D. Dobbelaere and V. Heussler, Transformation of leukocytes by Theileria parva and T. annulata, Annu Rev Microbiol, vol.53, p.10547684, 1999.

N. Echebli, M. Mhadhbi, M. Chaussepied, C. Vayssettes, D. Santo et al., Engineering attenuated virulence of a Theileria annulata-infected macrophage, PLoS Negl Trop Dis, vol.8, issue.11, p.25375322, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-01089412

R. P. Somerville, R. E. Adamson, C. G. Brown, and F. R. Hall, Metastasis of Theileria annulata macroschizontinfected cells in scid mice is mediated by matrix metalloproteinases, Parasitology, vol.116, p.9550215, 1998.

K. Cheeseman and J. B. Weitzman, Host-parasite interactions: an intimate epigenetic relationship, Cell Microbiol, vol.17, issue.8, p.26096716, 2015.

M. Chaussepied, N. Janski, M. Baumgartner, R. Lizundia, K. Jensen et al., TGF-b2 induction regulates invasiveness of Theileria-transformed leukocytes and disease susceptibility, PLoS Pathog, vol.6, issue.11, p.21124992, 2010.

H. A. Baylis, A. Megson, and R. Hall, Infection with Theileria annulata induces expression of matrix metalloproteinase 9 and transcription factor AP-1 in bovine leucocytes, Mol Biochem Parasitol, vol.69, issue.2, p.7770085, 1995.

R. Lizundia, M. Chaussepied, M. Huerre, D. Werling, D. Santo et al., c-Jun NH2-terminal kinase/c-Jun signaling promotes survival and metastasis of B lymphocytes transformed by Theileria, Cancer Res, vol.66, issue.12, p.16778183, 2006.

A. M. Cock-rada, S. Medjkane, N. Janski, N. Yousfi, M. Perichon et al., SMYD3 promotes cancer invasion by epigenetic upregulation of the metalloproteinase MMP-9, Cancer Res, vol.72, issue.3, p.22194464, 2012.

J. Marsolier, S. Pineau, S. Medjkane, M. Perichon, Q. Yin et al., OncomiR addiction is generated by a miR-155 feedback loop in Theileria-transformed leukocytes, PLoS Pathog, vol.9, issue.4, p.23637592, 2013.

M. I. Aslam, K. Taylor, J. H. Pringle, and J. S. Jameson, MicroRNAs are novel biomarkers of colorectal cancer, Br J Surg, vol.96, issue.7, p.19526617, 2009.

N. El-murr, Z. Abidi, K. Wanherdrick, M. Svrcek, M. P. Gaub et al., MiRNA genes constitute new targets for microsatellite instability in colorectal cancer, PLoS One, vol.7, issue.2, p.22348132, 2012.

C. Delay, F. Calon, P. Mathews, and S. S. Hebert, Alzheimer-specific variants in the 3'UTR of Amyloid precursor protein affect microRNA function, Mol Neurodegener, vol.6, p.21982160, 2011.

J. Kruger and M. Rehmsmeier, RNAhybrid: microRNA target prediction easy, fast and flexible, Nucleic Acids Res, vol.34, p.16845047, 2006.

M. Satoh, Y. Minami, Y. Takahashi, T. Tabuchi, and M. Nakamura, A cellular microRNA, let-7i, is a novel biomarker for clinical outcome in patients with dilated cardiomyopathy, J Card Fail, vol.17, issue.11, p.22041329, 2011.

F. Moretti, R. Thermann, and M. W. Hentze, Mechanism of translational regulation by miR-2 from sites in the 5' untranslated region or the open reading frame, RNA, vol.16, issue.12, p.20966199, 2010.

S. Kulkarni, R. Savan, Y. Qi, X. Gao, Y. Y. Bass et al., Differential microRNA regulation of HLA-C expression and its association with HIV control, Nature, vol.472, issue.7344, p.21499264, 2011.

A. M. Duursma, M. Kedde, M. Schrier, C. Le-sage, and R. Agami, miR-148 targets human DNMT3b protein coding region, RNA, vol.14, issue.5, p.18367714, 2008.

M. A. Hakimi and K. W. Deitsch, Epigenetics in Apicomplexa: control of gene expression during cell cycle progression, differentiation and antigenic variation, Curr Opin Microbiol, vol.10, issue.4, p.17719264, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00171528

C. C. Judice, C. Bourgard, A. C. Kayano, L. Albrecht, and F. T. Costa, MicroRNAs in the Host-Apicomplexan Parasites Interactions: A Review of Immunopathological Aspects, Front Cell Infect Microbiol, vol.6, p.26870701, 2016.

A. Eulalio, L. Schulte, and J. Vogel, The mammalian microRNA response to bacterial infections, RNA Biol, vol.9, issue.6, p.22664920, 2012.

C. Staedel and F. Darfeuille, MicroRNAs and bacterial infection, Cell Microbiol, vol.15, issue.9, p.23795564, 2013.

B. R. Cullen, Viruses and microRNAs: RISCy interactions with serious consequences, Genes Dev, vol.25, issue.18, p.21896651, 2011.

G. Lamonte, N. Philip, J. Reardon, J. R. Lacsina, W. Majoros et al., Translocation of sickle cell erythrocyte microRNAs into Plasmodium falciparum inhibits parasite translation and contributes to malaria resistance, Cell Host Microbe, vol.12, issue.2, p.22901539, 2012.

A. Schober, M. Nazari-jahantigh, Y. Wei, K. Bidzhekov, F. Gremse et al., MicroRNA-126-5p promotes endothelial proliferation and limits atherosclerosis by suppressing Dlk1, Nat Med, vol.20, issue.4, p.24584117, 2014.

A. J. Whitmarsh, The JIP family of MAPK scaffold proteins, Biochem Soc Trans, vol.34, pp.828-860, 2006.

M. Dickens, J. S. Rogers, J. Cavanagh, A. Raitano, Z. Xia et al., A cytoplasmic inhibitor of the JNK signal transduction pathway, Science, vol.277, issue.5326, p.9235893, 1997.

J. Yasuda, A. J. Whitmarsh, J. Cavanagh, M. Sharma, and R. J. Davis, The JIP group of mitogen-activated protein kinase scaffold proteins, Mol Cell Biol, vol.19, issue.10, p.10490659, 1999.

D. Nihalani, H. N. Wong, and L. B. Holzman, Recruitment of JNK to JIP1 and JNK-dependent JIP1 phosphorylation regulates JNK module dynamics and activation, J Biol Chem, vol.278, issue.31, p.12756254, 2003.

W. G. Stetler-stevenson, S. Aznavoorian, and L. A. Liotta, Tumor cell interactions with the extracellular matrix during invasion and metastasis, Annu Rev Cell Biol, vol.9, pp.541-73, 1993.

E. Bossy-wetzel, L. Bakiri, and M. Yaniv, Induction of apoptosis by the transcription factor c-Jun, EMBO J, vol.16, issue.7, p.9130714, 1997.

K. Kovary and R. Bravo, Existence of different Fos/Jun complexes during the G0-to-G1 transition and during exponential growth in mouse fibroblasts: differential role of Fos proteins, Mol Cell Biol, vol.12, issue.11, p.1406676, 1992.

L. Vandel, N. Montreau, E. Vial, C. M. Pfarr, B. Binetruy et al., Stepwise transformation of rat embryo fibroblasts: c-Jun, JunB, or JunD can cooperate with Ras for focus formation, but a c-Jun-containing heterodimer is required for immortalization, Mol Cell Biol, vol.16, issue.5, p.8628254, 1996.
URL : https://hal.archives-ouvertes.fr/ensl-00179738

M. Haidar, J. Whitworth, G. Noe, W. Q. Liu, M. Vidal et al., TGF-beta2 induces Grb2 to recruit PI3-K to TGF-RII that activates JNK/AP-1-signaling and augments invasiveness of Theileria-transformed macrophages, Sci Rep, vol.5, p.15688, 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.25516281, 2014.

T. J. Hardcastle and K. A. Kelly, baySeq: empirical Bayesian methods for identifying differential expression in sequence count data, BMC Bioinformatics, vol.11, p.20698981, 2010.

F. Cordero, M. Beccuti, M. Arigoni, S. Donatelli, and R. A. Calogero, Optimizing a massive parallel sequencing workflow for quantitative miRNA expression analysis, PLoS One, vol.7, issue.2, p.22363693, 2012.

J. E. Fish, M. M. Santoro, S. U. Morton, S. Yu, R. F. Yeh et al., miR-126 regulates angiogenic signaling and vascular integrity, Dev Cell, vol.15, issue.2, p.18694566, 2008.

J. Meister and M. H. Schmidt, miR-126 and miR-126*: new players in cancer, ScientificWorldJournal, vol.10, p.20953557, 2010.

S. Wang, A. B. Aurora, B. A. Johnson, X. Qi, J. Mcanally et al., The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis, Dev Cell, vol.15, issue.2, p.18694565, 2008.

D. Schmitter, J. Filkowski, A. Sewer, R. S. Pillai, E. J. Oakeley et al., Effects of Dicer and Argonaute down-regulation on mRNA levels in human HEK293 cells, Nucleic Acids Res, vol.34, issue.17, p.16971455, 2006.

M. Metheni, N. Echebli, M. Chaussepied, C. Ransy, C. Chereau et al., The level of H(2)O(2) type oxidative stress regulates virulence of Theileria-transformed leukocytes, Cell Microbiol, vol.16, issue.2, p.24112286, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-01075079

M. Yang, A. D. Haase, F. K. Huang, G. Coulis, K. D. Rivera et al., Dephosphorylation of tyrosine 393 in argonaute 2 by protein tyrosine phosphatase 1B regulates gene silencing in oncogenic RASinduced senescence, Mol Cell, vol.55, issue.5, p.25175024, 2014.

B. J. Goldstein, A. Bittner-kowalczyk, M. F. White, and M. Harbeck, Tyrosine dephosphorylation and deactivation of insulin receptor substrate-1 by protein-tyrosine phosphatase 1B. Possible facilitation by the formation of a ternary complex with the Grb2 adaptor protein, J Biol Chem, vol.275, issue.6, p.10660596, 2000.

M. Chaussepied, D. Lallemand, M. F. Moreau, R. Adamson, R. Hall et al., Upregulation of Jun and Fos family members and permanent JNK activity lead to constitutive AP-1 activation in Theileria-transformed leukocytes, Mol Biochem Parasitol, vol.94, issue.2, p.9747972, 1998.

Y. Galley, G. Hagens, I. Glaser, W. Davis, M. Eichhorn et al., Jun NH2-terminal kinase is constitutively activated in T cells transformed by the intracellular parasite Theileria parva, Proc Natl Acad Sci U S A, vol.94, issue.10, p.9144200, 1997.

A. M. Bode and Z. Dong, The functional contrariety of JNK, Mol Carcinog, vol.46, issue.8, p.17538955, 2007.

R. Adamson, M. Logan, J. Kinnaird, G. Langsley, and R. Hall, Loss of matrix metalloproteinase 9 activity in Theileria annulata-attenuated cells is at the transcriptional level and is associated with differentially expressed AP-1 species, Mol Biochem Parasitol, vol.106, issue.1, p.10743610, 2000.

A. M. Monteys, R. M. Spengler, J. Wan, L. Tecedor, K. A. Lennox et al., Structure and activity of putative intronic miRNA promoters, RNA, vol.16, issue.3, pp.495-505, 2010.

S. Singh, N. Khatri, A. Manuja, R. D. Sharma, D. V. Malhotra et al., Impact of field vaccination with a Theileria annulata schizont cell culture vaccine on the epidemiology of tropical theileriosis, Vet Parasitol, vol.101, issue.2, p.11587838, 2001.

S. Morzaria, P. Roeder, D. Roberts, D. Chasey, and T. Drew, Characteristics of a continuous suspension cell line derived from a calf with sporadic leukosis, pp.519-547, 1982.

B. R. Shiels, C. Mcdougall, A. Tait, and C. G. Brown, Identification of infection-associated antigens in Theileria annulata transformed cells, Parasite Immunol, vol.8, issue.1, p.2421227, 1986.

M. F. Moreau, J. L. Thibaud, L. B. Miled, M. Chaussepied, M. Baumgartner et al., Theileria annulata in CD5(+) macrophages and B1 B cells, Infect Immun, vol.67, issue.12, p.10569790, 1999.
URL : https://hal.archives-ouvertes.fr/pasteur-02024445

G. H. Theilen, J. D. Rush, N. Wa, D. L. Dungworth, R. J. Munn et al., Bovine leukemia: establishment and morphologic characterization of continuous cell suspension culture, BL-1, J Natl Cancer Inst, vol.40, issue.4, p.4171552, 1968.

S. Andrews, FastQC: a quality control tool for high throughput sequence data, 2010.

J. Wu, Q. Liu, X. Wang, J. Zheng, T. Wang et al., mirTools 2.0 for non-coding RNA discovery, profiling, and functional annotation based on high-throughput sequencing, RNA Biol, vol.10, issue.7, p.23778453, 2013.

R. Li, Y. Li, K. Kristiansen, and J. Wang, SOAP: short oligonucleotide alignment program, Bioinformatics, vol.24, issue.5, pp.713-717, 2008.

A. Kozomara, S. Griffiths-jones, and . Mirbase, annotating high confidence microRNAs using deep sequencing data, Nucleic Acids Res, vol.42, p.24275495, 2014.

E. P. Nawrocki, S. W. Burge, A. Bateman, J. Daub, R. Y. Eberhardt et al., Rfam 12.0: updates to the RNA families database, Nucleic Acids Res, vol.43, p.25392425, 2015.

M. W. Pfaffl, A new mathematical model for relative quantification in real-time RT-PCR, Nucleic Acids Res, vol.29, issue.9, p.11328886, 2001.

B. Okutucu, A. Dincer, O. Habib, and F. Zihnioglu, Comparison of five methods for determination of total plasma protein concentration, J Biochem Biophys Methods, vol.70, issue.5, p.17597224, 2007.