Browse > Article
http://dx.doi.org/10.5713/ajas.20.0567

Investigation of the efficacy of mycotoxin-detoxifying additive on health and growth of newly-weaned pigs under deoxynivalenol challenges  

Holanda, Debora Muratori (Department of Animal Science, North Carolina State University)
Kim, Sung Woo (Department of Animal Science, North Carolina State University)
Publication Information
Animal Bioscience / v.34, no.3_spc, 2021 , pp. 405-416 More about this Journal
Abstract
Objective: This study evaluated the effects of feeding diets naturally contaminated with deoxynivalenol (supplemental 2 mg/kg) on health, growth, and the effects of a mycotoxin-detoxifying additive in newly-weaned pigs. Methods: Thirty-six pigs (27 day-old) were housed individually and assigned to 3 treatments for 5 weeks: CON (diet containing minimal deoxynivalenol), MT (diet with supplemental 1.9 mg/kg of deoxynivalenol), and MT+D (MT + mycotoxin-detoxifying additive, 0.2%, MegaFix, ICC, São Paulo, Brazil). The mycotoxin-detoxifying additive included bentonite, algae, enzymes, and yeast. Blood was taken at week 2 and 5. Jejunal tissue were taken at week 5. Data were analyzed using the MIXED procedure of SAS. Results: Pigs fed MT+D tended to have decreased (p = 0.056) averaged daily feed intake during week 1 than MT. At week 2, serum aspartate aminotransferase/alanine aminotransferase in MT tended to be lower (p = 0.059) than CON, whereas it was increased (p<0.05) for MT+D than MT, indicating hepatic damages in MT and recovery in MT+D. Pigs fed MT had lower (p<0.05) blood urea nitrogen/creatinine than CON, supporting hepatic damage. At week 5, pigs fed MT tended to have reduced (p = 0.079) glucose than CON, whereas it was increased (p<0.05) for MT+D than MT, indicating impaired intestinal glucose absorption in MT, which was improved in MT+D. Pigs fed CON tended to have increased (p = 0.057) total glutathione in jejunum than MT, indicating oxidative stress in MT. Pigs fed MT+D had a reduced (p<0.05) proportion of Ki-67-positive cells in jejunum than MT, indicating lower enterocyte proliferation in MT+D. Conclusion: Feeding supplemental 1.9 mg/kg of deoxynivalenol reduced growth and debilitated hepatic health of pigs, as seen in leakage of hepatic enzymes, impaired nitrogen metabolism, and increase in oxidative stress. The mycotoxin-detoxifying enhanced hepatic health and glucose levels, and attenuated gut damage in pigs fed deoxynivalenol contaminated diets.
Keywords
Adsorbent; Deoxynivalenol; Enzyme; Gut Health; Pig; Probiotic;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Kim SW, Holanda DM, Gao X, Park I, Yiannikouris A. Efficacy of a yeast cell wall extract to mitigate the effect of naturally co-occurring mycotoxins contaminating feed ingredients fed to young pigs: impact on gut health, microbiome, and growth. Toxins 2019;11:633. https://doi.org/10.3390/toxins11110633   DOI
2 Huwig A, Freimund S, Kappeli O, Dutler H. Mycotoxin detoxication of animal feed by different adsorbents. Toxicol Lett 2001;122:179-88. https://doi.org/10.1016/S0378-4274(01)00360-5   DOI
3 Frobose HL, Stephenson EW, Tokach MD, et al. Effects of potential detoxifying agents on growth performance and deoxynivalenol (DON) urinary balance characteristics of nursery pigs fed DON-contaminated wheat. J Anim Sci 2017;95:327-37. https://doi.org/10.2527/jas.2016.0664   DOI
4 Alassane-Kpembi I, Pinton P, Hupe JF, et al. Saccharomyces cerevisiae boulardii reduces the deoxynivalenol-induced alteration of the intestinal transcriptome. Toxins 2018;10:199. https://doi.org/10.3390/toxins10050199   DOI
5 Springler A, Hessenberger S, Reisinger N, et al. Deoxynivalenol and its metabolite deepoxy-deoxynivalenol: multi-parameter analysis for the evaluation of cytotoxicity and cellular effects. Mycotoxin Res 2017;33:25-37. https://doi.org/10.1007/s12550-016-0260-z   DOI
6 Loi M, Fanelli F, Liuzzi VC, Logrieco AF, Mule G. Mycotoxin biotransformation by native and commercial enzymes: present and future perspectives. Toxins 2017;9:111. https://doi.org/10.3390/toxins9040111   DOI
7 Keller K, Pereyra C, Almeida T, Cavaglieri L, Rosa C. Zearalenone adsorption by a commercial seeweed meal (Lithothamnium sp.). In: Berthiller F, editor. ISM conference 2009: worldwide mycotoxin reduction in food and feed chains. Tulln, Austria: Taylor & Francis; 2010.
8 Kong C, Shin SY, Kim BG. Evaluation of mycotoxin sequestering agents for aflatoxin and deoxynivalenol: an in vitro approach. Springerplus 2014;3:346. https://doi.org/10.1186/2193-1801-3-346   DOI
9 Weaver AC, See MT, Hansen JA, et al. The use of feed additives to reduce the effects of aflatoxin and deoxynivalenol on pig growth, organ health and immune status during chronic exposure. Toxins 2013;5:1261-81. https://doi.org/10.3390/toxins5071261   DOI
10 Holanda DM, Kim SW. Efficacy of mycotoxin detoxifiers on health and growth of newly-weaned pigs under chronic dietary challenge of deoxynivalenol. Toxins 2020;12:311. https://doi.org/10.3390/toxins12050311   DOI
11 Gruber-Dorninger C, Jenkins T, Schatzmayr G. Global mycotoxin occurrence in feed: a ten-year survey. Toxins 2019;11:375. https://doi.org/10.3390/toxins11070375   DOI
12 Lessard M, Savard C, Deschene K, et al. Impact of deoxynivalenol (DON) contaminated feed on intestinal integrity and immune response in swine. Food Chem Toxicol 2015; 80:7-16. https://doi.org/10.1016/j.fct.2015.02.013   DOI
13 Official Journal of the European Union. Commission recommendation (EU) 2016/1319 of 29 July 2016 amending recommendation 2006/576/EC as regards deoxynivalenol, zearalenone and ochratoxin A in pet food (text with EEA relevance). Official Journal of the European Union; 2016.
14 Perali C, Magnoli AP, Aronovich M, Da Rocha Rosa CA, Cavaglieri LR. Lithothamnium calcareum (Pallas) Areschoug seaweed adsorbs aflatoxin B1 in vitro and improves broiler chicken's performance. Mycotoxin Res 2020;36:371-9. https://doi.org/10.1007/s12550-020-00402-y   DOI
15 Park SH, Kim J, Kim D, Moon Y. Mycotoxin detoxifiers attenuate deoxynivalenol-induced pro-inflammatory barrier insult in porcine enterocytes as an in vitro evaluation model of feed mycotoxin reduction. Toxicol In Vitro 2017;38:108-16. https://doi.org/10.1016/j.tiv.2016.10.003   DOI
16 Dersjant-Li Y, Verstegen MWA, Gerrits WJJ. The impact of low concentrations of aflatoxin, deoxynivalenol or fumonisin in diets on growing pigs and poultry. Nutr Res Rev 2003;16: 223-39. https://doi.org/10.1079/NRR200368   DOI
17 Rosero DS, Odle J, Moeser AJ, Boyd RD, van Heugten E. Peroxidised dietary lipids impair intestinal function and morphology of the small intestine villi of nursery pigs in a dose-dependent manner. Br J Nutr 2015;114:1985-92. https://doi.org/10.1017/S000711451500392X   DOI
18 Food and Drug Administration. Compliance program guidance manual. Silver Spring, MD, USA: FDA; 2005.
19 Jiang SZ, Yang ZB, Yang WR, et al. Effects of feeding purified zearalenone contaminated diets with or without clay enterosorbent on growth, nutrient availability, and genital organs in post-weaning female pigs. Asian-Australas J Anim Sci 2010; 23:74-81. https://doi.org/10.5713/ajas.2010.90242   DOI
20 Jiang SZ, Yang ZB, Yang WR, et al. Effects of purified zearalenone on growth performance, organ size, serum metabolites, and oxidative stress in postweaning gilts. J Anim Sci 2011;89: 3008-15. https://doi.org/10.2527/jas.2010-3658   DOI
21 Cheng YH, Weng CF, Chen BJ, Chang MH. Toxicity of different Fusarium mycotoxins on growth performance, immune responses and efficacy of a mycotoxin degrading enzyme in pigs. Anim Res 2006;55:579-90. https://doi.org/10.1051/animres:2006032   DOI
22 Swamy HVLN, Smith TK, MacDonald EJ, Karrow NA, Woodward B, Boermans HJ. Effects of feeding a blend of grains naturally contaminated with Fusarium mycotoxins on growth and immunological measurements of starter pigs, and the efficacy of a polymeric glucomannan mycotoxin adsorbent. J Anim Sci 2003;81:2792-803. https://doi.org/10.2527/2003.81112792x   DOI
23 Marin S, Ramos AJ, Cano-Sancho G, Sanchis V. Mycotoxins: occurrence, toxicology, and exposure assessment. Food Chem Toxicol 2013;60:218-37. https://doi.org/10.1016/j.fct.2013.07.047   DOI
24 Yu H, Zhou T, Gong J, et al. Isolation of deoxynivalenoltransforming bacteria from the chicken intestines using the approach of PCR-DGGE guided microbial selection. BMC Microbiol 2010;10:182. https://doi.org/10.1186/1471-2180-10-182   DOI
25 Chang C, Wang K, Zhou SN, Wang XD, Wu JE. Protective effect of Saccharomyces boulardii on deoxynivalenol-induced injury of porcine macrophage via attenuating p38 MAPK signal pathway. Appl Biochem Biotechnol 2017;182:411-27. https://doi.org/10.1007/s12010-016-2335-x   DOI
26 Gonzalez NFG. Aditivos anti-micotoxinas em dietas para frangos de corte [master's thesis]. Lavras, MG, Brazil: Universidade Federal de Lavras; 2013.
27 Young JC, Zhou T, Yu H, Zhu H, Gong J. Degradation of trichothecene mycotoxins by chicken intestinal microbes. Food Chem Toxicol 2007;45:136-43. https://doi.org/10.1016/j.fct.2006.07.028   DOI
28 Ahad R, Zhou T, Lepp D, Pauls KP. Microbial detoxification of eleven food and feed contaminating trichothecene mycotoxins. BMC Biotechnol 2017;17:30. https://doi.org/10.1186/s12896-017-0352-7   DOI
29 Liang Z, Ren Z, Gao S, et al. Individual and combined effects of deoxynivalenol and zearalenone on mouse kidney. Environ Toxicol Pharmacol 2015;40:686-91. https://doi.org/10.1016/j.etap.2015.08.029   DOI
30 Holanda DM, Yiannikouris A, Kim SW. Investigation of the efficacy of a postbiotic yeast cell wall-based blend on newly-weaned pigs under a dietary challenge of multiple mycotoxins with emphasis on deoxynivalenol. Toxins 2020;12:504. https://doi.org/10.3390/toxins12080504   DOI
31 Committee on Nutrient Requirements of Swine, National Research Council. Nutrition requirements of swine. 11th ed. Washington, DC, USA: National Academies Press; 2012.
32 Zhao Y, Kim SW. Oxidative stress status and reproductive performance of sows during gestation and lactation under different thermal environments. Asian-Australas J Anim Sci 2020;33:722-31. https://doi.org/10.5713/ajas.19.0334   DOI
33 Pestka JJ, Zhou HR, Moon Y, Chung YJ. Cellular and molecular mechanisms for immune modulation by deoxynivalenol and other trichothecenes: unraveling a paradox. Toxicol Lett 2004;153:61-73. https://doi.org/10.1016/j.toxlet.2004.04.023   DOI
34 Laskin JD, Heck DE, Laskin DL. The ribotoxic stress response as a potential mechanism for MAP kinase activation in xenobiotic toxicity. Toxicol Sci 2002;69:289-91. https://doi.org/10.1093/toxsci/69.2.289   DOI
35 Danicke S, Goyarts T, Doll S, Grove N, Spolders M, Flachowsky G. Effects of the Fusarium toxin deoxynivalenol on tissue protein synthesis in pigs. Toxicol Lett 2006;165:297-311. https://doi.org/10.1016/j.toxlet.2006.05.006   DOI
36 Weaver AC, See MT, Kim SW. Protective effect of two yeast based feed additives on pigs chronically exposed to deoxynivalenol and zearalenone. Toxins 2014;6:3336-53. https://doi.org/10.3390/toxins6123336   DOI
37 Weaver AC, Campbell JM, Crenshaw JD, Polo J, Kim SW. Efficacy of dietary spray dried plasma protein to mitigate the negative effects on performance of pigs fed diets with corn naturally contaminated with multiple mycotoxins. J Anim Sci 2014;92:3878-86. https://doi.org/10.2527/jas.2013-6939   DOI
38 Flannery BM, Clark ES, Pestka JJ. Anorexia induction by the trichothecene deoxynivalenol (vomitoxin) is mediated by the release of the gut satiety hormone peptide YY. Toxicol Sci 2012;130:289-97. https://doi.org/10.1093/toxsci/kfs255   DOI
39 He P, Young LG, Forsberg C. Microbial transformation of deoxynivalenol (vomitoxin). Appl Environ Microbiol 1992; 58:3857-63. https://doi.org/10.1128/AEM.58.12.3857-3863.1992   DOI
40 Kollarczik B, Gareis M, Hanelt M. In vitro transformation of the Fusarium mycotoxins deoxynivalenol and zearalenone by the normal gut microflora of pigs. Nat Toxins 1994;2:105-10. https://doi.org/10.1002/nt.2620020303   DOI
41 Sabater-Vilar M, Malekinejad H, Selman MHJ, van der Doelen MAM, Fink-Gremmels J. In vitro assessment of adsorbents aiming to prevent deoxynivalenol and zearalenone mycotoxicoses. Mycopathologia 2007;163:81-90. https://doi.org/10.1007/s11046-007-0093-6   DOI
42 Prelusky DB, Hartin KE, Trenholm HL, Miller JD. Pharmacokinetic fate of 14C-labeled deoxynivalenol in swine. Fundam Appl Toxicol 1988;10:276-86. https://doi.org/10.1016/0272-0590(88)90312-0   DOI
43 Reddy KE, Jeong JY, Song J, et al. Colon microbiome of pigs fed diet contaminated with commercial purified deoxynivalenol and zearalenone. Toxins 2018;10:347. https://doi.org/10.3390/toxins10090347   DOI
44 Chaytor AC, Hansen JA, van Heugten E, See MT, Kim SW. Occurrence and decontamination of mycotoxins in swine feed. Asian-Australas J Anim Sci 2011;24:723-38. https://doi.org/10.5713/ajas.2011.10358   DOI
45 Kim SW. Bio-fermentation technology to improve efficiency of swine nutrition. Asian-Australas J Anim Sci 2010;23:825-32. https://doi.org/10.5713/ajas.2010.r.02   DOI
46 Guaiquil VH, Vera JC, Golde DW. Mechanism of vitamin C inhibition of cell death induced by oxidative stress in glutathione-depleted HL-60 cells. J Biol Chem 2001;276:40955-61. https://doi.org/10.1074/jbc.M106878200   DOI
47 Webel DM, Finck BN, Baker DH, Johnson RW. Time course of increased plasma cytokines, cortisol, and urea nitrogen in pigs following intraperitoneal injection of lipopolysaccharide. J Anim Sci 1997;75:1514-20. https://doi.org/10.2527/1997.7561514x   DOI
48 Prelusky DB. Effect of intraperitoneal infusion of deoxynivalenol on feed consumption and weight gain in the pig. Nat Toxins 1997;5:121-5. https://doi.org/10.1002/1522-7189(1997)5:3<121::AID-NT7>3.0.CO;2-Y   DOI
49 Awad WA, Vahjen W, Aschenbach JR, Zentek J. A diet naturally contaminated with the Fusarium mycotoxin deoxynivalenol (DON) downregulates gene expression of glucose transporters in the intestine of broiler chickens. Livest Sci 2011;140:72-9. https://doi.org/10.1016/j.livsci.2011.02.014   DOI
50 Sougioultzis S, Simeonidis S, Bhaskar KR, et al. Saccharomyces boulardii produces a soluble anti-inflammatory factor that inhibits NF-κB-mediated IL-8 gene expression. Biochem Biophys Res Commun 2006;343:69-76. https://doi.org/10.1016/j.bbrc.2006.02.080   DOI
51 Niki E, Tsuchiya J, Tanimura R, Kamiya Y. Regeneration of vitamin E from α-chromanoxyl radical by glutathione and vitamin C. Chem Lett 1982;11:789-92. https://doi.org/10.1246/cl.1982.789   DOI
52 Almeida JS, Iriabho EE, Gorrepati VL, et al. ImageJS: personalized, participated, pervasive, and reproducible image bioinformatics in the web browser. J Pathol Inform 2012;3:25. https://doi.org/10.4103/2153-3539.98813   DOI