You are on page 1of 8

Ecotoxicology and Environmental Safety 194 (2020) 110420

Contents lists available at ScienceDirect

Ecotoxicology and Environmental Safety


journal homepage: www.elsevier.com/locate/ecoenv

Compound probiotics alleviating aflatoxin B1 and zearalenone toxic effects T


on broiler production performance and gut microbiota
Juan Changa,1, Tao Wanga,1, Ping Wanga, Qingqiang Yina,∗, Chaoqi Liua, Qun Zhub, Fushan Luc,
Tianzeng Gaod
a
College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
b
Henan Delin Biological Product Co., Ltd., Xinxiang, 453000, China
c
Henan Puai Feed Co., Ltd., Zhoukou, 466000, China
d
Henan Guangan Biotechnology Co., Ltd., Zhengzhou, 450001, China

ARTICLE INFO ABSTRACT

Keywords: In order to alleviate toxic effects of aflatoxins B1 (AFB1) and zearalenone (ZEA) on broiler production perfor-
Compound probiotics mance and gut microbiota, three kinds of compound probiotics (CP) were selected. The optimal ratios of Bacillus
Aflatoxin B1 subtilis, Lactobacillus casei and Candida utilis in broiler diets were 7, 5 and 6 log CFU/g for ZEA biodegradation
Zearalenone (CP1); 6, 7 and 7 log CFU/g for AFB1 biodegradation (CP2); 7, 6 and 7 log CFU/g for ZEA + AFB1 biode-
Detoxification
gradation (CP3). A total of 350 1-day-old Ross broilers were randomly divided into 7 groups. Group A was the
Microbiota
basal diet, group B-G contained ZEA, AFB1, ZEA + AFB1, ZEA + CP1, AFB1+CP2, ZEA + AFB1+CP3, re-
Broilers
spectively. The experiment showed that AFB1 or AFB1+ZEA significantly decreased broiler production perfor-
mance, damaged liver and jejunum, increased mycotoxin residues in broiler body; however, three kinds of
compound probiotics additions could alleviate mycotoxin negative effects on the above parameters (p < 0.05).
The gut microbiota analysis indicated that AFB1+ZEA increased jejunal microbial richness, but which were
decreased to almost the same level as the control group by CP3 addition. CP3 addition significantly increased
jejunal Firmicutes and Lactobacillus aviarius abundances. The correlative analysis showed that gut Lactobacillus
aviarius abundance was positively correlated with average daily gain (ADG) of broilers (p < 0.05), while
AFB1+ZEA addition decreased its relative abundance, indicating that CP3 addition increased broiler growth by
increasing Lactobacillus aviarius abundance. AFB1 and ZEA residues in broiler body were negatively correlated
with the gut beneficial bacterial abundances (p < 0.01), but positively correlated with the potentially harmful
bacterial abundances (p < 0.05), which inferred that CP3 addition could decrease mycotoxin residues through
positively regulating gut relative bacterial abundances. In conclusion, compound probiotics could keep gut
microbiota stable, degrade mycotoxins, alleviate histological lesions, increase production performance and re-
duce mycotoxin toxicity for broilers.

1. Introduction feeds to cause more serious toxic effect on human and animal health
(Freire and Sant’Ana, 2018). It has been reported that AFB1 and ZEA are
Mycotoxins such as aflatoxins B1 (AFB1) and zearalenone (ZEA) are the common harmful mycotoxins in food and feed chemistry (Wu,
toxic compounds mainly produced by Aspergillus and Fusarium species 2004). AFB1 and ZEA usually contaminate some grains together
(Luo et al., 2018). Mycotoxin contamination commonly found in feed (Kosicki et al., 2016), which may have greater hepatotoxicity than
and food commodities, climate change and the improper storage in- single mycotoxin (Sun et al., 2015); therefore, the effective co-de-
creases the pollution risk substantially (Dong et al., 2019). It is reported gradation of AFB1 and ZEA is necessary.
that approximately 25% of crops in the world have been contaminated AFB1 is well known for its teratogenic, carcinogenic and im-
by mycotoxins, leading to serious economic losses (Rice and Ross, munosuppressive effects on human and animals, it is mainly produced
1994). Moreover, several kinds of mycotoxins can co-occur in food and by Aspergillus flavus and Aspergillus parasiticus. Consumption of AFB1-

Corresponding author.

E-mail addresses: changjuan2000@126.com (J. Chang), wt9587@126.com (T. Wang), wangping850516@163.com (P. Wang), qqy1964@126.com (Q. Yin),
15093389011@163.com (C. Liu), Zhuqun1991@126.com (Q. Zhu), lfshn@126.com (F. Lu), zheng196604@126.com (T. Gao).
1
Juan Chang and Tao Wang contribute equally to this work.

https://doi.org/10.1016/j.ecoenv.2020.110420
Received 17 December 2019; Received in revised form 28 February 2020; Accepted 1 March 2020
Available online 06 March 2020
0147-6513/ © 2020 Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).
J. Chang, et al. Ecotoxicology and Environmental Safety 194 (2020) 110420

contaminated feeds by poultry may decrease feed intake and growth 69 μg/kg AFB1 and 60 μg/kg ZEA was used to prepare the mycotoxin-
performance as well as increase intestinal damage and mortality treated diets.
(Williams et al., 2011). In addition, AFB1 residue in animal edible tis-
sues and products is also a potential threat to human health.
ZEA is a regular worldwide contaminant of cereals and feeds, which 2.3. Animals and managements
is mainly produced by Fusarium strains such as F. graminearum and F.
culmorum (Luo et al., 2018). ZEA is an estrogenic mycotoxin to cause A total of 350 1-day-old Ross broilers were randomly divided into 7
reproduction disorders such as early sexual maturity, miscarriage, un- groups, 5 replicates for each group, 10 broilers (half male and half fe-
developed embryos, and so on (Green et al., 1990). male) in each replicate. All animals used in this experiment were
In order to degrade mycotoxins, many methods such as physical, managed according to the guidelines of Animal Care and Use Ethics
chemical and biological treatments have been used. The traditional Committee in Henan Agricultural University (SKLAB-B-2010-003-01).
physical and chemical methods have some drawbacks such as nutrient The feeding experiment was divided into 2 stages (1–21 d and 22–42 d).
losses, high equipment cost and bio-safety risk. Biodegradation is a The basal diets in both stages were prepared according to the re-
promising method for mycotoxin decontamination. Lactobacillus plan- commended standard (NRC, 1994). Feed and water were given to the
tarum was reported to have AFB1-degrading ability (Khaniana et al., birds ad libitum. During the experiment, body weight, feed intake,
2019), and Bacillus pumilus presented effectiveness in degrading ZEA mortality, diarrhea rates were recorded, and average daily feed intake
(Wang et al., 2017). Streptomyces sp. was found to be able to degrade (ADFI) and average daily gain (ADG) were measured. The feeding ex-
both AFB1 and ZEA (Harkai et al., 2016). Bacillus subtilis was reported to periment was designed as follows:
alleviate the negative effects of AFB1 and ZEA on egg production and Group A: Basal diet (14.45 μg/kg AFB1, 58.58 μg/kg ZEA).
feed intake (Jia et al., 2016). The previous research in our laboratory Group B: Basal diet with 500 μg/kg ZEA.
showed that the compound probiotics of Bacillus subtilis, Lactobacillus Group C: Basal diet with 50 μg/kg AFB1
casei and Candida utilis could synchronously degrade both AFB1 and Group D: Basal diet with 500 μg/kg ZEA+50 μg/kg AFB1
ZEA (Huang et al., 2018). However, few data have been reported about Group E: Basal diet with 500 μg/kg ZEA plus CP1.
the synchronous biodegradation of AFB1 and ZEA application in broi- Group F: Basal diet with 50 μg/kg AFB1 plus CP2.
lers as well as the interactions among probiotics, mycotoxins and gut Group G: Basal diet with 500 μg/kg ZEA+50 μg/kg AFB1 plus CP3.
microbiota. Note: AFB1 and ZEA used for adjusting mycotoxin concentrations in
In order to alleviate the toxic effects of AFB1, ZEA or AFB1+ZEA on the diets from group B to group G were purchased from Sigma (Sigma-
broiler growth and gut microbiota, the different kinds of compound Aldrich, St. Luis, USA).
probiotics were prepared in this study to reduce or eliminate the ha-
zards of AFB1 and ZEA in broiler production.
2.4. Determinations of nutrient metabolic rates
2. Materials and methods
At the end of feeding experiment, metabolic experiment was per-
formed using total fecal collection method. Fresh excreta were collected
2.1. Microbes and preparation
without contamination from 5 birds in each replicate for 3 d. The feed
intake and excreta were recorded daily. The fecal samples of each re-
After considering the beneficial microbes with high ability for de-
plicate were dried and ground. Crude protein, crude fat, calcium and
grading both AFB1 and ZEA, three species of microorganisms such as
phosphorus in feed and excreta were determined with Kjeldahl, ether
Bacillus subtilis (CGMCC1.0504), Lactobacillus casei (CGMCC1.2884),
extract, potassium permanganate (KMnO4) and ammonium molybdate
Candida utilis (CGMCC2.0615) were obtained from China General
((NH4)6Mo7O24), respectively (Jurgens, 1997). The AFB1 and ZEA re-
Microbiological Culture Collection Center (CGMCC). They were in-
sidues in excreta were also measured. The calculation of apparent nu-
cubated according to the published protocols (Huang et al., 2018) to
trient metabolic rates was made as follows: Nutrient apparent metabolic
make the powder by freeze drying. Based on the previous results ob-
rate (%) = 100 × (nutrient content in diet - nutrient content in ex-
tained with response surface regression design in our laboratory in vitro,
creta)/nutrient content in diet.
three kinds of compound probiotics (CP) were prepared. The optimal
ratios of Bacillus subtilis, Lactobacillus casei and Candida utilis in broiler
diets were 1 × 107, 1 × 105 and 1 × 106 colony-forming units (CFU)/g 2.5. AFB1 and ZEA residues analysis
for ZEA biodegradation (CP1); 1 × 106, 1 × 107 and 1 × 107 CFU/g
for AFB1 biodegradation (CP2); 1 × 107, 1 × 106 and 1 × 107 CFU/g At the end of feeding experiment, 3 broilers in each group were
for ZEA + AFB1 biodegradation (CP3), respectively. The above three selected to be sacrificed. The blood sample, small intestine, liver and
kinds of compound probiotics were prepared and added in broiler diets chest muscle were collected. The extraction method of AFB1 and ZEA in
according to the suggested microbial counts (CFU/g diet). excreta was the same as the above protocol. The tissues samples of
small intestine, liver and chest muscle for AFB1 and ZEA extraction
2.2. AFB 1 and ZEA determination were prepared as the following: 5 g tissues were added with 25 mL 70%
methanol (v/v), homogenized by an Ultra Turrax (T10, IKA Instrument
The contents of AFB 1 and ZEA were determined by ELISA method Company, Staufen, Germany) at 10,000 rpm for 2 min (Ghali et al.,
with RIDASCREEN® AFB1 and ZEA test kits (R-Biopharm, Darmstadt, 2008). The following procedure was the same as the above protocol.
Germany). The preparing process of samples for mycotoxin determi-
nation was as the following: About 5 g sample was ground, mixed with
25 mL 70% methanol (v/v), shaken for 3 min in a vortex mixer, filtered 2.6. Hepatic and jejunal sample preparation for microscopic analyses
with Whatman No.1 filter paper, and then 1 mL filtrate was diluted with
1 mL purified water. AFB1 and ZEA standard solutions were used for The hepatic internal lobes and the middle section of jejunum from
making the AFB1 and ZEA calibration curves (Zuo et al., 2013). The each group were taken, cut at 1 cm3, cleaned with physiological saline.
optical density was finally measured at 450 nm using ELISA 96-well The samples were fixed in formalin, dehydrated in ethanol, embedded
plate reader (BioTek Instruments Inc. Winooski, USA). The natural corn in paraffin, cut at thickness of 0.6 μm, stained with hematoxylin and
meal contaminated with 14 μg/kg AFB1 and 57 μg/kg ZEA was used to eosin, and then observed with a microscope (Olympus Co., Ltd. Tokyo,
prepare the basal diet, and the moldy corn meal contaminated with Japan).

2
J. Chang, et al. Ecotoxicology and Environmental Safety 194 (2020) 110420

2.7. Jejunal microbiota analysis production performance was presented in Tables 1 and 2. During both
feeding stages, ADFI and ADG in group E, F and G containing three
The jejunal contents of 3 broilers in group A, D and G were collected at kinds of compound probiotics and mycotoxins were better than that in
the age of 42 d, respectively; and put in liquid nitrogen for further mo- their corresponding groups (group B, C and D) with only mycotoxins
crobiota analysis. The total genomic DNA of each sample was extracted by (p < 0.05). The diarrhea rates were the highest in group C and D
the stool DNA Kit (Omega Biotek, Norcross, GA, USA) according to during both feeding stages (p < 0.05). Mortality was the highest in
manufacturer's instructions. The V3 and V4 variable regions of the bac- group B and C during the first feeding stage (1–21 d), and in group C
terial 16S rRNA genes were amplified by the universal primer pairs: 338F ( and D during the second feeding stage (22–42 d). During the second
5′-ACTCCTACGGGAGGCAGCA-3′) and 806R (5′-GGACTACHVGGGTWT- feeding stage, Table 3 showed that protein metabolic rates from high to
CTAAT-3′) (Liang et al., 2015). Reaction conditions consisted of an initial low were group B, E > group G > group A > group D, F > group C
95 °C for 3 min followed by 27 cycles of 95 °C for 30 s, 55 °C for 30 s, 72 °C (p < 0.05). Crude fat metabolic rate in group B was significantly
for 45 s, and a final extension of 72 °C for 10 min. PCR reactions were higher than that in the other groups (p < 0.05), calcium metabolic rate
conducted in 20 μL reaction mixtures containing 10 ng template DNA, in group E was significantly higher than that in group C (p < 0.05). In
0.8 μL each primer (5 μM), 2 μL 2.5 mM dNTPs, 0.4 μL FastPfu poly- general, three kinds of compound probiotics additions in broiler diets
merase, and 4 μL 5 × FastPfu buffer. The PCR products were extracted could significantly alleviate mycotoxin hazard, increase growth per-
from 2% agarose gels and purified using the AxyPrep DNA Extraction Kit formance and nutrient metabolic rates, decease diarrhea rates and
(Axygen Biosciences, CA, USA), and then quantified using QuantiFluor-ST mortality for broilers (P < 0.05).
(Promega Corporation, Madison, USA). The purified amplifications were AFB1 is one of the most predominant and highly toxic mycotoxins in
pooled in equimolar and paired-end sequenced on an Illumina platform the world, which has great adverse effects on animal production (Peng
(Biomarker Technology Co., Ltd. Beijing, China). The ACE and Chao1 et al., 2018). The previous report showed that the applications of diets
indexes were used to indicate the abundances of bacterial community, and contaminated with AFB1 significantly decreased ADG, ADFI and nu-
the Shannon and Simpson indexes were used to reflect the diversity of trient metabolic rates (Jahanian et al., 2016). In this study, the low
bacterial community. growth rates, high diarrhea rates and mortality induced by AFB1 or
AFB1+ZEA were found, in correspondence with the previous reports.
2.8. Bioinformatics analysis This research also indicated that AFB1 caused more serious damages
than ZEA for broiler growth, health and nutrient availability. The lower
The high-quality tags were clustered into the operational taxonomic ADG in the control group in the first-stage feeding experiment may be
units (OTUs) using UPARSE (version 7.1, http://drive5.com/uparse/) due to a little high AFB1 concentration in the basal diet and high sen-
with a similarity threshold of 0.97. The phylogenetic affiliation of each sitivity to AFB1 for the young broilers.
16S rRNA gene sequence was analyzed using the RDP Classifier ZEA can competitively bind to estrogen receptors to cause re-
(https://rdp.cme.msu.edu/) at 0.80 confidence level. The coverage productive problems (Conkova et al., 2003). The previous report
percentage was calculated by Good's method (1953). The abundance- showed that ZEA didn't influence growth performance of piglets (Marin
based coverage estimator (ACE), the Chao1 richness estimator and the et al., 2013), but another research indicated that small doses of ZEA
Shannon and Simpson indices were analyzed with the MOTHUR pro- stimulated body weight gain in pre-pubertal gilts when metabolic
gram (Schloss et al., 2009). processes were intensified (Rykaczewska et al., 2018). In this study,
ZEA addition alone increased young broiler growth, improved crude
protein and crude fat metabolic rates. The reason might be due to the
2.9. Statistical analyses
following two factors: (1) The broilers are more tolerant to ZEA (Fink-
Gremmels and Malekinejad, 2007); (2) ZEA is one kind of estrogen-like
Experimental data were represented as the means ± standard er-
mycotoxin, so it may be able to increase animal growth and feed
rors. The data were analyzed using the SPSS software (SPSS 19.0, IBM,
availability except for its negative effect on animal reproduction
IL, USA), one-way ANOVA was used to compare treatment means.
(Rykaczewska et al., 2018).
Differences were considered statistically significance at p < 0.05. The
The previous research showed that AFB1+ZEA had a synergistic
correlations between gut microbiota and toxin residues or ADG were
toxicity to reduce egg production and feed intake for layers, compared
assessed by Pearson's correlation test using GRAPHPAD PRISM version
with the single mycotoxin (Jia et al., 2016). It was also reported that the
5.00 (GRAPHPAD Software, CA, USA).
coexistence of AFB1 and ZEA could be more hepatotoxic than the in-
dividual one (Sun et al., 2015). However, the synergistic toxicity of
3. Results and discussion
AFB1+ZEA has not been found in this study, maybe due to the low ZEA
sensitiveness for broilers.
3.1. Broiler production performance and nutrient metabolic rates influenced
Several beneficial microorganisms have been used for reducing
by mycotoxins and compound probiotics
toxic effects on animal production. The inclusion of different compound
microorganism preparations in AFB1-contaminated diet significantly
The effect of three kinds of compound probiotics on broiler

Table 1
Effects of compound probiotics on production performance of 1-21d broilers (n = 5).
Groups Initial weight (g) Final weight (g) ADG (g) ADFI (g) ADFI/ADG Diarrhea rate (%) Mortality (%)

a d d d ab d
A 50.55 ± 0.46 561.65 ± 6.79 24.34 ± 0.34 42.40 ± 0.04 1.74 ± 0.02 2.92 ± 0.22 0
B 50.54 ± 0.38a 585.97 ± 5.91b 25.50 ± 0.28b 44.98 ± 0.01b 1.76 ± 0.02a 2.90 ± 0.40d 4
C 50.55 ± 0.21a 546.74 ± 1.82e 23.63 ± 0.08e 41.34 ± 0.07e 1.75 ± 0.01a 5.83 ± 0.67a 4
D 50.33 ± 0.16a 570.25 ± 7.77c 24.76 ± 0.37c 42.41 ± 0.01d 1.71 ± 0.02c 5.22 ± 0.56b 2
E 50.67 ± 0.15a 618.58 ± 3.51a 27.04 ± 0.17a 46.50 ± 0.03a 1.72 ± 0.01bc 3.26 ± 0.27d 0
F 50.73 ± 0.18a 572.41 ± 5.97c 24.84 ± 0.28c 42.49 ± 0.02d 1.71 ± 0.02c 4.47 ± 0.25c 0
G 50.32 ± 0.24a 577.48 ± 5.68c 25.10 ± 0.28c 43.84 ± 0.02c 1.75 ± 0.02a 4.51 ± 0.26c 0

Note: The different letters in the same columns indicate significant difference (p < 0.05), while the same letters in the same columns indicate insignificant difference
(p > 0.05).

3
J. Chang, et al. Ecotoxicology and Environmental Safety 194 (2020) 110420

Table 2
Effects of compound probiotics on production performance of 22–42 d broilers (n = 5).
Groups Initial weight (g) Final weight (g) ADG (g) ADFI (g) ADFI/ADG Diarrhea rate (%) Mortality (%)

a ab a ab ab d
A 560.00 ± 9.35 1723.00 ± 61.81 55.38 ± 2.60 116.58 ± 2.93 2.11 ± 0.06 2.29 ± 0.22 2
B 568.00 ± 7.58a 1741.00 ± 66.65a 55.86 ± 3.15a 116.48 ± 7.26ab 2.09 ± 0.06b 2.60 ± 0.37d 0
C 564.00 ± 8.22a 1644.00 ± 55.27b 51.43 ± 2.28b 112.20 ± 5.97b 2.18 ± 0.05a 3.83 ± 0.52a 8
D 566.00 ± 12.45a 1645.00 ± 95.07b 51.38 ± 4.31b 109.94 ± 9.01b 2.14 ± 0.04ab 3.86 ± 0.42a 4
E 568.00 ± 10.37a 1777.00 ± 72.33a 57.57 ± 3.17a 122.44 ± 6.68a 2.13 ± 0.02ab 2.70 ± 0.27cd 0
F 570.00 ± 6.12a 1770.00 ± 28.28a 57.14 ± 1.50a 121.06 ± 4.76a 2.12 ± 0.03ab 3.07 ± 0.26bc 0
G 557.00 ± 5.70a 1788.00 ± 61.60a 58.62 ± 3.13a 124.72 ± 5.36a 2.13 ± 0.05ab 3.47 ± 0.30ab 0

Table 3
Effect of compound probiotics on nutrient metabolic rates of broilers (%, n = 5).
Groups Crude protein Crude fat Calcium Phosphorus

A 53.67 ± 0.68cd 73.67 ± 0.01b 30.40 ± 0.83ab 39.24 ± 0.50a


B 55.17 ± 0.75ab 74.30 ± 0.60a 31.27 ± 0.41ab 39.76 ± 0.31a
C 50.38 ± 0.80e 73.51 ± 0.45b 30.05 ± 1.65b 39.52 ± 0.77a
D 52.65 ± 0.52d 73.24 ± 0.11b 30.16 ± 0.26ab 39.16 ± 0.96a
E 56.39 ± 1.22a 73.78 ± 0.29b 31.87 ± 0.88a 40.26 ± 0.66a
F 52.82 ± 0.61d 73.62 ± 0.06b 30.87 ± 0.72ab 40.47 ± 0.85a
G 54.02 ± 0.81bcd 73.32 ± 0.20b 31.04 ± 0.19ab 40.36 ± 1.16a

Note: The different letters in the same columns indicate significant difference (p < 0.05), while the same letters in the same columns indicate insignificant difference
(p > 0.05).

increased broiler growth (Zuo et al., 2013; Liu et al., 2018). Jia et al. able to reduce ZEA and AFB1 residues in broiler body.
(2016) reported that the supplementation of Bacillus subtilis in AFB1 and AFB1 and ZEA can transmit through farm animals to human food
ZEA-contaminated diet could significantly alleviate their negative ef- chain, possessing a potential threat to human health. In this study, AFB1
fects on feed intake and egg production of layers. In this study, the and ZEA contamination increased the corresponding mycotoxin re-
additions of three kinds of compound probiotics significantly relieved sidues in broilers. Moreover, AFB1+ZEA exerted synergistic effects on
the toxic effects of AFB1 and ZEA on broiler production performance, ZEA residues. Three kinds of compound probiotics additions to the ZEA
indicating their effective detoxification for broilers. and AFB1 contaminated diets were effective in reducing mycotoxin
residues. It was reported that AFB1 and ZEA were absorbed from the
gastrointestinal tract and extensively penetrated into various tissues
3.2. Effect of compound probiotics on AFB1 and ZEA residues for broilers
(Buranatragool et al., 2015). Bacillus subtilis was reported to have the
ability to germinate in animal intestinal tract, reduce AFB1 absorption
Table 4 showed that AFB1 residues in serum, excreta and various
and residues in the internal organs of broilers (Salem et al., 2018). The
tissues in group C and D were significantly higher than that in group A
compound probiotics of L. casei, B. subtilis and Pichia anomala were
(p < 0.05), indicating that the contamination of AFB1 or AFB1+ZEA in
shown to decrease AFB1 residues in serum, liver, and muscle of broilers
diets increased AFB1 residues in broilers. However, the supplement of
(Zuo et al., 2013). In this study, three kinds of compound probiotics
CP2 or CP3 in AFB1-contaminated diet in group F or G significantly
additions in broiler diets significantly decreased both AFB1 and ZEA
decreased the AFB1 residues in liver, chest muscle and small intestine,
residues in small intestine and liver, AFB1 in chest muscle, and ZEA in
compared with group C and D (p < 0.05).
serum and excreta. It is inferred that low AFB1 and ZEA residues by
Table 5 indicated that ZEA was not detected in chest muscle of
three kinds of compound probiotics additions are probably attributed to
broiler in all the groups, but ZEA residues in serum, excreta and other
mycotoxin absorption and biodegradation by the beneficial bacteria
tissues significantly increased in group B and D, compared with group A
(Salem et al., 2018; Liu et al., 2019).
(p < 0.05), indicating that the contamination of ZEA or ZEA + AFB1
in diets increased ZEA residues in broilers. In addition, ZEA residues in
all parts of group D were significantly higher than that of group B 3.3. Hepatic and jejunal villus tissue damages induced by AFB1 and ZEA
(p < 0.05), inferring that the addition of AFB1 increased ZEA residues and their restoration by CP3 addition
in serum, liver and small intestine. Nevertheless, CP1 or CP3 supple-
ment in ZEA-contaminated diets in group E or G significantly decreased The hepatic cell cords disorder, hepatocyte vacuolization and he-
ZEA residues in serum, liver, small intestine and excreta, compared patic sinusoid hypertrophy were induced by AFB1+ZEA addition. The
with the corresponding group B or D (p < 0.05). It could be concluded massive atrophy and degeneration of microvilli, low crypt depth and
that three kinds of compound probiotics additions in broiler diets were thin intestinal smooth muscle were observed by AFB1+ZEA addition.

Table 4
AFB1 residues in serum, liver, chest muscle, small intestine and excreta of broilers (μg/kg, n = 3).
Groups Serum Liver Chest muscle Small intestine Excreta

b c c d
A 1.81 ± 0.27 1.14 ± 0.37 0.91 ± 0.11 1.12 ± 0.26 2.30 ± 0.26c
B 1.83 ± 0.15b 1.13 ± 0.12c 0.80 ± 0.04c 1.12 ± 0.12d 2.36 ± 0.08c
C 2.85 ± 0.39a 9.50 ± 0.46a 3.05 ± 0.09ab 7.38 ± 0.66ab 10.01 ± 0.63ab
D 2.45 ± 0.89ab 9.13 ± 0.44ab 3.29 ± 0.19a 7.96 ± 0.40a 10.93 ± 0.65a
E 1.76 ± 0.10b 1.22 ± 0.16c 0.99 ± 0.11c 1.22 ± 0.19d 2.32 ± 0.20c
F 2.20 ± 0.43ab 8.86 ± 0.19b 3.03 ± 0.17b 6.69 ± 0.46c 9.14 ± 0.64b
G 1.71 ± 0.23b 8.85 ± 0.11b 3.02 ± 0.15b 7.02 ± 0.15bc 11.07 ± 0.97a

4
J. Chang, et al. Ecotoxicology and Environmental Safety 194 (2020) 110420

Table 5
ZEA residues in serum, liver, chest muscle, small intestine and excreta of broilers (μg/kg, n = 3).
Groups Serum Liver Chest muscle Small intestine Excreta

a d e
A ND 3.68 ± 0.31 ND 1.88 ± 0.57 38.75 ± 3.07d
B 8.72 ± 0.51b 8.72 ± 0.51bc ND 20.60 ± 0.66b 178.97 ± 7.09a
C ND 3.52 ± 0.35d ND 3.70 ± 0.43d 62.00 ± 1.35c
D 10.35 ± 0.24a 12.78 ± 1.97a ND 22.97 ± 2.31a 174.06 ± 5.70a
E ND 7.92 ± 0.29c ND 19.80 ± 0.50b 130.40 ± 9.90b
F ND 3.78 ± 0.14d ND 3.58 ± 0.40d 61.67 ± 0.48c
G 8.15 ± 0.49c 9.59 ± 0.60b ND 12.59 ± 0.60c 131.85 ± 8.38b

a
ND = Not detected.

However, CP3 addition could alleviate the toxic damages to make the It was reported that Bacillus subtilis significantly improved the pro-
hepatic and intestinal structure recover to the same status as the control portion of Proteobacteria, Bacteroidetes, Actinobacteria and Acidobacteria
group (Supplementary Fig. 1). in the jejunum of young broilers, and decreased Bacteroidetes propor-
Liver is the main target of mycotoxins. Histopathological examina- tion during growing phase (Li et al., 2019a); while Lactobacillus reuteri
tion can be used as an effective method to judge the toxic effect could increase Actinobacteria and Corynebacterium abundances, and
(Ellakany et al., 2011). Previous study showed that AFB1 caused hepatic decrease Proteobacteria and Campylobacterales abundances in broiler
architecture enlargement, bileduct hyperplasia, periportal fibrosis, he- ileum (Nakphaichit et al., 2011). The other report showed that ZEA
patocytic vacuolation and necrosis (He et al., 2013). Another report decreased the abundance of Actinobacteria and increased the abundance
showed that mycotoxin challenge had toxic effect on intestinal mor- of Cyanobacteria, Synergistetes and Proteobacteria in rabbit caecum (Li
phology with shorter and thinner villi (Jahanian et al., 2016). However, et al., 2018). It can be concluded that gut bacterial abundances at
the beneficial microbes and mycotoxin-degradation enzyme added in phylum level may be regulated by probiotics, mycotoxins, animal ages,
broiler diet could alleviate hepatic damage induced by AFB1 (Zuo et al., animal breeds, gut segments or their interactions.
2013), in agreement with this research. The reason why the compound At bacterial species level, the top 10 species of bacteria were pre-
probitotics treatment can alleviate the alteration of hepatic and jejunal sented in Fig. 1. The predominant bacterium in broiler jejunum among
histology induced by AFB1 and ZEA may be due to the low mycotoxin three groups was Lactobacillus aviarius. The relative abundance of Lac-
residues in tissues caused by CP3 addition. tobacillus aviarius in group G was higher than that in group D (p < 0.05)
and group A (p > 0.05), indicating that CP3 addition was able to in-
crease the relative abundance of Lactobacillus aviarius. The contamination
3.4. Gut microbial community influenced by compound probiotics and
of AFB1+ZEA significantly changed the relative abundances of
mycotoxins
uncultured_bacterium_g_Romboutsia, uncultured_bacterium_g_Terrisporobacter,
Clostridium_sensu_stricto_1, uncultured_bacterium_g_Lactobacillus, unculture-
The microbial abundance and diversity indices in Supplementary
d_bacterium_g_Turicibacter, bacterium_GC452011 (p < 0.05); however,
Table 1 showed that the microbial abundance (ACE and Chao1) in
CP3 addition significantly increased uncultured_bacterium_g_Lactobacillus
group D was higher than that in group A (p < 0.05); however, CP3
abundance.
addition in group G made the microbial abundance recover to the same
Previous study showed that AFB1 could induce dramatic changes in
level as group A (p > 0.05), indicating that CP3 addition could regain
the composition and metabolisms of intestinal microbiota (Wang et al.,
the normal gut microbiota disturbed by mycotoxins to keep broiler
2018). ZEA was also reported to affect the biodiversity of micro-
health. The previous research showed that ZEA could increase caecal
organisms, increase the Shannon's diversity index of gut microbiota in
microbial abundance for rabbits (Li et al., 2018). The other reports
porcine (Piotrowska et al., 2014), and decrease caecal Lactobacillus
indicated that probiotic Lactobacillus sp. added in swine and broiler
abundance of rabbits (Li et al., 2018). However, probiotics addition
diets increased microbial richness (ACE and Chao1) (Nakphaichit et al.,
could establish lactobacilli-enriched microbiota in broiler ileum and
2011; Shin et al., 2019). After considering the different results, it can be
swine feces (Nakphaichit et al., 2011; Shin et al., 2019), in agreement
concluded that microbial abundances increased by mycotoxins will be
with this research. Generally, supplementation with probiotics is con-
decreased by probiotics. In addition, Shannon and Simpson indexes
sidered as an effective method to treat the disorders of gut microbiota.
were not influenced by probiotics and mycotoxin supplements, in-
In this study, the high abundance of beneficial bacteria such as Lacto-
dicating that both of them had no effect on the diversity of bacterial
bacillus aviarius triggered by CP3 addition in group G may play an
community in broiler jejunum.
The relative abundances of bacteria at phylum level in
Supplementary Fig. 2 showed that Firmicutes, Bacteroidetes, Proteo-
bacteria, Actinobacteria and Campylobacterota were the main phyla in
broiler jejunum, accounting for over 90% of the total bacterial 16S
rDNA gene sequences in the samples. The relative abundance of Fir-
micutes in group G was significantly higher than that in group A and D
(p < 0.05), indicating that Firmicutes was easily influenced by the
external probiotics intervention. The previous researches showed that
probiotics addition could increase Firmicutes abundance (Li et al.,
2019a; Shin et al., 2019), corresponding with this study. The relative
abundances of Bacteroidetes, Proteobacteria and Actinobacteria in group
G were significantly lower than that in group D (p < 0.05). The re-
lative abundances of Actinobacteria, Fusobacteria, Cyanobacteria, Chlor-
oflexi, Acidobacteria and Gemmatimonadetes in group D were sig-
nificantly lower than that in group A (p < 0.05), while the relative
abundances of Fusobacteria, Cyanobacteria and Chloroflexi were regu-
lated to the same levels as group A by CP3 addition in group G. Fig. 1. Relative abundances of microbiota in broiler jejunum at species level.

5
J. Chang, et al. Ecotoxicology and Environmental Safety 194 (2020) 110420

Fig. 2. Correlations between jejunal microbial


abundances and mycotoxin residues or ADG. Note:
The different rectangles are colored based on the
Pearson correlation coefficients between microbial
abundances and mycotoxin residues or ADG. The
intensity of color represents the degree of correla-
tion, gray represents positive correlation, red re-
presents negative correlation. * indicates significant
correlation (p < 0.05), ** indicates highly sig-
nificant correlation (p < 0.01). AFB1-1: AFB1 re-
sidue in serum, AFB1-2: AFB1 residue in chest
muscle, AFB1-3: AFB1 residue in small intestine, ZEA-
1: ZEA residue in serum, ZEA-2: ZEA residue in small
intestine, ADG: average daily gain. . (For inter-
pretation of the references to color in this figure le-
gend, the reader is referred to the Web version of this
article.)

important role in re-establishing a new healthy microbial community can be inferred that CP3 addition in group G contributes to broiler body
for alleviating mycotoxin toxicity in broilers. weight gain by decreasing the ratio. Another report showed that Acti-
nobacteria and Proteobacteria could influence nutrient metabolism and
3.5. Gut microbial community analysis and its correlation with broiler disturb the normal microbial environment (Li et al., 2019b); therefore,
growth and mycotoxin residues both bacterial abundances decreased by CP3 addition may help to es-
tablish the healthy microbiota, which will be beneficial to broiler
In order to evaluate the correlation between jejunal microbiota (at growth and nutrient availability.
the species level) and mycotoxin residues or ADG, a Pearson's correla- In general, the intestinal microbiota plays an important role in an-
tion matrix was performed by calculating the Pearson's correlation imal health and production performance. An optimal microbiota pre-
coefficient (Fig. 2). This research showed that jejunal Lactobacillus vents colonisation of the intestinal epithelium by pathogens, modulates
aviarius abundance was positively correlated with ADG (p < 0.05), and the gut-associated systemic immunity, and influences gastrointestinal
negatively correlated with AFB1 residue in serum, illustrating that development (Mills et al., 2012). It can be inferred that compound
Lactobacillus aviarius plays an important role in promoting broiler probiotics additions are able to decrease AFB1 and ZEA residues and
growth and decreasing AFB1 residue. Lactobacillus aviarius has been increase broiler growth by increasing the proliferations of beneficial
found to be correlated with broiler growth (Torok et al., 2011), in bacteria such as Lactobacillus species and decreasing the proliferations
agreement with this study. The high abundance of lactic acid bacteria of detrimental bacteria such as Clostridium and Terrisporobacter.
has previously been used as an indicator of the healthy gut (Merrifield
et al., 2010). The other research showed that Lactobacillus had the 4. Conclusions
ability to produce bacteriocins and organic acids to inhibit pathogens
and inflammatory reactions for increasing body weight gain (Yang This study showed that AFB1 or AFB1+ZEA significantly decreased
et al., 2016). broiler production performance, induced hepatic and jejunal tissue
AFB1 and ZEA contamination significantly decreased the relative damages, disturbed the balance of gut microbiota. However, the com-
abundances of uncultured_bacterium_g_Lactobacillus and bacterium_GC452011 pound probiotics added in broiler diets could positively regulate gut
(p < 0.05), and both of them were negatively correlated with AFB1 and microbiota, decrease residues and cytotoxicity of AFB1 and ZEA, alle-
ZEA residues in serum, chest muscle and small intestine (except for AFB1 in viate mycotoxin negative effects on broiler production performance. It
serum) (p < 0.01); however, CP3 addition could enhance both bacterial was also found that gut microbiota was closely correlated with broiler
abundances. It can be speculated that the low mycotoxin residues caused by growth, mycotoxin metabolism and residues. This research will lay a
CP3 addition may be due to its function of increasing both bacterial theoretical foundation for probiotic application in detoxification and
abundances for mycotoxin biodegradation. Some strains of Lactobacillus animal health protection.
have been reported to have the ability of binding and removing AFB1 via
surface protein (Haskard et al., 2001), supporting this speculation. CRediT authorship contribution statement
The correlation analysis also revealed that Clostridium_sensu_
stricto_1, uncultured_bacterium_g_Turicibacter and uncultured_bacterium_g_ Juan Chang: Project administration, Investigation, Methodology,
Terrisporobacter abundances were positively correlated with the AFB1 and Writing - original draft. Tao Wang: Methodology, Investigation. Ping
ZEA residues in serum, chest muscle and small intestine (except for AFB1 Wang: Methodology, Investigation. Qingqiang Yin: Funding acquisi-
in serum) (p < 0.05). The Clostridium_sensu_stricto_1 and uncultured_ tion, Project administration, Methodology, Investigation, Writing - re-
bacterium_g_Terrisporobacter abundances were found to be negatively cor- view & editing. Qun Zhu: Methodology, Investigation. Fushan Lu:
related with ADG (p < 0.05), which was contrary to Lactobacillus aviarius. Data curation, Formal analysis. Tianzeng Gao: Data curation, Formal
Clostridium_sensu_stricto_1 belongs to genus Clostridium, which was reported analysis.
to be associated with intestinal disease and soft-tissue infections (Swartz,
2002). Turicibacter sp. was reported as a potential pathogen to have a Declaration of competing interest
negative impact on gastrointestinal health and physiological function
(Rettedal et al., 2009). Terrisporobacter was shown to be positively asso- The authors declare that they have no known competing financial
ciated with oxidative stress and inflammation for human being (Cai et al., interests or personal relationships that could have appeared to influ-
2019). However, the addition of CP3 could significantly decrease the re- ence the work reported in this paper.
lative abundances of these three potential harmful species, which will help
to alleviate mycotoxin toxicity for broilers. Acknowledgments
It was reported that the low ratio between Bacteroidetes and
Firmicutes was beneficial to fat deposition (Turnbaugh et al., 2006), so it This research was funded by the Natural Science Foundation of

6
J. Chang, et al. Ecotoxicology and Environmental Safety 194 (2020) 110420

Henan Province (182300410029), the Henan Key Scientific and Intestinal morphologic and microbiota responses to dietary Bacillus spp. in a broiler
Technological Project (171100110500), and Xinxiang Key Scientific chicken model. Front. Physiol. 9, 1968. https://doi.org/10.3389/fphys.2018.01968.
Li, S.S., Qi, Y.L., Chen, L.X., Qu, D., Li, Z.M., Gao, K., Chen, J.B., Sun, Y.S., 2019b. Effects
and Technological Project (ZD19005). of Panax ginseng polysaccharides on the gut microbiota in mice with antibiotic-as-
sociated diarrhea. Int. J. Biol. Macromol. 124, 931–937. https://doi.org/10.1016/j.
Appendix A. Supplementary data ijbiomac.2018.11.271.
Liang, X., Fu, Y., Liu, H., 2015. Isolation and characterization of enzyme-producing
bacteria of the silkworm larval gut in bioregenerative life support system. Acta
Supplementary data to this article can be found online at https:// Astronaut. 116, 247–253. https://doi.org/10.1016/j.actaastro.2015.07.010.
doi.org/10.1016/j.ecoenv.2020.110420. Liu, N., Wang, J.Q., Deng, Q.Q., Gu, K.T., Wang, J.P., 2018. Detoxification of aflatoxin B1
by lactic acid bacteria and hydrated sodium calcium aluminosilicate in broiler
chickens. Livest. Sci. 208, 28–32. https://doi.org/10.1016/j.livsci.2017.12.005.
References Liu, C.Q., Chang, J., Wang, P., Yin, Q.Q., Huang, W.W., Dang, X.W., Lu, F.S., Gao, T.Z.,
2019. Zearalenone biodegradation by the combination of probiotics with cell-free
extracts of Aspergillus oryzae and its mycotoxin-alleviating effect on pig production
Buranatragool, K., Poapolathep, S., Isariyodom, S., Imsilp, K., Klangkaew, N.,
performance. Toxins 11, 552. https://doi.org/10.3390/toxins11100552.
Poapolathep, A., 2015. Dispositions and tissue residue of zearalenone and its meta-
Luo, Y., Liu, X.J., Li, J.K., 2018. Updating techniques on controlling mycotoxins-A review.
bolites α-zearalenol and β-zearalenol in broilers. Toxicol. Rep. 2, 351–356. https://
Food Contr. 89, 123–132. https://doi.org/10.1016/j.foodcont.2018.01.016.
doi.org/10.1016/j.toxrep.2014.12.011.
Marin, D.E., Pistol, G.C., Neagoe, I.V., Calin, L., Taranu, I., 2013. Effects of zearalenone on
Cai, C.X., Zhang, Z.X., Morales, M., Wang, Y.N., Khafipour, E., Friel, J., 2019. Feeding
oxidative stress and inflammation in weanling piglets. Food Chem. Toxicol. 58,
practice influences gut microbiome composition in very low birth weight preterm
408–415. https://doi.org/10.1016/j.fct.2013.05.033.
infants and the association with oxidative stress: a prospective cohort study. Free
Merrifield, D.L., Dimitroglou, A., Foey, A., Davies, S.J., Baker, R.T.M., Bøgwald, J.,
Radical Biol. Med. 142, 146–154. https://doi.org/10.1016/j.freeradbiomed.2019.02.
Castex, M., Ringø, E., 2010. The current status and future focus of probiotic and
032.
prebiotic applications for salmonids. Aquaculture 302, 1–18. https://doi.org/10.
Conkova, E., Laciakova, A., Kovac, G., Seidel, H., 2003. Fusarial toxins and their role in
1016/j.aquaculture.2010.02.007.
animal diseases. Vet. J. 165, 214–220. https://doi.org/10.1016/S1090-0233(02)
Mills, S., Shanahan, F., Stanton, C., Hill, C., Coffey, A., Ross, R.P., 2012. Movers and
00127-2.
shakers: influence of bacteriophages in shaping the mammalian gut microbiota. Gut
Dong, H., Xian, Y.P., Xiao, K.J., Wu, Y.L., He, J.P., 2019. Development and comparison of
Microb. 4, 4–16. https://doi.org/10.4161/gmic.22371.
single-step solid phase extraction and QuEChERS clean-up for the analysis of 7 my-
Nakphaichit, M., Thanomwongwattana, S., Phraephaisarn, C., Sakamoto, N.,
cotoxins in fruits and vegetables during storage by UHPLC-MS/MS. Food Chem. 274,
Keawsompong, S., Nakayama, J., Nitisinprasert, S., 2011. The effect of including
471–479. https://doi.org/10.1016/j.foodchem.2018.09.035.
Lactobacillus reuteri KUB-AC5 during post-hatch feeding on the growth and ileum
Ellakany, H.F., Abuakkada, S.S., Oda, S.S., El-Sayed, Y.S., 2011. Infuence of low levels of
microbiota of broiler chickens. Poultry Sci. 90, 2753–2765. https://doi.org/10.3382/
dietary afatoxins on Eimeria tenella infections in broilers. Trop. Anim. Health Prod.
ps.2011-01637.
43, 249–257. https://doi.org/10.1007/s11250-010-9685-0.
National Research Council (NRC), 1994. Nutrient Requirements of Poultry. National
Fink-Gremmels, J., Malekinejad, H., 2007. Clinical effects and biochemical mechanisms
Academic Press, Washington, DC.
associated with exposure to the mycoestrogen zearalenone. Anim. Feed Sci. Technol.
Peng, W.X., Marchal, J.L.M., Poel, A.F.B.V.D., 2018. Strategies to prevent and reduce
137, 326–341. https://doi.org/10.1016/j.anifeedsci.2007.06.008.
mycotoxins for compound feed manufacturing. Anim. Feed Sci. Technol. 237,
Freire, L., Sant'Ana, A.S., 2018. Modified mycotoxins: an updated review on their for-
129–153. https://doi.org/10.1016/j.anifeedsci.2018.01.017.
mation, detection, occurrence, and toxic effects. Food Chem. Toxicol. 111, 189–205.
Piotrowska, M., Slizewska, K., Nowak, A., Zielonka, L., Zakowska, Z., Gajecka, M.,
https://doi.org/10.1016/j.fct.2017.11.021.
Gajecki, M., 2014. The effect of experimental Fusarium mycotoxicosis on microbiota
Ghali, R., Hmaissia-khlifa, K., Ghorbel, H., Maaroufi, K., Hedili, A., 2008. Incidence of
diversity in porcine ascending colon contents. Toxins 6, 2064–2681. https://doi.org/
aflatoxins, ochratoxin A and zearalenone in tunisian foods. Food Contr. 19, 921–924.
10.3390/toxins6072064.
https://doi.org/10.1016/j.foodcont.2007.09.003.
Rettedal, E., Vilain, S., Lindblom, S., Lehnert, K., Scofield, C., George, S., Clay, S.,
Good, I.J., 1953. The population frequencies of species and the estimation of population
Kaushik, R.S., Rosa, A.J., Francis, D., Brozel, V.S., 2009. Alteration of the ileal mi-
parameters. Biometrika 40, 237–264.
crobiota of weanling piglets by the growth-promoting antibiotic chlortetracycline.
Green, M.L., Diekman, M.A., Malayer, J.R., Scheidt, A.B., Long, G.G., 1990. Effect of
Appl. Environ. Microbiol. 75, 5489–5495. https://doi.org/10.1128/AEM.02220-08.
prepubertal consumption of zearalenone on puberty and subsequent reproduction of
Rice, L.G., Ross, F.B., 1994. Methods for detection and quantitation of fumonisins in corn,
gilts. J. Anim. Sci. 68, 171–178. https://doi.org/10.1051/gse:19900407.
cereal products and animal excreta. J. Food Protect. 57, 36–40. https://doi.org/10.
Harkai, P., Szabo, I., Cserhati, M., Krifaton, c., Risa, A., Rado, J., Balazs, A., Berta, k.,
4315/0362-028x-57.6.536.
Kriszt, B., 2016. Biodegradation of aflatoxin-B1 and zearalenone by Streptomyces sp.
Rykaczewska, A., Gajecka, M., Dabrowski, M., Wisniewska, A., Szczesniewska, J., Gajecki,
collection. Int. Biodeterior. Biodegrad. 108, 48–56. https://doi.org/10.1016/j.ibiod.
M.T., Zielonka, L., 2018. Growth performance, selected blood biochemical para-
2015.12.007.
meters and body weights of pre-pubertal gilts fed diets supplemented with different
Haskard, C.A., Elnezami, H.S., Kankaanpaa, P.E., Salminen, S., Ahokas, J.T., 2001.
doses of zearalenone (ZEN). Toxicon 152, 84–94. https://doi.org/10.1016/j.toxicon.
Surface binding of aflatoxin B1 by lactic acid bacteria. Appl. Environ. Microbiol. 67,
2018.07.013.
3086–3091. https://doi.org/10.1128/aem.67.7.3086-3091.2001.
Salem, R., El-Habashi, N., Fadl, S.E., Sakr, O.A., Elbialy, Z.I., 2018. Effect of probiotic
He, J., Zhang, K.Y., Chen, D.W., Ding, X.M., Feng, G.D., Ao, X., 2013. Effects of maize
supplement on aflatoxicosis and gene expression in the liver of broiler chicken.
naturally contaminated with afatoxin B1 on growth performance, blood profiles and
Environ. Toxicol. Pharmacol. 60, 118–127. https://doi.org/10.1016/j.etap.2018.04.
hepatic histopathology in ducks. Livest. Sci. 152, 192–199. https://doi.org/10.1016/
015.
j.livsci.2012.12.019.
Schloss, P.D., Westcott, S.L., Ryabin, T., Hall, J.R., Hartmann, M., Hollister, E.B.,
Huang, W.W., Chang, J., Wang, P., Liu, C.Q., Yin, Q.Q., Zhu, Q., Lu, F.S., Gao, T.Z., 2018.
Lesniewski, R.A., Oakley, B.B., Parks, D.H., Robinson, C.J., Sahl, J.W., Stres, B.,
Effect of the combined compound probiotics with mycotoxin-degradation enzyme on
Thallinger, G.G., Van Horn, D.J., Weber1, C.F., 2009. Introducing mothur: open-
detoxifying aflatoxin B1 and zearalenone. J. Toxicol. Sci. 43, 377–385. https://doi.
source, platform-independent, community-supported software for describing and
org/10.2131/jts.43.377.
comparing microbial communities. Appl. Environ. Microbiol. 75, 7537–7541.
Jahanian, E., Mahdavi, A.H., Asgary, S., Jahanian, R., 2016. Effect of dietary supple-
https://doi.org/10.1128/AEM.01541-09.
mentation of mannanoligosaccharides on growth performance, ileal microbial
Shin, D., Chang, S.Y., Bogere, P., Won, K., Choi, J.Y., Choi, Y.J., Lee, H.K., Hur, J., Park,
counts, and jejunal morphology in broiler chicks exposed to aflatoxins. Livest. Sci.
B.Y., Kim, Y., Heo, J., 2019. Beneficial roles of probiotics on the modulation of gut
190, 123–130. https://doi.org/10.1016/j.livsci.2016.05.008.
microbiota and immune response in pigs. PloS One 14, e0220843. https://doi.org/
Jia, R., Ma, Q.G., Fan, Y., Ji, C., Zhang, J.Y., Liu, T., Zhao, L.H., 2016. The toxic effects of
10.1371/journal.pone.0220843.
combined aflatoxins and zearalenone in naturally contaminated diets on laying
Sun, L., Lei, M., Zhang, N., Gao, X., Li, C., Krumm, C.S., Qi, D., 2015. Individual and
performance, egg quality and mycotoxins residues in eggs of layers and the protective
combined cytotoxic effects of aflatoxin B1, zearalenone, deoxynivalenol and fumo-
effect of Bacillus subtilis biodegradation product. Food Chem. Toxicol. 90, 142–150.
nisin B1 on BRL 3A rat liver cells. Toxicon 95, 6–12. https://doi.org/10.1016/j.
https://doi.org/10.1016/j.fct.2016.02.010.
toxicon.2014.12.010.
Jurgens, M.H., 1997. Animal Feeding and Nutrition, eighth ed. Kendall/Hunt Publishing
Swartz, M.N., 2002. Human diseases caused by foodborne pathogens of animal origin.
Company, Iowa.
Clin. Infect. Dis. 34, S111–S122. https://doi.org/10.1086/340248.
Khaniana, M., Karimi-Torshizia, M.A., Allamehb, A., 2019. Alleviation of aflatoxin-re-
Torok, V.A., Hughes, R.J., Mikkelsen, L.L., Perez-Maldonado, R., Balding, K., MacAlpine,
lated oxidative damage to liver and improvement of growth performance in broiler
R., Percy, N.J., Ophel-Keller, K., 2011. Identification and characterization of potential
chickens consumed Lactobacillus plantarum 299v for entire growth period. Toxicon
performance-related gut microbiotas in broiler chickens across various feeding trials.
158, 57–62. https://doi.org/10.1016/j.toxicon.2018.11.431.
Appl. Environ. Microbiol. 77, 5868–5878. https://doi.org/10.1128/AEM.00165-11.
Kosicki, R., Błajet-Kosicka, A., Grajewski, J., Twaruzek, M., 2016. Multiannual mycotoxin
Turnbaugh, P.J., Ley, R.E., Mahowald, M.A., Magrini, V., Mardis, E.R., Gordon, J.I., 2006.
survey in feed materials and feeding stuffs. Anim. Feed Sci. Technol. 215, 165–180.
An obesity-associated gut microbiome with increased capacity for energy harvest.
https://doi.org/10.1016/j.anifeedsci.2016.03.012.
Nature 444, 1027–1031. https://doi.org/10.1038/nature05414.
Li, P., Yang, S., Zhang, X., Huang, S., Wang, N., Wang, M., Long, M., He, J., 2018.
Wang, G., Yu, M.Z., Dong, F., Shi, J.R., Xu, J.H., 2017. Esterase activity inspired selection
Zearalenone changes the diversity and composition of caecum microbiota in weaned
and characterization of zearalenone degrading bacteria Bacillus pumilus ES-21. Food
rabbit. BioMed Res. Int. https://doi.org/10.1155/2018/3623274. Article ID
Contr. 77, 57–64. https://doi.org/10.1016/j.foodcont.2017.01.021.
3623274.
Wang, Y.L., Wang, B.J., Liu, M., Jiang, K.Y., Wang, M.Q., Wang, L., 2018. Aflatoxin B1
Li, C.L., Wang, J., Zhang, H.J., Wu, S.G., Hui, Q.R., Yang, C.B., Fang, R.J., Qi, G.H., 2019a.
(AFB1) induced dysregulation of intestinal microbiota and damage of antioxidant

7
J. Chang, et al. Ecotoxicology and Environmental Safety 194 (2020) 110420

system in pacific white shrimp (Litopenaeus vannamei). Aquaculture 495, 940–947. Yang, S.Y., Zheng, Y., Huang, Z., Min, W.X., Yang, H., 2016. Lactococcus nasutitermitis sp.
https://doi.org/10.1016/j.Toxicon.2018.09.005. nov. isolated from a termite gut. Int. J. Syst. Evol. Microbiol. 66, 518–522. https://
Williams, J.G., Deschl, U., Williams, G.M., 2011. DNA damage in fetal liver cells of Turkey doi.org/10.1099/ijsem.0.000743.
and chicken eggs dosed with aflatoxin B1. Arch. Toxicol. 85, 1167–1172. https://doi. Zuo, R.Y., Chang, J., Yin, Q.Q., Wang, P., Yang, Y.R., Wang, X., Wang, G.Q., Zheng, Q.H.,
org/10.1007/s00204-011-0653-x. 2013. Effect of the combined probiotics with aflatoxin B1-degrading enzyme on
Wu, F., 2004. Mycotoxin risk assessment for the purpose of setting international reg- aflatoxin detoxification, broiler production performance and hepatic enzyme gene
ulatory standards. Environ. Sci. Technol. 38, 4049–4055. https://doi.org/10.1021/ expression. Food Chem. Toxicol. 59, 470–475. https://doi.org/10.1016/j.fct.2013.
es035353n. 06.044.

You might also like