J. F. Bach, The effect of infections on susceptibility to autoimmune and allergic diseases, N Engl J Med, vol.347, pp.911-920, 2002.

D. E. Elliott, J. J. Urban, C. K. Argo, and J. V. Weinstock, Does the failure to acquire helminthic parasites predispose to Crohn's disease?, FASEB J, vol.14, pp.1848-1855, 2000.

D. P. Strachan, Hay fever, hygiene, and household size, BMJ, vol.299, pp.1259-1260, 1989.

D. C. Baumgart and W. J. Sandborn, Crohn's disease, Lancet, vol.380, pp.1590-1605, 2012.

M. Economou and G. Pappas, New global map of Crohn's disease: Genetic, environmental, and socioeconomic correlations, Inflamm Bowel Dis, vol.14, pp.709-720, 2008.

F. Guarner, Mechanisms of disease: the hygiene hypothesis revisited, Nat Clin Pract Gastr, vol.3, pp.275-284, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00294430

D. E. Elliott and J. V. Weinstock, Where are we on worms?, Curr Opin Gastroenterol, vol.28, pp.551-556, 2012.

D. Ramanan, Helminth infection promotes colonization resistance via type 2 immunity, Science, vol.352, pp.608-612, 2016.

R. W. Li, Alterations in the porcine colon microbiota induced by the gastrointestinal nematode Trichuris suis, Infect Immun, vol.80, pp.2150-2157, 2012.

A. J. Daveson, Effect of hookworm infection on wheat challenge in celiac disease-a randomised double-blinded placebo controlled trial, PLoS ONE, vol.6, p.17366, 2011.

P. Giacomin, Changes in duodenal tissue-associated microbiota following hookworm infection and consecutive gluten challenges in humans with coeliac disease, Sci Rep, vol.6, p.36797, 2016.

H. Helmby, Human helminth therapy to treat inflammatory disorders -where do we stand?, BMC Immunol, vol.16, p.12, 2015.

J. S. Pittman, G. Shepherd, B. J. Thacker, and G. H. Myers, Trichuris suis in finishing pigs: Case report and review, J Swine Health, vol.18, pp.306-313, 2010.

R. J. Beer, Morphological descriptions of the egg and larval stages of Trichuris suis Schrank, 1788, Parasitology, vol.67, pp.263-278, 1973.

R. J. Beer, The relationship between Trichuris trichiura (Linnaeus 1758) of man and Trichuris suis (Schrank 1788) of the pig, Res Vet Sci, vol.20, pp.47-54, 1976.

C. Zaph, P. J. Cooper, and N. L. Harris, Mucosal immune responses following intestinal nematode infection, Parasite Immunol, vol.36, pp.439-452, 2014.

D. E. Elliott and J. V. Weinstock, Helminth-host immunological interactions: prevention and control of immune-mediated diseases, Ann NY Acad Sci, vol.1247, pp.83-96, 2012.

M. Gerencer, A. Marinculic, D. Rapic, M. Frankovic, and I. Valpotic, Immunosuppression of in vivo and in vitro lymphocyte responses in swine induced by Trichinella spiralis or excretory-secretory antigens of the parasite, Vet Parasitol, vol.44, pp.263-273, 1992.

M. Langelaar, Suppression of dendritic cell maturation by Trichinella spiralis excretory/secretory products, Parasite Immunol, vol.31, pp.641-645, 2009.

X. Bai, Regulation of cytokine expression in murine macrophages stimulated by excretory/secretory products from Trichinella spiralis in vitro, Mol Cell Biochem, vol.360, pp.79-88, 2012.

C. Aranzamendi, Trichinella spiralis-secreted products modulate DC functionality and expand regulatory T cells in vitro, Parasite Immunol, vol.34, pp.210-223, 2012.

M. Segura, Z. Su, C. Piccirillo, and M. M. Stevenson, Impairment of dendritic cell function by excretory-secretory products: a potential mechanism for nematode-induced immunosuppression, Eur J Immunol, vol.37, pp.1887-1904, 2007.

R. M. Valanparambil, Production and analysis of immunomodulatory excretory-secretory products from the mouse gastrointestinal nematode Heligmosomoides polygyrus bakeri, Nat Protoc, vol.9, pp.2740-2754, 2014.

J. C. Massacand, Helminth products bypass the need for TSLP in Th2 immune responses by directly modulating dendritic cell function, Proc Natl Acad Sci, vol.106, pp.13968-13973, 2009.

H. J. Mcsorley, Suppression of type 2 immunity and allergic airway inflammation by secreted products of the helminth Heligmosomoides polygyrus, Eur J Immunol, vol.42, pp.2667-2682, 2012.

J. R. Grainger, Helminth secretions induce de novo T cell Foxp3 expression and regulatory function through the TGF-beta pathway, J Exp Med, vol.207, pp.2331-2341, 2010.

E. Vendelova, Proteomic analysis of excretory-secretory products of Mesocestoides corti Metacestodes reveals potential suppressors of dendritic cell functions, PLoS Negl Trop Dis, vol.10, p.5061, 2016.

S. M. Geiger, Stage-specific immune responses in human Necator americanus infection, Parasite Immunol, vol.29, pp.347-358, 2007.

I. H. Hiemstra, Excreted/secreted Trichuris suis products reduce barrier function and suppress inflammatory cytokine production of intestinal epithelial cells, Mol Immunol, vol.60, pp.1-7, 2014.

F. Ebner, Therapeutic potential of larval excretory/secretory proteins of the pig whipworm Trichuris suis in allergic disease, Allergy, vol.69, pp.1489-1497, 2014.

M. K. Ottow, The helminth Trichuris suis suppresses TLR4-induced inflammatory responses in human macrophages, Genes Immun, vol.15, pp.477-486, 2014.

, SCIENTIFIC RePoRTS |, vol.8, 2018.

E. J. Klaver, Trichuris suis soluble products induce Rab7b expression and limit TLR4 responses in human dendritic cells, Genes Immun, vol.16, pp.378-387, 2015.

M. A. Hoeksema, Treatment with Trichuris suis soluble products during monocyte-to-macrophage differentiation reduces inflammatory responses through epigenetic remodeling, FASEB J, vol.30, pp.2826-2836, 2016.

G. Kooij, Trichuris suis induces human non-classical patrolling monocytes via the mannose receptor and PKC: implications for multiple sclerosis, Acta Neuropathol Commun, vol.3, p.45, 2015.

A. Summan, P. Nejsum, and A. R. Williams, Modulation of human dendritic cell activity by Giardia and helminth antigens, Parasite Immunol, vol.40, p.12525, 2018.

S. Navarro, Hookworm recombinant protein promotes regulatory T cell responses that suppress experimental asthma, Sci Transl Med, vol.8, pp.362-143, 2016.

N. Xu, Recombinant Trichinella pseudospiralis serine protease inhibitors alter macrophage polarization in vitro, Front Microbiol, vol.8, p.1834, 2017.

E. Kraus, H. H. Kiltz, and U. F. Femfert, The specificity of proteinase K against oxidized insulin B chain, Hoppe Seylers Z Physiol Chem, vol.357, pp.233-237, 1976.

A. M. Blum, Heligmosomoides polygyrus bakeri induces tolerogenic dendritic cells that block colitis and prevent antigenspecific gut T cell responses, J Immunol, vol.189, pp.2512-2520, 2012.

M. Weng, Alternatively activated macrophages in intestinal helminth infection: effects on concurrent bacterial colitis, J Immunol, vol.179, pp.4721-4731, 2007.

C. D. Mills, K. Kincaid, J. M. Alt, M. J. Heilman, and A. M. Hill, M-1/M-2 macrophages and the Th1/Th2 paradigm, J Immunol, vol.164, pp.6166-6173, 2000.

F. O. Martinez and S. Gordon, The M1 and M2 paradigm of macrophage activation: time for reassessment, vol.6, p.13, 2014.

C. J. Punjabi, D. L. Laskin, D. E. Heck, and J. D. Laskin, Production of nitric oxide by murine bone marrow cells. Inverse correlation with cellular proliferation, J Immunol, vol.149, pp.2179-2184, 1992.

M. Perretti, C. Szabo, and C. Thiemermann, Effect of interleukin-4 and interleukin-10 on leucocyte migration and nitric oxide production in the mouse, Brit J Pharmacol, vol.116, pp.2251-2257, 1995.

C. Bogdan, H. Thuring, M. Dlaska, M. Rollinghoff, and G. Weiss, Mechanism of suppression of macrophage nitric oxide release by IL-13: influence of the macrophage population, J Immunol, vol.159, pp.4506-4513, 1997.

E. M. Coccia, E. Stellacci, G. Marziali, G. Weiss, and A. Battistini, IFN-gamma and IL-4 differently regulate inducible NO synthase gene expression through IRF-1 modulation, Int Immunol, vol.12, pp.977-985, 2000.

V. Bronte, Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards, Nat Commun, vol.7, p.12150, 2016.

T. Kobayashi, NFIL3 is a regulator of IL-12p40 in macrophages and mucosal immunity, J Immunol, vol.186, pp.4649-4655, 2011.

D. Ruffell, A CREB-C/EBPbeta cascade induces M2 macrophage-specific gene expression and promotes muscle injury repair, Proc Natl Acad Sci, vol.106, pp.17475-17480, 2009.

A. P. Hutchins, D. Diez, and D. Miranda-saavedra, The IL-10/STAT3-mediated anti-inflammatory response: recent developments and future challenges, Brief Funct Genomics, vol.12, pp.489-498, 2013.

Y. R. Na, D. Jung, B. R. Yoon, W. W. Lee, and S. H. Seok, Endogenous prostaglandin E2 potentiates anti-inflammatory phenotype of macrophage through the CREB-C/EBP-beta cascade, Eur J Immunol, vol.45, pp.2661-2671, 2015.

S. Sanduja, F. F. Blanco, L. E. Young, V. Kaza, and D. A. Dixon, The role of tristetraprolin in cancer and inflammation, Front Biosci, vol.17, pp.174-188, 2012.

A. M. Owyang, Interleukin 25 regulates type 2 cytokine-dependent immunity and limits chronic inflammation in the gastrointestinal tract, J Exp Med, vol.203, pp.843-849, 2006.

T. Setiawan, Heligmosomoides polygyrus promotes regulatory T-cell cytokine production in the murine normal distal intestine, Infect Immun, vol.75, pp.4655-4663, 2007.

C. Zaph, Epithelial-cell-intrinsic IKK-beta expression regulates intestinal immune homeostasis, Nature, vol.446, pp.552-556, 2007.

N. E. Humphreys, D. Xu, M. R. Hepworth, F. Y. Liew, and R. K. Grencis, IL-33, a potent inducer of adaptive immunity to intestinal nematodes, J Immunol, vol.180, pp.2443-2449, 2008.

B. C. Taylor, TSLP regulates intestinal immunity and inflammation in mouse models of helminth infection and colitis, J Exp Med, vol.206, pp.655-667, 2009.

L. Hang, Heligmosomoides polygyrus bakeri infection activates colonic Foxp3+ T cells enhancing their capacity to prevent colitis, J Immunol, vol.191, pp.1927-1934, 2013.

L. C. Laan, The whipworm (Trichuris suis) secretes prostaglandin E2 to suppress proinflammatory properties in human dendritic cells, FASEB J, vol.31, pp.719-731, 2017.

A. R. Jex, Genome and transcriptome of the porcine whipworm Trichuris suis, Nat Genet, vol.46, pp.701-706, 2014.

B. J. Foth, Whipworm genome and dual-species transcriptome analyses provide molecular insights into an intimate hostparasite interaction, Nat Genet, vol.46, pp.693-700, 2014.

F. A. Simao, R. M. Waterhouse, P. Ioannidis, E. V. Kriventseva, and E. M. Zdobnov, BUSCO: assessing genome assembly and annotation completeness with single-copy orthologs, Bioinformatics, vol.31, pp.3210-3212, 2015.

D. J. Gough, D. E. Levy, R. W. Johnstone, and C. J. Clarke, IFNgamma signaling-does it mean JAK-STAT?, Cytokine Growth F R, vol.19, pp.383-394, 2008.

J. A. Hamerman, Negative regulation of TLR signaling in myeloid cells-implications for autoimmune diseases, Immunological Rev, vol.269, pp.212-227, 2016.

F. Kratochvill, Tristetraprolin limits inflammatory cytokine production in tumor-associated macrophages in an mRNA decayindependent manner, Cancer Res, vol.75, pp.3054-3064, 2015.

T. Kobayashi, NFIL3-deficient mice develop microbiota-dependent, IL-12/23-driven spontaneous colitis, J Immunol, vol.192, pp.1918-1927, 2014.

L. Brys, Reactive oxygen species and 12/15-lipoxygenase contribute to the antiproliferative capacity of alternatively activated myeloid cells elicited during helminth infection, J Immunol, vol.174, pp.6095-6104, 2005.

R. M. Valanparambil, M. Tam, A. Jardim, T. G. Geary, and M. M. Stevenson, Primary Heligmosomoides polygyrus bakeri infection induces myeloid-derived suppressor cells that suppress CD4(+) Th2 responses and promote chronic infection, Mucosal Immunol, vol.10, pp.238-249, 2017.

R. M. Valanparambil, IRF-8 regulates expansion of myeloid-derived suppressor cells and Foxp3+ regulatory T cells and modulates Th2 immune responses to gastrointestinal nematode infection, PLoS Pathog, vol.13, p.1006647, 2017.

Q. Yang, A Schistosoma japonicum infection promotes the expansion of myeloid-derived suppressor cells by activating the JAK/STAT3 pathway, J Immunol, vol.198, pp.4716-4727, 2017.

C. F. Anderson, M. Oukka, V. J. Kuchroo, and D. Sacks, CD4(+)CD25(?)Foxp3(?) Th1 cells are the source of IL-10-mediated immune suppression in chronic cutaneous leishmaniasis, J Exp Med, vol.204, pp.285-297, 2007.

D. Jankovic, Conventional T-bet(+)Foxp3(?) Th1 cells are the major source of host-protective regulatory IL-10 during intracellular protozoan infection, J Exp Med, vol.204, pp.273-283, 2007.

, SCIENTIFIC RePoRTS |, vol.8, 2018.

A. Cope, G. Le-friec, J. Cardone, and C. Kemper, The Th1 life cycle: molecular control of IFN-gamma to IL-10 switching, Trends Imunol, vol.32, pp.278-286, 2011.

J. P. Hewitson, The secreted triose phosphate isomerase of Brugia malayi is required to sustain microfilaria production in vivo, PLoS Pathog, vol.10, p.1003930, 2014.

S. Bellafiore, Direct identification of the Meloidogyne incognita secretome reveals proteins with host cell reprogramming potential, PLoS Pathog, vol.4, p.1000192, 2008.

A. P. Yatsuda, J. Krijgsveld, A. W. Cornelissen, A. J. Heck, and E. De-vries, Comprehensive analysis of the secreted proteins of the parasite Haemonchus contortus reveals extensive sequence variation and differential immune recognition, J Biol Chem, vol.278, pp.16941-16951, 2003.

G. M. Knudsen, K. F. Medzihradszky, K. C. Lim, E. Hansell, and J. Mckerrow, Proteomic analysis of Schistosoma mansoni cercarial secretions, Mol Cell Proteomics: MCP, vol.4, pp.1862-1875, 2005.

J. F. Miranda-ozuna, The glycolytic enzyme triosephosphate isomerase of Trichomonas vaginalis is a surface-associated protein induced by glucose that functions as a laminin-and fibronectin-binding protein, Infec Immun, vol.84, pp.2878-2894, 2016.

L. A. Pereira, Analysis of the Paracoccidioides brasiliensis triosephosphate isomerase suggests the potential for adhesin function, FEMS Yeast Res, vol.7, pp.1381-1388, 2007.

R. Spooner and O. Yilmaz, Nucleoside-diphosphate-kinase: a pleiotropic effector in microbial colonization under interdisciplinary characterization, Microbes Infect, vol.14, pp.228-237, 2012.

J. H. Pedra, S. L. Cassel, and F. S. Sutterwala, Sensing pathogens and danger signals by the inflammasome, Curr Opin Immunol, vol.21, pp.10-16, 2009.

P. Pelegrin and A. Surprenant, Dynamics of macrophage polarization reveal new mechanism to inhibit IL-1beta release through pyrophosphates, EMBO J, vol.28, pp.2114-2127, 2009.

H. Sakaki, M. Tsukimoto, H. Harada, Y. Moriyama, and S. Kojima, Autocrine regulation of macrophage activation via exocytosis of ATP and activation of P2Y11 receptor, Plos One, vol.8, p.59778, 2013.

Y. Van-kooyk, C-type lectins on dendritic cells: key modulators for the induction of immune responses, Biochem Soc T, vol.36, pp.1478-1481, 2008.

J. Nigou, C. Zelle-rieser, M. Gilleron, M. Thurnher, and G. Puzo, Mannosylated lipoarabinomannans inhibit IL-12 production by human dendritic cells: evidence for a negative signal delivered through the mannose receptor, J Immunol, vol.166, pp.7477-7485, 2001.
URL : https://hal.archives-ouvertes.fr/hal-00177977

T. B. Geijtenbeek, Mycobacteria target DC-SIGN to suppress dendritic cell function, J Exp Med, vol.197, pp.7-17, 2003.

S. Tundup, L. Srivastava, and D. A. Harn, Polarization of host immune responses by helminth-expressed glycans, Ann NY Acad Sci, vol.1253, pp.1-13, 2012.

E. Van-liempt, Schistosoma mansoni soluble egg antigens are internalized by human dendritic cells through multiple C-type lectins and suppress TLR-induced dendritic cell activation, Mol Immunol, vol.44, pp.2605-2615, 2007.

R. Tyagi, Cracking the nodule worm code advances knowledge of parasite biology and biotechnology to tackle major diseases of livestock, Biotechnology Adv, vol.33, pp.980-991, 2015.

S. Gnerre, High-quality draft assemblies of mammalian genomes from massively parallel sequence data, Proc Natl Acad Sci, vol.108, pp.1513-1518, 2011.

T. M. Lowe and S. R. Eddy, tRNAscan-SE: a program for improved detection of transfer RNA genes in genomic sequence, Nucleic Acids Res, vol.25, pp.955-964, 1997.

A. F. Smit, R. Hubley, and P. Green, , 2010.

I. Korf, Gene finding in novel genomes, BMC Bioinformatics, vol.5, p.59, 2004.

A. A. Salamov and V. V. Solovyev, Ab initio gene finding in Drosophila genomic DNA, Genome Res, vol.10, pp.516-522, 2000.

M. Stanke, M. Diekhans, R. Baertsch, and D. Haussler, Using native and syntenically mapped cDNA alignments to improve de novo gene finding, Bioinformatics, vol.24, pp.637-644, 2008.

B. L. Cantarel, MAKER: an easy-to-use annotation pipeline designed for emerging model organism genomes, Genome Res, vol.18, pp.188-196, 2008.

Y. T. Tang, Genome of the human hookworm Necator americanus, Nat Genet, vol.46, pp.261-269, 2014.

L. Kall, A. Krogh, and E. L. Sonnhammer, A combined transmembrane topology and signal peptide prediction method, J Mol Biol, vol.338, pp.1027-1036, 2004.

L. Kall, A. Krogh, and E. L. Sonnhammer, Advantages of combined transmembrane topology and signal peptide prediction-the Phobius web server, Nucleic Acids Res, vol.35, pp.429-432, 2007.

J. D. Bendtsen, L. J. Jensen, N. Blom, G. Von-heijne, and S. Brunak, Feature-based prediction of non-classical and leaderless protein secretion, Protein Eng Des Sel, vol.17, pp.349-356, 2004.

T. Wylie, NemaPath: online exploration of KEGG-based metabolic pathways for nematodes, BMC Genomics, vol.9, p.525, 2008.

M. Kanehisa, S. Goto, Y. Sato, M. Furumichi, and M. Tanabe, KEGG for integration and interpretation of large-scale molecular data sets, Nucleic Acids Res, vol.40, pp.109-114, 2012.

M. Ashburner, Gene ontology: tool for the unification of biology. The Gene Ontology Consortium, Nat Genet, vol.25, pp.25-29, 2000.

S. Hunter, 2011: new developments in the family and domain prediction database, vol.40, pp.306-312, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00697960

E. Quevillon, InterProScan: protein domains identifier, Nucleic Acids Res, vol.33, pp.116-120, 2005.

K. Prufer, FUNC: a package for detecting significant associations between gene sets and ontological annotations, BMC Bioinformatics, vol.8, p.41, 2007.

S. Fischer, Using OrthoMCL to assign proteins to OrthoMCL-DB groups or to cluster proteomes into new ortholog groups, Current Protoc Bioinformatics, vol.6, pp.11-19, 2011.

D. A. Benson, Nucleic Acids Res, vol.43, pp.30-35, 2015.

B. A. Rosa, D. P. Jasmer, and M. Mitreva, Genome-wide tissue-specific gene expression, co-expression and regulation of co-expressed genes in adult nematode Ascaris suum, PLoS Negl Trop Dis, vol.8, p.2678, 2014.

S. N. Mcnulty, Systems biology studies of adult paragonimus lung flukes facilitate the identification of immunodominant parasite antigens, PLoS Negl Trop Dis, vol.8, p.3242, 2014.

D. Kim, TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions, Genome Biol, vol.14, p.36, 2013.

S. Anders, T. P. Pyl, and W. Huber, HTSeq -A Python framework to work with high-throughput sequencing data, Bioinformatics, vol.31, pp.166-169, 2014.

S. Anders and W. Huber, Differential expression analysis for sequence count data, Genome Biol, vol.11, p.106, 2010.

L. P. Leroux, Parasite manipulation of the invariant chain and the peptide editor H2-DM affects major histocompatibility complex class II antigen presentation during Toxoplasma gondii infection, Infect Immun, vol.83, pp.3865-3880, 2015.

N. E. Reiner, Methods in molecular biology. Macrophages and dendritic cells, Methods Mol Biol, vol.531, 2009.

B. J. Quah and C. R. Parish, The use of carboxyfluorescein diacetate succinimidyl ester (CFSE) to monitor lymphocyte proliferation, J Vis Exp, vol.44, p.2259, 2010.