Next Article in Journal
A Natural Variation of Fumonisin Gene Cluster Associated with Fumonisin Production Difference in Fusarium fujikuroi
Next Article in Special Issue
Time-Dependent Changes in the Intestinal Microbiome of Gilts Exposed to Low Zearalenone Doses
Previous Article in Journal
The Pressure of Fusarium Disease and Its Relation with Mycotoxins in The Wheat Grain and Malt
Previous Article in Special Issue
Occupational Exposure to Mycotoxins in Swine Production: Environmental and Biological Monitoring Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Novel Modified Hydrated Sodium Calcium Aluminosilicate (HSCAS) Adsorbent Can Effectively Reduce T-2 Toxin-Induced Toxicity in Growth Performance, Nutrient Digestibility, Serum Biochemistry, and Small Intestinal Morphology in Chicks

1
Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
2
Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province, Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
3
Animal Production Department, Faculty of Agriculture, Benha University, Benha 13736, Egypt
4
Sichuan Green Food Development Center, Chengdu 610041, China
5
Tianjin Animal Disease Prevention and Control Center, Tianjin 300402, China
6
Jiangsu Aomai Bio-Technology Co., Ltd., Nanjing 211226, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Toxins 2019, 11(4), 199; https://doi.org/10.3390/toxins11040199
Submission received: 5 March 2019 / Revised: 28 March 2019 / Accepted: 2 April 2019 / Published: 2 April 2019
(This article belongs to the Special Issue Mycotoxin Exposure and Related Diseases)

Abstract

:
The objective of this study was to evaluate the ability of a modified hydrated sodium calcium aluminosilicate (HSCAS) adsorbent to reduce the toxicity of T-2 toxin in broilers. Ninety-six one-day-old male broilers were randomly allocated into four experimental groups with four replicates of six birds each. The four groups, 1–4, received a basal diet (BD), a BD plus 6.0 mg/kg T-2 toxin, a BD plus 6.0 mg/kg T-2 toxin with 0.05% modified HSCAS adsorbent, and a BD plus 0.05% modified HSCAS adsorbent, respectively, for two weeks. Growth performance, nutrient digestibility, serum biochemistry, and small intestinal histopathology were analyzed. Compared to the control group, dietary supplementation of T-2 toxin decreased (p < 0.05) body weight gain, feed intake, and the feed conversion ratio by 11.4–31.8% during the whole experiment. It also decreased (p < 0.05) the apparent metabolic rates of crude protein, calcium, and total phosphorus by 14.9–16.1%. The alterations induced by T-2 toxin were mitigated (p < 0.05) by the supplementation of the modified HSCAS adsorbent. Meanwhile, dietary modified HSCAS adsorbent supplementation prevented (p < 0.05) increased serum aspartate aminotransferase by T-2 toxin at d 14. It also prevented (p < 0.05) T-2 toxin-induced morphological changes and damage in the duodenum, jejunum, and ileum of broilers. However, dietary supplementation of the modified HSCAS adsorbent alone did not affect (p > 0.05) any of these variables. In conclusion, these findings indicate that the modified HSCAS adsorbent could be used against T-2 toxin-induced toxicity in growth performance, nutrient digestibility, and hepatic and small intestinal injuries in chicks.
Key Contribution: This study reveals that the modified HSCAS adsorbent, AmdetoxTM, could be used as a promising adsorbent for the counteracting of T-2 toxin in practice.

1. Introduction

Trichothecenes are secondary fungal metabolites largely produced by Fusarium, Trichoderma, and Mycothecium species [1]. T-2 toxin has shown the highest toxicity of the commonly tested type A trichothecenes [1]. T-2 toxin has been detected in grains and animal feed all over the world [2,3]. Previous reports have shown that about 20–70% of European cereal samples, including maize, barley, and wheat, have T-2 toxin [2,3]. T-2 toxin can be quickly absorbed in the intestinal tract, and then causes severe damage to various organs of animals, especially the liver and the digestive system [4,5]. After consumption, T-2 toxin is known to reduce feed intake and weight gain in mice [6], broiler chickens [7], and pigs [8]. Furthermore, many studies have considered T-2 toxin impacts on the relative weight of organs [9], serum biochemistry [10], restrained protein synthesis [1], cell apoptosis [11], and the suppression of immune functions [1]. Therefore, the development of effective strategies to reduce T-2 toxicity has attracted much interest over the past few decades.
Generally, there are several methods to reduce the harmful effects of T-2 toxin, including physical, chemical, and biological procedures. Irradiation provides intense energy to break down T-2 toxin in grains [12], and strong alkaline solutions can inhibit T-2 toxin biological activity [13]. Furthermore, enzymatic treatment can also degrade T-2 toxin, destroying its 12,13-epoxide ring [14]. However, methods to remove T-2 toxin from feed and food can be unstable and expensive and can further affect grain quality [15]. Physical adsorption is more effective and directly detoxifies mycotoxins by inhibiting absorption in the gastrointestinal tract [16], but there is a lack of efficient adsorbent for T-2 toxin and deoxynivalenol (DON) [17]. Previous studies have reported that adsorbents contain aluminosilicates, such as bentonite [18], montmorillonite [19], and zeolite [20], displaying an ability to effectively protect against zearalenone [21], aflatoxin B1 (AFB1), and fumonisin B1 (FB1) [22] in several farm and experimental animals. Hydrated sodium calcium aluminosilicate (HSCAS) is a material obtained by using natural zeolite ore: It is purified through crushing, screening, and high-temperature treatment, allowing the structure to increase its size, surface area, and adsorption volume [23]. AmdetoxTM is an adsorbent that mainly contains HSCAS whose surface is modified by cetylpyridinium chloride and the intercalation of β-glucan. The modified HSCAS adsorbent increases surface area and might be able to increase adsorbing mycotoxins and avoid adsorbing the nutrients in feed. The objective of this study was to determine the ameliorative effects of AmdetoxTM detoxification on the toxicity induced by T-2 toxin.

2. Results

2.1. Growth Performance

Growth performance results are presented in Table 1. Nonsignificant differences in initial body weight were observed among the four groups (p > 0.05, Table 2). Compared to the control (group 1), dietary T-2 supplementation (group 2) decreased (p < 0.05) body weight gain and feed intake by 15.3–31.8% and 12.4–20.6%, respectively, during d 1–7, d 8–14, and d 1–14, while it increased (p < 0.05) feed intake by 11.4–15.9% during d 8–14 and d 1–14. Although dietary supplementation of AmdetoxTM (group 3) did not alleviate T-2 toxin-induced (group 2) adverse effects on body weight gain and feed intake in d 1–7, dietary supplementation of AmdetoxTM (group 3) alleviated T-2 toxin-induced (group 2) adverse effects on body weight gain and feed intake by 38.6–46.6% and 33.0–36.0%, respectively, during d 8–14 and d 1–14. Meanwhile, dietary supplementation of AmdetoxTM (group 3) mitigated the reduced feed/gain induced by T-2 toxin (group 2) throughout the experiment. Notably, dietary-supplemented AmdetoxTM alone (group 4) did not affect (p > 0.05) body weight gain, feed intake, and feed/gain compared to the control (group 1) throughout the experiment. Notably, the variation in growth performance was very low for each treatment, indicating that in the current study, the results from four replicates per treatment might be reliable.

2.2. Apparent Metabolic Rate

The nutrient metabolic rate results are shown in Table 2. Although dietary supplementation of T-2 toxin (group 2) did not affect (p > 0.05) the apparent metabolic rate of gross energy, it decreased (p < 0.05) the apparent metabolic rate of crude protein, calcium, and total phosphorus by 14.9%, 18.0%, and 16.1%, respectively. Notably, the changes in the apparent metabolic rates of crude protein and total phosphorus induced by T-2 toxin were alleviated by dietary supplementation of AmdetoxTM (group 3) when compared to the control (group 1). In addition, dietary-supplemented AmdetoxTM alone (group 4) did not affect (p > 0.05) the apparent metabolic rates of gross energy, crude protein, calcium, and total phosphorus compared to the control (group 1).

2.3. Serum Biochemistry and Histopathology

Serum biochemistry results are presented in Table 3. After two weeks of experimental treatments, although T-2 toxin (group 2) did not affect (p > 0.05) serum alanine aminotransferase (ALT), total protein (TP), and albumin (ALB), it increased (p < 0.05) aspartate aminotransferase (AST) (17.7%) relative to the control (group 1). Strikingly, this change was inhibited by dietary supplementation of AmdetoxTM (group 3). Dietary-supplemented AmdetoxTM alone (group 4) did not affect (p > 0.05) these serum biochemistry variables compared to the control (group 1). Additionally, the histological results showed that dietary T-2 toxin supplementation induced intestinal injury (Figure 1). Specifically, compared to the control (group 1), T-2 toxin (group 2) induced severe degeneration, necrosis, and desquamation of the villous epithelial cells; and increased inflammatory cells in the intestinal mucosa, congestion in the intestinal lamina propria, goblet cell hyperplasia in the intestinal gland, and/or edema and thickening in the serosa, with an infiltration of numerous lymphoid cells in the duodenum (Figure 1A), jejunum (Figure 1B), and ileum (Figure 1C). Intriguingly, dietary supplementation of AmdetoxTM (group 3) prevented T-2 toxin-induced (group 2) injury in the small intestine. In contrast, intestinal morphology was not affected by the supplementation of AmdetoxTM alone (group 4).

3. Discussion

The current study demonstrates that the modified HSCAS adsorbent could effectively counteract T-2 toxin-induced adverse effects on broilers. Chick consumption of T-2 toxin reduced body weight, feed intake, and feed conversion, which was in accordance with previous studies [7,24]. The poor growth performance of broilers, induced by T-2 toxin, was further proven to be associated with decreased metabolic rates of crude protein, calcium, and total phosphate in chicks in the current study. These outcomes are similar to previous studies that showed that Fusarium toxins can negatively affect nutrient digestibility in chicks [24,25]. Interestingly, the current study showed that dietary supplementation of 0.05% AmdetoxTM successfully reduced the negative effect induced by T-2 toxin. Notably, no negative effects in these productive parameters were found between broilers in the experimental group supplemented with AmdetoxTM alone and the control group, indicating that AmdetoxTM was nontoxic and safe.
The small intestine, including the duodenum, jejunum, and ileum, is the major part where most nutrient digestion and absorption takes place [26]. In this study, the pathological results showed that T-2 toxin caused serious small intestinal detriment in broilers, and these outcomes are in agreement with previous reports that T-2 toxin could induce intestine damage, thus decreasing nutrient utilization efficiency, as described above [27]. Interestingly, T-2 toxin-induced injury in the small intestine was mitigated by AmdetoxTM supplementation. Furthermore, the administration of T-2 toxin alone increased AST activity compared to the control group. The activity of serum enzymes such as AST and ALT, and concentrations of serum ALB and TP, have been described as valuable parameters of hepatic function and injury [28]. These outcomes are similar to previous studies that provided evidence that liver injury was induced by T-2 toxins in chicks [29,30]. However, some other reports have shown that T-2 toxin did not affect these parameters in poultry [31], pigs [32], and hamsters [33]. This discrepancy could be attributed to different experimental conditions, including exposure doses, duration, and animal species. The results obtained from the current study show that serum biochemical changes could be ameliorated by AmdetoxTM supplementation. Taken together, these results are in agreement with previous studies that reported that growth retardation induced by T-2 toxin was mainly due to induced intestinal and hepatic injury. Dietary AmdetoxTM supplementation, however, prevented T-2 toxin-induced poor growth performance, which was associated with the inhibition of intestinal and hepatic injury.
HSCAS adsorbent is a commercial feed additive that has been proven to have an effective ability to adsorb AFB1 [34,35], while several studies have reported that general HSCAS adsorbents could not effectively adsorb Trichothecenes such as T-2 toxin [36] and DON [37]. Interestingly, artificially modified substances of zeolites [38], glucomannan [39], montmorillonite [40], and diatomaceous earth [30] can enhance the adsorption and detoxification of mycotoxins. Therefore, a modified HSCAS adsorbent product, AmdetoxTM, was developed to prevent the harmful effects of T-2 toxin. This HSCAS adsorbent is surface-modified with cetylpyridinium chloride based on natural bentonite and is intercalated with yeast β-glucan. After the special bentonite interlayer cation and water molecules are replaced by modifiers, the spacing between the particles is significantly increased, and the surface of the particles changes from hydrophilic to hydrophobic. Therefore, the modified HSCAS adsorbent has a larger adsorption capacity and lipophilic hydrophobicity, and it could effectively adsorb various mycotoxins in the feed. As expected, the modified HSCAS adsorbent displayed an effective ability to prevent the negative effect of T-2 toxin in broilers. Previous studies have shown that 0.25% adsorbent or 2% polymeric glucomannans could alleviate the harmful effect of 1.0–2.0 mg/kg T-2 toxin [41,42]. An addition of only 0.05% AmdetoxTM to a diet could reduce the negative effect induced by 6.0 mg/kg T-2 toxin, indicating that the modified HSCAS adsorbent was quite effective. Meanwhile, consistent with previous studies [19,43], the modified HSCAS adsorbent did not affect nutrient metabolic rates and growth performance in the current study.

4. Conclusions

In summary, these data indicate that the modified HSCAS adsorbent, AmdetoxTM, could be used as a promising adsorbent for the detoxification of T-2 toxin in practice.

5. Materials and Methods

5.1. Birds, Treatment, and Growth Performance

Ninety-six one-day-old Cobb-500 male broiler chicks with similar body weights were randomly allocated into four groups with four replicates of six birds/cage. The four groups of birds were allowed free access to water and were fed a corn-soybean-based diet (BD, group 1) formulated to meet the nutritional requirements of broilers (National Research Council (NRC), 1994; Table 4) or BD supplemented with 6.0 mg/kg T-2 toxin (Pribolab Pte. Ltd., Singapore) (group 2), 6.0 mg/kg T-2 toxin with 0.05% of AmdetoxTM (Jiangsu Aomai Bio-technology Co., Ltd. Nanjing, China) (group 3), or 0.05% of AmdetoxTM (group 4). The T-2 toxin-contaminated diet was made through a stepwise dilution method. Briefly, 150 mg of T-2 toxin was dissolved in 50 mL of ethanol and then mixed with 500 g of corn. The mixed sample was subsequently dried at 65 °C in an oven, which was used to make the T-2 toxin-contaminated diets. The mortality of the birds was monitored daily, whereas feed intake and body weight were measured weekly. The total excreta of each pen were collected during d 8–14 to measure the apparent metabolic rates of gross energy, crude protein, calcium, and total phosphorus of chicks. The experiment lasted for two weeks. At the end of the experiment, eight birds from each treatment group were killed to collect blood and small intestine (duodenum, jejunum, and ileum) for serological and intestinal histological examinations, as previously described [44].

5.2. Serum Biochemistry and Histopathology

The serum was prepared by centrifugation of the blood at 1000× g at 4 °C for 10 min. The activities of ALT and AST, as well as the concentrations of TP and ALB, in the serum were measured with the use of an automatic biochemistry analyzer (Beckman Synchron CX4 PRO, CA, USA), as previously described [45]. The duodenum, jejunum, and ileum (n = 4/group) were removed, fixed in neutral-buffered 10% formalin, embedded in paraffin, sectioned at 5 μm, stained with hematoxylin and eosin (H&E), and examined microscopically for histopathology [46]. Briefly, intestinal tissue was examined for each chick by light microscopy for described lesions: Degeneration, necrosis, and desquamation of the villous epithelial cells; and edema, thickening in the serosa, or both. Sections with no, slight, moderate, or intense presence of lesions were given scores of 0, 1, 2, and 3, respectively [46].

5.3. Apparent Metabolic Rate

The apparent metabolic rates of gross energy, crude protein, calcium, and total phosphorus of chicks were measured and calculated as previously described [47]. Gross energy was analyzed using an adiabatic bomb calorimeter standardized (IKA C2000) with benzoic acid. Calcium, total phosphorus, and crude protein were measured following the permanganate titration method 990.03 (AOAC, 2000), the colorimetric determination method 985.01 (AOAC, 1990), and the Kjeldahl digestion method 984.13 (AOAC, 1990), respectively [48].

5.4. Statistical Analysis

A one-way ANOVA was used to test the main effects of the dietary effect. A Bonferroni t-test followed for multiple mean comparisons if there was a main effect. Data are presented as means ± SD, and the significance level was set at p < 0.05. The analyses were conducted using the SPSS Statistics 19.0 package (SPSS Inc., IBM, New York, Ny, USA).

5.5. Ethical

This research was approved by Scientific Ethic Committee of Huazhong Agricultural University on 20 March 2017. The project identification code is HZAUCH-2017-008.

Author Contributions

L.-H.S., J.-T.W., and Q.L. conceived and designed the experiments; K.-T.W., J.-T.W. and H.S. performed the experiments; J.-T.W. and K.-T.W. analyzed the data; Q.L., N.-Y.Z., D.-S.Q., J.-F.D., and Y.L. contributed reagents/materials/analysis tools; J.-T.W., K.-T.W., M.M.K. and L.-H.S. wrote the paper.

Funding

This research was funded by the National Key Research and Development Program of China, Project (2018YFD0500601 and 2016YFD0501207); the Open Project of the Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei Province (KLAMEMB-2017-05); the Innovation Group of the Hubei Natural Science Foundation (2018CFA020); and a gift from Jiangsu Aomai Bio-technology Co., Ltd.

Conflicts of Interest

All authors have read and approved the final manuscript and declare that no competing interests exist.

References

  1. Wu, Q.; Wang, X.; Nepovimova, E.; Miron, A.; Liu, Q.; Wang, Y.; Su, D.; Yang, H.; Li, L.; Kuca, K. Trichothecenes: Immunomodulatory effects, mechanisms, and anti-cancer potential. Arch. Toxicol. 2017, 91, 3737–3785. [Google Scholar] [CrossRef]
  2. Gareis, M.; Zimmermann, C.; Schothorst, R.C. Collection of Occurrence Data of Fusarium Toxin in Food and Assessment of Dietary Intake by the Population of EU Member States. 2003. Available online: https://ci.nii.ac.jp/naid/20001046845/ (accessed on 2 April 2019).
  3. Rasmussen, P.H.; Ghorbani, F.; Berg, T. Deoxynivalenol and other Fusarium toxins in wheat and rye flours on the Danish market. Food Addit. Contam. 2003, 20, 396. [Google Scholar] [CrossRef] [PubMed]
  4. Makowska, K.; Obremski, K.; Gonkowski, S. The impact of T-2 toxin on vasoactive intestinal polypeptide-like immunoreactive (VIP-LI) nerve structures in the wall of the porcine stomach and duodenum. Toxins 2018, 10, 138. [Google Scholar] [CrossRef] [PubMed]
  5. Makowska, K.; Obremski, K.; Zielonka, L.; Gonkowski, S. The influence of low doses of zearalenone and t-2 toxin on calcitonin gene related peptide-like immunoreactive (CGRP-LI) neurons in the ens of the porcine descending colon. Toxins 2017, 9, 98. [Google Scholar] [CrossRef]
  6. Zhang, J.; Zhang, H.; Liu, S.; Wu, W.; Zhang, H. Comparison of anorectic potencies of type A trichothecenes T-2 toxin, HT-2 toxin, Diacetoxyscirpenol, and Neosolaniol. Toxins 2018, 10, 179. [Google Scholar] [CrossRef] [PubMed]
  7. Chi, M.S.; Mirocha, C.J.; Kurtz, H.J.; Weaver, G.; Bates, F.; Shimoda, W. Subacute toxicity of T-2 toxin in broiler chicks. Poult. Sci. 1977, 56, 306. [Google Scholar] [CrossRef] [PubMed]
  8. Meissonnier, G.M.; Laffitte, J.; Raymond, I.; Benoit, E.; Cossalter, A.M.; Pinton, P.; Bertin, G.; Oswald, I.P.; Galtier, P. Subclinical doses of T-2 toxin impair acquired immune response and liver cytochrome P450 in pigs. Toxicology 2008, 247, 46–54. [Google Scholar] [CrossRef]
  9. Kubena, L.F.; Edrington, T.S.; Harvey, R.B.; Buckley, S.A.; Phillips, T.D.; Rottinghaus, G.E.; Casper, H.H. Individual and combined effects of fumonisin B1 present in Fusarium moniliforme culture material and T-2 toxin or deoxynivalenol in broiler chicks. Poult. Sci. 1997, 76, 1239–1247. [Google Scholar] [CrossRef]
  10. Manafi, M.; Mohan, K.; Ali, M.N. Effect of ochratoxin A on coccidiosis-challenged broiler chicks. World Mycotoxin J. 2011, 4, 177–181. [Google Scholar] [CrossRef]
  11. Wu, J.; Zhou, Y.; Yuan, Z.; Yi, J.; Chen, J.; Wang, N.; Tian, Y. Autophagy and apoptosis interact to modulate T-2 toxin-induced toxicity in liver cells. Toxins 2019, 11, 179. [Google Scholar] [CrossRef] [PubMed]
  12. Hooshmand, H.; Klopfenstein, C.F. Effects of gamma irradiation on mycotoxin disappearance and amino acid contents of corn, wheat, and soybeans with different moisture contents. Plant Foods Hum. Nutr. 1995, 47, 227–238. [Google Scholar] [CrossRef] [PubMed]
  13. Faifer, G.C.; Velazco, V.; Godoy, H.M. Adjustment of the conditions required for complete decontamination of T-2 toxin residues with alkaline sodium hypochlorite. Bull. Environ. Contam. Toxicol. 1994, 52, 102. [Google Scholar] [CrossRef] [PubMed]
  14. Diaz, G.J.; Cortes, A.; Roldan, L. Evaluation of the efficacy of four feed additives against the adverse effects of T-2 toxin in growing broiler chickens. J. Appl. Poult. Res. 2005, 14, 226–231. [Google Scholar] [CrossRef]
  15. Piva, A.; Galvano, F. Nutritional approaches to reduce the impact of mycotoxins. In Biotechnology in the Feed Industry; Nottingham University Press: Nottingham, UK, 1999. [Google Scholar]
  16. Huwig, A.; Freimund, S.; Käppeli, O.; Dutler, H. Mycotoxin detoxication of animal feed by different adsorbents. Toxicol. Lett. 2001, 122, 179. [Google Scholar] [CrossRef]
  17. Gerez, J.; Buck, L.; Marutani, V.; Calliari, C.; Bracarense, A. Low levels of chito-oligosaccharides are not effective in reducing deoxynivalenol toxicity in swine jejunal explants. Toxins 2018, 10, 276. [Google Scholar] [CrossRef] [PubMed]
  18. Santurio, J.M.; Mallmann, C.A.; Rosa, A.P.; Appel, G.; Heer, A.; Dageforde, S.; Bottcher, M. Effect of sodium bentonite on the performance and blood variables of broiler chickens intoxicated with aflatoxins. Br. Poult. Sci. 1999, 40, 115–119. [Google Scholar] [CrossRef]
  19. Desheng, Q.; Fan, L.; Yanhu, Y.; Niya, Z. Adsorption of aflatoxin B1 on montmorillonite. Poult. Sci. 2005, 84, 959–961. [Google Scholar] [CrossRef] [PubMed]
  20. Miazzo, R.; Rosa, C.A.; De Queiroz, C.E.; Magnoli, C.; Chiacchiera, S.M.; Palacio, G.; Saenz, M.; Kikot, A.; Basaldella, E.; Dalcero, A. Efficacy of synthetic zeolite to reduce the toxicity of aflatoxin in broiler chicks. Poult. Sci. 2000, 79, 1–6. [Google Scholar] [CrossRef]
  21. Abbes, S.; Salah-Abbes, J.B.; Ouanes, Z.; Houas, Z.; Othman, O.; Bacha, H.; Abdel-Wahhab, M.A.; Oueslati, R. Preventive role of phyllosilicate clay on the immunological and biochemical toxicity of zearalenone in Balb/c mice. Int. Immunopharmacol. 2006, 6, 1251–1258. [Google Scholar] [CrossRef]
  22. Mitchell, N.J.; Xue, K.S.; Lin, S.; Marroquin-Cardona, A.; Brown, K.A.; Elmore, S.E.; Tang, L.; Romoser, A.; Gelderblom, W.C.; Wang, J.S.; et al. Calcium montmorillonite clay reduces AFB1 and FB1 biomarkers in rats exposed to single and co-exposures of aflatoxin and fumonisin. J. Appl. Toxicol. 2014, 34, 795–804. [Google Scholar] [CrossRef]
  23. Girish, C.K.; Devegowda, G. Efficacy of Glucomannan-containing Yeast Product (Mycosorb 짰) and Hydrated Sodium Calcium Aluminosilicate in Preventing the Individual and Combined Toxicity of Aflatoxin and T-2 Toxin in Commercial Broilers. Asian-Australas. J. Anim. Sci. 2006, 19, 877–883. [Google Scholar] [CrossRef]
  24. Danicke, S.; Matthes, S.; Halle, I.; Ueberschar, K.H.; Doll, S.; Valenta, H. Effects of graded levels of Fusarium toxin-contaminated wheat and of a detoxifying agent in broiler diets on performance, nutrient digestibility and blood chemical parameters. Br. Poult. Sci. 2003, 44, 113–126. [Google Scholar] [CrossRef]
  25. Osborne, D.J.; Huff, W.E.; Hamilton, P.B.; Burmeister, H.R. Comparison of ochratoxin, aflatoxin, and T-2 toxin for their effects on selected parameters related to digestion and evidence for specific metabolism of carotenoids in chickens. Poult. Sci. 1982, 61, 1646–1652. [Google Scholar] [CrossRef] [PubMed]
  26. Vermeulen, B.; Backer, P.D.; Remon, J.P. Drug administration to poultry. Adv. Drug Deliv. Rev. 2002, 54, 795–803. [Google Scholar] [CrossRef]
  27. Yang, J.H.; Chen, H.Y.; Han, W.; Zhao, Z.H.; Sun, Z.Z.; Guo, W.B. Influence of T-2 toxin on nutrient apparent digestibility and small intestinal morphology in BALB/c Mice. Chin. J. Anim. Vet. Sci. 2015, 46, 1584–1592. [Google Scholar]
  28. Sun, L.H.; Lei, M.Y.; Zhang, N.Y.; Zhao, L.; Krumm, C.S.; Qi, D.S. Hepatotoxic effects of mycotoxin combinations in mice. Food Chem. Toxicol. 2014, 74, 289–293. [Google Scholar] [CrossRef] [PubMed]
  29. Shareef, A.M. Ineffectiveness of different adsorbents in alleviation of oral lesions induced by feeding T-2 toxin in broiler chickens. Iraqi J. Vet. Sci. 2007, 21, 75–86. [Google Scholar] [CrossRef]
  30. Shivashankar, B.P.; Narayanaswamy, H.D.; Satyanarayana, M.L.; Rao, S.; Rathnamma, D.; Rathnamma, H.K.; Sridhar, N.B. Effect of diatomaceous earth on performance, internalorgans and biochemical alterations in T-2 toxicosis of broiler chickens. J. Cell Tissue Res. 2015, 15, 4983–4988. [Google Scholar]
  31. Kutasi, J.; Papp, Z.; Jakab, L.; Brydl, E.; Rafai, P. Deactivation of T-2 toxin in broiler ducks by biotransformation. J. Appl. Poult. Res. 2012, 21, 13–20. [Google Scholar] [CrossRef]
  32. Frankič, T.; Salobir, J.; Rezar, V. The effect of vitamin E supplementation on reduction of lymphocyte DNA damage induced by T-2 toxin and deoxynivalenol in weaned pigs. Anim. Feed Sci. Technol. 2008, 141, 274–286. [Google Scholar] [CrossRef]
  33. Rajmon, R.; Sedmikova, M.; Jilek, F.; Koubkova, M.; Barta, I.; Smerak, P. Combined effects of repeated low doses of aflatoxin B1 and T-2 toxin on the Chinese hamster. Vet. Med. 2001, 46, 301–307. [Google Scholar] [CrossRef]
  34. Chen, S.S.; Li, Y.H.; Lin, M.F. Chronic exposure to the Fusarium mycotoxin deoxynivalenol: Impact on performance, immune organ, and intestinal integrity of slow-growing chickens. Toxins 2017, 9, 334. [Google Scholar] [CrossRef]
  35. Grant, P.G.; Phillips, T.D. Isothermal adsorption of aflatoxin B1 on HSCAS clay. J. Agric. Food Chem. 1998, 46, 599–605. [Google Scholar] [CrossRef]
  36. Bailey, R.H.; Kubena, L.F.; Harvey, R.B.; Buckley, S.A.; Rottinghaus, G.E. Efficacy of various inorganic sorbents to reduce the toxicity of aflatoxin and T-2 toxin in broiler chickens. Poult. Sci. 1998, 77, 1623–1630. [Google Scholar] [CrossRef]
  37. Patterson, R.; Young, L.G. Efficacy of hydrated sodium calcium aluminosilicate, screening and dilution in reducing the effects of mold contaminated corn in pigs. Can. J. Anim. Sci. 1993, 73, 615–624. [Google Scholar] [CrossRef]
  38. Tomašević-Čanović, M.; Daković, A.; Rottinghaus, G.; Matijašević, S.; Đuričić, M. Surfactant modified zeolites–new efficient adsorbents for mycotoxins. Microporous Mesoporous Mater. 2003, 61, 173–180. [Google Scholar] [CrossRef]
  39. Reddy, N.B.; Devegowda, G.; Shashidhara, R.G. Ability of modified glucomannan to sequestrate T-2 toxin in the gastrointestinal tract of chicken. Asian-Australas. J. Anim. Sci. 2004, 17, 259–262. [Google Scholar] [CrossRef]
  40. Lemke, S.L.; Mayura, K.; Reeves, W.R.; Wang, N.; Fickey, C.; Phillips, T.D. Investigation of organophilic montmorillonite clay inclusion in zearalenone-contaminated diets using the mouse uterine weight bioassay. J. Toxicol. Environ. Health 2001, 62, 243–258. [Google Scholar] [CrossRef]
  41. Fazekas, B.; Tóthné Hajdu, E.; Tanyi, J. Effect of Myco-ad on experimental T-2 toxicosis in broiler chickens. Magyar Állatorvosok Lapja 2000, 122, 412–416. [Google Scholar]
  42. Meissonnier, G.M.; Raymond, I.; Laffitte, J.; Cossalter, A.M.; Pinton, P.; Benoit, E.; Bertin, G.; Galtier, P.; Oswald, I.P. Dietary glucomannan improves the vaccinal response in pigs exposed to aflatoxin B1 or T-2 toxin. World Mycotoxin J. 2009, 2, 161–172. [Google Scholar] [CrossRef]
  43. Abbes, S.; Ouanes, Z.S.J.; Houas, Z.; Oueslati, R.; Bacha, H.; Othman, O. The protective effect of hydrated sodium calcium aluminosilicate against haematological, biochemical and pathological changes induced by Zearalenone in mice. Toxicon 2006, 47, 567. [Google Scholar] [CrossRef]
  44. Gao, X.; Xiao, Z.H.; Liu, M.; Zhang, N.Y.; Khalil, M.M.; Gu, C.Q.; Qi, D.S.; Sun, L.H. Dietary Silymarin Supplementation Alleviates Zearalenone-Induced Hepatotoxicity and Reproductive Toxicity in Rats. J. Nutr. 2018, 148, 1209–1216. [Google Scholar] [CrossRef]
  45. Sun, L.H.; Zhang, N.Y.; Zhu, M.K.; Zhao, L.; Zhou, J.C.; Qi, D.S. Prevention of aflatoxin B1 hepatoxicity by dietary selenium is associated with inhibition of cytochrome P450 isozymes and up-regulation of 6 selenoprotein genes in chick liver. J. Nutr. 2016, 146, 655–661. [Google Scholar] [CrossRef]
  46. Denli, M.; Blandon, J.C.; Guynot, M.E.; Salado, S.; Perez, J.F. Effects of dietary AflaDetox on performance, serum biochemistry, histopathological changes, and aflatoxin residues in broilers exposed to aflatoxin B1. Poult. Sci. 2009, 88, 1444–1451. [Google Scholar] [CrossRef]
  47. Namkung, H.; Leeson, S. Effect of phytase enzyme on dietary nitrogen-corrected apparent metabolizable energy and the ileal digestibility of nitrogen and amino acids in broiler chicks. Poult. Sci. 1999, 78, 1317–1319. [Google Scholar] [CrossRef]
  48. Zhao, L.; Zhang, N.Y.; Pan, Y.X.; Zhu, L.Y.; Batonon-Alavo, D.I.; Ma, L.B.; Khalil, M.M.; Qi, D.S.; Sun, L.H. Efficacy of 2-hydroxy-4-methylthiobutanoic acid compared to DL-Methionine on growth performance, carcass traits, feather growth, and redox status of Cherry Valley ducks. Poult. Sci. 2018, 97, 3166–3175. [Google Scholar] [CrossRef]
Figure 1. Effects of dietary T-2 toxin and AmdetoxTM on histopathology of the (A) duodenum, (B) jejunum, and (C) ileum of chicks. The sections were stained with hematoxylin and eosin. Photomicrographs are shown at 100× magnification. Group 1 = basal diet; group 2 = basal diet + 6 mg/kg T-2 toxin; group 3 = basal diet + 6 mg/kg T-2 toxin + 0.05% of AmdetoxTM; group 4 = basal diet + 0.05% of AmdetoxTM.
Figure 1. Effects of dietary T-2 toxin and AmdetoxTM on histopathology of the (A) duodenum, (B) jejunum, and (C) ileum of chicks. The sections were stained with hematoxylin and eosin. Photomicrographs are shown at 100× magnification. Group 1 = basal diet; group 2 = basal diet + 6 mg/kg T-2 toxin; group 3 = basal diet + 6 mg/kg T-2 toxin + 0.05% of AmdetoxTM; group 4 = basal diet + 0.05% of AmdetoxTM.
Toxins 11 00199 g001
Table 1. Effects of T-2 toxin and AmdetoxTM on growth performance of broilers. 1
Table 1. Effects of T-2 toxin and AmdetoxTM on growth performance of broilers. 1
ItemGroup 1Group 2Group 3Group 4
Initial body weight, g/bird54.3 ± 0.454.4 ± 0.754.5 ± 0.754.0 ± 0.2
d 1 to 7
Body weight gain, g/bird138.9 ± 6.2 a117.6 ± 5.4 b115.9 ± 8.7 b136.2 ± 18.4 a
Feed intake, g/bird167.6 ± 3.8 a146.9 ± 4.8 b149.1 ± 5.3 b166.3 ± 1.8 a
Feed/gain, g/g1.21 ± 0.051.25 ± 0.081.29 ± 0.061.24 ± 0.17
d 8 to 14
Body weight gain, g/bird280.2 ± 17.0 a191.1 ± 10.1 c232.6 ± 11.9 b289.8 ± 22.7 a
Feed intake, g/bird385.2 ± 11.9 a305.8 ± 10.7 c334.4 ± 28.9 b376.3 ± 18.8 a
Feed/gain, g/g1.38 ± 0.05 b1.60 ± 0.07 a1.44 ± 0.05 b1.30 ± 0.06 b
d 1 to 14
Body weight gain, g/bird419.2 ± 21.7 a308.7 ± 8.1 c351.3 ± 11.6 b426.0 ± 9.1 a
Feed intake, g/bird552.8 ± 11.9 a452.7 ± 14.5 b485.7 ± 30.7 b542.9 ± 17.9 a
Feed/gain, g/g1.32 ± 0.05 b1.47 ± 0.02 a1.38 ± 0.05 b1.27 ± 0.02 b
a–c Means within a row lacking a common superscript differ significantly (p < 0.05). 1 Results are reported as the mean ± SD, n = 4. Group 1 = basal diet; group 2 = basal diet + 6 mg/kg T-2 toxin; group 3 = basal diet + 6 mg/kg T-2 toxin + 0.05% of AmdetoxTM; group 4 = basal diet + 0.05% of AmdetoxTM.
Table 2. Effects of T-2 toxin and AmdetoxTM on the metabolic rates of gross energy, crude protein, calcium, and total phosphorus of broilers during d 8–14. 1
Table 2. Effects of T-2 toxin and AmdetoxTM on the metabolic rates of gross energy, crude protein, calcium, and total phosphorus of broilers during d 8–14. 1
ItemGroup 1Group 2Group 3Group 4
Gross energy, %67.1 ± 3.064.1 ± 2.867.0 ± 3.666.1 ± 2.6
Crude protein, %57.6 ± 2.5 a49.0 ± 5.0 b52.5 ± 6.9 ab55.3 ± 1.7 a
Calcium, %40.5 ± 5.2 a33.2 ± 4.9 b30.6 ± 1.1 b34.9 ± 10.9 ab
Total phosphorus, %52.7 ± 6.9 a44.2 ± 4.8 b48.4 ± 8.1 ab49.3 ± 7.8 ab
a–b Means within a row lacking a common superscript differ significantly (p < 0.05). 1 Results are reported as the mean ± SD, n = 4. Group 1 = basal diet; group 2 = basal diet + 6 mg/kg T-2 toxin; group 3 = basal diet + 6 mg/kg T-2 toxin + 0.05% of AmdetoxTM; group 4 = basal diet + 0.05% of AmdetoxTM.
Table 3. Effects of T-2 toxin and AmdetoxTM on serum biochemistry of broilers at d 14. 1
Table 3. Effects of T-2 toxin and AmdetoxTM on serum biochemistry of broilers at d 14. 1
ItemGroup 1Group 2Group 3Group 4
ALT/(U/L)2.68 ± 0.122.85 ± 0.342.47 ± 0.272.80 ± 0.19
AST/(U/L)196.7 ± 6.0 b231.5 ± 5.2 a203.0 ± 3.4 b200.6 ± 7.9 b
TP/(g/L)25.7 ± 1.627.2 ± 1.228.4 ± 1.027.4 ± 1.9
ALB/(g/L)12.7 ± 1.013.1 ± 0.813.3 ± 0.613.7 ± 1.3
a–b Means within a row lacking a common superscript differ significantly (p < 0.05). 1 Results are reported as the mean ± SD, n = 8. ALT = alanine transaminase; AST = aspartate aminotransferase; TP = total protein; ALB = albumin. Group 1 = basal diet; group 2 = basal diet + 6 mg/kg T-2 toxin; group 3 = basal diet + 6 mg/kg T-2 toxin + 0.05% of AmdetoxTM; group 4 = basal diet + 0.05% of AmdetoxTM.
Table 4. Formulation and nutritional content of the basal diet. 1
Table 4. Formulation and nutritional content of the basal diet. 1
Ingredients (%)Quantity (%)
Corn54.5
Soybean meal (48%)30.4
Fish meal (64.5%)5.6
Soybean oil5.9
Calcium hydrophosphate1.2
Limestone1.0
Salt0.2
DL-methionine0.2
Premix 11.0
Approximate composition of the test diets 2
Crude protein23.00
Metabolisable energy, (MJ/kg)13.38
Lysine1.40
Methionine0.58
Methionine + cysteine0.94
Calcium1.02
Available phosphorus0.47
1 The approximate composition provides the following per-kg diet: Vitamin A, 13800 IU/kg; vitamin D, 3600 IU; vitamin E, 24 IU/kg; vitamin K3, 3.6 mg; vitamin B1, 1.5 mg; vitamin B2, 6.6 mg; vitamin B6, 3 mg; vitamin B12, 0.015 mg; folate, 0.9 mg; biotin, 0.09 mg; D-pantothenic acid, 9.6 mg; nicotinamide, 36 mg; iron, 96 mg; zinc, 53.9 mg; manganese, 71.4 mg; copper, 12 mg; selenium, 0.3 mg; iodine, 0.42 mg. 2 Calculated value.

Share and Cite

MDPI and ACS Style

Wei, J.-T.; Wu, K.-T.; Sun, H.; Khalil, M.M.; Dai, J.-F.; Liu, Y.; Liu, Q.; Zhang, N.-Y.; Qi, D.-S.; Sun, L.-H. A Novel Modified Hydrated Sodium Calcium Aluminosilicate (HSCAS) Adsorbent Can Effectively Reduce T-2 Toxin-Induced Toxicity in Growth Performance, Nutrient Digestibility, Serum Biochemistry, and Small Intestinal Morphology in Chicks. Toxins 2019, 11, 199. https://doi.org/10.3390/toxins11040199

AMA Style

Wei J-T, Wu K-T, Sun H, Khalil MM, Dai J-F, Liu Y, Liu Q, Zhang N-Y, Qi D-S, Sun L-H. A Novel Modified Hydrated Sodium Calcium Aluminosilicate (HSCAS) Adsorbent Can Effectively Reduce T-2 Toxin-Induced Toxicity in Growth Performance, Nutrient Digestibility, Serum Biochemistry, and Small Intestinal Morphology in Chicks. Toxins. 2019; 11(4):199. https://doi.org/10.3390/toxins11040199

Chicago/Turabian Style

Wei, Jin-Tao, Kun-Tan Wu, Hua Sun, Mahmoud Mohamed Khalil, Jie-Fan Dai, Ying Liu, Qiang Liu, Ni-Ya Zhang, De-Sheng Qi, and Lv-Hui Sun. 2019. "A Novel Modified Hydrated Sodium Calcium Aluminosilicate (HSCAS) Adsorbent Can Effectively Reduce T-2 Toxin-Induced Toxicity in Growth Performance, Nutrient Digestibility, Serum Biochemistry, and Small Intestinal Morphology in Chicks" Toxins 11, no. 4: 199. https://doi.org/10.3390/toxins11040199

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop