S. Sankararaman, S. Mallick, M. Dannemann, K. Prüfer, J. Kelso et al., The genomic landscape of Neanderthal ancestry in present-day humans, Nature, vol.507, issue.7492, pp.354-357, 2014.

S. Sankararaman, S. Mallick, N. Patterson, and D. Reich, The Combined Landscape of Denisovan and Neanderthal Ancestry in Present-Day Humans, Current Biology, vol.26, issue.9, pp.1241-1247, 2016.

B. Vernot and J. M. Akey, Resurrecting Surviving Neandertal Lineages from Modern Human Genomes, Science, vol.343, issue.6174, pp.1017-1021, 2014.

B. Vernot, S. Tucci, J. Kelso, J. G. Schraiber, A. B. Wolf et al., Excavating Neandertal and Denisovan DNA from the genomes of Melanesian individuals, Science, vol.352, issue.6282, pp.235-239, 2016.

S. R. Browning, B. L. Browning, Y. Zhou, S. Tucci, and J. M. Akey, Analysis of Human Sequence Data Reveals Two Pulses of Archaic Denisovan Admixture, Cell, vol.173, issue.1, pp.53-61.e9, 2018.

K. Prüfer, F. Racimo, N. Patterson, F. Jay, S. Sankararaman et al., The complete genome sequence of a Neanderthal from the Altai Mountains, Nature, vol.505, issue.7481, pp.43-49, 2013.

R. C. Lee, R. L. Feinbaum, and V. Ambros, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14, Cell, vol.75, issue.5, pp.843-854, 1993.
URL : https://hal.archives-ouvertes.fr/in2p3-00597159

J. Krol, I. Loedige, and W. Filipowicz, The widespread regulation of microRNA biogenesis, function and decay, Nature Reviews Genetics, vol.11, issue.9, pp.597-610, 2010.

R. M. O'connell, D. S. Rao, and D. Baltimore, microRNA Regulation of Inflammatory Responses, Annual Review of Immunology, vol.30, issue.1, pp.295-312, 2012.

Y. Li and X. Shi, MicroRNAs in the regulation of TLR and RIG-I pathways, Cellular & Molecular Immunology, vol.10, issue.1, pp.65-71, 2012.

X. Luo, K. Ranade, R. Talker, B. Jallal, N. Shen et al., microRNA-mediated regulation of innate immune response in rheumatic diseases, Arthritis Research & Therapy, vol.15, issue.2, p.210, 2013.

E. Vigorito, S. Kohlhaas, D. Lu, and R. Leyland, miR-155: an ancient regulator of the immune system, Immunological Reviews, vol.253, issue.1, pp.146-157, 2013.

K. D. Taganov, M. P. Boldin, K. Chang, and D. Baltimore, NF- B-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses, Proceedings of the National Academy of Sciences, vol.103, issue.33, pp.12481-12486, 2006.

C. Xiao and K. Rajewsky, MicroRNA Control in the Immune System: Basic Principles, Cell, vol.136, issue.1, pp.26-36, 2009.

K. J. Siddle, M. Deschamps, L. Tailleux, Y. Nedelec, J. Pothlichet et al., A genomic portrait of the genetic architecture and regulatory impact of microRNA expression in response to infection, Genome Research, vol.24, issue.5, pp.850-859, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02348571

K. J. Siddle, Correction: Bacterial Infection Drives the Expression Dynamics of microRNAs and Their isomiRs, PLOS Genetics, vol.11, issue.6, p.e1005321, 2015.

A. A. Pai, Widespread Shortening of 3' Untranslated Regions and Increased Exon Inclusion Are Evolutionarily Conserved Features of Innate Immune Responses to Infection, PLoS Genet, vol.12, issue.9, p.1006338, 2016.

S. Zhang, J. Li, J. Li, Y. Yang, X. Kang et al., Up-regulation of microRNA-203 in influenza A virus infection inhibits viral replication by targeting DR1, Scientific Reports, vol.8, issue.1, p.6797, 2018.

C. Borel, S. Deutsch, A. Letourneau, E. Migliavacca, S. B. Montgomery et al., Identification of cis- and trans-regulatory variation modulating microRNA expression levels in human fibroblasts, Genome Research, vol.21, issue.1, pp.68-73, 2010.

M. Civelek, R. Hagopian, C. Pan, N. Che, W. Yang et al., Genetic regulation of human adipose microRNA expression and its consequences for metabolic traits, Human Molecular Genetics, vol.22, issue.15, pp.3023-3037, 2013.

T. Lappalainen, M. Sammeth, M. R. Friedländer, P. A. ?t-hoen, J. Monlong et al., Transcriptome and genome sequencing uncovers functional variation in humans, Nature, vol.501, issue.7468, pp.506-511, 2013.

S. Budach, M. Heinig, and A. Marsico, Principles of microRNA Regulation Revealed Through Modeling microRNA Expression Quantitative Trait Loci, Genetics, vol.203, issue.4, pp.1629-1640, 2016.

T. Huan, J. Rong, C. Liu, X. Zhang, K. Tanriverdi et al., Genome-wide identification of microRNA expression quantitative trait loci, Nature Communications, vol.6, issue.1, p.6601, 2015.

L. Parts, Å. K. Hedman, S. Keildson, A. J. Knights, C. Abreu-goodger et al., Extent, Causes, and Consequences of Small RNA Expression Variation in Human Adipose Tissue, PLoS Genetics, vol.8, issue.5, p.e1002704, 2012.

E. R. Gamazon, F. Innocenti, R. Wei, L. Wang, M. Zhang et al., A genome-wide integrative study of microRNAs in human liver, BMC Genomics, vol.14, issue.1, p.395, 2013.

E. R. Gamazon, D. Ziliak, H. K. Im, B. Lacroix, D. S. Park et al., Genetic Architecture of MicroRNA Expression: Implications for the Transcriptome and Complex Traits, The American Journal of Human Genetics, vol.90, issue.6, pp.1046-1063, 2012.

Y. Nedelec, Genetic Ancestry and Natural Selection Drive Population Differences in Immune Responses to Pathogens, Cell, vol.167, issue.3, p.17, 2016.

S. Li, Y. Liao, M. Ho, K. Tsai, C. Lai et al., miRNA arm selection and isomiR distribution in gastric cancer, BMC Genomics, vol.13, issue.Suppl 1, p.S13, 2012.

C. T. Neilsen, G. J. Goodall, and C. P. Bracken, IsomiRs ? the overlooked repertoire in the dynamic microRNAome, Trends in Genetics, vol.28, issue.11, pp.544-549, 2012.

S. Backes, J. S. Shapiro, L. R. Sabin, A. M. Pham, I. Reyes et al., Degradation of Host MicroRNAs by Poxvirus Poly(A) Polymerase Reveals Terminal RNA Methylation as a Protective Antiviral Mechanism, Cell Host & Microbe, vol.12, issue.2, pp.200-210, 2012.

S. L. Ameres and P. D. Zamore, Diversifying microRNA sequence and function, Nature Reviews Molecular Cell Biology, vol.14, issue.8, pp.475-488, 2013.

G. C. Tan, E. Chan, A. Molnar, R. Sarkar, D. Alexieva et al., 5? isomiR variation is of functional and evolutionary importance, Nucleic Acids Research, vol.42, issue.14, pp.9424-9435, 2014.

L. Li, Y. Song, X. Shi, J. Liu, S. Xiong et al., The landscape of miRNA editing in animals and its impact on miRNA biogenesis and targeting, Genome Research, vol.28, issue.1, pp.132-143, 2017.

M. J. De-hoon, R. J. Taft, T. Hashimoto, M. Kanamori-katayama, H. Kawaji et al., Cross-mapping and the identification of editing sites in mature microRNAs in high-throughput sequencing libraries, Genome Research, vol.20, issue.2, pp.257-264, 2010.

M. R. Jones, L. J. Quinton, M. T. Blahna, J. R. Neilson, S. Fu et al., Zcchc11-dependent uridylation of microRNA directs cytokine expression, Nature Cell Biology, vol.11, issue.9, pp.1157-1163, 2009.

T. Katoh, Y. Sakaguchi, K. Miyauchi, T. Suzuki, S. Kashiwabara et al., Selective stabilization of mammalian microRNAs by 3' adenylation mediated by the cytoplasmic poly(A) polymerase GLD-2, Genes & Development, vol.23, issue.4, pp.433-438, 2009.

D. Lee, D. Park, J. H. Park, J. H. Kim, and C. Shin, Poly(A)-specific ribonuclease sculpts the 3? ends of microRNAs, RNA, vol.25, issue.3, pp.388-405, 2018.

N. Cloonan, S. Wani, Q. Xu, J. Gu, K. Lea et al., MicroRNAs and their isomiRs function cooperatively to target common biological pathways, Genome Biology, vol.12, issue.12, p.R126, 2011.

S. L. Fernandez-valverde, R. J. Taft, and J. S. Mattick, Dynamic isomiR regulation in Drosophila development, RNA, vol.16, issue.10, pp.1881-1888, 2010.

F. Yu, K. A. Pillman, C. T. Neilsen, J. Toubia, D. M. Lawrence et al., Naturally existing isoforms of miR-222 have distinct functions, Nucleic Acids Research, vol.45, issue.19, pp.11371-11385, 2017.

C. Nejad, K. A. Pillman, K. J. Siddle, G. Pépin, M. Änkö et al., miR-222 isoforms are differentially regulated by type-I interferon, RNA, vol.24, issue.3, pp.332-341, 2017.

B. P. Lewis, C. B. Burge, and D. P. Bartel, Conserved Seed Pairing, Often Flanked by Adenosines, Indicates that Thousands of Human Genes are MicroRNA Targets, Cell, vol.120, issue.1, pp.15-20, 2005.

R. C. Friedman, N. H. Farh, C. B. Burge, and D. P. Bartel, Most mammalian mRNAs are conserved targets of microRNAs, Genome Research, vol.19, issue.1, pp.92-105, 2008.

A. M. Burroughs, Y. Ando, M. J. De-hoon, Y. Tomaru, T. Nishibu et al., A comprehensive survey of 3' animal miRNA modification events and a possible role for 3' adenylation in modulating miRNA targeting effectiveness, Genome Research, vol.20, issue.10, pp.1398-1410, 2010.

M. Hafner, M. Landthaler, L. Burger, M. Khorshid, J. Hausser et al., Transcriptome-wide Identification of RNA-Binding Protein and MicroRNA Target Sites by PAR-CLIP, Cell, vol.141, issue.1, pp.129-141, 2010.

S. W. Chi, J. B. Zang, A. Mele, and R. B. Darnell, Argonaute HITS-CLIP decodes microRNA?mRNA interaction maps, Nature, vol.460, issue.7254, pp.479-486, 2009.

A. E. Pasquinelli, MicroRNAs and their targets: recognition, regulation and an emerging reciprocal relationship, Nature Reviews Genetics, vol.13, issue.4, pp.271-282, 2012.

D. P. Bartel, MicroRNAs: Target Recognition and Regulatory Functions, Cell, vol.136, issue.2, pp.215-233, 2009.

R. Gumienny and M. Zavolan, Accurate transcriptome-wide prediction of microRNA targets and small interfering RNA off-targets with MIRZA-G: Figure 2., Nucleic Acids Research, vol.43, issue.18, pp.9095-9095, 2015.

M. Khorshid, J. Hausser, M. Zavolan, and E. Van-nimwegen, A biophysical miRNA-mRNA interaction model infers canonical and noncanonical targets, Nature Methods, vol.10, issue.3, pp.253-255, 2013.

L. Wang, J. Zhu, F. Deng, L. Wu, X. Mo et al., Correlation analyses revealed global microRNA?mRNA expression associations in human peripheral blood mononuclear cells, Molecular Genetics and Genomics, vol.293, issue.1, pp.95-105, 2017.

D. Gaidatzis, L. Burger, M. Florescu, and M. B. Stadler, Analysis of intronic and exonic reads in RNA-seq data characterizes transcriptional and post-transcriptional regulation, Nature Biotechnology, vol.33, issue.7, pp.722-729, 2015.

H. Kim, J. Kim, K. Kim, H. Chang, K. You et al., Bias-minimized quantification of microRNA reveals widespread alternative processing and 3? end modification, Nucleic Acids Research, vol.47, issue.5, pp.2630-2640, 2019.

I. Heo, C. Joo, Y. Kim, M. Ha, M. Yoon et al., TUT4 in Concert with Lin28 Suppresses MicroRNA Biogenesis through Pre-MicroRNA Uridylation, Cell, vol.138, issue.4, pp.696-708, 2009.

B. Kim, M. Ha, L. Loeff, H. Chang, D. K. Simanshu et al., TUT 7 controls the fate of precursor micro RNA s by using three different uridylation mechanisms, The EMBO Journal, vol.34, issue.13, pp.1801-1815, 2015.

L. Zhu, S. K. Kandasamy, and R. Fukunaga, Dicer partner protein tunes the length of miRNAs using base-mismatch in the pre-miRNA stem, Nucleic Acids Research, vol.46, issue.7, pp.3726-3741, 2018.

J. Ding, V. Tarokh, and Y. Yang, Model Selection Techniques: An Overview, IEEE Signal Processing Magazine, vol.35, issue.6, pp.16-34, 2018.

F. Bazzoni, Induction and regulatory function of miR-9 in human monocytes and neutrophils exposed to proinflammatory signals, Proc Natl Acad Sci, vol.106, issue.13, pp.5282-5289, 2009.

I. Wohlers, L. Bertram, and C. M. Lill, Evidence for a potential role of miR-1908-5p and miR-3614-5p in autoimmune disease risk using integrative bioinformatics, Journal of Autoimmunity, vol.94, pp.83-89, 2018.

J. Xiao, J. Tang, Q. Chen, D. Tang, M. Liu et al., miR-429 regulates alveolar macrophage inflammatory cytokine production and is involved in LPS-induced acute lung injury, Biochemical Journal, vol.471, issue.2, pp.281-291, 2015.

H. Tian and Z. He, miR-215 Enhances HCV Replication by Targeting TRIM22 and Inactivating NF-?B Signaling, Yonsei Medical Journal, vol.59, issue.4, p.511, 2018.

S. H. Najafi-shoushtari, MicroRNA-33 and the SREBP host genes cooperate to control cholesterol homeostasis, Science, vol.328, issue.5985, pp.1566-1575, 2010.

V. Zuber and K. Strimmer, High-Dimensional Regression and Variable Selection Using CAR Scores, Statistical Applications in Genetics and Molecular Biology, vol.10, issue.1, 2011.

J. J. Seeley, Induction of innate immune memory via microRNA targeting of chromatin remodelling factors, Nature, vol.559, issue.7712, pp.114-119, 2018.

H. Quach, Signatures of purifying and local positive selection in human miRNAs, Am J Hum Genet, vol.84, issue.3, pp.316-343, 2009.

M. Rantalainen, B. M. Herrera, G. Nicholson, R. Bowden, Q. F. Wills et al., MicroRNA Expression in Abdominal and Gluteal Adipose Tissue Is Associated with mRNA Expression Levels and Partly Genetically Driven, PLoS ONE, vol.6, issue.11, p.e27338, 2011.

J. W. Freimer, T. J. Hu, and R. Blelloch, Decoupling the impact of microRNAs on translational repression versus RNA degradation in embryonic stem cells, eLife, vol.7, issue.7, 2018.

B. Langmead, C. Trapnell, M. Pop, and S. L. Salzberg, Ultrafast and memory-efficient alignment of short DNA sequences to the human genome, Genome Biology, vol.10, issue.3, p.R25, 2009.

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.

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

W. E. Johnson, C. Li, and A. Rabinovic, Adjusting batch effects in microarray expression data using empirical Bayes methods, Biostatistics, vol.8, issue.1, pp.118-127, 2006.

A. A. Shabalin, Matrix eQTL: ultra fast eQTL analysis via large matrix operations, Bioinformatics, vol.28, issue.10, pp.1353-1358, 2012.

D. De-rie, Response to Gould et al. and Vincelli et al., Nature Biotechnology, vol.35, issue.4, pp.306-308, 2017.

C. Trapnell, The sva package for removing batch effects and other unwanted variation in high-throughput experiments, Nature Protocols, vol.7, issue.6, pp.882-885, 2012.

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, 2014.

J. Friedman, T. Hastie, and R. Tibshirani, Regularization Paths for Generalized Linear Models via Coordinate Descent, Journal of Statistical Software, vol.33, issue.1, 2010.

B. Hofner, L. Boccuto, and M. Göker, Controlling false discoveries in high-dimensional situations: boosting with stability selection, BMC Bioinformatics, vol.16, issue.1, p.144, 2015.

P. Carninci, M. Rehli, and A. Sandelin, An atlas of active enhancers across human cell types and tissues, Nature, vol.507, issue.7493, pp.455-461, 2014.

D. Backenroth, Z. He, K. Kiryluk, V. Boeva, L. Pethukova et al., FUN-LDA: A LATENT DIRICHLET ALLOCATION MODEL FOR PREDICTING TISSUE-SPECIFIC FUNCTIONAL EFFECTS OF NONCODING VARIATION, Am J Hum Genet, vol.102, issue.5, pp.920-942, 2016.

D. P. Bartel, MicroRNAs: Target Recognition and Regulatory Functions, Cell, vol.136, issue.2, pp.215-233, 2009.

W. Bateson, Heredity and Variation in Modern Lights, William Bateson, Naturalist, pp.215-232

A. Battle, S. Mostafavi, X. Zhu, J. B. Potash, M. M. Weissman et al., Characterizing the genetic basis of transcriptome diversity through RNA-sequencing of 922 individuals, Genome Research, vol.24, issue.1, pp.14-24, 2013.

M. L. Benton, S. C. Talipineni, D. Kostka, and J. A. Capra, Genome-wide Enhancer Maps Differ Significantly in Genomic Distribution, Evolution, and Function, p.176610, 2017.

J. J. Berg and G. Coop, A Population Genetic Signal of Polygenic Adaptation, PLoS Genetics, vol.10, issue.8, p.e1004412, 2014.

T. Bersaglieri, P. C. Sabeti, N. Patterson, T. Vanderploeg, S. F. Schaffner et al., Genetic Signatures of Strong Recent Positive Selection at the Lactase Gene, The American Journal of Human Genetics, vol.74, issue.6, pp.1111-1120, 2004.

J. L. Bischoff, R. W. Williams, R. J. Rosenbauer, A. Aramburu, J. L. Arsuaga et al., High-resolution U-series dates from the Sima de los Huesos hominids yields : implications for the evolution of the early Neanderthal lineage, Journal of Archaeological Science, vol.34, issue.5, pp.763-770, 2007.

S. R. Browning, B. L. Browning, Y. Zhou, S. Tucci, and J. M. Akey, Analysis of Human Sequence Data Reveals Two Pulses of Archaic Denisovan Admixture, Cell, vol.173, issue.1, pp.53-61.e9, 2018.

A. Buniello, J. A. Macarthur, M. Cerezo, L. W. Harris, J. Hayhurst et al., The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019, Nucleic Acids Research, vol.47, issue.D1, pp.D1005-D1012, 2018.

C. D. Campbell, J. X. Chong, M. Malig, A. Ko, B. L. Dumont et al., Estimating the human mutation rate using autozygosity in a founder population, Nature Genetics, vol.44, issue.11, pp.1277-1281, 2012.

J. A. Coyne and H. A. Orr, PATTERNS OF SPECIATION IN DROSOPHILA, Evolution, vol.43, issue.2, pp.362-381, 1989.

M. Dannemann and J. Kelso, The Contribution of Neanderthals to Phenotypic Variation in Modern Humans, The American Journal of Human Genetics, vol.101, issue.4, pp.578-589, 2017.

M. Dannemann, K. Prüfer, and J. Kelso, Functional implications of Neandertal introgression in modern humans, Genome Biology, vol.18, issue.1, p.61, 2017.

M. Dannemann and F. Racimo, Something old, something borrowed: Admixture and adaptation in human evolution, Curr Opin Genet Dev, vol.53, pp.1-8, 2018.

S. De-azevedo, M. F. González, C. Cintas, V. Ramallo, M. Quinto-sánchez et al., Nasal airflow simulations suggest convergent adaptation in Neanderthals and modern humans, Proceedings of the National Academy of Sciences, vol.114, issue.47, pp.12442-12447, 2017.

M. Deschamps, G. Laval, M. Fagny, Y. Itan, L. Abel et al., Genomic Signatures of Selective Pressures and Introgression from Archaic Hominins at Human Innate Immunity Genes, The American Journal of Human Genetics, vol.98, issue.1, pp.5-21, 2016.

H. L. Dibble, D. Sandgathe, P. Goldberg, S. Mcpherron, and V. Aldeias, Were Western European Neandertals Able to Make Fire?, Journal of Paleolithic Archaeology, vol.1, issue.1, pp.54-79, 2018.

T. Dobzhansky, Genetics and the origin of species, vol.11, 1982.

M. K. Donovan, A. D?antonio-chronowska, M. D?antonio, and K. A. Frazer, Cellular deconvolution of GTEx tissues powers eQTL studies to discover thousands of novel disease and cell-type associated regulatory variants, p.671040, 2019.

A. Durvasula and S. Sankararaman, Recovering signals of ghost archaic introgression in African populations, p.285734, 2018.

D. Enard and D. A. Petrov, Evidence that RNA Viruses Drove Adaptive Introgression between Neanderthals and Modern Humans, Cell, vol.175, issue.2, pp.360-371.e13, 2018.

A. J. Enright, B. John, U. Gaul, T. Tuschl, C. Sander et al., Dietary reconstruction of the el sidrón neandertal familial group (spain) in the context of other neandertal and modern huntergatherer groups. a molar microwear texture analysis, Journal of Human Evolution, vol.5, issue.1, pp.13-22, 2003.

A. Eyre-walker and P. D. Keightley, High genomic deleterious mutation rates in hominids, Nature, vol.397, issue.6717, pp.344-347, 1999.

L. Fang, J. K. Ahn, D. Wodziak, and E. Sibley, The human lactase persistence-associated SNP ?13910*T enables in vivo functional persistence of lactase promoter?reporter transgene expression, Human Genetics, vol.131, issue.7, pp.1153-1159, 2012.

F. Consortium, R. P. The, . Clst, A. R. Forrest, H. Kawaji et al., A promoter-level mammalian expression atlas, Nature, vol.507, issue.7493, pp.462-470, 2014.

D. P. Francis, Deadly AIDS policy failure by the highest levels of the US government: A personal look back 30 years later for lessons to respond better to future epidemics, Journal of Public Health Policy, vol.33, issue.3, pp.290-300, 2012.

H. B. Fraser, Gene expression drives local adaptation in humans, Genome Research, vol.23, issue.7, pp.1089-1096, 2013.

J. W. Freimer, T. J. Hu, and R. Blelloch, Decoupling the impact of microRNAs on translational repression versus RNA degradation in embryonic stem cells, eLife, vol.7, 2018.

Q. Fu, C. Posth, M. Hajdinjak, M. Petr, S. Mallick et al., The genetic history of Ice Age Europe, Nature, vol.534, issue.7606, pp.200-205, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01315112

J. M. Good, T. Giger, M. D. Dean, and M. W. Nachman, Widespread Over-Expression of the X Chromosome in Sterile F1 Hybrid Mice, PLoS Genetics, vol.6, issue.9, p.e1001148, 2010.

R. E. Green, J. Krause, A. W. Briggs, T. Maricic, U. Stenzel et al., A Draft Sequence of the Neandertal Genome, Science, vol.328, issue.5979, pp.710-722, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00920381

O. Reiner, Faculty Opinions recommendation of A draft sequence of the Neandertal genome., Science, vol.328, issue.5979, pp.710-722, 2010.

R. E. Green, A. Malaspinas, J. Krause, A. W. Briggs, P. L. Johnson et al., A Complete Neandertal Mitochondrial Genome Sequence Determined by High-Throughput Sequencing, Cell, vol.134, issue.3, pp.416-426, 2008.

G. Consortium, Enhancing GTEx by bridging the gaps between genotype, gene expression, and disease, Nature Genetics, vol.49, issue.12, pp.1664-1670, 2017.

G. Consortium, D. A. Laboratory, C. Center-analysis, G. ;. Working, G. G. Enhancing et al., Genetic effects on gene expression across human tissues, Nature, vol.550, issue.7675, pp.204-213, 2017.

W. Haerty and R. S. Singh, Gene Regulation Divergence Is a Major Contributor to the Evolution of Dobzhansky?Muller Incompatibilities between Species of Drosophila, Molecular Biology and Evolution, vol.23, issue.9, pp.1707-1714, 2006.

K. Harris and R. Nielsen, The Genetic Cost of Neanderthal Introgression, Genetics, vol.203, issue.2, pp.881-891, 2016.

K. Harvati, C. Röding, A. M. Bosman, F. A. Karakostis, R. Grün et al., Apidima Cave fossils provide earliest evidence of Homo sapiens in Eurasia, Nature, vol.571, issue.7766, pp.500-504, 2019.

M. Helmy, A. Hatlen, and A. Marco, The Impact of Population Variation in the Analysis of microRNA Target Sites, Non-Coding RNA, vol.5, issue.2, p.42, 2019.

T. Higham, K. Douka, R. Wood, C. B. Ramsey, F. Brock et al., The timing and spatiotemporal patterning of Neanderthal disappearance, Nature, vol.512, issue.7514, pp.306-309, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01952841

L. A. Hindorff, P. Sethupathy, H. A. Junkins, E. M. Ramos, J. P. Mehta et al., Potential etiologic and functional implications of genome-wide association loci for human diseases and traits, Proceedings of the National Academy of Sciences, vol.106, issue.23, pp.9362-9367, 2009.

D. L. Hoffmann, C. D. Standish, M. García-diez, P. B. Pettitt, J. A. Milton et al., U-Th dating of carbonate crusts reveals Neandertal origin of Iberian cave art, Science, vol.359, issue.6378, pp.912-915, 2018.

J. J. Hublin, The last Neanderthal, Proceedings of the National Academy of Sciences, vol.114, issue.40, pp.10520-10522, 2017.

E. Huerta-sánchez, X. Jin, . Asan, Z. Bianba, B. M. Peter et al., Altitude adaptation in Tibetans caused by introgression of Denisovan-like DNA, Nature, vol.512, issue.7513, pp.194-197, 2014.

C. J. Ingram, C. A. Mulcare, Y. Itan, M. G. Thomas, and D. M. Swallow, Lactose digestion and the evolutionary genetics of lactase persistence, Human Genetics, vol.124, issue.6, pp.579-591, 2008.

B. M. Javierre, O. S. Burren, S. P. Wilder, R. Kreuzhuber, S. M. Hill et al., Lineage-Specific Genome Architecture Links Enhancers and Non-coding Disease Variants to Target Gene Promoters, Cell, vol.167, issue.5, pp.1369-1384.e19, 2016.

M. R. Jones, L. J. Quinton, M. T. Blahna, J. R. Neilson, S. Fu et al., Zcchc11-dependent uridylation of microRNA directs cytokine expression, Nature Cell Biology, vol.11, issue.9, pp.1157-1163, 2009.

T. Katoh, Y. Sakaguchi, K. Miyauchi, T. Suzuki, S. Kashiwabara et al., Selective stabilization of mammalian microRNAs by 3' adenylation mediated by the cytoplasmic poly(A) polymerase GLD-2, Genes & Development, vol.23, issue.4, pp.433-438, 2009.

D. E. Kelly, M. E. Hansen, and S. A. Tishkoff, Global variation in gene expression and the value of diverse sampling, Current Opinion in Systems Biology, vol.1, pp.102-108, 2017.

B. Y. Kim, C. D. Huber, and K. E. Lohmueller, Deleterious variation shapes the genomic landscape of introgression, PLOS Genetics, vol.14, issue.10, p.e1007741, 2018.

W. King, The reputed fossil man of the neanderthal. The Quarterly journal of science, vol.1, pp.88-97, 1864.

A. Kozomara and S. Griffiths-jones, miRBase: integrating microRNA annotation and deep-sequencing data, Nucleic Acids Research, vol.39, issue.Database, pp.D152-D157, 2010.

J. Krause, Q. Fu, J. M. Good, B. Viola, M. V. Shunkov et al., The complete mitochondrial DNA genome of an unknown hominin from southern Siberia, Nature, vol.464, issue.7290, pp.894-897, 2010.

J. Krause, L. Orlando, D. Serre, B. Viola, K. Prüfer et al., Neanderthals in central Asia and Siberia, Nature, vol.449, issue.7164, pp.902-904, 2007.

M. Krings, H. Geisert, R. W. Schmitz, H. Krainitzki, and S. Paabo, DNA sequence of the mitochondrial hypervariable region II from the Neandertal type specimen, Proceedings of the National Academy of Sciences, vol.96, issue.10, pp.5581-5585, 1999.

M. Krings, A. Stone, R. W. Schmitz, H. Krainitzki, M. Stoneking et al., Neandertal DNA Sequences and the Origin of Modern Humans, Cell, vol.90, issue.1, pp.19-30, 1997.

G. V. Kryukov, L. A. Pennacchio, and S. R. Sunyaev, Most Rare Missense Alleles Are Deleterious in Humans: Implications for Complex Disease and Association Studies, The American Journal of Human Genetics, vol.80, issue.4, pp.727-739, 2007.

A. T. Clark, Spontaneous differentiation of germ cells from human embryonic stem cells in vitro, Human Molecular Genetics, vol.13, issue.7, pp.727-739, 2004.

S. Kudaravalli, J. Veyrieras, B. E. Stranger, E. T. Dermitzakis, and J. K. Pritchard, Gene Expression Levels Are a Target of Recent Natural Selection in the Human Genome, Molecular Biology and Evolution, vol.26, issue.3, pp.649-658, 2008.

M. Kuhlwilm, I. Gronau, M. J. Hubisz, C. De-filippo, J. Prado-martinez et al., Ancient gene flow from early modern humans into Eastern Neanderthals, Nature, vol.530, issue.7591, pp.429-433, 2016.

C. M. Lakhani, B. T. Tierney, A. K. Manrai, J. Yang, P. M. Visscher et al., Repurposing large health insurance claims data to estimate genetic and environmental contributions in 560 phenotypes, Nature Genetics, vol.51, issue.2, pp.327-334, 2019.

E. S. Lander, L. M. Linton, B. Birren, C. Nusbaum, M. C. Zody et al., Initial sequencing and analysis of the human genome, Nature, vol.409, issue.6822, pp.860-921, 2001.

T. Lappalainen, M. Sammeth, M. R. Friedländer, P. A. ?t-hoen, J. Monlong et al., Transcriptome and genome sequencing uncovers functional variation in humans, Nature, vol.501, issue.7468, pp.506-511, 2013.

P. J. Law, M. Timofeeva, C. Fernandez-rozadilla, P. Broderick, J. Studd et al., Association analyses identify 31 new risk loci for colorectal cancer susceptibility, Nature Communications, vol.10, issue.1, p.2154, 2019.

I. Lazaridis, D. Nadel, G. Rollefson, D. C. Merrett, N. Rohland et al., Genomic insights into the origin of farming in the ancient Near East, Nature, vol.536, issue.7617, pp.419-424, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02014294

D. Lee, D. Park, J. H. Park, J. H. Kim, and C. Shin, Poly(A)-specific ribonuclease sculpts the 3? ends of microRNAs, RNA, vol.25, issue.3, pp.388-405, 2018.

R. H. Lewinsky, T. G. Jensen, J. Møller, A. Stensballe, J. Olsen et al., T ?13910 DNA variant associated with lactase persistence interacts with Oct-1 and stimulates lactase promoter activity in vitro, Human Molecular Genetics, vol.14, issue.24, pp.3945-3953, 2005.

L. Li, Y. Song, X. Shi, J. Liu, S. Xiong et al., The landscape of miRNA editing in animals and its impact on miRNA biogenesis and targeting, Genome Research, vol.28, issue.1, pp.132-143, 2017.

S. Liu and C. Trapnell, Single-cell transcriptome sequencing: recent advances and remaining challenges, F1000Research, vol.5, p.182, 2016.

Y. Loh, S. V. Yi, and J. T. Streelman, Evolution of MicroRNAs and the Diversification of Species, Genome Biology and Evolution, vol.3, pp.55-65, 2010.

K. E. Lohmueller, A. Albrechtsen, Y. Li, S. Y. Kim, T. Korneliussen et al., Natural Selection Affects Multiple Aspects of Genetic Variation at Putatively Neutral Sites across the Human Genome, PLoS Genetics, vol.7, issue.10, p.e1002326, 2011.

K. L. Mack and M. W. Nachman, Gene Regulation and Speciation, Trends in Genetics, vol.33, issue.1, pp.68-80, 2017.

S. Mallick, H. Li, M. Lipson, I. Mathieson, M. Gymrek et al., The Simons Genome Diversity Project: 300 genomes from 142 diverse populations, Nature, vol.538, issue.7624, pp.201-206, 2016.

A. Marco, SeedVicious: Analysis of microRNA target and near-target sites, PLOS ONE, vol.13, issue.4, p.e0195532, 2018.

M. T. Maurano, R. Humbert, E. Rynes, R. E. Thurman, E. Haugen et al., Systematic Localization of Common Disease-Associated Variation in Regulatory DNA, Science, vol.337, issue.6099, pp.1190-1195, 2012.

R. C. Mccoy, J. Wakefield, and J. M. Akey, Impacts of Neanderthal-Introgressed Sequences on the Landscape of Human Gene Expression, Cell, vol.168, issue.5, pp.916-927.e12, 2017.

G. Mcvicker, D. Gordon, C. Davis, and P. Green, Widespread Genomic Signatures of Natural Selection in Hominid Evolution, PLoS Genetics, vol.5, issue.5, p.e1000471, 2009.

P. Mellars, Neanderthals and the modern human colonization of Europe, Nature, vol.432, issue.7016, pp.461-465, 2004.

M. Meyer, M. Kircher, M. Gansauge, H. Li, F. Racimo et al., A High-Coverage Genome Sequence from an Archaic Denisovan Individual, Science, vol.338, issue.6104, pp.222-226, 2012.

P. Michalak and M. A. Noor, Genome-Wide Patterns of Expression in Drosophila Pure Species and Hybrid Males, Molecular Biology and Evolution, vol.20, issue.7, pp.1070-1076, 2003.

L. S. Mogil, A. Andaleon, A. Badalamenti, S. P. Dickinson, X. Guo et al., Genetic architecture of gene expression traits across diverse populations, PLoS Genet, vol.14, issue.8, p.1007586, 2018.

H. Muller, Recessive genes causing interspecific sterility and other disharmonies between drosophila melanogaster and simulans, Genetics, p.157, 1942.

D. L. Nicolae, E. Gamazon, W. Zhang, S. Duan, M. E. Dolan et al., Trait-Associated SNPs Are More Likely to Be eQTLs: Annotation to Enhance Discovery from GWAS, PLoS Genetics, vol.6, issue.4, p.e1000888, 2010.

R. Nielsen, Molecular Signatures of Natural Selection, Annual Review of Genetics, vol.39, issue.1, pp.197-218, 2005.

A. Pacis, L. Tailleux, A. M. Morin, J. Lambourne, J. L. Macisaac et al., Bacterial infection remodels the DNA methylation landscape of human dendritic cells, Genome Research, vol.25, issue.12, pp.1801-1811, 2015.

D. Z. Pan, K. M. Garske, M. Alvarez, Y. V. Bhagat, J. Boocock et al., Integration of human adipocyte chromosomal interactions with adipose gene expression prioritizes obesity-related genes from gwas, 1512. Parts, vol.9, p.1002704, 2012.

M. Petr, S. Pääbo, J. Kelso, and B. Vernot, Limits of long-term selection against Neandertal introgression, Proceedings of the National Academy of Sciences, vol.116, issue.5, pp.1639-1644, 2019.

B. Piasecka, D. Duffy, A. Urrutia, H. Quach, E. Patin et al., Distinctive roles of age, sex, and genetics in shaping transcriptional variation of human immune responses to microbial challenges, Proceedings of the National Academy of Sciences, vol.115, issue.3, pp.E488-E497, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01721850

N. Pinzón, B. Li, L. Martinez, A. Sergeeva, J. Presumey et al., microRNA target prediction programs predict many false positives, Genome Research, vol.27, issue.2, pp.234-245, 2016.

V. Plagnol, J. D. Wall, T. J. Polderman, B. Benyamin, C. A. De-leeuw et al., Meta-analysis of the heritability of human traits based on fifty years of twin studies, Nature Genetics, vol.2, issue.7, p.702, 2006.

J. K. Pritchard, J. K. Pickrell, and G. Coop, The Genetics of Human Adaptation: Hard Sweeps, Soft Sweeps, and Polygenic Adaptation, Current Biology, vol.20, issue.4, pp.R208-R215, 2010.

K. Prüfer, C. De-filippo, S. Grote, F. Mafessoni, P. Korlevi? et al., A high-coverage Neandertal genome from Vindija Cave in Croatia, Science, vol.358, issue.6363, pp.655-658, 2017.

K. Prüfer, F. Racimo, N. Patterson, F. Jay, S. Sankararaman et al., The complete genome sequence of a Neanderthal from the Altai Mountains, Nature, vol.505, issue.7481, pp.43-49, 2013.

H. Quach, M. Rotival, J. Pothlichet, Y. Loh, M. Dannemann et al., Genetic Adaptation and Neandertal Admixture Shaped the Immune System of Human Populations, Cell, vol.167, issue.3, pp.643-656.e17, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01385620

J. Quintin, S. Cheng, J. W. Van-der-meer, and M. G. Netea, Innate immune memory: towards a better understanding of host defense mechanisms, Current Opinion in Immunology, vol.29, pp.1-7, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-02875454

F. Racimo, J. J. Berg, and J. K. Pickrell, Detecting Polygenic Adaptation in Admixture Graphs, Genetics, vol.208, issue.4, pp.1565-1584, 2018.

F. Racimo, D. Marnetto, and E. Huerta-sánchez, Signatures of archaic adaptive introgression in present-day human populations, Mol Biol Evol, vol.34, issue.2, pp.296-317, 2016.

M. Rantalainen, B. M. Herrera, G. Nicholson, R. Bowden, Q. F. Wills et al., Microrna expression in abdominal and gluteal adipose tissue is associated with mrna expression levels and partly genetically driven, PLoS One, vol.6, issue.11, pp.1053-60, 2010.

M. P. Richards, G. Taylor, T. Steele, S. P. Mcpherron, M. Soressi et al., Isotopic dietary analysis of a Neanderthal and associated fauna from the site of Jonzac (Charente-Maritime), France, Journal of Human Evolution, vol.55, issue.1, pp.179-185, 2008.
URL : https://hal.archives-ouvertes.fr/halshs-00430209

D. C. Rinker, C. Simonti, E. Mcarthur, D. Shaw, E. Hodges et al., Neanderthal introgression reintroduced functional alleles lost in the human out of africa bottleneck. bioRxiv, 533257. Roadmap Epigenomics Consortium, pp.317-347, 2015.

A. R. Rogers, R. J. Bohlender, and C. D. Huff, Early history of Neanderthals and Denisovans, Proceedings of the National Academy of Sciences, vol.114, issue.37, pp.9859-9863, 2017.

A. Saini, Superior : The return of race science, 2019.

A. J. Sams, A. Dumaine, Y. Nédélec, V. Yotova, C. Alfieri et al., Adaptively introgressed Neandertal haplotype at the OAS locus functionally impacts innate immune responses in humans, Genome Biol, vol.17, issue.1, pp.354-361, 2016.

S. Sankararaman, S. Mallick, N. Patterson, and D. Reich, The Combined Landscape of Denisovan and Neanderthal Ancestry in Present-Day Humans, Current Biology, vol.26, issue.9, pp.1241-1247, 2016.

D. Serre, A. Langaney, M. Chech, M. Teschler-nicola, M. Paunovic et al., No Evidence of Neandertal mtDNA Contribution to Early Modern Humans, Early Modern Humans at the Moravian Gate, vol.2, pp.491-503

K. J. Siddle, M. Deschamps, L. Tailleux, Y. Nedelec, J. Pothlichet et al., A genomic portrait of the genetic architecture and regulatory impact of microRNA expression in response to infection, Genome Research, vol.24, issue.5, pp.850-859, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02348571

K. J. Siddle, L. Tailleux, M. Deschamps, Y. Loh, C. Deluen et al., Bacterial Infection Drives the Expression Dynamics of microRNAs and Their isomiRs, PLOS Genetics, vol.11, issue.3, p.e1005064, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01236327

C. N. Simonti, B. Vernot, L. Bastarache, E. Bottinger, D. S. Carrell et al., The phenotypic legacy of admixture between modern humans and Neandertals, Science, vol.351, issue.6274, pp.737-741, 2016.

L. Skov, R. Hui, V. Shchur, A. Hobolth, A. Scally et al., The genome of the offspring of a neanderthal mother and a denisovan father, PLOS Genetics, vol.14, issue.9, pp.113-116, 2018.

J. D. Sommer, The Shanidar IV ?Flower Burial?: a Re-evaluation of Neanderthal Burial Ritual, Cambridge Archaeological Journal, vol.9, issue.1, pp.127-129, 1999.

B. E. Stranger, S. B. Montgomery, A. S. Dimas, L. Parts, O. Stegle et al., Patterns of Cis Regulatory Variation in Diverse Human Populations, PLoS Genetics, vol.8, issue.4, p.e1002639, 2012.

C. B. Stringer and J. Hublin, New age estimates for the Swanscombe hominid, and their significance for human evolution, Journal of Human Evolution, vol.37, issue.6, pp.873-877, 1999.

L. Ségurel and C. Bon, On the Evolution of Lactase Persistence in Humans, Annual Review of Genomics and Human Genetics, vol.18, issue.1, pp.297-319, 2017.

G. C. Tan, E. Chan, A. Molnar, R. Sarkar, D. Alexieva et al., 5? isomiR variation is of functional and evolutionary importance, Nucleic Acids Research, vol.42, issue.14, pp.9424-9435, 2014.

R. Grocock, S. Humphray, T. James, Z. Kingsbury, H. Lehrach et al., Integrating common and rare genetic variation in diverse human populations, Nature, vol.467, issue.7311, pp.52-58, 2010.

S. Thomas, V. Rouilly, E. Patin, C. Alanio, A. Dubois et al., The Milieu Intérieur study ? An integrative approach for study of human immunological variance, Clinical Immunology, vol.157, issue.2, pp.277-293, 2015.

S. A. Tishkoff, F. A. Reed, A. Ranciaro, B. F. Voight, C. C. Babbitt et al., Convergent adaptation of human lactase persistence in Africa and Europe, Nature Genetics, vol.39, issue.1, pp.31-40, 2006.

J. C. Venter, M. D. Adams, E. W. Myers, P. W. Li, R. J. Mural et al., , p.1304, 2001.

B. Vernot and J. M. Akey, Resurrecting Surviving Neandertal Lineages from Modern Human Genomes, Science, vol.343, issue.6174, pp.1017-1021, 2014.

B. Vernot, S. Tucci, J. Kelso, J. G. Schraiber, A. B. Wolf et al., Excavating Neandertal and Denisovan DNA from the genomes of Melanesian individuals, Science, vol.352, issue.6282, pp.235-239, 2016.

D. Villar, C. Berthelot, S. Aldridge, T. F. Rayner, M. Lukk et al., Enhancer Evolution across 20 Mammalian Species, Cell, vol.160, issue.3, pp.554-566, 2015.

B. F. Voight, S. Kudaravalli, X. Wen, and J. K. Pritchard, A Map of Recent Positive Selection in the Human Genome, PLoS Biology, vol.4, issue.3, p.e72, 2006.

U. Võsa, A. Claringbould, H. Westra, M. J. Bonder, P. Deelen et al., Unraveling the polygenic architecture of complex traits using blood eQTL metaanalysis, p.447367, 2018.

J. D. Wall, Detecting ancient admixture in humans using sequence polymorphism data, Genetics, vol.154, issue.3, pp.1271-1280, 2000.

J. D. Wall, K. E. Lohmueller, and V. Plagnol, Detecting Ancient Admixture and Estimating Demographic Parameters in Multiple Human Populations, Molecular Biology and Evolution, vol.26, issue.8, pp.1823-1827, 2009.

S. Wright, Evolution in mendelian populations, Genetics, vol.16, issue.2, pp.97-159, 1931.

B. S. Hewlett, Hunter-Gatherer Childhoods in the Congo Basin, Hunter-Gathers of the Congo Basin, pp.245-276, 2017.

G. H. Perry and P. Verdu, Genomic perspectives on the history and evolutionary ecology of tropical rainforest occupation by humans, Quaternary International, vol.448, pp.150-157, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02271452

S. Bahuchet, Changing Language, Remaining Pygmy, Human Biology, vol.84, issue.1, pp.11-43, 2012.

M. Lopez, A. Kousathanas, H. Quach, C. Harmant, P. Mouguiama-daouda et al., The demographic history and mutational load of African hunter-gatherers and farmers, Nature Ecology & Evolution, vol.2, issue.4, pp.721-730, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02271464

P. Verdu, F. Austerlitz, A. Estoup, R. Vitalis, M. Georges et al., Origins and Genetic Diversity of Pygmy Hunter-Gatherers from Western Central Africa, Current Biology, vol.19, issue.4, pp.312-318, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00397221

P. Hsieh, K. R. Veeramah, J. Lachance, S. A. Tishkoff, J. D. Wall et al., Whole-genome sequence analyses of Western Central African Pygmy hunter-gatherers reveal a complex demographic history and identify candidate genes under positive natural selection, Genome Research, vol.26, issue.3, pp.279-290, 2016.

E. Patin, G. Laval, L. B. Barreiro, A. Salas, O. Semino et al., Inferring the Demographic History of African Farmers and Pygmy Hunter?Gatherers Using a Multilocus Resequencing Data Set, PLoS Genetics, vol.5, issue.4, p.e1000448, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00397240

C. Batini, J. Lopes, D. M. Behar, F. Calafell, L. B. Jorde et al., Insights into the Demographic History of African Pygmies from Complete Mitochondrial Genomes, Molecular Biology and Evolution, vol.28, issue.2, pp.1099-1110, 2010.

K. R. Veeramah, D. Wegmann, A. Woerner, F. L. Mendez, J. C. Watkins et al., An Early Divergence of KhoeSan Ancestors from Those of Other Modern Humans Is Supported by an ABC-Based Analysis of Autosomal Resequencing Data, Molecular Biology and Evolution, vol.29, issue.2, pp.617-630, 2011.

C. Aim-e, G. Laval, E. Patin, P. Verdu, L. Chaix et al., Human genetic data reveal contrasting demographic patterns between sedentary and nomadic populations that predate the emergence of farming, Mol. Biol. Evol, vol.30, pp.2629-2644, 2013.

L. Quintana-murci, H. Quach, C. Harmant, F. Luca, B. Massonnet et al., Maternal traces of deep common ancestry and asymmetric gene flow between Pygmy hunter-gatherers and Bantu-speaking farmers, Proceedings of the National Academy of Sciences, vol.105, issue.5, pp.1596-1601, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00271643

E. Patin, K. J. Siddle, G. Laval, H. Quach, C. Harmant et al., The impact of agricultural emergence on the genetic history of African rainforest hunter-gatherers and agriculturalists, Nature Communications, vol.5, issue.1, p.3163, 2014.
URL : https://hal.archives-ouvertes.fr/halshs-01178754

J. K. Pickrell, G. Coop, J. Novembre, S. Kudaravalli, J. Z. Li et al., Signals of recent positive selection in a worldwide sample of human populations, Genome Research, vol.19, issue.5, pp.826-837, 2009.

J. P. Jarvis, L. B. Scheinfeldt, S. Soi, C. Lambert, L. Omberg et al., Patterns of Ancestry, Signatures of Natural Selection, and Genetic Association with Stature in Western African Pygmies, PLoS Genetics, vol.8, issue.4, p.e1002641, 2012.

A. B. Migliano, I. G. Romero, M. Metspalu, M. Leavesley, L. Pagani et al., Evolution of the Pygmy Phenotype: Evidence of Positive Selection from Genome-wide Scans in African, Asian, and Melanesian Pygmies, Human Biology, vol.85, issue.1-3, pp.251-284, 2013.

N. S. Becker, P. Verdu, M. Georges, P. Duquesnoy, A. Froment et al., The role of GHR and IGF1 genes in the genetic determination of African pygmies? short stature, European Journal of Human Genetics, vol.21, issue.6, pp.653-658, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02271435

T. J. Pemberton, P. Verdu, N. S. Becker, C. J. Willer, B. S. Hewlett et al., A genome scan for genes underlying adult body size differences between Central African hunter-gatherers and farmers, Human Genetics, vol.137, issue.6-7, pp.487-509, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02271453

J. Lachance, B. Vernot, C. C. Elbers, B. Ferwerda, A. Froment et al., Evolutionary History and Adaptation from High-Coverage Whole-Genome Sequences of Diverse African Hunter-Gatherers, Cell, vol.150, issue.3, pp.457-469, 2012.

I. Mendizabal, U. M. Marigorta, O. Lao, and D. Comas, Adaptive evolution of loci covarying with the human African Pygmy phenotype, Human Genetics, vol.131, issue.8, pp.1305-1317, 2012.

G. H. Perry, M. Foll, J. Grenier, E. Patin, Y. Nedelec et al., Adaptive, convergent origins of the pygmy phenotype in African rainforest hunter-gatherers, Proceedings of the National Academy of Sciences, vol.111, issue.35, pp.E3596-E3603, 2014.

C. E. Amorim, J. T. Daub, F. M. Salzano, M. Foll, and L. Excoffier, Detection of Convergent Genome-Wide Signals of Adaptation to Tropical Forests in Humans, PLOS ONE, vol.10, issue.4, p.e0121557, 2015.

S. Fan, D. E. Kelly, M. H. Beltrame, M. E. Hansen, S. Mallick et al., Correction to: African evolutionary history inferred from whole genome sequence data of 44 indigenous African populations, Genome Biology, vol.20, issue.1, p.82, 2019.

C. M. Bergey, M. Lopez, G. F. Harrison, E. Patin, J. A. Cohen et al., Polygenic adaptation and convergent evolution on growth and cardiac genetic pathways in African and Asian rainforest hunter-gatherers, Proceedings of the National Academy of Sciences, vol.115, issue.48, pp.E11256-E11263, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02352859

E. Patin, M. Lopez, R. Grollemund, P. Verdu, C. Harmant et al., Dispersals and genetic adaptation of Bantu-speaking populations in Africa and North America, Science, vol.356, issue.6337, pp.543-546, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02277558

M. Fagny, E. Patin, J. L. Macisaac, M. Rotival, T. Flutre et al., The epigenomic landscape of African rainforest hunter-gatherers and farmers, Nature Communications, vol.6, issue.1, p.10047, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01238387

D. H. Alexander, J. Novembre, and K. Lange, Fast model-based estimation of ancestry in unrelated individuals, Genome Research, vol.19, issue.9, pp.1655-1664, 2009.

A. Auton, L. D. Brooks, R. M. Durbin, E. P. Garrison, H. M. Kang et al., A global reference for human genetic variation, Nature, vol.526, issue.7571, pp.68-74, 2015.

X. Yi, Y. Liang, E. Huerta-sanchez, X. Jin, Z. X. Cuo et al., Sequencing of 50 Human Exomes Reveals Adaptation to High Altitude, Science, vol.329, issue.5987, pp.75-78, 2010.

P. C. Sabeti, P. Varilly, B. Fry, J. Lohmueller, E. Hostetter et al., Genome-wide detection and characterization of positive selection in human populations, Nature, vol.449, issue.7164, pp.913-918, 2007.

S. R. Grossman, K. G. Andersen, I. Shlyakhter, S. Tabrizi, S. Winnicki et al., Identifying Recent Adaptations in Large-Scale Genomic Data, Cell, vol.152, issue.4, pp.703-713, 2013.

K. A. Frazer, D. G. Ballinger, D. R. Cox, D. A. Hinds, L. L. Stuve et al., A second generation human haplotype map of over 3.1 million SNPs, Nature, vol.449, issue.7164, pp.851-861, 2007.

D. Gurdasani, T. Carstensen, F. Tekola-ayele, L. Pagani, I. Tachmazidou et al., The African Genome Variation Project shapes medical genetics in Africa, Nature, vol.517, issue.7534, pp.327-332, 2014.

A. Battle, C. D. Brown, B. E. Engelhardt, and S. B. Montgomery, GTEx Consortium; Laboratory, Data Analysis &Coordinating Center (LDACC)-Analysis Working Group; Statistical Methods groups-Analysis Working Group; Enhancing GTEx (eGTEx) groups; NIH Common Fund; NIH/NCI; NIH/NHGRI; NIH/NIMH

, Biospecimen Collection, Biospecimen Collection Source Site-NDRI, 2020.

, Biospecimen Collection, Biospecimen Collection Source Site-RPCI, 2020.

, Biospecimen Core Resource, Leidos Biomedical-Project Management, 2020.

J. Goecks, K. Li, D. Clements, T. G. Team, and J. Taylor, The Galaxy Track Browser: Transforming the genome browser from visualization tool to analysis tool, 2011 IEEE Symposium on Biological Data Visualization (BioVis)., 2011.

K. Ma, Scalable Data Management, Analysis, and Visualization (SDAV) Institute, Data Analysis &Coordinating Center (LDACC); NIH program management, 2019.

, Genetic effects on gene expression across human tissues, Nature, vol.550, issue.7675, pp.204-213, 2017.

K. G. Andersen, I. Shylakhter, S. Tabrizi, S. R. Grossman, C. T. Happi et al., Genome-wide scans provide evidence for positive selection of genes implicated in Lassa fever, Philosophical Transactions of the Royal Society B: Biological Sciences, vol.367, issue.1590, pp.868-877, 2012.

K. A. Fantauzzo and A. M. Christiano, Trps1 activates a network of secreted Wnt inhibitors and transcription factors crucial to vibrissa follicle morphogenesis, Development, vol.139, issue.1, pp.203-214, 2011.

M. Wuelling, F. J. Kaiser, L. A. Buelens, D. Braunholz, R. A. Shivdasani et al., Trps1, a regulator of chondrocyte proliferation and differentiation, interacts with the activator form of Gli3, Developmental Biology, vol.328, issue.1, pp.40-53, 2009.

N. Yosef, A. K. Shalek, J. T. Gaublomme, H. Jin, Y. Lee et al., Dynamic regulatory network controlling TH17 cell differentiation, Nature, vol.496, issue.7446, pp.461-468, 2013.

C. E. Dunn-fletcher, L. M. Muglia, M. Pavlicev, G. Wolf, M. Sun et al., Anthropoid primate?specific retroviral element THE1B controls expression of CRH in placenta and alters gestation length, PLOS Biology, vol.16, issue.9, p.e2006337, 2018.

B. J. Schmiedel, D. Singh, A. Madrigal, A. G. Valdovino-gonzalez, B. M. White et al., Impact of Genetic Polymorphisms on Human Immune Cell Gene Expression, Cell, vol.175, issue.6, pp.1701-1715.e16, 2018.

H. Quach, M. Rotival, J. Pothlichet, Y. Loh, M. Dannemann et al., Genetic Adaptation and Neandertal Admixture Shaped the Immune System of Human Populations, Cell, vol.167, issue.3, pp.643-656.e17, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01385620

H. T. Ngo, L. V. Pham, J. Kim, Y. Lim, and S. B. Hwang, Modulation of Mitogen-Activated Protein Kinase-Activated Protein Kinase 3 by Hepatitis C Virus Core Protein, Journal of Virology, vol.87, issue.10, pp.5718-5731, 2013.

J. Tillmanns, D. Hoffmann, Y. Habbaba, J. D. Schmitto, D. Sedding et al., Fibroblast activation protein alpha expression identifies activated fibroblasts after myocardial infarction, Journal of Molecular and Cellular Cardiology, vol.87, pp.194-203, 2015.

W. A. Andrade, A. M. Silva, V. S. Alves, A. P. Salgado, M. B. Melo et al., Early endosome localization and activity of RasGEF1b, a toll-like receptor-inducible Ras guanine-nucleotide exchange factor, Genes & Immunity, vol.11, issue.6, pp.447-457, 2010.

S. Nemec, M. Luxey, D. Jain, A. Huang-sung, T. Pastinen et al., Pitx1directly modulates the core limb development program to implement hindlimb identity, Development, vol.144, issue.18, pp.3325-3335, 2017.

S. Rüeger, A. Mcdaid, and Z. Kutalik, Evaluation and application of summary statistic imputation to discover new height-associated loci, PLOS Genetics, vol.14, issue.5, p.e1007371, 2018.

D. P. Szeto, C. Rodriguez-esteban, A. K. Ryan, S. M. O'connell, F. Liu et al., Role of the Bicoid-related homeodomain factor Pitx1 in specifying hindlimb morphogenesis and pituitary development, Genes & Development, vol.13, issue.4, pp.484-494, 1999.

M. Island, T. Mesplede, N. Darracq, M. Bandu, N. Christeff et al., Repression by Homeoprotein Pitx1 of Virus-Induced Interferon A Promoters Is Mediated by Physical Interaction and trans Repression of IRF3 and IRF7, Molecular and Cellular Biology, vol.22, issue.20, pp.7120-7133, 2002.

J. K. Pritchard, J. K. Pickrell, and G. Coop, The Genetics of Human Adaptation: Hard Sweeps, Soft Sweeps, and Polygenic Adaptation, Current Biology, vol.20, issue.4, pp.R208-R215, 2010.

E. A. Boyle, Y. I. Li, and J. K. Pritchard, An Expanded View of Complex Traits: From Polygenic to Omnigenic, Cell, vol.169, issue.7, pp.1177-1186, 2017.

C. Bycroft, C. Freeman, D. Petkova, G. Band, L. T. Elliott et al., The UK Biobank resource with deep phenotyping and genomic data, Nature, vol.562, issue.7726, pp.203-209, 2018.

A. B. Migliano, L. Vinicius, and M. M. Lahr, Life history trade-offs explain the evolution of human pygmies, Proceedings of the National Academy of Sciences, vol.104, issue.51, pp.20216-20219, 2007.

R. Walker, M. Gurven, K. Hill, A. Migliano, N. Chagnon et al., Growth rates and life histories in twenty-two small-scale societies, American Journal of Human Biology, vol.18, issue.3, pp.295-311, 2006.

M. Ashburner, C. A. Ball, J. A. Blake, D. Botstein, H. Butler et al., Gene Ontology: tool for the unification of biology, Nature Genetics, vol.25, issue.1, pp.25-29, 2000.

M. Kim, M. Rådinger, and A. M. Gilfillan, The multiple roles of phosphoinositide 3-kinase in mast cell biology, Trends in Immunology, vol.29, issue.10, pp.493-501, 2008.

T. R. Malek and I. Castro, Interleukin-2 Receptor Signaling: At the Interface between Tolerance and Immunity, Immunity, vol.33, issue.2, pp.153-165, 2010.

M. Deschamps, G. Laval, M. Fagny, Y. Itan, L. Abel et al., Genomic Signatures of Selective Pressures and Introgression from Archaic Hominins at Human Innate Immunity Genes, The American Journal of Human Genetics, vol.98, issue.1, pp.5-21, 2016.

D. Enard and D. A. Petrov, Evidence that RNA Viruses Drove Adaptive Introgression between Neanderthals and Modern Humans, Cell, vol.175, issue.2, pp.360-371.e13, 2018.

B. K. Maples, S. Gravel, E. E. Kenny, and C. D. Bustamante, RFMix: A Discriminative Modeling Approach for Rapid and Robust Local-Ancestry Inference, The American Journal of Human Genetics, vol.93, issue.2, pp.278-288, 2013.

S. Younis, M. Schönke, J. Massart, R. Hjortebjerg, E. Sundström et al., The ZBED6?IGF2 axis has a major effect on growth of skeletal muscle and internal organs in placental mammals, Proceedings of the National Academy of Sciences, vol.115, issue.9, pp.E2048-E2057, 2018.

S. Younis, W. Kamel, T. Falkeborn, H. Wang, D. Yu et al., Multiple nuclear-replicating viruses require the stress-induced protein ZC3H11A for efficient growth, Proceedings of the National Academy of Sciences, vol.115, issue.16, pp.E3808-E3816, 2018.

D. E. Humphries, G. W. Wong, D. S. Friend, M. F. Gurish, W. Qiu et al., Heparin is essential for the storage of specific granule proteases in mast cells, Nature, vol.400, issue.6746, pp.769-772, 1999.

S. Kudaravalli, J. Veyrieras, B. E. Stranger, E. T. Dermitzakis, and J. K. Pritchard, Gene Expression Levels Are a Target of Recent Natural Selection in the Human Genome, Molecular Biology and Evolution, vol.26, issue.3, pp.649-658, 2008.

P. Momeni, G. Glöckner, O. Schmidt, D. Von-holtum, B. Albrecht et al., Mutations in a new gene, encoding a zinc-finger protein, cause tricho-rhino-phalangeal syndrome type I, Nature Genetics, vol.24, issue.1, pp.71-74, 2000.

K. A. Fantauzzo, M. Tadin-strapps, Y. You, S. E. Mentzer, F. A. Baumeister et al., A position effect on TRPS1 is associated with Ambras syndrome in humans and the Koala phenotype in mice, Human Molecular Genetics, vol.17, issue.22, pp.3539-3551, 2008.

E. J. Hillmer, H. Zhang, H. S. Li, and S. S. Watowich, STAT3 signaling in immunity, Cytokine & Growth Factor Reviews, vol.31, pp.1-15, 2016.

K. Adhikari, M. Fuentes-guajardo, M. Quinto-sánchez, J. Mendoza-revilla, J. Camilo-chacón-duque et al., A genome-wide association scan implicates DCHS2, RUNX2, GLI3, PAX1 and EDAR in human facial variation, Nature Communications, vol.7, issue.1, p.11616, 2016.

M. Chopin, S. P. Preston, A. T. Lun, J. Tellier, G. K. Smyth et al., RUNX2 Mediates Plasmacytoid Dendritic Cell Egress from the Bone Marrow and Controls Viral Immunity, Cell Reports, vol.15, issue.4, pp.866-878, 2016.

A. Durvasula and S. Sankararaman, Recovering signals of ghost archaic introgression in African populations, 2018.

P. Hsieh, A. E. Woerner, J. D. Wall, J. Lachance, S. A. Tishkoff et al., Model-based analyses of whole-genome data reveal a complex evolutionary history involving archaic introgression in Central African Pygmies, Genome Research, vol.26, issue.3, pp.291-300, 2016.

Y. Foupouapouognigni, S. A. Sadeuh-mba, E. B. A-betsem, D. Rousset, A. Froment et al., Hepatitis B and C Virus Infections in the Three Pygmy Groups in Cameroon, Journal of Clinical Microbiology, vol.49, issue.2, pp.737-740, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00542157

M. P. Kowo, P. Goubau, E. Ndam, O. Njoya, S. Sasaki et al., Prevalence of hepatitis C virus and other blood-borne viruses in Pygmies and neighbouring Bantus in southern Cameroon, Transactions of the Royal Society of Tropical Medicine and Hygiene, vol.89, issue.5, pp.484-486, 1995.

J. J. Berg, A. Harpak, N. Sinnott-armstrong, A. M. Joergensen, H. Mostafavi et al., Author response: Reduced signal for polygenic adaptation of height in UK Biobank, vol.8, p.39725, 2018.

M. Sohail, R. M. Maier, A. Ganna, A. Bloemendal, A. R. Martin et al., Polygenic adaptation on height is overestimated due to uncorrected stratification in genome-wide association studies, eLife, vol.8, 2019.

M. Nordborg, N. H. Barton, and J. Hermisson, Decision letter: Polygenic adaptation on height is overestimated due to uncorrected stratification in genome-wide association studies, vol.8, p.39702, 2018.

N. S. Becker, P. Touraille, A. Froment, E. Heyer, and A. Courtiol, Short stature in African pygmies is not explained by sexual selection, Evolution and Human Behavior, vol.33, issue.6, pp.615-622, 2012.

G. H. Perry and N. J. Dominy, Evolution of the human pygmy phenotype, Trends in Ecology & Evolution, vol.24, issue.4, pp.218-225, 2009.

G. F. Harrison, J. Sanz, J. Boulais, M. J. Mina, J. Grenier et al., Natural selection contributed to immunological differences between hunter-gatherers and agriculturalists, Nature Ecology & Evolution, vol.3, issue.8, pp.1253-1264, 2019.
URL : https://hal.archives-ouvertes.fr/pasteur-02549704

K. A. Owers, P. Sjödin, C. M. Schlebusch, P. Skoglund, H. Soodyall et al., Adaptation to infectious disease exposure in indigenous Southern African populations, Proceedings of the Royal Society B: Biological Sciences, vol.284, issue.1852, p.20170226, 2017.

P. T. Hawkins and L. R. Stephens, PI3K signalling in inflammation, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1851, issue.6, pp.882-897, 2015.

B. D. Hopkins, C. Pauli, X. Du, D. G. Wang, X. Li et al., Suppression of insulin feedback enhances the efficacy of PI3K inhibitors, Nature, vol.560, issue.7719, pp.499-503, 2018.

D. A. Fruman, H. Chiu, B. D. Hopkins, S. Bagrodia, L. C. Cantley et al., The PI3K Pathway in Human Disease, Cell, vol.170, issue.4, pp.605-635, 2017.

J. I. Odegaard and A. Chawla, Pleiotropic Actions of Insulin Resistance and Inflammation in Metabolic Homeostasis, Science, vol.339, issue.6116, pp.172-177, 2013.

T. J. Smith, Insulin-Like Growth Factor-I Regulation of Immune Function: A Potential Therapeutic Target in Autoimmune Diseases?, Pharmacological Reviews, vol.62, issue.2, pp.199-236, 2010.

C. C. Chang, C. C. Chow, L. C. Tellier, S. Vattikuti, S. M. Purcell et al., Second-generation PLINK: rising to the challenge of larger and richer datasets, GigaScience, vol.4, issue.1, p.7, 2015.

A. Manichaikul, J. C. Mychaleckyj, S. S. Rich, K. Daly, M. Sale et al., Robust relationship inference in genome-wide association studies, Bioinformatics, vol.26, issue.22, pp.2867-2873, 2010.

H. Li and R. Durbin, Fast and accurate short read alignment with Burrows-Wheeler transform, Bioinformatics, vol.25, issue.14, pp.1754-1760, 2009.

M. A. Depristo, E. Banks, R. Poplin, K. V. Garimella, J. R. Maguire et al., A framework for variation discovery and genotyping using next-generation DNA sequencing data, Nature Genetics, vol.43, issue.5, pp.491-498, 2011.

O. Delaneau, J. Zagury, and J. Marchini, Improved whole-chromosome phasing for disease and population genetic studies, Nature Methods, vol.10, issue.1, pp.5-6, 2013.

B. N. Howie, P. Donnelly, and J. Marchini, A Flexible and Accurate Genotype Imputation Method for the Next Generation of Genome-Wide Association Studies, PLoS Genetics, vol.5, issue.6, p.e1000529, 2009.

B. K. Bulik-sullivan, P. Loh, H. K. Finucane, S. Ripke, J. Yang et al., LD Score regression distinguishes confounding from polygenicity in genome-wide association studies, Nature Genetics, vol.47, issue.3, pp.291-295, 2015.

D. V. Klopfenstein, L. Zhang, B. S. Pedersen, F. Ramírez, A. Warwick-vesztrocy et al., GOATOOLS: A Python library for Gene Ontology analyses, Scientific Reports, vol.8, issue.1, 2018.

S. R. Browning and B. L. Browning, Rapid and Accurate Haplotype Phasing and Missing-Data Inference for Whole-Genome Association Studies By Use of Localized Haplotype Clustering, The American Journal of Human Genetics, vol.81, issue.5, pp.1084-1097, 2007.

E. V. Davydov, D. L. Goode, M. Sirota, G. M. Cooper, A. Sidow et al., Identifying a High Fraction of the Human Genome to be under Selective Constraint Using GERP++, PLoS Computational Biology, vol.6, issue.12, p.e1001025, 2010.

G. A. Van-der-auwera, M. O. Carneiro, C. Hartl, R. Poplin, G. Del-angel et al., From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline, Curr. Protoc. Bioinformatics, vol.43, pp.1-33, 2013.

G. R. Abecasis, D. Altshuler, A. Auton, L. D. Brooks, R. M. Durbin et al., A map of human genome variation from population-scale sequencing, Nature, vol.467, issue.7319, pp.1061-1073, 2010.

D. M. Altshuler, R. A. Gibbs, L. Peltonen, D. M. Altshuler, R. A. Gibbs et al., Integrating common and rare genetic variation in diverse human populations, Nature, vol.467, issue.7311, pp.52-58, 2010.

S. Purcell, B. Neale, K. Todd-brown, L. Thomas, M. A. Ferreira et al., PLINK: A Tool Set for Whole-Genome Association and Population-Based Linkage Analyses, The American Journal of Human Genetics, vol.81, issue.3, pp.559-575, 2007.

L. L. Cavalli-sforza, African Pygmies, 1986.

J. M. Diamond, Why are pygmies small?, Nature, vol.354, issue.6349, pp.111-112, 1991.

B. T. Shea and R. C. Bailey, Allometry and adaptation of body proportions and stature in African pygmies, American Journal of Physical Anthropology, vol.100, issue.3, pp.311-340, 1996.

A. Froment, Hunter-gatherers: an interdisciplinary perspective, Hunter-Gatherers: An Interdisciplinary Perspective, pp.239-266, 2001.

N. S. Becker, P. Verdu, B. Hewlett, and S. Pavard, Can Life History Trade-Offs Explain the Evolution of Short Stature in Human Pygmies? A Response to Migliano et al. (2007), Human Biology, vol.82, issue.1, pp.17-27, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00674368

. International-hapmap-consortium, A haplotype map of the human genome, Nature, vol.437, issue.7063, pp.1299-1320, 2005.

, resource identifier, pp.140-1006

. N/a, http://www.avensonline.org/fulltextarticles/JSUR-2332-4139-S1-0001.html, Journal of Surgery, vol.87, pp.01-07, 2015.

, Subscription and Copyright Information, Trends in Cell Biology, vol.29, issue.10, p.e1, 2019.

M. Lopez, J. Choin, M. Sikora, K. Siddle, C. Harmant et al., Genomic Evidence for Local Adaptation of Hunter-Gatherers to the African Rainforest, Current Biology, vol.29, issue.17, pp.2926-2935.e4, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02271461