quality of genome, protein sequence, and taxonomy databases: a prerequisite for microbiome meta-omics 2.0, Proteomics, vol.15, issue.20, pp.3418-3423, 2015.

B. J. Haas, A. Papanicolaou, M. Yassour, M. Grabherr, P. D. Blood et al., De novo transcript sequence reconstruction from RNA-Seq: reference generation and analysis with Trinity, Nat. Protoc, vol.8, issue.8, 2013.

Y. Moreno, P. Gros, M. Tam, M. Segura, R. Valanparambil et al., Proteomic analysis of excretory-secretory products of heligmosomoides polygyrus assessed with next-generation sequencing transcriptomic information, PLoS Negl. Trop. Dis, vol.5, issue.10, p.1370, 2011.

G. Lopez-casado, P. A. Covey, P. A. Bedinger, L. A. Mueller, T. W. Thannhauser et al., Enabling proteomic studies with RNA-Seq: the proteome of tomato pollen as a test case, Proteomics, vol.12, issue.6, pp.761-774, 2012.

J. Trapp, O. Geffard, G. Imbert, J. C. Gaillard, A. H. Davin et al., Proteogenomics of gammarus fossarum to document the reproductive system of amphipods, Mol. Cell. Proteomics, vol.13, issue.12, pp.3612-3625, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02600748

J. Grossmann, H. Fernández, P. M. Chaubey, A. E. Valdés, V. Gagliardini et al., Proteogenomic analysis greatly expands the identification of proteins related to reproduction in the apogamous fern Dryopteris affinis ssp. affinis, Front, Plant Sci, vol.8, 2017.

A. Campos, G. Danielsson, A. P. Farinha, J. Kuruvilla, P. Warholm et al., Shotgun proteomics to unravel marine mussel (Mytilus edulis) response to longterm exposure to low salinity and propranolol in a Baltic Sea microcosm, J. Proteomics, vol.137, pp.97-106, 2016.

D. Gouveia, A. Chaumot, A. Charnot, H. Queau, J. Armengaud et al., Assessing the relevance of a multiplexed methodology for proteomic biomarker measurement in the invertebrate species Gammarus fossarum: a physiological and ecotoxicological study, Aquat. Toxicol, vol.190, pp.199-209, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01682339

P. V. Bondarenko, D. Chelius, and T. A. Shaler, Identification and relative quantitation of protein mixtures by enzymatic digestion followed by capillary reversed-phase liquid chromatography-tandem mass spectrometry, Anal. Chem, vol.74, issue.18, pp.4741-4749, 2002.

H. Liu, R. G. Sadygov, and J. R. Yates-3rd, A model for random sampling and estimation of relative protein abundance in shotgun proteomics, Anal. Chem, vol.76, issue.14, pp.4193-4201, 2004.

J. Trapp, J. Armengaud, O. Pible, J. C. Gaillard, K. Abbaci et al., Proteomic investigation of male Gammarus fossarum, a freshwater crustacean, in response to endocrine disruptors, J. Proteome Res, vol.14, issue.1, pp.292-303, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02600747

Y. T. Chen, H. W. Chen, C. F. Wu, L. J. Chu, W. F. Chiang et al., Development of a multiplexed liquid chromatography multiple-reaction-monitoring mass spectrometry (LC-MRM/MS) method for evaluation of salivary proteins as oral cancer biomarkers, Mol. Cell. Proteomics, vol.16, issue.5, pp.799-811, 2017.

A. N. Hoofnagle, J. O. Becker, M. N. Oda, G. Cavigiolio, P. Mayer et al., Multiple-reaction monitoring-mass spectrometric assays can accurately measure the relative protein abundance in complex mixtures, Clin. Chem, vol.58, issue.4, pp.777-781, 2012.

M. A. Gillette and S. A. Carr, Quantitative analysis of peptides and proteins in biomedicine by targeted mass spectrometry, Nat. Methods, vol.10, p.28, 2012.

P. Picotti and R. Aebersold, Selected reaction monitoring-based proteomics: workflows, potential, pitfalls and future directions, Nat. Methods, vol.9, issue.6, pp.555-566, 2012.

R. Simon, G. Jubeaux, A. Chaumot, J. Lemoine, O. Geffard et al., Mass spectrometry assay as an alternative to the enzyme-linked immunosorbent assay test for biomarker quantitation in ecotoxicology: application to vitellogenin in Crustacea (Gammarus fossarum), J. Chromatogr. A, vol.1217, issue.31, pp.5109-5115, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02593846

G. Jubeaux, R. Simon, A. Salvador, H. Quéau, A. Chaumot et al., Vitellogenin-like proteins in the freshwater amphipod Gammarus fossarum (Koch, 1835): functional characterization throughout reproductive process, potential for use as an indicator of oocyte quality and endocrine disruption biomarker in males, Aquat. Toxicol, pp.72-82, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00697838

P. He, E. K. Matich, L. T. Yonkos, A. E. Friedman, G. E. Atilla-gokcumen et al., Mass spectrometry based detection of common vitellogenin peptides across fish species for assessing exposure to estrogenic compounds in aquatic environments, Sci. Total Environ, vol.646, pp.400-408, 2019.

D. Gouveia, A. Chaumot, A. Charnot, C. Almunia, A. Francois et al., Ecotoxico-proteomics for aquatic environmental monitoring: first in situ application of a new proteomics-based multibiomarker assay using caged amphipods, Environ. Sci. Technol, vol.51, issue.22, pp.13417-13426, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01663631

A. Charnot, D. Gouveia, J. Armengaud, C. Almunia, A. Chaumot et al., Multiplexed assay for protein quantitation in the invertebrate Gammarus fossarum by liquid chromatography coupled to tandem mass spectrometry, Anal. Bioanal. Chem, vol.409, issue.16, pp.3969-3991, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01539762

A. Charnot, D. Gouveia, S. Ayciriex, J. Lemoine, J. Armengaud et al., On-line solid phase extraction liquid chromatography-mass spectrometry method for multiplexed proteins quantitation in an ecotoxicology test specie: gammarus fossarum, J. Appl. Bioanal, vol.4, issue.3, pp.81-101, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01860702

V. Vidova and Z. Spacil, A review on mass spectrometry-based quantitative proteomics: Targeted and data independent acquisition, Anal. Chim. Acta, vol.964, pp.7-23, 2017.

A. Bertsch, S. Jung, A. Zerck, N. Pfeifer, S. Nahnsen et al., Optimal de novo design of MRM experiments for rapid assay development in targeted proteomics, J. Proteome Res, vol.9, issue.5, pp.2696-2704, 2010.

C. Rudén, J. Adams, M. Ågerstrand, T. C. Brock, V. Poulsen et al., Assessing the relevance of ecotoxicological studies for regulatory decision making, Integr. Environ. Assess. Manag, vol.13, issue.4, pp.652-663, 2016.

G. Jubeaux, F. Audouard-combe, R. Simon, R. Tutundjian, A. Salvador et al., Vitellogenin-like proteins among invertebrate species diversity: potential of proteomic mass spectrometry for biomarker development, Environ. Sci. Technol, vol.46, issue.11, pp.6315-6323, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00875198

C. Tsangaris, I. Hatzianestis, V. Catsiki, K. A. Kormas, E. Strogyloudi et al., Active biomonitoring in Greek coastal waters: application of the integrated biomarker response index in relation to contaminant levels in caged mussels, Sci. Total Environ, pp.359-365, 2011.

J. Besse, M. Coquery, C. Lopes, A. Chaumot, H. Budzinski et al., Caged Gammarus fossarum (Crustacea) as a robust tool for the characterization of bioavailable contamination levels in continental waters: towards the determination of threshold values, Water Res, vol.47, issue.2, pp.650-660, 2013.

L. Charron, O. Geffard, A. Chaumot, R. Coulaud, H. Queau et al., Effect of water quality and confounding factors on digestive enzyme activities in Gammarus fossarum, vol.20, pp.9044-9056, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01920248

L. Charron, O. Geffard, A. Chaumot, R. Coulaud, A. Jaffal et al., Influence of molting and starvation on digestive enzyme activities and energy storage in Gammarus fossarum, PLoS One, vol.9, issue.4, p.96393, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01070508

L. Charron, O. Geffard, A. Chaumot, R. Coulaud, A. Jaffal et al., Consequences of lower food intake on the digestive enzymes activities, the energy reserves and the reproductive outcome in Gammarus fossarum, vol.10, p.125154, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01181614

I. Apraiz, J. Mi, and S. , Identification of proteomic signatures of exposure to marine pollutants in mussels, Mol. Cell. Proteomics, vol.5, issue.7, pp.1274-1285, 2006.

B. Mcdonagh and D. Sheehan, Effect of oxidative stress on protein thiols in the blue mussel Mytilus edulis: proteomic identification of target proteins, Proteomics, vol.7, issue.18, pp.3395-3403, 2007.

T. Knigge, T. Monsinjon, and O. K. Andersen, Surface-enhanced laser desorption/ionization-time of flight-mass spectrometry approach to biomarker discovery in blue mussels (Mytilus edulis) exposed to polyaromatic hydrocarbons and heavy metals under field conditions, Proteomics, vol.4, issue.9, pp.2722-2727, 2004.
URL : https://hal.archives-ouvertes.fr/hal-02022087

A. Campos, S. Tedesco, V. Vasconcelos, and S. , Proteomic research in bivalves. Towards the identification of molecular markers of aquatic pollution, J. Proteomics, vol.75, issue.14, pp.4346-4359, 2012.

A. Campos, I. Apraiz, R. R. Da-fonseca, and S. , Shotgun analysis of the marine mussel Mytilus edulis hemolymph proteome and mapping the innate immunity elements, Proteomics, vol.15, pp.4021-4029, 2015.

P. Y. Kunz, C. Kienle, and A. Gerhardt, Gammarus spp. in aquatic ecotoxicology and water quality assessment: toward integrated multilevel tests, Rev. Environ. Contam. Toxicol, vol.205, pp.1-76, 2010.

J. Trapp, J. Armengaud, J. C. Gaillard, O. Pible, A. Chaumot et al., Highthroughput proteome dynamics for discovery of key proteins in sentinel species: unsuspected vitellogenins diversity in the crustacean Gammarus fossarum, J. Proteomics, vol.146, pp.207-214, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02604683

J. Trapp, D. Gouveia, C. Almunia, O. Pible, D. Esposti et al., Digging deeper into the pyriproxyfen-response of the amphipod gammarus fossarum with a next-generation ultra-highfield orbitrap analyser: new perspectives for environmental toxicoproteomics, Front. Environ. Sci, vol.6, issue.54, 2018.

E. Gismondi, J. P. Thomé, N. Urien, E. Uher, D. Baiwir et al., Ecotoxicoproteomic assessment of the functional alterations caused by chronic metallic exposures in gammarids, Environ. Pollut, vol.225, pp.428-438, 2017.

K. Roland, P. Kestemont, M. Dieu, M. Raes, and F. Silvestre, Integrated Biomarker Proteomic" index to assess the effects of freshwater pollutants in European eel peripheral blood mononuclear cells, J. Proteomics, vol.137, pp.83-96, 2016.

A. Rhoads and K. F. Au, PacBio Sequencing and its applications, vol.13, pp.278-289, 2015.

D. Branton, D. W. Deamer, A. Marziali, H. Bayley, S. A. Benner et al., The potential and challenges of nanopore sequencing, Nat. Biotechnol, vol.26, issue.10, pp.1146-1153, 2008.

J. D. Chapman, D. R. Goodlett, and C. D. Masselon, Multiplexed and data-independent tandem mass spectrometry for global proteome profiling, Mass Spectrom. Rev, p.33
URL : https://hal.archives-ouvertes.fr/hal-02084749

F. Meier, P. E. Geyer, S. Winter, J. Cox, and M. Mann, BoxCar acquisition method enables single-shot proteomics at a depth of 10,000 proteins in 100 minutes, Nat. Methods, vol.15, issue.6, pp.440-448, 2018.

B. Rougemont, S. Gallo, S. Ayciriex, R. Carrière, H. Hondermarck et al., Scout-MRM: multiplexed targeted mass spectrometry-based assay without retention time scheduling exemplified by Dickeya Dadantii proteomic analysis during plant infection, Anal. Chem, vol.89, issue.3, pp.1421-1426, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01518912

L. C. Gillet, P. Navarro, S. Tate, H. Rost, N. Selevsek et al., Targeted data extraction of the MS/MS spectra generated by dataindependent acquisition: a new concept for consistent and accurate proteome analysis, Mol. Cell. Proteomics, vol.11, issue.6, p.18, 2012.

L. Cid-barrio, F. Calderón-celis, P. Abásolo-linares, M. L. Fernández-sánchez, J. M. Costa-fernández et al., Advances in absolute protein quantification and quantitative protein mapping using ICP-MS, TrAC Trends Anal. Chem, vol.104, pp.148-159, 2018.

B. Campos and J. K. Colbourne, How omics technologies can enhance chemical safety regulation: perspectives from academia, government, and industry: the perspectives column is a regular series designed to discuss and evaluate potentially competing viewpoints and research findings on current environmental issues, Environ. Toxicol. Chem, vol.37, issue.5, pp.1252-1259, 2018.