E. Shebani, S. Shahana, C. Janson, and G. Roomans, Attachment of columnar airway epithelial cells in asthma, Tissue and Cell, vol.37, issue.2, pp.145-52, 2005.
DOI : 10.1016/j.tice.2004.12.002

E. Puchelle, Airway secretions: new concepts and functions, Eur Respir J, vol.5, pp.3-4, 1992.

H. Nadel, D. Stringer, H. Levison, J. Turner, and J. Sturgess, The immotile cilia syndrome: radiological manifestations., Radiology, vol.154, issue.3, pp.651-656, 1985.
DOI : 10.1148/radiology.154.3.3969467

S. Varsano, C. Basbaum, L. Forsberg, D. Borson, G. Caughey et al., Dog Tracheal Epithelial Cells in Culture Synthesize Sulfated Macromolecular Glycoconjugates and Release Them from the Cell Surface upon Exposure to Extracellular Proteinases, Experimental Lung Research, vol.127, issue.2, pp.157-84, 1987.
DOI : 10.1016/B978-0-12-356104-6.50011-8

D. Zahm and L. Heimer, Two transpallidal pathways originating in the rat nucleus accumbens, The Journal of Comparative Neurology, vol.255, issue.3, pp.437-483, 1990.
DOI : 10.1146/annurev.ne.09.030186.002041

J. Rutland, W. Griffin, and P. Cole, Human ciliary beat frequency in epithelium from intrathoracic and extrathoracic airways, Am Rev Respir Dis, vol.125, pp.100-105, 1982.

P. Verdugo, Goblet Cells Secretion and Mucogenesis, Annual Review of Physiology, vol.52, issue.1, pp.157-76, 1990.
DOI : 10.1146/annurev.ph.52.030190.001105

P. Jeffery, Morphologic features of airway surface epithelial cells and glands, Am Rev Respir Dis, vol.128, pp.14-20, 1983.

M. Goodman, D. Link, W. Brown, and P. Nakane, Ultrastructural evidence of transport of secretory IgA across bronchial epithelium, Am Rev Respir Dis, vol.123, pp.115-124, 1981.

L. Reid, PATHOLOGY OF CHRONIC BRONCHITIS, The Lancet, vol.263, issue.6806, pp.275-278
DOI : 10.1016/S0140-6736(54)91030-2

M. Ayers and P. Jeffery, Proliferation and differentiation in mammalian airway epithelium, Eur Respir J, vol.1, pp.58-80, 1988.

P. Jeffery and D. Li, Airway mucosa: secretory cells, mucus and mucin genes, European Respiratory Journal, vol.10, issue.7, pp.1655-62, 1997.
DOI : 10.1183/09031936.97.10071655

URL : http://erj.ersjournals.com/content/erj/10/7/1655.full.pdf

G. Donnelly, D. Haack, and C. Heird, Tracheal epithelium: cell kinetics and differentiation in normal rat tissue, Cell Proliferation, vol.3, issue.2, pp.119-149, 1982.
DOI : 10.1016/B978-1-4832-3151-8.50011-7

J. Boers, A. Ambergen, and F. Thunnissen, Number and Proliferation of Basal and Parabasal Cells in Normal Human Airway Epithelium, American Journal of Respiratory and Critical Care Medicine, vol.133, issue.6, pp.2000-2006, 1998.
DOI : 10.1136/jcp.43.3.184

M. Rose and J. Voynow, Respiratory Tract Mucin Genes and Mucin Glycoproteins in Health and Disease, Physiological Reviews, vol.86, issue.1
DOI : 10.1172/JCI118372

W. Finkbeiner, Physiology and pathology of tracheobronchial glands, Respiration Physiology, vol.118, issue.2-3, pp.77-83, 1999.
DOI : 10.1016/S0034-5687(99)00080-8

C. Franken, C. Meijer, and J. Dijkman, Tissue distribution of antileukoprotease and lysozyme in humans J Histochem Cytochem, pp.493-501, 1989.

E. Puchelle, J. Zahm, and N. Bonnet, Imaging of airway cells, European Respiratory Journal, vol.12, issue.1, pp.3-5, 1998.
DOI : 10.1183/09031936.98.12010003

C. Plopper, Comparative morphologic features of bronchiolar epithelial cells. The Clara cell, Am Rev Respir Dis, vol.128, pp.37-41, 1983.

J. Widdicombe and R. Pack, The Clara cell, Eur J Respir Dis, vol.63, pp.202-222, 1982.

A. Mukherjee, G. Kundu, and G. Mantile-selvaggi, Uteroglobin: a novel cytokine?, Cellular and Molecular Life Sciences CMLS, vol.55, issue.5, pp.771-87, 1999.
DOI : 10.1007/s000180050331

B. A. Domin, T. R. Devereux, and R. M. Philpot, The cytochrome P-450 monooxygenase system of rabbit lung enzyme components, activities, and induction in the nonciliated bronchialor epithelial (clara) cell, alveolar type 2 cell, and alveolar macrophage, Mol. Pharmacol, vol.30, pp.296-303, 1986.

M. Weichselbaum, M. Sparrow, E. Hamilton, P. Thompson, and D. Knight, A confocal microscopic study of solitary pulmonary neuroendocrine cells in human airway epithelium, Respiratory Research, vol.150, issue.1, p.115, 2005.
DOI : 10.1002/ar.a.10007

E. Cutz, W. Chan, and N. Track, Bombesin, calcitonin and leu-enkephalin immunoreactivity in endocrine cells of human lung, Experientia, vol.28, issue.7, pp.765-772, 1981.
DOI : 10.1177/28.8.6160182

S. Fuchs, A. Hollins, M. Laue, U. Schaefer, K. Roemer et al., Differentiation of human alveolar epithelial cells in primary culture: morphological characterization and synthesis of caveolin-1 and surfactant protein-C, Cell and Tissue Research, vol.311, issue.1, pp.31-45, 2003.
DOI : 10.1007/s00441-002-0653-5

S. Rooney, S. Young, and C. Mendelson, Molecular and cellular processing of lung surfactant., The FASEB Journal, vol.8, issue.12, pp.957-67, 1994.
DOI : 10.1096/fasebj.8.12.8088461

C. Franken, J. Kramps, C. Meyer, and J. Dijkman, Bull Localization of a low molecular weight protease inhibitor in the respiratory tract, Eur Physiopathol Respir, vol.16, pp.231-237, 1980.

L. Willems, J. Kramps, T. Stijnen, P. Sterk, J. Weening et al., Antileukoprotease-containing Bronchiolar Cells: Relationship with Morphologic Disease of Small Airways and Parenchyma, American Review of Respiratory Disease, vol.238, issue.5, pp.1244-50, 1989.
DOI : 10.1111/j.1365-2362.1980.tb00048.x

P. Hiemstra, R. Maassen, J. Stolk, R. Heinzel-wieland, G. Steffens et al., Antibacterial activity of antileukoprotease, Infect Immun, vol.64, pp.4520-4524, 1996.

J. Sallenave, Antimicrobial activity of antiproteinases, Biochemical Society Transactions, vol.30, issue.2, pp.111-116, 2002.
DOI : 10.1042/bst0300111

T. Mcneely, M. Dealy, D. Dripps, J. Orenstein, S. Eisenberg et al., Secretory leukocyte protease inhibitor: a human saliva protein exhibiting anti-human immunodeficiency virus 1 activity in vitro., Journal of Clinical Investigation, vol.96, issue.1, pp.456-64, 1995.
DOI : 10.1172/JCI118056

T. Abe, N. Kobayasi, and K. Yoshimura, Expression of the secretory leukoprotease inhibitor gene in epithelial cells., Journal of Clinical Investigation, vol.87, issue.6, pp.2207-151, 1991.
DOI : 10.1172/JCI115255

J. V. Fahey and C. R. Wira, Effect of Menstrual Status on Antibacterial Activity and Secretory Leukocyte Protease Inhibitor Production by Human Uterine Epithelial Cells in Culture, The Journal of Infectious Diseases, vol.185, issue.11, pp.1606-1619, 2002.
DOI : 10.1086/340512

T. B. Mcneely, M. Dealy, and D. J. Dripps, Secretory leukocyte protease inhibitor: a human saliva protein exhibiting anti-human immunodeficiency virus 1 activity in vitro., Journal of Clinical Investigation, vol.96, issue.1, pp.456-464, 1995.
DOI : 10.1172/JCI118056

G. S. Ashcroft, K. Lei, and W. Jin, Secretory leukocyte protease inhibitor mediates non-redundant functionsnecessary for normal wound healing, Nature Medicine, vol.161, issue.10, pp.1147-53, 2000.
DOI : 10.1007/BF00491759

P. Hiemstra, R. Maassen, J. Stolk, R. Heinzel-wieland, G. Steffens et al., Antibacterial activity of antileukoprotease, Infect Immun, vol.64, pp.4520-4524, 1996.

J. Ashitani, N. Matsumoto, and M. Nakazato, Elevated ?-defensin levels in plasma of patients with pulmonary sarcoidosis, Respirology, vol.305, issue.3, pp.339-384, 2007.
DOI : 10.1007/PL00000282

G. Diamond, M. Zasloff, H. Eck, M. Brasseur, W. Maloy et al., Tracheal antimicrobial peptide, a cysteine-rich peptide from mammalian tracheal mucosa: peptide isolation and cloning of a cDNA., Proceedings of the National Academy of Sciences, vol.88, issue.9, pp.3952-3958, 1991.
DOI : 10.1073/pnas.88.9.3952

G. Diamond, D. Laube, and M. Klein-patel, Mammalian ?-defensins in mucosal defences.. Mammalian host defence peptides, pp.111-138, 2004.

N. Borregaard, Neutrophils, from Marrow to Microbes, Immunity, vol.33, issue.5, pp.657-70
DOI : 10.1016/j.immuni.2010.11.011

B. Borregaard, N. Sørensen, O. Theilgaard-mönch, and K. , Neutrophil granules: a library of innate immunity proteins, Trends in Immunology, vol.28, issue.8, pp.340-345, 2007.
DOI : 10.1016/j.it.2007.06.002

T. Németh and A. Mócsai, The role of neutrophils in autoimmune diseases, Immunology Letters, vol.143, issue.1, pp.9-19, 2012.
DOI : 10.1016/j.imlet.2012.01.013

J. Riegger, J. Ware, A. Eckart, S. Haidari, M. Rudelius et al., Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo, pp.819-854, 2012.

B. Geering and H. Simon, Peculiarities of cell death mechanisms in neutrophils, Cell Death & Differentiation, vol.62, issue.9, pp.1457-69, 2011.
DOI : 10.1074/jbc.M604051200

V. Witko-sarsat, M. Pederzoli-ribeil, E. Hirsch, S. Sozzani, and M. Cassatella, Regulating neutrophil apoptosis: new players enter the game, Trends in Immunology, vol.32, issue.3, pp.117-141, 2011.
DOI : 10.1016/j.it.2011.01.001

F. Bazzoni, M. Cassatella, F. Rossi, M. Ceska, B. Dewald et al., Phagocytosing neutrophils produce and release high amounts of the neutrophil-activating peptide 1/interleukin 8, Journal of Experimental Medicine, vol.173, issue.3, pp.771-775, 1991.
DOI : 10.1084/jem.173.3.771

D. Bratton and P. Henson, Neutrophil clearance: when the party is over, clean-up begins, Trends in Immunology, vol.32, issue.8, pp.350-357, 2011.
DOI : 10.1016/j.it.2011.04.009

O. Soehnlein and L. Lindbom, Phagocyte partnership during the onset and resolution of inflammation, Nature Reviews Immunology, vol.8, issue.6, pp.427-466, 2010.
DOI : 10.4049/jimmunol.180.10.6868

G. Kaiko, J. Horvat, K. Beagley, and P. Hansbro, Immunological decision-making: how does the immune system decide to mount a helper T-cell response?, Immunology, vol.177, issue.3, pp.326-364, 2008.
DOI : 10.1074/jbc.M207577200

P. Kidd and . Th1, Th2 balance: the hypothesis, its limitations, and implications for health and disease, Altern Med Rev, vol.8, pp.223-269, 2003.

N. Fujiwara and K. Kobayashi, Macrophages in Inflammation, Current Drug Target -Inflammation & Allergy, vol.4, issue.3, pp.281-287, 2005.
DOI : 10.2174/1568010054022024

K. Nazimek and K. Bryniarski, The biological activity of macrophages in health and disease, Post??py Higieny i Medycyny Do??wiadczalnej, vol.66, pp.507-527, 2012.
DOI : 10.5604/17322693.1004080

P. Holt and P. Stumbles, Regulation of immunologic homeostasis in peripheral tissues by dendritic cells: The respiratory tract as a paradigm, Journal of Allergy and Clinical Immunology, vol.105, issue.3, pp.421-429, 2000.
DOI : 10.1067/mai.2000.105010

M. Moser and K. Murphy, Dendritic cell regulation of TH1-TH2 development, Nature Immunology, vol.9, issue.3, pp.199-205, 2000.
DOI : 10.1016/S1074-7613(00)80671-8

S. Currie, E. Roberts, S. Spaethe, N. Roehm, and R. Kramer, in MCII mast cells mediated by high-affinity Fc receptor for IgE, Biochemical Journal, vol.304, issue.3, pp.923-931, 1994.
DOI : 10.1042/bj3040923

C. Brightling, P. Bradding, F. Symon, S. Holgate, A. Wardlaw et al., Mast-Cell Infiltration of Airway Smooth Muscle in Asthma, New England Journal of Medicine, vol.346, issue.22, pp.1699-1705, 2002.
DOI : 10.1056/NEJMoa012705

I. Akers, M. Parsons, and M. Hill, Mast cell tryptase stimulates human lung fibroblast proliferation via protease-activated receptor-2, American Journal of Physiology-Lung Cellular and Molecular Physiology, vol.129, issue.1, pp.193-201, 2000.
DOI : 10.1073/pnas.95.12.6642

C. Williams and S. Galli, The diverse potential effector and immunoregulatory roles of mast cells in allergic disease, Journal of Allergy and Clinical Immunology, vol.105, issue.5, pp.847-859, 2000.
DOI : 10.1067/mai.2000.106485

J. Fahy, Reducing IgE levels as a strategy for the treatment of asthma, Clinical <html_ent glyph="@amp;" ascii="&"/> Experimental Allergy, vol.162, issue.2, pp.16-21, 2000.
DOI : 10.1164/ajrccm.158.6.9712073

H. Milgrom, R. Fick, . Jr, and J. Su, Treatment of Allergic Asthma with Monoclonal Anti-IgE Antibody, New England Journal of Medicine, vol.341, issue.26
DOI : 10.1056/NEJM199912233412603

D. Spriggs, S. Deutsch, and D. Kufe, Genomic structure, induction, and production of TNF-alpha, Immunol Ser, vol.56, pp.3-34, 1992.

N. Mukaida and K. Matsushima, Regulation of IL-8 production and the characteristics of the receptors for IL-8, Cytokines, vol.4, pp.41-53, 1992.

H. Wajant, K. Pfizenmaier, and P. Scheurich, Tumor necrosis factor signaling, Cell Death and Differentiation, vol.10, issue.1, pp.45-65, 2003.
DOI : 10.1038/sj.cdd.4401189

M. Krönke and T. Mak, Mice deficient for the 55 kd tumor necrosis factor receptor are resistant to endotoxic shock, yet succumb to L. monocytogenes infection, Cell, vol.73, pp.457-467, 1993.

F. Bazzoni and B. Beutler, The Tumor Necrosis Factor Ligand and Receptor Families, New England Journal of Medicine, vol.334, issue.26, pp.1717-1742, 1996.
DOI : 10.1056/NEJM199606273342607

K. Van-zee, T. Kohno, E. Fischer, C. Rock, L. Moldawer et al., Tumor necrosis factor soluble receptors circulate during experimental and clinical inflammation and can protect against excessive tumor necrosis factor alpha in vitro and in vivo., Proceedings of the National Academy of Sciences, vol.89, issue.11, pp.4845-4894, 1992.
DOI : 10.1073/pnas.89.11.4845

M. Shalaby, A. Sundan, H. Loetscher, M. Brockhaus, W. Lesslauer et al., Binding and regulation of cellular functions by monoclonal antibodies against human tumor necrosis factor receptors, Journal of Experimental Medicine, vol.172, issue.5
DOI : 10.1084/jem.172.5.1517

T. Nomura, Y. Abe, Y. Yoshioka, S. Nakagawa, S. Tsunoda et al., Creation of TNFR1-selective Antagonist and Its Therapeutic Effects, YAKUGAKU ZASSHI, vol.130, issue.1, pp.63-71, 2010.
DOI : 10.1248/yakushi.130.63

K. Bianchi and P. Meier, A Tangled Web of Ubiquitin Chains: Breaking News in TNF-R1 Signaling, Molecular Cell, vol.36, issue.5, pp.736-778, 2009.
DOI : 10.1016/j.molcel.2009.11.029

M. Takeuchi, M. Rothe, and D. Goeddel, Anatomy of TRAF2, Journal of Biological Chemistry, vol.148, issue.33, pp.19935-19977, 1996.
DOI : 10.1016/S0962-8924(00)89088-1

L. Tartaglia, R. Weber, I. Figari, C. Reynolds, M. Palladino et al., The two different receptors for tumor necrosis factor mediate distinct cellular responses., Proceedings of the National Academy of Sciences, vol.88, issue.20, pp.9292-96, 1991.
DOI : 10.1073/pnas.88.20.9292

R. Heller, K. Song, N. Fan, and D. Chang, The p70 tumor necrosis factor receptor mediates cytotoxicity, Cell, vol.70, issue.1, pp.47-56, 1992.
DOI : 10.1016/0092-8674(92)90532-H

T. Hirano, K. Yasukawa, H. Harada, T. Taga, Y. Watanabe et al., Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin, Nature, vol.16, issue.6092, pp.73-79, 1986.
DOI : 10.1016/B978-0-12-057550-3.50033-8

K. Yamasaki, T. Taga, Y. Hirata, H. Yawata, Y. Kawanishi et al., Cloning and expression of the human interleukin-6 (BSF-2/IFN beta 2) receptor, Science, vol.241, issue.4867, pp.825-833, 1988.
DOI : 10.1126/science.3136546

M. Romano, M. Sironi, C. Toniatti, N. Polentarutti, P. Fruscella et al., Role of IL-6 and Its Soluble Receptor in Induction of Chemokines and Leukocyte Recruitment, Immunity, vol.6, issue.3, pp.315-340, 1997.
DOI : 10.1016/S1074-7613(00)80334-9

K. Matsushima, K. Morishita, T. Yoshimura, S. Lavu, Y. Kobayashi et al., Purification and characterization of a novel monocyte chemotactic and activating factor produced by a human myelomonocytic cell line, Journal of Experimental Medicine, vol.169, issue.4, pp.1485-90, 1988.
DOI : 10.1084/jem.169.4.1485

N. Mukaida, A newly discovered chemotactic factor for neutrophil: interleukin 8, Rinsho Byori, vol.40, pp.371-380, 1992.

M. Aihara, D. Tsuchimoto, H. Takizawa, A. Azuma, H. Wakebe et al., Mechanisms involved in Helicobacter pyloriinduced interleukin-8 production by a gastric cancer cell line, MKN45. Infect Immun, vol.65, pp.3218-3242, 1997.

A. Samanta, J. Oppenheim, and K. Matsushima, Interleukin 8 (monocyte-derived neutrophil chemotactic factor) dynamically regulates its own receptor expression on human neutrophils, J Biol Chem, vol.265, pp.183-189, 1990.

T. Jinquan, B. Moller, M. Storgaard, N. Mukaida, J. Bonde et al., Chemotaxis and IL-8 receptor expression in B cells from normal and HIV-infected subjects, J Immunol, vol.158, pp.475-484, 1997.

K. Woolley, E. Adelroth, M. Woolley, R. Ellis, J. M. et al., Granulocyte-macrophage colony-stimulating factor, eosinophils and eosinophil cationic protein in subjects with and without mild, stable, atopic asthma, European Respiratory Journal, vol.7, issue.9, pp.1576-84, 1994.
DOI : 10.1183/09031936.94.07091576

T. L. Noah, A. M. Paradiso, M. C. Madden, K. P. Mckinnon, and R. B. Devlin, The Response of a Human Bronchial Epithelial Cell Line to Histamine: Intracellular Calcium Changes and Extracellular Release of Inflammatory Mediators, American Journal of Respiratory Cell and Molecular Biology, vol.1, issue.5, pp.484-492, 1991.
DOI : 10.1073/pnas.82.14.4633

Y. Li, C. L. Stechschulte, D. Dileepan, and K. , Histamine-Induced Production of Interleukin-6 and Interleukin-8 by Human Coronary Artery Endothelial Cells Is Enhanced by Endotoxin and Tumor Necrosis Factor-??, Microvascular Research, vol.61, issue.3, pp.253-62, 2001.
DOI : 10.1006/mvre.2001.2304

F. Kohda, T. Koga, H. Uchi, K. Urabe, and M. Furue, Histamine-induced IL-6 and IL-8 production are differentially modulated by IFN-?? and IL-4 in human keratinocytes, Journal of Dermatological Science, vol.28, issue.1, pp.34-41, 2002.
DOI : 10.1016/S0923-1811(01)00147-5

C. Oh and K. Nakano, Inhibition by glucocorticoids of mitogen dependent histamine biosynthesis caused by histidine decaroboxylase incultured mouse spleen cells and peritoneal adherent cells, Immunology, vol.65, pp.433-439, 1988.

Y. Kubo and K. Nakano, Regulation of histamine synthesis in mouse CD4+ and CD8+ T lymphocytes, Inflammation Research, vol.48, issue.3, pp.149-53, 1999.
DOI : 10.1007/s000110050438

Y. Kitamura, A. Miyoshi, Y. Murata, B. Kalubi, H. Fukui et al., Effect of glucocorticoid on upregulation of histamine H1 receptor mRNA in nasal mucosa of rats sensitized by exposure to toluene diisocyanate

K. Karlstedt, T. Sallmén, K. Eriksson, M. Lintunen, P. Couraud et al., Receptor mRNA Levels by Dexamethasone in Cerebral Endothelial Cells, Journal of Cerebral Blood Flow & Metabolism, vol.269, issue.3, pp.321-351, 1999.
DOI : 10.1111/j.1471-4159.1989.tb02509.x

Z. Qiu, M. A. Gijo´n, M. S. De-carvalho, D. M. Spencer, and C. Leslie, Role of calcium and phosphorylation of cytosolic phospholipase A2 In regulating arachidonic acid release in macrophages, J

E. Dennis, Diversity of group types, regulation, and function of phospholipase A2, J Biol Chem, vol.269, pp.13057-60, 1994.

P. Vadas, J. Browning, J. Edelson, and W. Pruzanski, Extracellular phospholipase A2 expression and inflammation : the relationship with associated disease states, J Lipid Mediat, vol.8, pp.1-30, 1993.

G. Lambeau, J. Barhanin, H. Schweitz, J. Qar, and M. Lazdunski, Identification and properties of very high affinity brain membranebinding sites for a neurotoxic phospholipase from the taipan venom, J Biol Chem, vol.264, pp.11503-11513, 1989.

G. Lambeau, A. Schmid-alliana, M. Lazdunski, and J. Barhanin, Identification and purification of a very high affinity binding protein for toxic phospholipases A2 in skeletal muscle, J. Biol Chem, vol.265, pp.9526-9558, 1990.

E. Valentin and G. Lambeau, Increasing molecular diversity of secreted phospholipases A2 and their receptors and binding proteins, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1488, issue.1-2, pp.59-70, 2000.
DOI : 10.1016/S1388-1981(00)00110-4

J. Balsinde, I. D. Bianco, E. J. Ackermann, K. Conde-frieboes, and E. Dennis, Inhibition of calcium-independent phospholipase A2 prevents arachidonic acid incorporation and phospholipid remodeling in P388D1 macrophages., Proceedings of the National Academy of Sciences, vol.92, issue.18, pp.8527-8558, 1995.
DOI : 10.1073/pnas.92.18.8527

A. K. Roshak, E. A. Capper, C. Stevenson, C. Eichman, and L. A. Marshall, Mediates Lymphocyte Proliferation, Journal of Biological Chemistry, vol.92, issue.46, pp.35692-35700, 2000.
DOI : 10.1074/jbc.270.25.14855

B. Falkenburger, J. Jensen, E. Dickson, B. Suh, and B. Hille, SYMPOSIUM REVIEW: Phosphoinositides: lipid regulators of membrane proteins, The Journal of Physiology, vol.37, issue.17, pp.3179-85, 2010.
DOI : 10.1016/S0896-6273(03)00125-9

F. Spadaro, C. Ramoni, D. Mezzanzanica, S. Miotti, P. Alberti et al., Phosphatidylcholine-Specific Phospholipase C Activation in Epithelial Ovarian Cancer Cells, Cancer Research, vol.68, issue.16, pp.6541-6550, 2008.
DOI : 10.1158/0008-5472.CAN-07-6763

L. Zhang, H. Li, H. Li, J. Zhao, L. Su et al., Lipopolysaccharide activated phosphatidylcholine-specific phospholipase C and induced IL-8 and MCP-1 production in vascular endothelial cells, Journal of Cellular Physiology, vol.30, issue.6, pp.1694-701, 2011.
DOI : 10.1161/ATVBAHA.109.195768

C. Ramoni, F. Spadaro, B. Barletta, M. Dupuis, and F. Podo, Phosphatidylcholine-specific phospholipase C in mitogen-stimulated fibroblasts, Experimental Cell Research, vol.299, issue.2, pp.370-82, 2004.
DOI : 10.1016/j.yexcr.2004.05.037

M. A. Rao, A. Hatcher, and . Gusain, Tricyclodecan-9-yl-xanthogenate (D609) mechanism of actions

Z. Kiss and M. Tomono, Compound D609 inhibits phorbol ester-stimulated phospholipase D activity and phospholipase C-mediated phosphatidylethanolamine hydrolysis, Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism, vol.1259, issue.1, pp.105-113, 1995.
DOI : 10.1016/0005-2760(95)00148-6

P. Kahle, E. Shooter, and R. Johnson, Phosphatidylcholine-specific phospholipases inhibitor D609 differentially affects MAP kinases and immediate-early genes in PC12 cells. Verity AN.Cell Signal, 1998.

M. Van-dijk, F. Muriana, J. De-widt, H. Hilkmann, and W. Van-blittezswijk, Involvement of Phosphatidylcholine-specific Phospholipase C in Platelet-derived Growth Factor-induced Activation of the Mitogen-activated Protein Kinase Pathway in Rat-1 Fibroblasts, Journal of Biological Chemistry, vol.265, issue.17, pp.11011-11017, 1997.
DOI : 10.1073/pnas.83.18.6785

C. Ramoni, F. Spadaro, M. Menegon, and F. Podo, Cellular Localization and Functional Role of Phosphatidylcholine-Specific Phospholipase C in NK Cells, The Journal of Immunology, vol.167, issue.5, pp.2642-50, 2001.
DOI : 10.4049/jimmunol.167.5.2642

D. J. Hanahan and I. L. Chaikoff, The phosphorus-containing lipides of the carrot, J. Biol. Chem, vol.168, pp.233-240, 1947.

D. J. Hanahan and I. Chaikoff, On the nature of the phosphorus-containing lipides of cabbage leaves and their relation to a phospholipide-splitting enzyme contained in these leaves, Biol. Chem, vol.172, pp.191-198, 1948.

C. Morgan, H. Sengelov, J. Whatmore, N. Borregaard, and C. , ADP-ribosylation-factor-regulated phospholipase D activity localizes to secretory vesicles and mobilizes to the plasma membrane following N-formylmethionyl-leucyl-phenylalanine stimulation of human neutrophils, Biochemical Journal, vol.325, issue.3, pp.581-586, 1997.
DOI : 10.1042/bj3250581

J. Balsinde, M. Balboa, P. Insel, and E. Dennis, Differential regulation of phospholipase D and phospholipase A2 by protein kinase C in P388D1 macrophages, Biochemical Journal, vol.321, issue.3, pp.805-814, 1997.
DOI : 10.1042/bj3210805

L. Simon, Role and regulation of cyclooxygenase-2 during inflammation, The American Journal of Medicine, vol.106, issue.5, pp.37-42, 1999.
DOI : 10.1016/S0002-9343(99)00115-1

A. W. Ford-hutchinson, M. A. Bray, M. V. Doig, M. E. Shipley, and M. J. Smith, Leukotriene B, a potent chemokinetic and aggregating substance released from polymorphonuclear leukocytes, Nature, vol.32, issue.5770, pp.264-269, 1980.
DOI : 10.1111/j.2042-7158.1980.tb12946.x

L. H. Faccioli, S. Nourshargh, R. Moqbel, F. M. Williams, R. Sehmi et al., The accumulation of 111In-eosinophils induced by inflammatory mediators, in vivo, Immunology, vol.73, pp.222-229, 1991.

K. Goodarzi, M. Goodarzi, A. M. Tager, A. D. Luster, and U. H. Von-andrian, Leukotriene B4 and BLT1 control cytotoxic effector T cell recruitment to inflamed tissues, Nature Immunology, vol.188, issue.10, pp.965-73, 2003.
DOI : 10.1084/jem.188.6.1063

P. Davis, M. Drumm, and M. Konstan, Cystic fibrosis., American Journal of Respiratory and Critical Care Medicine, vol.154, issue.5, pp.1229-12561, 1996.
DOI : 10.1164/ajrccm.154.5.8912731

J. F. Chmiel, M. Berger, and M. W. Konstan, The Role of Inflammation in the Pathophysiology of CF Lung Disease, Clinical Reviews in Allergy & Immunology, vol.23, issue.1, pp.5-27, 2002.
DOI : 10.1385/CRIAI:23:1:005

M. Griese, M. Kappler, and A. Gaggar, Inhibition of airway proteases in cystic fibrosis lung disease, European Respiratory Journal, vol.32, issue.3, pp.783-95, 2008.
DOI : 10.1183/09031936.00146807

M. W. Konstan and P. B. , Pharmacological approaches for the discovery and development of new anti-inflammatory agents for the treatment of cystic fibrosis, Advanced Drug Delivery Reviews, vol.54, issue.11, pp.1409-1432, 2002.
DOI : 10.1016/S0169-409X(02)00146-1

I. G. Doring, T. F. Parameswaran, and . Murphy, Differential adaptation of microbial pathogens to airways of patients with cystic fibrosis and chronic obstructive pulmonary disease, FEMS Microbiology Reviews, vol.64, issue.1, pp.124-146, 2011.
DOI : 10.1111/j.1469-0691.2008.02659.x

J. Jacquot, O. Tabary, and P. L. Rouzic, Airway epithelial cell inflammatory signalling in cystic fibrosis, The International Journal of Biochemistry & Cell Biology, vol.40, issue.9, pp.1703-1718, 2008.
DOI : 10.1016/j.biocel.2008.02.002

S. Brennan, Innate immune activation and cystic fibrosis, Paediatric Respiratory Reviews, vol.9, issue.4, pp.271-279, 2008.
DOI : 10.1016/j.prrv.2008.05.008

L. Imundo, J. Barasch, and A. Prince, Cystic fibrosis epithelial cells have a receptor for pathogenic bacteria on their apical surface., Proceedings of the National Academy of Sciences, vol.92, issue.7, pp.3019-3042, 1995.
DOI : 10.1073/pnas.92.7.3019

B. K. Rubin, Mucus structure and properties in cystic fibrosis, Paediatric Respiratory Reviews, vol.8, issue.1, pp.4-7, 2007.
DOI : 10.1016/j.prrv.2007.02.004

A. Moran, D. Hardin, and D. Rodman, Diagnosis, screening and management of cystic fibrosis related diabetes mellitus, Diabetes Research and Clinical Practice, vol.45, issue.1, pp.61-73, 1999.
DOI : 10.1016/S0168-8227(99)00058-3

F. Collins, Cystic fibrosis: molecular biology and therapeutic implications, Science, vol.256, issue.5058, pp.774-783, 1992.
DOI : 10.1126/science.1375392

S. Rowe, S. Miller, and E. Sorscher, Cystic Fibrosis, New England Journal of Medicine, vol.352, issue.19, pp.1992-2001, 2005.
DOI : 10.1056/NEJMra043184

J. Navarro and G. Bellon, La mucoviscidose ; de la théorie à la pratique. 2 em edition, 2001.

A. E. Klinger, M. J. Smith, and . Welsh, Expression of cystic fibrosis transmembrane conductance regulator corrects defective chloride channel regulation in cystic fibrosis airway epithelial cells, Nature, vol.347, pp.358-63, 1990.

M. P. Anderson, D. P. Rich, R. J. Gregory, A. Smith, and M. J. Welsh, Generation of cAMP-activated chloride currents by expression of CFTR, Science, vol.251, issue.4994, pp.679-82, 1991.
DOI : 10.1126/science.1704151

. Welsh and . Science, Demonstration that CFTR is a chloride channel by alteration of its anion selectiVity, pp.202-204, 1991.

C. E. Bear, C. Li, N. Kartner, R. J. Bridges, T. J. Jensen et al., Purification and functional reconstitution of the cystic fibrosis transmembrane conductance regulator (CFTR), Cell, vol.68, issue.4, pp.809-827
DOI : 10.1016/0092-8674(92)90155-6

L. Tsui, Mutations and sequence variations detected in the cystic fibrosis transmembrane conductance regulator (CFTR) gene: A report from the cystic fibrosis genetic analysis consortium, Human Mutation, vol.49, issue.3
DOI : 10.1007/978-1-4684-5934-0_48

S. H. Cheng, D. P. Rich, J. Marshall, R. J. Gregory, M. J. Welsh et al., Phosphorylation of the R domain by cAMP-dependent protein kinase regulates the CFTR chloride channel, Cell, vol.66, issue.5, pp.1027-1063, 1991.
DOI : 10.1016/0092-8674(91)90446-6

F. Seibert, T. Loo, D. Clarke, and J. Riordan, Cystic fibrosis: channel, catalytic, and folding properties of the CFTR protein, Journal of Bioenergetics and Biomembranes, vol.29, issue.5, pp.429-471, 1997.
DOI : 10.1023/A:1022478822214

D. Sheppard and M. Welsh, Structure and Function of the CFTR Chloride Channel, Physiological Reviews, vol.14, issue.1, pp.23-45, 1999.
DOI : 10.1126/science.2122520

M. Stutts, C. Canessa, J. Olsen, M. Hamrick, J. Cohn et al., CFTR as a cAMP-dependent regulator of sodium channels, Science, vol.261, issue.2, pp.847-50, 1995.
DOI : 10.1089/hum.1992.3.3-253

R. Boucher, Cystic fibrosis: a disease of vulnerability to airway surface dehydration, Trends in Molecular Medicine, vol.13, issue.6, pp.231-271, 2007.
DOI : 10.1016/j.molmed.2007.05.001

Z. Zhou, J. Duerr, B. Johannesson, S. Schubert, D. Treis et al., The ENaC-overexpressing mouse as a model of cystic fibrosis lung disease, Journal of Cystic Fibrosis, vol.10, pp.172-82, 2011.
DOI : 10.1016/S1569-1993(11)60021-0

D. Nelson, CFTR regulates phagosome acidification in macrophages and alters bactericidal activity, Nat Cell Biol, vol.8, pp.933-977, 2006.

S. Gabriel, L. Clarke, R. Boucher, and M. Stutts, CFTR and outward rectifying chloride channels are distinct proteins with a regulatory relationship, Nature, vol.363, issue.6426, pp.263-271, 1993.
DOI : 10.1038/363263a0

E. Schwiebert, D. Benos, M. Egan, M. Stutts, and W. Guggino, CFTR Is a Conductance Regulator as well as a Chloride Channel, Physiological Reviews, vol.266, issue.1, pp.145-66, 1999.
DOI : 10.1152/ajpcell.1994.266.3.C809

E. Schwiebert, D. Gruenert, W. Guggino, and B. Stanton, G protein G alpha i-2 inhibits outwardly rectifying chloride channels in human airway epithelial cells, American Journal of Physiology-Cell Physiology, vol.265, issue.2, pp.451-457, 1995.
DOI : 10.1073/pnas.88.12.5277

G. Loussouarn, S. Demolombe, R. Mohammad-panah, D. Escande, and I. Baró, Expression of CFTR controls cAMP-dependent activation of epithelial K+ currents, American Journal of Physiology-Cell Physiology, vol.142, issue.5, pp.1565-73, 1996.
DOI : 10.1093/hmg/2.8.1253

P. Linsdell and J. Hanrahan, Glutathione permeability of CFTR, American Journal of Physiology-Cell Physiology, vol.4, issue.40, pp.323-329, 1998.
DOI : 10.1085/jgp.61.6.687

M. Hodson, A. Penketh, and J. Batten, AEROSOL CARBENICILLIN AND GENTAMICIN TREATMENT OF PSEUDOMONAS AERUGINOSA INFECTION IN PATIENTS WITH CYSTIC FIBROSIS, The Lancet, vol.318, issue.8256, pp.1137-1146, 1981.
DOI : 10.1016/S0140-6736(81)90588-2

F. Bressolle, J. De-la-coussaye, and R. Ayoub, Endotracheal and aerosol administrations of ceftazidime in patients with nosocomial pneumonia: pharmacokinetics and absolute bioavailability., Antimicrobial Agents and Chemotherapy, vol.36, issue.7, 1992.
DOI : 10.1128/AAC.36.7.1404

G. Nolan, P. Moivor, and H. Levison, Antibiotic prophylaxis in cystic fibrosis: Inhaled cephaloridine as an adjunct to oral cloxacillin, The Journal of Pediatrics, vol.101, issue.4, pp.626-656, 1982.
DOI : 10.1016/S0022-3476(82)80726-9

J. Schentag, Clinical Significance of Antibiotic Tissue Penetration, Clinical Pharmacokinetics, vol.16, issue.Supplement 1, pp.25-31, 1989.
DOI : 10.2165/00003088-198900161-00005

G. Aubert-tulkens, F. Van-hoof, and P. Tulkens, Gentamicin-induced lysosomal phospholipidosis in cultured rat fibroblasts, Lab Invest, vol.40, pp.481-93, 1979.

J. Nukiwa, Clarithromycin suppresses lipopolysaccharide-induced interleukin-8 production by human monocytes through AP-1 and NF-?B transcription factors, Antimicrob. Chemother, vol.49, pp.745-755, 2002.

J. Tamaoki, J. Nakata, E. Tagaya, and K. Konno, Effects of roxithromycin and erythromycin on interleukin 8-induced neutrophil recruitment and goblet cell secretion in guinea pig tracheas

J. Costerton, Cystic fibrosis pathogenesis and the role of biofilms in persistent infection, Trends in Microbiology, vol.9, issue.2, pp.50-52, 2001.
DOI : 10.1016/S0966-842X(00)01918-1

R. L. Henry, C. M. Mellis, and L. Petrovic, MucoidPseudomonas aeruginosa is a marker of poor survival in cystic fibrosis, Pediatric Pulmonology, vol.96, issue.3, pp.158-161, 1992.
DOI : 10.1001/archpedi.1958.02060060008002

H. Curschmann, Ueber Bronchiolitis exsudative und ihr Verhaltniss zum Asthma nervosum, Dtsch Arch Klin Med, vol.32, pp.1-34, 1882.

W. Busse, R. Lemanske, and . Asthma, Asthma, New England Journal of Medicine, vol.344, issue.5, pp.350-62, 2001.
DOI : 10.1056/NEJM200102013440507

URL : https://hal.archives-ouvertes.fr/inserm-00796149

K. Woolley, P. Gibson, K. Carty, A. Wilson, S. Twaddell et al., Eosinophil apoptosis and the resolution of airway inflammation in asthma., American Journal of Respiratory and Critical Care Medicine, vol.154, issue.1, pp.237-280, 1996.
DOI : 10.1164/ajrccm.154.1.8680686

D. Raeburn and S. Webber, Proinflammatory potential of the airway epithelium in bronchial asthma, European Respiratory Journal, vol.7, issue.12, pp.2226-2259, 1994.
DOI : 10.1183/09031936.94.07122226

M. Versnel, Modulation of glucocorticoid receptor expression in human bronchial epithelial cell lines by IL- 1 beta, TNF-a and LPS, Eur Respir J, vol.9, pp.2036-2079, 1996.

T. Levan, F. Behr, K. Adkins, R. Miesfeld, and J. Bloom, Glucocorticoid receptor signaling in a bronchial epithelial cell line, American Journal of Physiology-Lung Cellular and Molecular Physiology, vol.272, issue.5, pp.838-843, 1997.
DOI : 10.1152/ajplung.1997.272.5.L838

T. Levan, E. Babin, H. Yamamura, and J. Bloom, Pharmacological characterization of glucocorticoid receptors in primary human bronchial epithelial cells, Biochemical Pharmacology, vol.57, issue.9, pp.1003-1012, 1999.
DOI : 10.1016/S0006-2952(99)00008-8

I. Adcock, T. Gilbey, C. Gelder, K. Chung, and P. Barnes, Glucocorticoid receptor localization in normal and asthmatic lung., American Journal of Respiratory and Critical Care Medicine, vol.154, issue.3, pp.771-82, 1996.
DOI : 10.1164/ajrccm.154.3.8810618

L. Pujols, J. Mullol, J. Roca-ferrer, A. Torrego, A. Xaubet et al., Expression of glucocorticoid receptor ??- and ??-isoforms in human cells and tissues, American Journal of Physiology-Cell Physiology, vol.266, issue.4, pp.1324-1355, 2002.
DOI : 10.1111/j.1365-2222.1994.tb00240.x

R. H. Oakley, M. Sar, and J. A. Cidlowski, The Human Glucocorticoid Receptor b

. Isoform, Expression, biochemical properties, and putative function. The journal of biological, chemistry, vol.16, pp.9550-9559, 1996.

M. De-castro, S. Elliot, T. Kino, C. Bamberger, M. Karl et al., The Non-Ligand Binding f3-Isoform of the Human Glucocorticoid Receptor (hGRf): Tissue Levels, Mechanism of Action, and Potential Physiologic Role, Mol Med, vol.2, pp.597-607, 1996.

R. Oakley, C. Jewell, M. Yudt, and D. Bofetiado, The Dominant Negative Activity of the Human Glucocorticoid Receptor ?? Isoform, Journal of Biological Chemistry, vol.2, issue.39, pp.27857-66, 1999.
DOI : 10.1210/mend-5-11-1707

C. Stellato, Post-transcriptional and Nongenomic Effects of Glucocorticoids, Proceedings of the American Thoracic Society, vol.1, issue.3, pp.255-63, 2004.
DOI : 10.1513/pats.200402-015MS

L. Bladh, J. Liden, A. Pazirandeh, I. Rafter, K. Dahlman-wright et al., Identification of Target Genes Involved in the Antiproliferative Effect of Glucocorticoids Reveals a Role for Nuclear Factor-??B Repression, Molecular Endocrinology, vol.19, issue.3, pp.632-675, 2005.
DOI : 10.1210/me.2004-0294

J. Brewer, B. Khor, S. Vogt, L. Muglia, H. Fujiwara et al., T-cell glucocorticoid receptor is required to suppress COX-2-mediated lethal immune activation, Nature Medicine, vol.21, issue.10, pp.1318-1340, 2003.
DOI : 10.1002/eji.1830211009

P. Barnes, Anti-inflammatory Actions of Glucocorticoids: Molecular Mechanisms, Clinical Science, vol.94, issue.6, pp.557-72, 1998.
DOI : 10.1042/cs0940557

P. Barnes, Molecular mechanisms of corticosteroids in all_ergic diseases., Allergy, vol.166, issue.10, pp.928-964, 2001.
DOI : 10.4049/jimmunol.166.3.1975

L. Schwiebert, C. Stellato, and R. Schleimer, The Epithelium as a Target of Glucocorticoid Action in the Treatment of Asthma, American Journal of Respiratory and Critical Care Medicine, vol.150, issue.2_pt_2, pp.16-25, 1996.
DOI : 10.1172/JCI112937

C. Hubeau, L. Naour, R. Abély, M. Hinnrasky, J. Guenounou et al., Dysregulation of IL-2 and IL-8 production in circulating T lymphocytes from young cystic fibrosis patients, Clinical and Experimental Immunology, vol.152, issue.3, pp.528-562, 2004.
DOI : 10.1128/IAI.68.4.2142-2147.2000

URL : https://hal.archives-ouvertes.fr/inserm-00145671

R. Crystal, Modulation of airway inflammation in cystic fibrosis In vivo suppression of interleukin-8 levels on the respiratory epithelial surface by aerosolization of recombinant secretory leukoprotease inhibitor, J Clin Invest, vol.90, pp.1296-301, 1992.

T. Machen, Innate immune response in CF airway epithelia: hyperinflammatory?, American Journal of Physiology-Cell Physiology, vol.291, issue.2, pp.218-248, 2006.
DOI : 10.1128/IAI.73.11.7151-7160.2005

J. Richman-eisenstat, P. Jorens, C. Hébert, I. Ueki, and J. Nadel, Interleukin-8: an important chemoattractant in sputum of patients with chronic inflammatory airway diseases, American Journal of Physiology-Lung Cellular and Molecular Physiology, vol.264, issue.4, pp.413-421, 1993.
DOI : 10.1152/ajplung.1993.264.4.L413

C. Francoeur and M. Denis, Nitric oxide and interleukin-8 as inflammatory components of cystic fibrosis, Inflammation, vol.107, issue.8457, pp.587-98, 1995.
DOI : 10.1007/BF01539138

T. Noah, H. Black, P. Cheng, R. Wood, and M. Leigh, Nasal and Bronchoalveolar Lavage Fluid Cytokines in Early Cystic Fibrosis, Journal of Infectious Diseases, vol.175, issue.3, pp.638-685, 1997.
DOI : 10.1093/infdis/175.3.638

R. Tirouvanziam, S. De-bentzmann, C. Hubeau, J. Hinnrasky, J. Jacquot et al., Inflammation and Infection in Naive Human Cystic Fibrosis Airway Grafts, American Journal of Respiratory Cell and Molecular Biology, vol.156, issue.2, pp.121-128, 2000.
DOI : 10.1164/ajrccm.152.3.7663805

N. Reiniger, J. Ichikawa, and G. Pier, Influence of Cystic Fibrosis Transmembrane Conductance Regulator on Gene Expression in Response to Pseudomonas aeruginosa Infection of Human Bronchial Epithelial Cells, Infection and Immunity, vol.73, issue.10
DOI : 10.1128/IAI.73.10.6822-6830.2005

L. Wiszniewski, L. Jornot, T. Dudez, A. Pagano, T. Rochat et al., Long-Term Cultures of Polarized Airway Epithelial Cells from Patients with Cystic Fibrosis, American Journal of Respiratory Cell and Molecular Biology, vol.34, issue.1, pp.39-48, 2006.
DOI : 10.1128/IAI.72.7.4188-4199.2004

P. Scheid, L. Kempster, U. Griesenbach, J. Davies, A. Dewar et al., Inflammation in cystic fibrosis airways: relationship to increased bacterial adherence, European Respiratory Journal, vol.17, issue.1, pp.27-35, 2001.
DOI : 10.1183/09031936.01.17100270

A. Massengale, F. Quinn, . Jr, J. Yankaskas, D. Weissman et al., Reduced Interleukin-8 Production by Cystic Fibrosis Airway Epithelial Cells, American Journal of Respiratory Cell and Molecular Biology, vol.267, issue.5, pp.1073-80, 1999.
DOI : 10.1126/science.2472008

Y. Fong, K. Tracey, L. Moldawer, D. Hesse, K. Manogue et al., Antibodies to cachectin/tumor necrosis factor reduce interleukin 1 beta and interleukin 6

M. Konstan, R. Walenga, K. Hilliard, and J. Hilliard, Markedly Elevated in the Epithelial Lining Fluid of Patients with Cystic Fibrosis, American Review of Respiratory Disease, vol.256, issue.11, pp.896-901, 1993.
DOI : 10.1172/JCI111570

B. Rigas, J. Korenberg, W. Merrill, and L. Levine, Prostaglandins E2 and E2 alpha are elevated in saliva of cystic fibrosis patients, Am J Gastroenterol, vol.84, pp.1408-1420, 1989.

J. Chen, X. Jiang, H. Chen, J. Guo, L. Tsang et al., positive feedback loop in inflammation, Journal of Cellular Physiology, vol.11, issue.6, pp.2759-66, 2012.
DOI : 10.1186/1465-9921-11-49

J. Carlstedt-duke, M. Brönnegård, and B. Strandvik, Pathological regulation of arachidonic acid release in cystic fibrosis: the putative basic defect., Proceedings of the National Academy of Sciences, vol.83, issue.23, pp.9202-9208, 1986.
DOI : 10.1073/pnas.83.23.9202

S. Freedman, M. Katz, E. Parker, M. Laposata, M. Urman et al., A membrane lipid imbalance plays a role in the phenotypic expression of cystic fibrosis in cftr-/- mice, Proceedings of the National Academy of Sciences, vol.26, issue.9, pp.13995-4000, 1999.
DOI : 10.1159/000177530

J. Carlstedt-duke, M. Brönnegård, and B. Strandvik, Pathological regulation of arachidonic acid release in cystic fibrosis: the putative basic defect., Proceedings of the National Academy of Sciences, vol.83, issue.23, pp.9202-9208, 1986.
DOI : 10.1073/pnas.83.23.9202

F. Alonso, P. M. Henson, L. , and C. , A cytosolic phospholipase in human neutrophils that hydrolyzes arachidonoyl-containing phosphatidylcholine, Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism, vol.878, issue.2, pp.273-80, 1986.
DOI : 10.1016/0005-2760(86)90156-6

D. Riendeau, J. Guay, P. Weech, F. Laliberté, J. Yergey et al., Arachidonyl trifluoromethyl ketone, a potent inhibitor of 85-kDa phospholipase A2, blocks production of arachidonate and 12 hydroxyeicosa-tetraenoic acid by calcium ionophore-challenged platelets, J Biol Chem, vol.269, pp.15619-15643, 1994.

L. Miele, E. Cordella-miele, M. Xing, R. Frizzell, and A. Mukherjee, Cystic fibrosis gene mutation (deltaF508) is associated with an intrinsic abnormality in Ca2+-induced arachidonic acid release by epithelial cells, DNA Cell Biol, vol.16, pp.749-59, 1997.

W. Kammouni, B. Moreau, F. Becq, A. Saleh, A. Pavirani et al., A Cystic Fibrosis Tracheal Gland Cell Line, CF-KM4, American Journal of Respiratory Cell and Molecular Biology, vol.15, issue.4, pp.684-691, 1999.
DOI : 10.1006/bbrc.1997.7720

J. Liao, K. L. Kirk, E. J. Sorscher, J. Hanrahan, and J. P. Clancy, A macromolecular complex of ?2 adrenergic receptor, CFTR, and ezrin/radixin/moesin-binding phosphoprotein 50 is regulated by PKA

B. Pamela, P. Davis, J. Shelhamer, and M. Kaliner, Abnormal adrenergic and cholinergic sensitivity in cystic fibrosis, N Eng J Med, vol.302, pp.1453-1459, 1980.

S. Pippig, S. Andexinger, K. Daniel, M. Puzicha, M. Caron et al., Overexpression of beta-arrestin and beta-adrenergic receptor kinase augment desensitization of beta 2-adrenergic receptors

M. Schlador and N. Nathanson, Synergistic Regulation of m2 Muscarinic Acetylcholine Receptor Desensitization and Sequestration by G Protein-coupled Receptor Kinase-2 and ??-Arrestin-1, Journal of Biological Chemistry, vol.12, issue.30, pp.18882-90, 1997.
DOI : 10.1016/0306-4522(92)90253-X

D. Diviani, A. Lattion, N. Larbi, P. Kunapuli, A. Pronin et al., Effect of different G protein-coupled receptor kinases on phosphorylation and desensitization of the alpha1B-adrenergic receptor, J Biol Chem, vol.271, pp.5049-58, 1996.

K. Haga, K. Kameyama, T. Haga, U. Kikkawa, K. Shiozaki et al., Phosphorylation of Human m1 Muscarinic Acetylcholine Receptors by G Protein-coupled Receptor Kinase 2 and Protein Kinase C, Journal of Biological Chemistry, vol.271, issue.5, pp.2776-82, 1996.
DOI : 10.1074/jbc.271.5.2776

M. Lohse, J. Benovic, J. Codina, M. Caron, and R. Lefkowitz, beta-Arrestin: a protein that regulates beta-adrenergic receptor function, Science, vol.248, issue.4962, pp.1547-50, 1990.
DOI : 10.1126/science.2163110

S. Medjane, B. Raymond, Y. Wu, and L. Touqui, expression by pulmonary epithelial cells, American Journal of Physiology-Lung Cellular and Molecular Physiology, vol.289, issue.5, 2005.
DOI : 10.1165/ajrcmb/4.4.313

Z. Kiss and M. Tomono, Compound D609 inhibits phorbol ester-stimulated phospholipase D activity and phospholipase C-mediated phosphatidylethanolamine hydrolysis, Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism, vol.1259, issue.1, pp.105-113, 1995.
DOI : 10.1016/0005-2760(95)00148-6

A. Agro, C. Langdon, F. Smith, and C. Richards, Prostaglandin E2 enhances interleukin 8 (IL-8) and IL-6 but inhibits GMCSF production by IL-1 stimulated human synovial fibroblasts in vitro, J Rheumatol, vol.23, pp.862-870, 1996.

U. Berger and M. Hediger, Distribution of peptide transporter PEPT2 mRNA in the rat nervous system, Anatomy and Embryology, vol.199, issue.5
DOI : 10.1007/s004290050242

D. Groneberg, F. Doring, S. Theis, M. Nickolaus, A. Fischer et al., Peptide transport in the mammary gland: expression and distribution of PEPT2 mRNA and protein, American Journal of Physiology-Endocrinology and Metabolism, vol.277, issue.5, pp.1172-1181, 2002.
DOI : 10.1111/j.1476-5381.1995.tb15958.x

H. Lu and C. Klaassen, Tissue distribution and thyroid hormone regulation of Pept1 and Pept2 mRNA in rodents, Peptides, vol.27, issue.4, pp.850-857, 2006.
DOI : 10.1016/j.peptides.2005.08.012

T. Zhu, X. Chen, A. Steel, M. Hediger, and D. Smith, Differential recognition of ACE inhibitors in Xenopus laevis oocytes expressing rat PEPT1 and PEPT2, Pharmaceutical Research, vol.17, issue.5, pp.526-528, 2000.
DOI : 10.1023/A:1007556630189

C. Yang, A. Dantzig, and C. Pidgeon, Intestinal peptide transport systems and oral drug availability, Pharmaceutical Research, vol.16, issue.9, pp.1331-1374, 1999.
DOI : 10.1023/A:1018982505021

D. Groneberg, A. Fischer, K. Chung, and H. Daniel, Molecular Mechanisms of Pulmonary Peptidomimetic Drug and Peptide Transport, American Journal of Respiratory Cell and Molecular Biology, vol.30, issue.3, pp.251-260, 2004.
DOI : 10.1124/jpet.102.040295

M. Brandsch, I. Knütter, and E. Bosse-doenecke, Pharmaceutical and pharmacological importance of peptide transporters, Journal of Pharmacy and Pharmacology, vol.12, issue.5, pp.543-85, 2008.
DOI : 10.1023/A:1016254514167

H. Daniel and G. Kottra, The proton oligopeptide cotransporter family SLC15 in physiology and pharmacology, Pflugers Arch, vol.447, pp.610-618, 2004.

F. Döring, D. Dorn, U. Bachfischer, S. Amasheh, M. Herget et al., Functional analysis of a chimeric mammalian peptide transporter derived from the intestinal and renal isoforms., The Journal of Physiology, vol.497, issue.3, pp.773-782, 1996.
DOI : 10.1113/jphysiol.1996.sp021808

Y. Fei, J. Liu, T. Fujita, R. Liang, V. Ganapathy et al., Identification of a Potential Substrate Binding Domain in the Mammalian Peptide Transporters PEPT1 and PEPT2 Using PEPT1-PEPT2 and PEPT2-PEPT1 Chimeras, Biochemical and Biophysical Research Communications, vol.246, issue.1, pp.39-44, 1998.
DOI : 10.1006/bbrc.1998.8566

T. Terada, H. Saito, K. Sawada, Y. Hashimoto, and K. Inui, N-terminal halves of rat H+/peptide transporters are responsible for their substrate recognition, Pharmaceutical Research, vol.17, issue.1, pp.15-20, 2000.
DOI : 10.1023/A:1007554105597

S. Newstead, D. Drew, A. Cameron, V. Postis, X. Xia et al., Crystal structure of a prokaryotic homologue of the mammalian oligopeptide-proton symporters, PepT1 and PepT2, The EMBO Journal, vol.105, issue.2, pp.417-443, 2011.
DOI : 10.1073/pnas.0800825105

D. Meredith and R. Price, Molecular Modeling of PepT1 ??? Towards a Structure, Journal of Membrane Biology, vol.250, issue.Suppl, pp.79-88, 2006.
DOI : 10.1007/s00232-006-0876-6

D. Meredith and C. A. Boyd, Oligopeptide transport by epithelial cells, The Journal of Membrane Biology, vol.145, issue.1, pp.1-12, 1995.
DOI : 10.1007/BF00233302

H. Daniel, First insights into the operational mode of epithelial peptide transporters., The Journal of Physiology, vol.498, issue.3, p.561, 1997.
DOI : 10.1113/jphysiol.1997.sp021882

C. S. Temple, J. R. Bronk, P. D. Bailey, and C. A. Boyd, Substrate-charge dependence of stoichiometry shows membrane potential is the driving force for proton-peptide cotransport in rat renal cortex, Pfl???gers Archiv European Journal of Physiology, vol.19, issue.5, pp.825-834, 1995.
DOI : 10.1007/BF00386182

F. Doring, J. Walter, J. Will, M. Fo¨cking, M. Boll et al., Delta-aminolevulinic acid transport by intestinal and renal peptide transporters and its physiological and clinical implications., Journal of Clinical Investigation, vol.101, issue.12, pp.2761-67, 1998.
DOI : 10.1172/JCI1909

H. Lu and C. Klaassen, Tissue distribution and thyroid hormone regulation of Pept1 and Pept2 mRNA in rodents, Peptides, vol.27, issue.4, pp.850-857, 2006.
DOI : 10.1016/j.peptides.2005.08.012

I. Rubio-aliaga, M. Boll, and H. Daniel, Cloning and Characterization of the Gene Encoding the Mouse Peptide Transporter PEPT2, Biochemical and Biophysical Research Communications, vol.276, issue.2, pp.734-775, 2000.
DOI : 10.1006/bbrc.2000.3546

H. Søndergaard, B. Brodin, and C. Nielsen, hPEPT1 is responsible for uptake and transport of Gly-Sar in the human bronchial airway epithelial cell-line Calu-3, Pfl??gers Archiv - European Journal of Physiology, vol.47, issue.3, pp.611-613, 2008.
DOI : 10.1016/S0002-9440(10)64013-8

S. Endtera, C. Francombea, M. Ehrhardtb, and . Gumbleton, RT-PCR analysis of ABC, SLC and SLCO drug transporters in human lung epithelial cell models, Journal of Pharmacy and Pharmacology, vol.vol. VII, issue.5, pp.583-591, 2009.
DOI : 10.1053/gast.2001.21176

K. Bleasby, Expression profiles of 50 xenobiotic transporter genes in humans and pre-clinical species: A resource for investigations into drug disposition, Xenobiotica, vol.33, issue.2, pp.963-88, 2006.
DOI : 10.1124/dmd.104.001354

H. Saito, T. Terada, M. Okuda, S. Sasaki, and K. Inui, Molecular cloning and tissue distribution of rat peptide transporter PEPT2, Biochimica et Biophysica Acta (BBA) - Biomembranes, vol.1280, issue.2, pp.173-180, 1996.
DOI : 10.1016/0005-2736(96)00024-7

S. Ayyadurai, M. A. Charania, E. Xiao, D. Viennoisand, and . Merlin, PepT1 expressed in immune cells has an important role in promoting the immune response during experimentally induced colitis, Laboratory Investigation, vol.32, issue.8, pp.888-899, 2013.
DOI : 10.1016/j.it.2011.02.002

Y. Fei, Y. Kanai, S. Nussberger, V. Ganapathy, F. H. Leibach et al., Expression cloning of a mammalian proton-coupled oligopeptide transporter, Nature, vol.368, issue.6471, pp.563-566, 1994.
DOI : 10.1038/368563a0

P. Luckner and M. Brandsch, Interaction of 31 ??-lactam antibiotics with the H+/peptide symporter PEPT2: analysis of affinity constants and comparison with PEPT1, European Journal of Pharmaceutics and Biopharmaceutics, vol.59, issue.1, pp.17-24, 2005.
DOI : 10.1016/j.ejpb.2004.07.008

G. Müns, J. Vishwanatha, and R. , Regulation of angiotensin I-converting enzyme in cultured bovine bronchial epithelial cells, Journal of Cellular Biochemistry, vol.1, issue.4, pp.352-361, 1993.
DOI : 10.1165/ajrcmb/2.3.245

W. Kammouni, C. Figarella, S. Marchand, and M. Merten, Altered cytokine production by cystic fibrosis tracheal gland serous cells, Infect Immun, vol.65, pp.5176-83, 1997.

W. Kammouni, D. Naïmi, W. Renaud, N. Bianco, C. Figarella et al., High lysosomal activities in cystic fibrosis tracheal gland cells corrected by adenovirus-mediated CFTR gene transfer, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, vol.1453, issue.1, pp.14-22, 1999.
DOI : 10.1016/S0925-4439(98)00084-2

S. El-chemaly, R. Ali-rachedi, C. Gerson, M. Cobas, R. Forteza et al., Lactoperoxidase and human airway host defense, Am J Respir Cell Mol Biol, vol.29, pp.206-218, 2003.

S. Stabler, Deficiency, New England Journal of Medicine, vol.368, issue.2, pp.149-60, 2013.
DOI : 10.1056/NEJMcp1113996

T. Robak, . Gladalska, E. Stepie?, and . Robak, Serum levels of interleukin-6 type cytokines and soluble interleukin-6 receptor in patients with rheumatoid arthritis, Mediators of Inflammation, vol.7, issue.5, pp.347-353, 1998.
DOI : 10.1080/09629359890875

C. Feghali and T. Wright, Cytokines in acute and chronic inflammation, Front Biosci, vol.1, pp.12-26, 1997.

F. Xi-mi and Z. , Hypomethylation of interleukin-4 and -6 promoters in T cells from systemic lupus erythematosus patients, Acta Pharmacologica Sinica, vol.29, issue.1, pp.105-107, 2008.

E. Joseph, B. De-larco, R. K. Beverly, D. Wuertz, B. L. Yee et al., Atypical methylation of the interleukin-8 gene correlates strongly with the metastatic potential of breast carcinoma cells, PNAS, vol.100, pp.24-13988, 2003.

R. Ross, Atherosclerosis ??? An Inflammatory Disease, New England Journal of Medicine, vol.340, issue.2, pp.115-141, 1999.
DOI : 10.1056/NEJM199901143400207

M. Hofmann, Hyperhomocysteinemia enhances vascular inflammation and accelerates atherosclerosis in a murine model, Journal of Clinical Investigation, vol.107, issue.6, pp.675-83, 2001.
DOI : 10.1172/JCI10588

M. Hofmann, S. Drury, C. Fu, W. Qu, A. Taguchi et al., RAGE Mediates a Novel Proinflammatory Axis, Cell, vol.97, issue.7, pp.889-901, 1999.
DOI : 10.1016/S0092-8674(00)80801-6

C. Wheatley, The return of the Scarlet Pimpernel: cobalamin in inflammation II ??? cobalamins can both selectively promote all three nitric oxide synthases (NOS), particularly iNOS and eNOS, and, as needed, selectively inhibit iNOS and nNOS, Journal of Nutritional & Environmental Medicine, vol.44, issue.3, pp.181-211, 2007.
DOI : 10.1073/pnas.95.13.7631

G. Biolo, G. Toigo, and B. Ciocchi, Metabolic response to injury and sepsis: changes in protein metabolism, Nutrition, vol.13, issue.9, pp.52-57, 1997.
DOI : 10.1016/S0899-9007(97)83044-4

C. Obled, I. Papet, and D. Breuille, Metabolic bases of amino acid requirements in acute diseases, Current Opinion in Clinical Nutrition and Metabolic Care, vol.5, issue.2, pp.189-97, 2002.
DOI : 10.1097/00075197-200203000-00012

P. Reeds and F. Jahoor, The amino acid requirements of disease, Clinical Nutrition, vol.20, pp.15-22, 2001.
DOI : 10.1054/clnu.2001.0402

K. Storch, D. Wagner, J. Burke, and V. Young, Quantitative study in vivo of methionine cycle in humans using [methyl-2H3]- and [1-13C]methionine, American Journal of Physiology-Endocrinology and Metabolism, vol.101, issue.3, pp.322-331, 1988.
DOI : 10.1152/jappl.1984.56.1.230

J. Haley, Compositions de cobalamine et procedes destines au traitement ou a la prevention de la mucosite, 2007.

B. C. Shin, Compositions pour application externe contenant de l'adenosyl cobalamine, destinees au traitement des maladies de la peau, 2007.

K. Nilsson, B. Hultberg, and L. Gustafson, Lack of Association between Plasma Homocysteine and Inflammation in Psychogeriatric Patients, Dementia and Geriatric Cognitive Disorders, vol.14, issue.3, pp.151-156, 2002.
DOI : 10.1159/000063602

D. M. Stuart, Method for treating generalized and focal peripheral neuropathies locales, 2008.

U. Funada, M. Wada, T. Kawata, N. Tanaka, T. Tadokoro et al., Effect of Cobalamin on the Allergic Response in Mice, Bioscience, Biotechnology, and Biochemistry, vol.64, issue.10, pp.2053-58, 2000.
DOI : 10.1271/bbb.64.2053

J. Wang, R. J. Homer, Q. Chen, and J. A. Elias, Endogenous and Exogenous IL-6 Inhibit Aeroallergen-Induced Th2 Inflammation, The Journal of Immunology, vol.165, issue.7, pp.4051-61, 2000.
DOI : 10.4049/jimmunol.165.7.4051

URL : http://www.jimmunol.org/content/jimmunol/165/7/4051.full.pdf

. Atopy, asthma, and lung function in relation to folate and vitamin B12 in adults, Allergy, vol.65, pp.1446-54, 2010.

S. Hodge, G. Hodge, M. Holmes, R. Flower, and R. Scicchitano, Interleukin-4 and tumour necrosis factor-?? inhibit transforming growth factor-?? production in a human bronchial epithelial cell line: Possible relevance to inflammatory mechanisms in chronic obstructive pulmonary disease, Respirology, vol.176, issue.3, pp.205-216, 2001.
DOI : 10.1084/jem.176.5.1381

H. Takizawa, T. Ohtoshi, T. Kikutani, H. Okazaki, N. Akiyama et al., Histamine Activates Bronchial Epithelial Cells to Release Inflammatory Cytokines in vitro, International Archives of Allergy and Immunology, vol.108, issue.3, pp.260-267, 1995.
DOI : 10.1159/000237162

Y. Aoki, D. Qiu, G. Zhao, and P. Kao, Leukotriene B4 mediates histamine induction of NF-kappaB and IL-8

Y. Li, L. Chi, D. J. Stechschulte, and K. N. Dileepan, Histamine-Induced Production of Interleukin-6 and Interleukin-8 by Human Coronary Artery Endothelial Cells Is Enhanced by Endotoxin and Tumor Necrosis Factor-??, Microvascular Research, vol.61, issue.3, pp.253-62, 2001.
DOI : 10.1006/mvre.2001.2304

S. Lee, A. Tsou, H. Chan, J. Thomas, K. Petrie et al., Glucocorticoids selectively inhibit the transcription of the interleukin 1 beta gene and decrease the stability of interleukin 1 beta mRNA., Proceedings of the National Academy of Sciences, vol.85, issue.4, pp.1204-1212, 1988.
DOI : 10.1073/pnas.85.4.1204

S. Arya, F. Wong-staal, and R. Gallo, Dex-mediated inhibition of human T cell growth factor and gammainterferon messenger RNA, J Immunol, vol.133, pp.273-279, 1984.

N. Mukaida, G. Gussella, T. Kasahara, Y. Ko, C. Zachariae et al., Molecular analysis of the inhibition of interleukin-8 production by dexamethasone in a human fibrosarcoma cell line

N. Mukaida, M. Morita, Y. Ishikawa, N. Rice, S. Okamoto et al., Novel mechanism of glucocorticoid-mediated gene repression. Nuclear factor-kappa B is target for glucocorticoid-mediated interleukin 8 gene repression, J Biol Chem, vol.269, pp.13289-95, 1994.

A. Ray, K. Laforge, and P. Sehgal, On the mechanism for efficient repression of the interleukin-6 promoter by glucocorticoids: enhancer, TATA box, and RNA start site (Inr motif) occlusion., Molecular and Cellular Biology, vol.10, issue.11, pp.5736-5782, 1990.
DOI : 10.1128/MCB.10.11.5736

A. So, C. Chaivorapol, E. Bolton, H. Li, and K. Yamamoto, Determinants of Cell- and Gene-Specific Transcriptional Regulation by the Glucocorticoid Receptor, PLoS Genetics, vol.32, issue.6, p.94, 2007.
DOI : 10.1371/journal.pgen.0030094.st004

O. Eickelberg, M. Roth, R. Lörx, V. Bruce, J. Rüdiger et al., Ligand-independent Activation of the Glucocorticoid Receptor by b2-Adrenergic Receptor Agonists in Primary Human Lung Fibroblasts and Vascular Smooth Muscle Cells, THE JOURNAL OF BIOLOGICAL CHEMISTRY, pp.1005-1015, 1999.

D. Bosscher and G. Haegeman, Molecular Endocrinology. Minireview: Latest Perspectives on Antiinflammatory Actions of Glucocorticoids, pp.281-310, 2009.

J. C. Webster, C. M. Jewell, J. E. Bodwell, A. Munck, M. Sar et al., Mouse Glucocorticoid Receptor Phosphorylation Status Influences Multiple Functions of the Receptor Protein, Journal of Biological Chemistry, vol.260, issue.14
DOI : 10.1210/mend-2-12-1256

R. D. Blind and M. J. Garabedian, Differential recruitment of glucocorticoid receptor phospho-isoforms to glucocorticoid-induced genes, The Journal of Steroid Biochemistry and Molecular Biology, vol.109, issue.1-2, pp.150-157, 2008.
DOI : 10.1016/j.jsbmb.2008.01.002

W. Chen, T. Dang, R. D. Blind, Z. Wang, C. N. Cavasotto et al., Glucocorticoid Receptor Phosphorylation Differentially Affects Target Gene Expression, Molecular Endocrinology, vol.22, issue.8
DOI : 10.1210/me.2007-0219

URL : https://academic.oup.com/mend/article-pdf/22/8/1754/8941373/mend1754.pdf

Z. Wang, W. Chen, E. Kono, T. Dang, and M. Garabedian, Modulation of Glucocorticoid Receptor Phosphorylation and Transcriptional Activity by a C-Terminal-Associated Protein Phosphatase, Molecular Endocrinology, vol.21, issue.3, pp.625-659, 2007.
DOI : 10.1210/me.2005-0338

K. V. Rao and C. F. Fox, Epidermal growth factor stimulates tyrosine phosphorylation of human glucocorticoid receptor in cultured cells, Proc. Natl. Acad. Sci, pp.10762-67, 1987.
DOI : 10.1016/S0006-291X(87)80539-9

S. Battaglia-hsu, N. Akchiche, N. Noel, J. Alberto, E. Jeannesson et al., Vitamin B12 deficiency reduces proliferation and promotes differentiation of neuroblastoma cells and up-regulates PP2A, proNGF, and TACE, Proceedings of the National Academy of Sciences, vol.391, issue.1, pp.21930-21935, 2009.
DOI : 10.1007/s00216-008-1953-8

Y. Kobayashi, N. Mercado, P. Barnes, and K. Ito, Defects of Protein Phosphatase 2A Causes Corticosteroid Insensitivity in Severe Asthma, PLoS ONE, vol.6, issue.12, p.27627, 2011.
DOI : 10.1371/journal.pone.0027627.s002

K. Ronacher, K. Hadley, C. Avenant, E. Stubsrud, S. Simons et al., Ligand-selective transactivation and transrepression via the glucocorticoid receptor: Role of cofactor interaction, Molecular and Cellular Endocrinology, vol.299, issue.2, pp.219-250, 2009.
DOI : 10.1016/j.mce.2008.10.008

A. Chen, S. Innis, A. Davidson, and S. James, Phosphatidylcholine and lysophosphatidylcholine excretion is increased in children with cystic fibrosis and is associated with plasma homocysteine, S-adenosylhomocysteine, and S-adenosylmethionine, The American Journal of Clinical Nutrition, vol.16, issue.suppl, pp.686-91, 2005.
DOI : 10.1007/BF02534900

V. Marguerite, E. Gkikopoulou, J. Alberto, J. Guéant, and M. Merten, Phospholipase D activation mediates cobalamin-induced downregulation of Multidrug Resistance-1 gene and increase in sensitivity to vinblastine in HepG2 cells, The International Journal of Biochemistry & Cell Biology, vol.45, issue.2, pp.213-233, 2013.
DOI : 10.1016/j.biocel.2012.09.018

P. Analyse-par and L. De-l-'adn-immunoprécipité, Toutes les réactions de PCR ont été réalisées dans un volume final de 25 ?L contenant : 0,2 mM de chacun des désoxyribonucléotides (dNTP), le tampon de la Taq concentré une fois 5 ?L de la solution d'ADN (diluée 8 fois pour les Input) et 1 ?M de chacune des amorces sens et antisens, p.2

P. Dans-chaque-série-de, Les ADN amplifiés sont migrés sur un gel d'agarose à 2% contenant 0,05 ?g/mL de bromure d'éthidium. Le gel est alors scanné. Les bandes ont été quantifiées par densitométrie et analysées avec le logiciel « Image J » developpé par « National Institutes of Health ». L'enrichissement en ADN immunoprécipité (IP) est déterminé en divisant la densité des produits de PCR IP par celle des inputs (INP), pour les échantillons contrôles et traités puis en comparant le rapport entre les valeurs obtenues. La PCR est réalisée selon le protocole suivant : une incubation à 94°C pendant 3 min, pp.585-586