C. T. Ong and V. G. Corces, Enhancer function: new insights into the regulation of tissue-specific gene expression, Nat Rev Genet, vol.12, pp.283-293, 2011.

P. Moreau, The SV40 72 base repair repeat has a striking effect on gene expression both in SV40 and other chimeric recombinants, Nucleic acids research, vol.9, pp.6047-6068, 1981.

V. Haberle and A. Stark, Eukaryotic core promoters and the functional basis of transcription initiation, Nat Rev Mol Cell Biol, vol.19, pp.621-637, 2018.

D. Hnisz, K. Shrinivas, R. A. Young, A. K. Chakraborty, and P. A. Sharp, A Phase Separation Model for Transcriptional Control, Cell, vol.169, pp.13-23, 2017.

M. A. Schaub, A. P. Boyle, A. Kundaje, S. Batzoglou, and M. Snyder, Linking disease associations with regulatory information in the human genome, Genome research, vol.22, pp.1748-1759, 2012.

H. Chen, Dynamic interplay between enhancer-promoter topology and gene activity, Nat Genet, vol.50, pp.1296-1303, 2018.

L. A. Pennacchio, W. Bickmore, A. Dean, M. A. Nobrega, and G. Bejerano, Enhancers: five essential questions, Nat Rev Genet, vol.14, pp.288-295, 2013.

K. K. Farh, Genetic and epigenetic fine mapping of causal autoimmune disease variants, Nature, 2014.

P. V. Kharchenko, Comprehensive analysis of the chromatin landscape in Drosophila melanogaster, Nature, vol.471, pp.480-485, 2011.

C. E. Romanoski, V. M. Link, S. Heinz, and C. K. Glass, Exploiting genomics and natural genetic variation to decode macrophage enhancers, Trends Immunol, vol.36, pp.507-518, 2015.

A. Sicard, Standing genetic variation in a tissue-specific enhancer underlies selfing-syndrome evolution in Capsella, Proceedings of the National Academy of Sciences of the United States of America, vol.113, pp.13911-13916, 2016.

J. Macarthur, The new NHGRI-EBI Catalog of published genome-wide association studies (GWAS Catalog), Nucleic acids research, vol.45, pp.896-901, 2017.

S. Neph, An expansive human regulatory lexicon encoded in transcription factor footprints, Nature, vol.489, pp.83-90, 2012.

N. Weinhold, A. Jacobsen, N. Schultz, C. Sander, and W. Lee, Genome-wide analysis of noncoding regulatory mutations in cancer, Nat Genet, vol.46, pp.1160-1165, 2014.

S. Heinz, Effect of natural genetic variation on enhancer selection and function, Nature, vol.503, pp.487-492, 2013.

T. D. Capellini, Ancient selection for derived alleles at a GDF5 enhancer influencing human growth and osteoarthritis risk, Nat Genet, vol.49, pp.1202-1210, 2017.

J. Jacobs, The transcription factor Grainy head primes epithelial enhancers for spatiotemporal activation by displacing nucleosomes, Nat Genet, vol.50, pp.1011-1020, 2018.

T. Sagai, M. Hosoya, Y. Mizushina, M. Tamura, and T. Shiroishi, Elimination of a long-range cis-regulatory module causes complete loss of limb-specific Shh expression and truncation of the mouse limb, Development, vol.132, pp.797-803, 2005.

S. Smemo, Regulatory variation in a TBX5 enhancer leads to isolated congenital heart disease, Hum Mol Genet, vol.21, pp.3255-3263, 2012.

C. D. Arnold, Quantitative genome-wide enhancer activity maps for five Drosophila species show functional enhancer conservation and turnover during cis-regulatory evolution, Nat Genet, vol.46, pp.685-692, 2014.

J. Vierstra, Mouse regulatory DNA landscapes reveal global principles of cis-regulatory evolution, Science, vol.346, pp.1007-1012, 2014.

L. F. Franchini and K. S. Pollard, Can a few non-coding mutations make a human brain, Bioessays, vol.37, pp.1054-1061, 2015.

S. K. Behura, High-throughput cis-regulatory element discovery in the vector mosquito Aedes aegypti, BMC genomics, vol.17, 2016.

K. Mysore, P. Li, and M. Duman-scheel, Identification of Aedes aegypti cis-regulatory elements that promote gene expression in olfactory receptor neurons of distantly related dipteran insects, Parasit Vectors, vol.11, 2018.

J. L. Ruiz, Chromatin changes in Anopheles gambiae induced by Plasmodium falciparum infection, Epigenetics Chromatin, vol.12, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01998017

D. A. O'brochta, K. L. Pilitt, R. A. Harrell, C. Aluvihare, and R. T. Alford, Gal4-based enhancer-trapping in the malaria mosquito Anopheles stephensi, vol.2, pp.1305-1315, 2012.

G. D. Weedall, A cytochrome P450 allele confers pyrethroid resistance on a major African malaria vector, reducing insecticidetreated bednet efficacy, Sci Transl Med, vol.11, 2019.

A. P. Mcgregor, Morphological evolution through multiple cis-regulatory mutations at a single gene, Nature, vol.448, pp.587-590, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00169725

B. Prud'homme, N. Gompel, and S. B. Carroll, Emerging principles of regulatory evolution, Proceedings of the National Academy of Sciences of the United States of America, vol.104, issue.1, pp.8605-8612, 2007.

C. D. Arnold, Genome-wide quantitative enhancer activity maps identified by STARR-seq, Science, vol.339, pp.1074-1077, 2013.

D. Bieli, The Drosophila melanogaster Mutants apblot and apXasta Affect an Essential apterous Wing Enhancer, G3, vol.5, pp.1129-1143, 2015.

E. Preger-ben-noon, Comprehensive Analysis of a cis-Regulatory Region Reveals Pleiotropy in Enhancer Function, Cell reports, vol.22, pp.3021-3031, 2018.

D. Bieli, Establishment of a Developmental Compartment Requires Interactions between Three Synergistic Cis-regulatory Modules, PLoS Genet, vol.11, 2015.

C. Anopheles-gambiae-genomes, Genetic diversity of the African malaria vector Anopheles gambiae, Nature, vol.552, pp.96-100, 2017.

E. Z. Kvon, Progressive Loss of Function in a Limb Enhancer during Snake Evolution, Cell, vol.167, pp.633-642, 2016.

Y. F. Chan, Adaptive evolution of pelvic reduction in sticklebacks by recurrent deletion of a Pitx1 enhancer, Science, vol.327, pp.302-305, 2010.

D. Schmidt, Five-vertebrate ChIP-seq reveals the evolutionary dynamics of transcription factor binding, Science, vol.328, pp.1036-1040, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01615156

A. Rada-iglesias, S. L. Prescott, and J. Wysocka, Human genetic variation within neural crest enhancers: molecular and phenotypic implications, Philos Trans R Soc Lond B Biol Sci, vol.368, 2013.

M. M. Riehle, A cryptic subgroup of Anopheles gambiae is highly susceptible to human malaria parasites, Science, vol.331, pp.596-598, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-01971785

F. Santolamazza, Insertion polymorphisms of SINE200 retrotransposons within speciation islands of Anopheles gambiae molecular forms, Malaria journal, vol.7, 2008.

H. M. Muller, G. Dimopoulos, C. Blass, and F. C. Kafatos, A hemocyte-like cell line established from the malaria vector Anopheles gambiae expresses six prophenoloxidase genes, The Journal of biological chemistry, vol.274, pp.11727-11735, 1999.

C. Fanello, F. Santolamazza, and A. Della-torre, Simultaneous identification of species and molecular forms of the Anopheles gambiae complex by PCR-RFLP, Medical and Veterinary Entomology, vol.16, 2002.

H. Thorvaldsdottir, J. T. Robinson, and J. P. Mesirov, Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration, Briefings in bioinformatics, vol.14, pp.178-192, 2013.

C. Harris, Polymorphisms in Anopheles gambiae immune genes associated with natural resistance to Plasmodium falciparum, PLoS pathogens, vol.6, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-02011022

S. N. Redmond, Association mapping by pooled sequencing identifies TOLL 11 as a protective factor against Plasmodium falciparum in Anopheles gambiae, BMC genomics, vol.16, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-02008320

S. Kumar, G. Stecher, M. Li, C. Knyaz, K. Tamura et al., Molecular Evolutionary Genetics Analysis across Computing Platforms, Mol Biol Evol, vol.35, pp.1547-1549, 2018.

A. Mckenna, The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data, Genome research, vol.20, pp.1297-1303, 2010.

V. Narasimhan, BCFtools/RoH: a hidden Markov model approach for detecting autozygosity from next-generation sequencing data, Bioinformatics, vol.32, pp.1749-1751, 2016.

M. A. Larkin, Clustal W and Clustal X version 2.0, Bioinformatics, vol.23, pp.2947-2948, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00206210

A. Larsson, AliView: a fast and lightweight alignment viewer and editor for large datasets, Bioinformatics, vol.30, pp.3276-3278, 2014.