S. A. Adamo, M. Jensen, and M. Younger, Changes in lifetime immunocompetence in male and female Gryllus texensis (formerly G. integer): trade-offs between immunity and reproduction, Animal Behaviour, vol.62, pp.417-425, 2001.

C. M. Adema, Fibrinogen-Related Proteins (FREPs) in Mollusks. Results and Problems in Cell Differentiation, vol.57, pp.111-129, 2015.

F. R. Adler and R. Karban, Defended fortresses or moving targets? Another model of inducible defenses inspired by military metaphors, American Naturalist, vol.144, pp.813-832, 1994.

A. A. Agrawal, C. Laforsch, and R. Tollrian, Transgenerational induction of defences in animals and plants, Nature, vol.401, pp.60-63, 1999.

S. Akira, S. Uematsu, and O. Takeuchi, Pathogen recognition and innate immunity, Cell, vol.124, pp.783-801, 2006.

K. Allander and P. Schmid-hempel, Immune defence reaction in bumble-bee workers after a previous challenge and parasitic coinfection, Functional Ecology, vol.14, pp.711-717, 2000.

R. M. Allen, Y. M. Buckley, and D. J. Marshall, Offspring size plasticity in response to intraspecific competition: an adaptive maternal effect across life-history stages, The American Naturalist, vol.171, pp.225-237, 2007.

J. André and S. Gandon, Vaccination, within-host dynamics, and virulence evolution, Evolution, vol.60, p.13, 2006.

C. R. Archer, F. Zajitschek, S. K. Sakaluk, N. J. Royle, and J. Hunt, Sexual selection affects the evolution of lifespan and ageing in the decorated cricket Gryllodes sigillatus, Evolution, vol.66, pp.3088-3100, 2012.

J. S. Ayres and D. S. Schneider, A signaling protease required for melanization in Drosophila affects resistance and tolerance of infections, PLoS Biology, vol.6, pp.2764-2773, 2008.

J. S. Ayres and D. S. Schneider, The role of anorexia in resistance and tolerance to infections in Drosophila, PLoS Biology, vol.7, p.1000150, 2009.

S. Asgari, MicroRNA functions in insects, Insect Biochemistry and Molecular Biology, vol.43, pp.388-397, 2013.

A. Ashe, P. Sarkies, J. Le-pen, M. Tanguy, and E. A. Miska, Antiviral RNA interference against Orsay virus is neither systemic nor transgenerational in Caenorhabditis elegans, Journal of Virology, vol.89, pp.12035-12046, 2015.

F. W. Avila, L. K. Sirot, B. A. Laflamme, C. D. Rubinstein, and M. F. Wolfner, Insect seminal fluid proteins: identification and function, Annual Review of Entomology, vol.56, pp.21-40, 2011.

A. P. Bascuñán-garcía, C. Lara, and A. Córdoba-aguilar, Immune investment impairs growth, female reproduction and survival in the house cricket, Acheta domesticus, Journal of Insect Physiology, vol.56, pp.204-211, 2010.

O. L. Baron, P. Van-west, B. Industri, M. Ponchet, G. Dubreuil et al.,

M. Coustau and C. , Parental transfer of the antimicrobial protein LBP/BPI protects Biomphalaria glabrata eggs against oomycete infections, PLoS Pathogens, vol.9, p.1003792, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00938929

A. I. Barnes and M. T. Siva-jothy, Density-dependent prophylaxis in the mealworm beetle Tenebrio molitor L. (Coleoptera: Tenebrionidae): cuticular melanization is an indicator of investment in immunity, Proceedings of the Royal Society of London Series B: Biological Sciences, vol.267, pp.177-182, 2000.

R. Barrangou and L. A. Marraffini, CRISPR-Cas systems: Prokaryotes upgrade to adaptive immunity, Molecular Cell, vol.54, pp.234-244, 2014.

S. M. Barribeau, P. Schmid-hempel, and B. M. Sadd, Royal decree: Gene expression in trans-generationally immune primed bumblebee workers mimics a primary immune response, PLoS One, vol.11, p.159635, 2016.

A. N. Becalska and E. R. Gavis, Lighting up mRNA localization in Drosophila oogenesis, Development, vol.136, pp.2493-2503, 2009.

W. A. Becker, Manual of Quantitative Genetics, 1984.

S. Behrens, R. Peuss, B. Milutinovic, H. Eggert, D. Esser et al., Infection routes matter in population-specific responses of the red flour beetle to the entomopathogen Bacillus thuringiensis, BMC Genomics, p.445, 2014.

G. Bell, The costs of reproduction and their consequences, American Naturalist, vol.116, pp.45-76, 1980.

T. G. Benton, S. J. Plaistow, A. P. Beckerman, C. T. Lapsley, and S. Littlejohns, Changes in maternal investment in eggs can affect population dynamics, Proceedings of the Royal Society of London B: Biological Sciences, vol.272, pp.1351-1356, 2005.

S. J. Berry, Maternal direction of oogenesis and early embryogenesis in insects, Annual Review of Entomology, vol.27, pp.205-227, 1982.

J. Bernardo, Maternal effects in animal ecology, American Zoologist, vol.36, pp.83-105, 1996.

B. Beutler, Innate immunity: an overview, Molecular immunology, vol.40, pp.845-859, 2004.

M. Biczkowski and F. Dittman, Translation of maternal mRNA during early oogenesis in a telotrophic-meroistic insect (Dysdercus intermedius), Journal of Insect Physiology, vol.41, pp.1133-1139, 1995.

H. G. Boman, I. Nilsson, and B. Rasmuson, Inducible antibacterial defence system in Drosophila, Nature, vol.237, pp.232-235, 1972.

M. Boots and K. E. Roberts, Maternal effects in disease resistance: poor maternal environment increases offspring resistance to an insect virus, Proceedings of the Royal Society B: Biological Sciences, vol.279, pp.4009-4014, 2012.

T. Boulinier and V. Staszewski, Maternal transfer of antibodies: raising immuno-ecology issues, Trends in Ecology & Evolution, vol.23, pp.282-288, 2008.

D. M. Bouts, A. C. Melo, A. L. Andrade, M. A. Silva-neto, O. Paiva-silva-gde et al., Biochemical properties of the major proteins from Rhodnius prolixus eggshell, Insect Biochemistry and Molecular Biology, vol.37, pp.1207-1221, 2007.

D. Brites and L. Du-pasquier, Somatic and germline diversification of a putative immunoreceptor within one phylum: Dscam in Arthropods. Results and Problems in Cell Differentiation, vol.57, pp.131-158, 2015.

J. Broggi, R. Soriguer, and J. Figuerola, Transgenerational effects enhance specific immune response in a wild passerine, PeerJ, vol.4, p.1766, 2016.

P. Bulet and R. Stöcklin, Insect antimicrobial peptides: Structures, properties and gene regulation, Protein and Peptide Letters, vol.12, pp.3-11, 2005.

D. G. Capco and W. R. Jeffery, Origin and spatial distribution of maternal messenger RNA during oogenesis of an insect, Oncopeltus fasciatus, Journal of Cell Science, vol.39, pp.63-76, 1979.

J. Capinera, P. Barbosa, and H. Hagedorn, Yolk and yolk depletion of Gypsy moth eggs: Implications for population quality 1, 2. Annals of the, vol.70, pp.40-42, 1977.

C. Castro-vargas, C. Linares-lópez, A. López-torres, K. Wrobel, J. C. Torres-guzmán et al., , 2017.

, Methylation on RNA: A potential mechanism related to immune priming within but not across generations, Frontiers in Microbiology, vol.8, p.473

L. Cerenius and K. Söderhäll, The prophenoloxidase-activating system in invertebrates, Immunological Reviews, vol.198, pp.116-126, 2004.

M. Charlet, M. Lagueux, J. M. Reichhart, D. Hoffman, A. Braun et al., Cloning of the gene encoding the antibacterial peptide drosocin involved in Drosophila immunity: expression studies during the immune response, European Journal of Biochemistry, vol.241, pp.699-706, 1996.

M. Chase, K. Raina, J. Bruno, and M. Sugumaran, Purification, characterization and molecular cloning of prophenoloxidase from Sarcophaga bullata, Insect Biochemestry and Molecular Biology, vol.30, pp.953-967, 2000.

P. S. Chen, E. Stumm-zollinger, T. Aigaki, J. Balmer, M. Blenz et al., A male accessory gland peptide that regulates reproductive behavior of female D. melanogaster, Cell, vol.54, pp.291-296, 1988.

M. M. Clark and B. G. Galef, Prenatal influences on reproductive life history strategies, Trends in Ecology & Evolution, vol.10, pp.151-153, 1995.

V. Corby-harris, K. E. Habel, F. G. Ali, and D. E. Promislow, Alternative measures of response to Pseudomonas aeruginosa infection in Drosophila melanogaster, Journal of Evolutionary Biology, vol.20, pp.526-533, 2007.

C. Coustau, J. Kurtz, and Y. Moret, A novel mechanism of immune memory unveiled at the invertebrate-parasite interface, Trends in Parasitology, vol.32, pp.353-355, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01315801

G. Cumming and S. Finch, Inference by eye: confidence intervals and how to read pictures of data, American Psychologist, vol.60, p.170, 2005.

J. R. David, P. Gibert, H. Legout, G. Pétavy, P. Capy et al., Isofemale lines in Drosophila: an empirical approach to quantitative trait analysis in natural populations, Heredity, vol.94, pp.3-12, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00427726

M. Dettloff, D. Wittwer, C. Weise, and A. Wiesner, Lipophorin of lower density is formed during immune responses in the lepidopteran insect Galleria mellonella, Cell Tissue Research, vol.306, pp.449-458, 2001.

J. Dhinaut, M. Chogne, and Y. Moret, Immune priming specificity within and across generations reveals the range of pathogens affecting evolution of immunity in an insect, Journal of Animal Ecology, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01728826

J. Dhinaut, M. Chogne, and Y. Moret, Immune priming specificity within and across generations reveals the range of pathogens affecting evolution of immunity in an insect, Dryad Digital Repository, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01728826

J. Dhinaut, M. Chogne, and Y. Moret, Trans-generational immune priming in the mealworm beetle protects eggs through pathogen-dependent mechanisms imposing no immediate fitness cost for the offspring
URL : https://hal.archives-ouvertes.fr/hal-01631046

E. V. Domanitskaya, H. Liu, S. Chen, and E. Kubli, The hydroxyproline motif of male sex peptide elicits the innate immune response in Drosophila females, The FEBS Journal, vol.274, pp.5659-5668, 2007.

Y. Dong, H. E. Taylor, and G. Dimopoulos, AgDscam, a hypervariable immunoglobulin domain-containing receptor of the Anopheles gambiae innate immune system, PLoS Biology, vol.4, p.229, 2006.

R. N. Du and M. Laing, Determination of insecticidal toxicity of three species of entomopathogenic spore-forming bacterial isolates against Tenebrio molitor L. (Coleoptera: Tenebrionidae), African Journal of Microbiology Research, vol.5, pp.2222-2228, 2011.

S. Dupas, S. Morand, and P. Eslin, Evolution of hemocyte concentration in the melanogaster subgroup species, Comptes Tendus Biologies, vol.327, pp.139-147, 2004.

L. Du-pasquier, Germline and somatic diversification of immune recognition elements in Metazoa, Immunology Letters, vol.104, pp.2-17, 2006.

A. Dubuffet, C. Zanchi, G. Boutet, J. Moreau, M. Teixeira et al., Transgenerational immune priming protects the eggs only against gram-positive bacteria in the mealworm beetle, PLoS Pathogens, p.11, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01214091

H. Eggert, . Diddens-de, M. F. Buhr, and J. Kurtz, A temperature shock can lead to transgenerational immune priming in the Red Flour Beetle, Tribolium castaneum, Ecology and Evolution, vol.5, pp.1318-1326, 2015.

H. Eggert, J. Kurtz, and M. F. Diddens-de-buhr, Different effects of paternal transgenerational immune priming on survival and immunity in step and genetic offspring, Proceedings of the Royal Society of London Series B: Biological Sciences, vol.281, 2014.

J. D. Ericsson, A. F. Janmaat, C. Lowenberger, and J. H. Myers, Is decreased generalized immunity a cost of Bt resistance in cabbage loopers Trichoplusia ni, Journal of Invertebrate Pathology, vol.100, pp.61-67, 2009.

E. Esteves, A. C. Fogaca, R. Maldonado, F. D. Silva, P. P. Manso et al.,

D. Valle and S. Daffre, Antimicrobial activity in the tick Rhipicephalus (Boophilus) microplus eggs: Cellular localization and temporal expression of microplusin during oogenesis and embryogenesis, Developmental & Comparative Immunology, vol.33, pp.913-919, 2009.

P. Fehlbaum, P. Bulet, L. Michaut, M. Lagueux, W. F. Broekaert et al., Insect immunity. Septic injury of Drosophila induces the synthesis of a potent antifungal peptide with sequence homology to plant antifungal peptides, Journal of Biological Chemistry, vol.269, pp.33159-33163, 1994.

M. A. Felix, A. Ashe, J. Piffaretti, G. Wu, I. Nuez et al., Natural and experimental infection of Caenorhabditis nematodes by novel viruses related to nodaviruses, PLoS Biology, vol.9, p.1000586, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00566941

A. Fenton, J. Lello, and M. B. Bonsall, Pathogen responses to host immunity: the impact of time delays and memory on the evolution of virulence, Proceedings of the Royal Society B: Biological Sciences, vol.273, pp.2083-2090, 2006.

M. Fischer, C. Regitz, M. Kahl, M. Werthebach, M. Boll et al., , 2012.

, Phytoestrogens genistein and daidzein affect immunity in the nematode Caenorhabditis elegans via alterations of vitellogenin expression, Molecular Nutrition & Food Research, vol.56, pp.957-965

J. J. Fisher and A. E. Hajek, Maternal exposure of a beetle to pathogens protects offspring against fungal disease, PLoS One, vol.10, p.125197, 2015.

I. Folstad and A. J. Karter, Parasites, bright males, and the immunocompetence handicap, The American Naturalist, vol.139, pp.603-622, 1992.

C. W. Fox, M. S. Thakar, and T. A. Mousseau, Egg size plasticity in a seed beetle: an adaptive maternal effect, The American Naturalist, vol.149, pp.149-163, 1997.

S. A. Frank, Immunology and evolution of infectious disease, 2002.

S. Fraune, R. Augustin, F. Anton-erxleben, J. Wittlieb, C. Gelhaus et al., In an early branching metazoan, bacterial colonization of the embryo is controlled by maternal antimicrobial peptides, vol.107, pp.18067-18072, 2010.

D. Freitak, D. G. Heckel, and H. Vogel, Dietary-dependent trans-generational immune priming in an insect herbivore, Proceedings of the Royal Society B: Biological Sciences, vol.276, p.2617, 2009.

D. Freitak, H. Schmidtberg, F. Dickel, G. Lochnit, H. Vogel et al., The maternal transfer of bacteria can mediate trans-generational immune priming in insects, 2014.

, Virulence, vol.5, pp.547-554

D. A. Galbraith, X. Yang, E. L. Nino, S. Yi, and C. Grozinger, Parallel epigenomic and transcriptomic responses to viral infection in honey bees (Apis mellifera), PLoS Pathogens, vol.11, p.1004713, 2015.

T. L. Geister, M. X. Lorenz, K. H. Hoffmann, and K. Fisher, Adult nutrition and butterfly fitness: effects of diet quality on reproductive output, egg composition, and egg hatching success, Frontiers in Zoology, vol.5, p.10, 2008.

M. Gibbs, C. J. Breuker, and H. Van-dyck, Flight during oviposition reduces maternal egg provisioning and influences offspring development in Pararge aegeria (L.), Physiological Entomology, vol.35, pp.29-39, 2010.

V. V. Glupov, M. F. Khvoshevskaya, Y. L. Lozinskaya, I. M. Dubovski, and V. Martemyanov,

V. Sokolova and J. Y. , Application of the nitroblue tetrazolium-reduction method for studies on the production of reactive oxygen species in insect haemocytes, Cytobios, vol.106, issue.2, pp.165-178, 2001.

M. J. Gorman, P. Kankanala, and M. R. Kanost, Bacterial challenge stimulates innate immune responses in extra-embryonic tissues of tobacco hornworm eggs, Insect Molecular Biology, vol.13, pp.19-24, 2004.

M. J. Gorman and S. M. Paskewitz, Serine proteases as mediators of mosquito immunity, Insect Biochemestry and Molecular Biology, vol.31, pp.257-262, 2001.

D. A. Gray, Sex differences in susceptibility of house crickets, Acheta domesticus, to experimental infection with Serratia liquefaciens, Journal of Invertebrate Pathology, vol.71, pp.288-289, 1998.

T. J. Green, K. Helbig, P. Speck, and D. A. Raftos, Primed for success: Oyster parents treated with poly(I:C) produce offspring with enhanced protection against Ostreid herpesvirus type I infection, Molecular Immunology, vol.78, pp.113-120, 2016.

J. L. Grindstaff, I. Brodie, E. D. Ketterson, and E. D. , Immune function across generations: integrating mechanism and evolutionary process in maternal antibody transmission, Proceedings of the Royal Society of London B: Biological Sciences, vol.270, pp.2309-2319, 2003.

J. L. Grindstaff, Initial levels of maternally derived antibodies predict persistence time in offspring circulation, Journal of Ornithology, vol.151, pp.423-428, 2010.

E. R. Haine, Y. Moret, M. T. Siva-jothy, and J. Rolff, Antimicrobial defense and persistent infection in insects, Science, vol.322, pp.1257-1259, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00343225

E. R. Haine, L. C. Pollitt, Y. Moret, M. T. Siva-jothy, and J. Rolff, Temporal patterns in immune responses to a range of microbial insults (Tenebrio molitor), Journal of Insect Physiology, vol.54, pp.1090-1097, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00293378

K. Hammerschmidt, P. Deines, A. J. Wilson, and J. Rolff, Quantitative genetics of immunity and life history under different photoperiods, Heredity, vol.108, pp.569-576, 2012.

S. Hangartner, S. H. Sbilordo, ?. Michalczyk, M. J. Gage, and O. Y. Martin, Are there genetic trade-offs between immune and reproductive investments in Tribolium castaneum? Infection, Genetics and Evolution, vol.19, pp.45-50, 2013.

P. C. Hanington and S. M. Zhang, The primary role of fibrinogen-related proteins in invertebrates is defense, not coagulation, Journal of Innate Immunity, vol.3, pp.17-27, 2011.

Z. G. Hao, I. Kasumba, and S. Aksoy, Proventriculus (cardia) plays a crucial role in immunity in tsetse fly (Diptera: Glossinidiae), Insect Biochemestry and Molecular Biology, vol.33, pp.1155-1164, 2003.

K. C. Harding, B. J. Hansen, and S. J. Goodman, Acquired immunity and stochasticity inepidemic intervals impede the evolution of host disease resistance, The American Naturalist, vol.166, pp.722-730, 2005.

J. F. Harris, S. Micheva-viteva, N. Li, and E. Hong-geller, Small RNA-mediated regulation of host-pathogen interactions, Virulence, vol.4, pp.785-795, 2013.

C. D. Harvell, The ecology and evolution of inducible defenses, Quaterly Review of Biology, vol.65, pp.323-340, 1990.

D. Hasselquist and J. Nilsson, Maternal transfer of antibodies in vertebrates: transgenerational effects on offspring immunity, Philosophical Transactions of the Royal Society BBiological Sciences, vol.364, pp.51-60, 2009.

H. Havukainen, D. Munch, A. Baumann, S. Zhong, O. Halskau et al., Vitellogenin recognizes cell damage through membrane binding and shields living cells from reactive oxygen species, The Journal of Biological Chemistry, vol.288, pp.28369-28381, 2013.

J. Hernández-lópez, W. Schuehly, K. Crailsheim, and U. Riessberger-gallé, Transgenerational immune priming in honeybees, Proceedings of the Royal Society B: Biological Sciences, vol.281, 2014.

D. H. Ho and W. W. Burggren, Epigenetics and transgenerational transfer: A physiological perspective, The Journal of Experimental Biology, vol.213, pp.3-16, 2010.

A. A. Hoffmann and P. A. Parsons, The analysis of quantitative variation in natural populations with isofemale strains, Génétique, Sélection, Evolution, vol.20, pp.87-98, 1988.
URL : https://hal.archives-ouvertes.fr/hal-00893747

J. A. Hoffman, The immune response of Drosophila, Nature, vol.426, pp.33-38, 2003.

B. Huang, L. Zhang, L. Li, X. Tang, and G. Zhang, Highly diverse fibrinogen-related proteins in the Pacific oyster Crassostrea gigas, Fish & Shellfish Immunology, vol.43, pp.485-490, 2015.

C. C. Huang and Y. L. Song, Maternal transmission of immunity to white spot syndrome associated virus (WSSV) in shrimp (Penaeus monodon), Developmental & Comparative Immunology, vol.23, pp.545-552, 1999.

D. Hultmark, Exs, vol.75, pp.87-102, 1995.

H. B. Huttenhuis, C. P. Grou, A. J. Taverne-thiele, N. Taverne, and J. H. Rombout, , 2006.

. Carp, Cyprinus carpio L.) innate immune factors are present before hatching, Fish & Shellfish Immunology, vol.20, pp.586-596

T. Ichikawa and T. Kurauchi, Larval cannibalism and pupal defense against cannibalism in two species of Tenebrionid beetles, Zoological Science, vol.26, pp.525-529, 2009.

C. Jacobs, Y. Wang, H. Vogel, A. Vilcinskas, M. Van-der-zee et al., Egg survival is reduced by grave-soil microbes in the carrion beetle, Nicrophorus vespilloides, BMC Evolutionary Biology, vol.14, p.208, 2014.

C. G. Jacobs, J. D. Gallagher, S. E. Evison, D. G. Heckel, A. Vilcinskas et al., Endogenous egg immune defenses in the yellow mealworm beetle, 2017.

, Developmental & Comparative Immunology, vol.70, pp.1-8

C. G. Jacobs, H. P. Spaink, and M. Van-der-zee, The extraembryonic serosa is a frontier epithelium providing the insect egg with a full-range innate immune response, Elife, vol.3, p.4111, 2014.

C. G. Jacobs and M. Van-der-zee, Immune competence in insect eggs depends on the extraembryonic serosa, Developmental & Comparative Immunology, vol.41, pp.263-269, 2013.

C. A. Janeway, . Jr, and R. Medzhitov, Innate immune recognition, Annual Review of Immunology, vol.20, pp.197-216, 2002.

O. Johnstone and P. Lasko, Translational regulation and RNA localization in Drosophila oocytes and embryos, Annual Review of Genetics, vol.35, pp.365-406, 2001.

J. Jokela, Transgenerational immune priming as cryptic parental care, Journal of Animal Ecology, vol.79, pp.305-307, 2010.

J. Jung, A. Heo, Y. W. Park, Y. J. Kim, H. Koh et al., Gut microbiota of Tenebrio molitor and their response ton environmental change, Journal of Microbiology and Biotechnology, vol.24, pp.888-897, 2014.

J. L. Jurat-fuentes and T. Jackson, Bacterial entomopathogens. Insect Pathology, pp.265-349, 2012.

G. P. Kaaya, Inducible humoral antibacterial immunity in insects, Insect immunity, pp.69-89, 1993.

J. E. Kinsella, Metabolic patterns of the fatty acids of Periplanteta americana (L.) during its embryonic development, Canadian Journal of Biochemistry, vol.44, pp.247-258, 1966.

M. Kirkpatrick and R. Lande, The evolution of maternal characters, Evolution, vol.43, pp.485-503, 1989.

E. Knorr, H. Schmidtberg, D. Arslan, L. Bingsohn, and A. Vilcinskas, Translocation of bacteria from the gut to the eggs triggers maternal transgenerational immune priming in Tribolium castaneum, Biology Letters, p.11, 2015.

J. C. Koella and C. Boëte, A genetic correlation between age at pupation and melanization immune response of the yellow fever mosquito Aedes aegypti, Evolution, vol.56, pp.1074-1079, 2002.

I. Krams, J. Daukste, T. Krama, and M. J. Rantala, Previous encapsulation response enhances within individual protection against fungal parasite in the mealworm beetle Tenebrio molitor, Insect Science, vol.20, pp.771-777, 2013.

J. Kurtz, Specific memory within innate immune systems, Trends in Immunology, vol.26, pp.186-192, 2005.

K. Kvell, E. L. Cooper, P. Engelmann, J. Bovari, and P. Nemeth, Blurring borders: innate immunity with adaptive features, Clinical & Developmental Immunology, p.83671, 2007.

A. M. Lackie, G. Takle, and L. Tetley, Haemocytic encapsulation in the locust Schistocerca gregaria (Orthoptera) and the cockroach Periplaneta americana, Cell and Tissue Research, vol.240, pp.343-351, 1985.

J. D. Lambeth, Nox enzymes, ROS and chronic disease: an example of antagonistic pleiotropy, Free Radical Biology and Medicine, vol.43, pp.332-347, 2007.

J. D. Lambeth, T. Kawahara, and B. Diebold, Regulation of Nox and Dox enzymatic activity and expression, Free Radical Biology and Medicine, vol.43, pp.319-331, 2007.

M. D. Lavine, G. Chen, and M. R. Strand, Immune challenge differentially affects transcript abundance of three antimicrobial peptides in hemocytes from the moth Pseudoplusia includens, Insect Biochemistry and Molecular Biology, vol.35, pp.1335-1346, 2005.

B. P. Lazzaro, Natural selection on the Drosophila antimicrobial immune system, Current Opinion in Microbiology, vol.11, pp.284-289, 2008.

B. Lemaître, J. M. Reichart, and J. A. Hoffmann, Drosophila host defense: differential induction of antimicrobial peptide genes after infection by various classes of microorganisms, Proceedings of the National Academy of Sciences of USA, vol.94, pp.14614-14619, 1997.

Z. Li, S. Zhang, and Q. Liu, Vitellogenin functions as a multivalent pattern recognition receptor with an opsonic activity, PLoS One, vol.3, p.1940, 2008.

J. E. Linder and D. E. Promislow, Cross-generational fitness effects of infection in Drosophila melanogaster, Fly, vol.3, pp.143-150, 2009.

J. Lindström, Early development and fitness in birds and mammals, Trends in Ecology & Evolution, vol.14, pp.343-348, 1999.

T. J. Little, D. Hultmark, and A. F. Read, Invertebrate immunity and the limits of mechanistic immunology, Nature Immunology, vol.6, pp.651-654, 2005.

T. J. Little and A. R. Kraaijeveld, Ecological and evolutionary implications of immunological priming in invertebrates, Trends in Ecology & Evolution, vol.19, pp.58-60, 2004.

T. J. Little, B. O'connor, N. Colegrave, K. Watt, and A. F. Read, Maternal transfer of strain-specific immunity in an invertebrate, Current Biology, vol.13, pp.489-492, 2003.

J. E. Littlefair, A. M. Laughton, and R. J. Knell, Maternal pathogen exposure causes dietand pathogen-specific transgenerational costs, Oikos, 2016.
DOI : 10.1111/oik.03430

H. Liu and E. Kubli, Sex-peptide is the molecular basis of the sperm effect in Drosophila melanogaster, Proceedings of the National Academy of Sciences of the United States of America, vol.100, pp.9929-9933, 2003.

L. M. Lorenz and J. C. Koella, Maternal environment shapes the life history and susceptibility to malaria of Anopheles gambiae mosquitoes, Malaria Journal, vol.10, p.382, 2011.

S. Luckhart, Y. Vodovotz, L. Cui, and R. Rosenberg, The mosquito Anopheles stephensi limits malaria parasite development with inducible synthesis of nitric oxide, Proceedings of the National Academy of Sciences, vol.95, pp.5700-5705, 1998.

M. J. Mackinnon and A. F. Read, Immunity promotes virulence evolution in a malaria model, PLoS Biology, vol.2, p.230, 2004.
DOI : 10.1371/journal.pbio.0020230

URL : https://journals.plos.org/plosbiology/article/file?id=10.1371/journal.pbio.0020230&type=printable

D. R. Maddison and K. S. Schulz, The Tree of Life Web Project, 2007.

O. Makarova, A. Rodriguez-rojas, M. Eravci, C. Weise, A. Dobson et al., Antimicrobial defence and persistent infection in insects revisited, Philosophical Transactions of the Royal Society B: Biological Sciences, vol.371, 2016.
DOI : 10.1098/rstb.2015.0296

URL : http://europepmc.org/articles/pmc4874393?pdf=render

L. J. Manseau and T. Schüpbach, Cappuccino and spire: two unique maternal-effect loci required for both the anteroposterior and the dorsoventral patterns of the Drosophila embryo, Genes and Development, vol.3, pp.1437-1452, 1989.

D. Marchini, A. G. Manetti, M. Rosetto, L. F. Bernini, J. L. Telford et al., cDNA sequence and expression of the ceratotoxin gene encoding an antibacterial sex-specific peptide from the medfly Ceratitis-capitata (Diptera), Journal of Biological Chemistry, vol.270, pp.6199-6204, 1995.

D. Marchini, L. Marri, M. Rosetto, A. G. Manetti, and R. Dallai, Presence of antibacterial peptides on the laid egg horion of the medfly Ceratitis capitata, Biochemical and Biophysical Research Communications, vol.240, pp.657-663, 1997.

D. Marshall and T. Uller, When is a maternal effect adaptive? Oikos, vol.116, pp.1957-1963, 2007.

N. E. Martins, V. G. Faria, L. Teixeira, S. Magalhaes, and E. Sucena, Host adaptation is contingent upon the infection route taken by pathogens, PLoS Pathogens, vol.9, p.1003601, 2013.

L. Masri, A. Branca, A. E. Sheppard, A. Papkou, D. Laehnemann et al.,

M. Saebelfeld, J. Hollensteiner, H. Liesegang, E. Brzuszkiewicz, R. Daniel et al.,

R. D. Schulte, J. Kurtz, P. Rosenstiel, A. Telschow, E. Bornberg-bauer et al., Host-pathogen coevolution: The selective qdvantage of Bacillus thuringiensis virulence and its cry toxin genes, PLoS Biology, p.1002169, 2015.

L. Masri and S. Cremer, Individual and social immunisation in insects, Trends in Immunology, vol.35, pp.471-482, 2014.

P. Matzinger, Tolerance, danger and the extended family, Annual Review of Immunology, vol.12, pp.991-1045, 1994.

K. A. Mckean and B. P. Lazzaro, The costs of immunity and the evolution of immunological defense mechanisms, 2011.

K. B. Mcnamara, E. Van-lieshout, and L. W. Simmons, The effect of maternal and paternal immune challenge on offspring immunity and reproduction in a cricket, Journal of Evolutionary Biology, vol.27, pp.1020-1028, 2014.

B. Milutinovi? and J. Kurtz, Immune memory in invertebrates, Seminars in Immunology, vol.28, pp.328-342, 2016.

S. E. Mitchell and A. F. Read, Poor maternal environment enhances offspring disease resistance in an invertebrate, Proceedings of the Royal Society of London Series B: Biological Sciences, vol.272, pp.2601-2607, 2005.

S. L. Moore and K. Wilson, Parasites as a viability cost of sexual selection in natural populations of mammals, Science, vol.297, pp.2015-2018, 2002.

J. Moreau, G. Martinaud, J. P. Troussard, C. Zanchi, and Y. Moret, Trans-generational immune priming is constrained by the maternal immune response in an insect, Oikos, vol.121, pp.1828-1832, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00756764

M. Moreno-garcia, R. Conde, R. Bello-bedoy, and H. Lanz-mendoza, The damage threshold hypothesis and the immune strategies of insects, Infection, Genetics and Evolution, vol.24, pp.25-33, 2014.

Y. Moret, Explaining variable costs of the immune response: selection for specific versus non-specific immunity and facultative life history change, Oikos, vol.102, pp.213-215, 2003.

Y. Moret, Trans-generational immune priming: specific enhancement of the antimicrobial immune response in the mealworm beetle, Tenebrio molitor, Proceedings of the Royal Society B: Biological Sciences, vol.273, pp.1399-1405, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00258228

Y. Moret and P. Schmid-hempel, Survival for immunity: The price of immune system activation for bumblebee workers, Science, vol.290, pp.1166-1168, 2000.

Y. Moret and M. T. Siva-jothy, Adaptive innate immunity? Responsive-mode prophylaxis in the mealworm beetle, Tenebrio molitor, Proceedings of the Royal Society of London Series B: Biological Sciences, vol.270, pp.2475-2480, 2003.

T. A. Mousseau and C. W. Fox, Maternal effects as adaptations, 1998.

J. Moya-laraño, Senescence and food limitation in a slowly ageing spider, Functional Ecology, vol.16, pp.734-741, 2002.

K. Mukherjee, R. Fischer, and A. Vilcinskas, Histone acetylation mediates epigenetic regulation of transcriptional reprogramming in insects during metamorphosis, wounding and infection, Frontiers in Zoology, vol.9, p.25, 2012.

K. Murphy and C. Weaver, Janeway's Immunobiology. Garland Science, 2016.

C. F. Nathan and J. B. Hibbs, Role of nitric oxide synthesis in macrophage antimicrobiol activity, Current Opinion in Immunology, vol.3, pp.65-70, 1991.

M. G. Netea, L. A. Joosten, E. Latz, K. H. Mills, G. Natoli et al.,

A. Xavier and R. J. , Trained immunity: A program of innate immune memory in health and disease, Science, vol.352, 1098.

M. G. Netea, J. Quintin, and J. W. Van-der-meer, Trained immunity: a memory for innate host defense, Cell Host & Microbe, vol.9, pp.355-361, 2011.

P. Norouzitallab, K. Baruah, P. Biswas, D. Vanrompay, and P. Bossier, Probing the phenomenon of trained immunity in invertebrates during a transgenerational study, using brine shrimp Artemia as a model system, Scientific Reports, vol.6, p.21166, 2016.

P. Norouzitallab, P. Biswas, K. Baruah, and P. Bossier, Multigenerational immune priming in an invertebrate parthenogenetic Artemia to a pathogenic Vibrio campbellii, Fish & Shellfish Immunology, vol.42, pp.426-429, 2015.

B. P. Oldroyd, M. H. Allsopp, K. M. Roth, E. J. Remnant, R. A. Drewell et al.,

, A parent-of-origin effect on honeybee worker ovary size, Proceedings of the Royal Society of London B: Biological Sciences, vol.281, 20132388.

Z. Pancer and M. D. Cooper, The evolution of adaptive immunity, Annual Review of Immunology, vol.24, pp.497-518, 2006.

R. Pant, S. Kumar, and S. D. Singh, Changes in carbohydrates and lipids during embryonic development of Antheraea mylitta (Lepidoptera), Journal of Biosciences, vol.1, pp.27-33, 1979.

J. Peng, P. Zipperlen, and E. Kubli, Drosophila sex-peptide stimulates female innate immune system after mating via the Toll and Imd pathways, Current Biology, vol.15, pp.1690-1694, 2005.

L. N. Pham, M. S. Dionne, M. Shirasu-hiza, and D. S. Schneider, A specific primed immune response in Drosophila is dependent on phagocytes, PLoS Pathogens, vol.3, p.26, 2007.

L. N. Pham and D. S. Schneider, Evidence for specificity and memory in the insect innate immune response, Insect Immunology, pp.97-127, 2008.

R. Pigeault, R. Garnier, A. Rivero, and S. Gandon, Evolution of transgenerational immunity in invertebrates, Proceedings of the Royal Society of London Series B: Biological Sciences, vol.283, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01942644

R. Pigeault, J. Vezilier, A. Nicot, S. Gandon, and A. Rivero, Transgenerational effect of infection in Plasmodium-infected mosquitoes, Biology Letters, vol.11, 2015.

S. Pinaud, J. Portela, D. Duval, F. C. Nowacki, M. A. Olive et al., A Shift from cellular to humoral responses contributes to innate immune memory in the vector snail Biomphalaria glabrata, PLoS Pathogens, vol.12, p.1005361, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02191477

S. J. Plaistow, . St, J. J. Clair, J. Grant, and T. G. Benton, How to put all your eggs in one basket: empirical patterns of offspring provisioning throughout a mother's lifetime, The American Naturalist, vol.170, pp.520-529, 2007.

R. Poulin, Sexual inequalities in helminth infections: a cost of being a male?, The American Naturalist, vol.147, pp.287-295, 1996.

T. Pradeu and E. L. Cooper, The danger theory: 20 years later, Frontiers in Immunology, vol.3, p.287, 2012.

T. Pradeu, S. Jaeger, and E. Vivier, The speed of change: towards a discontinuity theory of immunity?, Nature Reviews Immunology, vol.13, pp.764-769, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01343694

E. R. Pursall and J. Rolff, Immune responses accelerate ageing: proof-of-principle in an insect model, PLoS One, vol.6, p.19972, 2011.

A. Qvarnström and T. D. Price, Maternal effects, paternal effects and sexual selection, Trends in Ecology & Evolution, vol.16, pp.95-100, 2001.

L. Råberg, A. L. Graham, and A. F. Read, Decomposing health: tolerance and resistance to parasites in animals, Philosophical Transactions of the Royal Society B-Biological Sciences, vol.364, pp.37-49, 2009.

L. Råberg, M. Vestberg, D. Hasselquist, R. Holmdahl, E. Svensson et al., , 2002.

, Basal metabolic rate and the evolution of the adaptive immune system, Proceedings of the Royal Society B: Biological Sciences, vol.269, pp.817-821

M. M. Rahman, H. L. Roberts, M. Sarjan, S. Asgari, and O. Schmidt, Induction and transmission of Bacillus thuringiensis tolerance in the flour moth Ephestia kuehniella, Proceedings of the, vol.101, pp.2696-2699, 2004.

A. S. Raikhel and T. S. Dhadialla, Accumulation of yolk proteins in insect oocytes, Annual Review of Entomology, vol.37, pp.217-251, 1992.

A. S. Raikhel, V. A. Kokoza, J. S. Zhu, D. Martin, S. F. Wang et al., Molecular biology of mosquito vitellogenesis: from basic studies to genetic engineering of antipathogen immunity, Insect Biochemistry and Molecular Biology, vol.32, pp.1275-1286, 2002.

A. Rivero, Nitric oxide: an antiparasitic molecule of invertebrates, Trends in Parasitology, vol.22, pp.219-225, 2006.
DOI : 10.1016/j.pt.2006.02.014

J. M. Robinson and J. A. Badwey, Production of active oxygen species by phagocytic leukocytes, Immunology, vol.60, pp.159-178, 1994.
DOI : 10.1007/s00418-008-0461-4

URL : https://link.springer.com/content/pdf/10.1007%2Fs00418-008-0461-4.pdf

J. Rodrigues, F. A. Brayner, L. C. Alves, R. Dixit, and C. Barillas-mury, Hemocyte differentiation mediates innate immune memory in Anopheles gambiae mosquitoes, Science, vol.329, pp.1353-1355, 2010.
DOI : 10.1126/science.1190689

URL : http://europepmc.org/articles/pmc3510677?pdf=render

D. A. Roff, The evolution of life histories, 1992.

J. Rolff, S. A. Armitage, and D. W. Coltman, Genetic constraints and sexual dimorphism in immune defense, Evolution, vol.59, pp.1844-1850, 2005.
DOI : 10.1554/04-747.1

J. Rolff, Bateman's principle and immunity, Proceedings of the Royal Society of London Series B: Biological Sciences, vol.269, pp.867-872, 2002.
DOI : 10.1098/rspb.2002.1959

URL : http://europepmc.org/articles/pmc1690964?pdf=render

R. B. Rosengaus, N. Hays, C. Biro, J. Kemos, M. Zaman et al., Pathogen-induced maternal effects result in enhanced immune responsiveness across generations, Ecology and Evolution, vol.7, pp.2925-2935, 2017.
DOI : 10.1002/ece3.2887

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1002/ece3.2887

M. C. Rossiter, Maternal effects generate variation in life history: consequences of egg weight plasticity in the Gypsy Moth, Functional Ecology, vol.5, pp.386-393, 1991.

O. Roth, G. Joop, H. Eggert, J. Hilbert, J. Daniel et al., Paternally derived immune priming for offspring in the red flour beetle, Tribolium castaneum, Journal of Animal Ecology, vol.79, pp.403-413, 2010.

O. Roth and J. Kurtz, Phagocytosis mediates specificity in the immune defence of an invertebrate, the woodlouse Porcellio scaber (Crustacea: Isopoda), Developmental & Comparative Immunology, vol.33, pp.1151-1155, 2009.

O. Roth, B. M. Sadd, P. Schmid-hempel, and J. Kurtz, Strain-specific priming of resistance in the red flour beetle, Tribolium castaneum, Proceedings of the Royal Society B: Biological Sciences, vol.276, pp.145-151, 2009.

B. M. Sadd, Y. Kleinlogel, R. Schmid-hempel, and P. Schmid-hempel, Transgenerational immune priming in a social insect, Biology Letters, vol.1, pp.386-388, 2005.
DOI : 10.1098/rsbl.2005.0369

URL : http://europepmc.org/articles/pmc1626361?pdf=render

B. M. Sadd and P. Schmid-hempel, Insect immunity shows specificity in protection upon secondary pathogen exposure, Current Biology, vol.16, pp.1206-1210, 2006.
DOI : 10.1016/j.cub.2006.04.047

URL : https://doi.org/10.1016/j.cub.2006.04.047

B. M. Sadd and P. Schmid-hempel, Facultative but persistent trans-generational immunity via the mother's eggs in bumblebees, Current Biology, vol.17, pp.1046-1047, 2007.
DOI : 10.1016/j.cub.2007.11.007

URL : https://doi.org/10.1016/j.cub.2007.11.007

B. M. Sadd and P. Schmid-hempel, A distinct infection cost associated with transgenerational priming of antibacterial immunity in bumble-bees, Biology Letters, vol.5, pp.798-801, 2009.

B. M. Sadd and P. Schmid-hempel, Ecological and evolutionary implications of specific immune responses, Insect Infection and Immunity: Evolution, Ecology, and Mechanisms, 2009.
DOI : 10.1093/acprof:oso/9780199551354.003.0014

J. Rolff and S. Reynolds, , p.272

S. Saeed, J. Quintin, H. H. Kerstens, N. A. Rao, A. Aghajanirefah et al.,

G. , Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity, Science, vol.345, p.1251086, 2014.

H. Salmela, G. V. Amdam, and D. Freitak, Transfer of immunity from mother to offspring is mediated via egg-yolk protein vitellogenin, PLoS Pathogens, vol.11, p.1005015, 2015.
DOI : 10.1371/journal.ppat.1005015

URL : https://journals.plos.org/plospathogens/article/file?id=10.1371/journal.ppat.1005015&type=printable

G. Schalk and M. R. Forbes, Male biases in parasitism of mammals: effects of study type, host age, and parasite taxon, Oikos, vol.78, pp.67-74, 1997.

P. Schmid-hempel, Variation in immune defence as a question of evolutionary ecology, Proceedings of the Royal Society of London B: Biological Sciences, vol.270, pp.357-366, 2003.

P. Schmid-hempel, Evolutionary ecology of insect immune defenses, Annual Review of Entomology, vol.50, pp.529-551, 2005.

P. Schmid-hempel, Immune defence, parasite evasion strategies and their relevance for 'macroscopic phenomena' such as virulence, Philosophical Transactions of the Royal Society B: Biological Sciences, vol.364, pp.85-98, 2009.

D. S. Schneider and J. S. Ayres, Two ways to survive infection: what resistance and tolerance can teach us about treating infectious diseases, Nature Reviews Immunology, vol.8, pp.889-895, 2008.

E. Schnepf, N. Crickmore, J. Van-rie, D. Lereclus, J. Baum et al., Bacillus thuringiensis and its pesticidal crystal proteins, Microbiology and Molecular Biology Reviews, vol.62, pp.775-806, 1998.

G. A. Schwarzenbach, D. J. Hosken, and P. I. Ward, Sex and immunity in the yellow dung fly Scathophaga stercoraria, Journal of Evolutionary Biology, vol.18, pp.455-463, 2005.

S. C. Seehuus, K. Norberg, U. Gimsa, T. Krekling, and G. V. Amdam, Reproductive protein protects functionally sterile honey bee workers from oxidative stress, Proceedings of the, vol.103, pp.962-967, 2006.

M. Seppola, H. Johnsen, S. Mennen, B. Myrnes, and H. Tveiten, Maternal transfer and transcriptional onset of immune genes during ontogenesis in Atlantic cod, Developmental Comparative Immunology, vol.33, pp.205-211, 2009.

B. C. Sheldon and S. Verhulst, Ecological immunology: costly parasite defences and trade-offs in evolutionary ecology, Trends in Ecology & Evolution, vol.11, pp.317-321, 1996.

Z. H. Shi, Y. T. Lin, and Y. M. Hou, Mother-derived trans-generational immune priming in the red palm weevil, Rhynchophorus ferrugineus Olivier (Coleoptera, Dryophthoridae), Bulletin of Entomological Research, vol.104, pp.742-750, 2014.

I. Shikano, K. N. Hua, and J. S. Cory, Baculovirus-challenge and poor nutrition inflict within-generation fitness costs without triggering transgenerational immune priming, Journal of Invertebrate Pathology, vol.136, pp.35-42, 2016.

M. S. Singer, P. A. Mason, and A. M. Smilanich, Ecological immunology mediated by diet in herbivorous insects, Integrative and Comparative Biology, vol.54, pp.913-921, 2014.

N. K. Singh, B. C. Pakkianathan, M. Kumar, T. Prasad, M. Kannan et al., Vitellogenin from the silkworm, Bombyx mori: an effective anti-bacterial agent, PLoS One, vol.8, p.73005, 2013.

M. T. Siva-jothy, Y. Moret, and J. Rolff, Insect immunity: An evolutionary ecology perspective, Advances in Insect Physiology, vol.32, pp.1-48, 2005.

K. Söderhäll and L. Cerenius, Role of the prophenoloxidase-activating system in invertebrate immunity, Current Opinion in Immunology, vol.10, pp.23-28, 1998.

R. J. ?rám, B. Binková, J. Dejmek, and M. Bobak, Ambient air pollution and pregnancy outcomes: a review of the literature, Environmental Health Perspectives, vol.113, pp.375-382, 2005.

S. C. Stearns, The evolution of life histories, 1992.

D. Stein, J. S. Goltz, J. Jurcsak, and L. Stevens, The Dorsal-related immunity factor (Dif) can define the dorsal-ventral polarity in the Drosophila embryo, Development, vol.125, pp.2159-2169, 1998.

M. G. Sterken, L. B. Snoek, K. J. Bosman, J. Daamen, J. A. Riksen et al.,

P. Kammenga and J. E. , A heritable antiviral RNAi response limits Orsay virus infection in Caenorhabditis elegans N2, PLoS One, vol.9, p.89760, 2014.

E. D. Sternberg, J. C. De-roode, and M. D. Hunter, Trans-generational parasite protection associated with paternal diet, The Journal of Animal Ecology, vol.84, pp.310-321, 2015.

J. Stoops, S. Crauwels, M. Waud, J. Claes, B. Lievens et al., , 2016.

, Microbial community assessment of mealworm larvae (Tenebrio molitor) and grasshoppers (Locusta migratoria migratorioides) sold for human consumption, Food Microbiology, vol.53, pp.122-127

M. Sugumaran, K. Nellaiappan, and K. Valivittan, A new mechanism for the control of phenoloxidase activity: inhibition and complex formation with quinone isomerase, Archives of Biochemistry and Biophysics, vol.379, pp.252-260, 2000.

M. Sugumaran, Comparative biochemistry of eumelanogenesis and the protective roles of phenoloxidase and melanin in insects, Pigment Cell Research, vol.15, pp.2-9, 2002.

J. C. Sun, S. Ugolini, and E. Vivier, Immunological memory within the innate immune system, The EMBO Journal, vol.33, pp.1295-1303, 2014.

V. O. Sysoev, B. Fischer, C. K. Frese, I. Gupta, J. Krijgsveld et al., Global changes of the RNA-bound proteome during the maternal-tozygotic transition in Drosophila, Nature Communications, vol.7, p.12128, 2016.

N. Takamatsu, T. Shiba, K. Muramoto, and H. Kamiya, Molecular cloning of the defense factor in the albumen gland of the sea hare Aplysia kurodai, FEBS Letters, vol.377, pp.373-376, 1995.

A. T. Tate, The interaction of immune priming with different modes of disease transmission, Frontiers in Microbiology, vol.7, p.1102, 2016.

A. T. Tate and A. L. Graham, Trans-generational priming of resistance in wild flour beetles reflects the primed phenotypes of laboratory populations and is inhibited by co-infection with a common parasite, Functional Ecology, vol.29, pp.1059-1069, 2015.

A. T. Tate and V. H. Rudolf, Impact of life stage specific immune priming on invertebrate disease dynamics, Oikos, vol.121, pp.1083-1092, 2012.

G. Tetreau, J. Dhinaut, B. Gourbal, and Y. Moret, Trans-generational immune priming in invertebrates: where are we now and where must we go?

U. Theopold, S. Ekengren, and D. Hultmark, HLH106, a Drosophila transcription factor with similarity to the vertebrate sterol responsive element binding protein, Proceedings of the National Academy of Sciences, vol.93, pp.1195-1199, 1996.

U. Theopold, M. Rissler, M. Fabbri, O. Schmidt, and S. Natori, Insect glycobiology: a lectin multigen family in Drosophila melanogaster, Biochemical and Biophysical Research Communications, vol.261, pp.923-927, 1999.

R. Thornhill and J. Alcock, The evolution of insect mating system, 1983.

H. J. Tidbury, A. Best, and M. Boots, The epidemiological consequences of immune priming, Proceedings of the Royal Society of London B: Biological Sciences, vol.279, pp.4505-4512, 2012.

H. J. Tidbury, A. B. Pedersen, and M. Boots, Within and transgenerational immune priming in an insect to a DNA virus, Proceedings of the Royal Society B: Biological Sciences, vol.278, pp.871-876, 2011.

T. Ö. Tingvall, E. Roos, and Y. Engström, The GATA factor Serpent is required for the onset of the humoral immune response in Drosophila embryos, Proceedings of the National Academy of Sciences U.S.A, vol.98, pp.3884-3888, 2001.

C. J. Torgerson, Publication Bias: The Achilles' heel of systematic reviews?, British Journal of Educational Studies, vol.54, pp.89-102, 2010.

U. Trauer-kizilelma and M. Hilker, Impact of transgenerational immune priming on the defence of insect eggs against parasitism, Developmental & Comparative Immunology, vol.51, pp.126-133, 2015.

U. Trauer-kizilelma and M. Hilker, Insect parents improve the anti-parasitic and antibacterial defence of their offspring by priming the expression of immune-relevant genes, Insect Biochemistry and Molecular Biology, vol.64, pp.91-99, 2015.

U. Trauer and M. Hilker, Parental legacy in insects: variation of transgenerational immune priming during offspring development, PLoS One, vol.8, p.63392, 2013.

A. M. Triggs, R. J. Knell, and L. Martin, Parental diet has strong transgenerational effects on offspring immunity, Functional Ecology, vol.26, pp.1409-1417, 2012.

S. Tsakas and V. J. Marmaras, Insect immunity and its signalling: an overview, 2010.

, Invertebrate Survival Journal, vol.7, pp.228-238

M. Tseng, Interactions between the parasite's previous and current environment mediate the outcome of parasite infection, The American Naturalist, vol.168, pp.565-571, 2006.

M. Tufail and M. Takeda, Molecular characteristics of insect vitellogenins, Journal of Insect Physiology, vol.54, pp.1447-1458, 2008.

V. Vachon, R. Laprade, and J. L. Schwartz, Current models of the mode of action of Bacillus thuringiensis insecticidal crystal proteins: a critical review, Journal of Invertebrate Pathology, vol.111, pp.1-12, 2012.

A. Vantaux, K. R. Dabire, A. Cohuet, and T. Lefevre, A heavy legacy: offspring of malaria-infected mosquitoes show reduced disease resistance, Malaria Journal, vol.13, p.442, 2014.

A. Vilcinskas, The role of epigenetics in host-parasite coevolution: lessons from the model host insects Galleria mellonella and Tribolium castaneum, Zoology (Jena), vol.119, pp.273-280, 2016.

E. Vivier and B. Malissen, Innate and adaptive immunity: specificities and signaling hierarchies revisited, Nature Immunology, vol.6, pp.17-21, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00165761

J. Vizioli and M. Salzet, Antimicrobial peptides from animals: focus on invertebrates, Trends in Pharmacological Sciences, vol.23, pp.494-496, 2002.

M. J. Voordouw, L. Lambrechts, and J. C. Koella, No maternal effects after stimulation of the melanization response in the yellow fever mosquito Aedes aegypti, Oikos, vol.117, pp.1269-1279, 2008.

C. Vorburger, S. E. Gegenschatz, G. Ranieri, and P. Rodriguez, Limited scope for maternal effects in aphid defence against parasitoids, Ecological Entomology, vol.33, pp.189-196, 2008.

L. Wang, F. Yue, X. Song, and L. Song, Maternal immune transfer in mollusc, 2015.

, Developmental & Comparative Immunology, vol.48, pp.354-359

Y. Wang and Y. Zhang, Investigation of gut-associated bacteria in Tenebrio molitor (Coleoptera: Tenebrionidae) larvae using culture-dependent and DGGE methods, Annals of the Entomological Society of America, vol.108, pp.941-949, 2015.

F. L. Watson, R. Püttmann-holgado, F. Thomas, D. L. Lamar, M. Hughes et al., Extensive diversity of Ig-superfamily proteins in the immune system of insects, Science, vol.309, pp.1874-1878, 2005.

C. Wedekind and P. J. Jakobsen, Male-biased susceptibility to helminth infection: an experimental test with a copepod, Oikos, vol.81, pp.458-462, 1998.

J. Weiske and A. Wiesner, Stimulation of NO synthase activity in the immune-competent lepidopteran Estigmene acraea hemocyte line, Nitric Oxide Biology and Chemestry, vol.3, pp.123-131, 1999.

M. M. Whitten and N. A. Ratcliffe, In vitro superoxide activity in the haemolymph of the West Indian leaf cockroach, Blaberus discoidalis, Journal of Insect Physiology, vol.45, pp.667-675, 1999.

A. K. Wiklund and B. Sundelin, Impaired reproduction in the amphipods Monoporeia affinis and Pontoporeia femorata as a result of moderate hypoxia and increased temperature, 2001.

, Marine Ecology Progress Series, vol.222, pp.131-141

K. Wilson and R. I. Graham, Transgenerational effects modulate density-dependent prophylactic resistance to viral infection in a lepidopteran pest, Biology Letters, vol.11, 2015.

J. B. Wolf, I. Brodie, E. D. Cheverud, J. M. Moore, A. J. Wade et al., Evolutionary consequences of indirect genetic effects, Trends in Ecology & Evolution, vol.13, pp.64-69, 1998.

G. Wu, L. Xu, and Y. Yi, Galleria mellonella larvae are capable of sensing the extent of priming agent and mounting proportionatal cellular and humoral immune responses, 2016.

, Immunology Letters, vol.174, pp.45-52

F. Yue, Z. Zhou, L. Wang, Z. Ma, J. Wang et al., Maternal transfer of immunity in scallop Chlamys farreri and its trans-generational immune protection to offspring against bacterial challenge, Developmental & Comparative Immunology, vol.41, pp.569-577, 2013.

C. Zanchi, J. P. Troussard, G. Martinaud, J. Moreau, and Y. Moret, Differential expression and costs between maternally and paternally derived immune priming for offspring in an insect, Journal of Animal Ecology, vol.80, pp.1174-1183, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00634641

C. Zanchi, J. P. Troussard, J. Moreau, and Y. Moret, Relationship between maternal transfer of immunity and mother fecundity in an insect, Proceedings of the Royal Society B: Biological Sciences, vol.279, pp.3223-3230, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00717793

M. Zuk, Reproductive strategies and disease susceptibility: an evolutionary viewpoint, Parasitology Today, vol.6, pp.231-233, 1990.

M. Zuk and K. A. Mckean, Sex differences in parasite infections: patterns and processes, A dietary carotenoid reduces immunopathology and enhances, vol.26, pp.1009-1024, 1996.

:. J. Authors, A. Dhinaut, M. Balourdet, M. Teixeira, Y. Chogne et al., Evolutionary causes and consequences of immunopathology, Scientific Reports References, vol.36, pp.373-397, 2005.

G. Sorci and B. Faivre, Inflammation and oxidative stress in vertebrate host-parasite systems, Phil. Trans. Roy. Soc. Lond. B, vol.364, pp.71-83, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00356762

N. T. Ashley, Z. M. Weil, and R. J. Nelson, Inflammation: mechanisms, costs, and natural variation, Annu. Rev. Ecol. Evol. Syst, vol.43, pp.385-406, 2012.

C. A. Janeway, . Jr, and R. Medzhitov, Innate immune recognition, Ann. Rev. Immunol, vol.20, pp.197-216, 2002.

B. Stramer, W. Wood, M. J. Galko, M. J. Redd, A. Jacinto et al., Live imaging of wound inflammation in Drosophila embryos reveals key roles for small GTPases during in vivo cell migration, J. Cell. Biol, vol.168, pp.567-573

S. Libert, Y. F. Chao, X. W. Chu, and S. D. Pletcher, Trade-offs between longevity and pathogen resistance in Drosophila melanogaster are mediated by NF kappa B signalling, Aging Cell, vol.6, pp.533-543

E. R. Pursall and J. Rolff, Immune responses accelerate ageing: proof-of-principle in an insect model, PLoS ONE, vol.6, p.19972, 2011.

C. E. Finch and E. M. Crimmins, Inflammatory exposure and historical changes in human lifespans, Science, vol.305, pp.1736-1739, 2004.

F. Licastro, Innate immunity and inflammation in aging: a key for understanding agerelated diseases, Immunity & Ageing, vol.2, pp.8-22, 2005.

G. Pawelec, D. Goldeck, and E. Derhovanessian, Inflammation, ageing and chronic disease, Curr. Opin. Immunol, vol.29, pp.23-28, 2014.
DOI : 10.1016/j.coi.2014.03.007

B. P. Chew and J. S. Park, Carotenoid action on the immune response, J. Nutr, vol.134, pp.257-261, 2004.

G. W. Felton and C. B. Summers, Antioxidants systems in insects, Arch Insect Biochem, vol.29, pp.187-197, 1995.

C. Catoni, A. Peters, and H. M. Schaefer, Life history trade-offs are influenced by the diversity, availability and interactions of dietary antioxidants, Anim. Behav, vol.76, pp.1107-1119, 2008.

M. Flores, F. Diaz, R. Medina, A. D. Re, and A. Licea, Physiological, metabolic and haematological responses in white shrimp Litopenaeus vannamei (Boone) juveniles fed diets supplemented with astaxanthin acclimated to low-salinity, Aquac. Res, vol.38, pp.740-747, 2007.

J. S. Park, J. H. Chyun, Y. K. Kim, L. L. Line, and B. P. Chew, Astaxanthin decreased oxidative stress and inflammation and enhanced immune response in humans, Nutr. Metabol, vol.7, pp.1-18, 2010.

A. El-agamey, Carotenoid radical chemistry and antioxidant/pro-oxidant properties, Arch. Biochem. Biophys, vol.430, pp.37-48, 2004.
DOI : 10.1016/j.abb.2004.03.007

S. Walrand, In vivo and in vitro evidence that carotenoids could modulate the neutrophil respiratory burst during dietary manipulation, Eur. J. Nutr, vol.44, pp.114-120, 2005.

J. H. Kim, Y. S. Kim, G. G. Song, J. J. Park, and H. I. Chang, Protective effect of astaxanthin on naproxen-induced gastric antral ulceration in rats, Eur. J. Pharmacol, vol.514, pp.53-59, 2005.

D. H. Lee, C. S. Kim, and Y. I. Lee, Astaxanthin protects against MPTP/MPP plus-induced mitochondrial dysfunction and ROS production in vivo and in vitro, Food Chem. Toxicol, vol.49, pp.271-280, 2011.

A. Babin, Limiting immunopathology: interaction between carotenoids and enzymatic antioxidant defences, Dev. Comp. Immunol, vol.49, pp.278-281, 2015.
DOI : 10.1016/j.dci.2014.12.007

URL : https://hal.archives-ouvertes.fr/hal-01100356

V. A. Olson and I. P. Owens, Costly sexual signals: are carotenoids rare, risky, or required, Trends Ecol. Evol, vol.13, pp.510-515, 1998.
DOI : 10.1016/s0169-5347(98)01484-0

K. A. Huggins, K. J. Navara, M. T. Mendonça, and E. H. Geoffrey, Detrimental effects of carotenoid pigments: the dark side of bright coloration, Naturwissenschaften, vol.97, pp.637-644, 2010.

M. J. Simons, M. Briga, B. Leenknegt, and S. Verhulst, Context-dependent effects of carotenoid supplementation on reproduction in zebra finches, Behav. Ecol, vol.25, pp.945-950, 2014.

G. Hussein, U. Sankawa, H. Goto, K. Matsumoto, and H. Watanabe, Astaxanthin, a Carotenoid with Potential in Human Health and Nutrition, J. Nat. Prod, vol.69, pp.443-449, 2006.

S. Cornet, C. Biard, and Y. Moret, Is there a role for antioxidant carotenoids in limiting selfharming immune response in invertebrates?, Biol. Lett, vol.3, pp.284-288, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00168948

A. Babin, C. Biard, and Y. Moret, Dietary supplementation with carotenoids improves immunity without increasing its cost in a crustacean, Am. Nat, vol.176, pp.234-241, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00503833

P. D. Fraser and P. M. Bramley, The biosynthesis and nutritional uses of carotenoids, Prog. Lipid. Res, vol.43, pp.228-265, 2004.

S. Grünwald, I. V. Adam, A. M. Gurmai, L. Bauer, M. Boll et al., The red flour beetle Tribolium castaneum as a model to monitor food safety and functionality, Adv. Biochem. Eng. Biotechnol, vol.135, pp.111-122, 2013.

K. Söderhäll and L. Cerenius, Role of the prophenoloxidase-activating system in invertebrate immunity, Curr. Opin. Immunol, vol.10, pp.23-28, 1998.

M. T. Siva-jothy, Y. Moret, and J. Rolff, Insect immunity: an evolutionary ecology perspective, Adv. Insect Physiol, vol.32, pp.1-48, 2005.
DOI : 10.1016/s0065-2806(05)32001-7

A. J. Nappi, E. Vass, F. Frey, and Y. Carton, Nitric oxide involvement in Drosophila immunity, Nitric Oxide-Biol. Chem, vol.4, pp.423-430, 2000.
DOI : 10.1006/niox.2000.0294

A. J. Nappi and E. Ottaviani, Cytotoxicity and cytotoxic molecules in invertebrates, BioEssays, vol.22, pp.469-480, 2000.

A. Rivero, Nitric oxide: an antiparasitic molecule of invertebrates, Trends Parasitol, vol.22, pp.219-225, 2006.

S. Luckhart, Y. Vodovotz, L. Cui, and R. Rosenberg, The mosquito Anopheles stephensi limits malaria parasite development with inducible synthesis of nitric oxide, Proc. Natl. Acad. Sci. USA, vol.95, pp.5700-5705, 1998.

M. M. Whitten and N. A. Ratcliffe, In vitro superoxide activity in the hemolymph of the West Indian leaf cockroach, Blaberus discoidalis, J. Insect Physiol, vol.45, pp.667-675, 1999.

B. M. Sadd and M. T. Siva-jothy, Self-harm caused by an insect's innate immunity, Proc. R. Soc. B-Biol. Sci, vol.273, pp.2571-2574, 2006.
DOI : 10.1098/rspb.2006.3574

URL : http://europepmc.org/articles/pmc1634900?pdf=render

I. Khan, D. Agashe, and J. Rolff, Early inflammation, immunopathology and aging, Proc. R. Soc. B-Biol. Sci, vol.284, p.20170125, 2017.
DOI : 10.1101/076828

URL : https://www.biorxiv.org/content/biorxiv/early/2016/09/22/076828.full.pdf

R. S. Howard, The biology of the grain beetle Tenebrio molitor with particular reference to its behaviour, Ecology, vol.36, pp.262-269, 1955.

C. Tognetti, M. Moliné, M. Van-broock, and D. Libkind, Favored isolation and rapid identification of the astaxanthin-producing yeats Xanthophyllomyces dendrohous (Phaffia rhozyma) from environmental samples, J. Basic Microbiol, vol.53, pp.766-772, 2013.

M. M. Shah, Y. M. Liang, J. J. Cheng, and M. Daroch, Astaxanthin-producing green microalga Haemotococcus pluvialis: from single cell to high commercial products, Front. Plant Sci, vol.7, p.531, 2016.

E. R. Haine, Y. Moret, M. T. Siva-jothy, and J. Rolff, Antimicrobial defense and persistent infection in insects, Science, vol.322, pp.1257-1259, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00343225

J. L. Jurat-fuentes and T. Jackson, Bacterial Entomopathogens in Insect Pathology, 2012.

I. P. Angeles, Y. Chien, and A. V. Yambot, Effect of infected astaxanthin on survival, antioxidant capacity, and immune response of the giant freshwater prawn Macrobrachium rosenbergii (De Man, 1879) challenged with Lactococcus garvieae, J. Shellfish Res, vol.28, pp.931-937, 2009.

V. Kumar, B. R. Pillai, P. K. Sahoo, J. Mohanty, and S. Mohanty, Effect of dietary astaxanthin on growth and immune response of Giant freshwater prawn Macrobrachium rosenbergii (de man), Asian. Fish. Sci, vol.22, pp.61-69, 2009.

N. M. Wade, J. Gabaudan, and B. D. Glencross, A review of carotenoid utilisation and function in crustacean aquaculture, Rev. Aquac, vol.0, pp.1-16, 2015.

M. Imamura, J. Yang, and M. Yamakawa, cDNA cloning, characterization and gene expression of nitric oxide synthase from the silkworm, Bombyx mori, Insect Mol. Biol, vol.11, pp.257-265, 2002.

A. R. Kraaijeveld, N. P. Elrayes, H. Schuppe, and P. L. Newland, L-arginine enhances immunity to parasitoids in Drosophila melanogaster and increases NO production in lamellocytes

, Comp. Immunol, vol.35, pp.857-864, 2011.

I. Eleftherianos, Nitric oxide levels regulate the immune response of Drosophila melanogaster reference laboratory strains to bacterial infections, Infect. Immun, vol.82, pp.4169-4181, 2014.

U. Sanzhaeva, Y. Vorontsova, Y. Glazachev, and I. Slepneva, Dual effect of nitric oxide on phenoloxidase-mediated melanization, J. Enzyme Inhib. Med. Chem, vol.31, pp.1063-1068, 2016.

G. Hussein, Astaxanthin ameliorates features of metabolic syndrome in SHR/NDmcr-cp, Life Sci, vol.80, pp.522-529, 2007.

Y. Yang, Astaxanthin-rich extract from the green alga Haematococcus pluvialis lowers plasma lipid concentrations and enhances antioxidant defense in apolipoprotein E knockout mice, J. Nutr, vol.141, pp.1611-1617, 2011.

Y. Jia, Ursolic acid is a PPAR-? agonist that regulates hepatic lipid metabolism, Bioorg. Med. Chem. Lett, vol.21, pp.5876-5880, 2011.

P. Palozza, S. Serini, F. Di-nicuolo, and G. Calviello, Modulation of apoptotic signalling by carotenoids in cancer cells, Arch. Biochem. Biophys, vol.430, pp.104-109, 2004.

Y. Shao, Y. Ni, J. Yang, X. Lin, J. Li et al., Astaxanthin inhibits proliferation and induces apoptosis and cell cycle arrest of mice H22 hepatoma cells, Med. Sci. Monit, vol.22, pp.2152-2160, 2016.

A. Bergmann and H. Steller, Apoptosis, stem cells, and tissue regeneration, Sci. Signal, vol.3, p.8, 2010.

A. Dubuffet, C. Zanchi, G. Boutet, J. Moreau, M. Teixeira et al., Trans-generational immune priming protects the eggs only against Gram-positive bacteria in the mealworm beetle, PLoS Pathog, vol.11, p.1005178, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01214091

J. Dhinaut, M. Chogne, and Y. Moret, Immune priming specificity within and across generations reveals the range of pathogens affecting evolution of immunity in an insect, J. Anim. Ecol, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01728826

Y. Moret, Trans-generational immune priming: specific enhancement of the antimicrobial immune response in the mealworm beetle, Tenebrio molitor, Proc. R. Soc. B-Biol. Sci, vol.273, pp.1399-1405, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00258228

C. Zanchi, J. Troussard, G. Martinaud, J. Moreau, and Y. Moret, Differential expression and costs between maternally and paternally derived immune priming for offspring in an insect, J. Anim. Ecol, vol.80, pp.1174-1183, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00634641