M. Alhede, T. Bjarnsholt, M. Givskov, A. , and M. , Pseudomonas aeruginosa Biofilms, Adv. Appl. Microbiol, vol.86, pp.1-40, 2014.
DOI : 10.1016/B978-0-12-800262-9.00001-9

M. Alhede, T. Bjarnsholt, P. Ø. Jensen, R. K. Phipps, C. Moser et al., Pseudomonas aeruginosa recognizes and responds aggressively to the presence of polymorphonuclear leukocytes, Microbiology, vol.155, issue.11, pp.3500-3508, 2009.
DOI : 10.1099/mic.0.031443-0

N. M. Allewell, Introduction to Biofilms Thematic Minireview Series, Journal of Biological Chemistry, vol.291, issue.24, 2016.
DOI : 10.1074/jbc.R115.707257

URL : http://www.jbc.org/content/291/24/12527.full.pdf

M. E. Alvarez, J. I. Fuxman-bass, J. R. Geffner, P. X. Fernández-calotti, M. Costas et al., Neutrophil Signaling Pathways Activated by Bacterial DNA Stimulation, The Journal of Immunology, vol.177, issue.6, pp.4037-4046, 1950.
DOI : 10.4049/jimmunol.177.6.4037

URL : http://www.jimmunol.org/content/jimmunol/177/6/4037.full.pdf

R. Amino, S. Thiberge, S. Blazquez, P. Baldacci, O. Renaud et al., Imaging malaria sporozoites in the dermis of the mammalian host, Nature Protocols, vol.73, issue.7, pp.1705-1712120, 2007.
DOI : 10.1111/j.1365-2818.1989.tb00554.x

R. Amino, S. Thiberge, B. Martin, S. Celli, S. Shorte et al., Quantitative imaging of Plasmodium transmission from mosquito to mammal, Nature Medicine, vol.21, issue.2, pp.220-224, 1128.
DOI : 10.1038/ni1134

N. M. Bernthal, J. R. Pribaz, A. I. Stavrakis, F. Billi, J. S. Cho et al., Protective role of IL-1?? against post-arthroplasty Staphylococcus aureus infection, Journal of Orthopaedic Research, vol.434, issue.Suppl. 3, pp.1621-1626, 2011.
DOI : 10.1097/01.blo.0000155079.29604.d4

T. Bjarnsholt, P. Ø. Jensen, M. Burmølle, M. Hentzer, J. A. Haagensen et al., Pseudomonas aeruginosa tolerance to tobramycin, hydrogen peroxide and polymorphonuclear leukocytes is quorum-sensing dependent, Microbiology, vol.151, issue.2, pp.373-383, 2005.
DOI : 10.1099/mic.0.27463-0

URL : http://mic.microbiologyresearch.org/deliver/fulltext/micro/151/2/373.pdf?itemId=/content/journal/micro/10.1099/mic.0.27463-0&mimeType=pdf&isFastTrackArticle=

V. Brinkmann, U. Reichard, C. Goosmann, B. Fauler, Y. Uhlemann et al., Neutrophil Extracellular Traps Kill Bacteria, Science, vol.303, issue.5663, pp.1532-1535, 2004.
DOI : 10.1126/science.1092385

M. B. Carneiro, L. S. Hohman, J. G. Egen, and N. C. Peters, Use of two-photon microscopy to study Leishmania major infection of the skin, Methods, vol.127, pp.45-52, 2017.
DOI : 10.1016/j.ymeth.2017.04.012

L. D. Christensen, C. Moser, P. Ø. Jensen, T. B. Rasmussen, L. Christophersen et al., Impact of Pseudomonas aeruginosa quorum sensing on biofilm persistence in an in vivo intraperitoneal foreign-body infection model, Microbiology, vol.153, issue.7, pp.2312-2320006122, 2007.
DOI : 10.1099/mic.0.2007/006122-0

A. Corrado, P. Donato, S. Maccari, R. Cecchi, T. Spadafina et al., Staphylococcus aureus-dependent septic arthritis in murine knee joints: local immune response and beneficial effects of vaccination, Scientific Reports, vol.10, issue.1, 2016.
DOI : 10.1371/journal.pone.0123027

R. M. Corrigan, J. C. Abbott, H. Burhenne, V. Kaever, and A. Gründling, c-di-AMP Is a New Second Messenger in Staphylococcus aureus with a Role in Controlling Cell Size and Envelope Stress, PLoS Pathogens, vol.263, issue.9, 2011.
DOI : 10.1371/journal.ppat.1002217.s001

J. W. Costerton, P. S. Stewart, and E. P. Greenberg, Bacterial Biofilms: A Common Cause of Persistent Infections, Science, vol.284, issue.5418, 1999.
DOI : 10.1126/science.284.5418.1318

A. Dudeck, J. Dudeck, J. Scholten, A. Petzold, S. Surianarayanan et al., Mast Cells Are Key Promoters of Contact Allergy that Mediate the Adjuvant Effects of Haptens, Immunity, vol.34, issue.6, pp.973-984, 2011.
DOI : 10.1016/j.immuni.2011.03.028

N. Faust, F. Varas, L. M. Kelly, S. Heck, and T. Graf, Insertion of enhanced green fluorescent protein into the lysozyme gene creates mice with green fluorescent granulocytes and macrophages, Blood, vol.96, pp.719-726, 2000.

F. Bass, J. I. Gabelloni, M. L. Alvarez, M. E. Vermeulen, M. E. Russo et al., Characterization of bacterial DNA binding to human neutrophil surface, Laboratory Investigation, vol.58, issue.9, pp.926-93759, 2008.
DOI : 10.1128/JB.186.14.4449-4456.2004

T. Geiger, P. Francois, M. Liebeke, M. Fraunholz, C. Goerke et al., The Stringent Response of Staphylococcus aureus and Its Impact on Survival after Phagocytosis through the Induction of Intracellular PSMs Expression, PLoS Pathogens, vol.176, issue.11, 2012.
DOI : 10.1371/journal.ppat.1003016.s002

Y. Guo, R. I. Ramos, J. S. Cho, N. P. Donegan, A. L. Cheung et al., In Vivo Bioluminescence Imaging To Evaluate Systemic and Topical Antibiotics against Community-Acquired Methicillin-Resistant Staphylococcus aureus-Infected Skin Wounds in Mice, Antimicrobial Agents and Chemotherapy, vol.57, issue.2, pp.855-863, 1128.
DOI : 10.1128/AAC.01003-12

M. L. Hanke, A. Angle, and T. Kielian, MyD88-Dependent Signaling Influences Fibrosis and Alternative Macrophage Activation during Staphylococcus aureus Biofilm Infection, PLoS ONE, vol.7, issue.8, 2012.
DOI : 10.1371/journal.pone.0042476.g009

URL : https://doi.org/10.1371/journal.pone.0042476

M. L. Hanke, C. E. Heim, A. Angle, S. D. Sanderson, and T. Kielian, Targeting macrophage activation for the prevention and treatment of Staphylococcus aureus biofilm infections, J. Immunol, pp.2159-2168, 1950.

M. L. Hanke and T. Kielian, Deciphering mechanisms of staphylococcal biofilm evasion of host immunity, Frontiers in Cellular and Infection Microbiology, vol.2, 2012.
DOI : 10.3389/fcimb.2012.00062

F. Hanses, S. Park, J. Rich, L. , and J. C. , Reduced Neutrophil Apoptosis in Diabetic Mice during Staphylococcal Infection Leads to Prolonged Tnf?? Production and Reduced Neutrophil Clearance, PLoS ONE, vol.164, issue.8, 2011.
DOI : 10.1371/journal.pone.0023633.g008

C. E. Heim, D. Vidlak, T. D. Scherr, J. A. Kozel, M. Holzapfel et al., Myeloid-Derived Suppressor Cells Contribute to Staphylococcus aureus Orthopedic Biofilm Infection, The Journal of Immunology, vol.192, issue.8, pp.3778-3792, 1950.
DOI : 10.4049/jimmunol.1303408

URL : http://www.jimmunol.org/content/jimmunol/192/8/3778.full.pdf

R. Jain and W. Weninger, Shedding light on cutaneous innate immune responses: the intravital microscopy approach, Immunology and Cell Biology, vol.16, issue.4, 2013.
DOI : 10.1016/j.chembiol.2009.10.009

T. H. Jakobsen, M. Van-gennip, R. K. Phipps, M. S. Shanmugham, L. D. Christensen et al., ABSTRACT, Antimicrobial Agents and Chemotherapy, vol.56, issue.5, pp.2314-2325, 1128.
DOI : 10.1128/AAC.05919-11

C. V. Jakubzick, G. J. Randolph, and P. M. Henson, Monocyte differentiation and antigen-presenting functions, Nature Reviews Immunology, vol.595, issue.6, pp.349-362, 2017.
DOI : 10.1016/j.intimp.2012.11.016

P. Ø. Jensen, M. Givskov, T. Bjarnsholt, and C. Moser, The immune system vs. Pseudomonas aeruginosa biofilms, FEMS Immunol. Med. Microbiol, vol.59, 2010.

P. Ø. Jensen, C. Moser, O. Kobayashi, H. P. Hougen, A. Kharazmi et al., Faster activation of polymorphonuclear neutrophils in resistant mice during early innate response to Pseudomonas aeruginosa lung infection, Clinical and Experimental Immunology, vol.21, issue.3, 2004.
DOI : 10.1164/ajrccm.152.6.8520783

. Exp, S. W. Kashem, M. Haniffa, and D. H. Kaplan, Antigen-presenting cells in the skin, Annu. Rev. Immunol, vol.137, issue.35, pp.478-485, 2017.

J. S. Kavanaugh and A. R. Horswill, Impact of Environmental Cues on Staphylococcal Quorum Sensing and Biofilm Development, Journal of Biological Chemistry, vol.5, issue.24, pp.12556-12564, 2016.
DOI : 10.1038/nrg3182

D. Lebeaux, J. Ghigo, and C. Beloin, Biofilm-Related Infections: Bridging the Gap between Clinical Management and Fundamental Aspects of Recalcitrance toward Antibiotics, Microbiology and Molecular Biology Reviews, vol.78, issue.3, pp.510-543, 2014.
DOI : 10.1128/MMBR.00013-14

URL : https://hal.archives-ouvertes.fr/pasteur-01370744

J. L. Li, C. C. Goh, J. L. Keeble, J. S. Qin, B. Roediger et al., Intravital multiphoton imaging of immune responses in the mouse ear skin, Nature Protocols, vol.165, issue.2, pp.221-234, 2012.
DOI : 10.4049/jimmunol.165.6.3284

R. L. Lindquist, G. Shakhar, D. Dudziak, H. Wardemann, T. Eisenreich et al., Visualizing dendritic cell networks in vivo, Nature Immunology, vol.7, issue.12, pp.1243-1250, 2004.
DOI : 10.1214/aos/1176344552

C. Marquès, J. Tasse, A. Pracros, V. Collin, C. Franceschi et al., Effects of antibiotics on biofilm and unattached cells of a clinical Staphylococcus aureus isolate from bone and joint infection, Journal of Medical Microbiology, vol.64, issue.9, pp.1021-1026, 2015.
DOI : 10.1099/jmm.0.000125

D. E. Moormeier and K. W. Bayles, biofilm: a complex developmental organism, Molecular Microbiology, vol.212, issue.3, pp.365-376, 2017.
DOI : 10.1093/infdis/jiv319

URL : http://onlinelibrary.wiley.com/doi/10.1111/mmi.13634/pdf

C. Moser, H. T. Pedersen, C. J. Lerche, M. Kolpen, L. Line et al., Biofilms and host response - helpful or harmful, APMIS, vol.21, issue.Suppl 1, pp.320-338, 2017.
DOI : 10.1016/j.cmi.2014.10.024

URL : http://onlinelibrary.wiley.com/doi/10.1111/apm.12674/pdf

L. G. Ng, J. S. Qin, B. Roediger, Y. Wang, R. Jain et al., Visualizing the Neutrophil Response to Sterile Tissue Injury in Mouse Dermis Reveals a Three-Phase Cascade of Events, Journal of Investigative Dermatology, vol.131, issue.10, pp.2058-2068, 2011.
DOI : 10.1038/jid.2011.179

K. T. Nguyen, A. K. Seth, S. J. Hong, M. R. Geringer, P. Xie et al., Deficient cytokine expression and neutrophil oxidative burst contribute to impaired cutaneous wound healing in diabetic, biofilm-containing chronic wounds, Wound Repair and Regeneration, vol.73, issue.Suppl. 1, pp.833-841, 2013.
DOI : 10.1189/jlb.0802406

K. Nishitani, W. Sutipornpalangkul, K. L. De-mesy-bentley, J. J. Varrone, S. N. Bello-irizarry et al., osteomyelitis in mice to identify critical pathogen and host factors, Journal of Orthopaedic Research, vol.57, issue.9, pp.1311-1319, 2015.
DOI : 10.1128/AAC.01003-12

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4529770/pdf

J. A. Niska, J. A. Meganck, J. R. Pribaz, J. H. Shahbazian, E. Lim et al., Monitoring bacterial burden, inflammation and bone damage longitudinally using optical and µCT imaging in an orthopaedic implant infection in mice, PLOS ONE, vol.7, 2012.

M. Otto, Staphylococcal Biofilms, Curr. Top. Microbiol. Immunol, vol.322, pp.207-228, 2008.
DOI : 10.1007/978-3-540-75418-3_10

A. E. Paharik and A. R. Horswill, The Staphylococcal Biofilm: Adhesins, Regulation, and Host Response, Microbiol. Spectr, vol.4, 2016.
DOI : 10.1128/microbiolspec.VMBF-0022-2015

URL : http://europepmc.org/articles/pmc4887152?pdf=render

K. Papenfort and B. L. Bassler, Quorum sensing signal???response systems in Gram-negative bacteria, Nature Reviews Microbiology, vol.176, issue.9, pp.576-588, 2016.
DOI : 10.1371/journal.ppat.1000883

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5056591/pdf

S. S. Pedersen, A. Kharazmi, F. Espersen, and N. Høiby, Pseudomonas aeruginosa alginate in cystic fibrosis sputum and the inflammatory response, Infect. Immun, vol.58, pp.3363-3368, 1990.

S. Periasamy, H. Joo, A. C. Duong, T. L. Bach, V. Y. Tan et al., How Staphylococcus aureus biofilms develop their characteristic structure, Proceedings of the National Academy of Sciences, vol.202, issue.10, pp.1281-1286, 2012.
DOI : 10.1086/656915

URL : http://www.pnas.org/content/109/4/1281.full.pdf

A. Peschel, M. Otto, N. C. Peters, J. G. Egen, N. Secundino et al., Phenol-soluble modulins and staphylococcal infection, Nature Reviews Microbiology, vol.39, issue.10, pp.667-673, 1038.
DOI : 10.1042/BST0390807

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4780437/pdf

R. Prabhakara, J. M. Harro, J. G. Leid, A. D. Keegan, M. L. Prior et al., ABSTRACT, Infection and Immunity, vol.79, issue.12, pp.5010-5018, 1128.
DOI : 10.1128/IAI.05571-11

J. R. Pribaz, N. M. Bernthal, F. Billi, J. S. Cho, R. I. Ramos et al., Mouse model of chronic post-arthroplasty infection: Noninvasive in vivo bioluminescence imaging to monitor bacterial burden for long-term study, Journal of Orthopaedic Research, vol.30, issue.Suppl 3, 2012.
DOI : 10.1016/S8756-3282(01)00632-9

T. D. Scherr, M. L. Hanke, O. Huang, D. B. James, A. R. Horswill et al., Staphylococcus aureus biofilms induce macrophage dysfunction through leukocidin AB and Alpha-toxin, pp.1021-1501021, 2015.
DOI : 10.1128/mbio.01021-15

URL : http://mbio.asm.org/content/6/4/e01021-15.full.pdf

T. D. Scherr, C. E. Heim, J. M. Morrison, and T. Kielian, Hiding in Plain Sight: Interplay between Staphylococcal Biofilms and Host Immunity, Frontiers in Immunology, vol.5, 2014.
DOI : 10.3389/fimmu.2014.00037

URL : https://www.frontiersin.org/articles/10.3389/fimmu.2014.00037/pdf

P. R. Secor, L. A. Michaels, K. S. Smigiel, M. G. Rohani, L. K. Jennings et al., ABSTRACT, Infection and Immunity, vol.85, issue.1, pp.648-664, 1128.
DOI : 10.1128/IAI.00648-16

S. M. Shiels, K. M. Bedigrew, and J. C. Wenke, Development of a hematogenous implant-related infection in a rat model, BMC Musculoskeletal Disorders, vol.14, issue.6, pp.255-265, 2015.
DOI : 10.1302/2046-3758.36.2000293

G. Silva-santana, K. C. Lenzi-almeida, V. G. Lopes, and F. Aguiar-alves, Biofilm formation in catheter-related infections by Panton-Valentine leukocidin-producing Staphylococcus aureus, Int. Microbiol, vol.19, pp.199-207, 2016.

D. T. Snyder, J. F. Hedges, and M. A. Jutila, Getting ???Inside??? Type I IFNs: Type I IFNs in Intracellular Bacterial Infections, Journal of Immunology Research, vol.162, issue.4, p.9361802, 2017.
DOI : 10.1586/14787210.2013.840534

URL : http://doi.org/10.1155/2017/9361802

S. Sprangers, T. J. De-vries, and V. Everts, Monocyte Heterogeneity: Consequences for Monocyte-Derived Immune Cells, Journal of Immunology Research, vol.115, issue.3, 2016.
DOI : 10.1016/j.gene.2012.07.071

URL : http://doi.org/10.1155/2016/1475435

N. Sumaria, B. Roediger, L. G. Ng, J. Qin, R. Pinto et al., Cutaneous immunosurveillance by self-renewing dermal gammadelta T cells, 2011.
DOI : 10.1084/jem.20101824

URL : http://jem.rupress.org/content/jem/208/3/505.full.pdf

J. Tavares, P. Formaglio, S. Thiberge, E. Mordelet, N. Van-rooijen et al., Role of host cell traversal by the malaria sporozoite during liver infection, The Journal of Experimental Medicine, vol.923, issue.5, pp.905-915, 2013.
DOI : 10.1111/j.1462-5822.2004.00474.x

S. S. Tay, B. Roediger, P. L. Tong, S. Tikoo, and W. Weninger, The Skin-Resident Immune Network, Current Dermatology Reports, vol.5, issue.23363980, pp.13-22, 2014.
DOI : 10.1038/nri3365

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3931970/pdf

T. Teng, A. Ji, X. Ji, L. , and Y. , Neutrophils and Immunity: From Bactericidal Action to Being Conquered, Journal of Immunology Research, vol.4, issue.4, 2017.
DOI : 10.1074/jbc.m115.695866

URL : http://doi.org/10.1155/2017/9671604

L. R. Thurlow, M. L. Hanke, T. Fritz, A. Angle, A. Aldrich et al., Staphylococcus aureus Biofilms Prevent Macrophage Phagocytosis and Attenuate Inflammation In Vivo, The Journal of Immunology, vol.186, issue.11, pp.6585-6596, 1950.
DOI : 10.4049/jimmunol.1002794

URL : http://www.jimmunol.org/content/jimmunol/186/11/6585.full.pdf

P. L. Tong, B. Roediger, N. Kolesnikoff, M. Biro, S. S. Tay et al., The Skin Immune Atlas: Three-Dimensional Analysis of Cutaneous Leukocyte Subsets by Multiphoton Microscopy, Journal of Investigative Dermatology, vol.135, issue.1, pp.84-93, 2015.
DOI : 10.1038/jid.2014.289

A. Torre, M. Bacconi, C. Sammicheli, B. Galletti, D. Laera et al., Four-Component Staphylococcus aureus Vaccine 4C-Staph Enhances Fc?? Receptor Expression in Neutrophils and Monocytes and Mitigates S. aureus Infection in Neutropenic Mice, Infection and Immunity, vol.83, issue.8, pp.3157-3163, 1128.
DOI : 10.1128/IAI.00258-15

URL : http://iai.asm.org/content/83/8/3157.full.pdf

H. Trøstrup, K. Thomsen, L. J. Christophersen, H. P. Hougen, T. Bjarnsholt et al., biofilm aggravates skin inflammatory response in BALB/c mice in a novel chronic wound model, Wound Repair and Regeneration, vol.23, issue.2A, pp.292-299, 2013.
DOI : 10.1111/j.1365-2249.2004.02554.x

M. Valentini and A. Filloux, and Other Bacteria, Journal of Biological Chemistry, vol.6, issue.24, pp.12547-12555, 2016.
DOI : 10.1111/j.1462-2920.2011.02595.x

J. Valle, C. Solano, B. García, A. Toledo-arana, and I. Lasa, Biofilm switch and immune response determinants at early stages of infection, Trends in Microbiology, vol.21, issue.8, pp.364-371, 2013.
DOI : 10.1016/j.tim.2013.05.008

M. Van-gennip, L. D. Christensen, M. Alhede, K. Qvortrup, P. Ø. Jensen et al., ABSTRACT, Infection and Immunity, vol.80, issue.8, pp.2601-260706215, 2012.
DOI : 10.1128/IAI.06215-11

M. Van-gennip, C. Moser, L. D. Christensen, T. Bjarnsholt, H. Calum et al., Augmented effect of early antibiotic treatment in mice with experimental lung infections due to sequentially adapted mucoid strains of Pseudomonas aeruginosa, Journal of Antimicrobial Chemotherapy, vol.55, issue.5, pp.1241-1250, 1997.
DOI : 10.1093/jac/dki079

D. Vidlak and T. Kielian, Infectious Dose Dictates the Host Response during Staphylococcus aureus Orthopedic-Implant Biofilm Infection, Infection and Immunity, vol.84, issue.7, pp.1957-1965, 2016.
DOI : 10.1128/IAI.00117-16

URL : http://iai.asm.org/content/84/7/1957.full.pdf

C. Wagner, K. Kondella, T. Bernschneider, V. Heppert, A. Wentzensen et al., Post-Traumatic Osteomyelitis: Analysis of Inflammatory Cells Recruited into the Site of Infection, Shock, vol.20, issue.6, pp.503-510, 2003.
DOI : 10.1097/01.shk.0000093542.78705.e3

Y. Wang, L. I. Cheng, D. R. Helfer, A. G. Ashbaugh, R. J. Miller et al., biofilm infection reveals therapeutic targets, Proceedings of the National Academy of Sciences, vol.2, pp.5094-5102, 2017.
DOI : 10.1128/AAC.06291-11

C. Watters, K. Deleon, U. Trivedi, J. A. Griswold, M. Lyte et al., Pseudomonas aeruginosa biofilms perturb wound resolution and antibiotic tolerance in diabetic mice, Medical Microbiology and Immunology, vol.326, issue.1, pp.131-141, 2013.
DOI : 10.1124/jpet.108.138891

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3567255/pdf

C. Watters, J. A. Everett, C. Haley, A. Clinton, R. et al., ABSTRACT, Infection and Immunity, vol.82, issue.1, pp.92-10000651, 2014.
DOI : 10.1128/IAI.00651-13

C. Watters, D. Fleming, D. Bishop, R. , and K. P. , Host Responses to Biofilm, Prog. Mol. Biol. Transl. Sci, vol.142, 2016.
DOI : 10.1016/bs.pmbts.2016.05.007

J. M. Yarwood and P. M. Schlievert, Quorum sensing in Staphylococcus infections, Journal of Clinical Investigation, vol.112, issue.11, pp.1620-1625, 2003.
DOI : 10.1172/JCI200320442

URL : http://www.jci.org/articles/view/20442/files/pdf