M. Zasloff, Antimicrobial peptides of multicellular organisms, Nature, vol.415, pp.389-95, 2002.

M. Seo, H. Won, J. Kim, T. Mishig-ochir, and B. Lee, Antimicrobial peptides for therapeutic applications: a review, Molecules, vol.17, pp.12276-86, 2012.

K. Matsuzaki, Why and how are peptide-lipid interactions utilized for selfdefense? Magainins and tachyplesins as archetypes, Biochim Biophys Acta, vol.1462, pp.1-10, 1999.

Y. Shai, Mechanism of the binding, insertion and destabilization of phospholipid bilayer membranes by alpha-helical antimicrobial and cell nonselective membrane-lytic peptides, Biochim Biophys Acta, vol.1462, pp.55-70, 1999.

K. A. Brogden, Antimicrobial peptides: pore formers or metabolic inhibitors in bacteria?, Nat Rev Microbiol, vol.3, pp.238-50, 2005.

K. A. Brogden, M. Ackermann, P. B. Mccray, and B. F. Tack, Antimicrobial peptides in animals and their role in host defences, Int J Antimicrob Agents, vol.22, pp.465-78, 2003.

H. Jenssen, P. Hamill, and R. Hancock, Peptide antimicrobial agents, Clin Microbiol Rev, vol.19, pp.491-511, 2006.

A. L. Hilchie, K. Wuerth, and R. Hancock, Immune modulation by multifaceted cationic host defense (antimicrobial) peptides, Nat Chem Biol, vol.9, pp.761-769, 2013.

S. C. Mansour, O. M. Pena, and R. Hancock, Host defense peptides: front-line immunomodulators, Trends Immunol, vol.35, pp.443-50, 2014.

A. Nijnik and R. Hancock, Host defence peptides: antimicrobial and immunomodulatory activity and potential applications for tackling antibiotic-resistant infections, Emerg Health Threats J, vol.2, p.1, 2009.

D. Smet, K. Contreras, and R. , Human antimicrobial peptides: defensins, cathelicidins and histatins, Biotechnol Lett, vol.27, pp.1337-1384, 2005.

D. Yang, O. Chertov, and J. J. Oppenheim, Participation of mammalian defensins and cathelicidins in anti-microbial immunity: receptors and activities of human defensins and cathelicidin (LL-37), J Leukoc Biol, vol.69, pp.691-698, 2001.

A. Nijnik, J. Pistolic, A. Wyatt, S. Tam, and R. Hancock, Human cathelicidin peptide LL-37 modulates the effects of IFN-gamma on APCs, J Immunol, vol.183, pp.5788-98, 2009.

D. Bowdish, D. J. Davidson, D. P. Speert, and R. Hancock, The human cationic peptide LL-37 induces activation of the extracellular signal-regulated kinase and p38 kinase pathways in primary human monocytes, J Immunol, vol.172, pp.3758-65, 2004.

A. Elssner, M. Duncan, M. Gavrilin, and M. D. Wewers, A novel P2X7 receptor activator, the human cathelicidin-derived peptide LL37, induces IL-1 beta processing and release, J Immunol, vol.172, pp.4987-94, 2004.

J. Yu, N. Mookherjee, K. Wee, D. Bowdish, J. Pistolic et al., Host defense peptide LL-37, in synergy with inflammatory mediator IL-1beta, augments immune responses by multiple pathways, J Immunol, vol.179, pp.7684-91, 2007.

S. M. Alalwani, J. Sierigk, C. Herr, O. Pinkenburg, R. Gallo et al., The antimicrobial peptide LL-37 modulates the inflammatory and host defense response of human neutrophils, Eur J Immunol, vol.40, pp.1118-1144, 2010.

M. G. Scott, D. J. Davidson, M. R. Gold, D. Bowdish, and R. Hancock, The human antimicrobial peptide LL-37 is a multifunctional modulator of innate immune responses, J Immunol, vol.169, pp.3883-91, 2002.

X. Tang, D. Basavarajappa, J. Z. Haeggstrom, and M. Wan, P2X7 receptor regulates internalization of antimicrobial peptide LL-37 by human macrophages that promotes intracellular pathogen clearance, J Immunol, vol.195, pp.1191-201, 2015.

P. G. Barlow, Y. Li, T. S. Wilkinson, D. Bowdish, Y. E. Lau et al., The human cationic host defense peptide LL-37 mediates contrasting effects on apoptotic pathways in different primary cells of the innate immune system, J Leukoc Biol, vol.80, pp.509-529, 2006.

P. G. Barlow, P. E. Beaumont, C. Cosseau, A. Mackellar, T. S. Wilkinson et al., The human cathelicidin LL-37 preferentially promotes apoptosis of infected airway epithelium, Am J Respir Cell Mol Biol, vol.43, pp.692-702, 2010.

M. Carretero, M. J. Escámez, M. García, B. Duarte, A. Holguín et al., In vitro and in vivo wound healing-promoting activities of human cathelicidin LL-37, J Invest Dermatol, vol.128, pp.223-259, 2008.

R. Koczulla, G. Von-degenfeld, C. Kupatt, F. Krötz, S. Zahler et al., An angiogenic role for the human peptide antibiotic LL-37/hCAP-18, J Clin Invest, vol.111, pp.1665-72, 2003.

M. D. Salvado, D. Gennaro, A. Lindbom, L. Agerberth, B. Haeggström et al., Cathelicidin LL-37 induces angiogenesis via PGE2-EP3 signaling in endothelial cells, in vivo inhibition by aspirin, Arterioscler Thromb Vasc Biol, vol.33, pp.1965-72, 2013.

M. B. Pucca, F. A. Cerni, P. El, K. F. Zoccal, C. F. De-bordon et al., Non-disulfide-bridged peptides from Tityus serrulatus venom: evidence for proline-free ACE-inhibitors, Peptides, vol.82, pp.44-51, 2016.

R. Tavano, D. Segat, M. Gobbo, and E. Papini, The honeybee antimicrobial peptide apidaecin differentially immunomodulates human macrophages, monocytes and dendritic cells, J Innate Immun, vol.3, pp.614-636, 2011.

D. Y. Null, Q. Chen, A. P. Schmidt, G. M. Anderson, J. M. Wang et al., LL-37, the neutrophil granule-and epithelial cell-derived cathelicidin, utilizes formyl peptide receptor-like 1 (FPRL1) as a receptor to chemoattract human peripheral blood neutrophils, monocytes, and T cells, J Exp Med, vol.192, pp.1069-74, 2000.

R. Smithrithee, F. Niyonsaba, C. Kiatsurayanon, H. Ushio, S. Ikeda et al., Human ?-defensin-3 increases the expression of interleukin-37 through CCR6 in human keratinocytes, J Dermatol Sci, vol.77, pp.46-53, 2015.

M. Wan, C. Godson, P. J. Guiry, B. Agerberth, and J. Z. Haeggström, Leukotriene B4/ antimicrobial peptide LL-37 proinflammatory circuits are mediated by BLT1 and FPR2/ALX and are counterregulated by lipoxin A4 and resolvin E1, FASEB J, vol.25, pp.1697-705, 2011.

M. Wan, A. M. Van-der-does, X. Tang, L. Lindbom, B. Agerberth et al., Antimicrobial peptide LL-37 promotes bacterial phagocytosis by human macrophages, J Leukoc Biol, vol.95, pp.971-81, 2014.

A. Rifflet, S. Gavalda, N. Téné, J. Orivel, J. Leprince et al., Identification and characterization of a novel antimicrobial peptide from the venom of the ant Tetramorium bicarinatum, Peptides, vol.38, pp.363-70, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00996401

A. Mantovani, A. Sica, S. Sozzani, P. Allavena, A. Vecchi et al., The chemokine system in diverse forms of macrophage activation and polarization, Trends Immunol, vol.25, pp.677-86, 2004.

F. O. Martinez, A. Sica, A. Mantovani, and M. Locati, Macrophage activation and polarization, Front Biosci, vol.13, pp.453-61, 2008.

A. Coste, M. Dubourdeau, M. D. Linas, S. Cassaing, J. Lepert et al., PPARgamma promotes mannose receptor gene expression in murine macrophages and contributes to the induction of this receptor by IL-13, Immunity, vol.19, pp.329-368, 2003.

A. Galès, A. Conduché, J. Bernad, L. Lefevre, D. Olagnier et al., PPAR? controls dectin-1 expression required for host antifungal defense against Candida albicans, PLoS Pathog, vol.6, p.1000714, 2010.

L. Lefèvre, G. Lugo-villarino, E. Meunier, A. Valentin, D. Olagnier et al., The C-type lectin receptors dectin-1, MR, and SIGNR3 contribute both positively and negatively to the macrophage response to Leishmania infantum, Immunity, vol.38, pp.1038-1087, 2013.

L. Lefèvre, H. Authier, S. Stein, C. Majorel, B. Couderc et al., LRH-1 mediates anti-inflammatory and antifungal phenotype of IL-13-activated macrophages through the PPAR? ligand synthesis, Nat Commun, vol.6, p.6801, 2015.

M. G. Netea, N. Gow, C. A. Munro, S. Bates, C. Collins et al., Immune sensing of Candida albicans requires cooperative recognition of mannans and glucans by lectin and Toll-like receptors, J Clin Invest, vol.116, pp.1642-50, 2006.

S. Saijo and Y. Iwakura, Dectin-1 and dectin-2 in innate immunity against fungi, Int Immunol, vol.23, pp.467-72, 2011.

P. R. Taylor, S. V. Tsoni, J. A. Willment, K. M. Dennehy, M. Rosas et al., Dectin-1 is required for ?-glucan recognition and control of fungal infection, Nat Immunol, vol.8, pp.31-39, 2007.

F. L. Van-de-veerdonk, L. Joosten, I. Devesa, H. M. Mora-montes, T. Kanneganti et al., Bypassing pathogen-induced inflammasome activation for the regulation of interleukin-1beta production by the fungal pathogen Candida albicans, J Infect Dis, vol.199, pp.1087-96, 2009.

B. A. Wevers, T. Geijtenbeek, and S. I. Gringhuis, C-type lectin receptors orchestrate antifungal immunity, Future Microbiol, vol.8, pp.839-54, 2013.

A. Coste, C. Lagane, C. Filipe, H. Authier, A. Gales et al., IL-13 attenuates gastrointestinal candidiasis in normal and immunodeficient RAG-2-/-mice via peroxisome proliferator-activated receptor-activation, J Immunol, vol.180, pp.4939-4986, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00283071

R. A. Daynes and D. C. Jones, Emerging roles of PPARS in inflammation and immunity, Nat Rev Immunol, vol.2, pp.748-59, 2002.

S. A. Kliewer, J. M. Lenhard, T. M. Willson, I. Patel, D. C. Morris et al., A prostaglandin J2 metabolite binds peroxisome proliferator-activated receptor gamma and promotes adipocyte differentiation, Cell, vol.83, pp.813-822, 1995.

L. Lefèvre, A. Galès, D. Olagnier, J. Bernad, L. Perez et al., PPAR? ligands switched high fat diet-induced macrophage M2b polarization toward M2a thereby improving intestinal Candida elimination, PLoS One, vol.5, p.12828, 2010.

L. Marec, O. Neveu, C. Lefranc, B. Dubessy, C. Boutin et al., Structure-activity relationships of a series of analogues of the RFamide-related peptide 26RFa, J Med Chem, vol.54, pp.4806-4820, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01962818

A. Berry, P. Balard, A. Coste, D. Olagnier, C. Lagane et al., IL-13 induces expression of CD36 in human monocytes through PPARgamma activation, Eur J Immunol, vol.37, pp.1642-52, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00166681

O. Gross, H. Poeck, M. Bscheider, C. Dostert, N. Hannesschläger et al., Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence, Nature, vol.459, pp.433-439, 2009.

K. Gupta, H. Subramanian, and H. Ali, Modulation of host defense peptide-mediated human mast cell activation by LPS, Innate Immun, vol.22, pp.21-30, 2016.

P. Pundir, A. Catalli, C. Leggiadro, S. E. Douglas, and M. Kulka, Pleurocidin, a novel antimicrobial peptide, induces human mast cell activation through the FPRL1 receptor, Mucosal Immunol, vol.7, pp.177-87, 2014.

M. Wan, O. Soehnlein, X. Tang, A. M. Van-der-does, E. Smedler et al., Cathelicidin LL-37 induces time-resolved release of LTB4 and TXA2 by human macrophages and triggers eicosanoid generation in vivo, FASEB J, vol.28, pp.3456-67, 2014.

Y. Zhang, Y. Yu, Y. Zhang, W. Zhang, and B. Yu, The modulatory effect of TLR2 on LL-37-induced human mast cells activation, Biochem Biophys Res Commun, vol.470, pp.368-74, 2016.

A. M. Van-der-does, H. Beekhuizen, B. Ravensbergen, T. Vos, T. Ottenhoff et al., LL-37 directs macrophage differentiation toward macrophages with a proinflammatory signature, J Immunol, vol.185, pp.1442-1451, 2010.

J. Willems, W. Noppe, L. Moerman, J. Van-der-walt, and F. Verdonck, Cationic peptides from scorpion venom can stimulate and inhibit polymorphonuclear granulocytes, Toxicon, vol.40, pp.183-189, 2002.

B. N. Gantner, R. M. Simmons, and D. M. Underhill, Dectin-1 mediates macrophage recognition of Candida albicans yeast but not filaments, EMBO J, vol.24, pp.1277-86, 2005.

J. A. Willment and G. D. Brown, C-type lectin receptors in antifungal immunity, Trends Microbiol, vol.16, pp.27-32, 2008.

J. I. Odegaard, R. R. Ricardo-gonzalez, M. H. Goforth, C. R. Morel, V. Subramanian et al., Macrophage-specific PPAR? controls alternative activation and improves insulin resistance, Nature, vol.447, pp.1116-1136, 2007.

L. P. Erwig and N. Gow, Interactions of fungal pathogens with phagocytes, Nat Rev Microbiol, vol.14, pp.163-76, 2016.

L. Maródi, H. M. Korchak, and R. B. Johnston, Mechanisms of host defense against Candida species. I. Phagocytosis by monocytes and monocyte-derived macrophages, J Immunol, vol.146, pp.2783-2792, 1991.

Y. R. Bommineni, G. H. Pham, L. T. Sunkara, M. Achanta, and G. Zhang, Immune regulatory activities of fowlicidin-1, a cathelicidin host defense peptide, Mol Immunol, vol.59, pp.55-63, 2014.

N. Mookherjee, K. L. Brown, D. Bowdish, S. Doria, R. Falsafi et al., Modulation of the TLR-mediated inflammatory response by the endogenous human host defense peptide LL-37, J Immunol, vol.176, pp.2455-64, 2006.

F. Niyonsaba, L. Madera, N. Afacan, K. Okumura, H. Ogawa et al., The innate defense regulator peptides IDR-HH2, IDR-1002, and IDR-1018 modulate human neutrophil functions, J Leukoc Biol, vol.94, pp.159-70, 2013.

H. Subramanian, K. Gupta, Q. Guo, R. Price, and H. Ali, Mas-related gene X2 (MrgX2) is a novel G protein-coupled receptor for the antimicrobial peptide LL-37 in human mast cells: resistance to receptor phosphorylation, desensitization, and internalization, J Biol Chem, vol.286, pp.44739-44788, 2011.

H. B. Yu and B. B. Finlay, The caspase-1 inflammasome: a pilot of innate immune responses, Cell Host Microbe, vol.4, pp.198-208, 2008.

J. El-benna, P. Dang, G. , and M. , Braut-Boucher F. p47phox, the phagocyte NADPH oxidase/NOX2 organizer: structure, phosphorylation and implication in diseases, Exp Mol Med, vol.41, p.217, 2009.

F. Osorio and C. Reis-e-sousa, Myeloid C-type lectin receptors in pathogen recognition and host defense, Immunity, vol.34, pp.651-64, 2011.

, Frontiers in Immunology | www.frontiersin.org November, vol.8, p.1650, 2017.

F. Martinon, A. Mayor, and J. Tschopp, The inflammasomes: guardians of the body, Annu Rev Immunol, vol.27, pp.229-65, 2009.

C. Dostert, V. Pétrilli, R. Van-bruggen, C. Steele, B. T. Mossman et al., Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica, Science, vol.320, pp.674-681, 2008.

J. Tschopp and K. Schroder, NLRP3 inflammasome activation: the convergence of multiple signalling pathways on ROS production?, Nat Rev Immunol, vol.10, pp.210-215, 2010.

R. Zhou, A. S. Yazdi, P. Menu, and J. Tschopp, A role for mitochondria in NLRP3 inflammasome activation, Nature, vol.469, pp.221-226, 2011.

J. M. Kim, Antimicrobial proteins in intestine and inflammatory bowel diseases, Intest Res, vol.12, pp.20-33, 2014.

L. Moerman, F. Verdonck, J. Willems, J. Tytgat, and S. Bosteels, Antimicrobial peptides from scorpion venom induce Ca(2+) signaling in HL-60 cells, Biochem Biophys Res Commun, vol.311, pp.90-97, 2003.

J. D. Clark, L. L. Lin, R. W. Kriz, C. S. Ramesha, L. A. Sultzman et al., A novel arachidonic acid-selective cytosolic PLA2 contains a Ca(2+)-dependent translocation domain with homology to PKC and GAP, Cell, vol.65, pp.1043-51, 1991.

R. M. Kramer, E. F. Roberts, J. Manetta, and J. E. Putnam, The Ca2(+)-sensitive cytosolic phospholipase A2 is a 100-kDa protein in human monoblast U937 cells, J Biol Chem, vol.266, pp.5268-72, 1991.