, WHO. World malaria report, 2017.

D. Vlachou, T. Schlegelmilch, G. K. Christophides, and F. C. Kafatos, Functional genomic analysis of midgut epithelial responses in Anopheles during Plasmodium invasion, Curr Biol, vol.15, issue.13, pp.1185-95, 2005.

S. Blandin and E. A. Levashina, Mosquito immune responses against malaria parasites, Curr Opin Immunol, vol.16, issue.1, pp.16-20, 2004.

L. Akman-anderson, M. Olivier, and S. Luckhart, Induction of nitric oxide synthase and activation of signaling proteins in Anopheles mosquitoes by the malaria pigment, hemozoin, Infect Immun, vol.75, issue.8, pp.4012-4021, 2007.

S. A. Blandin, E. Marois, and E. A. Levashina, Antimalarial responses in Anopheles gambiae: from a complement-like protein to a complement-like pathway, Cell Host Microbe, vol.3, issue.6, pp.364-74, 2008.

L. C. Bartholomay and K. Michel, Mosquito immunobiology: the intersection of vector health and vector competence, Ann Rev Entomol, vol.63, pp.145-67, 2018.

. Ruiz, Epigenetics & Chromatin, vol.12, p.5, 2019.

L. C. Pollitt, J. T. Bram, S. Blanford, M. J. Jones, and A. F. Read, Existing infection facilitates establishment and density of malaria parasites in their mosquito vector, PLoS Pathog, vol.11, issue.7, p.1005003, 2015.

M. L. Simões and G. Dimopoulos, A mosquito mediator of parasite-induced immune priming, Trends Parasitol, vol.31, issue.9, pp.402-406, 2015.

L. Lambrechts, J. M. Chavatte, G. Snounou, and J. C. Koella, Environmental influence on the genetic basis of mosquito resistance to malaria parasites, Proc Biol Sci, vol.273, pp.1501-1507, 1593.

T. Lefevre, A. Vantaux, K. R. Dabire, K. Mouline, and A. Cohuet, Non-genetic determinants of mosquito competence for malaria parasites, PLoS Pathog, vol.9, issue.6, p.1003365, 2013.

R. Feil and M. F. Fraga, Epigenetics and the environment: emerging patterns and implications, Nat Rev Genet, vol.13, issue.2, pp.97-109, 2012.

P. Gut and E. Verdin, The nexus of chromatin regulation and intermediary metabolism, Nature, vol.502, issue.7472, pp.489-98, 2013.

T. Kouzarides, Chromatin modifications and their function, Cell, vol.128, issue.4, pp.693-705, 2007.

H. Yin, S. Sweeney, D. Raha, M. Snyder, and H. Lin, A high-resolution wholegenome map of key chromatin modifications in the adult Drosophila melanogaster, PLoS Genet, vol.7, issue.12, p.1002380, 2011.

S. H. Merkling, A. W. Bronkhorst, J. M. Kramer, G. J. Overheul, A. Schenck et al., The epigenetic regulator G9a mediates tolerance to RNA virus infection in Drosophila, PLoS Pathog, vol.11, issue.4, p.1004692, 2015.

I. Anreiter, J. M. Kramer, and M. B. Sokolowski, Epigenetic mechanisms modulate differences in Drosophila foraging behavior, Proc Natl Acad Sci, vol.114, issue.47, pp.12518-12541, 2017.

A. J. Dobson, M. Ezcurra, C. E. Flanagan, A. C. Summerfield, M. Piper et al., Nutritional programming of Lifespan by FOXO inhibition on sugar-rich diets, Cell Rep, vol.18, issue.2, pp.299-306, 2017.

F. Bantignies, C. Grimaud, S. Lavrov, M. Gabut, and G. Cavalli, Inheritance of Polycomb-dependent chromosomal interactions in Drosophila, Genes Dev, vol.17, pp.2406-2426, 2003.

R. Margueron and D. Reinberg, Chromatin structure and the inheritance of epigenetic information, Nat Rev Genet, vol.11, issue.4, pp.285-96, 2010.

E. I. Campos, J. M. Stafford, and D. Reinberg, Epigenetic inheritance: histone bookmarks across generations, Trends Cell Biol, vol.24, issue.11, pp.664-74, 2014.

H. Stroud, S. C. Su, S. Hrvatin, A. W. Greben, W. Renthal et al., Early-life gene expression in neurons modulates lasting epigenetic states, Cell, vol.171, issue.5, pp.1151-64, 2017.

R. T. Coleman and G. Struhl, Causal role for inheritance of H3K27me3 in maintaining the OFF state of a Drosophila HOX gene, Science, vol.356, issue.6333, p.8236, 2017.

A. J. Osborne and P. K. Dearden, A 'phenotypic hangover': the predictive adaptive response and multigenerational effects of altered nutrition on the transcriptome of Drosophila melanogaster, Environ Epigenet, vol.3, issue.4, p.19, 2017.

F. Ciabrelli, F. Comoglio, S. Fellous, B. Bonev, M. Ninova et al., Stable Polycomb-dependent transgenerational inheritance of chromatin states in Drosophila, Nat Genet, vol.49, issue.6, p.876, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01529240

E. Gomez-diaz, M. Jorda, M. A. Peinado, and A. Rivero, Epigenetics of host-pathogen interactions: the road ahead and the road behind, PLoS Pathog, vol.8, issue.11, p.1003007, 2012.

E. Gómez-díaz, A. Rivero, F. Chandre, and V. G. Corces, Insights into the epigenomic landscape of the human malaria vector Anopheles gambiae, Front Genet, vol.5, p.277, 2014.

B. Pérez-zamorano, S. Rosas-madrigal, O. Lozano, M. C. Méndez, and V. Valverde-garduño, Identification of cis-regulatory sequences reveals potential participation of lola and Deaf1 transcription factors in Anopheles gambiae innate immune response, PLoS ONE, vol.12, issue.10, p.186435, 2017.

S. Zakovic and E. A. Levashina, NF-?B-like signaling pathway REL2 in immune defenses of the Malaria vector Anopheles gambiae, Front Cell Infect Microbiol, vol.7, p.258, 2017.

F. Tripet, F. Aboagye-antwi, and H. Hurd, Ecological immunology of mosquitomalaria interactions, Trends Parasitol, vol.24, issue.5, pp.219-246, 2008.

Y. Zhang, T. Liu, C. A. Meyer, J. Eeckhoute, D. S. Johnson et al., Model-based analysis of ChIP-Seq (MACS), Genome Biol, vol.9, issue.9, p.137, 2008.

J. Ernst and M. Kellis, ChromHMM: automating chromatin-state discovery and characterization, Nat Methods, vol.9, issue.3, pp.215-221, 2012.

P. V. Kharchenko, A. A. Alekseyenko, Y. B. Schwartz, A. Minoda, N. C. Riddle et al., Comprehensive analysis of the chromatin landscape in Drosophila melanogaster, Nature, vol.471, issue.7339, p.480, 2011.

L. Shen, N. Shao, X. Liu, I. Maze, J. Feng et al., diffReps: detecting differential chromatin modification sites from ChIP-seq data with biological replicates, PLoS ONE, vol.8, issue.6, p.65598, 2013.

R. M. Brucker, L. J. Funkhouser, S. Setia, R. Pauly, and S. R. Bordenstein, Insect innate immunity database (IIID): an annotation tool for identifying immune genes in insect genomes, PLoS ONE, vol.7, issue.9, p.45125, 2012.

R. M. Waterhouse, E. V. Kriventseva, S. Meister, Z. Xi, K. S. Alvarez et al., Evolutionary dynamics of immune-related genes and pathways in disease-vector mosquitoes, Science, vol.316, issue.5832, pp.1738-1781, 2007.

L. Souvannaseng, L. V. Hun, H. Baker, J. M. Klyver, B. Wang et al., Inhibition of JNK signaling in the Asian malaria vector Anopheles stephensi extends mosquito longevity and improves resistance to Plasmodium falciparum infection, PLoS Pathog, vol.14, issue.11, p.1007418, 2018.

L. Kumar and M. E. Futschik, Mfuzz: a software package for soft clustering of microarray data, Bioinformation, vol.2, issue.1, pp.5-7, 2007.

D. C. Rinker, R. J. Pitts, and L. J. Zwiebel, Disease vectors in the era of next generation sequencing, Genome Biol, vol.17, issue.1, p.95, 2016.

B. E. Bernstein, T. S. Mikkelsen, X. Xie, M. Kamal, D. J. Huebert et al., A bivalent chromatin structure marks key developmental genes in embryonic stem cells, Cell, vol.125, issue.2, pp.315-341, 2006.

P. Voigt, W. W. Tee, and D. Reinberg, A double take on bivalent promoters, Genes Dev, vol.27, issue.12, pp.1318-1356, 2013.

M. P. Creyghton, A. W. Cheng, G. G. Welstead, T. Kooistra, B. W. Carey et al., Histone H3K27ac separates active from poised enhancers and predicts developmental state, Proc Natl Acad Sci, vol.107, issue.50, pp.21931-21937, 2010.

M. M. Pradeepa, Causal role of histone acetylations in enhancer function, Transcription, vol.8, issue.1, pp.40-47, 2017.

B. Lemaitre and J. Hoffmann, The host defense of Drosophila melanogaster, Annu Rev Immunol, vol.25, pp.697-743, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00167467

E. Marois, The multifaceted mosquito anti-Plasmodium response, Curr Opin Microbiol, vol.14, issue.4, pp.429-464, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02346282

T. Habtewold, Z. Groom, and G. K. Christophides, Immune resistance and tolerance strategies in malaria vector and non-vector mosquitoes. Parasites Vectors, vol.10, p.186, 2017.

L. A. Baton and L. C. Ranford-cartwright, Ookinete destruction within the mosquito midgut lumen explains Anopheles albimanus refractoriness to Plasmodium falciparum (3D7A) oocyst infection, Int J Parasitol, vol.42, issue.3, pp.249-58, 2012.

R. C. Smith and C. Barillas-mury, Plasmodium oocysts: overlooked targets of mosquito immunity, Trends Parasitol, vol.32, issue.12, pp.979-90, 2016.

N. Negre, C. D. Brown, L. Ma, C. A. Bristow, S. W. Miller et al., A cis-regulatory map of the Drosophila genome, Nature, vol.471, issue.7339, pp.527-558, 2011.

S. K. Behura, J. Sarro, P. Li, K. Mysore, D. W. Severson et al., High-throughput cis-regulatory element discovery in the vector mosquito Aedes aegypti, BMC Genom, vol.17, p.341, 2016.

S. Govind, Innate immunity in Drosophila: pathogens and pathways, Insect Sci, vol.15, issue.1, pp.29-43, 2008.

D. E. Reed, X. M. Huang, J. A. Wohlschlegel, M. S. Levine, and K. Senger, DEAF-1 regulates immunity gene expression in Drosophila, Proc Natl Acad Sci, vol.105, issue.24, pp.8351-8357, 2008.

H. Alout, I. Djègbè, F. Chandre, L. S. Djogbénou, R. K. Dabiré et al., Insecticide exposure impacts vector-parasite interactions in insecticide-resistant malaria vectors, Proc R Soc B, vol.281, p.20140389, 1786.

A. Vantaux, K. R. Dabiré, A. Cohuet, and T. Lefèvre, A heavy legacy: offspring of malaria-infected mosquitoes show reduced disease resistance, Malar J, vol.13, issue.1, p.442, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02411103

D. F. Da, T. S. Churcher, R. S. Yerbanga, B. Yaméogo, I. Sangaré et al., Experimental study of the relationship between Plasmodium gametocyte density and infection success in mosquitoes; implications for the evaluation of malaria transmissionreducing interventions, Exp Parasitol, vol.149, pp.74-83, 2015.

M. Gendrin, F. H. Rodgers, R. S. Yerbanga, J. B. Ouédraogo, M. Basáñez et al., Antibiotics in ingested human blood affect, vol.18, p.18

. Ruiz, 12:5 ? fast, convenient online submission ? thorough peer review by experienced researchers in your field ? rapid publication on acceptance ? support for research data, including large and complex data types ? gold Open Access which fosters wider collaboration and increased citations maximum visibility for your research: over 100M website views per year ? At BMC, research is always in progress. Learn more biomedcentral.com/submissions Ready to submit your research ? Choose BMC and benefit from: the mosquito microbiota and capacity to transmit malaria, Epigenetics & Chromatin, vol.6, p.5921, 2015.

G. I. Giraldo-calderon, S. J. Emrich, R. M. Maccallum, G. Maslen, E. Dialynas et al., VectorBase: an updated bioinformatics resource for invertebrate vectors and other organisms related with human diseases, Nucleic Acids Res, vol.43, pp.707-720, 2015.

D. Kim, G. Pertea, C. Trapnell, H. Pimentel, R. Kelley et al., TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions, Genome Biol, vol.14, issue.4, p.36, 2013.

H. Li, B. Handsaker, A. Wysoker, T. Fennell, J. Ruan et al., Genome project data processing S: the sequence alignment/map format and SAMtools, Bioinformatics, vol.25, issue.16, pp.2078-2087, 2009.

K. Okonechnikov, A. Conesa, and F. Garcia-alcalde, Qualimap 2: advanced multi-sample quality control for high-throughput sequencing data, Bioinformatics, vol.32, issue.2, pp.292-296, 2016.

S. Anders, P. T. Pyl, and W. Huber, HTSeq-a python framework to work with high-throughput sequencing data, Bioinformatics, vol.31, issue.2, pp.166-175, 2015.

M. I. Love, W. Huber, and S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, vol.15, issue.12, p.550, 2014.

X. Jiao, B. T. Sherman, D. W. Huang, R. Stephens, M. W. Baseler et al., DAVID-WS: a stateful web service to facilitate gene/protein list analysis, Bioinformatics, vol.28, issue.13, pp.1805-1811, 2012.

D. W. Huang, B. T. Sherman, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nat Protoc, vol.4, issue.1, p.44, 2008.

S. K. Bowman, M. D. Simon, A. M. Deaton, M. Tolstorukov, M. L. Borowsky et al., Multiplexed Illumina sequencing libraries from picogram quantities of DNA, BMC Genom, vol.14, issue.1, p.466, 2013.

F. Ramirez, D. P. Ryan, B. Gruning, V. Bhardwaj, F. Kilpert et al., deepTools2: a next generation web server for deepsequencing data analysis, Nucleic Acids Res, vol.44, issue.W1, pp.160-165, 2016.

B. Langmead and S. L. Salzberg, Fast gapped-read alignment with Bowtie 2, Nat Methods, vol.9, issue.4, pp.357-366, 2012.

A. R. Quinlan and I. M. Hall, BEDTools: a flexible suite of utilities for comparing genomic features, Bioinformatics, vol.26, issue.6, pp.841-843, 2010.

J. G. Chitpin and A. Awdeh, Perkins TJ: RECAP reveals the true statistical significance of ChIP-seq peak calls. bioRxiv, p.260687, 2018.

J. T. Robinson, H. Thorvaldsdottir, W. Winckler, M. Guttman, E. S. Lander et al., Integrative genomics viewer, Nat Biotechnol, vol.29, issue.1, pp.24-30, 2011.

H. Thorvaldsdottir, J. T. Robinson, and J. P. Mesirov, Integrative genomics viewer (IGV): high-performance genomics data visualization and exploration, Brief Bioinform, vol.14, issue.2, pp.178-92, 2013.

, MACS. Build signal track

S. Heinz, C. Benner, N. Spann, E. Bertolino, Y. C. Lin et al., Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities, Mol Cell, vol.38, issue.4, pp.576-89, 2010.

P. D. Thomas, M. J. Campbell, A. Kejariwal, H. Mi, B. Karlak et al., PANTHER: a library of protein families and subfamilies indexed by function, Genome Res, vol.13, issue.9, pp.2129-2170, 2003.

H. Mi, Q. Dong, A. Muruganujan, P. Gaudet, S. Lewis et al., PANTHER version 7: improved phylogenetic trees, orthologs and collaboration with the gene ontology consortium, Nucleic Acids Res, vol.38, pp.204-214, 2010.

A. Taudt, M. A. Nguyen, M. Heinig, F. Johannes, and M. Colome-tatche, chromstaR: tracking combinatorial chromatin state dynamics in space and time, bioRxiv, p.38612, 2016.

L. Shen, N. Shao, X. Liu, and N. E. Ngs, plot: quick mining and visualization of next-generation sequencing data by integrating genomic databases, BMC Genom, vol.15, p.284, 2014.

E. Gomez-diaz, R. S. Yerbanga, T. Lefevre, A. Cohuet, M. J. Rowley et al., Epigenetic regulation of Plasmodium falciparum clonally variant gene expression during development in Anopheles gambiae, Sci Rep, vol.7, p.40655, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02010957

X. Zhou, C. E. Cain, M. Myrthil, N. Lewellen, K. Michelini et al., Epigenetic modifications are associated with inter-species gene expression variation in primates, Genome Biol, vol.15, issue.12, p.547, 2014.

X. Dong, M. C. Greven, A. Kundaje, S. Djebali, J. B. Brown et al., Modeling gene expression using chromatin features in various cellular contexts, Genome Biol, vol.13, issue.9, p.53, 2012.

K. Barto?, Multi-model inference. R package version, vol.1, p.42, 2018.

J. Fox and S. Weisberg, An R companion to applied regression, 2018.

U. Grömping, Relative importance for linear regression in R: the package relaimpo, J Stat Softw, vol.17, issue.1, pp.1-27, 2006.

A. N. Schep and S. K. Kummerfeld, iheatmapr: interactive complex heatmaps in R, J Open Sour Softw, vol.2, issue.16, p.359, 2017.

H. Wickham, ggplot2: elegant graphics for data analysis, 2016.

, Harrell FE with contributions from Charles Dupont and many others, vol.4, pp.0-3, 2017.