• Keine Ergebnisse gefunden

VII. Results

2. Materials and methods

3.2 ELISA based binding studies

In order to verify the functionality of the newly produced ovine CTLR hFc-fusion proteins, ELISA-based binding studies were performed. Given the observed amino acid sequence similarity of CTLRs in different species (Fig. 1), we hypothesized that known ligands of murine and human CTLRs might be recognized by ovine CTLRs as well. For Dectin-1 testing, a β-glucan zymosan (Saijo and Iwakura, 2011) derived from Saccharomyces cerevisiae yeast, was selected. Zymosan is a common fungal and yeast cell wall constituent. For Dectin-2 and Langerin testing, α-linked mannose polysaccharide mannan (Hanske et al., 2017; Saijo and Iwakura, 2011) prepared from Saccharomyces cerevisiae cell wall via alkaline extraction was selected. For Mincle testing, a synthetic mycobacterial cord factor analog Trehalose-6,6-dibehenate (TDB) (Ishikawa et al., 2009) was applied. Indeed, substantial binding to the respective ligands could be observed for ovine and murine Dectin-1, Dectin-2 and Mincle, (Fig.

3A, B, D). In contrast, only marginal binding of ovine Langerin to mannan was detected (Fig.

3C), thus suggesting a potentially different ligand preference of ovine Langerin compared to its murine orthologue.

55 3.3 Flow cytometry based binding study

For the comparison of binding preferences between ovine and murine DNGR-1, a flow cytometry-based approach was chosen. To this end, CHO-cells were subjected to two freeze-thaw cycles in order to expose intracellular F-actin filaments, which act as potent DAMPs and are a known ligand of the murine DNGR-1 (Ahrens et al., 2012). Hierarchical gating strategy was applied to discriminate live cells from dead cells

and debris (Suppl. Fig. 1). The F-actin filaments were stained by murine as well as ovine DNGR-1 hFc-fusion proteins. The intact cells on the other hand did not reveal any substantial binding to murine or ovine DNGR-1 (Fig. 3E).

3.4 Pathogen screening

The applicability of the ovine CTLR hFc-fusion protein library for a flow-cytometry based pathogen screening was verified with Mycoplasma mycoides subsp. capri (Mmc). In the flow-cytometric analysis, DNA-positive cells were considered intact and gated accordingly for CTLR binding assessment (Fig. 4A).

Remarkably, selective binding of ovine DCIR, MCL and MICL hFc-fusion proteins to Mmc was observed (Fig. 4B) which calls for further examination. In contrast, no substantial binding was detected for Dectin-1, Dectin-2, DNGR-1, Langerin or Mincle, which might indicate a limited role of these CTLRs in Mmc recognition. However, we cannot formally exclude that certain binding epitopes on the Mmc surface may have been masked by the fixation procedure.

Fig. 4. Ovine CTLR hFc-fusion protein library application to screen for pathogen/CTLR interactions. Flow cytometry-based binding study with the ovine CTLR hFc-fusion protein library and Mycoplasma mycoides subsp. capri (Mmc) GM12 was performed. Mmc was gated for DNA-positive events to distinguish intact Mmc from debris (A). Binding of ovine CTLR hFc-fusion proteins to DNA-positive Mmc was investigated (B). Two independent experiments were performed in duplicates each.

4. Discussion

To screen for pathogen-host interactions, a comprehensive library of murine CTLR hFc-fusion protein receptors was previously established (Maglinao et al., 2014; Mayer et al., 2018;

Monteiro et al., 2019). In order to meet the need for comparable research tools aiming at the elucidation of pathogen-host interactions in large animals, in this study sheep CTLRs were selected. The importance of sheep farming for developing and developed countries as well as sheep’s role as vectors of multiple zoonotic diseases (Ganter, 2015), such as Rift Valley Fever, Tuberculosis and Brucellosis, highlights the relevance of small ruminant immunology research.

Chimeric CTLRs produced as dimeric human hFc-fusion proteins in a mammalian cell system exploits the advances of multivalent ligand recognition while at the same time mimicking the multimerization or high density clustering observed for some CTLRs in vivo (Dam and Brewer, 2010; Richardson and Williams, 2014). The versatile hFc-tag supports standard methods of fusion protein purification and detection while being compatible with CHO-cell protein expression system which provides a mammalian-type glycosylation. It has indeed been shown that glycosylation may impact CTLR binding capacity in vitro (Bloem et al., 2013; Su et al., 2005). The occurrence of recombinant proteins of different sizes, as observed for ovine Langerin and MICL, suggests the presence of alternative glycoforms, which may impact the functionality of the respective CTLR (Su et al., 2005) and which has been observed for human MICL in vivo (Marshall et al., 2006). Different CTLR variants characterized by SNPs in the respective CTLDs might feature alternative ligand preferences or affected binding capacities (Kumar et al., 2019). The capability of ovine Dectin-2, Mincle and DNGR-1 (one SNP each) to bind distinct ligands as well as the pathogen binding capacity of the ovine MICL (three distinct SNPs), all discussed in detail below, however, suggest a limited role of these point mutations for the function of respective CTLRs. The occurrence of CTLR polymorphisms in distinctive sheep populations (Zhou et al., 2010) still remains largely unknown. Furthermore, the sheep genome assembly Oar_v4.0 referenced in our study, which is built upon the genome sequencing data of a female and a male Texel sheep (Archibald et al., 2010), does not necessarily feature the most common CTLR variants in different sheep races.

The higher degree of morphological similarity of ruminant CTLRs to the human rather than to the murine orthologs (Fig. 1) allude that ligand preferences of ovine CTLRs might be even more similar to human CTLRs compared to murine ones and should be investigated in the future. The functional evaluation of the selected CTLR-hFc fusion protein library members nevertheless revealed comparable ligand binding preferences for most tested ovine CTLRs and their murine orthologs. The barely detectable binding of the ovine Langerin to mannan in

contrast to the substantial binding of the murine Langerin is in accordance with the recently discovered species-specific differences in murine and human Langerin ligand preferences (Hanske et al., 2017). This finding, as well as the newly-discovered species-specific variety in signal transduction and immune stimulation shown for murine and human Dectin-1 (Takano et al., 2017), and differences seen in Mincledependent TDB-mediated immune stimulation in humans, mice and cattle (Thakur et al., 2018) highlight the need for further studies on target species’ CTLRs and respective ligand interactions.

In our proof-of-principle flow cytometry-based pathogen binding study with the ovine CTLR hFc-fusion protein library, binding of ovine DCIR, MCL and MICL to Mmc was observed.

Being primarily associated with DAMP recognition and downregulation of the immune response (Fujikado et al., 2008; Redelinghuys et al., 2016), these CTLRs have also been shown to play an important role in PAMP recognition and related immune signaling (Bloem et al., 2014; Raulf et al., 2019; Wang et al., 2015). The elucidation of associated molecular mechanisms alongside with the investigation of these CTLRs involvement in Mmc infections in vivo thus can become the scope of further studies.

Neither Mmc, nor other mycoplasmas have yet been reported to interact with transmembrane CTLRs of any species. However, for human and rat lung surfactant protein D (SP-D), a soluble C-type lectin, specific binding to Mycoplasma pneumoniae has been described (Chiba et al., 2002). Binding studies with CTLRs of further species as well as the application of the ovine CTLR hFc-fusion protein library for screenings with additional ruminant and non-ruminant Mycoplasma species might provide useful insights into veterinary relevant mycoplasmoses.

5. Conclusion

The here generated ovine CTLR hFc-fusion protein library is a versatile tool allowing for the identification of pathogen interactions with ovine CTLRs. However, it may not only provide insights into ligand preferences of single ovine CTLRs, but it also enables a comparison with CTLRs across different species. Further screenings with bacterial and viral pathogens of sheep and other species as well as the extension of the library with additional ovine CTLRs such as MDL-1, MGL-1, Clec2L and Clec15B, and comparisons between different CTLR isoforms are conceived.

6. Funding

This work was supported by the “Nationale Forschungsplattform für Zoonosen”

(DLR/BMBF, Fkz. 01KI1724). We also acknowledge support from the Niedersachsen-Research Network on Neuroinfectiology (NRENNT-2).

7. Declaration of Competing Interest

No author has any competing interests or conflicts of interests.

8. Acknowledgement

We thank Julian Glanz and Anna Sternberg for their assistance in bioinformatic analysis and Silke Schöneberg for technical assistance. We thank the Clinic for Swine, Small Ruminants and Forensic Medicine and the Institute for Pathology, University for Veterinary Medicine Hannover, Foundation, for the provided biological materials.

9. Appendix A. Supplementary data

Supplementary material related to this article can be found, in the online version, at doi:https://doi.org/10.1016/j.vetimm.2020.110047.

10. References

Ahrens, S., Zelenay, S., Sancho, D., Hanč, P., Kjær, S., Feest, C., Fletcher, G., Durkin, C., Postigo, A., Skehel, M., Batista, F., Thompson, B., Way, M., Reis e Sousa, C., Schulz, O., 2012. F-actin is an evolutionarily conserved damage-associated molecular pattern recognized by DNGR-1, a receptor for dead cells. Immunity 36, 635–645.

Archibald, A.L., Cockett, N.E., Dalrymple, B.P., Faraut, T., Kijas, J.W., Maddox, J.F.,

McEwan, J.C., Hutton Oddy, V., Raadsma, H.W., Wade, C., Wang, J., Wang, W., Xun, X., 2010. The sheep genome reference sequence: a work in progress. Anim. Genet. 41, 449–453.

Behler, F., Steinwede, K., Balboa, L., Ueberberg, B., Maus, R., Kirchhof, G., Yamasaki, S., Welte, T., Maus, U.A., 2012. Role of Mincle in alveolar macrophage-dependent innate immunity against mycobacterial infections in mice. J. Immunol. 189, 3121–3129.

Bloem, K., Vuist, I.M., van den Berk, M., Klaver, E.J., van Die, I., Knippels, L.M., Garssen, J., Garcia-Vallejo, J.J., van Vliet, S.J., van Kooyk, Y., 2014. DCIR interacts with ligands from both endogenous and pathogenic origin. Immunol. Lett. 158, 33–41.

Bloem, K., Vuist, I.M., van der Plas, A.-J., Knippels, L.M.J., Garssen, J., García-Vallejo, J.J., van Vliet, S.J., van Kooyk, Y., 2013. Ligand binding and signaling of dendritic cell immunoreceptor (DCIR) is modulated by the glycosylation of the carbohydrate recognition domain. PLoS One 8, e66266.

Bonkobara, M., Hoshino, M., Yagihara, H., Tamura, K., Isotani, M., Tanaka, Y., Washizu, T., Ariizumi, K., 2006. Identification and gene expression of bovine C-type lectin dectin-2. Vet.

Immunol. Immunopathol. 110, 179–186.

Cheng, A.C., Yang, K.Y., Chen, N.J., Hsu, T.L., Jou, R., Hsieh, S.L., Tseng, P.H., 2017.

CLEC9A modulates macrophage-mediated neutrophil recruitment in response to heat-killed Mycobacterium tuberculosis H37Ra. PLoS One 12, e0186780.

Chiba, H., Pattanajitvilai, S., Evans, A.J., Harbeck, R.J., Voelker, D.R., 2002. Human surfactant protein D (SP-D) binds Mycoplasma pneumoniae by high affinity interactions with lipids. J.

Biol. Chem. 277, 20379–20385.

Dam, T.K., Brewer, C.F., 2010. Lectins as pattern recognition molecules: the effects of epitope density in innate immunity. Glycobiology 20, 270–279.

DaMassa, A.J., Brooks, D.L., Adler, H.E., 1983. Caprine mycoplasmosis: widespread infection in goats with Mycoplasma mycoides subsp mycoides (large-colony type). Am. J. Vet. Res.

44, 322–325.

de Jong, M.A., Vriend, L.E., Theelen, B., Taylor, M.E., Fluitsma, D., Boekhout, T., Geijtenbeek, T.B., 2010. C-type lectin Langerin is a beta-glucan receptor on human Langerhans cells that recognizes opportunistic and pathogenic fungi. Mol. Immunol.

47, 1216–1225.

Feinberg, H., Jegouzo, S.A., Rowntree, T.J., Guan, Y., Brash, M.A., Taylor, M.E., Weis, W.I., Drickamer, K., 2013. Mechanism for recognition of an unusual mycobacterial glycolipid by the macrophage receptor mincle. J. Biol. Chem. 288, 28457–28465.

Ferwerda, B., Ferwerda, G., Plantinga, T.S., Willment, J.A., van Spriel, A.B., Venselaar, H., Elbers, C.C., Johnson, M.D., Cambi, A., Huysamen, C., 2009. Human dectin-1 deficiency and mucocutaneous fungal infections. N. Engl. J. Med. 361, 1760–1767.

Fischer, A., Shapiro, B., Muriuki, C., Heller, M., Schnee, C., Bongcam-Rudloff, E., Vilei, E.M., Frey, J., Jores, J., 2012. The origin of the’ Mycoplasma mycoides cluster’ coincides with domestication of ruminants. PLoS One 7, e36150.

Fujikado, N., Saijo, S., Yonezawa, T., Shimamori, K., Ishii, A., Sugai, S., Kotaki, H., Sudo, K., Nose, M., Iwakura, Y., 2008. Dcir deficiency causes development of autoimmune diseases in mice due to excess expansion of dendritic cells. Nat. Med. 14, 176–180.

Ganter, M., 2015. Zoonotic risks from small ruminants. Vet. Microbiol. 181, 53–65.

Hanske, J., Schulze, J., Aretz, J., McBride, R., Loll, B., Schmidt, H., Knirel, Y., Rabsch, W., Wahl, M.C., Paulson, J.C., Rademacher, C., 2017. Bacterial polysaccharide specificity of the pattern recognition receptor Langerin is highly species-dependent. J. Biol.

Chem. 292, 862–871.

Hattori, Y., Morita, D., Fujiwara, N., Mori, D., Nakamura, T., Harashima, H., Yamasaki, S., Sugita, M., 2014. Glycerol monomycolate is a novel ligand for the human, but not mouse macrophage inducible C-type lectin, Mincle. J. Biol. Chem. 289, 15405–15412.

Heinsbroek, S.E., Brown, G.D., Gordon, S., 2005. Dectin-1 escape by fungal dimorphism.

Trends Immunol. 26, 352–354.

Hernandez, L., Lopez, J., St-Jacques, M., Ontiveros, L., Acosta, J., Handel, K., 2006.

Mycoplasma mycoides subsp. capri associated with goat respiratory disease and high flock mortality. Can. Vet. J. 47, 366–369.

Heukeshoven, J., Dernick, R., 1988. Improved silver staining procedure for fast staining in phastsystem development Unit. I. Staining of sodium dodecyl sulfate gels.

Electrophoresis 9, 28–32.

Iborra, S., Martínez-López, M., Cueto, F.J., Conde-Garrosa, R., Del Fresno, C., Izquierdo, H.M., Abram, C.L., Mori, D., Campos-Martín, Y., Reguera, R.M., Kemp, B., Yamasaki, S., Robinson, M.J., Soto, M., Lowell, C.A., Sancho, D., 2016. Leishmania uses Mincle to target an inhibitory ITAM signaling pathway in dendritic cells that dampens adaptive immunity to infection. Immunity 45, 788–801.

Iliev, I.D., Funari, V.A., Taylor, K.D., Nguyen, Q., Reyes, C.N., Strom, S.P., Brown, J., Becker, C.A., Fleshner, P.R., Dubinsky, M., 2012. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science 336, 1314–1317.

Ishikawa, E., Ishikawa, T., Morita, Y.S., Toyonaga, K., Yamada, H., Takeuchi, O., Kinoshita, T., Akira, S., Yoshikai, Y., Yamasaki, S., 2009. Direct recognition of the mycobacterial glycolipid, trehalose dimycolate, by C-type lectin Mincle. J. Exp. Med. 206, 2879.

Jin, X., Zhang, M., Cao, G.-F., Yang, Y.-F., 2019. Saccharomyces cerevisiae mannan induces sheep beta-defensin-1 expression via Dectin-2-Syk-p38 pathways in ovine ruminal epithelial cells. Vet. Res. 50, 8.

Kapoor, P.K., Garg, D.N., Mahajan, S.K., 1989. Isolation of Mycoplasma mycoides subsp.

mycoides (LC variant, Y-Goat) from naturally aborted bovine fetuses. Theriogenology 32, 683–691.

Kumar, S., Kumar, S., Singh, R.V., Chauhan, A., Kumar, A., Bharati, J., Singh, S.V., 2019.

Association of Bovine CLEC7A gene polymorphism with host susceptibility to paratuberculosis disease in Indian cattle. Res. Vet. Sci. 123, 216–222.

Larsen, F.T., Bed’Hom, B., Guldbrandtsen, B., Dalgaard, T.S., 2019. Identification and tissue-expression profiling of novel chicken c-type lectin-like domain containing proteins as potential targets for carbohydrate-based vaccine strategies. Mol.Immunol. 114, 216–225.

Letunic, I., Bork, P., 2018. 20 years of the SMART protein domain annotation resource. Nucleic Acids Res. 46, D493–D496.

Maglinao, M., Eriksson, M., Schlegel, M.K., Zimmermann, S., Johannssen, T., Gotze, S., Seeberger, P.H., Lepenies, B., 2014. A platform to screen for C-type lectin receptorbinding carbohydrates and their potential for cell-specific targeting and immune modulation. J.

Control. Release 175, 36–42.

Marshall, A.S., Willment, J.A., Pyz, E., Dennehy, K.M., Reid, D.M., Dri, P., Gordon, S., Wong, S.Y., Brown, G.D., 2006. Human MICL (CLEC12A) is differentially glycosylated and is down-regulated following cellular activation. Eur. J. Immunol. 36, 2159–2169.

Mayer, S., Moeller, R., Monteiro, J.T., Ellrott, K., Josenhans, C., Lepenies, B., 2018. C-type lectin receptor (CLR)–fc fusion proteins as tools to screen for novel CLR/Bacteria interactions: an exemplary study on preselected Campylobacter jejuni isolates. Front.

Immunol. 9.

Mayer, S., Raulf, M.K., Lepenies, B., 2017. C-type lectins: their network and roles in pathogen recognition and immunity. Histochem. Cell Biol. 147, 223–237.

Monteiro, J.T., Schön, K., Ebbecke, T., Goethe, R., Ruland, J., Baumgärtner, W., Becker, S.C., Lepenies, B., 2019. The CARD9-Associated C-Type lectin, mincle, recognizes La crosse virus (LACV) but plays a limited role in early antiviral responses against LACV. Viruses 11, 303.

Neumann, K., Castineiras-Vilarino, M., Hockendorf, U., Hannesschlager, N., Lemeer, S., Kupka, D., Meyermann, S., Lech, M., Anders, H.J., Kuster, B., Busch, D.H., Gewies, A., Naumann, R., Gross, O., Ruland, J., 2014. Clec12a is an inhibitory receptor for uric acid crystals that regulates inflammation in response to cell death. Immunity 40, 389–399.

Ng, W.C., Londrigan, S.L., Nasr, N., Cunningham, A.L., Turville, S., Brooks, A.G., Reading, P.C., 2015. The C-type lectin Langerin functions as a receptor for attachment and infectious entry of influenza a virus. J. Virol. 90, 206–221.

Nicholas, R., Ayling, R., McAuliffe, L., 2008. Respiratory diseases of small ruminants. In:

Nicholas, R., Ayling, R., McAuliffe, L. (Eds.), Mycoplasma Diseases of Ruminants.

CABI, Wallingford, UK, pp. 169–198.

Osorio, F., Reis e Sousa, C., 2011. Myeloid C-type lectin receptors in pathogen recognition and host defense. Immunity 34, 651–664.

Paradis, E., Schliep, K., 2018. Ape 5.0: an environment for modern phylogenetics and evolutionary analyses in R. Bioinformatics (Oxford, England).

Perreau, P., Bind, J., 1981. Infection naturelle du veau par Mycoplasma mycoides subsp.

mycoides (biotype chevre). Bulletin de l’Académie Vétérinaire de France.

Core Team, R., 2018. R: a language and environment for statistical computing. R Foundation for Statistical Computing. , Vienna, Austria. Available online at. https:// www.R-project.org/.

Raulf, M.K., Johannssen, T., Matthiesen, S., Neumann, K., Hachenberg, S., MayerLambertz, S., Steinbeis, F., Hegermann, J., Seeberger, P.H., Baumgartner, W., Strube, C., Ruland, J., Lepenies, B., 2019. The C-type lectin receptor CLEC12A recognizes plasmodial hemozoin and contributes to cerebral malaria development. Cell Rep. 28, 30–38 e35.

Redelinghuys, P., Whitehead, L., Augello, A., Drummond, R.A., Levesque, J.-M., Vautier,S., Reid, D.M., Kerscher, B., Taylor, J.A., Nigrovic, P.A., Wright, J., Murray, G.I.,Willment, J.A., Hocking, L.J., Fernandes, M.J.G., De Bari, C., McInnes, I.B., Brown, G.D., 2016.

MICL controls inflammation in rheumatoid arthritis. Ann. Rheum. Dis. 75, 1386–1391.

Richardson, M.B., Williams, S.J., 2014. MCL and mincle: C-Type lectin receptors that sense damaged self and pathogen-associated molecular patterns. Front. Immunol. 5, 288.

Saijo, S., Iwakura, Y., 2011. Dectin-1 and Dectin-2 in innate immunity against fungi. Int.

Immunol. 23, 467–472.

Sattler, S., Ghadially, H., Hofer, E., 2012. Evolution of the C-type lectin-like receptor genes of the DECTIN-1 cluster in the NK gene complex. ScientificWorldJournal 2012, 931386.

Schliep, K.P., 2011. phangorn: phylogenetic analysis in R. Bioinformatics (Oxford, England) 27, 592–593.

Schoenen, H., Bodendorfer, B., Hitchens, K., Manzanero, S., Werninghaus, K.,

Nimmerjahn, F., Agger, E.M., Stenger, S., Andersen, P., Ruland, J., Brown, G.D., Wells, C., Lang, R., 2010. Cutting edge: Mincle is essential for recognition and adjuvanticity of the mycobacterial cord factor and its synthetic analog trehalose-dibehenate. J. Immunol. 184, 2756–2760.

Sonck, E., Stuyven, E., Goddeeris, B., Cox, E., 2009. Identification of the porcine C-type lectin dectin-1. Vet. Immunol. Immunopathol. 130, 131–134.

Stothard, P., 2000. The sequence manipulation suite: javascript programs for analyzing and formatting protein and DNA sequences. BioTechniques 28 (1102), 1104.

Su, Y., Bakker, T., Harris, J., Tsang, C., Brown, G.D., Wormald, M.R., Gordon, S., Dwek, R.A., Rudd, P.M., Martinez-Pomares, L., 2005. Glycosylation influences the lectin activities of the macrophage mannose receptor. J. Biol. Chem. 280, 32811–32820.

Takano, T., Motozono, C., Imai, T., Sonoda, K.H., Nakanishi, Y., Yamasaki, S., 2017. Dectin-1 intracellular domain determines species-specific ligand spectrum by modulating receptor sensitivity. J. Biol. Chem. 292, 16933–16941.

Tang, D., Kang, R., Coyne, C.B., Zeh, H.J., Lotze, M.T., 2012. PAMPs and DAMPs: signal 0s that spur autophagy and immunity. Immunol. Rev. 249, 158–175.

Thakur, A., Andrea, A., Mikkelsen, H., Woodworth, J.S., Andersen, P., Jungersen, G., Aagaard, C., 2018. Targeting the Mincle and TLR3 receptor using the dual agonist cationic adjuvant formulation 9 (CAF09) induces humoral and polyfunctional memory T cell responses in calves. PloS one 13, e0201253.

Thiaucourt, F., Bolske, G., 1996. Contagious caprine pleuropneumonia and other pulmonary mycoplasmoses of sheep and goats. Revue scientifique et technique 15, 1397–1414.

Trichard, C.J., Jordaan, P., Prozesky, L., Jacobsz, E.P., Henton, M.M., 1993. The identification of Mycoplasma mycoides mycoides LC as the aetiological agent of balanoposthitis and vulvovaginitis in sheep in South Africa. Onderstepoort J. Vet. Res. 60, 29–37.

Troegeler, A., Mercier, I., Cougoule, C., Pietretti, D., Colom, A., Duval, C., Vu Manh, T.P., Capilla, F., Poincloux, R., Pingris, K., Nigou, J., Rademann, J., Dalod, M., Verreck, F.A., Al Saati, T., Lugo-Villarino, G., Lepenies, B., Hudrisier, D., Neyrolles, O., 2017. C-type lectin receptor DCIR modulates immunity to tuberculosis by sustaining type I interferon signaling in dendritic cells. Proc. Natl. Acad. Sci. U. S. A. 114, E540–e549.

Ulrich-Lynge, S.L., Juul-Madsen, H.R., Kjaerup, R.B., Okimoto, R., Abrahamsen, M.S., Maurischat, S., Sorensen, P., Dalgaard, T.S., 2016. Broilers with low serum Mannosebinding Lectin show increased fecal shedding of Salmonella enterica serovar Montevideo. Poult. Sci.

95, 1779–1786.

Wang, H., Li, M., Lerksuthirat, T., Klein, B., Wuthrich, M., 2015. The C-Type lectin receptor MCL mediates vaccine-induced immunity against infection with blastomyces dermatitidis.

Infect. Immun. 84, 635–642.

Wang, S., Hu, D., Wang, C., Tang, X., Du, M., Gu, X., Suo, J., Hu, M., Fang, R., Zhu, X., Zhang, X., Du, A., Suo, X., Liu, X., 2019. Transcriptional profiling of innate immune responses in sheep PBMCs induced by Haemonchus contortus soluble extracts.

Parasit. Vectors 12, 182.

Weldearegay, Y.B., Muller, S., Hanske, J., Schulze, A., Kostka, A., Ruger, N., HewickerTrautwein, M., Brehm, R., Valentin-Weigand, P., Kammerer, R., Jores, J., Meens, J., 2019. Host-pathogen interactions of Mycoplasma mycoides in Caprine and bovine precision-cut lung slices (PCLS) models. Pathogens (Basel, Switzerland) 8.

Wevers, B.A., Kaptein, T.M., Zijlstra-Willems, E.M., Theelen, B., Boekhout, T., Geijtenbeek, T.B., Gringhuis, S.I., 2014. Fungal engagement of the C-type lectin Mincle suppresses dectin-1-induced antifungal immunity. Cell Host Microbe 15, 494–505.

Willcocks, S., Yamakawa, Y., Stalker, A., Coffey, T.J., Goldammer, T., Werling, D., 2006.

Identification and gene expression of the bovine C-type lectin Dectin-1. Vet. Immunol.

Immunopathol. 113, 234–242.

Wright, E.S., 2015. DECIPHER: harnessing local sequence context to improve protein multiple sequence alignment. BMC Bioinf. 16, 322.

Yamakawa, Y., Pennelegion, C., Willcocks, S., Stalker, A., MacHugh, N., Burt, D., Coffey, T.J., Werling, D., 2008. Identification and functional characterization of a bovine orthologue to DC-SIGN. J. Leukoc. Biol. 83, 1396–1403.

Yang, K., Park, C.G., Cheong, C., Bulgheresi, S., Zhang, S., Zhang, P., He, Y., Jiang, L., Huang, H., Ding, H., Wu, Y., Wang, S., Zhang, L., Li, A., Xia, L., Bartra, S.S., Plano, G.V., Skurnik, M., Klena, J.D., Chen, T., 2015. Host Langerin (CD207) is a receptor for Yersinia pestis phagocytosis and promotes dissemination. Immunol. Cell Biol. 93, 815–824.

Yao, W.R., Yu, L., Li, D., Yang, G.B., 2017. Molecular cloning and characterization of DNGR-1 in rhesus macaques. Mol. Immunol. 87, 2DNGR-17–226.

Ye, J., Coulouris, G., Zaretskaya, I., Cutcutache, I., Rozen, S., Madden, T.L., 2012.

PrimerBLAST: a tool to design target-specific primers for polymerase chain reaction. BMC Bioinf. 13, 134.

Yonekawa, A., Saijo, S., Hoshino, Y., Miyake, Y., Ishikawa, E., Suzukawa, M., Inoue, H., Tanaka, M., Yoneyama, M., Oh-hora, M., 2014. Dectin-2 is a direct receptor for mannose-capped lipoarabinomannan of mycobacteria. Immunity 41, 402–413.

Zhou, H., Hu, J., Luo, Y., Hickford, J.G., 2010. Variation in the ovine C-type lectin dectin1 gene (CLEC7A). Dev. Comp. Immunol. 34, 246–249.

Zhu, L.L., Zhao, X.Q., Jiang, C., You, Y., Chen, X.P., Jiang, Y.Y., Jia, X.M., Lin, X., 2013.

Ctype lectin receptors Dectin-3 and Dectin-2 form a heterodimeric pattern-recognition receptor for host defense against fungal infection. Immunity 39, 324–334.

VIII. Discussion

This thesis addresses one of the limitations of current research on CTLs in veterinary glycoimmunology, namely the lack of reliable tools to investigate CTL/ligand interactions of veterinary species in vitro. The need for specific tools in immunologic research is apparent, since major differences in PRRs ligand preferences have recently been revealed among phylogenetically distinct animals [142, 155], questioning the applicability of results obtained in model species [156]. Lacking more suitable alternatives, current studies in veterinary immunology are yet often compelled to using recombinant human PRRs [157] or mouse models [158-160] (Tab. 3).

Name Species Length Coverage Identity Accession Nr.

Dectin-1 Ovis aries 246 Querry Querry XP_027823677.1

Homo sapiens 247 100% 71,26% NP_922938.1

Mus musculus 244 99% 55,51% NP_064392.2

DNGR-1 Ovis aries 241 Querry Querry XP_004006925.3

Homo sapiens 241 100% 68,46% NP_997228.1

Homo sapiens 241 100% 68,46% NP_997228.1