Deoksyniwalenol - czynnikiem wspomagającym bakteryjne martwicze zapalenia jelit u drobiu


Magdalena Gajęcka, Michał Dąbrowski, Łukasz Zielonka, Maciej Gajęcki


Zarówno Clostridium perfringens jak i mikotoksyny a wśród nich deoksyniwalenol (DON), indukują martwicze zapalenie jelit u drobiu i mają coraz większy wpływ na światową produkcję drobiarską (Antonissen i wsp., 2014). Patogeny oporne na antybiotyki oraz rosnące obawy obecności pozostałości chemicznych w mięsie drobiowym, w postaci antybiotyków, tradycyjnie stosowanych w celu zapobiegania i kontroli np. martwiczego zapalenia jelit u drobiu, są ograniczane lub zaczyna się myśleć o ich wycofaniu ze stosowania w produkcji drobiarskiej (Tian i wsp., 2016).
Z drugiej strony, trichoteceny zasadniczo występują w niskich stężeniach (Bryła i wsp., 2016), są one jednak trudne do wykrycia. Analiza pasz nie zawsze daje dokładną ocenę występujących w niej metabolitów grzybów pleśniowych. Spowodowane to jest faktem: (i) niedoskonałością procedur analitycznych; lub (ii) obecnością nowych, nieznanych jeszcze tego typu związków; oraz (iii) brakiem umiejętności rozpoznawania objawów klinicznych u zwierząt narażonych na obecność mikotoksyn w paszach, co nie pozwala na wykonanie odpowiednich/właściwych analiz diagnostycznych (Krska i wsp., 2008).
Celem przedstawionej pracy jest próba uzasadnienia wpływu jednego z czynników predysponujących, jakim jest DON, do rozwoju martwiczego zapalenia jelit.

Literatura: 
 

Alassane-Kpembi, I., Kolf-Clauw, M., Gauthier, T., Abrami, R., Abiola, F.A., Oswald, I.P., Puel, O., New insights into mycotoxin mixtures: The toxicity of low doses of Type B trichothecenes on intestinal epithelial cells is synergistic, Toxicol. Appl. Pharm. 2013, 272, 191-198.
Antonissen, G., Van Immerseel, F., Pasmans, F., Ducatelle, R., Haesebrouck, F., Timbermont, L., Verlinden, M., Janssens, G.P.J., Eeckhaut, V., De Saeger, S., Hessenberger, S., Martel, A., Croubels, S., The Mycotoxin Deoxynivalenol Predisposes for the Development of Clostridium perfringens-Induced Necrotic Enteritis in Broiler Chickens, PLoS ONE 2014, 9 (9): e108775. doi: 10.1371 / journal.pone.0108775
Avantaggiato, G., Havenaar, R., Visconti, A., Evaluation of the intestinal absorption of deoxynivalenol and nivalenol by an in vitro gastrointestinal model, and the binding efficacy ofactivated carbon and other adsorbent materials, Food Chem. Toxicol. 2004, 42, 817-824.
Awad, W., Ghareeb, K., Böhm, J., Zentek, J., The Toxicological Impacts of the Fusarium Mycotoxin, Deoxynivalenol, in Poultry Flocks with Special Reference to Immunotoxicity, Toxins 2013, 5, 912-925; doi:10.3390/toxins5050912
Bezerra da Rocha, M.E., da Chagas, Oliveira Freire, F., Feitosa, Maia, F.E., Florindo, Guedes, M.I., Rondina, D., Mycotoxins and their effects on human and animal health, Food Control 2014, 36, 159-165.
Boermans, H.J., Leung, M.C., Mycotoxins and the pet food industry: toxicological evidence and risk assessment, Int. J. Food Microbiol. 2007, 119, 95-102.
Brandon, E.F.A., Bulder, A.S., van Engelen, J.G.M., Mahieu, C.M., Mennes, W.C., Pronk, M.E.J., Rietveld, A.G., van de Ven, B.M., ten Voorde, S.E.C.G., Wolterink, G., Slob, W., Zeilmaker, M.J., Bessems, J.G.M., Does EU legislation allow the use of the Benchmark dose (BMD) approach for risk assessment? Regul. Toxicol. Pharm. 2013, 67, 182-188.
Bryden, W.L., Mycotoxin contamination of the feed supply chain: Implications for animal productivity and feed security, Anim. Feed Sci. Tech. 2012, 173, 134-158.
Bryła, M.,  Waśkiewicz, A., Podolska, G., Szymczyk, K., Jędrzejczak, R., Damaziak, K., Sułek, A., Occurrence of 26 Mycotoxins in the Grain of Cereals Cultivated in Poland, Toxins 2016, 8, 160; doi:10.3390/toxins8060160
Calabrese, E.J., Paradigm lost, paradigm found: the re-emergence of hormesis as a fundamental dose response model in the toxicological sciences, Environ. Pollut. 2005, 138, 378-411.
De Angelis, E., Monaci, L., Visconti, A., Investigation on the stability of deoxynivalenol and DON-3 glucoside during gastro-duodenal in vitro digestion of a naturally contaminated bread model food, Food Control 2014, 43, 270-275.
del Cacho, E., Gallego, M., Lee, S.H., Lillehoj, H.S., Quilez, J., Lillehoj, E.P., Sánchez-Acedoa, C. Induction of Protective Immunity against Eimeria tenella, Eimeria maxima, and Eimeria acervulina Infections Using Dendritic Cell-Derived Exosomes, Infect. Immun. 2012, 80 (5), 1909–1916. doi:10.1128/IAI.06413-11
Dobrzyński, L., Fornalski, K.W., Hormesis - Natural phenomenon of answer of organism on stress Proceeding, VII International Scientific Conference: Veterinary Feed Hygiene – The Effects of Mycotoxins on Gastrointestinal Function. 23-24 September 2011, Olsztyn, Poland 2011, 6-14.
Du, E., Wang, W., Gan, L., Li, Z., Guo, S., Guo, Y., Effects of thymol and carvacrol supplementation on intestinal integrity and immune responses of broiler chickens challenged with Clostridium perfringens, J. Anim. Sci. Biotech. 2016, 7:19. DOI 10.1186/s40104-016-0079-7
EFSA – 17th Scientific Colloquium Summary Report. Low-dose response in toxicology and risk assessment, 14-15 June 2012, Parma, Italy.
Frizzell, C., Ndossi, D., Verhaegen, S., Dahl, E., Eriksen, G., Sřrlie, M., Ropstad, E., Muller, M., Elliott, C.T., Connolly, L., Endocrine disrupting effects of zearalenone, alpha- and beta-zearalenol at the level of nuclear receptor binding and steroidogenesis, Toxicol. Lett. 2011, 206, 210-217.
Frizzell, C., Verhaegen, S., Ropstad, E., Elliott, C.T., Connolly, L., Endocrine  disrupting  effects  of  ochratoxin  A  at  the  level  of  nuclear  receptor activation  and  steroidogenesis, Toxicol. Lett. 2013, 217, 243-250.
Gajęcka, M., Stopa, E., Tarasiuk, M., Zielonka, Ł., Gajęcki, M., The expression of type-1 and type-2 nitric oxide synthase in selected tissues of the gastrointestinal tract during mixed mycotoxicosis, Toxins 2013, 5(11), 2281-2292.
Grenier, B., Applegate, T.J., Modulation of intestinal functions following mycotoxin ingestion: meta-analysis of published experiments in animals, Toxins 2013, 5, 396-430.
Guerre, P., Fusariotoxins in Avian Species: Toxicokinetics, Metabolism and Persistence in Tissues, Toxins 2015, 7, 2289-2305; doi:10.3390/toxins7062289
Hickey, G.L., Craig, P.S., Luttik, R., de Zwart, D., On the quantification of intertest variability in ecotoxicity data with application to species sensitivity distributions, Environ. Toxicol. Chem. 2012, 31(8), 1903-1910.
Jiang, S.Z., Yang, Z.B., Yang, W.R., Gao, J., Liu, F.X., Broomhead, J., Chi, F., Effects of purified zearalenone on growth performance, organ size, serum metabolites, and oxidative stress in postweaning gilts, J. Anim. Sci. 2011, 89, 3008-3015.
Kędziora, S., Słotwiński, R.. Molecular mechanisms associated with recognition of pathogens by receptors of innate immunity, Post. Hig. 2009, 63, 30-38.
Krska, R., Schubert-Ullrich, P., Molinelli, A., Sulyok, M., Macdonald, S., Crews, C., Mycotoxin analysis: An update, Food Addit. Contam. 2008, 25(2), 152-163.
Lathe, R., Kotelevtsev, Y., Steroid signaling: ligand-binding promiscuity molecular symmetry, and the need for gating, Steroids 2014, 82, 14-22.
Lathe, R., Kotelevtsev, Y., Mason, J.I., Steroid promiscuity: Diversity of enzyme action, J. Steroid. Biochem. 2015, 151, 1-2.
Lun, Y., Xia, H., Zhang, Q., Yu, C., Chen, N., Li, X., Liu, S., Lei, L., Anti-inflammatory and immunosuppressive activities of 1,3-dicyclopentyl-1,2,3,6-tetrahydropyrimidine-4,5-dicarboxylic acid diethyl ester (ZL-5015), Int. Immunopharmacol. 2013, 17, 168-177.
Maresca, M., From the gut to the brain: journey and pathophysiological effects of the food-associated trichothecene mycotoxin deoxynivalenol, Toxins 2013, 5, 784-820.
Maresca, M., Fantini, J., Some food-associated mycotoxins as potential risk factors in humans predisposed to chronic intestinal inflammatory diseases, Toxicon 2010, 56, 282-294.
Maresca, M., Yahi, N., Younès-Sakr, L., Boyron, M., Caporiccio, B., Fantini, J., Both direct and indirect effects account for the proinflammatory activity of enteropathogenic mycotoxins on the human intestinal epithelium: stimulation of interleukin-8 secretion, potentiation of interleukin-1beta effect and increase in the transepithelial passage of commensal bacteria, Toxicol. Appl. Pharm. 2008, 228, 84-92.
Marroquín-Cardona, A.G., Johnson, N.M., Phillips, T.D., Hayes, A.W., Mycotoxins in a changing global environment – A review, Food Chem. Toxicol. 2014, 69, 220-230.
Netea, M.G., van de Veerdonk, F.L., Kullberg, B.J., Van der Meer, J.W., Joosten, L.A., The role of NLRs and TLRs in the activation of the inflammasome, Expert Opin. Biol. Th. 2008, 8, 1867-1872.
Pestka, J.J., Deoxynivalenol: Mechanisms of action, human exposure, and toxicological relevance, Arch. Toxicol. 2010, 84, 663-679.
Piotrowska, M., Śliżewska, K., Nowak, A., Zielonka, Ł., Żakowska, Z., Gajęcka, M., Gajęcki, M., The effect of experimental Fusarium mycotoxicosis on microbiota diversity in porcine ascending colon contents, Toxins 2014, 6, 2064-2081.
Pinton, P., Oswald, I.P., Effect of deoxynivalenol and other type B trichothecenes on the intestine: A Review, Toxins 2014, 6, 1615-1643.
Pinton, P., Tsybulskyy, D., Lucioli, J., Laffitte, J., Cllu, P., Lyazhri, F., Grosjean, F., Bracarense, A.P., Kolf-Clauw, M., Oswald, I.P., Toxicity of Deoxynivalenol and Its Acetylated Derivatives on the Intestine: Differential Effects on Morphology, Barrier Function, Tight Junction Proteins, and Mitogen-Activated Protein Kinases, Toxicol. Sci. 2012, 130(1), 180-190.
Schwartz-Zimmermann, H.E., Fruhmann, P., Dänicke, S., Wiesenberger, G., Caha, S., Weber, J., Berthiller, F., Metabolism of Deoxynivalenol and Deepoxy-Deoxynivalenol in Broiler Chickens, Pullets, Roosters and Turkeys, Toxins 2015, 7, 4706-4729; doi:10.3390/toxins7114706
Smith, M.C., Madec, S., Coton, E., Hymery, N., Natural Co-Occurrence of Mycotoxins in Foods and Feeds and Their in vitro Combined Toxicological Effects, Toxins 2016, 8, 94; doi:10.3390/toxins8040094
Strugala, V., Dettmar, P.W., Pearson, J.P., Thickness and continuity of the adherent colonic mucus barrier in active and quiescent ulcerative colitis and Crohn’s disease, Inter. J. Clin. Pract. 2008, 62, 762-769.
Tian, X., Shao, Y., Wang, Z., Guo, Y., Effects of dietary yeast β-glucans supplementation on growth performance, gut morphology, intestinal Clostridium perfringens population and immune response of broiler chickens challenged with necrotic enteritis, Anim. Feed Sci. Tech. 2016, 215, 144–155.
Vandenberg, L.N., Colborn, T., Hayes, T.B., Heindel, J.J., Jacobs, D.R., Lee, D.-H., Shioda, T., Soto, A.M., vom Saal, F.S., Welshons, W.V., Zoeller, R.T., Myers, J.P., Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses, Endoc. Rev. 2012, 33, 378-455.
Waśkiewicz, A., Beszterda, M., Kostecki, M., Zielonka, Ł., Goliński, P., Gajęcki, M., Deoxynivalenol in gastrointestinal tract of immature gilts under per os toxin application, Toxins 2014, 6, 973-987.
Wen, K., Azevedo, M.S.P., Gonzalez, A., Zhang, W., Saif, L.J., Li, G., Yousef, A., Yuan, L., Toll-like receptor and innate cytokine responses induced by lactobacilli colonization and human rotavirus infection in gnotobiotic pigs, Vet. Immunol. Immunop. 2009, 15, 3-4.
Yanga, W., Yu, M., Fu, J., Bao, W., Wang, D., Hao, L., Yao, P., Nüssler, A.K., Yan, H., Liu, L., Deoxynivalenol induced oxidative stress and genotoxicity in human peripheral blood lymphocytes, Food Chem. Toxicol. 2014, 64, 383-396.
Zachariasova, M., Dzumana, Z., Veprikova, Z., Hajkovaa, K., Jiru, M., Vaclavikova, M., Zachariasova, A., Pospichalova, M., Florian, M., Hajslova, J., Occurrence of multiple mycotoxins in European feeding stuffs, assessment of dietary intake by farm animals, Anim. Feed Sci. Technol. 2014, 193, 124-140.
Zhang, L., Lu, L., Li, S., Zhang, G., Ouyang, L., Robinson, K., Tang, Y., Zhu, Q., Li, D., Hu, Y., Liu, Y., 1,25-Dihydroxyvitamin-D3 Induces Avian β-Defensin Gene Expression in Chickens, PLoS ONE 2016, 11(5): e0154546. doi:10.1371/ journal.pone.0154546
Zielonka, Ł., Jakimiuk, E., Obremski, K., Gajęcka, M., Dąbrowski, M., Gajęcki, M., An evaluation of the proliferative activity of immunocompetent cells in the jejunal and iliac lymph nodes of prepubertal female wild boars diagnosed with mixed mycotoxicosis, Bull. Vet. Inst. Pulawy 2015, 59, 197-203, DOI: 10.1515/bvip-2015-0030

 

Zielonka, Ł., Zwierzchowski, W., Obremski, K., Gajęcka, M., Jakimiuk, E., Gajęcki, M., Evaluation of selected indicators of immune response (IL-1β, IL-4, IL-6, SAA, and Hp) in pigs fed a diets containing deoxynivalenol, T-2 toxin, and zearalenone, Bull. Vet. Inst. Pulawy 2010, 54, 631-635.

Wstecz

Partnerzy

Zakup czasopisma