T. H. Mogensen, Pathogen Recognition and Inflammatory Signaling in Innate Immune Defenses, Clinical Microbiology Reviews, vol.22, issue.2, pp.240-273, 2009.
DOI : 10.1128/CMR.00046-08

H. H. Niller and J. Minarovits, Patho-epigenetics of Infectious Diseases Caused by Intracellular Bacteria, Patho-Epigenetics Infect. Dis
DOI : 10.1007/978-3-319-24738-0_6

S. M. Peterson, J. A. Thompson, M. L. Ufkin, P. Sathyanarayana, L. Liaw et al., Common features of microRNA target prediction tools, Frontiers in Genetics, vol.5, pp.1-10, 2014.
DOI : 10.3389/fgene.2014.00023

P. Bouloc and F. Repoila, Fresh layers of RNA-mediated regulation in Gram-positive bacteria, Current Opinion in Microbiology, vol.30, pp.30-35, 2016.
DOI : 10.1016/j.mib.2015.12.008

URL : https://hal.archives-ouvertes.fr/hal-01412635

I. Caldelari, Y. Chao, P. Romby, and J. Vogel, RNA-Mediated Regulation in Pathogenic Bacteria, Cold Spring Harbor Perspectives in Medicine, vol.3, issue.9, 2013.
DOI : 10.1101/cshperspect.a010298

N. Sesto, M. Koutero, and P. Cossart, infection, Future Microbiology, vol.9, issue.9, 2014.
DOI : 10.2217/fmb.14.79

URL : https://hal.archives-ouvertes.fr/pasteur-01161885

E. G. Wagner and P. Romby, Small RNAs in Bacteria and Archaea, Adv. Genet, 2015.
DOI : 10.1016/bs.adgen.2015.05.001

Y. Ding, Z. Zhang, Y. Liu, C. Shi, J. Zhang et al., Relationship of long noncoding RNA and viruses, Genomics, vol.107, issue.4, 2016.
DOI : 10.1016/j.ygeno.2016.01.007

C. J. Brown, J. L. Ballabio, R. G. Rupert, M. Lafreniere, R. Grompe et al., A gene from the region of the human X inactivation centre is expressed exclusively from the inactive X chromosome, Nature, vol.349, issue.6304, pp.349-387, 1991.
DOI : 10.1038/349038a0

N. Brockdorff, A. Ashworth, G. F. Kay, V. M. Mccabe, D. P. Norris et al., The product of the mouse Xist gene is a 15 kb inactive X-specific transcript containing no conserved ORF and located in the nucleus, Cell, vol.71, issue.3, pp.515-526, 1992.
DOI : 10.1016/0092-8674(92)90519-I

M. Quan, J. Chen, and D. Zhang, Exploring the Secrets of Long Noncoding RNAs, International Journal of Molecular Sciences, vol.16, issue.3, pp.5467-5496, 2015.
DOI : 10.3390/ijms16035467

S. Geisler and J. Coller, RNA in unexpected places: long non-coding RNA functions in diverse cellular contexts, Nature Reviews Molecular Cell Biology, vol.472, issue.11, pp.699-712, 2013.
DOI : 10.1038/nrm3679

M. Guttman and J. L. Rinn, Modular regulatory principles of large non-coding RNAs, Nature, vol.136, issue.7385, pp.339-385, 2012.
DOI : 10.1038/nature10887

R. C. Friedman, K. K. Farh, C. B. Burge, and D. P. , Most mammalian mRNAs are conserved targets of microRNAs, Genome Research, vol.19, issue.1, pp.92-105, 2009.
DOI : 10.1101/gr.082701.108

R. M. O-'connell, D. S. Rao, A. A. Chaudhuri, D. Baltimore, M. J. Bueno et al., Physiological and pathological roles for microRNAs in the immune system, Nature Reviews Immunology, vol.435, issue.2, pp.111-122, 2008.
DOI : 10.1038/nri2708

M. Kedde and R. Agami, Interplay between microRNAs and RNA-binding proteins determines developmental processes, Cell Cycle, vol.7, issue.7, pp.899-9035644, 2008.
DOI : 10.4161/cc.7.7.5644

A. V. Orang, R. Safaralizadeh, and M. Kazemzadeh-bavili, Mechanisms of miRNA-mediated gene regulation from common downregulation to mRNA-specific upregulation, Int. J. Genomics, vol.2014970607, pp.10-1155, 2014.

J. E. Parkes, P. J. Day, H. Chinoy, and J. A. Lamb, The role of microRNAs in the idiopathic inflammatory myopathies, Current Opinion in Rheumatology, vol.27, issue.6, pp.608-623, 2015.
DOI : 10.1097/BOR.0000000000000225

P. Gurha, MicroRNAs in cardiovascular disease, Current Opinion in Cardiology, vol.31, issue.3, pp.31-2016
DOI : 10.1097/HCO.0000000000000280

W. Huang, J. Lin, and H. Zhang, miR-126: A novel regulator in colon cancer (Review), Biomedical Reports, 2015.
DOI : 10.3892/br.2015.549

L. Navarro, P. Dunoyer, F. Jay, B. Arnold, N. Dharmasiri et al., A Plant miRNA Contributes to Antibacterial Resistance by Repressing Auxin Signaling, Science, vol.312, issue.5772, pp.3121-436, 2006.
DOI : 10.1126/science.1126088

URL : https://hal.archives-ouvertes.fr/hal-00093086

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.12481-12487, 2006.
DOI : 10.1073/pnas.0605298103

W. Jin, E. M. Ibeagha-awemu, G. Liang, F. Beaudoin, X. Zhao et al., Transcriptome microRNA profiling of bovine mammary epithelial cells challenged with Escherichia coli or Staphylococcus aureus bacteria reveals pathogen directed microRNA expression profiles, BMC Genomics, vol.15, issue.1, pp.181-191, 2014.
DOI : 10.1093/nar/22.22.4673

F. R. Zeng, L. J. Tang, Y. He, and R. C. Garcia, An update on the role of miRNA-155 in pathogenic microbial infections, Microbes and Infection, vol.17, issue.9, pp.613-621, 2015.
DOI : 10.1016/j.micinf.2015.05.007

C. Staedel and F. Darfeuille, MicroRNAs and bacterial infection, Cellular Microbiology, vol.7, issue.9, pp.1496-1507, 2013.
DOI : 10.1111/cmi.12159

A. Eulalio, L. Schulte, and J. Vogel, The mammalian microRNA response to bacterial infections, RNA Biology, vol.9, issue.6, pp.742-750, 2012.
DOI : 10.4161/rna.20018

C. Maudet, M. Mano, and A. Eulalio, MicroRNAs in the interaction between host and bacterial pathogens, FEBS Letters, vol.2, issue.22, pp.4140-4147, 2014.
DOI : 10.1016/j.febslet.2014.08.002

K. J. Siddle, L. Tailleux, M. Deschamps, Y. H. Loh, C. Deluen et al., Bacterial Infection Drives the Expression Dynamics of microRNAs and Their isomiRs, PLOS Genetics, vol.25, issue.3, pp.1-25, 2015.
DOI : 10.1371/journal.pgen.1005064.s020

URL : https://hal.archives-ouvertes.fr/hal-01236327

S. Griffiths-jones, J. H. Hui, A. Marco, and M. Ronshaugen, MicroRNA evolution by arm switching, EMBO reports, vol.2, issue.2, pp.172-177, 2011.
DOI : 10.1111/j.1525-142X.2008.00302.x

L. N. Schulte, A. J. Westermann, and J. Vogel, Differential activation and functional specialization of miR-146 and miR-155 in innate immune sensing, Nucleic Acids Research, vol.41, issue.1, pp.542-553, 2013.
DOI : 10.1093/nar/gks1030

C. U. Duerr and M. W. Hornef, The mammalian intestinal epithelium as integral player in the establishment and maintenance of hostmicrobial homeostasis, Semin. Immunol, vol.24, 2012.

R. M. O-'connell, K. D. Taganov, M. P. Boldin, G. Cheng, and D. Baltimore, MicroRNA-155 is induced during the macrophage inflammatory response, Proceedings of the National Academy of Sciences, vol.104, issue.5, pp.1604-1613, 2007.
DOI : 10.1073/pnas.0610731104

E. Tili, J. Michaille, A. Cimino, S. Costinean, C. D. Dumitru et al., Modulation of miR-155 and miR-125b Levels following Lipopolysaccharide/TNF-?? Stimulation and Their Possible Roles in Regulating the Response to Endotoxin Shock, The Journal of Immunology, vol.179, issue.8, pp.5082-5091, 2007.
DOI : 10.4049/jimmunol.179.8.5082

R. M. O-'connell, A. A. Chaudhuri, D. S. Rao, and D. Baltimore, Inositol phosphatase SHIP1 is a primary target of miR-155, Proceedings of the National Academy of Sciences, vol.106, issue.17, pp.7113-7121, 2009.
DOI : 10.1073/pnas.0902636106

T. J. Cremer, D. H. Ravneberg, C. D. Clay, M. G. Piper-hunter, C. B. Marsh et al., MiR-155 Induction by F. novicida but Not the Virulent F. tularensis Results in SHIP Down-Regulation and Enhanced Pro-Inflammatory Cytokine Response, PLoS ONE, vol.26, issue.12, 2009.
DOI : 10.1371/journal.pone.0008508.g007

A. T. Billeter, J. Hellmann, H. Roberts, D. Druen, S. A. Gardner et al., MicroRNA-155 potentiates the inflammatory response in hypothermia by suppressing IL-10 production, The FASEB Journal, vol.28, issue.12, pp.5322-5336, 2014.
DOI : 10.1096/fj.14-258335

M. Kurowska-stolarska, S. Alivernini, L. E. Ballantine, D. L. Asquith, N. L. Millar et al., MicroRNA-155 as a proinflammatory regulator in clinical and experimental arthritis, Proceedings of the National Academy of Sciences, vol.108, issue.27, pp.11193-11201, 2011.
DOI : 10.1073/pnas.1019536108

C. E. Mccoy, F. J. Sheedy, J. E. Qualls, S. L. Doyle, S. R. Quinn et al., IL-10 Inhibits miR-155 Induction by Toll-like Receptors, Journal of Biological Chemistry, vol.285, issue.27, pp.20492-20498, 2010.
DOI : 10.1074/jbc.M110.102111

R. Hu, D. A. Kagele, T. B. Huffaker, M. C. Runtsch, M. Alexander et al., miR-155 Promotes T Follicular Helper Cell Accumulation during Chronic, Low-Grade Inflammation, Immunity, vol.41, issue.4, pp.605-619, 2014.
DOI : 10.1016/j.immuni.2014.09.015

G. Wan, W. Xie, Z. Liu, W. Xu, Y. Lao et al., is a potent autophagy inducer by targeting multiple players in the MTOR pathway, Autophagy, vol.14, issue.1, pp.70-79, 2014.
DOI : 10.1371/journal.pone.0020849

J. Wang, K. Yang, L. Zhou, M. Wu, Y. Wu et al., MicroRNA-155 Promotes Autophagy to Eliminate Intracellular Mycobacteria by Targeting Rheb, PLoS Pathogens, vol.277, issue.10, 2013.
DOI : 10.1371/journal.ppat.1003697.g009

S. Clare, V. John, A. W. Walker, J. L. Hill, C. Abreu-goodger et al., Enhanced Susceptibility to Citrobacter rodentium Infection in MicroRNA-155-Deficient Mice, Infection and Immunity, vol.81, issue.3, pp.723-73200969, 2013.
DOI : 10.1128/IAI.00969-12

E. F. Lind, A. R. Elford, and P. S. Ohashi, Micro-RNA 155 Is Required for Optimal CD8+ T Cell Responses to Acute Viral and Intracellular Bacterial Challenges, The Journal of Immunology, vol.190, issue.3, pp.1210-1216, 2013.
DOI : 10.4049/jimmunol.1202700

A. Rodriguez, E. Vigorito, S. Clare, M. Warren, P. Couttet et al., Requirement of bic/microRNA-155 for Normal Immune Function, Science, vol.316, issue.5824, pp.981-608, 2007.
DOI : 10.1126/science.1139253

T. Imaizumi, H. Tanaka, A. Tajima, Y. Yokono, T. Matsumiya et al., IFN-?? and TNF-?? Synergistically Induce microRNA-155 Which Regulates TAB2/IP-10 Expression in Human Mesangial Cells, American Journal of Nephrology, vol.32, issue.5, pp.462-468, 2010.
DOI : 10.1159/000321365

X. M. Chen, P. L. Splinter, S. P. O-'hara, and N. F. Larusso, Infection, Journal of Biological Chemistry, vol.282, issue.39, pp.28929-28938, 2007.
DOI : 10.1074/jbc.M702633200

URL : https://hal.archives-ouvertes.fr/pasteur-01179295

B. Izar, G. K. Mannala, M. A. Mraheil, T. Chakraborty, and T. Hain, microRNA Response to Listeria monocytogenes Infection in Epithelial Cells, International Journal of Molecular Sciences, vol.13, issue.12, pp.1173-1185, 2012.
DOI : 10.3390/ijms13011173

L. N. Schulte, A. Eulalio, H. Mollenkopf, R. Reinhardt, and J. Vogel, family, The EMBO Journal, vol.88, issue.10, pp.1977-1989, 2011.
DOI : 10.1038/emboj.2011.94

G. Gen-teng, W. Hong-wang, Y. Dai, S. Wang, Y. Xiang-chu et al., Let-7b Is Involved in the Inflammation and Immune Responses Associated with Helicobacter pylori Infection by Targeting Toll-Like Receptor 4, PLoS One, vol.8, 2013.

G. Hu, A. Y. Gong, A. L. Roth, B. Q. Huang, H. D. Ward et al., Release of Luminal Exosomes Contributes to TLR4-Mediated Epithelial Antimicrobial Defense, PLoS Pathogens, vol.270, issue.4, 2013.
DOI : 10.1371/journal.ppat.1003261.s006

D. Iliopoulos, H. A. Hirsch, and K. Struhl, An Epigenetic Switch Involving NF-??B, Lin28, Let-7 MicroRNA, and IL6 Links Inflammation to Cell Transformation, Cell, vol.139, issue.4, 2009.
DOI : 10.1016/j.cell.2009.10.014

K. Matsushima, H. Isomoto, N. Inoue, T. Nakayama, T. Hayashi et al., MicroRNA signatures in Helicobacter pylori-infected gastric mucosa, International Journal of Cancer, vol.18, issue.2, pp.361-370, 2011.
DOI : 10.1002/ijc.25348

Y. Hayashi, M. Tsujii, J. Wang, J. Kondo, T. Akasaka et al., CagA mediates epigenetic regulation to attenuate let-7 expression in Helicobacter pylori-related carcinogenesis, Gut, pp.1536-1546, 2012.

M. Kumar, S. K. Sahu, R. Kumar, A. Subuddhi, R. K. Maji et al., MicroRNA let-7 Modulates the Immune Response to Mycobacterium tuberculosis Infection via Control of A20, an Inhibitor of the NF-??B Pathway, Cell Host & Microbe, vol.17, issue.3, pp.345-356, 2015.
DOI : 10.1016/j.chom.2015.01.007

A. Marcais, R. Blevins, J. Graumann, A. Feytout, G. Dharmalingam et al., microRNA-mediated regulation of mTOR complex components facilitates discrimination between activation and anergy in CD4 T cells, The Journal of Experimental Medicine, vol.162, issue.11, pp.2281-95, 2014.
DOI : 10.1084/jem.20080707

URL : https://hal.archives-ouvertes.fr/pasteur-01091088

F. Ma, S. Xu, X. Liu, Q. Zhang, X. Xu et al., The microRNA miR-29 controls innate and adaptive immune responses to intracellular bacterial infection by targeting interferon-??, Nature Immunology, vol.51, issue.9, pp.861-869, 2011.
DOI : 10.1038/ni.1742

A. K. Schnitger, A. Machova, R. U. Mueller, A. Androulidaki, B. Schermer et al., Listeria monocytogenes Infection in Macrophages Induces Vacuolar-Dependent Host miRNA Response, PLoS ONE, vol.6, issue.11, 2011.
DOI : 10.1371/journal.pone.0027435.s009

S. Liu, A. P. Da-cunha, R. M. Rezende, R. Cialic, Z. Wei et al., The Host Shapes the Gut Microbiota via Fecal MicroRNA, Cell Host & Microbe, vol.19, issue.1, pp.32-43, 2016.
DOI : 10.1016/j.chom.2015.12.005

J. A. Weber, D. H. Baxter, S. Zhang, D. Y. Huang, K. H. Huang et al., The MicroRNA Spectrum in 12 Body Fluids, Clinical Chemistry, vol.56, issue.11, pp.1733-1741, 2010.
DOI : 10.1373/clinchem.2010.147405

M. Alexander, R. Hu, M. C. Runtsch, D. Kagele, T. L. Mosbruger et al., Exosome-delivered microRNAs modulate the inflammatory response to endotoxin, Nature Communications, vol.103
DOI : 10.1038/ncomms8321

K. Wu, C. Zhu, Y. Yao, X. Wang, J. Song et al., MicroRNA-155-enhanced autophagy in human gastric epithelial cell in response to Helicobacter Pylori, Saudi Journal of Gastroenterology, vol.22, issue.1, pp.30-36, 173756.
DOI : 10.4103/1319-3767.173756

C. Chakraborty and S. Das, Profiling cell-free and circulating miRNA: a clinical diagnostic tool for different cancers, Tumor Biology, vol.10, issue.2, pp.10-1007, 2016.
DOI : 10.1155/2011/597145

C. Guay and R. Regazzi, Circulating microRNAs as novel biomarkers for diabetes mellitus, Nature Reviews Endocrinology, vol.39, issue.9, 2013.
DOI : 10.1038/nrendo.2013.86

B. Ueberberg, M. Kohns, E. Mayatepek, and M. Jacobsen, Are microRNAs suitable biomarkers of immunity to tuberculosis?, Molecular and Cellular Pediatrics, vol.67, issue.Suppl, 2014.
DOI : 10.1186/s40348-014-0008-9

C. Hsieh, C. Rau, J. C. Jeng, Y. Chen, T. Lu et al., Whole blood-derived microRNA signatures in mice exposed to lipopolysaccharides, Journal of Biomedical Science, vol.19, issue.1, pp.10-1186, 2012.
DOI : 10.1007/s00134-002-1397-0

C. Hsieh, J. C. Yang, J. C. Jeng, Y. Chen, T. Lu et al., Circulating microRNA signatures in mice exposed to lipoteichoic acid, Journal of Biomedical Science, vol.20, issue.1
DOI : 10.1093/nar/gkq601

C. Rau, S. Wu, J. Yang, T. Lu, Y. Wu et al., Profiling the circulating miRNAs in mice exposed to grampositive and gram-negative bacteria by Illumina small RNA deep sequencing, J. Biomed. Sci, vol.22

M. C. Runtsch, J. L. Round, and R. M. , MicroRNAs and the regulation of intestinal homeostasis, Frontiers in Genetics, vol.11, issue.e61250, 2014.
DOI : 10.1038/nrmicro3089

C. Archambaud, O. Sismeiro, J. Toedling, G. Soubigou, C. Bécavin et al., The Intestinal Microbiota Interferes with the microRNA Response upon Oral Listeria Infection, mBio, vol.4, issue.6, 2013.
DOI : 10.1128/mBio.00707-13

URL : https://hal.archives-ouvertes.fr/hal-01350904

C. Archambaud, M. Nahori, G. Soubigou, C. Bécavin, L. Laval et al., Impact of lactobacilli on orally acquired listeriosis, Proceedings of the National Academy of Sciences, vol.109, issue.41, pp.16684-16689, 2012.
DOI : 10.1073/pnas.1212809109

URL : https://hal.archives-ouvertes.fr/hal-01003361

L. E. Reddick and N. M. Alto, Bacteria Fighting Back: How Pathogens Target and Subvert the Host Innate Immune System, Molecular Cell, vol.54, issue.2, 2014.
DOI : 10.1016/j.molcel.2014.03.010

Y. Zhang, J. M. Noto, C. E. Hammond, J. L. Barth, W. S. Argraves et al., Helicobacter pylori-induced posttranscriptional regulation of H-K-ATPase ??-subunit gene expression by miRNA, AJP: Gastrointestinal and Liver Physiology, vol.306, issue.7, pp.606-613, 2014.
DOI : 10.1152/ajpgi.00333.2013

C. Maudet, M. Mano, U. Sunkavalli, M. Sharan, M. Giacca et al., Functional high-throughput screening identifies the miR-15 microRNA family as cellular restriction factors for Salmonella infection, Nature Communications, vol.26
DOI : 10.1038/ncomms5718

C. Belair, J. Baud, S. Chabas, C. M. Sharma, J. Vogel et al., Helicobacter pylori interferes with an embryonic stem cell micro RNA cluster to block cell cycle progression, Silence, vol.2, issue.1, pp.7-10, 2011.
DOI : 10.1038/ng1914

URL : https://hal.archives-ouvertes.fr/inserm-00639316

Y. Zhu, Q. Jiang, X. Lou, X. Ji, Z. Wen et al., MicroRNAs Up-Regulated by CagA of Helicobacter pylori Induce Intestinal Metaplasia of Gastric Epithelial Cells, PLoS ONE, vol.19, issue.4, 2012.
DOI : 10.1371/journal.pone.0035147.s008

N. Li, B. Tang, E. D. Zhu, B. S. Li, Y. Zhuang et al., -associated gastric cancer correlated with tumor progression by promoting cancer cell proliferation and targeting RECK, FEBS Letters, vol.12, issue.6, pp.722-728, 2012.
DOI : 10.1016/j.febslet.2012.01.025

URL : https://hal.archives-ouvertes.fr/tel-00807841

Z. Zhang, Z. Li, C. Gao, P. Chen, J. Chen et al., miR-21 plays a pivotal role in gastric cancer pathogenesis and progression, Laboratory Investigation, vol.1, issue.12, pp.1358-1366, 2008.
DOI : 10.1016/S0092-8674(01)00597-9

T. Zhang, J. Yu, Y. Zhang, L. Li, Y. Chen et al., Salmonella enterica Serovar Enteritidis Modulates Intestinal Epithelial miR-128 Levels to Decrease Macrophage Recruitment via Macrophage Colony-Stimulating Factor, Journal of Infectious Diseases, vol.209, issue.12, pp.2000-2011, 2014.
DOI : 10.1093/infdis/jiu006

B. Ni, M. V. Rajaram, W. P. Lafuse, M. B. Landes, and L. S. Schlesinger, Decreases Human Macrophage IFN-?? Responsiveness through miR-132 and miR-26a, The Journal of Immunology, vol.193, issue.9, pp.4537-4584, 2014.
DOI : 10.4049/jimmunol.1400124

M. V. Rajaram, B. Ni, J. D. Morris, M. N. Brooks, T. K. Carlson et al., Mycobacterium tuberculosis lipomannan blocks TNF biosynthesis by regulating macrophage MAPK-activated protein kinase 2 (MK2) and microRNA miR-125b, Proceedings of the National Academy of Sciences, vol.108, issue.42, pp.17408-17421, 2011.
DOI : 10.1073/pnas.1112660108

Y. Singh, V. Kaul, A. Mehra, S. Chatterjee, S. Tousif et al., Mycobacterium tuberculosis Controls MicroRNA-99b (miR-99b) Expression in Infected Murine Dendritic Cells to Modulate Host Immunity, Journal of Biological Chemistry, vol.288, issue.7, pp.5056-5061, 2013.
DOI : 10.1074/jbc.C112.439778

S. Yang, F. Li, S. Jia, K. Zhang, W. Jiang et al., Early Secreted Antigen ESAT-6 of <b><i>Mycobacterium Tuberculosis</i></b> Promotes Apoptosis of Macrophages via Targeting the MicroRNA155-SOCS1 Interaction, Cellular Physiology and Biochemistry, vol.35, issue.4, pp.1276-1288, 2015.
DOI : 10.1159/000373950

A. Grundhoff and C. S. Sullivan, Virus-encoded microRNAs, Virology, pp.325-343, 2011.

H. Liu, X. Wang, H. Wang, J. Wu, J. Ren et al., Escherichia coli noncoding RNAs can affect gene expression and physiology of Caenorhabditis elegans, Nature Communications, vol.161, issue.418
DOI : 10.1038/ncomms2071

A. Shmaryahu, M. Carrasco, and P. D. Valenzuela, Prediction of Bacterial microRNAs and possible targets in human cell transcriptome, Journal of Microbiology, vol.155, issue.6, pp.482-489, 2014.
DOI : 10.1007/s12275-014-3658-3

Y. Furuse, R. Finethy, H. A. Saka, A. M. Xet-mull, D. M. Sisk et al., Search for MicroRNAs Expressed by Intracellular Bacterial Pathogens in Infected Mammalian Cells, PLoS ONE, vol.150, issue.9, 2014.
DOI : 10.1371/journal.pone.0106434.s007

P. Lefevre, J. Witham, C. E. Lacroix, P. N. Cockerill, and C. Bonifer, The LPS-Induced Transcriptional Upregulation of the Chicken Lysozyme Locus Involves CTCF Eviction and Noncoding RNA Transcription, Molecular Cell, vol.32, issue.1, pp.32-129, 2008.
DOI : 10.1016/j.molcel.2008.07.023

N. E. Ilott, J. Heward, B. Roux, E. Tsitsiou, P. S. Fenwick et al., Corrigendum: Long non-coding RNAs and enhancer RNAs regulate the lipopolysaccharide-induced inflammatory response in human monocytes, Nature Communications, vol.6
DOI : 10.1038/ncomms7814

H. Cui, N. Xie, Z. Tan, S. Banerjee, V. J. Thannickal et al., The human long noncoding RNA lnc-IL7R regulates the inflammatory response, European Journal of Immunology, vol.22, issue.7, pp.2085-2095, 2014.
DOI : 10.1002/eji.201344126

J. A. Gomez, O. L. Wapinski, Y. W. Yang, S. Gopinath, D. M. Monack et al., The NeST Long ncRNA Controls Microbial Susceptibility and Epigenetic Activation of the Interferon-?? Locus, Cell, vol.152, issue.4, pp.152-743, 2013.
DOI : 10.1016/j.cell.2013.01.015

A. Mao, J. Shen, and Z. Zuo, Expression and regulation of long noncoding RNAs in TLR4 signaling in mouse macrophages, BMC Genomics, vol.16, issue.1, 2015.
DOI : 10.1093/nar/gks1193

A. J. Westermann, K. U. Förstner, F. Amman, L. Barquist, Y. Chao et al., Dual RNA-seq unveils noncoding RNA functions in host?pathogen interactions Functional Demarcation of Active and Silent Chromatin Domains in Human HOX Loci by Noncoding RNAs, Nature. Cell, pp.129-1311, 2007.

H. Wu, J. Liu, W. Li, G. Liu, and Z. Li, LncRNA-HOTAIR promotes TNF-?? production in cardiomyocytes of LPS-induced sepsis mice by activating NF-??B pathway, Biochemical and Biophysical Research Communications, vol.471, issue.1, pp.471-240, 2016.
DOI : 10.1016/j.bbrc.2016.01.117

M. Guttman, I. Amit, M. Garber, C. French, M. F. Lin et al., Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals, Nature, vol.322, issue.7235, pp.458-223, 2009.
DOI : 10.1038/nature07672

S. Carpenter, D. Aiello, M. K. Atianand, E. P. Ricci, P. Gandhi et al., A Long Noncoding RNA Mediates Both Activation and Repression of Immune Response Genes, Science, vol.341, issue.6147, pp.789-792, 2013.
DOI : 10.1126/science.1240925

N. A. Rapicavoli, K. Qu, J. Zhang, M. Mikhail, R. M. Laberge et al., A mammalian pseudogene lncRNA at the interface of inflammation and antiinflammatory therapeutics, pp.1-16, 2013.

K. Pawar, C. Hanisch, S. Eliseo, P. Vera, R. Einspanier et al., Down regulated lncRNA MEG3 eliminates mycobacteria in macrophages via autophagy, Scientific Reports, vol.34, issue.1, pp.1-13, 2016.
DOI : 10.1038/nmeth.2089

L. Salmena, L. Poliseno, Y. Tay, L. Kats, and P. P. Pandolfi, A ceRNA Hypothesis: The Rosetta Stone of a Hidden RNA Language?, Cell, vol.146, issue.3, 2011.
DOI : 10.1016/j.cell.2011.07.014

Z. Du, T. Sun, E. Hacisuleyman, T. Fei, X. Wang et al., Integrative analyses reveal a long noncoding RNA-mediated sponge regulatory network in prostate cancer, Nature Communications, vol.339
DOI : 10.1038/ncomms10982