Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

METABOLISM AND NUTRITION

A novel aflatoxin-binding Bacillus probiotic: Performance, serum


biochemistry, and immunological parameters in Japanese quail

F. Bagherzadeh Kasmani,* M. A. Karimi Torshizi,*1 A. Allameh,† and F. Shariatmadari*

*Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran; and
†Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran

ABSTRACT Two experiments were performed to screen significant decrease in serum albumin, total protein,
bacilli isolated from quails for their aflatoxin removal and glucose and cholesterol levels but a significant in-
potential and to assess the efficiency of their ameliora- crease in serum uric acid, urea, creatinin and phospho-
tion of experimental aflatoxicosis. Nonhemolytic bacilli rus (P < 0.05). Treatment of birds on AFB1 with Bl
were selected for in vitro aflatoxin B1 (AFB1) removal restored these to their original levels (P < 0.05). AFB1
and conventional probiotic tests. The isolate with the + Bl-fed birds had serum aspartate aminotransferase,
highest scores was selected for assessment in field ex- alanine aminotransferase, lactate dehydrogenase, and
periments and was identified as Berevibacillus laterospo- alkaline phosphatase enzyme activity similar to control
rus (Bl). In the second experiment, 125 male Japanese birds (P < 0.05). Antibody titer against Newcastle dis-
quails (21 d old) were divided into 5 groups with 5 rep- ease virus was found to be lowest in the AFB1 group
lications to compare the toxin removal efficiency of Bl but highest in the Bl group (P < 0.05). Antibody pro-
with that of a commercial toxin binder, improved Mill- duction against sheep red blood cells was lower in the
bond-TX (IMTX). The experimental groups were as AFB1 group compared with the AFB1 + Bl group (P
follows: Control (without any feed additive or AFB1); < 0.05). Berevibacillus laterosporus supplementation of
AFB1 (2.5 mg/kg); AFB1 + Bl (2.5 mg/kg + 108 cfu/ the AFB1 diet restored the skin response to 2,4-dinitro
mL); AFB1+IMTX (2.5 mg/kg + 2.5 g/kg); and Bl 1-chlorobenzene to levels comparable with control birds
(108 cfu/mL). The greatest BW gain and slaughter and (P < 0.05). It can be concluded that selected indig-
carcass weights were found in the Bl group and the enous Bl is a promising probiotic with AFB1 removal
lowest values were observed in the AFB1 group (P < potential.
0.05). Feeding AFB1 alone to the chicks resulted in a
Key words: Japanese quail, aflatoxicosis, Berevibacillus laterosporus, performance, immune system
2012 Poultry Science 91:1846–1853
http://dx.doi.org/10.3382/ps.2011-01830

INTRODUCTION al., 2002), inorganic phosphorus, and calcium (Harvey


et al., 1990) are often reported due to aflatoxin poison-
Aflatoxins are a group of mycotoxins that are largely ing in avian and porcine species. Biochemical changes
produced by the fungi Aspergillus flavus and A. para- are sensitive indicators of aflatoxicosis and precede
siticus (Diaz et al., 2002). Aflatoxin B1 (AFB1) is the performance impairment and the emergence of clinical
most toxic and most prevalent compound, followed by symptoms. Immune suppression by aflatoxin has been
AFG1, AFB2, and AFG2 with decreasing toxicity (Bus- well documented in poultry as a consequence of vac-
by and Wogan, 1984). Aflatoxins can easily contaminate cination failure in affected commercial flocks (Thaxton
various types of crops and cause heavy economic losses; et al., 1974). Contact sensitivity to 2,4-dinitro 1-chlo-
they have a wide range of biological effects on different robenzene (DNCB) has been shown to induce a de-
species (Karaman et al., 2005). Significant changes in layed hypersensitivity reaction in chickens and used as
serum biochemistry parameters are generally regarded a convenient model to investigate cellular immune re-
as indicative of aflatoxicosis (Basmacioglu et al., 2005). sponse and its regulation in the chicken (Awadhiya et
Decreased serum concentrations of total protein, albu- al., 1982; Huynh and Chubb, 1987).
min, cholesterol (Rosa et al., 2001), uric acid (Oğuz et Several methods have been developed to reduce afla-
toxin contamination in animal feed. However, most of
©2012 Poultry Science Association Inc. these are not of practical value (Diaz et al., 2002). Re-
Received August 30, 2011.
Accepted April 26, 2012.
cently, the interest of researchers has been focused on
1 Corresponding author: karimitm@modares.ac.ir biological methods to take advantage of their simplicity

1846
AFLATOXIN-BINDING BACILLUS PROBIOTIC 1847
and affordability. The main step of these methods is the Pathogenicity Test. Aerobic spore-forming isolates
choice of microorganisms for diminishing the presence were cultured by streaking on blood agar media (Difco,
of aflatoxins in contaminated feed and food supplies Franklin Lakes, NJ) containing 5% defibrinated sheep
(Teniola et al., 2005). blood and incubated for 24 h at 37°C to evaluate their
Probiotics are defined by Fuller as “live microbial possible hemolytic activity (Wolfenden et al., 2010).
food supplements which beneficially affect the host Only γ-hemolytic (nonhemolytic) bacteria were re-
by improving its intestinal microbial balance” (Fuller, tained.
1991). Furthermore, lactic acid bacteria are important Aflatoxin B1 Binding Assay. Aflatoxin B1 (Sigma-
aflatoxin reducer microorganisms, as has been empha- Aldrich, Steinheim, Germany) was suspended in ben-
sized recently (el-Nezami et al., 1998; Peltonen et al., zene/acetonitrile (97:3 vol/vol) to obtain a concentra-
2000, 2001; Lee et al., 2003; Shahin, 2007; Hernandez- tion of 2 mg/mL. A working solution of 5 μg/mL of
Mendoza et al., 2009). It appears that AFB1 is bound AFB1 was prepared in PBS and the benzene/aceto-
to the surface components of probiotic bacteria (Has- nitrile was evaporated by gently heating in a water
kard et al., 2001). The destruction of specific compo- bath. The cell count of an overnight culture of Bacil-
nents of the bacterial cell wall, such as carbohydrates lus isolates obtained in a shaking incubator (150 rpm,
and proteins, reduced AFB1 binding by L. rhamnosus 37°C) was adjusted to 7th McFarland tube, equal to
strain GG, suggesting the importance of the cellular approximately 1 to 1.5 × 1010 cfu/mL. The culture was
envelope in AFB1 binding (Haskard et al., 2000; Her- then centrifuged (3,000 × g, 15 min) and the bacterial
nandez-Mendoza et al., 2009). It is likely, however, that pellets were washed twice with 5 mL of Milli-Q water.
multiple mechanisms are involved in AFB1 binding. Bacterial pellets were resuspended in 1.5 mL of AFB1
Members of the Bacillus genus are considered the working solution and incubated at 37°C for 4 h. Cells
most promising as probiotics for use in animal feeds were pelleted (3,000 × g, 15 min) and aliquots (1 mL)
because of their extraordinary extended shelf life and of the supernatant were collected for AFB1 quantifica-
resistance to environmental conditions compared with tion (Peltonen et al., 2000). The AFB1 concentration
more conventional probiotics based on lactic acid bacte- was estimated by ELISA (Ridascreen Aflatoxin B1 Art.
ria (Shivaramaiah et al., 2011). Wolfenden et al. (2010) No. 1211, R-Biopharm, Darmstadt, Germany).
screened potential Bacillus probiotics based on heat/ Selection and Identification. To choose a candi-
ethanol treatment, hemolytic activity, and antimicro- date isolate for field experiments, conventional probi-
bial activity against some pathogens. otic tests were performed on the isolates that already
Although mycotoxin removal is well established in showed superior AFB1 removal ability. Tests included
lactobacilli species, there has been no published report antagonistic activity, cell surface hydrophobicity, co-
on mycotoxin removal by poultry probiotics based on aggregation, aggregation, enzymatic activities, low pH
bacilli. Hence, the present study was performed to as- tolerance, bile salt tolerance, and antibiotic sensitiv-
sess the AFB1 removal potential of a Berevibacillus lat- ity (Kirby et al., 1957; Kim et al., 2007; Surachon et
erosporus (Bl) probiotic in vitro and in vivo. al., 2011). Based on the results obtained in the AFB1-
binding and conventional probiotic tests, an isolate was
MATERIALS AND METHODS selected for further assessment in in vivo experiments.
Identification of the selected isolate was performed
Experiment 1 using standard taxonomic descriptions from Sneath
(1986) with commercially available strips (API 50CHB,
Isolation and Growth Conditions. The animal use API Laboratory Products Ltd., Biomerieux, France).
protocol was approved by the institutional animal care The results were analyzed using the API Web database
of Tarbiat Modares University. Forty healthy Japanese (https://apiweb.biomerieux.com) for species-level iden-
quails aged 21 d that were not receiving any medical tification.
treatment and had not received antibiotics were select-
ed. The quails had free access to water for 24 h before Experiment 2
slaughter. Tissue samples were taken from different
parts of the gastrointestinal tract (crop, jejunum, ileum, Experimental Birds and Treatments. In total, 125
and cecca) under aseptic conditions. Homogenized sam- male Japanese quails (Coturnix japonica), aged 21 d,
ples were transferred to buffered peptone water (1:10 were randomly assigned to one of 5 treatment groups
vol/vol). Samples were placed in a water bath at 80°C with 5 replicates of 5 birds each. All birds were fed a
for 15 min and then plated on nutrient agar medium similar base diet formulated to meet or exceed the nu-
(Merck; Darmstadt, Germany). Plates were incubated tritional requirements of Japanese quails (NRC, 1994;
aerobically at 37°C for 24 to 48 h (Barbosa et al., 2005; Table 1). The experimental groups included the con-
Guo et al., 2006). Any isolate with colony morphology trol (without any feed additive or AFB1), AFB1 (2.5
consistent with that of the Bacillus cereus group (B. mg/kg), AFB1 + Bl (2.5 mg/kg + 108 cfu/mL), AFB1
cereus, B. mycoides, B. thurigensis, and B. anthrasis) + IMTX (2.5 mg/kg + 2.5 g/kg) and Bl (108 cfu/
was excluded from further investigations (Wolfenden mL) groups. Supplementation was continued for 4 wk.
et al., 2010). The IMTX (Milwhite Inc., Brownsville, TX) is an inert
1848 Bagherzadeh Kasmani et al.
Table 1. Composition of the basal diets Serum Biochemical Analysis
Grower
Item (21–49 d)
Samples of approximately 1 mL of whole blood were
drawn from 10 birds in each treatment through punc-
Ingredient (%) ture of a wing vein on d 49. Concentrations of albu-
  Yellow corn 42.32
  Soybean meal (44% CP) 40.20 min, total protein, glucose, cholesterol, uric acid, urea,
  Vegetable oil 7.48 creatinine and phosphorus were determined in serum
  Fish meal (65% CP) 7.30 samples. The serum activities of aspartate aminotrans-
 CaCO3 1.21
  Dicalcium phosphate 0.01 ferase (AST), alanine aminotransferase (ALT), lac-
  Sodium chloride 0.28 tate dehydrogenase (LDH), and alkaline phosphatase
  Mineral and vitamin premix1 0.50 (ALP) were also determined. The analyses of the se-
  dl-Methionine 0.03
  Washed sand 0.67 rum samples were performed immediately by spectro-
 Total 100 photometric methods using commercially available kits
  Calculated value2   (Parsazmun, Tehran, Iran).
  ME (kcal/kg) 3,130
  CP (%) 25.90
  Lys (%)
  Met + Cys (%)
1.40
0.81
Humoral Immune Response: Antibody
  Calcium (%) 0.86 Production Against Newcastle Disease
  Nonphytate phosphorus (%) 0.32
Virus and Sheep Red Blood Cells
1Supplied the following per kilogram of diet: retinyl acetate, 9,000 IU;
cholecalciferol, 2,000 IU; dl-α-tocopheryl acetate, 12.5 IU; menadione Vaccination against the Newcastle disease (ND) vi-
sodium bisulfite, 1.76 mg; biotin, 0.12 mg; thiamine, 1.2 mg; riboflavin,
3.2 mg; calcium d-pantothenate, 6.4 mg; pyridoxine, 1.97 mg; nicotinic rus was performed on d 41 using an eye dropper (Live
acid, 28 mg; cyanocobalamine, 0.01 mg; choline chloride, 320 mg; folic B1 strain; Vetrina; Zagreb, Croatia). The anti-ND titer
acid, 0.38 mg; MnSO4∙H2O, 60 mg; FeSO4∙7H2O, 80 mg; ZnO, 51.74 mg; was assessed by a hemagglutination inhibition test on
CuSO4∙5H2O, 8 mg; iodized NaCl, 0.8 mg; Na2SeO3, 0.2 mg.
2Calculated from NRC (1994). sera obtained on d 48.
Birds were also injected into the breast muscle with
SRBC (5% vol/vol in sterile PBS, 0.2 mL/chick) at 41
d. Blood samples were drawn 7 d after the injection.
montmorillonite clay-based adsorbent that is obtained The antibody levels against SRBC were determined by
from natural clay deposits (Miles and Henry, 2007). hemagglutination test. Heat inactivated plasma (56°C
Aflatoxin Production. Aflatoxin was produced for 30 min) was analyzed for anti-SRBC titer as previ-
from an A. parasiticus PTCC-5286 culture (obtained ously described (Qureshi and Havenstein, 1994).
from the Iranian Research Organization for Science
and Technology) by fermentation of rice grains and its Cellular Immune Response: Skin
AFB1 content was determined by the method of Sho- Response to 2,4-Dinitro 1-Chlorobenzene
twell et al. (1966). The contaminated rice powder was
incorporated into the base diet to provide 2.5 mg of 2,4-Dinitro 1-chlorobenzene (Merck; Darmstadt,
AFB1/kg of feed. Germany) was dissolved in a mixture of acetone and
Performance Parameters. The birds were weighed olive oil (4:1 vol/vol) to a final concentration of 1 mg/
at weekly intervals for up to 4 wk on a pen basis and mL. On d 48, the skin of the birds was anointed with
the feed intake per pen was recorded at weekly inter- 0.1 mL of DNCB solution. An area of approximately 10
vals. The feed conversion ratio was calculated from cm2 without feathers on the left lateral abdomen was
feed intake and BW gain data. Slaughter and carcass chosen for the challenge with DNCB. The same area on
weights were measured at the end of the experiment the right side was treated with the solvent alone. The
on d 49. skin thickness (on both sides) before and 24 h after the

Table 2. Effect of aflatoxin B1 and feed additives on performance of Japanese quail


Aflatoxin B1 BW gain Feed conversion Slaughter Carcass
Group (mg/kg) Additive1 (g/bird/d) ratio (g/g) weight (g) weight (g)

Control 0 — 3.21a 4.10ab 259.97a 166.57a


Bl2 0 Bl 3.33a 3.89b 266.30a 171.63a
AFB1 2.5 — 2.10b 5.77a 204.22c 132.63c
AFB1+Bl 2.5 Bl 2.98a 4.33ab 256.97a 169.81a
AFB1+IMTX3 2.5 IMTX 2.90a 4.65ab 231.87b 149.64b
SEM   0.133 0.265 5.66 3.78
P-value   0.022 0.048 <0.001 <0.001
a–cDifferentsuperscripts in each column show significant differences (P < 0.05).
1Additive: Bl (108 cfu/mL of drinking water); IMTX (2.5 g/kg of feed).
2Bl: Berevibacillus laterosporus.
3IMTX: Commercial toxin binder (Milwhite Inc., Brownsville, TX).
AFLATOXIN-BINDING BACILLUS PROBIOTIC 1849
challenge was measured to assess reactions. Differences
before and after the challenge were calculated to deter-

Phosphorus
(mg/dL)
6.74ab

7.11ab
mine the mean increase in skin thickness in each bird.

0.044
5.92b

6.09b
7.55a

0.21
For each bird, the average of 3 repeat measurements
was used for analysis (Verma et al., 2004).
nitrogen (mg/dL)
Statistical Analysis
Blood urea

<0.001
2.90d

3.80b
3.90b
4.50a
3.30c

1.00
The data were analyzed by a general linear model
for a completely randomized experimental design us-
ing SAS software (SAS Institute Inc., 1990); the means
were compared by Duncan’s multiple range test (P ≤
Uric acid
(mg/dL)

<0.001
5.90b
4.80b

7.90b

0.05).
19.80a

18.05a
1.75

RESULTS
Experiment 1: Isolation, Hemolysis Test,
Creatinine
(mg/dL)

0.008
0.004
0.54b

0.54b
0.54b
0.56a

0.56a

AFB1 Binding, and Identification


Three hundred fifty nonhemolytic spore-forming iso-
lates were prescreened from the 40 quail samples. All
isolates shared the typical characteristics of Bacillus
Cholesterol

106.66bc
(mg/dL)

<0.001
69.50d

115.05b
146.77a
93.22c

7.03

species; that is, they were gram-positive, rod-shaped,


catalase-positive, and spore-forming aerobic bacteria.
After conducting the AFB1 removal test as well as
some conventional probiotic tests consisting of acid and
bile tolerance, antagonistic activity, and hydrophobic-
(mg/dL)

0.002
182.61b

182.24b
186.59b
Glucose

209.06a

163.04c

4.43

ity, one isolate was selected for field experiments with


growing quails. The selected isolate was able to remove
57.3% of the AFB1 and was identified as Berevibacillus
laterosporus.
Table 3. Effect of aflatoxin B1 and feed additives on some serum biochemical variables
Triglyceride
(mg/dL)

<0.001
90.07b
82.44c
67.93c

13.81
115.2b
206.1a

Experiment 2
Performance. The results showed that AFB1 has a
significant effect on quail performance (Table 2). The
Albumin

<0.001
2.35b

1.37d
3.36a

3.42a
2.25c
(g/dL)

greatest BW gain was found in the Bl group and the


0.20

lowest in the AFB1 group. Body weight gain in AFB1 +


Bl (108 cfu/mL of drinking water); IMTX (25 g/kg of feed).

Bl birds was not significantly different from the control


3IMTX: Commercial toxin binder (Milwhite Inc., Brownsville, TX).
Total protein

group (P > 0.05). The feed conversion ratio was signifi-


superscripts show significant differences (P < 0.05).
(g/dL)

cantly affected by both AFB1 and Bl (P < 0.05), and


0.001
1.96b

2.60b
3.50a
3.70a

4.10a

0.22

AFB1 group had higher value than the other groups.


The highest slaughter and carcass weights were found
in the Bl group and vice versa for the AFB1 group (P
Additive1

< 0.05).
IMTX


Bl

Bl

Serum Biochemistry and Enzyme Activity. The


effects of AFB1, Bl, and IMTX on serum biochemical
variables are shown in Tables 3 and 4. The serum total
protein and albumin significantly reduced and serum
B1 (mg/kg)
Aflatoxin

uric acid increased in AFB1 group compared with the


2Bl: Berevibacillus laterosporus.
2.5
2.5
2.5
0
0

AFB1 + Bl, Bl, and control groups (P < 0.05). Add-


ing IMTX to AFB1-contaminated feed significantly in-
creased serum uric acid (P < 0.05; Table 3). The high-

est value of serum phosphorus and the lowest amount


of glucose was observed in the AFB1 group (P < 0.05).
AFB1+IMTX3

a–dDifferent

Serum AST activity in the AFB1 group was signifi-


1Additive:

cantly higher than that in the other groups (P < 0.05).


AFB1+Bl
Control

P-value

Adding IMTX to the contaminated diet increased the


Group

AFB1

SEM

activity of AST, LDH, and ALP enzymes compared


Bl2
1850 Bagherzadeh Kasmani et al.
Table 4. Effect of aflatoxin B1 and feed additives on selected serum enzyme activity
Aspartate Alanine Lactate Alkaline
Aflatoxin Additive1 amino-transferase amino-transferase dehydragenase phosphatase
Group B1 (mg/kg) (mg/kg) (IU/L) (IU/L) (IU/L) (IU/L)

Control 0 — 141.59b 5.95b 590.00c 375.34c


Bl2 0 Bl 130.01c 6.12b 576.67c 361.60c
AFB1 2.5 — 160.60a 10.25a 753.33b 425.12b
AFB1+Bl 2.5 Bl 122.07d 6.28b 626.66c 383.35c
AFB1+IMTX3 2.5 IMTX 132.33c 11.57a 1,146.66a 458.65a
SEM   3.58 0.669 57.44 10.04
P-value   <0.001 <0.001 <0.001 <0.001
a–cDifferentsuperscripts show significant differences (P < 0.05).
1Additive: Bl (108 cfu/mL of drinking water); IMTX (25 g/kg of feed).
2Bl: Berevibacillus laterosporus.
3IMTX: Commercial toxin binder (Milwhite Inc., Brownsville, TX).

with the other groups (P < 0.05). Supplementation of stock. Failure to gain BW will lead to economic losses.
Bl in birds fed AFB1 restored the elevated activity of The relationship between commercial performance of
these enzymes (Table 4). broilers and aflatoxin contamination of diets has been
reviewed by Shane (1994). Aflatoxin effects on feed in-
Humoral Immune Response take, BW gain, and the feed conversion ratio may be a
result of anorexia, reluctance and prevention of protein
Antibody production against ND on d 48 was lower synthesis, and lipogenesis (Oğuz et al., 2000; Parlat et
in the AFB1 group compared with the Bl group (Ta- al., 2001). The aflatoxin effects on growth performance
ble 5). Antibody titer against SRBC on d 48 was sig- in this study are in accordance with those in previous
nificantly different among groups (P < 0.05), and the studies (Allameh et al., 2005; Shi et al., 2006; Pasha et
AFB1 group had a lower titer than the AFB1 + Bl al., 2007).
group (P < 0.05). Serum total protein, albumin, creatinine, urea, and
glucose concentrations have also been described as valu-
Cellular Immune Response able parameters of hepatic injury and function (Mathur
et al., 2001). In our study, AFB1-contaminated feed
Increases in skin thickness in response to DNCB were resulted in an increase in serum creatinine, uric acid,
significantly different among groups (P < 0.05; Table urea, and phosphorus concentrations and a decrease
5). The lowest skin response was observed in the AFB1 in concentrations of serum albumin, total protein, glu-
group and there was no significant difference between cose, and triglyceride. Madheswaran et al. (2004) also
the AFB1 + Bl and control groups (P < 0.05). reported a decrease in serum total protein, albumin,
globulin, glucose, and cholesterol when Japanese quails
DISCUSSION were fed 3 ppm of AFB1 alone or in combination with
T2 toxin for 35 d. It is worth mentioning that they did
Prescreening of Bacillus isolates by amplification in not observe any change in serum urea, uric acid, cre-
broth before agar isolation has been reported previous- atinine, or phosphorus. Nazar et al. (2012) observed a
ly (Földes et al., 2000; Wolfenden et al., 2010; Shivara- significant reduction in protein, albumin, and globulin
maiah et al., 2011). However, selection of bacilli from concentrations in the plasma of Japanese quails fed an
the digestive tract of poultry to determine their AFB1 AFB1-contaminated diet. The biochemical changes dur-
removal ability is reported here for the first time. Pro- ing aflatoxicosis could be due to the inhibition of pro-
biotics with high mycotoxin-binding ability have good tein synthesis in the liver along with other damage to
prospects as mycotoxin-binding organisms. Practical the liver and kidney (Tung et al., 1975; Kubena et al.,
use of binder strains is anticipated in the near future 1993; Shi et al., 2006). Aflatoxins produce a large num-
(Shetty and Jespersen, 2006). The mycotoxin binding ber of active metabolites during a biological conversion
property of probiotic bacteria is attributed to cell wall and these bind to DNA and RNA and reduce protein
moieties (el-Nezami et al., 1998; Haskard et al., 2000; production (Doerr et al., 1983). Impaired protein and
Hernandez-Mendoza et al., 2009). albumin biosynthesis was observed in chickens fed an
Dietary inclusion of AFB1 reduced the growth of aflatoxin-contaminated diet (Oğuz et al., 2002; Basma-
quails and supplementation of Bl to birds prevented cioglu et al., 2005). Supplementation of contaminated
the loss in BW gain and feed conversion. Bacillus iso- feed with Bl could ameliorate the aflatoxin-induced in-
lates have previously been shown to increase BW gain crease in uric acid and creatinine concentrations and
(Shivaramaiah et al., 2011; Wolfenden et al., 2011). return these to levels similar to controls. The similarity
Low growth rate and poor performance are the most of serum metabolites in the AFB1 + Bl and control
prevalent symptoms of aflatoxicosis in poultry and live- groups could be the result of toxin binding by Bl as in
AFLATOXIN-BINDING BACILLUS PROBIOTIC 1851
Table 5. Effect of aflatoxin B1 and feed additives on immune response to Newcastle disease virus, sheep red blood cells, and 2,4-di-
nitro 1-chlorobenzene
Increase in mean
Aflatoxin B1 Anti-NDV2 Anti-SRBC skin thickness
Group (mg/kg) Additive1 titer (log2) titer (log2) to DNCB3 (mm)

Control 0 — 1.75bc 3.2b 0.492b


Bl4 0 Bl 3.20a 4.1a 0.950a
AFB1 2.5 — 1.15c 2.4c 0.076d
AFB1+Bl 2.5 Bl 1.70bc 3.2b 0.520b
AFB1+IMTX5 2.5 IMTX 2.30b 2.8bc 0.182c
SEM   0.062 0.165 0.134
P-value   <0.001 <0.001 <0.001
a–dDifferent superscripts show significant differences (P < 0.05).
1Additive: Bl (108 cfu/mL of drinking water); IMTX (25 g/kg of feed).
2NDV: Newcastle disease virus.
3DNCB: dinitrochlorobenzene.
4Bl: Berevibacillus laterosporus.
5IMTX: Commercial toxin binder (Milwhite Inc., Brownsville, TX).

experiment 1 this activity was evidenced (57.3% AFB1 restored to normal when birds on contaminated feed
removal). were supplemented with Bl. Several researchers used
Activity of serum enzymes such as ALP, ALT, AST, topical DNCB to evaluate the cell mediated immuno-
and LDH provides a sensitive and specific measure of suppressive effect of mycotoxins in broilers (Singh et
hepatic function or injury (Kubena et al., 1997; Mathur al., 1990; Bakshi, 1991; Verma et al., 2004; Nazar et al.,
et al., 2001; Abbès et al., 2006). Feeding AFB1 alone in- 2012). However, cell mediated immune-stimulant effect
creased AST, ALT, LDH, and ALP activity compared of probiotic administration via drinking water on skin
with the control diet. Supplementation of Bl in con- response to DNCB was reported by Karimi Torshizi et
taminated feed restored the activity of these enzymes. al. (2010).
Surprisingly, IMTX supplementation of AFB1-contam- In conclusion, the Bacillus probiotic isolated in the
inated feed resulted in an increase in serum LDH and present study proved its AFB1-binding activity in vi-
ALP activity (P < 0.05); this increase in ALT was nu- tro. In vivo results verified its AFB1-binding activity
meric. In the AFB1 + IMTX group, AST activity was in quails with regard to performance, serum biochem-
decreased in comparison to the AFB1 group (P < 0.05). istry, and immune responses. The common list of pro-
Aravind et al. (2003) reported that broilers fed a natu- biotic selection criteria could be amended by inserting
rally AFB1-contaminated diet had significant increases a mycotoxin-binding property, which could result in
in the activity of these enzymes at 21 d of age. Similar the introduction of powerful multifunctional probiotic
results were observed by others (Shi et al., 2006; Gowda preparations in the near future.
et al., 2008), which suggests that mycotoxins exert a di-
rect toxic effect on animal livers. The supplementation
REFERENCES
of Bl to birds fed AFB1-contaminated feed proved to be
an effective means of protecting against aflatoxicosis as Abbès, S., Z. Ouanes, J. Salah-Abbes, Z. Houas, R. Oueslati, H.
judged by serum metabolites and enzyme activity. Bacha, and O. Othman. 2006. The protective effect of hydrated
sodium calcium aluminosilicate against haematological, biochem-
The immunomodulatory effects of probiotics have ical and pathological changes induced by Zearalenone in mice.
been reported frequently (Kabir et al., 2004; Haghighi Toxicon 47:567–574.
et al., 2005, 2006; Nayebpor and Hashemi, 2007; Apata, Allameh, A., A. Safamehr, S. Mirhadi, M. Shivazad, M. Razzaghi-
2008; Brisbin et al., 2008). Interestingly, the immune Abyaneh, and A. Afshar-Naderi. 2005. Evaluation of biochemical
and production parameters of broiler chicks fed ammonia treated
response of the AFB1 + Bl group was similar to that of aflatoxin contaminated maize grains. Anim. Feed Sci. Technol.
the control group. Our results confirm previous reports 12:289–301.
that stated that consumption of aflatoxin-contaminat- Apata, D. F. 2008. Growth performance, nutrient digestibility and
ed feed in broilers reduced the production of antibodies immune response of broiler chicks fed diets supplemented with a
culture of Lactobacillus bulgaricus. J. Sci. Food Agric. 88:1253–
(Qureshi et al., 1998; Verma et al., 2004; Tessari et 1258.
al., 2006). Weakening of the immune system may be Aravind, K. L., V. S. Patil, G. Devegowda, B. Umakantha, and S.
the result of the prevention of protein synthesis, which P. Ganpule. 2003. Efficacy of esterified glucomannan to counter-
act mycotoxicosis in naturally contaminated feed on performance
includes a reduction in IgG and IgA, a reduced number and serum biochemical and hematological parameters in broilers.
of lymphocytes, and an effect on the bursa (Sur and Poult. Sci. 82:571–576.
Celik, 2003). Awadhiya, R. P., J. L. Vegad, and G. N. Kolte. 1982. Eosinophil
Birds of AFB1 group had the lowest skin thickness af- leukocytic response in dinitrochlorobenzene skin hypersensitivity
reaction in chicken. Avian Pathol. 11:187–194.
ter challenge with DNCB, which confirmed the results Bakshi, C. S. 1991. Studies on the effect of graded dietary levels of
of earlier studies (Singh et al., 1990; Bakshi, 1991). The aflatoxin on immunity in commercial broilers. M.V.Sc. Thesis.
AFB1-induced decrease in skin response to DNCB was Indian Veterinary Research Institute, Izatnagar, U.P. India.
1852 Bagherzadeh Kasmani et al.
Barbosa, T. M., C. R. Serra, R. M. La Ragione, M. J. Woodward, Karimi Torshizi, M. A., A. R. Moghaddam, Sh. Rahimi, and N.
and A. O. Henriques. 2005. Screening for Bacillus isolates in the Mojgani. 2010. Assessing the effect of administering probiotics
broiler gastrointestinal tract. Appl. Environ. Microbiol. 71:968– in water or as a feed supplement on broiler performance and im-
978. mune response. Br. Poult. Sci. 51:178–184.
Basmacioglu, H., H. Oguz, M. Ergun, R. Col, and Y. O. Bardane. Kim, P. I.,, M. Y. Jung, Y. Chang, S. Kim, S. Kim, and Y. Park.
2005. Effect of dietary esterified glucomannan on performance, 2007. Probiotic properties of Lactobacillus and Bifidobacterium
serum biochemistry and hematology in broiler exposed to afla- strains isolated from porcine gastrointestinal tract. Appl. Micro-
toxin. Czech J. Anim. Sci. 50:31–39. biol. Biotechnol. 74:1103–1111.
Brisbin, J. T., H. Zhou, J. Gong, P. Sabour, M. R. Akbari, H. R. Kirby, W. M. M., G. M. Yoshihara, K. S. Sundsted, and J. H. War-
Haghighi, H. Yu, A. Clarke, A. J. Sarson, and S. Sharif. 2008. ren. 1957. Clinical usefulness of a single disc method for antibi-
Gene expression profiling of chicken lymphoid cells after treat- otic sensitivity testing. Antibiot. Annu. 892–897.
ment with Lactobacillus acidophilus cellular components. Dev. Kubena, L. F., R. B. Harvey, S. A. Buckley, T. S. Edrington, and
Comp. Immunol. 32:563–574. G. E. Rottinghaus. 1997. Individual and combined effects of mo-
Busby, W. F., and G. N. Wogan. 1984. Aflatoxins. Pages 946–1135 niliformin present in Fusarium fujikuroi culture material and af-
in Chemical Carcinogens. 2nd ed. Am. Chem. Soc., Washington, latoxin in broiler chicks. Poult. Sci. 76:265–270.
DC. Kubena, L. F., R. B. Harvey, W. E. Huff, M. H. Elissald, A. G.
Diaz, D. E., W. M. Hagler Jr., B. A. Hopkins, and L. W. Whitlow. Yersin, T. D. Philips, and G. E. Rottinghaus. 1993. Efficacy of a
2002. Aflatoxin Binders I: In vitro binding assay for aflatoxin hydrated sodium calcium aluminosilicate to reduce the toxicity of
B1 by several potential sequestering agents. Mycopathologia aflatoxin and diacetoxyscripenol. Poult. Sci. 72:51–59.
156:223–226. Lee, Y. K., H. El-Nezami, C. A. Haskard, S. Gratz, K. Y. Puong,
Doerr, J. A., C. J. Huff, G. W. Wabeck, J. D. Ghaloupak, and J. W. S. Salminen, and H. Mykkanen. 2003. Kinetics of adsorption and
Markely. 1983. Effects of low level chronic aflatoxicosis in broiler desorption of aflatoxin B1 by viable and nonviable bacteria. J.
chicken. Poult. Sci. 62:1971–1977. Food Prot. 66:426–430.
el-Nezami, H., P. S. Kankaanpää, S. Salminen, and J. Ahokas. 1998. Madheswaran, R., C. Balachandran, and B. Murali Manohar. 2004.
Physicochemical alterations enhance the ability of dairy strains Influence of dietary culture material containing aflatoxin and
of lactic acid bacteria to remove aflatoxin from contaminated T2 toxin on certain serum biochemical constituents in Japanese
media. J. Food Prot. 61:466–468. quail. Mycopathologia 158:337–341.
Földes, T., I. Banhegyi, Z. Herpai, L. Varga, and J. Szigeti. 2000. Mathur, S., P. D. Constable, and R. M. Eppley. 2001. Fumonisin B1
Isolation of Bacillus strains from the rhizosphere of cereals and is hepatotoxic and nephrotoxic in milk-fed calves. Toxicol. Sci.
in vitro screening for antagonism against phytopathogenic, food- 60:385–396.
borne pathogenic and spoilage microorganisms. J. Appl. Micro- Miles, R. D., and P. R. Henry. 2007. Safety of improved Milbond-TX
biol. 89:840–846. when fed in broiler diets limiting in available phosphorus or con-
Fuller, R. 1991. Probiotics in human medicine. Gut 32:439–442. taining variable levels of metabolizable energy. J. Appl. Poult.
Gowda, N. K. S., D. R. Ledoux, G. E. Rottinghaus, A. J. Bermudez, Res. 16:412–419.
and Y. C. Chen. 2008. Efficacy of turmeric (Curcuma longa), NRC. 1994. Nutrient Requirements of Poultry. 9th rev. ed. Natl.
containing a known level of curcumin, and a hydrated sodium Acad. Press, Washington, DC.
calcium aluminosilicate to ameliorate the adverse effects of afla- Nayebpor, M. P. F., and A. Hashemi. 2007. Effects of different levels
toxin in broiler chicks. Poult. Sci. 87:1125–1130. of direct fed microbial (Primalac) on growth performance and
Guo, X., D. Li, W. Lu, X. Piao, and X. Chen. 2006. Screening of humoral immune response in broiler chickens. J. Anim. Vet.
Bacillus strains as potential probiotics and subsequent confirma- Adv. 6:1308–1313.
tion of the in vivo effectiveness of Bacillus subtilis MA139 in pigs. Nazar, F. N., A. P. Magnoli, A. M. Dalcero, and R. H. Marin. 2012.
Anton. Leeuw. Int. J. G. 90:139–146. Effect of feed contamination with aflatoxin B1 and administra-
Haghighi, H. R., J. Gong, C. L. Gyles, M. A. Hayes, B. Sanei, P. tion of exogenous corticosterone on Japanese quail biochemical
Parvizi, H. Gisavi, J. R. Chambers, and S. Sharif. 2005. Modu- and immunological parameters. Poult. Sci. 91:47–54.
lation of antibody-mediated immune response by probiotics in Oğuz, H., T. Kececi, F. Birdane, F. Onder, and V. Kurtoglu. 2000.
chickens. Clin. Diagn. Lab. Immunol. 12:1387–1392. Effect of clinoptilolite on serum biochemical and haematological
Haghighi, H. R., J. Gong, C. L. Gyles, M. A. Hayes, H. Zhou, B. characters of broiler chickens during aflatoxicosis. Res. Vet. Sci.
Sanei, J. R. Chambers, and S. Sharif. 2006. Probiotics stimulate 69:89–93.
production of natural antibodies in chickens. Clin. Vaccine Im- Oğuz, H., F. Kurtoglu, V. Kurtoglu, and O. Birdane. 2002. Evalua-
munol. 13:975–980. tion of biochemical characters of broiler chickens during dietary
Harvey, R. B., L. F. Kubena, W. E. Huff, D. E. Corrier, G. E. aflatoxin (50 and 100 ppb) and clinoptilolite exposure. Res. Vet.
Rottinghaus, and T. D. Phillips. 1990. Effects of treatment of Sci. 73:101–103.
growing swine with aflatoxin and T-2 toxin. Am. J. Vet. Res. Parlat, S. S., M. Ozcan, and H. Oguz. 2001. Biological suppres-
51:1688–1693. sion of aflatoxicosis in Japanese quil (Coturnix japonica) by di-
Haskard, C., C. Binnion, and J. Ahokas. 2000. Factors affecting the etary addition of yeast (Saccharomyces cerevisiae). Res. Vet. Sci.
sequestration of aflatoxin by Lactobacillus rhamnosus strain GG. 71:207–211.
Chem. Biol. Interact. 128:39–49. Pasha, T. N., M. U. Farooq, F. M. Khattak, M. A. Jabbar, and
Haskard, C. A., H. El-Nezami, P. Kankaanpää, S. Salminen, and A. D. Khan. 2007. Effectiveness of sodium bentonite and two
J. Ahokas. 2001. Surface binding of aflatoxin B1 by lactic acid commercial products as aflatoxin absorbents in diets for broiler
bacteria. Appl. Environ. Microbiol. 67:3086–3091. chickens. Anim. Feed Sci. Technol. 132:103–110.
Hernandez-Mendoza, A., H. S. Garcia, and J. L. Steele. 2009. Screen- Peltonen, K., H. El-Nezami, C. Haskard, J. Ahokas, and S. Sal-
ing of Lactobacillus casei strains for their ability to bind aflatoxin minen. 2001. Aflatoxin B1 binding by dairy strains of lactic acid
B1. Food Chem. Toxicol. 47:1064–1068. bacteria and bifidobacteria. J. Dairy Sci. 84:2152–2156.
Huynh, V., and R. C. Chubb. 1987. The induction of delayed type Peltonen, K., H. El-Nezami, S. Salminen, and J. Ahokas. 2000. Bind-
hypersensitivity to dinitrochlorobenzene in the chicken. Avian ing of aflatoxin B1 by probiotic bacteria. J. Sci. Food Agric.
Pathol. 16:383–393. 80:1942–1945.
Kabir, S. M. L., M. Rahman, M. B. Rahman, and S. U. Ahmed. Qureshi, M. A., J. Brake, P. B. Hamilton, W. M. Hagler, and S.
2004. The dynamics of probiotics on growth performance and Nesheim. 1998. Dietary exposure of broiler breeders to aflatoxin
immune response in broilers. Int. J. Poult. Sci. 3:361–364. results in immune dysfunction in progeny chicks. Poult. Sci.
Karaman, M., H. Basmacioglu, M. Ortatatli, and H. Oguz. 2005. 77:812–819.
Evaluation of the detoxifying effect of yeast glucomannan on af- Qureshi, M. A., and G. B. Havenstein. 1994. A comparison of the
latoxicosis in broiler as assessed by gross examination and histo- immune performance of a 1991 commercial broiler with a 1957
pathology. Br. Poult. Sci. 46:394–400. random bred strain when fed typical 1957 and 1991 broiler diets.
Poult. Sci. 73:1805–1812.
AFLATOXIN-BINDING BACILLUS PROBIOTIC 1853
Rosa, C. A., R. Miazzo, C. Magnoli, M. Salvano, S. M. Chiac, S. Sur, E., and I. Celik. 2003. Effect of aflatoxin B1 on the development
Ferrero, M. Saenz, E. C. Carvalho, and A. Dalcero. 2001. Evalu- of the bursa of Fabricius and blood lymphocyte acid phosphatase
ation of the efficacy of bentonite from the south of Argentina to of the chicken. Br. Poult. Sci. 44:558–566.
ameliorate the toxic effects of aflatoxin in broiler. Poult. Sci. Surachon, P., P. Sukon, P. Chaveerach, P. Waewdee, and C. Soikum.
80:139–144. 2011. Screening of lactic acid bacteria isolated from chicken ceca
Shahin, A. A. M. 2007. Removal of aflatoxin B1 from contaminated for in vitro growth inhibition of Salmonella enteritica serovar En-
liquid media by dairy lactic acid bacteria. Int. J. Agric. Biol. teritidis. J. Anim. Vet. Adv. 10:939–944.
9:71–75. Teniola, O. D., P. A. Addo, I. M. Brost, P. Farber, K. D. Jany, J. F.
Shane, S. M. 1994. Economic issues associated with aflatoxins. Pages Alberts, W. H. Vanzyl, P. S. Steyn, and W. H. Holzapfel. 2005.
513–527 in The Toxicology of Aflatoxins. L. D. Eaton and D. J. Degradation of aflatoxin B1 by cell-free extracts of Rhodococ-
Groopman, ed. Acad. Press, New York, NY. cus erythropolis and Mycobacterium fluoranthenivorans sp. nov.
Shetty, P. H., and L. Jespersen. 2006. Saccharomyces cerevisiae DSM44556T. Int. J. Food Microbiol. 105:111–117.
and lactic acid bacteria as potential mycotoxin decontaminating Tessari, E. N. C., C. A. F. Oliveira, A. L. S. P. Cardoso, D. R.
agents. Trends Food Sci. Technol. 17:48–55. Ledoux, and G. R. Rottinghaus. 2006. Effect of aflatoxin B1 and
Shi, Y. H., Z. R. Xu, J. L. Feng, and C. Z. Wang. 2006. Efficacy fumonisin B1 on body weight, antibody titres and histology of
of modified montmorillonite nanocomposite to reduce the tox- broiler chicks. Br. Poult. Sci. 47:357–364.
icity of aflatoxin in broiler chicks. Anim. Feed Sci. Technol. Thaxton, J. P., H. T. Tung, and P. B. Hamilton. 1974. Immuno-
129:138–148. suppression in thrombocytes during aflatoxicosis. Poult. Sci.
Shivaramaiah, S., N. R. Pumford, M. J. Morgan, R. E. Wolfenden, 58:562–566.
A. D. Wolfenden, A. Torres-Rodriguez, B. M. Hargis, and G. Tung, H. T., F. W. Cook, R. D. Wyatt, and P. B. Hamilton. 1975.
Tellez. 2011. Evaluation of Bacillus species as potential candi- The anemia caused by aflatoxin. Poult. Sci. 54:1962–1969.
dates for direct-fed microbials in commercial poultry. Poult. Sci. Verma, J., T. S. Johri, D. B. K. Swain, and S. Ameena. 2004. Ef-
90:1574–1580. fect of graded levels of aflatoxin and their combination on the
Shotwell, O. L., C. V. Hesseltine, R. D. Stubblefield, and W. G. performance and immune response of broilers. Br. Poult. Sci.
Sorenson. 1966. Production of aflatoxin on rice. Appl. Microbiol. 45:512–518.
14:425–428. Wolfenden, R. E., N. R. Pumford, M. J. Morgan, S. Shivaramaiah,
Singh, G. S. P., H. V. S. Chauhan, G. J. Jha, and K. K. Singh. A. D. Wolfenden, C. M. Pixley, J. Green, G. Tellez, and B. M.
1990. Immunosuppression due to chronic ochratoxicosis in broiler Hargis. 2011. Evaluation of selected direct-fed microbial candi-
chicks. J. Comp. Pathol. 103:399–410. dates on live performance and Salmonella reduction in commer-
Sneath, P. H. A. 1986. Endospore-forming Gram-positive rods and cial turkey brooding houses. Poult. Sci. 90:2627–2631.
cocci. Pages 1104–1139 in Bergey’s Manual of Systematic Bac- Wolfenden, R. E., N. R. Pumford, M. J. Morgan, S. Shivaramaiah,
teriology. J. G. Holt, ed. Williams and Wilkins Co., Baltimore, A. D. Wolfenden, G. Téllez, and B. M. Hargis. 2010. Evaluation
MD. of a screening and selection method for Bacillus isolates for use
SAS Institute Inc. 1990. SAS User’s Guide: Statistics. SAS Institute as effective direct fed microbials in commercial poultry. Int. J.
Inc., Cary, NC. Poult. Sci. 9:317–323.

You might also like