163
Investigation of the Role of Probiotics on Toxicity of Aflatoxin B1 Muhammad Khalid Tipu Thesis submitted in the partial fulfilment for the requirement of the degree of Doctor of Philosophy May, 2015. University College of Pharmacy, University of the Punjab, Lahore, Pakistan.

Investigation of the Role of Probiotics on Toxicity of

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Investigation of the Role of Probiotics on Toxicity of Aflatoxin B1

Muhammad Khalid Tipu

Thesis submitted in the partial fulfilment for the requirement

of the degree of Doctor of Philosophy

May, 2015.

University College of Pharmacy,

University of the Punjab, Lahore, Pakistan.

Investigation of the Role of Probiotics on Toxicity of Aflatoxin B1

Muhammad Khalid Tipu

Thesis submitted in the partial fulfilment for the requirement

of the degree of Doctor of Philosophy

May, 2015.

University College of Pharmacy,

University of the Punjab, Lahore, Pakistan.

ii

DEDICATED TO MY ALL STUDENTS

iii

Acknowledgments

The first and foremost thanks, prayers and gratitude are for Almighty Allah,

most benevolent and merciful who enables me to accomplish the huge task

with flying color. I pay homage and respects to Holy Prophet Hazart

Muhammad (Peace be upon him) who is source of eternal guidance for whole

mankind in this world and hereafter

I have been lucky enough to have the research supervision of an eminent

researcher and experienced academician, Prof. Dr. Bashir Ahmad(R),

University College of Pharmacy, University of Punjab, Lahore, Pakistan. His

support and guidance in achieving this goal are highly appreciable. He was

always there to be a source of inspiration and encouragement, which guide me

to complete my doctorate thesis.

I am very much obliged to Prof. Dr. Khushi Muhammad, Dean, Faculty of

Veterinary Sciences, University of Veterinary and Animal Science, for his

supervision and personal efforts in my research work and thesis write-up. I am

very thankful to Prof. (R) Dr. Syed Saeed-ul-Hassan, who encouraged me a lot

in completion of my research work.

I am personally grateful to Prof. Dr. Khalid Hussain, Department of

Pharmaceutical Chemistry, University College of Pharmacy, University of the

Punjab, Lahore, Pakistan, for his efforts and contributions in the preparation

of research proposal and the present research work. I also express my

thanks to Prof. Dr. Nadeem Irfan Bokhari, Department of Pharmaceutics,

University College of Pharmacy, University of the Punjab,Lahore,

iv

Pakistan, for his contribution in statistical analysis of the data. I am in very much

debt to Prof. Dr. Mobasher Ahmad, Dean and Principal, University College of

Pharmacy, University of Punjab, Lahore, Pakistan, for facilitating the serum

biochemical analysis.

I am highly grateful to Prof. Dr. Gul Majeed Khan, who allowed me to use the

premises and laboratory facilities of the Department of Pharmacy, Quaid-i-

Azam University, Islamabad, Pakistan, to conduct my research work. I feel

lucky enough to have the company of Dr. Ihsan-ul-Haq (Lecturer) and Dr.

Tofeeq-ur- Rehma (Assistant Professor) at the Department of Pharmacy,

Quaid-i-Azam University, Islamabad, Pakistan. Their encouragement and

fruitful discussion were a source of energy for me to achieve this goal.

I am also grateful to Dr. Zahid Hussain, Livestock Officer, Chiniot,

Pakistan, who facilitated me in immunological analysis of samples. I am

thankful to M r . Abdul Muqeet Khan, Analyst at Quality Operation Lab at

University of Veterinary and Animal Sciences, Lahore, Pakistan, for

assisting me in analysis of the aflatoxin B1 in isolated l iver tissues. I am

also grateful to Dr. Imtiaz Khan, Associate Professor of Pathobiology at Pir

Mahar University of Arid Agriculture, Rawalpindi for histopathological

examination of the tissues. I am highly obliged to Dr. Muhammad Imran,

Assistant Professor, Department of Microbiology, Quaid-i-Azam University, and

Islamabad-Pakistan for his contribution a n d guidance in microbial

characterization of lactobacilli in yogurt (Dhai).

I am personally thankful to Mr. Zahid Hussain (Lab Assistant) and Mr. Faisal

(Lab Attendant) at the Department of Pharmacy, Quaid-i-Azam University,

v

Islamabad, Pakistan, and Mr. Ejaz (Lab Assistant), Noor Hussain and Javed in

Pharmacology Section at University College of Pharmacy, University of Punjab,

Lahore, Pakistan, for their assistance in my research work.

I am in personal debt to Mr. Daud Butt, Mr. Fahad Khan and Mr. Syed

Naqeebullah students of mine at the Department of Pharmacy, Quaid-i-Azam

University, Islamabad, Pakistan for their selfless efforts in fermentation of rice

for AFB production. I thank Mr. Abdul Hadi, owner of Pakeeza Chicks,

Islamabad, Pakistan a n d his team for providing the housing facilities for

broiler birds during my research work.

Last but not least I am also thankful to Ms. Zainab, Mr. Ali Hassan and Mr.

Shaheer who because of t h e i r innocent queries/question, kept me charged

and determined to complete the task in time.

Muhammad Khalid Tipu

vi

TABLE OF CONTENTS

Page

CERTIFICATE OF APPROVAL i

DEDICATION ii

ACKNOWLEDGMENTS iii

LIST OF TABLES ix

LIST OF FIGURES xi

LIST OF PLATES xiii

LIST OF ABBREVIATIONS xiv

ABSTRACT xvi

CHAPTER 1 Introduction 1

CHAPTER 2 Review of Literature 5

2.1 Aflatoxins 5

2.1.1 Historical Review 5

2.1.2 Toxicokinetics 6

2.1.3 Toxicodynamics 7

2.1.4 Aflatoxicosis 8

2.1.4.1 Acute aflatoxicosis 8

2.1.4.2 Chronic aflatoxicosis 9

2.1.4.3 Occurrence AFB in Pakistan 16

2.2 Control of Aflatoxins 17

2.2..1 Agricultural Control 18

2.2.1.1 Pre-harvest control 18

2.2.1.2 Post-Harvest control 19

2.2..2. Dietary control 19

2.2..2.1 Chemopreventive agents 20

vii

2.2.2.2 Adsorbents 24

2.2.2.2.1 Mycosorb (MYC) 25

2.2.2.2.2 Probiotics (Lactobacilli) 29

2.3 Justification 37

2.4 Aims and Objectives 39

CHAPTER 3 Material and Methods 41

3.1 Plan of Study 41

3.2 Preparation of feed containing different treatment 41

3.2.1 Production of AFB 41

3.2.2 Preparation of PBT 44

3.2.3 Preparation of SLM containing feed 44

3.2.4 Preparation of MYC containing feed 45

3.3 Sampling 45

3.3.1 Collection of blood and serum 45

3.3.2 Collection of internal organ 45

3.4 Analysis of samples 45

3.4.1 Determination of NDV antibody response 45

3.4.1.1 Preparation of RBCs 46

3.4.1.2 Preparation of 4HA unit of virus 46

3.4.1.3 Determination of BDV antibody titre 47

3.4.2. Determination of AFB residue in liver 51

3.4.3 Clinical biochemistry 55

3.4.3.1 Total Serum protein 55

3.4.3.2 Serum Albumin 55

3.4.3.3 Serum bilirubin 56

3.4.3.4 Serum Glutamate pyruvate transferase 56

viii

3.4.3.5 Serum creatinine 57

3.4.3.6 Serum blood urea nitrogen 57

3.4.4 Histopathological examination 58

3.5 Statistical analysis 58

Chapter 4 Results 60

4.1 NDV antibody titre 60

4.2 Total leukocytes count 64

4.3 Relative weight of bursa of Fabricius 67

4.4 Relative weight of spleen 69

4.5 Relative weight of liver 72

4.6 Total body weight 73

4.7 Total serum protein 77

4.8 Serum albumin 80

4.9 Serum glutamate pyruvate transferase 82

4.10 Serum bilirubin 85

4.11 Serum blood urea nitrogen 88

4.12 Serum Creatinine 90

4.13 AFB-residue in liver 92

4.14 Histopathological examination 94

Chapter 5 Discussion and Conclusion 99

Chapter 6 References 111

ix

LIST OF TABLES

Table No. Title Page

No.

2.1 Dietary Control of Aflatoxins 21

3.1 General plan for Hemagglutination (HA) test 48

3.2 General plan for Hemagglutination inhibition (HI) test 49

3.3 Table of Brugh 50

4.1 Mean of LOG of antibody titer of birds taking different

therapies for two week 60

4.2 The Distribution of serum of birds given various

treatments according to Well Number in HI Test 61

4.3 Mean of total leukocytes counts (cells×1000/µl) in birds

taking different therapies for two week 66

4.4 Mean relative weight (g/Kg) of bursa of Fabricius of birds

taking different therapies for two week 67

4.5 Mean relative weight (g/Kg)of spleen of birds taking

different therapies for two week 69

4.6 Mean relative weight of liver (g/Kg) of birds taking

different therapies for two week 73

4.7 Mean total body weight (Kg) of birds taking different

therapies for two week 75

4.8 Mean total serum protein level (g/dl) of birds taking

different therapies for two week 77

4.9 Mean total serum albumin level (g/dl) of birds taking 80

x

different therapies for two week

4.10 Mean SGPT (U/L) of birds taking different therapies for

two week 83

4.11 Mean serum bilirubin level (mg/dl) of birds taking different

therapies for two week 85

4.12 Mean serum BUN level (mg/dl) of birds taking different

therapies for two week 88

4.13 Mean serum creatinine level (mg/dl) of birds taking

different therapies for two week 90

4.14 Mean of AFB residue in liver of birds taking different

therapies for two week 92

xi

LIST OF FIGURES

Figure

No. Title

Page

No.

2.1 Chemical Structure of Aflatoxins 5

2.2 Metabolism of Aflatoxins 7

2.3 Mechanism of AFB toxicity 8

2.4 Mechanism of action of adsorbents 24

2.5 Mechanism of Lactobacilli in aflatoxicosis 32

3.1 Standard of AFB 52

3.2 AFB in bird of group III at 4th week of age 53

3.3 AFB in bird of group VI at 4th week of age 53

3.4 AFB in bird of group VI at 6th week of age 54

3.5 AFB in bird of group VII at 6th week of age 54

3.6 AFB in bird of group IV at 4th week of age 55

4.1 Effect of different treatments on NDV antibody titre 62

4.2 Effect of different treatments on total leukocytes counts 65

4.3 Effect of different treatments on relative weight of bursa of

Fabricius

68

4.4 Effect of different treatments on relative weight of spleen 70

4.5 Effect of different treatment on relative weight of liver 74

4.6 Effect of different treatment on total body weight 76

4.7 Effect of different treatment on total serum protein 78

4.8 Effect of different treatment on serum albumin level 81

4.9 Effect of treatment on serum GPT level 84

xii

4.10 Effect of treatment on serum bilirubin level 87

4.11 Effect of treatment on serum BUN level 89

4.12 Effect of treatment on serum Creatinine level 91

4.13 Effect of treatment on liver AFB residue level 93

xiii

LIST OF PLATES

Page No.

a) Bursal follicle (10X) in PBT treated Birds 96

b) Bursal follicle (40X) in PBT treated Birds 96

c) Depleted Bursal follicle (10X) in AFB treated Birds 96

d) Depleted Bursal follicle (40X) in AFB treated Birds 96

e) Kidney (40X)of MYC for two weeks treated 97

f) Kidney (40X)of AFB for two weeks treated 97

g) Kidney (40X)of SLM for two weeks treated 97

h) Kidney (40X)of PBT for two weeks treated 97

i) Liver (10X) of AFB treated birds for two weeks 98

j) Liver (40X)of AFB treated birds for two weeks 98

k) Liver(10X) of PBT treated birds for two weeks 98

l) Liver (40X)of PBT treated birds for two weeks 98

xiv

LIST OF ABBREVIATIONS

AFB Aflatoxin B1

AFM Aflatoxin M1

ALT Alanine aminotransferase

AOAC Association of analytical chemists

AST Aspartate transaminase

BF Bursa of Fabricius

BUN Blood urea nitrogen

CYP Cytochrome

EG Esterified glucomannan

FAO Food and Agricultural organization

FDA Food and Drug Administration

GIT Gastrointestinal tract

GMT Geometric mean titre

HA Heamagglutination

HCS Hydro pericardium syndrome

HI Heamagglutination inhibition

HPLC High performance liquid chromatography

HSCAS Hydrated sodium calcium aluminosilicate

IARC International Agency for Cancer

research

IB Infectious bronchitis

IBD Infectious bursal disease

xv

MYC Mycosorb

NDV Newcastle disease virus

PBT Probiotics

ppb Part per billion

ROS Reactive oxygen species

SGOT Serum glutamic oxaloacetate transferase

SGPT Serum glutamic pyruvic transaminase

SLM Silymarin

SOD Super oxide dismutase

TLC Total leukocytes counts

TBW Total body weight

TSP Total serum Protein

UV/VIS Ultraviolet and visible spectroscopy

WHO World Health Organization

xvi

Investigation of the Role of Probiotics on Toxicity of Aflatoxin B1

Abstract

The aflatoxins are unavoidable contaminant of feed and food commodities.

Among these, aflatoxin B1 (AFB) has established hepatotoxic, immuno-toxic and

carcinogenic properties. AFB is grouped as class I carcinogenic by International

Agency for Cancer Research (IARC). Various methods to prevent dietary exposure

of AFB are: use of chemopreventive agents like Silymarin (SLM) and adsorbents

e.g. mycosorb (MYC). Among adsorbents, Probiotics (Live Lactobacilli) have

exhibited good AFB binding properties in vitro as well as in vivo. Traditional

Pakistani yogurt (Dhai) has been reported to be good source of such Lactobacilli.

The current study was designed to evaluate protective effect of these Lactobacilli

(PBT) species against AFB toxicity on liver, immunity and kidneys.

One day old broilers (n=240) were reared under standard environmental

conditions. On 3rd week of age, the broilers were segregated into different treatment

groups: I (Basal diet), II (300 g of yogurt [PBT 1X] ), III (400 ppb of AFB), IV ( 600 g

of yogurt [PBT 2X]+400 ppb of AFB), V (300 g of yogurt [PBT 1X]+400 ppb of AFB),

VI (SLM 600mg/Kg body weight+400 ppb of AFB), VII (MYC 1g/Kg of feed+400 ppb

of AFB). The above treatments were continued for two weeks i.e. 4th & 5th week of

age. All birds were vaccinated against regional prevailing diseases such as

Newcastle disease, Infectious bursal disease, etc.

Two ml blood was collected on 4th, 5th, 6th and 7th weeks of age for

measuring leukocytes count, serum antibody titre against Newcastle Disease Virus

(NDV) and clinical chemistry. Birds after weighing were euthanized and internal

xvii

organs such as liver, spleen, bursa of Fabricius and kidney were collected for

histopathological examination and determination of AFB residue in the liver. Data

thus obtained were analyzed by two way ANOVA with LSD test (α=0.05) for multiple

comparison. Ingestion of AFB resulted in significant decline in total body weight and

relative weight of spleen and bursa of Fabricius, serum NDV antibody titer, total

serum proteins and serum albumin. Moreover, significant rise in relative weight of

liver, serum GPT, bilirubin and AFB residue in the liver were recorded in AFB-treated

birds (p<0.05). Histopathological examination revealed vacuolar degeneration, bile

duct hyperplasia and loss of hepatic chord in the AFB-treated birds.

MYC intake significantly restored the negative effects of AFB in birds by its

adsorptive action during exposure (p<0.05). The SLM intake caused substantial

protection against harmful effects of AFB but the effect appeared on second week of

exposure (p<0.05). However, protective effects of both SLM and MYC were lost

when intake was stopped. Birds receiving PBT showed better NDV-antibody titer,

normalized organs weight, serum total proteins, albumin, GPT and bilirubin (p<0.05)

Histopathological findings also reflected shielding effects of PBT. The protective

action of PBT was observed during the exposure as well as post exposure. It is

concluded that Lactobacilli (PBT) in the Pakistani food significantly ameliorate the

negative effect of AFB on immune system and liver presumably via its adsorptive

properties which result in declined AFB bioavailability. Furthermore, investigations

are needed to elucidate mechanism of protective action of such PBT persisting even

after stopping of intake of PBT.

1

1. INTRODUCTION

Aflatoxin B1, B2, G1 and G2, are fungal secondary metabolites, which

are produced by some strains of Aspergillus flavus and Aspergillus

parasiticus (Wei and Jong, 1986; Karimi Torshizi et al., 2010). These are natural

contaminants of raw materials used for preparation of feed and food

commodities. Among these aflatoxin B1 (AFB) is classified as group I

carcinogen in humans (IARC, 2002). Additionally, it has ability to produce

hepatotoxic, carcinogenic and immunotoxic effects. These toxins enter into

the body through contaminated feeds/foods, and are either metabolized or

deposited in liver to cause deleterious effects. In lactating animals AFB and

aflatoxin B2 are excreted in milk such as aflatoxin M1 (AFM) and aflatoxin M2

(El-Nezami et al., 1995; Prandini et al., 2009). First record about the toxicity of

aflatoxins appeared in literature in 1960 when increased rate of death in turkey

poultry and ducklings was found to be due to contaminated peanut oil used in

the production of poultry feed, because the aflatoxins were isolated from the

feed (Hartley et al., 1963). The toxins were found to be involved in outbreak of

liver disorders and other symptoms of aflatoxicosis resulting in human deaths

in various part of the world such as India (Krishnamachari et al., 1975a; Tandon

et al., 1978) and Kenya (Ngindu et al., 1982; Azziz-Baumgartner et al., 2005).

The source was found to be the contaminated g ra in used for the

preparation o f food.

Aflatoxicosis is of two types: acute poisoning resulting in hemorrhagic

necrosis of liver, proliferation of bile duct, edema and lethargy and death

due to liver cirrhosis (Tandon et al., 1978; Azziz-Baumgartner et al., 2005)

2

and chronic sub-clinical exposure causing nutritional disorder (Gong et al.,

2002), immunosuppression (Meissonnier et al., 2008b), mutagenicity (Wild and

Turner, 2002) and carcinogenic consequences (Williams et al., 2004). These

toxins can cause toxicity in respiratory tract by exposure through inhalation

(Kelly et al., 1997). The immunosuppressant effects are the major health and

economic problems for both animals as well as humans which results in failure

of immune-prophylaxis (Dimitri and Gabal, 1996; Gabal and Dimitri, 1998).

Because of their profound toxicity, amount of these toxins in food is

essential to be regulated. The permissible limit by FDA is 20 ppb of AFB for

human food with exception of milk which is 0.5 ppb for AFM (JECFA, 2001). The

maximum level set by the European Union is 2 µg/Kg of AFB in food stuff

intended for direct human consumption whereas 0.10 µg/Kg of the AFB in food

for infants and special medical purposes. According to OJL, the level of AFM

is 0.05 µg/Kg and 0.025 µg/Kg for milk products for infant and special medical

purposes, respectively (Regulation, 2006).

Several strategies have been established to remove these toxins from

food commodities such as proper maturing of crops, minimizing post-harvest

damage and improving storage conditions. Sequestering a gen t s (adsorbents)

are also used in poultry feed and animal ration to reduce the aflatoxins

exposure. However, s ome poor quality adsorbents also adsorb essential

nutrients like vitamins resulting in stunted growth of animals, and

also show irreproducible results in experiments (Chung et al., 1990). On

account of these limitations these adsorbents are not used in human food.

Therefore, many researchers have started using live cultured microbes

(Probiotics) l i ke bacteria, yeast and molds to reduce the level of the toxins in

foods and feeds.

3

The term probiotics (PBT) was first used by Lilly and Stillwell in

1965 (Lilly and Stillwell, 1965). The concept of PBT arouse from study of Nobel

laureate Eli Metchnikoff, who linked the longer longevity of Bulgarian peasants

with consumption of fermented milk products (Gupta and Garg, 2009).

According to the Food and Agriculture Organization (FAO) and the World

Health Organization (WHO) PBT are live micro-organisms which, when

administered in adequate amounts confer health benefits on the host

(Quigley, 2010). The most commonly used bacterial genera as PBT are

Lactobacillus, Bifidobacterium, Enterococcus, and Streptococcus. Some fungal

strains belonging to Saccharomyces have also been used as PBT (Jin et al.,

2000; Alvarez-Olmos and Oberhelman, 2001). PBT are beneficial in

different pathological conditions such as antibiotic associated diarrhea

(D'Souza, 2002), Helicobacter pylori infection (Lesbros-Pantoflickova et al.,

2007) and cancer (Aso et al., 1995). Apart from these effects, Lactobacillus

and Bifidobacteria are reported to reduces level of free mycotoxin, cyanotoxin

and heavy metals in aqueous solution (Salminen et al., 2010a). The same

has been reported by various scientists that different strains of

Lactobacilli and Bifidobacteria on incubation with aflatoxins containing culture

media reduce the level of the free aflatoxins, either by binding or metabolizing

the toxin (Peltonen et al., 2001b; Fazeli et al., 2009b; Hernandez-Mendoza et al.,

2009a).

Due to humid and warm climate in Pakistan, grains/foods are more

susceptible to aflatoxins contamination (Mobeen, 2011). In Pakistan, all four

aflatoxins have identified at level higher than permissible limit in both various

unprocessed and processed food (Mushtaq et al., 2012). The wheat, the staple

food of Pakistan, is harvested and stored during the summer. The stores houses

4

in the country lack proper system to control temperature and humidity which

encourage the fungal growth. The Dhai is traditional dairy food product of

Pakistan and subcontinent which is prepared by fermentation like kefir and

yogurt in Europe. The traditional fermented milk product is prepared by mixing

raw and heat treated milk with starter culture (Jhaag: local term) containing

Lactobacillales as dominating microbial group while it also have other undefined

complex microbial consortia for fermentation. Dhai serves as very good and

economical source of probiotic Lactobacillales (Aslam and Qazi, 2010).

In Ayurveda, Dahi is advised in various liver and kidney aliments;

moreover it is also suggested in various type of intoxication as folklore remedy.

The probiotics from Dahi (Yogurt) for example Lactobacilli, have been used in

alcoholic liver disease, non-alcoholic liver disease, minimal hepatic

encephalopathy and overt hepatic encephalopathy (Hitchins and McDonough,

1989; Lewis and Freedman, 1998; Bajaj et al., 2008; Nabavi et al., 2015).

We hypothesized that low priced freely available fermented milk product

containing probiotics will be beneficial in combating aflatoxicosis related

economical and health issues of a developing country like Pakistan. The current

study was designed to evaluate whether the traditional product offers a

comparable benefit to established strategies (Silymarin and mycosorb) for

prevention /treatment of aflatoxicosis.

5

2. Review of Literature

2.1. Aflatoxins

Mycotoxins are the secondary metabolites produced by certain fungal

species. These highly toxic compounds are produced as natural protectant. But

these become a serious health and economical problem when fungus

contaminates the crops/grains used for human and animal food. Aflatoxins are

produced by mainly Aspergillus flavus and Asergillus parasiticus (Wei and Jong,

1986). The word ‘Afla’ is combination of ‘A’ from Aspergillus and ‘fla’ from flavus

(Schoental, 1967). There are about 18 different mycotoxins identified, but AFB,

aflatoxin B2, G1 and G2 are more common. AFM and aflatoxin M2 are metabolites

of AFB and aflatoxin B2 (Wu et al., 2009). These compounds are coumarone

derivative showed intense blue and green fluorescence in UV light. Hence named

B (blue) and G (green) are used. The chemical structures of these are shown in

Fig 2.1.

Fig. 2.1: Chemical Structures of Aflatoxins (Williams et al., 2004)

2.1.1. Historical Review

6

The first record about aflatoxins appeared in 1960 when unexpected high

death rate observed in turkey poultry, was traced to consumption of highly

contaminated feed (Nesbitt et al., 1962). These compounds were discovered by

Hartley in 1963. Hartley reported the isolation and chemical characteristics of the

toxic metabolites of Aspergillus flavus. The four toxic compounds were isolated

from sterilized groundnuts which were inoculated by the fungus. The mixture of

AFB, aflatoxins-B2, -G1 and -G2 were separated and recrystallized using various

solvents by column chromatography. Then AFB was re-crystallized using

trichloroethylene and chloroform (Hartley et al., 1963). In 1966, Shotwell and his

co-workers reported a method of production of the aflatoxins by culturing the

fungus (NRRL 2999) on fermented rice. AFB was separated from almost all

impurities and from the other aflatoxins by chromatography on silica gel with 1%

ethyl alcohol in chloroform which was further purified by recrystallization

(Shotwell et al., 1966).

2.1.2. Toxicokinetics

Because of lipophilicity and low molecular weight, AFB is rapidly absorbed

in gastrointestinal tract. It reversibly bound mainly to albumin in plasma. After

distribution, AFB is mainly accumulated in liver, muscle, and Kidney (Smith and

Henderson, 1991). In another study higher concentration (3 µg/Kg) of aflatoxins

were found in kidney, gizzard and liver of broilers given mixture of AFB and

aflatoxin B2 for four weeks (Wolzak et al., 1986). In a study, Mico and his

colleagues found detectable amount of AFB, AFM, aflatoxin B2 and aflatoxicol in

liver, kidney and thigh muscle of broiler fed diet containing 50 ppb of aflatoxins

(Micco et al., 1988). AFB is oxidized by cytochrome P450 enzyme especially

CYP2A6 and CYP1A1 to various metabolites: AFB-8,9-exo-epoxide, AFB-8,9

endo-epoxide, AFM, aflatoxin P1 and aflatoxin Q1. Reduction of AFB produces

7

aflatoxicol which can be detected in liver (Fernández et al., 1994; Diaz et al.,

2010; Yunus et al., 2011a).The AFB-8,9-epoxide is detoxified by glutathione-S-

transferase to AFB-mercapturic acid which is excreted in urine. Metabolites of

aflatoxin P1, aflatoxin Q1 and aflatoxicol are inactive and excreted by renal and

biliary route after conjugation with glucuronic acid or sulfate (Yunus et al., 2011a;

Gross-Steinmeyer and Eaton, 2012). The metabolism of AFB is illustrated in Fig

.2.2.

Fig.2.2 Metabolism of AFB, GST: Glutathione-S-transferase, UDPGT: Uridine-diphosphate glucuronosyltransferase, ST: Sulphotransferase, CYP450(s): Cytochromes P450.

2.1.3. Toxicodynamics

The toxic effects of AFB are exerted by its epoxide metabolite. The highly

reactive metabolite covalently binds with macromolecules. It forms AFB-Lysine

adduct with serum albumin which is one of biomarker for aflatoxin exposure. The

epoxide metabolite is converted to dihydrodiol by epo-hydrolase. Binding of both

8

epoxide and dihydrodiol with cellular proteins result in cell death and toxicity

(Gross-Steinmeyer and Eaton, 2012). Secondly the epoxide metabolite form

adduct with N-7 in guanine in DNA. The adduct strand either undergoes

depurination or the AFB-N(7)-guanine adduct is converted to open ring

formamidopyrimidine. Both forms cause mutation in p-53 gene or activate the ras

oncogene (Eaton and Gallagher, 1994; Smela et al., 2002; Bedard and Massey,

2006). The mechanism of toxicity is shown in Fig. 2.3

Fig.2.3 Mechanism of AFB toxicity

2.1.4. Aflatoxicosis

The exposure to the aflatoxins causes deleterious manifestation in

humans, animals and birds. These effects are collectively termed as aflatoxicosis.

The aflatoxicosis is of two types:

2.1.4.1. Acute Aflatoxicosis

9

Acute aflatoxicosis occurs due to excessive intake of aflatoxins particularly

AFB in contaminated diet. Symptoms include vomiting, abdominal pain, hepatitis

and death. The lethal dose of AFB is about 10-20 mg for adult (Etzel, 2002).

Different outbreaks of aflatoxicosis have been reported by various scientists in

different part of world. Krishnamachari and his colleagues described an outbreak

of hepatitis in 1975. The outbreak affected the human and even street dogs. The

symptoms observed were jaundice, ascites and portal hypertension with high

death rate. The autopsy revealed the proliferation of bile duct and giant cells. The

cause was traced to consumption of 2-6 mg of the aflatoxins for a month due to

use of highly contaminated maize as staple food (Krishnamachari et al., 1975b).

Another outbreak of liver disease with jaundice and ascites was investigated by

Tandon and his coworkers in some rural area of India. The corn sample had

shown high level of AFB (Tandon et al., 1978). In 1982 Similarly Ngindu with his

colleagues observed hepatitis in 20 people with 60 % mortality. The liver of

deceased contained about 89 ppb of AFB (Ngindu et al., 1982).

In 1991 an outbreak of aflatoxicosis was described in Malaysia. There was

death of 13 children associated with consumption of local food contaminated with

the aflatoxins. The cause of death was acute hepatic and renal failure (Cheng,

1992). An outbreak of aflatoxicosis was recorded in 2004 which claimed 317

sufferings with 125 deaths. The symptoms followed somewhat same pattern as

described by Etzel, 2000. The causative agent was AFB, confirmed by analysis of

corn consumed, AFB-lysine adduct and positive Hepatitis B surface antigen in

blood of affected ones (Azziz-Baumgartner et al., 2005; Probst et al., 2007).

2.1.4.2. Chronic Aflatoxicosis

The chronic aflatoxicosis results from low but continuous ingestion of AFB

from environment. The low level contamination does not cause sudden necrosis

10

or other sign of acute toxicity. Rather it results in different effects with prolonged

consequences. These are:

I. Immunosuppressive Effects

The laboratory and farm animals (chronic exposure) are main source of

immunological effects. The variability of the aflatoxins level in food commodities

and biomarkers of the toxins are main hindrance for generalization of the results.

However these studies are linked to human in broader term (Williams et al.,

2004). The deleterious effects of AFB on various cells of immune system are as

follow:

a. Effects on Dendritic Cells

Dendritic cells play a central role in immune response to pathogens. They

are produced by distinct hematopoietic lineage (Merad et al., 2013). The classical

type is present in most of the tissue and triggered immune response by

presenting the antigen to T-lymphocytes, phagocytic property and production of

cytokines (Geissmann et al., 2010). AFB ingestion resulted in impairment of their

phagocytic activity and T-cell stimulating ability in porcine (Mehrzad et al., 2014)

b. Effect on Lymphocytes

. AFB had detrimental effects on lymphocytes. AFB produced oxidative

stress indicated by reduced glutathione peroxidase, glutathione reductase and

catalase activity in broiler chickens (Chen et al., 2013a). AFB caused decrease in

cells proliferation in Jurkat T-cell line and increased expression of IL-8 (Luongo et

al., 2013). Similarly in human lymphocytes, AFB induced cytotoxic effects. It

caused decrease in cellular oxygen consumption and mediated apoptosis as

indicated by intracellular activity of caspase and annexin-V positive cells. These

effects were produced within two hours at 100 µM concentration and 24 hours at

50 µM concentration (Al-Hammadi et al., 2014).

11

c. Effects on Macrophages:

Macrophages have imperative role in both innate and acquired immunity.

They not only have role in phagocytosis but also act as antigen presenting cells

(Rossi et al., 1986). In swine, AFB exposure at concentration of 1.5 µg/ml for 24

hours reduced viability of alveolar macrophages to 41% and expression of

apoptosis-related heat shock protein 72 (HSP 72). The reduction in phagocytic

activity (36% of control) was observed in same at level of 100 ng/ml for 24 hours

(Liu et al., 2002). The secretions of various cytokines was also affected by AFB.

It reduced anti-inflammatory cytokine IL-10 while enhanced the pro-inflammatory

cytokine IL-6. The expression of toll like receptor 2 (TLR2) and differentiation

CD14 was reduced significantly by AFB in murine macrophages (Bruneau et al.,

2012).

d. Cell Mediated Immunity:

Dimitri and Gabal had described the immunosuppressant activity of the

aflatoxins in rabbits where AFB showed reduction in lymphocyte stimulation and

abolished the tuberculin test in the treated rabbits. The lymphocyte stimulation

indices and diameter of skin reactions were significantly reduced in the AFB-

treated groups. In addition, rabbits (25%) from the AFB-treated groups failed to

produce any detectable response to the tuberculin test. It was concluded that

AFB inhibited the lymphocyte proliferation and negatively influenced the

tuberculin skin test (Gabal and Dimitri, 1998).

Similarly Meissonnier and colleagues studied the impact of dietary AFB on

cell mediated immunity in pigs. The results of experiment indicated an increase in

the level of pro inflammatory and regulatory cytokines. These findings suggested

that AFB causes reduction in cell mediated immunity coupled with inflammatory

response (Meissonnier et al., 2008b).

12

e. Humoral Immunity:

AFB exposure decreased the antibody titer (enzyme-linked

immunosorbent assay) and antibody level (protein electrophoresis) in a study

conducted by Gabal and Dimitri (1998). It was found that antibody titer and the

immunoglobulin levels were significantly decreased in the aflatoxin-treated

groups. Chen and co-workers reported decrease in serum immunoglobulin,

relative weight of bursa of Fabricius. Moreover, increase in percentage of

apoptotic bursal cells was observed (Chen et al., 2014a)

II. Hepatotoxic Effects

a. Liver Functions:

Chronic exposure to the aflatoxins especially AFB produced a direct injury

to hepatocytes causing macroscopic, microscopic lesions and impairing their

functions and consequently the serum profile. Exposure to 100 ppb of AFB in

broilers resulted in elevated level of gamma glutamyl transferase (GGT),

glutamate-oxaloacetate transferase (GOT) and glutamate-pyruvate transferase

(GPT). There was significant accumulation of AFB residue in liver than muscle

(Bintvihok and Kositcharoenkul, 2006). Similarly, Chen and his colleagues

observed the negative effect of AFB in duckling. They reported that AFB (110 to

210 ppb) given for 14 days, reduced serum glucose, protein and albumin level in

duckling. While the serum aminotransferases, alkaline phosphatase (ALP) and

serum urea nitrogen were elevated by increasing the AFB level (Chen et al.,

2014e).

In a study conducted in broilers by Denli and colleagues, AFB (1mg/Kg of

feed) significantly increased the liver weight and serum alkaline phosphatase.

Perilobular inflammation along with vacuolar degeneration was also observed in

histopathological examination of liver (Denli et al., 2009). Similarly, Weaver and

13

co-workers reported the hyperplasia of liver bile ductule and karyomegaly in pig

upon exposure to combination of 150 ppb of AFB and 1100 ppb of

deoxynivalenol (Weaver et al., 2013). Yang and colleagues studied effect of

contaminated corn in broilers. They reported that an increment in contamination

level adversely affected the liver histology with rise in apoptosis. Reduction in

total hepatic proteins and glutathione peroxidase were also observed at more

than 50% of contamination level. Whereas, rise in activity of glutathione-S-

transferase (GST), glutathione reductase and superoxide dismutase (SOD) along

with high hepatic malanodialdehyde level were also observed (Yang et al.,

2012).

b. Carcinogenesis:

Liu and Wu reported that long term exposure to AFB was linked to

increased risk of hepatic cell carcinoma (HCC), (Liu and Wu, 2010). William and

co-workers described the higher HCC prevalence (16 to 32 times) in developing

countries especially in sub-Saharan Africa and Asia-Pacific region was related to

higher exposure of AFB (Williams et al., 2004). Wild and Turner described the

liver as major site for activation of AFB (Wild and Turner, 2002). The metabolic

activation of AFB into AFB-8, 9-exo-epooxide metabolite mainly by CYP3A4 was

studied by Kamdem and colleagues. They also studied relative contribution of

CYPs in activation and detoxification of AFB. They suggested that inhibition of

CYP3A4 might be alternative chemoprevention (Kamdem et al., 2006). Mulder

and his colleagues studied role of AFB metabolite-DNA adduct in mutation and

consequently carcinogenesis by using the p53 gene knockout mice (Mulder et al.,

2014). Kirk and his team described that mutation by the AFB metabolite-adduct

at codon 249 (AGG to AGT: Arginine to Serine) in tumour suppressor p53 gene

and infection with hepatitis B virus had synergistic effect in HCC (Kirk et al.,

14

2005). William and his colleagues also reported an increment in odd ratio with

both AFB exposure and hepatitis B virus (HBV) infection than AFB alone

(Williams et al., 2004). Kew studied the impact of HBV X gene and protein in

induction of HCC. He studied the anti-apoptotic role of HBV X protein and its

inhibitory effect on the p-53 gene. The protein also interfered with other

nucleotide excision repair mechanism either p-53 gene dependent or

independent (Kew, 2011).

III. Nutritional Toxicity

Consumption of the aflatoxins resulted in reduced weight gain and

negative effect on nutritional status. Marin with his colleagues reported dose

dependent reduction in weight gain in weaning piglet given 140 and 280 ppb of

AFB (Marin et al., 2002). Similarly significant weigh reduction, feed consumption

and feed conversion ratio (FCR) were reported by Verma and co-workers in

broiler chickens (Verma et al., 2004). In a cross sectional study, Gong and her

colleagues observed stunted growth and low weight in children less than 5 year

in Benin and Togo. They determined the AFB exposure by measuring the AFB-

albumin adducts which ranged from 5 to 1064 Pico g/mg of albumin in 99% of

children (Gong et al., 2002).

Obuseh and co-workers studied the relation between AFB-albumin adduct,

vitamin A and E levels in HBV infected patient having either Human

Immunodeficiency virus (HIV) or none. They measured the level of AFB-albumin

adduct, vitamin A and E in HIV positive and negative patients. Low serum vitamin

A and E were correlated with the higher AFB-albumin adduct level (Obuseh et al.,

2011). Tang with his colleagues reported the negative co-relation between

vitamin A and E level with AFB exposure. They determined the AFB-albumin

adduct, vitamin A and vitamin E level in 507 Ghanaian peoples. High exposures

15

of AFB caused decrease in serum vitamin A and E level which influenced the

aflatoxin deleterious effects (Tang et al., 2009). Hendrickse reported that AFB

exposure modulated the recovery from Kwashiorkor by affecting the protein

synthesis (Hendrickse, 1997) .

Costanzo and co-workers studied the impact of AFB on vitamin D receptor

in osteosarcoma cell line SAOA-2. They reported that exposure to 5 to 50 ng of

AFB down regulated vitamin D receptor (58% and 86% respectively) in the cell

line. They suggested that early exposure with AFB might increase the risk of

rickets in African children (Costanzo et al., 2014). Hussein and his colleagues

studied the relation between AFB-adduct and oxidative stress in wheat milling

workers. They measured AFB-adduct, GOT, GPT, GGT, ALP, GST, SOD,

malaonodialdehyde, zinc and vitamin C. Significant correlation of zinc with GST

and SOD activities showed the role of Zn in oxidative stress induced by AFB.

Similarly positive correlation of vitamin C with GST while negative with the

malaonodialdehyde, indicated the role of the antioxidant vitamins in AFB toxicity

(Saad-Hussein et al., 2014).

IV. Inhalation Toxicity

Apart from all these deleterious effects, inhalation of the AFB in worker of

poultry and concerned industries, is becoming alarming. Kelly and co-workers

studied the impact of inhalation exposure to AFB on human lung because

inhalation of AFB in certain occupations was considerable. The data indicated

that human lung microsomes activated AFB to form the exo-AFB-8, 9-epooxide

and that cytochrome P450 (3A subfamily) might be responsible for this activity.

The relatively low amount of AFB activation in human lung compared to that

in human liver could be explained by the scarcity of cytochrome P450 containing

cells in the lung. In situ AFB activation and resultant carcinogenic risk were

16

distinctly possible in occupational settings where inhalation of AFB contaminated

dusts occurred (Kelly et al., 1997). Shen and his team studied toxicity of aflatoxin

G1 in rats. He administered the toxin through intra-tracheal route. After

determination of surfactant protein, they conferred that alveolar type II cells were

the target for acute toxicity (Shen et al., 2012). Study conducted on poultry

workers in Portugal necessitated the assessment and prevention of inhaled AFB

exposure to worker in such work places (Viegas et al., 2012).

2.1.4.3. Occurrence of AFB/Aflatoxicosis in Pakistan

Due to humid and warm climate in Pakistan, grains/foods are more

susceptible to aflatoxins contamination (Mobeen, 2011). In Pakistan, all four

aflatoxins have been identified at level higher than permissible limit in both

various unprocessed and processed food (Mushtaq et al., 2012). A study was

conducted to evaluate contamination of aflatoxins in animal feed and raw feed

from 2006 to 2009. It was revealed that 61% of samples collected were

contaminated with AFB and other aflatoxins. Moreover, the contamination level

was more than the permissible limit i.e. 20 ppb (Khan et al., 2011).

Another study revealed that more than 80% of wheat samples were

contained AFB more than the permissible limit. Impact of weather was significant

on AFB level in wheat grains (Rashid et al., 2012). Anjum and colleagues

analyzed the 100 sample of starter feed for broiler and layer for aflatoxin and

ochratoxin. It was found that 40 % of samples contained higher level of both

mycotoxins (Anjum et al., 2011). A study was conducted to determine occurrence

of aflatoxicosis in broilers. Sick and dead birds (n=1105) were examined from

June 2009 to May 2010. It indicated that 8.78 % of birds were aflatoxicosis

positive. Necropsy findings showed pale liver, atrophied bursa and thymus.

17

Occurrence was more in autumn season as compared to winter (Rashid et al.,

2013).

Aslam and his colleagues studied the level of AFM in milk supplied

through various supply chains. It was found that AFM concentration was higher

than permissible limit. AFM concentration was 2.60, 2.59 and 1.93 ppb in

autumn, rainy (monsoon) and summer season respectively (Aslam et al., 2016).

An incident of aflatoxicosis was reported bovine farm. It affected 45 animals with

30% mortality. Postmortem investigation revealed visceral hemorrhage, prolapse

and damaged hepatic portal system (Sohooa et al., 2015).

Despite of high level of aflatoxin and aflatoxicosis in broiler and cattle,

Aflatoxicosis either acute or chronic in Pakistani population has not been

reported. However, Qureshi and colleagues suggested aflatoxins contamination

(10-17%) was positively correlated with higher incidence of hepatocellular

carcinoma (Qureshi et al., 1990). Qazi and Fayyaz described a link between the

long term ingestion of aflatoxin in food with liver cancer in Karachi (Qazi and

Fayyaz, 2006).

2.2. Control of Aflatoxins

Because of the profound toxicity, the AFB level in foods is

required to be regulated. Its level in food and food ingredients may not

exceed the permissible limit, which is 20 ppb for human food with exception of

milk which is 0.5 ppb for AFM (JECFA, 2001). The maximum level of AFB as per

European Union is 2 µg/Kg in food stuff intended for direct human consumption

and is 0.10 µg/Kg in food for infants and special medical purposes. According

to OJEUL (2006) the level of AFM is 0.05 µg/Kg and 0.025 µg/Kg for milk

products for infant and special medical purposes, respectively (Regulation,

18

2006). Several strategies have been established to remove such toxins from

food commodities at different levels. These include:

2.2.1. Agricultural Control

At this level emphasis is laid down on agricultural practices to reduce

contamination/infection by toxigenic fungus. Such methods include: proper

maturing of crops, minimizing post-harvest damage and improving storage

conditions (Hell et al., 2000).

2.2.1.1 Pre-harvest Control

In this method, genetically resistant varieties against Aspergillus infection,

e.g. Mp420 and Mp13E were developed by Brown and his team (Brown et al.,

1999). After identifying the source of resistance to such pathogens, the

incorporated African maize varieties were used in Nigeria (Menkir et al., 2006).

Similar transgenic approach was implied in cotton utilizing the antifungal gene

from maize (Cary et al., 2011). Another approach in transgenic crops is to use

insect resistant variety. As insect infestation also promote the fungal growth by

increasing moisture contents and damage to Kernel. Bt maize variety and later on

new Bt varieties were developed to provide protection against harmful insects.

These interventions significantly reduced the aflatoxins level with resistance

against insects like European corn borer and southwestern corn borer (Wu,

2008).

Utilization of atoxigenic strains is one of the methods to minimize the

damage caused by the aflatoxins. These strains reduce level of the wild strains in

ecosystem by competing with wild toxigenic strains. Probst and co-workers

isolated the 96 atoxigenic strain from four province of Kenya. These strains were

tested by co-inoculation with toxigenic strain to assess the ability to reduce the

aflatoxins level. Among these 12 strains showed comparable result with

19

commercial NRL 21882 USA isolate (Probst et al., 2010). Weaver and

colleagues conducted the field trial in Mississippi for atoxigenic strains. They

suggested the use of atoxigenic strains at early stage markedly improve the

quality and safety (Weaver et al., 2015). Wu and Khlangwist showed cost

effectiveness of biocontrol method over the post-harvest strategies (Wu and

Khlangwiset, 2010). After completion of genome sequencing of Aspergillus

flavus, gene coding for enzymes involved in synthesis and regulation of aflatoxins

would be valuable for devising new strategies in biocontrol (Bhatnagar et al.,

2006).

2.2.1.2. Post-Harvest Control

In this approach, harvested crops are protected by minimizing the risk of

fungal contamination. It is achieved with application of rapid drying to reduce the

moisture content, sorting, smoking and good protection against pests during

storage (Hell et al., 2000; Hell et al., 2010). Certain chemicals like ammonia,

acids, oxidizing and reducing agents, herbicides, pesticides and surfactants were

used to decrease the fungal growth and toxin production by different researchers

as described by Kabak and colleagues (Kabak et al., 2006).

Antagonistic bacteria, yeast and fungi were also used to prevent growth of

Aspergillus, hence the toxin formation. Various species of lactic acid producing

bacteria, Leuconostoc mesenteroids inhibited the growth and the toxins formation

in Aspergillus flavus, were reported by different scientists. Shantha, investigated

the effect of different Rhizopus, Sporotrichum and Phoma species and their cell

free extract on the aflatoxins biosynthesis and degradation. The cell free extracts

exhibited better results than culture filtrate (Shantha, 1999).

2.2.2. Dietary Control

20

Due to economical constrains and lack of awareness, appropriate

measures at agricultural level are not adequate to keep the aflatoxins level within

the permissible limits. Secondly extreme weather conditions especially in tropical

and subtropical region increase the likelihood of fungal growth and thus the

aflatoxins contamination (Phillips et al., 2002b; Williams et al., 2004; Huebner et

al., 2008; Phillips et al., 2008a). Moreover the diverse cooking habits/ways in

these regions cannot assure the required reduction of aflatoxins content

(Bullerman and Bianchini, 2007; Mohamadi Sani et al., 2012). In order to reduce

the food-borne exposure of the aflatoxins, both chemical and physical

(Adsorption) approaches are utilized by researches for detoxification as shown in

Table 2.1. Reducing absorption of the aflatoxins and preventing toxicity in

animals and humans is becoming a vital strategy for effective prevention of

toxicity of the aflatoxins in animal and human as well.

2.2.2.1 Chemopreventive agents

These substances either natural or synthetic modulate or reduce

carcinogenic effect of the aflatoxins. Oltipraz an antischistosomal drug was

reported to reduce the carcinogenicity of AFB. In a clinical trial it showed

detoxification of AFB by increasing conjugation of its reactive metabolite AFB-

8,9-epoxide by glutathione S-transferase. It also inhibited the formation of the

epoxide.

Chlorophyllins and green tea polyphenols have shown reasonable

anticancer properties in different studies. In a study , chlorophyllin alone and in

combination with fermented milk, exhibited hepato-protective effect against AFB

in male Wister rats (Kumar et al., 2012b). Similarly copper complex of

chlorophyllin, a food colorant was reported by various researchers to show

21

reasonable health benefits in term of anti-mutagenic, anti-carcinogenic and anti-

oxidant action. (Tumolo and Lanfer-Marquez, 2012).

Among these various chemopreventive substances, milk thistle (silymarin)

by virtue of its hepato-protectant and antioxidant actions, has shown beneficial

role in detoxification of aflatoxins (Rawal et al., 2010).

Table 2.1 Dietary Control of Aflatoxins

CH

EMO

PREV

ENTI

ON

Origin Agents Mode of action

Natural Milk Thistle: Silymarin Anti-oxidants (Tedesco et al., 2004b)

Chlorophylls Antioxidants (Kumar et al., 2012a)

Synthetic Oltipraz Decreased formation of epoxide Increased Conjugation of epoxide (Sporn and Suh, 2002)

AD

SOR

BTI

ON

Non-Microbial Origin

Bentonite Adsorption (Rao and Chopra, 2001) Zeolite Adsorption (Miazzo et al., 2000) HCAHS Adsorption (Wang et al., 2008) Novasil Adsorption (Phillips et al., 2008b)

Microbial Origin

Esterified Glucomannan (Mycosorb)

Adsorption (Yildirim et al., 2011)

Live Culture: Saccharomyces cerviase

Adsorption (Pizzolitto et al., 2013)

Lactobacilli species Adsorption & metabolism (Fazeli et al., 2009b; Hernandez-Mendoza et al., 2009b)

a. Silymarin (SLM)

Silymarin (SLM) a flavonolignan obtained from milk thistle (Silybum

marianum) from family Compositae is indigenous to Mediterranean region. It is

found all over the Europe, North America, India, China, Africa and Australia

(Luper, 1998).

i. Chemistry

22

SLM is polyphenolic flavonolignan extracted from the milk thistle by 95%

ethanol. SLM is complex consisting of: silybin (most active), silychristin, silydianin

and taxifolin (Saller et al., 2001).

ii. Pharmacokinetics

Being insoluble in water, it is administered in encapsulated form. Oral

absorption is about 50%. Time to reach peak plasma level is 6-8 hours in human

and animals. After conjugation and sulfation in liver, the drug is excreted in bile

(Morazzoni et al., 1993; Kshirsagar et al., 2009).

iii. Mode of action

Various studies indicated the multiple mechanisms responsible for wide

variety of pharmacological action of SLM. These include antioxidant, free radical

scavenging with subsequent inhibition of lipid peroxidation, membrane

permeability, inhibition of cirrhosis as described by Pandey (Kshirsagar et al.,

2009; Pandey, 2014). These actions help in neutralization of oxidative stress

produced by AFB exposure and hence the toxicity.

iv. Role in hepato-toxicity

SLM is an established hepato-protectant used clinically. Various studies

showing this action has been reported by various researchers. The protective

role of SLM on hepatocytes when exposed to carbon tetrachloride was

reported by Muriel and Mourelle. They studied the protective effects of SLM

against the toxicity induced by carbon tetrachloride on the liver plasma

membrane through its antioxidant properties (Muriel and Mourelle, 1990).

Rastogi and co-workers studied the hepato-curative effects of SLM and picroliv

in AFB induced hepatotoxicity in rats. They concluded that hepato-curative

effect of picroliv and silymarin, was comparable (Rastogi et al., 2000). Tedesco

23

and colleagues studied the effects of SLM on the excretion of AFM in cows

upon the consumption of AFB contaminated diets. The animals were given

SLM extract (10 g/day) in first phase and (30 g/day) in second phase. The

treated animals exhibited significant reduction in AFM level (p<0.01) on day 3

in phase I and on day 11 in phase II (p<0.05) (Tedesco et al., 2003). Later on

they reported the beneficial effects of SLM on body weight gain and feed intake

in broiler chicken, intoxicated with AFB (Tedesco et al., 2004b).

In study conducted by Dumari and colleagues, milk thistle (SLM) at dose

1% in feed, exhibited protective effect against elevation of GOT and GPT in

broilers caused by ingestion of 500 ppb of AFB (Dumari et al., 2014). In another

study Muhammad and his team investigated the role of SLM in aflatoxicosis and

compared with commercial toxin binders. AFB (80 ppb) was fed during first week

and 520 ppb onward to broilers. Intake of silymarin alleviated the negative effect

of AFB on weight gain and feed conversion ratio. The levels of GOT and GPT

were significantly lower than intoxicated birds (Muhammad et al., 2012).

v. Role in immuno-toxicity

By virtue of its antioxidant and free radicals scavenging action, silymarin is

of value in counteracting the immune-toxicity of AFB and other mycotoxins.

Silymarin was reported to enhance the antibody titer against NDV and IBD in a

study conducted by Chand and co-workers (Chand et al., 2011). Similarly, in a

study conducted by Khatoon and her colleagues SLM neutralized the immune-

toxicity in layer hens induced by ochratoxin A at level of 1 mg/Kg alone and in

combination with vitamin E (Khatoon et al., 2013).

In the light of these protective effects on liver and immune systems,

SLM can be used as positive control in immune- and hepato-protective studies,

therefore, in current study silymarin would be used to compare the protective

24

effect of probiotics on liver and immune system against toxicity of AFB.

2.2.2.2. Adsorbents

Latest approach for reducing the exposure to the aflatoxins in animals and

humans is the usage of adsorbents in feed and food commodities. These

substances adsorb the aflatoxins either in ready to use feeds and foods or in

gastrointestinal tract (GIT) of ingesting organism. It results in a decrease in

bioavailability of the toxins as shown in Fig. 2.4.

Rao and Chopra studied the effect of sodium bentonite and charcoal for

reduction of AFM in goats fed with combination of 100µg of AFB alone and in

combination with sodium bentonite and charcoal. They reported that AFB

contaminated feed (100 ppb) alone showed no signs of AFB toxicity except

excretion of AFM in milk. However the treated goats had shown significantly

lower AFM level (Rao and Chopra, 2001).

Fig.2.4 Mechanism of action of Adsorbents

In another study, Zeolite (microporous aluminosilicate mineral) neutralized

some deleterious effects of AFB in broiler given combination of 2.5 mg/Kg of

Aflatoxin

Adsorption

Adsorben

Adsorbed Aflatoxin B1

(Reduction in Bioavailability)

25

AFB contaminated feed and 1% Zeolite from 21 to 42 days of age (Miazzo et al.,

2000). As described by Philips and co-workers adsorbent clays: Hydrated

sodium calcium aluminosilicates (HSCAS) were reported to decrease the level of

AFB and other mycotoxins (Phillips et al., 2002a; Phillips et al., 2008b). Neeff

and co-workers investigated the role of HSCAS in decreasing the toxicity of AFB

in broilers. They fed AFB (2.5 mg/Kg) of feed supplemented with 0.5 % of

HSCAS for 21 days. HSCAS prevented accumulation of AFB in liver of the

broilers. But it did not show any ample protective effects on liver toxicity (Neeff et

al., 2013).

Similarly Chen and colleague reported protective effect of HSCAS in

broiler fed with 0.5 to 2 mg of AFB/Kg of feed. Ingestion of HSCAS prevented the

decrease in cumulative weight gain in the birds. Liver weight of HSCAS treated

birds remained significantly lower along with better impact on clinical

biochemistry. HSCAS also enhanced expression of catalase and SOD (Chen et

al., 2014c).

However, sometime poor quality adsorbents also adsorb essential

nutrients like vitamins resulting in stunted growth of animals, and

also show irreproducible results in experiments (Chung et al., 1990). To

overcome it, uniform particle size novasil (microcrystalline cellulose) was used by

various researchers to avoid the nutritional consequences while protecting from

aflatoxicosis in animals as well as humans (Marroquín-Cardona et al., 2011;

Mitchell et al., 2014). On account of these limitations, adsorbents of microbial

origin became the focus of research for detoxification of aflatoxins (Jard et al.,

2011).

2.2.2.2.1 Mycosorb (MYC): Esterified Glucomannan (EG)

26

The yeast fermentation is a well-accepted method in region of high

exposure limit (Africa and Asia) for food processing and preservation.

Saccharomyces cerevisiae (SC), an important edible yeast is used in various

fermentation processes. SC has reasonable nutritive value having 40-45% of

protein and vitamin B Complex (Çelýk et al., 2003; Jespersen, 2003). Shetty

and co-workers demonstrated AFB adsorbing properties of SC cells. They also

studied the impact of different strains on binding capacities. It was also reported

by them that SC cells could bind AFB at concentration of 20 µg/ml (Shetty et

al., 2007).

Later on Armando and co-workers isolated SC stains from pigs feed, gut

and feces. They studied binding ability of strains at 50, 100 and 500 ng/ml

concentration and the survival of these SC strains in gastrointestinal

environment. They suggested the SC strain RC-016 and -008 were the potential

candidate to be used as feed additive (Armando et al., 2011). Similarly, SC

strain CECT 1891 when added to drinking water, significantly ameliorated the

deleterious effects of AFB on growth performance parameters, histopathology,

relative weight of liver and clinical chemistry (Pizzolitto et al., 2013). Recently

SC derived product (Dried Yeast) from sugar cane fermentation was reported to

be of important value as aflatoxin binder (Gonçalves et al., 2015).

Bejaoui and colleagues showed that heat or acid treated SC has high

binding capacity than the live culture. Moreover, no degradation products were

isolated revealing the adsorption as main mechanism for detoxification rather

the metabolism. Phosphorylated mannan oligosaccharides (Glucomannan); a

cell wall component of the yeast, was thought to be responsible for efficient

binding of mycotoxins (Raju and Devegowda, 2000b). Different researchers had

reported the binding capacity of esterified glucomannan (EG) with AFB,

27

ochratoxin and T-2 toxin (Hathout and Aly, 2014). Esterified Glucomannan

showed protective effects against toxicity caused by ingestion of AFB.

i. Alleviation of hepatotoxic effects

Scientists utilized the toxin binding ability of EG to prevent the hepatic

toxicity of AFB. Raju and Devegowda, investigated the effect of EG (1 g/Kg) in

broilers given diet containing AFB (300 ppb), ochratoxin (2 mg/Kg) and T-2 toxin

(3 mg/Kg). They reported that EG abolished the deteriorating effects of the

toxins. EG addition caused increase in body weight (2.26%) and feed intake

(1.6%), reduced weights of liver (32.5%) and activity of serum GGT (8.7%). It

also improved serum proteins (14.7%), cholesterol (21.9%), Blood urea nitrogen

(BUN) (20.8%) and blood hemoglobin level (3.1%) (Raju and Devegowda,

2000b). Arvind with his team studied the role of EG in broilers fed with

contaminated diet containing AFB (168 ppb), ochratoxin (8.4 ppb), zearalenone

(54 ppb) and T-2 toxin (32 ppb). Addition of EG ameliorated the weight loss and

increase in relative weight of liver and gizzard caused by consumption of the

contaminated diet (Aravind et al., 2003).

Cao and Wang studied effect of EG and its combination with astaxanthin

in broilers fed AFB for about three weeks. The toxin feeding in the broilers

caused the increase in liver weight, leukocyte counts, Hemoglobin and BUN.

Liver SOD activity was also elevated. Both EG and astaxanthin in combination

and alone alleviated the negative effects of AFB on parameter studied (Cao and

Wang, 2014). Yilidrim and colleagues also studied the role of EG in broilers

given 0.75 g/Kg of EG, 2mg/Kg of AFB for 21 days. EG prevented the negative

effect of AFB on body weight gain and feed intake. EG administration

significantly alleviated toxic effects on blood urea, creatinine, plasma GOT while

histopathological lesions remained unaffected (Yildirim et al., 2011).

28

ii. Alleviation of immunosuppressive effects

Immunosuppression caused by AFB was also prevented by EG in

various studies. Hasan and co-workers studied the role of EG, sodium

bentonite, and humic acid on Newcastle Disease Virus (NDV) antibody titer in

broilers. Diet containing 254 ppb of AFB was given to the birds from 28 to 35

days of age in different treatment groups: 0.2, 0.4, 0.6, 0.8 and 1% of humic

acid, 0.5 % of sodium bentonite and 0.1% EG. Results of their study indicated

strong immunosuppression in intoxicated birds. All feed additives showed

protection against AFB induced immunosuppression. But effect of humic acid

was better among all three treatments (Hasan et al., 2010). Similarly, Ghahri

with his colleagues studied role of EG, humate and sodium bentonite on

immunization against infectious bursal disease (IBD) and infectious bronchitis

(IB) in broiler birds. Humate (1%), sodium bentonite (0.5%) and EG (0.1%) were

given to birds with AFB (200 ppb) contaminated diet. Data of the study indicated

the protective effect by all treatments against AFB on IBD and IB antibody titers

(Ghahri et al., 2009).

Similarly, a study conducted by Mogadam and Azizpour, showed

significant EG protective effect when given to birds taking diet (250 ppb of AFB).

They gave EG, sodium bentonite alone and in combination to the intoxicated

birds. EG supplementation (0.1%) showed better result on humoral antibody

response against NDV (Mogadam and Azizpour, 2013).

Along with toxins binder properties, EG showed protective effect in pigs

when 82 ppb of T-2 were fed to pig for 18 days. After 18 days, pigs were

inoculated by Salmonella typhmuirium. Findings of studies indicated that EG

prevents toxicity of the toxin in animals but it also showed binding of the

infecting bacteria (Verbrugghe et al., 2012).

29

2.2.2.2.2. Probiotics (PBT): Lactobacilli

The concept of PBT was originated from the study of Nobel laureate Eli

Metchnikoff, who linked the longer longevity of Bulgarian peasants with

consumption of fermented milk products (Gupta et al., 2000). The term

“probiotics” was first used by Lilly and Stillwell in 1965 (Lilly and Stillwell,

1965). According to the Food and Agriculture Organization (FAO) and the

World Health Organization (WHO) probiotics are live micro-organisms

which, when administered in adequate amounts confer health benefits on the

host (Quigley, 2010).

Various bacterial species from Lactobacillus, Bifidobacterium,

Enterococcus, Streptococcus genera and some fungal species belonging to

genera Saccharomyces have been utilized as PBT by various researchers (Jin

et al., 2000; Alvarez-Olmos and Oberhelman, 2001). PBT have shown

beneficial role in different pathological conditions such as necrotizing

enterocolitis (Hoyos, 1999) and antibiotic associated diarrhea (D'Souza, 2002).

PBT also exhibited improvement in Helicobacter pylori infection and

inflammatory bowel syndrome (Gupta et al., 2000; Lesbros-Pantoflickova et al.,

2007). Apart from these beneficial effects, PBT were reported to decrease level

of free mycotoxins, cyanotoxins and heavy metals in aqueous solutions

(Salminen et al., 2010a; Zoghi et al., 2014).

Similarly it is also reported by various scientists that different

species of Lactobacilli, Bifidobacteria and Saccharomyces upon incubation

with culture media containing AFB, reduced the level of free AFB, either by

adsorpt ion or metabolizing the toxin (Peltonen et al., 2001b; Fazeli et al.,

2009b; Hernandez-Mendoza et al., 2009b; Hernandez-Mendoza et al., 2011).

30

Lactobacillus is one of the important genus of lactic acid producing

bacteria. It contains gram positive rod shaped facultative anaerobic or

microaerophilic bacteria. These bacteria ferment sugar mainly lactose and

produce lactic acid hence called as Lactobacilli (Makarova et al., 2006).

Different species of Lactobacillus have been reported to have beneficial role in

certain clinical disorder. Different Lactobacilli isolated from human gut include:

Lactobacillus rhamnosus GG, L. casei, L. johnsonii, L. acidophilus and L.

reuteri. Various researchers studied the beneficial role of Lactic acid bacteria in

different clinical conditions like gastrointestinal disorders, infections, autoimmune

disorders, malignancy and etc. These bacteria along with Bacteriodes,

Bifidobacteria, and Clostridium are one of the important component of human

microbiota (Neish, 2009).

i. Gastrointestinal Disorders

Evidence for positive health benefits of Lactobacilli is associated to few

strains which had commercial applications. Lactobacillus rhamnosus G G , a

variant of Lactobacillus casei, was studied extensively in adults and children.

When consumed as a dairy product or as a lyophilized powder, Lactobacillus

rhamnosus GG colonized in the gastrointestinal tract for 1-3 days in most of

the individuals and up to 7 days in about 30% of subjects. Lactobacillus

rhamnosus GG alleviated traveler’s diarrhea, antibiotic-associated diarrhea and

relapsing Clostridium difficile colitis. Similarly, in infantile diarrhea: the

severity and duration of the attack were reported to be reduced. It also

facilitates antigen transport to underlying lymphoid cells, which resulted in

enhanced antigen uptake in Peyer's patches (Gupta et al., 2000).

In double blind randomized clinical trial in Taiwan, Lactobacilli intake

significantly reduced the incidence of necrotizing entercolitis in low weight

31

preterm babies (Lin et al., 2008). In another study, cells free supernatant

obtained from L. acidophilus and L. casei were reported to prevent colon

cancer cell invasion in cultured metastatic colorectal carcinoma cells. Analysis

of supernatant suggested that key inhibitory molecule might be protein or

polysaccharides (Escamilla et al., 2012).

ii. Effect on Immune system

Lactobacilli having direct contact with intestinal epithelial lining play vital

role in modulating the immune responses. The effects on immune responses

depend upon the method of administration, strain and dose of probiotics. Karimi

and colleagues studied the effect of route of administration on probiotic

protective action. They administered probiotic to two groups of broilers: one

through drinking water (0.5 g/L) and other via feed (1 g/Kg). Growth

parameters, relative organ weight and T-cells function were evaluated. Results

indicated a strong impact of route of administration on parameter studied.

Administration through water appeared to be superior to in-feed

supplementation (Karimi Torshizi et al., 2010)

Rizzello and co-workers has reviewed the immunomodulatory role of

Lactobacilli. They described stimulatory role of Lactobacilli on natural killer

cells. These cells regulate population of dendritic cells pivotally involved in

generation of adaptive immune responses (Rizzello et al., 2011). Brisbin and

his colleagues administered three Lactobacilli specie: L. acidophilus, L. reuteri

and L. salivarius to birds on day 1-, 14- and 21 of age. Immunization was done

by sheep red blood cells, NDV and IBD vaccines. Findings of experiment

revealed that L. acidophilus enhanced humoral immune response. Cell

mediated immunity as assessed by INF-Ɣ determination was not affected

(Brisbin et al., 2011). Poorbaghi and co-workers investigated the role of

32

encapsulated form of L. acidophilus and inulin (prebiotic) alone or in

combination in broilers on viral shedding and antibody titer. Encapsulated form

of L. acidophilus showed remarkable effect on viral shedding (Poorbaghi et al.,

2014).

iii. Detoxification of Mycotoxins

Apart from these potential health benefits, Lactobacilli species bind

selectively to different mycotoxins and heavy metals in solution. It results in

decreased absorption and subsequent reduction in toxicity of these chemicals.

Salminen and his colleagues studied the beneficial impact of probiotics on

detoxification and human health. Lactic acid bacteria and Bifidobacteria have

been reported to remove heavy metals, cyanotoxins and mycotoxins from

aqueous solutions in vitro. The binding processes appear to be specie and

strain specific (Salminen et al., 2010a). The mechanism of action of Lactobacilli is

shown in Fig. 2.5.

Fig. 2.5 Mechanism of Lactobacilli in Aflatoxicosis

iv. Mechanism of Binding

The negatively charged cell surface of Lactobacilli plays vital role in

Probiotics (Lactobacilli)

Reduced Bioavailability of AFB

Adsorption Degradation AFB

33

binding of the mycotoxins. It comprises of peptidoglycans, teichoic acid, S protein

layer and polysaccharides: both neutral cell wall polysaccharides and

exopolysaccharides. The binding of AFB with bacterial cell wall largely depends

upon the teichoic acid and exopolysaccharides (Hernandez-Mendoza et al.,

2009b).

Bovo and co-workers studied the capacity of non-viable dried L.

rhamnosus in decreasing the AFB from contaminated medium. Bacteria were

cultured on de Man Rogosa and Sharpe (MRS) agar and then dried by either

spray drying or freeze drying after autoclaving. Binding ability was assessed

using 109 cells suspension either sprays dried or lyophilized. The cells

suspension was incubated with AFB solution (1µg/ml) for 60 minutes. Results

showed a lack of binding capacity in spray dried suspension due to loss of cell

wall architecture whereas, lyophilized cell retained their binding characteristics

(Bovo et al., 2014).

v. In-vitro Binding of Toxins

Lactobacilli exhibited strong binding affinity with various mycotoxins in

various studies. The binding of AFB by strains of Lactobacilli and

Bifidobacetria when incubated together was reported by Peltonen and his team.

The study involved the determination of binding of AFB in solution by 20 strains

of Lactobacilli and Bifidobacetria. Two Lactobacillus amylovorus strains and

one Lactobacillus rhamnosus strain removed more than 50% AFB and were

selected for further study. Bacterial binding of AFB by these strains was

rapid, and more than 50% AFB was bound throughout an incubation period of

72-hours. Binding was reversible, and AFB w a s released by repeated

aqueous washes. These findings further supported the ability of specific

strains of lactic acid bacteria to bind selected dietary contaminants (Peltonen

34

et al., 2001b).

Fazeli and colleagues studied the AFB reduction by autochthonous

strains of lactic acid bacteria (Lactobacillus casei, Lactobacillus plantarum and

Lactobacillus fermentum) which were isolated from traditional Iranian

sourdough and dairy products. All the strains were reported to be capable of

removing AFB, and the reduction of the AFB ranged from 25 to 61%

throughout the incubation period. Removal of AFB was rapid process, with

approximately 61% and 56% of the toxin taken instantly by Lactobacillus

fermentum and Lactobacillus plantarum, respectively. These findings

suggested the use of certain novel probiotic bacteria with high aflatoxin

binding capacity for detoxification of foods (Fazeli et al., 2009b).

The ability of different strains of Lactobacillus casei to reduce free AFB

by binding from aqueous solution was studied by Hernandez and colleagues.

The strains exhibited different degrees of AFB binding; the strain with the

highest AFB binding was Lactobacillus casei L30, which bound 49.2% of the

available AFB (4.6 µg/ml). In general, the human isolates bound the most AFB

while cheese isolates the least. Stability of the bacterial-AFB complex was

also assessed by repeated washings. Exposure of the bacterial cells to bile

significantly increased AFB binding and reduced the differences between the

strains (Hernandez-Mendoza et al., 2009a). In another study Hernandez-

Mendoza and co-workers utilized the flow cytometry to study the binding of

AFB with Lactobacilli reuteri. Results of the study indicated that binding of AFB

causes changes in surface of the bacterial cell wall. They suggested the

technique might be of value for detection of human exposure to AFB

(Hernandez-Mendoza et al., 2011).

vi. In-vivo Binding of Toxins

35

Gratz with colleague investigated the role L. rhamnosus GG in Caco-2 cell

line adopted to express CYP 3A4. The cells were grown as mono layer on

transmembrane filter for 21 days. AFB caused significant reduction in trans-

epithelial resistance at 24, 48 and 72 hour of incubation. Co-incubation of the

bacteria at (1×1010 and 5×1010 CFU/ml) significantly restored the trans-epithelial

resistance. Moreover, DNA fragmentation caused by AFB was absent in

Lactobacilli treated cells (Gratz et al., 2007). Similarly in another study

detoxification role of L. acidophilus CRL1014 and L. reuteri CRL 1098 was

determined in human peripheral blood mononuclear cells. When challenged with

ochratoxin, a fall in level of tumor necrotic factor (TNF-α) and IL-10 was

observed. Addition of the bacteria prevented apoptosis in cells but had no effect

on decreased IL-10 production (Mechoud et al., 2012).

vii. Use in Aflatoxicosis

Hathout and colleagues studied the role of L. casei and L. reuteri in AFB

induced oxidative stress in rats. They gave dose of 10 ml/kg body weight having

1×1011 cells of both bacteria to rats which were intoxicated with 3mg/Kg of AFB.

Findings of the study revealed the protective role of both bacteria against AFB

induced biochemical and histological alterations. L. reuteri showed better

performance than L. casei (Hathout et al., 2011). Kumar and his team studied the

role of fermented milk (containing both L. rhamnosus and L. casei) and

chlorophyllin in rats given 450 µg/kg of AFB intraperitoneally per animal twice a

week for 6 weeks. Both chlorophyllin and fermented milk alone or in combination

alleviated the negative effect of intraperitoneal AFB as evidenced by increased

level of glutathione peroxidase, SOD, catalase and glutathione-S-transferase

(Kumar et al., 2012a).

36

Similarly in a study conducted by Hernandez and co-workers L. casei

Shirota reduced the bioavailability of AFB given for three weeks. The AFB-

albumin adduct (biological marker to assess the exposure) was significantly lower

in animal receiving PBT bacteria. Fluorescent monoclonal antibody staining

technique clearly revealed the binding of AFB to cell surface (Hernandez-

Mendoza et al., 2010). Rawal with his colleagues studied the protective action

Lactobacilli in turkeys fed with 1ppm of AFB. The decline in body weight and

increased relative liver weight were not observed in PBT treated birds. However,

hepato-toxicity markers and MHC genes remained unaffected. Researchers

supported the use of probiotic keeping in mind the intensity of exposure and

extreme sensitivity of turkeys to AFB (Rawal et al., 2014).

viii. Safety of Lactobacilli

Lactobacilli usage appears to be safe even in human population. Quigley

described a few incidences of bacteremia and sepsis with use of the lactobacilli in

special group of patient: particularly in children, immuno-compromised, elderly

and critically-ill patients (Quigley, 2010). A case of bacteremia was reported in

child given L. rhamnosus GG strain having short gut syndrome. Strain typing and

gel electrophoresis investigation confirmed the identity between bacterial-tablet

and blood stream isolates (De Groote et al., 2005). Similarly case of sepsis was

reported in patient given L. rhamnosus before Aortic valve replacement (Kochan

et al., 2011). In double blind randomized clinical trial in 144 patients,

multispecies probiotic treatment resulted in intestinal ischemia in 9 patients with

high mortality. However, no direct link was established with probiotics (Besselink

et al., 2008).

ix. Food containing probiotics in Pakistan

37

The Dhai /Yogurt are traditional dairy food product of Pakistan and

subcontinent which is prepared by fermentation like kefir and yogurt in Europe.

The traditional fermented milk product is prepared by mixing raw and heat treated

milk with starter culture (Jhaag: local term) containing Lactobacillales as

dominating microbial group while it also have other undefined complex microbial

consortia for fermentation. The dhai serves as very good and economical source

of probiotic Lactobacillales. Aslam and Qazi (2010) isolated L. Bulgaricus, L.

casei, L. Acidophilus and L. salivarius from local yogurt (Dhai) sample (Aslam

and Qazi, 2010). In another study milk samples were collected from cow, buffalo

and sheep to isolate lactic acid bacteria. Among all isolates, overall occurrence of

lactic acid bacteria was 66% with highest frequency in cow milk. Milk from buffalo

and cow contained reasonable amount L. bulgaricus and L. acidophilus along

with other species of Lactococus and streptococcus (Aziz et al., 2009).

2.3. Justification

Aflatoxins are unavoidable contaminants of food commodities. Long

term low level exposure causes mutagenic, carcinogenic and

immunosuppressive effects. The aflatoxins producing fungi grow when humidity

is above 12% in storage areas. These toxins produced are not destroyed by

autoclaving. Due to toxic effects, the level of aflatoxins in food and food

ingredients is regulated by international regulatory bodies through

implementation of standard practices and procedures. It resulted in reduction

of exposure t o such toxins in developed countries.

In Pakistan, grains/foods are more susceptible to aflatoxins contamination

due humidity and high temperature (Mobeen, 2011). Moreover, lack of modern

harvesting and storage technology coupled with extreme weather conditions

38

result in increased likelihood of the fungal growth with subsequent AFB

contamination All of the four aflatoxins have identified at level higher than

permissible limit in both various unprocessed and processed food (Mushtaq et

al., 2012).

In Pakistan the control of aflatoxins production in feed and food

ingredients is not cost effective. It results in increased level of the toxins in

raw material used for poultry feed and animal ration. The consumption of

contaminated meat / milk of such animals results in human exposure to these

toxins. The addition of sequestering agents in animal ration and poultry

feed is common practice to reduce the level of the aflatoxins in feed and food

commodities. However, there are two main problems concerning the use of

sequestering agents; first interaction of sequestering agent with vital micro-

nutrients that result in the stunted growth in livestock and poultry and second

the use of these sequestering agents (adsorbent) is not advisable in human

due to poor quality and other deleterious physiological effects. In this study we

will evaluate the utilization of lactobacilli bacteria from fermented milk as PBT to

reduce the bioavailability of AFB in vivo. Since, it is hypothesized that usage of

PBT may reduce the bioavailability of aflatoxins from intestine resulting in

decreased hepato-toxicity, renal toxicity and immunosuppression. Moreover,

l ack of interaction with micronutrients like vitamins, low cost, ease of availability

and control of Lactobacilli culture may suggest the PBT as an economical and

practical approach to reduce toxicity of such toxins in under developed (African

countries) and developing countries, like Pakistan.

39

2.4 Aims and Objectives

Pakistani traditional food Dhai has been reported to contain several

species of Lactobacilli by various researchers which fulfill the prerequisite of PBT.

Current study was designed to evaluate ability of the Lactobacilli (PBT) from

Pakistani traditional food (Dhai) to prevent toxicity of AFB on liver and immune

system. Moreover, it provided comparison of the PBT with other established

methods for detoxification of the AFB i.e. hepato-protectant (Silymarin) and

adsorption (Mycosorb). Following set of experiments were performed to achieve

these objectives:

2.4.1 Production and Measurement of Aflatoxin

The toxigenic fungus was grown on fermented rice from the spore

obtained from culture center. After a period of 28 day, AFB was extracted from

the culture and quantified by HPLC method.

2.4.2 Administration of Treatments to Birds

A day old broiler chickens (n=240) were segregated into group I to VII and kept

for period of 49 day. The different treatments were administered from 3rd to 5th

week of age.

2.4.3 Sampling

Blood (2ml) was collected from wing vein of each bird of each group

weekly from 4th to 7th week of age. Six birds from each group were killed weekly

from 4th to 7th week of age to collect bursa of Fabricius, liver, spleen and kidney

for histopathological examination. Liver samples collected from the killed birds

were stored at -80 ºC.

2.4.4 Validation of analytical methods and analysis of Samples

1. Serum thus obtained was analyzed for:

a. NDV Antibody titer

40

b. Serum Biochemistry for assessing of hepatic and renal functions.

2. Liver, spleen, bursa of Fabricius and kidney was processed for

histopathological examinations.

3. Liver was also analyzed for AFB level by HPLC method..

41

3. Materials and methods

3.1. Plan of study

A day old broiler chickens (n=240) were purchased from well reputed local

hatchery. The birds were kept in semi-closed environmental shed under standard

environmental condition of temperature and relative humidity. The feed and water

were provided ad libitum. At age of three weeks, these birds are segregated into

following treatment groups:

1. Group I : (Basal Feed without aflatoxin)

2. Group II : (PBT [109CFU] + Basal Feed without aflatoxin)

3. Group III : (400 ppb of AFB)

4. Group IV : (PBT [2×109 CFU] + 400 ppb of AFB )

5. Group V : (PBT [109 CFU] + 400 ppb of AFB )

6. Group VI : (SLM [600mg/Kg body weight] + 400 ppb of AFB)

7. Group VII : (MYC [1g/Kg of Feed] + 400 ppb of AFB)

The birds were fed above mentioned treatments for two week i.e. on 4th to 5th

week of age. The birds of each group were primed with Newcastle disease virus

(NDV) (LaSota strain) vaccine (Solvay®) and boosted with the same vaccine at

day one and 30 of age respectively. Moreover, each bird was vaccinated against

prevailing disorders such as infectious bronchitis (IB), infectious bursal disease

(IBD), hydropericardium syndrome (HCS), and etc.

3.2. Preparation of feed containing different treatments

Feed containing AFB, PBT, MYC or SLM was prepared as followings:

3.2.1. Production of AFB

42

The production of AFB was optimized after using different media ranging

from SD broth (liquid) to solids including maize, broiler feed and rice with either

continuous shaking or without shaking. AFB for current study was produced

through rice fermentation with continuous shaking by using the method of

Shotwell et al., and Yunus and Böhm, with slight modifications (Shotwell et al.,

1966; Yunus and Böhm, 2011). The detail of method was as follow:

a. Fermentation of rice

Aspergillus flavus culture was obtained from Department of Microbiology,

University of Veterinary & Animal Sciences, Lahore Pakistan, and was used to

inoculate the Sabouraud dextrose agar (SDA) from Sigma-Aldrich, Germany, in

petri plates. The plates were incubated at 28 ± 2°C in humidified incubator for 5 to

7 days. Basmati rice (32gm) purchased from local market were soaked in 16 ml

of distilled water for 2 hours in 250 ml conical flasks. The flasks containing the

rice were autoclaved at 121°C for 15 minutes at 15 psi. Sterile glass rod was used

to separate the rice kernels in case of clumping. The spore suspension was

produced by using 6 ml of 0.1 % aqueous solution of tween 80. The flasks were

inoculated aseptically by one ml of the spore suspension. After thorough shaking

the flasks were placed in horizontal flask shaker (PAMCO, Faisalabad) in dark at

290 rpm for five days. The temperature was maintained at 26 ± 2°C. Color

change from white to brownish indicated the fungal growth. The flasks containing

infected rice were removed from shaker and autoclaved at 121°C for 15 minutes

at 15 psi. The autoclaved rice were removed from the flask and dried at 80°C.

The dried rice were powdered in blender for uniformity of the aflatoxins contents.

Five days cycle of rice fermentation was repeated 92 times (470 days) to produce

required amount of AFB for the study.

b. Determination of AFB in fermented rice

43

AFB was determined by slight modification of method of Iqbal et al.,

(2010).The powdered contaminated rice were extracted with 100 ml mixture of

acetonitrile (Sigma, Germany) and distilled water (86:14) by shaking for 35

minutes at 50 rpm in 250 ml conical flasks. Then solution was filtered through

Whatman filter paper No.5. Acetic acid from Sigma, Germany (70 µl) was added

to 9 ml aliquot of filtrate. The filtrate was passed through immunoaffinity cleanup

column (MycoSep® [COCMY2226: Romer Labs] at rate of 2 ml/minute for

purification. AFB retained in column was eluted by 2 ml of methanol (Merck,

Darmstadt, Germany).The elute was evaporated to dryness under the stream of

nitrogen gas.

To the eluted AFB and AFB standard solutions (Romer Labs, Rawalpindi-

Pakistan), 100 µl of trifluroacetic acid was added and kept at room temperature

for 20 minutes in dark for pre-column derivatization. 400 µl of acetonitrile/water

(1:9) mixture was added to the tubes. Mobile phase consisting of the acetonitrile,

methanol and double distilled water in (20:20:60) was used after degassing by

sonication. The 20 µl of sample was injected into HPLC with following

specification:

Pump: Quaternary pump

Column: C-18 Merck 4.6×250 mm×5 micron diameter

Colum oven: 40° C

Detector: Florescence limits are excitation 360, emission 440

Flow rate: 1ml / min

Software: Agilent software

c. Mixing of AFB with feed

44

The appropriate amount of the infected powdered rice was geometrically

mixed with the feed to achieve the level of 400 ppb of AFB in feed.

3.2.2. Preparation of probiotic (PBT)

Traditionally fermented Dahi was obtained from a cottage scale producer.

Selected Dahi sample was microbiologically characterized by using

metagenomics based analysis of bacterial diversity (Farah, 2016). In brief,

Lactobacillus sp. was about 85.3 % followed by Streptococcus sp. of about

10.74%, while all other species were less than one percent. The dominating

microbial species were identified based on 16srRNA sequencing and data was

submitted in NCBI gene bank for obtaining accession number. Selective strains

were identified as Streptococcus thermophilus QAUSt01 (Accession number:

KT021870) and Lactobacillus sp. QAULb01 (Accession number: KT021869). This

research work was done with our collaborative group at Department of

Microbiology, Quaid-i-Azam University in parallel to our work.

Before application to broiler chicken Lactobacillus CFU was measured by

using deMan Rogosa Sharpe agar (MRSA) media (Sigma-Aldrich, Germany). An

amount of the fermented milk (300g) equivalent to 109 colony forming units (CFU)

of Lactobacilli was defined to be used as 1X and subsequently, fermented milk

(600g) equivalent to 2 x 109 CFU was defined as 2X (Peltonen et al., 2001b;

Gratz et al., 2006; Gratz et al., 2007). The required amount of fresh fermented

milk was administered to birds in drinking water.

3.2.3. Preparation of SLM containing feed

SLM was administered at dose 600 mg /Kg body weight of bird (Tedesco

et al., 2004b). For 30 birds in the group 18g/day (90 tablets) of silymarin was

needed. Siliver tablets (200 mg of silymarin) of Abbot® (Karachi, Pakistan) were

purchased from local Pharmacy. For two weeks, 1,260 tablets were powdered

45

after measuring the average weight. The appropriate amount of powder was

mixed uniformly with feed to produce the required dose.

3.2.4. Preparation of MYC containing feed

MYC from Alltech®(Nicholasville, Kentucky US) was purchased from local

market and mixed thoroughly with feed to produce level of 1g/Kg of feed (Girish

and Devegowda, 2006).

3.3. Sampling

3.3.1. Collection of blood and serum

Two milliliters of blood was collected by 3 ml BD® syringe from wing vein of

six birds from each group on 4th, 5th, 6th and 7th weeks of age. One ml of the

blood from the syringe was transferred to heparinized vacutainer for hematology

while the rest of blood was transferred to the vacutainer containing clot activator for

separation of serum. The later was placed at ambient (25±3°C) temperature

overnight. The serum thus separated was stored in 2ml eppendorf (Eppendorf®) at

temperature of -80°C for determination of Newcastle disease virus (NDV) antibody

titer and serum biochemistry.

3.3.2. Collection of internal body organs

Six birds from each group were weighed and then slaughtered on 4th,

5th, 6th and 7th weeks of age for collection of liver, spleen, bursa of Fabricius and

kidney. All organs except liver were stored in 10% formalin solution for

histopathological examination. However among six livers thus obtained, three

were kept in 10% formalin solution for histopathological study while three were

stored at -80°C for AFB residue determination.

3.4. Analysis of samples

3.4.1. Determination of NDV antibody titer

46

Serum NDV antibody titer was measured by hemagglutination inhibition

(HI) test (Allan and Gough, 1974). Table of Brugh (Table-3.3) was used to

calculate Geometric mean titer (GMT) of the antibodies (Brugh, 1978). The

method included:

3.4.1.1. Preparation of RBCs

Blood (5ml) was collected from wing vein of unvaccinated broiler chicken

in 10 ml syringe BD® (Karachi, Pakistan) containing 4% sodium citrate (Sigma)

solution. The citrated blood was centrifuged at 1500 rpm for 5 minutes. The

plasma layer and buffy coat was removed by pasture pipette and replaced by

phosphate buffer saline (PBS) from Sigma, Germany. After thorough mixing, the

RBC suspension was re-centrifuged at 1500 rpm for 5 minutes. The supernatant

was discarded and the sediment was re-suspended in the PBS. The process was

repeated thrice. RBC suspension (5%) was prepared by adding washed RBCs to

the PBS.

3.4.1.2. Preparation of 4HA (hemagglutination) unit of NDV

PBS (50 µl) was added in each of 12 wells in row of 96 wells plate having

round bottom. In 1st well, 50 µl of virus suspension was pipetted and mixed

thoroughly. The diluted viral suspension (50 µl) was transferred from 1st well to

10th well and mixed thoroughly. Well no. 11th and 12th were kept free from virus.

RBCs suspension (1%) prepared from above mentioned RBC suspension (5%)

was added from 1st to 11th wells as shown in Table.3.1. The plate is incubated for

30 minutes at 25 ± 2°C.

The inhibition of agglutination was confirmed by formation of clear button

at bottom, while presence of uniform thin layer indicated agglutination of RBCs by

virus. The highest dilution of virus showing hemagglutination was regarded as

one HA unit. The highest well number in which agglutination observed was 8th.

47

For preparation 4HA units following calculation were used:

Highest Well number with Agglutination……………..8th

Dilution in well # 8…..…………………………………..28=256

For 4 HA titer ……. ……………………………………..256/4 =64

1ml of the viral suspension was diluted with 63 ml of the PBS. The viral

suspension thus obtained had 4HA titer.

3.4.1.3. Determination of NDV antibody titer

PBS (50 µl) was added to each 12 wells in row of 96 wells plate having

round bottom. The serum (50 µl) was pipetted in 1st well of the row and mixed

thoroughly. Then 50 µl of the mixed serum was transferred from 1st well to 2nd to

produce two folds dilution. The diluted serum was further diluted by same

process till 10th well of row. After which the 50 µl serum was discarded. No serum

was added in last two wells (11th and 12th) of the row.

4HA unit of Viral suspension (50 µl) was added and thoroughly mixed in each

well of row except 12th well (positive control). The plate was incubated for 30

minutes at 25°C (for antigen and antibody interaction). After incubation, 50 µl of

the 1% RBC suspension was added in each well of row. The 12th well had only

RBC with no serum and viral suspension (negative control) as shown in Table-

3.2. The plate was again incubated for 30 minute at 25 ± 2°C.

After incubation, positive result was formation of clear button at

bottom due to inhibition of hemagglutination in 12th well and negative result was

formation of uniformed layer (complete agglutination) in 11th well. The results thus

recorded were processed as follow: The highest well number having

hemagglutination inhibition (button formation by RBCs) was recorded for each

serum sample processed.

48

Table- 3.1

GENERAL PLAN OF HAEMAGGUTINATION (HA) TEST

Hemagglutination well number Control

Well 1 2 3 4 5 6 7 8 9 10

Normal

Saline

50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl

Virus 50 µl - - - - - - - - - -

Qty. of mix

transferred

- 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl -

Qty.

discarded

- - - - - - - - - 50 µl -

Qty. of 1%

RBC

50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl 50 µl

Final virus

dilution

1:2 1:4 1:8 1:16 1:32 1:64 1:128 1:256 1:512 1:1024 -

49

Table-3.2

GENERAL PLAN OF HEMAGGLUTINATION INHIBITION (HI) TEST

Hemagglutination well number Control Well

1 2 3 4 5 6 7 8 9 10 +ve -ve

Normal Saline

50 µl

50 µl

50 µl

50 µl

50 µl

50 µl 50µl 50µl 50µl 50µl 50µl 50µl

Serum 50 µl - - - - - - - - - -

Qty. of mix transferred - 50µl 50µl 50µl 50µl 50

µl 50µl 50µl 50µl 50µl - -

Qty. discarded - - - - - - - - - 50µl -

Qty. of 4 HA virus

50 µl 50µl 50µl 50µl 50µl 50µl 50µl 50µl 50µl 50µl 50µl

Qty. of 1% RBC

50 µl 50µl 50µl 50µl 50µl 50µl 50µl 50µl 50µl 50µl 50µl 50µl

HI Titer / Result 1:2 1:4 1:8 1:16 1:32 1:64 1:128 1:256 1:512 1:1024 Negative Positive

50

Table-3.3 (Table of Burgh)

51

The corresponding NDV antibody titer was calculated from formula:

• HI Titer = 2n (n is highest well # showing hemagglutination inhibition)

Geometric mean titer (GMT) was measured from the Table developed by Burgh

(Table-3.3). The wells numbers showing clear hemagglutination inhibition were

recorded for each treatment groups. The arithmetic mean of well numbers in

each group was calculated. For example arithmetic mean of toxicated birds in the

group I was 5.1 at 5th week of age. The whole number (integer) thus obtained i.e.

5 was found in column (1/20) to the left of the Table. The decimal fraction (0.1)

was then located in the box heads of the rest of the Table. The value obtained

from the Table was 343. For two fold dilution value was divided by 10 to

determine the GMT (34.3) of the birds in that group.

Antibody titer of each bird was expressed in form of well # of immune-plate

that was whole number so was transformed into continuous data using LOG10.

The continuous data of each group was processed to calculate mean and

standard error. The means of groups were compared using two way ANOVA and

LSD’s multiple range (α=0.05) using SPSS version 16.

3.4.2. Determination of AFB residue in liver

AFB was measured in liver of the birds by slight modification of method of

Chiavaro et al. (Chiavaro et al., 2005). Liver sample (25 gm) was added to 100 ml

of mixture of methanol (Sigma-Aldrich) and distilled water (80:16), containing 5 g

of sodium chloride (Sigma-Aldrich) in 250 ml conical flask. The mixture was

homogenized by homogenizer for uniform mixing. The homogenized mixture was

placed in shaking water bath for an hour at 100 to 110 rpm and 25-28°C. After

shaking the filtration was done by using Whatman filter paper # 41. The volume

of filtrate was measured.

52

Ten ml aliquot of the filtrate was diluted with 40 ml of distilled water and

passed uniformly through the immuno-affinity column (Aflastar® from Romer Lab,

Rawalpindi-Pakistan) at 1.5 ml / minute. The AFB was eluted with 2 ml of

methanol (Sigma-Aldrich, Germany) in glass tube. Elute was evaporated to

dryness under nitrogen stream.

To the eluted AFB and the standards solutions, 100 µl of trifluroacetic acid

(Sigma-Aldrich, Germany) was added and kept at room temperature for 20

minutes in dark for pre-column derivatization. Mixture (400 µl) consisting of

acetonitrile / water (1:9) was added to tubes. Mobile phase consisting of the

acetonitrile / methanol / double distilled water (20:20:60) was degassed by

sonication. The 20 µl of sample was injected into HPLC with specification as

described in section 3.2.2.1 (b). The concentration of AFB in liver was calculated

comparing the peak area with peak of AFB standard. Some chromatographs are

shown in following figures.

Fig. 3.1 Standard of AFB

53

Fig. 3.2 AFB in bird of group III at 4th week of age

Fig. 3.3 AFB in bird of group VI at 4th week of age

54

Fig.3.4 AFB in bird of group VI at 5th week of age

Fig. 3.5 AFB in bird of group VII at 5th week of age

55

Fig.3.6 AFB in bird of group IV at 4th week of age

3.4.3. Clinical biochemistry

All biochemical tests were performed on Humalyzer® 3000 by using the

specific kit from Human®, Germany to assess the hepatic and renal functions.

3.4.3.1. Total serum protein

Total serum protein was determined using specific Kit from the Human®

which contained: Reagent composed of: sodium hydroxide 200 mM/L; potassium

sodium tartrate 32 mM/L; copper sulphate 12 mM/L and potassium iodide 30

mM/L. Standard solution contained protein 80 g/L and sodium azide 0.095%.

The standard solution (20 µl) was mixed with 1000 µl of the reagent and

incubated for 10 minutes at 25°C. Absorbance of the solution was measured at

546 nm using the Humalyzer 3000. The same was repeated with serum sample.

Standard curve and calculation was performed automatically by the instrument

with following formula:

• Protein (g/dl) = Absorbance of Sample / Absorbance of standard × 8

3.4.3.2. Serum Albumin

56

Serum albumin was measured photo-metrically using kit for serum albumin

from the Human®. The kit contained: Reagent composed of: bromocresol green

260 µM/L and citrate buffer 30 µM/L of pH 4.2. Standard solution contained

albumin 40 g/L of albumin and sodium azide 0.095%. The standard solution was

mixed with the reagent and incubated at 25°C for five minutes. Absorbance of the

solution was determined at 546 nm using the Humalyzer 3000. Serum sample

was treated similarly and the absorbance was determined using same parameter.

Standard curve and calculation was performed automatically by instrument with

following formula:

• Albumin (g/dl) = Absorbance of Sample / Absorbance of Standard × 4

3.4.3.3. Serum Bilirubin

Bilirubin in serum was measured by photometric method using Kit from the

Human® which contained: DCA containing 3.0 mM/L of 2, 4-dichloroaniline; 95

mM / L of hydrochloric Acid and 70 g/L of detergent. NIT solution contained 2.5

mM of sodium nitrite. BLK solution composed of 1.5 mM /L of 2,4-dichloroaniline;

57.5 mM/L hydrochloric Acid and 35 g/L of detergent. Working Reagent was

prepared by mixing DCA and NIT in 1:1 ratio and allowed to stand for 15 minutes

in dark.

The BLK (100 µl) was mixed with working reagent and kept for 10 minute

at 20-25°C in dark. Its absorbance was measured at 546 nm using the Humalyzer

3000®. The serum sample (100 µl) was mixed with the working reagent and

treated as that of BLK. Absorbance of sample was measured on same parameter

using the Humalyzer®. Serum bilirubin was measured by the formula:

Bilirubin (mg/dl) = Δ Absorbance of sample × 12.5

Δ Absorbance of sample = Absorbance of sample – Absorbance of Blank

3.4.3.4. Serum Glutamate-pyruvate transferase (SGPT)

57

Serum GPT level was measured by kinetic method using the kit from the

Human® containing: Buffer/ Enzyme reagent (BUF) composed of 125 mM /L of

TRIS buffer (pH 7.4); 625 mM / L of l-alanine; LDH (≥ 1.5 kU / L) and 0.095% of

sodium azide. Substrate (SUB) solution composed of: 2-oxoglutarate (75 mM / L);

0.9 mM of NADH and o.095% of sodium azide (0.095%).

Two ml of the SUB solution was added to 8 ml of the BUF solution and

mixed thoroughly to prepare reagent. The serum (200 µl) was added to 1000 µl of

the BUF solution and incubated for five minutes at 37°C. Then 250 µl of the SUB

solution was added. After proper mixing, absorbance was measured at 365 nm

exactly after one and two minutes against air (decreasing absorbance). SGPT

level was measured as:

SGPT (U/L) = Δ Absorbance / min × 2184

3.4.3.5. Serum Creatinine

Serum creatinine was measured through photometric test by kinetic

measurement using kit from the Human®. The kit contained: 26 mM / L of Picric

acid Solution and 1.6 mM / L of Sodium Hydroxide. Creatinine (Standard) solution

contained 176.8 mM / L. Sodium hydroxide solution was diluted with distilled

water in ration of 1+7. Picric acid solution was mixed with diluted sodium

hydroxide solution in 1:1 ratio.

Standard (100 µl) was mixed with 1000 µl of working reagent at 37°C.

Absorbance was measured at 490 nm after 30 seconds (A1) and then exactly

after two minutes (A2). Similarly 100 µl of sample was added to the 1000 µl of the

working reagent and the A1 and A2 of serum sample were measured at 490 nm

with the same parameter. The serum creatinine concentration was determined as

Serum Creatinine (mg/dl) = (A2-A1) of sample / (A2-A1) of Blank × 2.0

3.4.3.6. Serum Blood Urea Nitrogen (BUN)

58

Serum urea concentration was measured by enzymatic photometric test

with kit from the Human®. The kit contained: Reagent 1 (RGT1) which contains:

120 mM/L of Phosphate Buffer (pH 7.0); Sodium Salicylate (60 mM / L); Sodium

Nitroprusside (5 mM/L) and EDTA (1 mM/L). Reagent 2 (RGT2) contained: 120

mM/L of Phosphate Buffer (pH < 13) ; 0.6 g/ L of Hypochlorite and Enzyme

(ENZ) containing Urease (>500KU/L). Standard (STD) contained 80 mg/dl of

Urea with (0.095%) of Sodium azide.

Whole of the enzyme solution (ENZ) was poured into the RGT1 to prepare

the enzyme reagent 1a. Standard solution (10 µl) was mixed with the enzyme

reagent 1a and kept for three minutes at 37°C. The Reagent 2 (RGT2) (1000 µl)

was added and kept for 5 minutes at 37°C. Absorbance of solution was

measured at 546nm against reagent blank within 60 minutes. The same was

repeated with 10 µl of serum sample to measure the absorbance (ASAMPLE). The

urea in serum was determined by formula:

Serum Urea (mg/dl) = (ASAMPLE / ASTD) × 80

Serum BUN was then measured using conversion factor as under:

Serum BUN = 0.466 × Serum Urea (mg/dl)

3.4.4. Histopathological Examination

Formalin preserved liver, spleen, bursa of Fabricius and kidney were

processed for histopathological studies. The tissues were dehydrated by passing

through ascending series of ethanol. After passing through xylene, the tissues

were fixed in paraffin wax. Cross section (5 µm thick) was stained by hematoxylin

and eosin stain (Bankcroft and Gamble, 2007)

3.5. Statistical Analysis

All data are reported as mean ± SE. However, for analysis of

discrete data, it was transformed to continuous data by taking LOG10 of the data

59

for calculation of mean and standard error. All means were compared by two-

way analysis of variance (ANOVA) and a Least Significant Difference test was

applied to compare (α=0.05) the results using SPSS 16 (SPSS for Windows,

v16.0. SPSS Inc., Chicago, IL).

60

4. Results

4.1. Newcastle disease virus (NDV) antibody titer

Antibody titer of NDV vaccinated birds receiving different treatments

is shown in Table 4.2 while mean (n=8) of Log of NDV antibody titer of each

group is depicted in Table 4.1 and Fig. 4.1. The Log value was determined to

convert geometric data to arithmetic one for measuring the mean and variance.

NDV antibody titer of birds in different treatments on 4th, 5th, 6th, and 7th week of

age were significantly different (p<0.05).

Table-4.1

Mean of Log of antibody titer of birds taking different therapies for two weeks

Treatments 4th Week 5th Week 6th Week 7th Week

Group I (Control) 1.96±0.11b 1.88±0.11a 1.39±0.20a 2.82±0.06a

Group II (PBT1X) 2.30±0.16a 1.92±0.21a 1.20±0.20a 2.63±0.08a

Group III (AFB) 2.60±0.11a 1.13±0.12c 0.41±0.22c 1.81±0.20c

Group IV (PBT2X+AFB) 1.69±0.14b 1.73±0.08b 1.45±0.21a 2.22±0.11b

Group V (PBT1X+AFB) 1.02±0.17c 1.54±0.13b 1.09±0.18a 2.90±0.11a

Group VI (SLM + AFB) 2.44±0.07a 1.84±0.19a 0.68±0.20b 2.71±0.10a

Group VII (MYC + AFB ) 1.69±0.10b 2.22±0.14a 0.72±0.14b 2.79±0.08a

Each value represents mean of Log (10) of NDV antibody titer± Standard Error (n=8).AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The figures in the same column having similar superscript are not significantly different (p>0.05).Antibody response in form of well no. of each bird of each group was transformed into value of LOG10. That was continuous datum. The thus obtained was processed for calculation of mean and standard error. Means of each group was analyzed through two way analysis of variance followed by LSD test for comparison of means.

The NDV antibody titer determined in birds of group II (PBT1X) was

significantly higher than that of birds in the group I (Control) at age of 4th week.

Afterward, it remained comparable to NDV antibody titer measured in the group I

61

Table-4.2

The Distribution of serum of birds given various treatments according to Well Number in HI Test

PBT: Probiotics, AFB: Aflatoxin B1, SLM: Silymarin, MYC: Mycosorb

Age

Distribution of broilers (n=8) on the basis of HI titers GMT

Well # 0 1 2 3 4 5 6 7 8 9 10 n= average Well #

GMT = 2n

4th W

eek

Control - - - - - 2 1 4 1 - - 6.5 90.6

PBT1X - - - - - - 3 - 3 1 1 7.6 194.0

AFB - - - - - - - 2 - 5 1 9.9 388.6

PBT2X+AFB - - - - 2 2 1 3 - - - 5.6 42.5

PBT1X+AFB - 1 1 3 1 1 1 - - - - 3.4 10.6

SLM+AFB - - - - - - - 1 5 Tw

- 8.1 274.4

MYC+AFB - - - - - 3 4 1 - - - 5.8 55.7

5th W

eek

Control - - - - - 2 3 2 1 - - 6.3 78.8

PBT1X - - - 1 1 - - 5 - 1 - 6.4 84.4

AFB - - 1 3 1 3 - - - - - 3.8 13.9

PBT2X+AFB - - - - - 3 4 1 - - - 5.8 55.7

PBT1X+AFB - - - 4 1 2 - - 1 - 5.3 39.4

SLM+AFB - - - 1 - 2 1 3 1 - 6.1 68.6

MYC+AFB - - - - - 2 1 4 1 7.3 157.6

6th W

eek

Control - - - 4 1 1 2 - - - - 4.1 34.3

PBT1X 1 - - - 5 2 - - - - - 3.5 17.5

AFB 4 1 1 1 1 - - - - - 1.5 2.8

PBT2X+AFB - - 2 4 1 1 - - - 4.0 21.1

PBT1X+AFB - 2 3 3 - - - - - 3.6 12.1

SLM+AFB 2 1 1 2 1 1 - - - - - 2.3 4.9

MYC+AFB 1 4 1 2 - - - - - - 2.4 5.3

7th o

f wee

k

Control - - - - - - - - - 5 3 10.6 1552.1

PBT1X - - - - - - - 1 - 7 - 8.8 445.7

AFB - - 1 - 1 - 3 1 2 - - 5.9 59.7

PBT2X+AFB - - - - - - 2 2 3 1 - 7.4 168.9

PBT1X+AFB - - - - - - - 1 - - 7 9.6 776.0

SLM+AFB - - - - - - - 1 - 5 2 9.0 512.0

MYC+AFB - - - - - - - - 1 4 3 9.2 588.1

62

Each value represents mean of Log (10) of -NDV antibody titer± Standard Error (n=8); AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. Antibody response in form of well no. of each bird of each group was transformed into value of LOG10. That was continuous datum. The thus obtained was processed for calculation of mean and standard error. Means of each group was analyzed through two way analysis of variance followed by LSD test for comparison of means. The groups in the same week having similar numerical superscript are not significantly different (p>0.05).

Fig.4.1

63

from 5th to 7th week of age. The NDV antibody titer measured in birds of the group

III (AFB) was significantly higher than the birds in the group I (Control) at the age

of 4th week. Furthermore, a significant decrease in the antibody titer was

observed in birds of the group III than birds of the group I from age of 5th to 7th

week. Birds in group VI (SLM+AFB) showed no significant difference in the

antibody titer with birds of the group III at the age of 4th week. There was a

significant increase in the antibody titer of birds in the group VI than the birds in

the group III from 5th to 7th week of age. However, the rise in antibody titer of

these birds was comparable to birds in the group I at the age of 5th and 7th week.

Whereas, the significant (p<0.05) decrease in antibody titer was shown by birds

of the group when compared to birds in the group I at the age of 6th week. Birds

of the group VII (MYC+AFB) indicated antibody titer comparable to that of birds in

the group I at the age of 4th, 5th and 7th week. Likewise the birds of the group VI

(SLM+AFB), the antibody titer was significantly higher than titer observed in birds

of the group III but was not comparable to birds in the group I (Control) at the age

of 6th week (p<0.05).

No significant difference was observed in birds of group IV (PBT2X+AFB)

when compared to that of birds in the group I at the age of 4th and 6th week.

However, birds in the group IV showed significant (p<0.05) decrease in the

antibody titer than the birds in the group I at the 5th and 7th week of age. Birds in

the group IV indicated significant (p<0.05) decline in the antibody titer than that of

birds in the group III at the age of 4th week. Thereafter, the antibody titer

increased significantly (p<0.05) than that of birds in the group III from 5th to 7th

week of age. Moreover, titer measured in birds of the group IV was significantly

higher than that of birds in the group VI (SLM+AFB) at age of 4th & 6th week

(p<0.05). Furthermore, same birds indicated comparable titer with birds of the

64

group VII (MYC+AFB) on 4th week of age. But, the titer in these birds remained

significantly (p<0.05) decreased than that of birds in the group VII at the age of

5th and 7th of week. However, titer was significantly (p<0.05) greater than that of

birds in the group VII at age of 6th week.

The birds in the group V (PBT1X+AFB) indicated significant decrease in

antibody titer than that of birds in the group I on 4th and 5th week of age (p<0.05).

Thereafter, titer in the birds of the group V became comparable to that of birds in

the group I (p≥0.05).,Similar trend in increase in the antibody titer was observed

in birds of the group V when compared to birds receiving SLM or MYC with AFB

(group VI and VII respectively). Moreover, the antibody titer in birds of the group

IV was significantly p<0.05) higher than that of birds in the group V at the age of

4th week. Later, no significant difference in antibody titers was shown by birds

given either high or low dose PBT (group IV&V respectively) from 5th to 7th week

of age.

4.2. Total Leukocyte Count (TLC)

Log10 of total leukocyte count (Cells × 103/µl of blood) of each bird

receiving different treatments was calculated to convert discrete data to

continuous data for measurement of mean and standard error. Later means were

compared by two way ANOVA using LSD for multiple comparison. The value α

was taken as 0.05. Mean leukocytes count (n=6) of the birds given various

treatments is shown in Table 4.3 and Fig. 4.2. During the administration of

treatments (4th to 5th week of age), significant difference was observed in

leukocyte count. The birds in the group II (PBT1X) exhibited non-significant

variation in leukocyte count to the birds in group I (Control) from 4th to 7th week of

age. TLC in birds of the group III (AFB) exhibited significant decrease in TLC than

that of birds receiving other treatments at the age of 4th and 5th week.

65

Each value represents mean of total leukocyte count (Cell×1000/µl) ± Standard Error (n=6).AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The Log value of cell count of birds in each group was determined to convert geometric data to arithmetic one for measuring of mean and standard error. Means of each group was analyzed through two way analysis of variance followed by LSD test for comparison of means. The groups in the same week having similar numerical superscript are not significantly different (p>0.05)

Fig. 4.3

66

The birds in group VI (SLM + AFB) showed comparable leukocyte count

with birds in the group I at the age of 4th and 5th week. While birds in group VII

(MYC+ AFB) indicated leukocyte count equivalent to that of birds in the group I at

age of 4th week. However, at the age of 5th week, birds in the group VII showed

significant reduction (p<0.05) in the count than that of birds in the group I,

although it was significantly higher (p<0.05) than birds in the group III.

Table-4.3

Mean of Total Leukocyte counts (cell ×1000/µl) in birds taking different therapies for

two weeks

Each value represents mean of total leukocyte count (Cell×1000/µl) ± Standard Error (n=6).AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb; The figures in the same column having similar superscript are not significantly different (p>0.05).The Log value of cell count of birds in each group was determined to convert geometric data to arithmetic one for measuring of mean and standard error. Means of each group was analyzed through two way analysis of variance followed by LSD test for comparison of means.

Birds in group IV (PBT2X+AFB) indicated comparable count to that of birds

in the group-I, -VI and -VII at the age of 4th and 5th week. However, no significant

difference in TLC was observed in group V (PBT1X+AFB) when compared to the

Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 8.40 ± 0.03 a 8.91 ± 0.04 a 7.22 ± 0.12 a 7.45 ± 0.06 a

Group II (PBT1X) 8.37 ± 0.15 a 8.96 ± 0.13 a 7.15 ± 0.07 a 7.55 ± 0.10 a

Group III (AFB) 7.82 ± 0.39 b 7.45 ± 0.17c 7.10 ± 0.10 a 7.52 ± 0.07 a

Group IV (PBT2X+AFB) 8.07 ± 0.08 a 9.08 0.02 a 7.03 ± 0.03 a 7.48 ± 0.05 a

Group V (PBT1X+AFB) 7.74 ± 0.07 b 8.93 ± 0.04 a 7.08 ± 0.09 a 7.49 ± 0.06 a

Group VI (SLM + AFB) 8.15 ± 0.07 a 9.08 ± 0.12 a 7.03 ± 0.08 a 7.58 ± 0.05 a

Group VII (MYC + AFB ) 8.10 ± 0.08 a 8.53 ± 0.18b 7.00 ±0.08 a 7.47 ± 0.10 a

67

group III at the age of 4th week. However, on 5th week of age the TLC in these

birds became comparable to that of birds in the group I. Likewise these

observations are in line with our previous result of antibody titer at the age of 4th

week. Thereafter, no significant difference in leukocyte count was observed from

5th to 7th week in these groups.

4.3. Relative weight of bursa of Fabricius

Mean relative weight of bursa of Fabricius (BF) of birds (n=6) receiving

different treatment is shown in Table 4.4 and Fig. 4.3.

Table-4.4

Mean relative weight of bursa of Fabricius (g/Kg) of birds taking different therapies for

two weeks

Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 2.95 ± 0.17 a 0.78 ± 0.06 a 0.95 ± 0.12 b 0.61 ± 0.06 a

Group II (PBT1X) 1.76 ± 0.24 b 0.89 ± 0.07 a 1.36 ± 0.06 a 0.86 ± 0.04 a

Group III (AFB) 1.21 ± 0.09 c 0.67 ± 0.03 a 0.55 ± 0.06 c 0.41 ± 0.02 a

Group IV (PBT2X+AFB) 1.89 ± 0.04 b 1.00 ± 0.04 a 0.97 ± 0.04 b 0.57 ± 0.01 a

Group V (PBT1X+AFB) 1.98 ± 0.25 b 0.89 ± 0.04 a 0.78 ± 0.06 b 0.50 ± 0.02 a

Group VI (SLM + AFB) 1.51 ± 0.15 b 0.84 ± 0.05 a 0.86 ± 0.03 b 0.67 ± 0.04 a

Group VII (MYC + AFB ) 1.56 ± 0.24 b 0.82 ± 0.09 a 0.89 ± 0.05 b 0.71 ± 0.03 a

Each value represent mean of relative weight of bursa of Fabricius (g/Kg) ± Standard Error (n=6) AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The figures in the same column having similar superscript are not significantly different (p>0.05).

Birds in group II (PBT1X) exhibited significant decrease in relative weight of BF

than that of birds in group I (Control) at the age of 4th week. Thereafter, no

significant difference was observed in relative weight of BF when compared to

birds of the group I from 5th to 7th week of age. The birds in group III (AFB)

showed significant reduction (p<0.05) in relative weight of BF than that of birds in

group I (Control) at the age of 4th and 6th week. However no significant decrease

68

Each value represent mean of relative weight of Bursa of Fabricius (g/Kg) ± Standard Error (n=6).AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The groups in the same week having similar numerical superscript are not significantly different (p>0.05).

Fig. 4.3

69

was observed in birds of the group III when compared to birds in the group I at

the age of 5th and 7th week.

Birds in group VI (SLM+AFB) and group VII (MYC +AFB) indicated relative

weight of BF comparable to that of birds in the group I at the age of 5th to 7th

week. While at age of 4th week, birds of these groups exhibited significant

decrease (p<0.05) in relative weight of BF than that of birds in the group I

although it was significantly higher than the relative weight measured in birds of

the group III. The birds receiving both high and low dose PBT (group IV & V

respectively) showed similar trend in relative weight of BF like that of birds in the

group VI and VII. Moreover, no significant difference in relative weight of BF was

observed between birds of the group IV and V.

4.4. Relative Weight of Spleen

The mean relative weight of spleen in birds (n=6) taking various treatments

on 4,-5,-6 and -7 week of age is shown in Table 4.5 and Fig.4.4.

Table-4.5

Mean relative weight of spleen (g/Kg) of birds taking different therapies for two

weeks and immunized at day 1 & 30

Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 0.72 ± 0.06 a 1.12 ± 0.12 a 0.94 ± 0.06 a 1.00 ± 0.04 a

Group II (PBT1X) 0.60 ± 0.06 a 0.97 ± 0.06 a 0.73 ± 0.05 b 0.91 ± 0.05 a

Group III (AFB) 0.57 ± 0.02 a 0.67 ± 0.03 c 0.52 ± 0.04 d 0.96 ± 0.04 a

Group IV (PBT2X+AFB) 0.60 ± 0.05 a 0.84 ± 0.09 c 1.14 ± 0.05 c 1.08 ± 0.03 a

Group V (PBT1X+AFB) 0.67 ± 0.04 a 0.76 ± 0.04 c 1.20 ± 0.07 c 1.06 ± 0.08 a

Group VI (SLM + AFB) 0.62 ± 0.04 a 0.86 ± 0.05 b 0.67 ± 0.15 d 1.10 ± 0.03 a

Group VII (MYC + AFB ) 0.72 ± 0.06 a 1.05 ± 0.07 a 0.63 ± 0.16 d 0.91 ± 0.05 a

Each value represent mean of relative weight of spleen (g/Kg) ± Standard Error (n=6). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The figures in the same column having similar superscript are not significantly different (p>0.05).

70

Each value represent mean of relative weight of spleen (g/Kg) ± Standard Error (n=6). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The groups in the same week having similar numerical superscript are not significantly different (p>0.05).

Fig.4.4

71

No significant difference was shown in relative weight of spleen by birds

receiving various treatments at the age of 4th and 7th week. Moreover, no

significant difference in weight of spleen was observed in birds of group I (control)

and group II (PBT1X) at the age of 5th week. Furthermore, significant decrease

was shown in relative weight of spleen by birds in the group II (PBT1X) than that

of birds in the group I (Control) at the 6th week of age. However, relative weight of

spleen in birds of the group III (AFB) was significantly lower (p<0.05) than that of

birds in the group I at the age of 5th and 6th week of age. Birds in group VI (SLM +

AFB) exhibited significantly greater relative weight of spleen than the birds in the

group III although it was not comparable to relative weight of spleen in birds of

the group I at 5th week of age (p<0.05). Thereafter, no significant difference was

shown by birds in the group VI when compared to birds in the group III at the age

of 6th week. While, birds in group VII (MYC+AFB) showed relative weight of

spleen comparable to that of birds in the group I on 5th week of age. Likewise,

birds in the group VI, no significant increase was observed in these birds than the

birds of the group III at the age of 6th week.

No significant difference in relative weight of spleen in birds receiving high

and low dose PBT (group IV & V respectively) was observed to that of birds in the

group III (AFB) at the age of 5th week. Whereas, at the age of 6th week, relative

weight of spleen in birds taking PBT (both high and low doses), showed

significant rise (p<0.05) than that of the birds of the group III. In comparison to

birds given SLM or MYC with AFB, weight of spleen was significantly reduced

than weight of spleen in birds treated with PBT either high or low dose (group IV

& V respectively) at the age of 5th week. Whereas, significant increase in weight

of spleen was observed in birds given PBT2X or PBT1X with AFB than that of

birds in the group VI and VII at the age of 6th week (p>0.05). Weight of spleen

72

observed in the group IV and V respectively were comparable (p≥0.05) with each

other from 4th to 7th week of age.

4.5. Relative weight of Liver

Mean relative weight of liver in birds (n=6) given different treatments for

two weeks is shown in Table 4.6 and Fig. 4.5. No significant difference in liver

weight was observed in birds of group I (control) and II (PBT1X) from 4th to 7th

week of age. Birds in group III (AFB) remained significantly higher (p<0.05) than

that of liver weight shown by birds in the group I from 4th to 7th week of age.

Birds in group VI (SLM+AFB) showed weight of liver analogous to that of

birds in the group III at the age of 4th and 5th week. Later, it reduced significantly

than that of birds in the group III but was significantly greater (p<0.05) than the

liver weight shown by birds of the group I at age of 6th. However, it rose

significantly (p<0.05) and became comparable to that of birds in the group III at

age of 7th week. Whereas, no significant increase in liver weight of birds in group

VII (MYC+AFB) was observed than in birds of the group I from 4th to 7th week of

age. Birds of the group IV (PBT2X+AFB) indicated significant increase in liver

weight than the birds in the group I at age of 4th week. However, liver weight in

these birds was significantly lower than the birds in the group III at age of 4th

week. Afterward, it became comparable to that of birds in the group I from 5th to

7th week of age. Birds in group V (PBT1X+AFB) indicated relative weight of liver

comparable to that of birds in the group III at the age of 4th week. Furthermore,

birds of the group V indicated similar trend like that of birds in the group IV from

5th to 7th week of age. Birds given high dose of PBT (group IV) exhibited

significant decrease in liver weight than that of birds of the group VI (SLM + AFB)

from 4th to 7th week of age (p<0.05).

73

Table-4.6

Mean relative weight of liver (g/Kg) of birds taking different therapies for two

weeks

Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 32.12 ± 1.48 c 27.76 ± 1.48 b 22.42 ± 1.47 c 26.82 ± 0.88a

Group II (PBT1X) 31.33 ± 0.83 c 27.97 ± 2.02 b 27.39 ± 0.90 c 27.36 ± 1.25 a

Group III (AFB) 43.51 ± 2.69 a 34.67 ± 1.86 a 33.30 ± 2.98 a 35.95 ± 1.13 b

Group IV (PBT2X+AFB) 39.54 ± 1.33 b 27.00 ± 1.08 b 27.33 ± 0.71 c 29.07 ± 0.54 a

Group V (PBT1X+AFB) 40.22 ± 1.94 a 28.12 ± 1.30 b 25.56 ± 1.11 c 29.32 ± 1.10 a

Group VI (SLM + AFB) 45.87 ± 4.80 a 32.32 ± 0.88 a 28.41 ± 1.42 b 34.61 ± 0.89 b

Group VII (MYC + AFB ) 36.1 4± 1.17 c 27.24 ± 1.85 b 23.91 ± 1.58 c 29.05 ± 1.60 a

Each value represent mean of relative weight of liver (g/Kg) ± Standard Error (n=6).AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The figures in the same column having similar superscript are not significantly different (p>0.05).

Moreover, birds given low dose PBT (group V) indicated no significant decrease

in liver weight than that of birds in the group VI at 4th week of age. Afterward,

significant decrease was shown in liver weight of these birds than the birds of the

group V when compared to liver weight in birds of the group VI. Moreover, birds

in the group IV & V indicated significant rise (p<0.05) in liver weight than that of

bird in the group VII at 4th week of age. Thereafter, the liver weight observed in

these birds was comparable with birds of the group VII. However, significant rise

(p<0.05) was observed in liver weight of birds in the group V than weight of liver

in birds of the group IV at the age of 4th week. Afterward, no significant difference

was seen in birds of the either group.

4.6. Total Body Weight (TBW)

Mean total body weight of birds given various treatments is shown

in Table 4.7 and Fig. 4.6.

74

Each value represent mean of relative weight of Liver (g/Kg) ± Standard Error (n=6).AFB: Aflatoxin B1,PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The groups in the same week having similar numerical superscript are not significantly different (p>0.05).

Fig. 4.5

75

Table-4.7

Mean total body weight (Kg) of birds taking different therapies for two weeks

Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 1.02 ± 0.03 a 1.34 ± 0.05 a 1.56 ± 0.08 a 1.94 ± 0.07a

Group II (PBT1X) 1.06 ± 0.02 a 1.42 ± 0.05 a 1.64 ± 0.07 a 1.94 ± 0.10a

Group III (AFB) 0.72 ± 0.02 b 1.00 ± 0.03 b 1.17 ± 0.06 b 1.55 ± 0.04b

Group IV (PBT2X+AFB) 0.88 ± 0.03 a 1.26 ± 0.03 a 1.48 ± 0.02 a 1.97 ± 0.03a

Group V (PBT1X+AFB) 0.89 ± 0.04 a 1.32 ± 0.04 a 1.51 ± 0.07 a 1.93 ± 0.08a

Group VI (SLM + AFB) 0.87 ± 0.03 b 1.22 ± 0.03 a 1.48 ± 0.06 a 1.79 ± 0.05a

Group VII (MYC + AFB ) 0.97 ± 0.04 a 1.32 ± 0.06 a 1.46 ± 0.08 a 1.71 ± 0.09b

Each value represent mean total body weight (Kg) ± Standard Error (n=6).AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The figures in the same column having similar superscript are not significantly different (p>0.05)..

No significant decrease in TBW was shown by birds of the group I

(Control) and group II (PBT1X). Birds in group III (AFB) exhibited significant

reduction (p<0.05) in TBW than that of birds in the group I from 4th to 7th week of

age. Birds taking SLM with AFB (group VI) indicated significant reduction

(p<0.05) in TBW than that of birds in the group I at the age of 4th week.

Furthermore, no significant decrease of TBW was observed in birds of the group

VI when compared to birds in the group I from 5th to 7th week of age. Birds given

MYC with AFB contaminated feed (group VII) displayed TBW comparable to that

of birds in the group I from 4th to 6th week of age (p≥0.05). However, birds

showed significant decline in TBW than the birds of the group I at the age of 7th

week.

Birds receiving both high and low dose of PBT with AFB contaminated

feed (group IV & V respectively) depicted comparable TBW to that of observed in

birds of the group I from 4th to 7th week of age. The body weight indicated by

birds in these two group was significantly higher than that of birds given SLM

76

Each value represent mean of total body weight (Kg) ± Standard Error (n=6). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The groups in the same week having similar numerical superscript are not significantly different (p>0.05).

Fig. 4.6

77

(group VI) at the age of 4th week (p<0.05). Afterward, it became comparable to

TBW in birds of the group VI from 5th to 7th week of age. In comparison to birds

given MYC, the body weight observed in PBT treated birds was comparable to

that of birds in the group VII from 4th to 6th week of age. However, TBW was

significantly higher (p>0.05) than MYC treated birds at the age of 7th week

4.7. Total Serum Protein (TSP)

Mean total serum protein (g/dl) of birds receiving various treatments for

two weeks is shown in Table-4.8 and Fig.4.7. Total serum protein concentration

in birds of the group II (PBT1X) was significantly (p<0.05) lower than that of birds

in the group I (Control) at the age of 6th week. While it remained comparable to

birds of the group I on 4th, 5th and 7th week of age. TSP concentration was

significantly lower in birds fed with AFB contaminated feed (group III) than that of

birds received basal feed (group I) during study except at 5th week of age

(p>0.05).

Table-4.8

Mean total serum protein level (g/dl) of birds taking different therapies for two weeks

Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 2.5 ± 0.12 a 2.8 ± 0.15 a 3.7 ± 0.14 a 4.1 ± 0.15 a

Group II (PBT1X) 2.6 ± 0.42 a 3.2 ± 0.17 a 2.7 ± 0.16 b 3.6 ± 0.07 a

Group III (AFB) 1.8 ± 0.17 b 1.5 ± 0.11 b 1.8 ± 0.20 c 2.3 ± 0.18 c

Group IV (PBT2X+AFB) 3.1 ± 0.41 a 2.4 ± 0.11 a 3.5 ± 0.33 a 3.7 ± 0.21 a

Group V (PBT1X+AFB) 3.0 ± 0.23 a 2.8 ± 0.25 a 3.0 ± 0.37 b 3.3 ± 0.14 b

Group VI (SLM + AFB) 2.2 ± 0.27 b 2.9 ± 0.30 a 2.5 ± 0.24 b 3.4 ± 0.27 a

Group VII (MYC + AFB ) 2.7 ± 0.21 a 3.2 ± 0.15 a 3.4 ± 0.14 a 3.5 ± 0.12 a

Each value represent mean total serum protein (g/100ml) ± Standard Error (n=6). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The figures in the same column having similar superscript are not significantly different (p>0.05)

78

Each value represent mean of total serum protein (g/100ml) ± Standard Error (n=6).AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The groups in the same week having similar numerical superscript are not significantly different (p>0.05)

Fig. 4.7

79

While on 5th week of age the TSP was comparable to that of birds in the

group I (p≥0.05).Birds given SLM with AFB contaminated feed (Group VI)

indicated comparable TSP concentration to that of birds in the group I on 5th and

7th week of age. While at 4th week of age it was similar to the concentration

recorded in birds of the group III (p≥0.05). However, it was significantly higher

(p<0.05) than birds of the group III on 6th week of age although it was not

equivalent to the birds of the group I. The birds receiving MYC with AFB

contaminated feed (group VII) indicated comparable TSP concertation to that of

birds in the group I from 4th to 7th week of age.

Birds given high dose of PBT with AFB contaminated feed (group IV)

indicated the comparable total serum protein concentration to that of birds in the

group I from 4th to 7th week of age. The serum protein concentration in these

birds was equivalent to that of birds given MYC (group VII). In comparison to

birds of the group VI, no significant decrease was observed in TSP concentration

at the age of 4th, 5th and 7th week. However, TSP concentration was significantly

(p<0.05) higher than birds of the group VI at 6th week of age.

Birds given low dose PBT with AFB (group V) indicated significant

(p<0.05) rise in TSP than that of birds in the group III. TSP concentration in these

birds was analogue to the concentration observed in birds of the group I & VII on

4th and 5th week of age. However, on 6th and 7th week of age, serum protein

concentration indicated significant decrease in TSP concentration as compared

to that birds in the group I and VII. Moreover, results recorded in these birds were

significantly higher than the birds given SLM on 4th week of age. Later, the

protein concentration in these birds became comparable to birds of the group VI

at age of 5th and 6th week. . Later, TSP concentration in birds of the group V

80

declined and became significantly lesser than that of birds given other

interventions.

4.8. Serum Albumin

Mean serum albumin concentration in birds given different treatments for

two weeks is shown in Table 4.9 and Fig. 4.8. The serum albumin level in birds of

the group II (PBT1X) was comparable to that of birds given basal feed (group I) at

the age of 4th and 6th week. However, serum albumin concentration was

significantly higher than the birds of the group I at the age of 5th and 7th week.

Birds receiving AFB contaminated feed (group III) depicted significant (p<0.05)

decrease in serum albumin concentration than that of observed in bird fed basal

feed (group I) from 4th to 7th age of week.

Table-4.9

Mean serum albumin level (g/100ml) of birds taking different therapies for two weeks Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 1.9 ± 0.07 b 1.9 ± 0.02 b 2.0 ± 0.09 a 2.2 ± 0.10 b

Group II (PBT1X) 2.0 ± 0.34 b 2.6 ± 0.17 a 1.9 ± 0.02 a 2.8 ± 0.07 a

Group III (AFB) 1.3 ± 0.15 c 1.0 ± 0.15 c 1.0 ± 0.14 b 1.4 ± 0.12 c

Group IV (PBT2X+AFB) 2.7 ± 0.31 a 1.0 ± 0.18 c 1.2 ± 0.26 b 1.9 ± 0.09 b

Group V (PBT1X+AFB) 1.7 ± 0.18 c 1.5 ± 0.12 c 1.0 ± 0.16 b 1.6 ± 0.08 b

Group VI (SLM + AFB) 1.6 ± 0.16 c 1.4 ± 0.22 c 1.2 ± 0.38 b 1.6 ± 0.20 b

Group VII (MYC + AFB ) 1.9 ± 0.12 b 1.1 ± 0.17 c 1.5 ± 0.15 a 2.4 ± 0.19 b

Each value represent mean serum albumin (g/100ml) ± Standard Error (n=6).AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The figures in the same column having similar superscript are not significantly different (p>0.05).

Birds taking SLM with AFB contaminated feed (group VI) indicated serum

albumin concentration comparable to birds in the group III on 4th and 5th week of

age. Later, serum albumin concentration in these birds raised significantly

(p<0.05) than that of birds in the group III but was not equivalent

81

Each value represent mean of serum albumin (g/dl) ± Standard Error (n=6);AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The groups in the same week having similar numerical superscript are not significantly different (p>0.05)

Fig. 4.8

82

to serum albumin concentration in birds of the group I. Birds given MYC with AFB

contaminated feed (group VII) showed serum albumin concentration analogue to

that of birds in the group I at the age of 4th, 6th and 7th week. However, at the age

of 5th week the serum albumin concertation in these birds indicated decline and

was comparable to that of birds in the group III. The birds given high dose PBT

with AFB (group IV) indicated serum albumin level significantly (p<0.05) higher

than that of shown by birds given basal feed (group I), SLM +AFB (group VI) and

MYC+AFB (group VII) at the age of 4th week. However, serum albumin

concertation in these birds declined and became comparable to that of birds

given AFB (group III) on 5th and 6th week of age (p≥0.05). Later, on 7th week of

age it raised significantly (p<0.05) higher than serum albumin level shown by

birds in the group III and became analogue to that of birds in the group I.

Likewise, birds receiving low dose PBT with AFB contaminated feed (group V)

showed serum albumin level equivalent to that of birds in the group III from 4th to

6th week of age. However, it became comparable to the serum concentration

observed in birds of the group I on 7th week of age.

4.9. Serum Glutamate-pyruvate Transferase (SGPT)

SGPT concentration (U/L) is depicted in Table 4.10 and Fig. 4.9. No

significant rise was observed in birds of the group II when compared to SGPT

concentration in birds of the group I. SGPT concentration in birds given AFB

contaminated feed (group III) was significantly (p<0.05) higher than that of birds

given basal feed only (group I) from 4th to 7th week of age. Birds given SLM with

AFB (group VI) showed SGPT concentration analogue to that of birds in the

group III on 4th, 6th and 7th week of age (p≥0.05). However, at 5th week of age, it

was significantly lower level than birds of the group III but was not analogous to

birds in the group I (p<0.05).

83

Table-4.10

Mean SGPT level (U/L) of birds taking different therapies for two weeks

Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 13.60 ± 0.50 b 14.52 ± 1.18 c 13.73 ± 0.72 c 12.55 ± 0.74 b

Group II (PBT1X) 14.87 ± 1.94 b 12.05 ± 0.59 c 17.08 ± 0.82 c 11.23 ± 2.91 b

Group III (AFB) 28.40 ± 4.54 a 42.28 ± 1.53 a 37.82 ± 1.71 a 33.25 ± 1.08 a

Group IV (PBT2X+AFB) 15.78 ± 2.04 b 20.25 ± 1.64 c 22.38 ± 2.03 b 18.57 ± 1.24 b

Group V (PBT1X+AFB) 17.15 ± 1.93 b 23.10 ± 5.49 b 25.67 ± 5.08 b 14.77 ± 1.45 b

Group VI (SLM + AFB) 24.03 ± 1.50 a 34.57 ± 5.25 b 32.38 ± 1.27 a 29.65 ± 7.16 a

Group VII (MYC + AFB ) 12.92 ± 0.49 b 27.78 ± 1.84 b 31.92 ± 0.94 a 27.58 ± 1.07 a

Each value represent mean serum GPT (U/L) ± Standard Error (n=6). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The figures in the same column having similar superscript are not significantly different (p>0.05)

Birds given MYC with AFB (group VII) exhibited significantly (p<0.05)

lower SGPT concentration than that of birds in the group III at the age of 4th and

5th week. However on 6th and 7th week of age, increase in SGPT concentration in

these birds was comparable to that of birds in the group III. The Birds receiving

high dose of PBT with AFB (group IV) indicated comparable concentration of

SGPT to the birds receiving basal feed (group I) at the age of 4th, 5th and 7th

week. Whereas, at the age of 6th week, it was significantly (p<0.05) lower than

that of birds in the group III although was not comparable to birds in the group I.

The SGPT concentration in these birds was comparable to birds given MYC

(group VII) on 4th week of age. Thereafter, it became significantly lower than that

of birds receiving MYC (p<0.05). Moreover, protective effect of PBT in these birds

was significantly (p<0.05) lower than the birds fed SLM with AFB (group VI) from

4th to 7th week of age.

Birds given low dose PBT (group V) indicated significant (p<0.05)

reduction in SGPT concentration than that of birds of the group III from 4th to 7th

84

Each value represent mean of SGPT (U/l) ± Standard Error (n=6). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The groups in the same week having similar numerical superscript are not significantly different (p>0.05).

Fig. 4.9

85

week of age. However, the SGPT level in these birds was comparable to that of

birds in the group I on 4th, 7th week of age. The SGPT concentration in such birds

was comparable to birds given SLM with AFB at the age of 5th week. Moreover,

there was significant decrease in SGPT concentration in these birds than that of

birds in the group VI on 4th, 6th and 7th week of age. Furthermore, birds of the

group V indicated SGPT concentration similar to that of birds in the group VII on

4th and 5th week of age. Whereas, on 6th and 7th week of age, it was significantly

lower than the birds of the group VII. Birds receiving PBT either low or high dose

exhibited comparable SGPT concentration at the age of 4th, 6th and 7th week. At

5th week of age, SGPT concentration in birds of the group V was significantly

higher than the birds in the group IV.

4.10. Serum Bilirubin

Serum bilirubin level of broilers received different treatments for two weeks

is shown in Table 4.11 and graphically in Fig. 4.10.

Table-4.11

Mean serum bilirubin level (mg/dl) of birds taking different therapies for two weeks Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 0.35 ± 0.01 b 0.38 ± 0.02 b 0.37 ± 0.02 b 0.36 ± 0.02 a

Group II (PBT1X) 0.27 ± 0.03 b 0.36 ± 0.05 b 0.40 ± 0.02 b 0.40 ± 0.03 a

Group III (AFB) 0.71 ± 0.10 a 0.77 ± 0.08 a 0.63 ± 0.02 a 0.52 ± 0.02 a

Group IV (PBT2X+AFB) 0.33 ± 0.02 a 0.39 ± 0.06 b 0.38 ± 0.06 b 0.36 ± 0.06 a

Group V (PBT1X+AFB) 0.31 ± 0.04 b 0.38 ± 0.09 b 0.38 ± 0.08 b 0.36 ± 0.05 a

Group VI (SLM + AFB) 0.61 ± 0.02 a 0.49 ± 0.08 b 0.31 ± 0.05 b 0.29 ± 0.03 a

Group VII (MYC + AFB ) 0.43 ± 0.06 b 0.46 ± 0.11 b 0.41 ± 0.10 b 0.38 ± 0.01 a

Each value represent mean serum bilirubin level (mg/dl) ± Standard Error (n=6). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The figures in the same column having similar superscript are not significantly different (p>0.05).

86

No significant rise in serum bilirubin level was observed in birds of the

group II (PBT1X) as compared to birds in group I (Control). Significantly (p<0.05)

elevated concentration of serum bilirubin was measured in birds fed AFB with

basal feed (group III) as compared to birds given basal feed (group I) from 4th to

6th week of age. However it was normalized at age of 7th week. Birds taking SLM

with AFB (group VI) indicated serum bilirubin level analogous to birds in the

group III at the age 4th week. Later it normalized and became comparable to that

of birds in the group I. No significant increase in serum bilirubin was exhibited in

birds of group VII (MYC+AFB) than that of birds in the group I from 4th to 6th week

of age.

Birds given high dose PBT with AFB (group IV) displayed serum bilirubin

concentration equivalent to that of birds in the group I, VI and VII on 5th and 6th

week of age. However on 4th week of age the serum bilirubin concentration was

analogue to birds fed AFB contaminated feed (group III). While birds receiving

low dose PBT (group VI) indicated serum bilirubin concentration comparable to

that birds in the group I and VII from 4th to 6th week of age. In comparison to SLM

taking birds (group VI) serum bilirubin level was significantly (p<0.05) lower at the

age of 4th week. However, it was comparable to that of birds in the group VI at the

age of 5th and 6th week. Moreover, serum bilirubin concentration in birds given

high or low dose PBT exhibited non-significant difference at from 5th to 6th week

of age. Furthermore, significant rise in serum bilirubin level was observed in birds

of the group IV than that of birds in the group V on 4th week of age. At the age of

7th week, no significant difference was shown in serum bilirubin level among birds

receiving various interventions.

87

Each value represent mean of serum bilirubin (mg/dl) ± Standard Error (n=6) AFB: Aflatoxin B1,PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The groups in the same week having similar numerical superscript are not significantly different (p>0.05)

Fig. 4.10

88

4.11. Serum Blood Urea Nitrogen

Effect of various treatments on serum blood urea nitrogen (BUN) is shown

in Table 4.12 and graphically in Fig.4.11. Significant difference was observed

among birds given various treatments on 4th and 5th week of age (p<0.05). Birds

given PBT1X (group II) indicated serum BUN level analogous to birds receiving

basal feed (group I) at 4th and 5th week of age. Birds fed AFB with basal feed

(group III) indicated significant decline in serum BUN than the birds in the group I

at 4th and 5th week of age. The Birds given SLM with AFB (group VI) displayed

equivalent serum BUN concentration to that of birds in the group III at the age of

4th week. While, birds given MYC with AFB (group VII) showed significantly

(p<0.05) higher serum BUN concentration than birds in the group III at the 4th

week of age but it was not analogue to the serum BUN concentration of birds in

the group I.

Table-4.12

Mean serum BUN level (mg/dl) of birds taking different therapies for two weeks

Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 6.15 ± 1.10 a 6.20 ± 0.34 a 5.03 ± 0.88 a 4.47 ± 0.74 a

Group II (PBT1X) 6.10 ± 0.70 a 6.21 ± 0.41 a 5.11 ± 0.85 a 4.38 ± 0.61 a

Group III (AFB) 4.06 ± 0.40 c 3.43± 0.34 b 4.02 ± 0.94 a 4.19 ± 0.61 a

Group IV (PBT2X+AFB) 5.81 ± 0.63 a 3.24 ± 0.48 b 4.75 ± 0.42 a 4.83 ± 0.29 a

Group V (PBT1X+AFB) 6.57 ± 0.57 a 4.31 ± 0.32 b 4.71 ± 0.27 a 4.95 ± 0.28 a

Group VI (SLM + AFB) 4.53 ± 1.03 c 2.42 ± 0.40 b 3.88 ± 0.29 a 4.05 ± 0.61 a

Group VII (MYC + AFB ) 6.10 ± 0.89 b 4.31 ± 0.28 b 4.83 ± 0.36 a 4.40 ± 0.40 a

Each value represent mean serum BUN level (mg/dl) ± Standard Error (n=6). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb, BUN: Blood Urea Nitrogen The figures in the same column having similar superscript are not significantly different (p>0.05).

Birds given PBT (both high and low dose) showed serum BUN

89

Each value represent mean of serum BUN (mg/dl) ± Standard Error (n=6) AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb, BUN:

Blood Urea Nitrogen. The groups in the same week having similar numerical superscript are not significantly different (p>0.05).

Fig. 4.11

90

concentration comparable to that of birds in the group I at the age of 4th week.

The serum BUN concentration was significantly higher in these birds than that of

birds in the group VI and VII. Moreover, serum BUN concentration was alike in

birds in either group IV or V at the age of 4th week. Nevertheless, on 5th week of

age, birds reeving either of interventions indicated similar serum BUN

concentration as that of observed in birds of the group III. Later, birds given

different treatments showed non-significant variation in serum BUN concentration

on 6th and 7th week of age.

4.12. Serum Creatinine

The effect of various treatments given for two weeks on mean serum

creatinine level is shown in Table 4.13 and Fig.4.12. Non-significant variation was

recorded among birds given various treatments with that of birds fed bsal feed

(p≥0.05).

Table-4.13

Mean serum creatinine level (mg/dl) of birds taking different therapies for two

weeks

Treatments Week 4 Week 5 Week 6 Week 7

Group I (Control) 0.32 ± 0.05 a 0.35 ± 0.02 a 0.37 ± 0.03 a 0.33 ± 0.04 a

Group II (PBT1X) 0.38 ± 0.03 a 0.38 ± 0.06 a 0.37 ± 0.04 a 0.32 ± 0.04 a

Group III (AFB) 0.40 ± 0.04 a 0.45 ± 0.03 a 0.42 ± 0.05 a 0.35 ± 0.03 a

Group IV (PBT2X+AFB) 0.30 ± 0.05 a 0.35 ± 0.03 a 0.27 ± 0.02 a 0.25 ± 0.02 a

Group V (PBT1X+AFB) 0.33 ± 0.06 a 0.38 ± 0.04 a 0.33 ± 0.03 a 0.28 ± 0.03 a

Group VI (SLM + AFB) 0.37 ± 0.08 a 0.45 ± 0.04 a 0.37 ± 0.02 a 0.35 ± 0.02 a

Group VII (MYC + AFB ) 0.40 ± 0.04 a 0.40 ± 0.04 a 0.35 ± 0.06 a 0.30 ± 0.03 a

Each value represent mean serum creatinine level (mg/dl) ± Standard Error (n=6). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The figures in the same column having similar superscript are not significantly different (p>0.05)

91

Each value represent mean of serum creatinine (mg/dl) ± Standard Error (n=6) AFB: Aflatoxin B1,PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb. The groups in the same week having similar numerical superscript are not significantly different (p>0.05)

Fig. 4.12

92

4.13. AFB Residue in liver

Table 4.14 and Fig. 4.13 depicted the mean AFB residue in liver of birds

given various treatments for two weeks. Birds in group I (Control) and group II

(PBT1X) did not indicate any detectable residue in liver from 4th to 7th week of

age. While, Birds given AFB contaminated feed (group III) exhibited detectable

level of AFB residue in the liver from 4th to 7th week of age.

Table-4.14

Mean of AFB residue (ng/g) in livers of birds taking different therapies for two weeks

Treatments 4th Week 5th Week 6th Week 7th Week

Group I (Control) ND ND ND ND

Group II (PBT1X) ND ND ND ND

Group III (AFB) 2.46±0.09 a 9.22±0.8 a 7.65±0.56 a 5.52± 0.6 a

Group IV (PBT2X+AFB) ND ND ND ND

Group V (PBT1X+AFB) ND 0.77±0.02 b ND ND

Group VI (SLM + AFB) 1.98±0.06 a 4.49±0.25 b 2.06±0.65 b ND

Group VII (MYC+ AFB ) ND 0.77±0.04 b 0.66±0.04 b ND

Each value represents mean of AFB residue in livers ± Standard Error (n=4). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb, ND: Non detectable. The figures in the same column having similar superscript are not significantly different (p>0.05).

Birds taking SLM with AFB (group VI) indicated measurable AFB residue

from 4th to 6th week of age. Whereas, birds fed MYC with AFB (group VII)

indicated measurable level of AFB in liver on 5th and 6th week of age. The

measureable level in birds of both the groups was significantly (p<0.05) lower

than the birds of the group III. Birds given high dose of PBT with AFB (group IV)

did not show any detectable level of AFB in liver from 4th to 7th week of age.

However, birds given low level of PBT (group V) displayed detectable level of

AFB in liver on 5th week of study only which was equivalent to that of birds in the

group VI and VII (p≥0.05).

93

Each value represents mean of AFB residue in livers ± Standard Error (n=4.). AFB: Aflatoxin B1, PBT: Probiotics, SLM: Silymarin, MYC: Mycosorb The groups in the same week having similar numerical superscript are not significantly different (p>0.05).

Fig. 4.1

94

4.14. Histopathological Examination

4.14.1. Liver

At 4th week of age, intake of AFB cause moderate degenerative changes

in liver of birds (group III) as compared to that of birds given basal feed only

(group I). Microscopic examination revealed vacuolar degeneration and early sign

of bile duct hyperplasia. Further exposure resulted in sever degenerative

changes in liver parenchyma (Plate i and j), vacuolar degeneration, pycnotic

nuclei and hepatocellular degeneration. Bile duct hyperplasia along with loss of

hepatic plate structure was also observed in these birds. These degenerative

changes persisted even at 6th week of age. However, at 7th week of age, the

hepatic tissues showed little recovery: from sever to moderate degenerative

changes.

Birds given SLM with AFB (group VI) and MYC +AFB (group VII) indicated

moderate degree of damage on 4th, 5th, 6th and 7th weeks of age. Liver of birds

given both high and low dose PBT (groups IV and V respectively) showed

moderate damage on 4th week of age. However on 5th, 6th and 7th weeks of age,

liver of the birds in these groups exhibited mild hepatocellular changes besides

normal parenchyma of cells (Plate k& l).

4.14.2. Bursa of Fabricius (BF)

On 4th week of age bursa of Fabricius of birds in the group III (AFB)

showed mild lymphoid degenerative changes as compared with the BF in the

group I and II. Later on 5th week of age BF showed depletion of bursal follicles,

loss of cortical cells and decreased cellular density. These atrophic changes were

also present on 6th week of birds in the group III (Plate a & b). Birds in group VI

(SLM +AFB) and group VII (MYC+AFB) showed moderate damage on 4th and 5th

95

week of age whereas, mild degenerative changes were observed on 6th and 7th

week of age (Plate c and d). Similarly, birds given high or low dose of PBT

(groups IV, V respectively) indicated mild loss of bursal follicle on 5th week of age

but indicated non-significant damage on 4th, 6th and 7th week of age i.e. Cells in

cortex remained intact,.

4.14.3 Spleen

On spleen, effect was mild but lymphocytic degeneration was also seen in

birds fed AFB (group III) on 4th and 5th week of age. On week 6th and 7th of age,

damage is not significant. Spleen of birds given various treatments exhibited non-

significant damage.

4.14.4. Kidney

Kidney of birds taking AFB (group III) showed moderated degree of

damage with atrophy of bowman corpuscle, loss of luminal limning and

desquamation of luminal epithelial cells in tubules from 4th to 6th week of age.

However, On 7th week of age, kidney showed regenerative changes with mild

degree of restoration in tubular lining cells in the birds of the group III (plates e

and f ) Birds in the groups given SLM or MYC with AFB (group VI and VII

respectively) showed atrophied glomerulus on 5th of age. Whereas, Birds in the

group IV (PBT2X+AFB) and V(PBT1X+AFB) indicated intact luminal lining of

tubules and improved appearance of bowman corpuscles on 5th week of age

(Plate g)..

96

(a) (b)

(c) (d)

a. Bursal follicle (10X) in PBT treated Birds for two weeks b. Bursal follicle (40X) in PBT treated Birds for two weeks c. Depleted Bursal follicle (10X) in AFB treated Birds for two weeks d. Depleted Bursal follicle (40X) in AFB treated Birds for two weeks

97

(e) (f)

(g) (h)

e. Kidney (40X)of MYC for two weeks treated (2week of exposure) f. Kidney (40X)of AFB for two weeks treated (2week of exposure) g. Kidney (40X)of SLM for two weeks treated (2week of exposure) h. Kidney (40X)of PBT for two weeks treated (2week of exposure)

98

(i) (j)

(k) (l)

i. Liver of AFB treated birds for two weeks (10X) j. Liver of AFB treated birds for two weeks (40X) k. Liver of PBT treated birds for two weeks (10X) l. Liver of PBT treated birds for two weeks (40X)

99

5. Discussions & Conclusion

Aflatoxin B1 (AFB), a secondary metabolite produced by many species of

Aspergillus, is classified as group I carcinogen by IARC (Wei and Jong, 1986;

IARC, 2002). AFB to be used in study was produced through rice fermentation.

The rice fermentation by Asperigllus flavus on flask shaker produced a level of

700 to 800 ppb of the toxin, which is in line of observations reported by Shotwell

et al. (1966) and Yunus and Böhm (2011).

To assess the immunosuppression serum NDV-antibody titer, leukocytes

count, relative weight of lymphoid organ such as bursa of Fabricius and spleen

were determined. Initially an increase in NDV antibody titer was observed after

one week of AFB exposure (at week 4 of age) in bird of the group III. This

temporary rise in humoral response by AFB was reported and described by

various researchers (Giambrone et al., 1985; Yunus et al., 2011b). The exact

mechanism of the initial increase in immune response is not known and may be

due to body’s protective response to the intoxication. However, AFB induced a

strong immuno-suppressive effect as observed by significant low titer values of

the NDV-antibody titer in birds of the group III after two weeks of exposure (week

5 of age) and even one week after cessation of exposure (week 6 of age). The

suppressed NDV-antibody level even after stoppage of intoxication may be due

toxicity of preformed AFB guanine adduct. The observed immuno-toxicity of AFB

is in line with previous reports (Meissonnier et al., 2008a; Chen et al., 2013b). A

rise in antibody titer in AFB treated birds after two weeks of stopping the

ingestion of AFB (week 7 of age) shows recovery of the birds from the

100

immunosuppression within two weeks after termination of exposure which

suggests that immunosuppressive effect of AFB is reversible.

Silymarin is an established hepato-protectant through its antioxidant action

(Muriel and Mourelle, 1990). It does not offer any protective effect on NDV-

antibody response, against AFB on week 4 of age in birds. The lack of protective

action at week 4 of age (1st week of exposure) indicates that SLM does not affect

the absorption of AFB. But SLM showed protective action on NDV-antibody titer

on 5-, 6- and 7-week of age. The later improved responses in birds of the group

VI although significantly lesser than PBT treated birds, can be explained through

its neutralizing effect via scavenging of free radicals and restoration of normal

glutathione level (Rastogi et al., 2001; Tedesco et al., 2004a; Chand et al., 2011).

MYC showed a protective role on NDV antibody response against

deleterious effects of AFB during the exposure by adsorption in birds of the group

VII. However, the protective effect was lost during the first week of terminating

the exposure (week 6 of age). It suggests that continuous intake of MYC may be

required to maintain protective effects of the MYC. These findings were

supported by observation of other researchers (Ghahri et al., 2009; Hasan et al.,

2010; Verbrugghe et al., 2012; Mogadam and Azizpour, 2013)

Both concentrations of PBT (1X and 2X) ameliorated the immuno-

suppressive effects of AFB in birds of the group IV and V as demonstrated by

significantly elevated NDV-antibody titer values in birds of these groups than the

birds in the group III. Secondly comparable NDV- antibody titer of birds in the

group II with that of birds in the group I, reflected lack of immunomodulation, if

given alone. It clearly reveals that higher NDV antibody titer observed in birds of

the group IV and V is due to the toxin adsorptive properties of PBT. The data

clearly demonstrate the adsorptive properties of Lactobacilli against AFB as

101

reported earlier (Peltonen et al., 2001a; Fazeli et al., 2009a; Salminen et al.,

2010b; Kumar et al., 2011). The protective action of PBT was equivalent to that of

MYC and SLM. The restoration of NDV antibody titer at the 6th week indicates

protective effect persisted even after stopping of intake of PBT. This effect could

be result of either decline in absorption during exposure or enhancing the

elimination of toxic metabolites.

Leukocytes plays central role in generation of immune response either

humoral or cellular. The data of present study show that administration of low

dose of AFB to the broilers for two week causes mild leukopenia which is in

accordance to previously published reports (Samuel et al., 2009). The AFB-

induced leukopenia is reversible and subsides as AFB-intake is stopped. Co-

administration of PBT (Lactobacilli) caused complete recovery from the AFB-

induced reduction in leukocyte count. The protective effect of lactobacilli during

the AFB exposure results in detoxification of AFB presumably via adsorption

(Fazeli et al., 2009b; Salminen et al., 2010a; Kumar et al., 2011) and beneficial

effect of PBT on leukocyte count as reported previously (Apata, 2008; Salim et

al., 2013). Intake of SLM ameliorates the leukopenia caused by AFB which is

attributed to its antioxidant properties reported by different researchers (Tedesco

et al., 2004a; Lutsenko et al., 2008; Lee et al., 2010). Similarly addition of MYC in

AFB contaminated diet prevents the negative effect of AFB on leukocytes count

which is line with observation of other workers (Basmacioglu et al., 2005; Che et

al., 2011; Xu et al., 2011)

Bursa of Fabricius is an important lymphoid organ in broilers which is

usually regressed slowly with age and vanishes upon maturity. An abrupt

reduction in weight of bursa of Fabricius was observed in birds of the group III

compared to that of birds in the group I. The atrophied changes in bursa of

102

Fabricius were not recovered during one week post-exposure period (week 6 of

age).These findings are supported by the observation of various researchers

(Smith et al., 1992; Sur and Celİk, 2003; Wang et al., 2009; Chen et al., 2014a).

Likewise, intake of MYC also exhibited improvement in the bursal weight of birds

of the group VII on week-4 and -6 which are in line of the previous studies

(Karaman et al., 2005; Girish and Devegowda, 2006; He et al., 2013b). Feeding

of SLM to birds also ameliorated the negative effect of AFB which are related to

its antioxidant abilities supported by the findings of other researchers (Abidin et

al., 2011; Chand et al., 2011).

PBT administration in diet significantly prevented the AFB-induced

regression and prevented cellular damage in bursa of Fabricius. The bursal

weight recorded in birds of the group IV and V was appeared to be better than

that of birds in the group VI and VIII. Birds in these groups indicated mild

degenerative changes in bursa of Fabricius. The ameliorative effect of PBT at

one week of exposure can be explained on basis of adsorptive properties of the

bacterial cell wall as reported by other scientists (Gratz et al., 2007; Hernandez-

Mendoza et al., 2009a; Hernandez-Mendoza et al., 2011; Bovo et al., 2014). The

suppressive actions on leukocyte count and bursa of Fabricius is also supported

by significant decrease in NDV antibody titer measured in these birds. Moreover,

protection after stoppage of intake can be explained through colonization of

Lactobacilli in intestine of birds and their positive effect on immune system.

Spleen an important organ of reticuloendothelial system involved in

immunity (white pulp). AFB reversibly causes reduction in spleen weight which

returns to normal within two weeks after stopping the toxin intake. The reversible

suppression of spleen weight was supported by findings of other researchers

(Chaytor et al., 2011; Chen et al., 2014b). Histopathological findings suggest

103

resistance of spleen to AFB’s negative effect. Similarly comparable spleen weight

in birds of the group VII on week-5 of age (week-1 exposure) indicated the toxin

binding properties of MYC as reported by other workers (Raju and Devegowda,

2000b; Basmacioglu et al., 2005; Armando et al., 2011; Zoghi et al., 2014;

Gonçalves et al., 2015). Lack of protective effect after stopping the MYC

ingestion necessitates continuous intake of MYC for protective action (Girish and

Devegowda, 2006; Matur et al., 2011; Mogadam and Azizpour, 2013). Likewise,

SLM administration produced better impact on spleen weight on week-5 of age.

The improved value in birds of the group VI indicated beneficial effect of SLM in

AFB toxicity on spleen which is attributed to protective action against oxidative

stress produced by AFB as reported earlier (Tedesco et al., 2004b; Kalorey et al.,

2005; Chand et al., 2011).

Feeding of PBT with contaminated diet did not prevent reduction in relative

weight of spleen during the AFB exposure; however, PBT accelerated the

recovery of spleen weight during post-exposure period indicated by normalized

spleen weight on week-6 of age. This beneficial effect on week-1 of exposure (5th

week of age) was linked to reduced bioavailability of AFB through adsorption by

the Lactobacilli. The reduced absorption results in decrease in formation of the

toxic metabolites. These findings are supported by observation of various

investigators (Fazeli et al., 2009b; Bovo et al., 2014; Zoghi et al., 2014). Positive

effect at 6th week of age suggests impact of Lactobacilli on either AFB activation

or elimination of epoxide metabolite.

Total body weight is an important parameter to assess growth and

nutritional consequences in AFB-induced toxicity. AFB ingestion caused

reduction in total body weight in birds which remain significantly lower from 4th to

7th week of age. The decline in total body weight in the birds of the group III is

104

linked with decreased feed intake due to AFB toxicity. These findings are in line

of previous studies conducted in various species by various scientists (He et al.,

2013a; Indresh et al., 2013; Kumar et al., 2013; Weaver et al., 2013; Chen et al.,

2014d).

MYC intake in birds significantly guards against AFB toxicity during

exposure and one week post exposure. But it fails to exhibit any protection after

two week of stopping the ingestion (week-7 of age). These findings are in line

with findings of researchers in broilers as well as other species (Karaman et al.,

2005; Indresh et al., 2013; Zychowski et al., 2013; Selim et al., 2014). SLM upon

ingestion showed shielding effect against AFB on week-5, -6 and -7 of age. The

lack of effect on toxin absorption can be reason for the absence of protection on

week 4 of age (one week-exposure). But later significant ameliorative action is

attributed to its anti-oxidative properties as studied by different scientists

(Tedesco et al., 2004a; Chand et al., 2011; Dumari et al., 2014; Kamali and

Mostafaei, 2014).

Co-administration of PBT ameliorated the harmful effect of AFB during the

exposure and post-exposure. The protective action from 4th to 5th week of age

was comparable to birds in the group VII can be explained via adsorptive

properties of these Lactobacilli. The adsorptive abilities were reported by various

researchers (Thongsong et al.; Peltonen et al., 2001b; Fazeli et al., 2009b;

Hernandez-Mendoza et al., 2009a; Nikbakht Nasrabadi et al., 2013). However,

amelioration from cumulative effect at the age of 6th to 7th week of age suggests

mechanism other than adsorption.

The liver is major site of AFB activation and more susceptible to AFB

toxicity (Wild and Turner, 2002). Ingestion of AFB to birds resulted in significant

rise in relative weight of liver (hepatomegaly) during the exposure and it remained

105

higher even after stopping the toxin intake. These observation are in line with

previous findings (Vekiru et al.; Magnoli et al., 2011; Chen et al., 2014c; Fowler et

al., 2014). Administration of PBT (Group IV) in high dose combination prevented

the hepatomegaly during exposure (4 to 5 week of age) as well as post-exposure

(6 to 7 week of age). However low dose PBT combination (Group V) showed

protection after week-2 of exposure (5th week of age) and maintained this effect

till 7th week of age. The persistent rise after the exposure shows cumulative

toxicity of AFB on hepatomegaly. Feeding of PBT with basal diet (group II) has

no significant influence on liver weight in hosts. The beneficial effect of PBT in

presence of toxin can be explained through its AFB binding abilities which have

been studied by various researches (Gratz et al., 2006; Fazeli et al., 2009b;

Hernandez-Mendoza et al., 2009a; Hernandez-Mendoza et al., 2010; Hathout et

al., 2011; Zoghi et al., 2014).

Intake of silymarin does not indicate any beneficial AFB during the

exposure. These finding demonstrates that SLM does not affect the absorption of

AFB. These observations are in line with previous reports (Tedesco et al., 2004a;

Suchý et al., 2008). Protective action recorded at week-6 of age indicates the

recovery from hepatomegaly but rise in liver weight at week-7 indicates lack of

protection against the cumulative action of AFB. SLM showed the ameliorative

action against the AFB toxicity through its antioxidant actions and scavenging of

free radical as reported by different scientists (Tedesco et al., 2004b; Kalorey et

al., 2005; Chand et al., 2011). Administration of MYC significantly prevented the

deleterious effect of the AFB on hepatomegaly during exposure (week 4 to 5) and

week one post-exposure (week-6 of age). However, lack protection at week-7 of

age proposes the continuous intake to provide amelioration. These observation

are supported by outcomes of several researchers (Raju and Devegowda, 2000a;

106

Aravind et al., 2003; Yildirim et al., 2011; Verbrugghe et al., 2012; Cao and

Wang, 2014)

Serum protein and albumin are an early indicator of hepatic injury during

aflatoxicosis (Huff et al., 1986). Both total serum protein and albumin has been

decreased in birds of the group III. The decline was significant during exposure

(week-4 to 5) and in post-exposure (week-6 to 7). The hypoproteinemia and

hypoalbuminemia in hosts was in line with previous findings (Karabacak et al.;

Manafi et al., 2012; Shamsudeen and Shrivastava, 2013; Chen et al., 2014d;

Chibanga et al., 2014).. The protective action in birds given PBT can be ascribed

to the toxin neutralizing properties of Lactobacilli presumably via adsorption.

These adsorptive /neutralizing properties were studied by several researchers

(Fazeli et al., 2009b; Hernandez-Mendoza et al., 2009a; Hernandez-Mendoza et

al., 2011; Bagherzadeh Kasmani et al., 2012; Zoghi et al., 2014). The protective

action of PBT on TSP and serum albumin was less than MYC.

Similarly protection offered by SLM was significantly lower than that of

PBT treated birds. Similarly early failure of the protective effect is associated with

negligible effect on the toxin adsorption and hence absorption. However, delayed

and weak protection in SLM fed birds, is due to its action against oxidative stress

and free radicals. These findings have also been studied in other species and

reported by various scientists (Rastogi et al., 2000; Saller et al., 2001; Tedesco et

al., 2003; Tedesco et al., 2004b; Dumari et al., 2014). Ingestion pf MYC displayed

amelioration against negative effect AFB on total serum protein and albumin

level. The protection is based on its effect to reduce the toxin bioavailability. This

adsorptive action of MYC was previously reported by other researchers (Aravind

et al., 2003; Karaman et al., 2005; Ghahri et al., 2009; Hasan et al., 2010; Yildirim

et al., 2011; Verbrugghe et al., 2012). Likewise, lower proteins concentration in

107

post-exposure time period demands continuous feeding of such detoxifying

agents and shows delayed toxicity of AFB even after intake was stopped.

SGPT is one of the transaminase present in hepatocytes which enters the

blood /serum during hepatic necrosis. Bilirubin is an important product of

hepatocytes which may enters the circulation during biliary tract obstruction. AFB

intake results in rise of both SGPT and bilirubin level as sign of hepatic injury.

The AFB produced rise in the level of SGPT and bilirubin was observed during

the toxin intake and even after stopping the ingestion of the toxin. These

observations are in line with previous reports of other investigators (Petkov et al.,

1985; Valchev et al., 2014). Same is true for other species (Cheng et al., 2001;

Rastogi et al., 2001). Histopathological examination of liver revealed severs

degenerative changes including vacuolar degeneration, karyolysis, bile duct

hyperplasia and loss of hepatic plate. Theses finding are supported by raised

concentration of bilirubin and SGPT. These histopathological findings are

supported by studies of different scientists (Denli et al., 2009; Yang et al., 2012;

Weaver et al., 2013).

Ingestion of SLM failed to show any protection after week-1 of exposure

(week-4 of age) on SGPT and bilirubin level like that of serum albumin and total

proteins. Failure of the protection on early phase indicates lack of effect on toxin

adsorption and so its bioavailability. However, later amelioration on week-5 and -

6 was due to its effect on oxidative stress caused by AFB. The observed

antioxidant action of SLM in the study is previously reported by various scientists

(Rastogi et al., 2000; Saller et al., 2001; Dumari et al., 2014). Similarly, MYC an

established mycotoxin adsorbent showed protection during the exposure but

protective action was lost after stopping the intake. The protective action seen

was due to absorption and is in line with previous findings of other researchers

108

(Basmacioglu et al., 2005; Karaman et al., 2005; Girish and Devegowda, 2006;

Yildirim et al., 2011). PBT intake resulted in normalized level of SGPT and

bilirubin during exposure as well as post exposure. Similarly mild to moderate

damage was observed in liver of birds in the group IV and V on week-5,-6 and -7

of age. Protective action of PBT was better than MYC or SLM. The significant

amelioration of the AFB toxicity by ingestion of PBT in the birds through feed can

be associated with the toxin adsorption during exposure by such bacteria. The

adsorptive properties are reported previously by several researchers (Peltonen et

al., 2001b; Gratz et al., 2006; Hernandez-Mendoza et al., 2010; Salminen et al.,

2010a; Hathout et al., 2011). However, protection in post-exposure phase

suggests some additional mechanism along with adsorption. Moreover, non-

significant impact of feeding PBT with basal feed (group II) also endorses the

above findings.

Serum creatinine and blood urea nitrogen were used to investigate

toxic effects on kidney by AFB and hence its amelioration. Lack of any significant

effect on serum creatinine by AFB during the exposure indicates that kidney is

not the main target of AFB toxicity. Blood urea nitrogen is an indicator of liver and

kidney function in early age of birds. Ingestion of AFB for two weeks produce

decline in serum BUN but this action was reversible upon stoppage of ingestion.

The decline in the BUN was reported by different researchers (Raju and

Devegowda, 2000a; Aravind et al., 2003; Li et al., 2012). AFB ingestion produced

atrophied bowman corpuscle, loss of luminal limning and desquamation of

luminal epithelial cells in tubules which were mildly recovered upon stopping of

the toxin ingestion. These findings were supported by Kamel (Kamel, 2013).

SLM administration does not show any protection on one week exposure.

However, later observed shielding effect can be explained by its anti-oxidant role.

109

The anti-oxidant role is reported previously by various investigators (Rastogi et

al., 2000; Tedesco et al., 2004b; Lutsenko et al., 2008). The protective action of

MYC during exposure on serum BUN is linked to its adsorptive action which is in

line with previous findings (Girish and Devegowda, 2006; Ghahri et al., 2009;

Hasan et al., 2010; Yildirim et al., 2011; Verbrugghe et al., 2012). However,

Kidney of birds in the group VI and VII showed atrophied glomeruli.

PBT treatments indicate improvement in histological feature on week-5 of

age and produced beneficial impact which was linked to the adsorptive ability of

Lactobacilli. It reduced the toxin absorption and hence the toxicity. The adsorptive

action of Lactobacilli is supported by studies of different researchers (El-Nezami

et al., 1998; Haskard et al., 2001; Peltonen et al., 2001b; Fazeli et al., 2009b;

Salminen et al., 2010a; Zhong et al., 2014).

5.1. Conclusions

The data of the current study demonstrate that live Lactobacilli present in

Pakistani traditional food significantly ameliorated the AFB immuno-toxicity as

well as hepatotoxicity presumably through its adsorptive action thus reducing the

toxin bioavailability. Moreover, persistent protective action after stopping the

intake shows advantage over other intervention (SLM or MYC). Furthermore, it

also suggests some additional mechanisms for neutralization of absorbed toxin or

its toxic metabolite rather than simple adsorption.

It is also suggested that addition of Dhai (source of PBT) in food or feed

decrease the likelihood of aflatoxicosis in human or birds in such regions where

prevention of fungal contamination is not practically feasible due to either cost or

unawareness of community. Moreover, addition of such probiotics in the diet of

patients suffering from hepatitis C or B patients results in delay of progression

toward HCC in such regions. Furthermore, lack of interaction with micronutrients,

110

low cost, ease of availability and control of Lactobacilli make such source of PBT

as an economical and practical remedy to reduce toxicity of the aflatoxins in

under-developed (African countries) and developing countries like Pakistan etc.

In future, further research is needed to elucidate the any additional

mechanism for AFB neutralization besides adsorption of the toxin by Lactobacilli.

111

6. References

1. Abidin, Z., Khatoon, A., Numan, M., 2011. Mycotoxins in broilers:

pathological alterations induced by aflatoxins and ochratoxins, diagnosis

and determination, treatment and control of mycotoxicosis. World's Poultry

Science Journal 67, 485-496.

2. Al-Hammadi, S., Marzouqi, F., Al-Mansouri, A., Shahin, A., Al-Shamsi, M.,

Mensah-Brown, E., Souid, A.K., 2014. The cytotoxicity of aflatoxin b1 in

human lymphocytes. Sultan Qaboos Univ Med J 14, e65-e71.

3. Allan, W., Gough, R., 1974. A standard haemagglutination inhibition test

for Newcastle disease. (1). A comparison of macro and micro methods.

Veterinary Record 95, 120-123.

4. Alvarez-Olmos, M.I., Oberhelman, R.A., 2001. Probiotic agents and

infectious diseases: a modern perspective on a traditional therapy. Clin

Infect Dis 32, 1567-1576.

5. Anjum, M., Sahota, A., Akram, M., Ali, I., 2011. Prevalence of mycotoxins

in poultry feeds and feed ingredients in Punjab (Pakistan). The Journal of

Animal and Plant Sciences 2, 117-120.

6. Apata, D.F., 2008. Growth performance, nutrient digestibility and immune

response of broiler chicks fed diets supplemented with a culture of

Lactobacillus bulgaricus. Journal of the Science of Food and Agriculture

88, 1253-1258.

7. Aravind, K., Patil, V., Devegowda, G., Umakantha, B., Ganpule, S., 2003.

Efficacy of esterified glucomannan to counteract mycotoxicosis in naturally

contaminated feed on performance and serum biochemical and

hematological parameters in broilers. Poultry Science 82, 571-576.

112

8. Armando, M., Dogi, C., Pizzolitto, R., Escobar, F., Peirano, M., Salvano,

M., Sabini, L., Combina, M., Dalcero, A., Cavaglieri, L., 2011.

Saccharomyces cerevisiae strains from animal environment with in vitro

aflatoxin B&lt;sub&gt;1&lt;/sub&gt; binding ability and anti-pathogenic

bacterial influence. World Mycotoxin Journal 4, 59-68.

9. Aslam, N., Tipu, M.Y., Ishaq, M., Cowling, A., McGill, D., Warriach, H.M.,

Wynn, P., 2016. Higher Levels of Aflatoxin M1 Contamination and Poorer

Composition of Milk Supplied by Informal Milk Marketing Chains in

Pakistan. Toxins 8.

10. Aslam, S., Qazi, J.I., 2010. Isolation of acidophilic lactic acid bacteria

antagonistic to microbial contaminants. Pakistan Journal of Zoology 42,

567-573.

11. Aso, Y., Akaza, H., Kotake, T., Tsukamoto, T., Imai, K., Naito, S., 1995.

Preventive effect of a Lactobacillus casei preparation on the recurrence of

superficial bladder cancer in a double-blind trial. The BLP Study Group.

Eur Urol 27, 104-109.

12. Aziz, T., Khan, H., Bakhtair, S., Naurin, M., 2009. Incidence and relative

abundance of lactic acid bacteria in raw milk of buffalo, cow and sheep. J

Anim Plant Sci 19, 168-173.

13. Azziz-Baumgartner, E., Lindblade, K., Gieseker, K., Rogers, H.S., Kieszak,

S., Njapau, H., Schleicher, R., McCoy, L.F., Misore, A., DeCock, K., 2005.

Case–control study of an acute aflatoxicosis outbreak, Kenya, 2004.

Environmental Health Perspectives 113, 1779.

14. Bagherzadeh Kasmani, F., Karimi Torshizi, M.A., Allameh, A.,

Shariatmadari, F., 2012. A novel aflatoxin-binding Bacillus probiotic:

113

Performance, serum biochemistry, and immunological parameters in

Japanese quail. Poultry Science 91, 1846-1853.

15. Bajaj, J.S., Saeian, K., Christensen, K.M., Hafeezullah, M., Varma, R.R.,

Franco, J., Pleuss, J.A., Krakower, G., Hoffmann, R.G., Binion, D.G.,

2008. Probiotic Yogurt for the Treatment of Minimal Hepatic

Encephalopathy. Am J Gastroenterol 103, 1707-1715.

16. Bankcroft, J., Gamble, M., 2007. Theory and Practice of Histological

Technique. 5th ed. Churchill Livingstone,London.

17. Basmacioglu, H., Oguz, H., Ergul, M., Col, R., Birdane, Y., 2005. Effect of

dietary esterified glucomannan on performance, serum biochemistry and

haematology in broilers exposed to aflatoxin. Czech Journal of Animal

Science 50, 31-39.

18. Bedard, L.L., Massey, T.E., 2006. Aflatoxin B1-induced DNA damage and

its repair. Cancer Lett 241, 174-183.

19. Besselink, M.G., van Santvoort, H.C., Buskens, E., Boermeester, M.A.,

van Goor, H., Timmerman, H.M., Nieuwenhuijs, V.B., Bollen, T.L., van

Ramshorst, B., Witteman, B.J., Rosman, C., Ploeg, R.J., Brink, M.A.,

Schaapherder, A.F., Dejong, C.H., Wahab, P.J., van Laarhoven, C.J., van

der Harst, E., van Eijck, C.H., Cuesta, M.A., Akkermans, L.M., Gooszen,

H.G., 2008. Probiotic prophylaxis in predicted severe acute pancreatitis: a

randomised, double-blind, placebo-controlled trial. Lancet 371, 651-659.

20. Bhatnagar, D., Cary, J.W., Ehrlich, K., Yu, J., Cleveland, T.E., 2006.

Understanding the genetics of regulation of aflatoxin production and

Aspergillus flavus development. Mycopathologia 162, 155-166.

21. Bintvihok, A., Kositcharoenkul, S., 2006. Effect of dietary calcium

propionate on performance, hepatic enzyme activities and aflatoxin

114

residues in broilers fed a diet containing low levels of aflatoxin B1. Toxicon

47, 41-46.

22. Bovo, F., Franco, L.T., Rosim, R.E., Trindade, C.S.F., De Oliveira, C.A.F.,

2014. The Ability of Lactobacillus rhamnosus in Solution, Spray-Dried or

Lyophilized to Bind Aflatoxin B1. Journal of Food Research 3, p35.

23. Brisbin, J.T., Gong, J., Orouji, S., Esufali, J., Mallick, A.I., Parvizi, P.,

Shewen, P.E., Sharif, S., 2011. Oral Treatment of Chickens with

Lactobacilli Influences Elicitation of Immune Responses. Clinical and

Vaccine Immunology 18, 1447-1455.

24. Brown, R., Chen, Z.-Y., Cleveland, T., Russin, J., 1999. Advances in the

development of host resistance in corn to aflatoxin contamination by

Aspergillus flavus. Phytopathology 89, 113-117.

25. Brugh, M., Jr., 1978. A simple method for recording and analyzing

serological data. Avian Dis 22, 362-365.

26. Bruneau, J.C., Stack, E., O'Kennedy, R., Loscher, C.E., 2012. Aflatoxins

B(1), B(2) and G(1) modulate cytokine secretion and cell surface marker

expression in J774A.1 murine macrophages. Toxicol In Vitro 26, 686-693.

27. Bullerman, L.B., Bianchini, A., 2007. Stability of mycotoxins during food

processing. International Journal of Food Microbiology 119, 140-146.

28. Cao, J., Wang, W., 2014. Effects of astaxanthin and esterified

glucomannan on hematological and serum parameters, and liver

pathological changes in broilers fed aflatoxin-B1-contaminated feed.

Animal Science Journal 85, 150-157.

29. Cary, J.W., Rajasekaran, K., Brown, R.L., Luo, M., Chen, Z.-Y.,

Bhatnagar, D., 2011. Developing Resistance to Aflatoxin in Maize and

Cottonseed. Toxins 3, 678-696.

115

30. Çelýk, K., Denlý, M., Savas, T., 2003. Reduction of toxic effects of

aflatoxin B1 by using baker yeast (Saccharomyces cerevisiae) in growing

broiler chicks diets. Revista Brasileira de Zootecnia 32, 615-619.

31. Chand, N., Muhammad, D., Durrani, F., Qureshi, M.S., Ullah, S.S., 2011.

Protective effects of milk thistle (Silybum marianum) against aflatoxin B1 in

broiler chicks. Asian-Australasian Journal of Animal Sciences 24, 1011-

1018.

32. Chaytor, A.C., See, M.T., Hansen, J.A., de Souza, A.L.P., Middleton, T.F.,

Kim, S.W., 2011. Effects of chronic exposure of diets with reduced

concentrations of aflatoxin and deoxynivalenol on growth and immune

status of pigs1. J. Anim. Sci. 89, 124-135.

33. Che, Z., Liu, Y., Wang, H., Zhu, H., Hou, Y., Ding, B., 2011. The protective

effects of different mycotoxin adsorbents against blood and liver

pathological changes induced by mold-contaminated feed in broilers.

Asian–Aust J Anim Sci 24, 250-257.

34. Chen, J., Chen, K., Yuan, S., Peng, X., Fang, J., Wang, F., Cui, H., Chen,

Z., Yuan, J., Geng, Y., 2013a. Effects of aflatoxin B1 on oxidative stress

markers and apoptosis of spleens in broilers. Toxicol Ind Health 4, 4.

35. Chen, K., Fang, J., Peng, X., Cui, H., Chen, J., Wang, F., Chen, Z., Zuo,

Z., Deng, J., Lai, W., Zhou, Y., 2014a. Effect of selenium supplementation

on aflatoxin B1-induced histopathological lesions and apoptosis in bursa of

Fabricius in broilers. Food and Chemical Toxicology 74, 91-97.

36. Chen, K., Peng, X., Fang, J., Cui, H., Zuo, Z., Deng, J., Chen, Z., Geng,

Y., Lai, W., Tang, L., 2014b. Effects of dietary selenium on

histopathological changes and T cells of spleen in broilers exposed to

116

aflatoxin B1. International journal of environmental research and public

health 11, 1904-1913.

37. Chen, K., Shu, G., Peng, X., Fang, J., Cui, H., Chen, J., Wang, F., Chen,

Z., Zuo, Z., Deng, J., Geng, Y., Lai, W., 2013b. Protective role of sodium

selenite on histopathological lesions, decreased T-cell subsets and

increased apoptosis of thymus in broilers intoxicated with aflatoxin B(1).

Food. Chem. Toxicol 59, 446-454.

38. Chen, X., Horn, N., Applegate, T.J., 2014c. Efficiency of hydrated sodium

calcium aluminosilicate to ameliorate the adverse effects of graded levels

of aflatoxin B1 in broiler chicks. Poultry Science.

39. Chen, X., Horn, N., Cotter, P., Applegate, T., 2014d. Growth, serum

biochemistry, complement activity, and liver gene expression responses of

Pekin ducklings to graded levels of cultured aflatoxin B1. Poultry Science,

PS3904.

40. Chen, X., Horn, N., Cotter, P.F., Applegate, T.J., 2014e. Growth, serum

biochemistry, complement activity, and liver gene expression responses of

Pekin ducklings to graded levels of cultured aflatoxin B1. Poult Sci 93,

2028-2036.

41. Cheng, C.T., 1992. Perak, Malaysia, mass poisoning. Tale of the Nine

Emperor Gods and rat tail noodles. Am J Forensic Med Pathol 13, 261-

263.

42. Cheng, Y.H., Shen, T.F., Pang, V.F., Chen, B.J., 2001. Effects of aflatoxin

and carotenoids on growth performance and immune response in mule

ducklings. Comp Biochem Physiol C Toxicol Pharmacol 128, 19-26.

43. Chiavaro, E., Cacchioli, C., Berni, E., Spotti, E., 2005. Immunoaffinity

clean-up and direct fluorescence measurement of aflatoxins B1 and M1 in

117

pig liver: comparison with high-performance liquid chromatography

determination. Food Addit Contam 22, 1154-1161.

44. Chibanga, J.F., Banda-Nyirenda, D., Simbaya, J., 2014. Effects of

Supplementing Different Levels of Vitamin A to Aflatoxin B1 Contaminated

Dietson the Performance of Broiler Chickens. Asian Journal of Natural &

Applied Sciences Vol 3, 4.

45. Chung, T.K., Erdman, J.W., Jr., Baker, D.H., 1990. Hydrated sodium

calcium aluminosilicate: effects on zinc, manganese, vitamin A, and

riboflavin utilization. Poult Sci 69, 1364-1370.

46. Costanzo, P., Santini, A., Fattore, L., Novellino, E., Ritieni, A., 2014.

Toxicity of aflatoxin B1 towards the vitamin D receptor (VDR). Food Chem

Toxicol 4, 00498-00490.

47. De Groote, M.A., Frank, D.N., Dowell, E., Glode, M.P., Pace, N.R., 2005.

LACTOBACILLUS RHAMNOSUS GG BACTEREMIA ASSOCIATED WITH

PROBIOTIC USE IN A CHILD WITH SHORT GUT SYNDROME. The

Pediatric Infectious Disease Journal 24, 278-280.

48. Denli, M., Blandon, J.C., Guynot, M.E., Salado, S., Perez, J.F., 2009.

Effects of dietary AflaDetox on performance, serum biochemistry,

histopathological changes, and aflatoxin residues in broilers exposed to

aflatoxin B1. Poultry Science 88, 1444-1451.

49. Diaz, G.J., Murcia, H.W., Cepeda, S.M., 2010. Cytochrome P450 enzymes

involved in the metabolism of aflatoxin B1 in chickens and quail. Poult Sci

89, 2461-2469.

50. Dimitri, R.A., Gabal, M.A., 1996. Immunosuppressant activity of aflatoxin

ingestion in rabbits measured by response to Mycobacterium bovis

118

antigen I. Cell mediated immune response measured by skin test reaction.

Vet Hum Toxicol 38, 333-336.

51. Dumari, M.A., Sarir, H., Makki, O.F., Afzali, N., 2014. Effect of Milk Thistle

(Silybum marianum L.) on Biochemical Parameters and Immunity of

Broiler Chicks Fed Aflatoxin B1 after Three Weeks. Iranian Journal of

Toxicology Volume 8.

52. Eaton, D.L., Gallagher, E.P., 1994. Mechanisms of aflatoxin

carcinogenesis. Annu Rev Pharmacol Toxicol 34, 135-172.

53. El-Nezami, H., Kankaanpaa, P., Salminen, S., Ahokas, J., 1998. Ability of

dairy strains of lactic acid bacteria to bind a common food carcinogen,

aflatoxin B-1. Food and chemical toxicology 36, 321-323.

54. El-Nezami, H., Nicoletti, G., Neal, G., Donohue, D., Ahokas, J., 1995.

Aflatoxin M< sub> 1</sub> in human breast milk samples from Victoria,

Australia and Thailand. Food and chemical toxicology 33, 173-179.

55. Escamilla, J., Lane, M.A., Maitin, V., 2012. Cell-Free Supernatants from

Probiotic Lactobacillus casei and Lactobacillus rhamnosus GG Decrease

Colon Cancer Cell Invasion In Vitro. Nutrition and Cancer 64, 871-878.

56. Etzel, R.A., 2002. MYcotoxins. JAMA 287, 425-427.

57. Farah, N., Sadia, M, Shakira, G.S.S, Tahir, N, Rauf, M.I., et al., 2016.

Isolation, characterization and application of indigenous lactic acid bacteria

in milk fermentation. Int. J. Biosci 9, 415-430.

58. Fazeli, M.R., Hajimohammadali, M., Moshkani, A., Samadi, N., Jamalifar,

H., Khoshayand, M.R., Vaghari, E., Pouragahi, S., 2009a. Aflatoxin B1

binding capacity of autochthonous strains of lactic acid bacteria. J. Food

Prot 72, 189-192.

119

59. Fazeli, M.R., Hajimohammadali, M., Moshkani, A., Samadi, N., Jamalifar,

H., Khoshayand, M.R., Vaghari, E., Pouragahi, S., 2009b. Aflatoxin B1

binding capacity of autochthonous strains of lactic acid bacteria. J Food

Prot 72, 189-192.

60. Fernández, A., Verde, M.T., Gascón, M., Ramos, J.J., Gómez, J., 1994.

Aflatoxin and its metabolites in tissues from laying hens and broiler

chickens fed a contaminated diet. Journal of the Science of Food and

Agriculture 65, 407-414.

61. Fowler, H.M., Barrientos-Velazquez, A., Deng, Y., Bailey, C., 2014.

Utilization of a Spray-Applied Calcium Bentonite Clay to Ameliorate the

Effects of Low-Levels of Aflatoxinin Starter Broiler Diets Containing DDGS.

Natural Products Chemistry & Research.

62. Gabal, M.A., Dimitri, R.A., 1998. Humoral immunosuppressant activity of

aflatoxin ingestion in rabbits measured by response to Mycobacterium

bovis antigens using enzyme-linked immunosorbent assay and serum

protein electrophoresis. Mycoses 41, 303-308.

63. Geissmann, F., Manz, M.G., Jung, S., Sieweke, M.H., Merad, M., Ley, K.,

2010. Development of monocytes, macrophages, and dendritic cells.

Science 327, 656-661.

64. Ghahri, H., Talebi, A., Chamani, M., Lotfollahian, H., Afzali, N., 2009.

Ameliorative effect of esterified glucomannan, sodium bentonite, and

humic acid on humoral immunity of broilers during chronic aflatoxicosis.

Turkish Journal of Veterinary and Animal Sciences 33, 419-425.

65. Giambrone, J.J., Diener, U.L., Davis, N.D., Panangala, V.S., Hoerr, F.J.,

1985. Effects of aflatoxin on young turkeys and broiler chickens. Poult. Sci

64, 1678-1684.

120

66. Girish, C., Devegowda, G., 2006. Efficacy of glucomannan-containing

yeast product (Mycosorb®) and hydrated sodium calcium aluminosilicate

in preventing the individual and combined toxicity of aflatoxin and T-2 toxin

in commercial broilers. ASIAN AUSTRALASIAN JOURNAL OF ANIMAL

SCIENCES 19, 877.

67. Gonçalves, B.L., Rosim, R.E., de Oliveira, C.A.F., Corassin, C.H., 2015.

The in vitro ability of different Saccharomyces cerevisiae – Based products

to bind aflatoxin B1. Food Control 47, 298-300.

68. Gong, Y.Y., Cardwell, K., Hounsa, A., Egal, S., Turner, P.C., Hall, A.J.,

Wild, C.P., 2002. Dietary aflatoxin exposure and impaired growth in young

children from Benin and Togo: cross sectional study. Bmj 325, 20-21.

69. Gratz, S., Taubel, M., Juvonen, R.O., Viluksela, M., Turner, P.C.,

Mykkanen, H., El-Nezami, H., 2006. Lactobacillus rhamnosus strain GG

modulates intestinal absorption, fecal excretion, and toxicity of aflatoxin B-

1 in rats. Applied and environmental microbiology 72, 7398-7400.

70. Gratz, S., Wu, Q.K., El-Nezami, H., Juvonen, R.O., Mykkanen, H., Turner,

P.C., 2007. Lactobacillus rhamnosus strain GG reduces aflatoxin B-1

transport, metabolism, and toxicity in caco-2 cells. Applied and

environmental microbiology 73, 3958-3964.

71. Gross-Steinmeyer, K., Eaton, D.L., 2012. Dietary modulation of the

biotransformation and genotoxicity of aflatoxin B1. Toxicology 299, 69-79.

72. Gupta, P., Andrew, H., Kirschner, B.S., Guandalini, S., 2000. Is

Lactobacillus GG helpful in children with Crohn's disease? Results of a

preliminary, open-label study. Journal of pediatric gastroenterology and

nutrition 31, 453-457.

73. Gupta, V., Garg, R., 2009. Probiotics. Indian J Med Microbiol 27, 202-209.

121

74. Hartley, R., Nesbitt, B.F., O'kelly, J., 1963. Toxic metabolites of Aspergillus

flavus. Nature 198, 1056-1058.

75. Hasan, G., Toloei, T., Habibi, M., 2010. Efficacy of esterified

glucomannan, sodium bentonite and humic acid to counteract

experimental aflatoxicosis on antibody titers against Newcastle disease in

broilers. African Journal of Biotechnology 9, 4127-4131.

76. Haskard, C.A., El-Nezami, H.S., Kankaanpaa, P.E., Salminen, S., Ahokas,

J.T., 2001. Surface binding of aflatoxin B-1 by lactic acid bacteria. Applied

and environmental microbiology 67, 3086-3091.

77. Hathout, A.S., Aly, S.E., 2014. Biological detoxification of mycotoxins: a

review. Annals of Microbiology 64, 905-919.

78. Hathout, A.S., Mohamed, S.R., El-Nekeety, A.A., Hassan, N.S., Aly, S.E.,

Abdel-Wahhab, M.A., 2011. Ability of Lactobacillus casei and Lactobacillus

reuteri to protect against oxidative stress in rats fed aflatoxins-

contaminated diet. Toxicon 58, 179-186.

79. He, J., Zhang, K.Y., Chen, D.W., Ding, X.M., Feng, G.D., Ao, X., 2013a.

Effects of maize naturally contaminated with aflatoxin B1 on growth

performance, blood profiles and hepatic histopathology in ducks. Livestock

Science 152, 192-199.

80. He, J., Zhang, K.Y., Chen, D.W., Ding, X.M., Feng, G.D., Ao, X., 2013b.

Effects of vitamin E and selenium yeast on growth performance and

immune function in ducks fed maize naturally contaminated with aflatoxin

B1. Livestock Science 152, 200-207.

81. Hell, K., Cardwell, K.F., Setamou, M., Poehling, H., 2000. The influence of

storage practices on aflatoxin contamination in maize in four

122

agroecological zones of Benin, west Africa. J Stored Prod Res 36, 365-

382.

82. Hell, K., Mutegi, C., Fandohan, P., 2010. Aflatoxin control and prevention

strategies in maize for Sub-Saharan Africa. Julius-Kühn-Archiv, S. 534.

83. Hendrickse, R.G., 1997. Of sick turkeys, kwashiorkor, malaria, perinatal

mortality, heroin addicts and food poisoning: research on the influence of

aflatoxins on child health in the tropics. Ann Trop Med Parasitol 91, 787-

793.

84. Hernandez-Mendoza, A., Garcia, H., Steele, J., 2009a. Screening of< i>

Lactobacillus casei</i> strains for their ability to bind aflatoxin B< sub>

1</sub>. Food and chemical toxicology 47, 1064-1068.

85. Hernandez-Mendoza, A., Guzman-de-Peña, D., Garcia, H.S., 2009b. Key

role of teichoic acids on aflatoxin B1 binding by probiotic bacteria. Journal

of Applied Microbiology 107, 395-403.

86. Hernandez-Mendoza, A., Guzman-De-Peña, D., González-Córdova, A.F.,

Vallejo-Córdoba, B., Garcia, H.S., 2010. In vivo assessment of the

potential protective effect of Lactobacillus casei Shirota against aflatoxin

B1. Dairy Sci. Technol. 90, 729-740.

87. Hernandez-Mendoza, A., Rivas-Jimenez, L., Garcia, H.S., 2011.

Assessment of Aflatoxin B1 Binding to Lactobacillus reuteri by Microscopy

and Fluorescence Techniques. Food Biotechnology 25, 140-150.

88. Hitchins, A.D., McDonough, F.E., 1989. Prophylactic and therapeutic

aspects of fermented milk. Am J Clin Nutr 49, 675-684.

89. Hoyos, A.B., 1999. Reduced incidence of necrotizing enterocolitis

associated with enteral administration of< i> Lactobacillus acidophilus</i>

123

and< i> Bifidobacterium infantis</i> to neonates in an intensive care unit.

International journal of infectious diseases 3, 197-202.

90. Huebner, H.J., Herrera, P., Phillips, T.D., 2008. Clay-Based Interventions

for the Control of Chemical and Microbial Hazards in Food and Water,

Preharvest and Postharvest Food Safety, Blackwell Publishing, pp. 389-

402.

91. Huff, W., Kubena, L., Harvey, R., Corrier, D., Mollenhauer, H., 1986.

Progression of aflatoxicosis in broiler chickens. Poultry Science 65, 1891-

1899.

92. IARC, 2002. Some traditional herbal medicines, some mycotoxins,

naphthalene and styrene, IARC Monographs on the Evaluation of

Carcinogenic Risk of Chemicals to Man, World Health Organization, pp. 1-

556.

93. Indresh, H., Devegowda, G., Ruban, S.W., Shivakumar, M., 2013. Effects

of high grade bentonite on performance, organ weights and serum

biochemistry during aflatoxicosis in broilers. Vet World 6, 313-317.

94. Jard, G., Liboz, T., Mathieu, F., Guyonvarc'h, A., Lebrihi, A., 2011. Review

of mycotoxin reduction in food and feed: from prevention in the field to

detoxification by adsorption or transformation. Food Addit Contam Part A

Chem Anal Control Expo Risk Assess 28, 1590-1609.

95. JECFA, 2001. Joint FAO/WHO Expert Committee on Food Additives.

Safety

96. evaluation of certain food additives and contaminants. World Health

Organization.

124

97. Jespersen, L., 2003. Occurrence and taxonomic characteristics of strains

of Saccharomyces cerevisiae predominant in African indigenous

fermented foods and beverages. FEMS Yeast Research 3, 191-200.

98. Jin, L.Z., Marquardt, R.R., Zhao, X., 2000. A strain of Enterococcus

faecium (18C23) inhibits adhesion of enterotoxigenic Escherichia coli K88

to porcine small intestine mucus. Appl Environ Microbiol 66, 4200-4204.

99. Kabak, B., Dobson, A.D.W., Var, I., 2006. Strategies to Prevent Mycotoxin

Contamination of Food and Animal Feed: A Review. Critical Reviews in

Food Science and Nutrition 46, 593-619.

100. Kalorey, D., Kurkure, N., Ramgaonkar, J., Sakhare, P., Warke, S.,

Nigot, N., 2005. Effect of polyherbal feed supplement “Growell “during

induced aflatoxicosis, ochratoxicosis and combined mycotoxicoses in

broilers. Asian-Australasian J. Anim. Sci 18, 375-383.

101. Kamali, M., Mostafaei, A., 2014. Hepatoprotective effect of Silybum

marianum on experimental hepatotoxicity in broilers. Comparative Clinical

Pathology 23, 967-973.

102. Kamdem, L.K., Meineke, I., Godtel-Armbrust, U., Brockmoller, J.,

Wojnowski, L., 2006. Dominant contribution of P450 3A4 to the hepatic

carcinogenic activation of aflatoxin B-1. Chemical Research in Toxicology

19, 577-586.

103. Kamel, E.S., 2013. Histological study on the effect of aflatoxin B1

on the renal tubules of adult rats. Egyptian Journal of Histology 36, 246-

252.

104. Karabacak, M., Eraslan, G., Kanbur, M., Sarıca, Z.S., Effects of

Tarantula cubensis D6 on aflatoxin-induced injury in biochemical

parameters in rats. Homeopathy.

125

105. Karaman, M., Basmacioglu, H., Ortatatli, M., Oguz, H., 2005.

Evaluation of the detoxifying effect of yeast glucomannan on aflatoxicosis

in broilers as assessed by gross examination and histopathology. Br Poult

Sci 46, 394-400.

106. Karimi Torshizi, M.A., Moghaddam, A.R., Rahimi, S., Mojgani, N.,

2010. Assessing the effect of administering probiotics in water or as a feed

supplement on broiler performance and immune response. British Poultry

Science 51, 178-184.

107. Kelly, J.D., Eaton, D.L., Guengerich, F.P., Coulombe, R.A., Jr.,

1997. Aflatoxin B1 activation in human lung. Toxicol Appl Pharmacol 144,

88-95.

108. Kew, M.C., 2011. Hepatitis B virus x protein in the pathogenesis of

hepatitis B virus-induced hepatocellular carcinoma. Journal of

Gastroenterology and Hepatology 26, 144-152.

109. Khan, S.H., Shamsul, H., Rozina, S., Muhammad, A., 2011.

Occurrence of aflatoxin B1 in poultry feed and feed ingredients in

Pakistan. International Journal for Agro Veterinary and Medical Sciences

5, 30-42.

110. Khatoon, A., Zargham Khan, M., Khan, A., Saleemi, M.K., Javed, I.,

2013. Amelioration of Ochratoxin A-induced immunotoxic effects by

silymarin and vitamin E in White Leghorn cockerels. Journal of

Immunotoxicology 10, 25-31.

111. Kirk, G.D., Lesi, O.A., Mendy, M., Szymanska, K., Whittle, H.,

Goedert, J.J., Hainaut, P., Montesano, R., 2005. 249(ser) TP53 mutation

in plasma DNA, hepatitis B viral infection, and risk of hepatocellular

carcinoma. Oncogene 24, 5858-5867.

126

112. Kochan, P., Chmielarczyk, A., Szymaniak, L., Brykczynski, M.,

Galant, K., Zych, A., Pakosz, K., Giedrys-Kalemba, S., Lenouvel, E.,

Heczko, P.B., 2011. Lactobacillus rhamnosus administration causes

sepsis in a cardiosurgical patient—is the time right to revise probiotic

safety guidelines? Clinical Microbiology and Infection 17, 1589-1592.

113. Krishnamachari, K.A., Bhat, R.V., Nagarajan, V., Tilak, T.B., 1975a.

Hepatitis due to aflatoxicosis. An outbreak in Western India. Lancet 1,

1061-1063.

114. Krishnamachari, K.A.V.R., Nagarajan, V., Bhat, R., Tilak, T.B.G.,

1975b. HEPATITIS DUE TO AFLATOXICOSIS: An Outbreak in Western

India. The Lancet 305, 1061-1063.

115. Kshirsagar, A., Ingawale, D., Vyawahare, N., Ashok, P., 2009.

Silymarin: A Comprehensive Review.

116. Kumar, D.S., Rao, S., Satyanaryana, M., Kumar, P.P., Anitha, N.,

2013. Amelioration of hepatotoxicity induced by aflatoxin using citrus fruit

oil in broilers (Gallus domesticus). Toxicology and Industrial Health.

117. Kumar, M., Verma, V., Nagpal, R., Kumar, A., Behare, P., Singh, B.,

Aggarwal, P., 2012a. Anticarcinogenic effect of probiotic fermented milk

and chlorophyllin on aflatoxin-B1-induced liver carcinogenesis in rats.

British Journal of Nutrition 107, 1006-1016.

118. Kumar, M., Verma, V., Nagpal, R., Kumar, A., Behare, P.V., Singh,

B., Aggarwal, P.K., 2012b. Anticarcinogenic effect of probiotic fermented

milk and chlorophyllin on aflatoxin-B1-induced liver carcinogenesis in rats.

British Journal of Nutrition 107, 1006-1016.

119. Kumar, M., Verma, V., Nagpal, R., Kumar, A., Gautam, S.K.,

Behare, P.V., Grover, C.R., Aggarwal, P.K., 2011. Effect of probiotic

127

fermented milk and chlorophyllin on gene expressions and genotoxicity

during AFB(1)-induced hepatocellular carcinoma. Gene 490, 54-59.

120. Lee, S.H., Lillehoj, H.S., Hong, Y.H., Jang, S.I., Lillehoj, E.P.,

Ionescu, C., Mazuranok, L., Bravo, D., 2010. In vitro effects of plant and

mushroom extracts on immunological function of chicken lymphocytes and

macrophages. British Poultry Science 51, 213-221.

121. Lesbros-Pantoflickova, D., Corthesy-Theulaz, I., Blum, A.L., 2007.

Helicobacter pylori and probiotics. J Nutr 137, 812S-818S.

122. Lewis, S.J., Freedman, A.R., 1998. Review article: the use of

biotherapeutic agents in the prevention and treatment of gastrointestinal

disease. Alimentary Pharmacology & Therapeutics 12, 807-822.

123. Li, Y., Liu, Y.H., Yang, Z.B., Wan, X.L., Chi, F., 2012. The efficacy

of clay enterosorbent to ameliorate the toxicity of aflatoxin B1 from

contaminated corn (Zea mays) on hematology, serum biochemistry, and

oxidative stress in ducklings. The Journal of Applied Poultry Research 21,

806-815.

124. Lilly, D.M., Stillwell, R.H., 1965. Probiotics: Growth-Promoting

Factors Produced by Microorganisms. Science 147, 747-748.

125. Lin, H.-C., Hsu, C.-H., Chen, H.-L., Chung, M.-Y., Hsu, J.-F., Lien,

R.-i., Tsao, L.-Y., Chen, C.-H., Su, B.-H., 2008. Oral probiotics prevent

necrotizing enterocolitis in very low birth weight preterm infants: a

multicenter, randomized, controlled trial. Pediatrics 122, 693-700.

126. Liu, B.H., Yu, F.Y., Chan, M.H., Yang, Y.L., 2002. The effects of

mycotoxins, fumonisin B1 and aflatoxin B1, on primary swine alveolar

macrophages. Toxicol Appl Pharmacol 180, 197-204.

128

127. Liu, Y., Wu, F., 2010. Global burden of aflatoxin-induced

hepatocellular carcinoma: a risk assessment. Environ Health Perspect

118, 818-824.

128. Luongo, D., Russo, R., Balestrieri, A., Marzocco, S., Bergamo, P.,

Severino, L., 2013. In vitro study of AFB and AFM effects on human

lymphoblastoid Jurkat T-cell model. J Immunotoxicol 22, 22.

129. Luper, S., 1998. A review of plants used in the treatment of liver

disease: part 1. Alternative medicine review: a journal of clinical

therapeutic 3, 410.

130. Lutsenko, S., Kashnikova, T., Khmyrov, A., Ledeshkova, O.,

Fel’dman, N., Luzhnov, N., Lutsenko, E., 2008. Study of the effect of a

liposomal form of silymarin on biochemical indices of the blood serum and

productivity of broiler chicks. Russian Agricultural Sciences 34, 415-417.

131. Magnoli, A.P., Monge, M.P., Miazzo, R.D., Cavaglieri, L.R.,

Magnoli, C.E., Merkis, C.I., Cristofolini, A.L., Dalcero, A.M., Chiacchiera,

S.M., 2011. Effect of low levels of aflatoxin B1 on performance,

biochemical parameters, and aflatoxin B1 in broiler liver tissues in the

presence of monensin and sodium bentonite. Poultry Science 90, 48-58.

132. Makarova, K., Slesarev, A., Wolf, Y., Sorokin, A., Mirkin, B., Koonin,

E., Pavlov, A., Pavlova, N., Karamychev, V., Polouchine, N., Shakhova,

V., Grigoriev, I., Lou, Y., Rohksar, D., Lucas, S., Huang, K., Goodstein,

D.M., Hawkins, T., Plengvidhya, V., Welker, D., Hughes, J., Goh, Y.,

Benson, A., Baldwin, K., Lee, J.H., Díaz-Muñiz, I., Dosti, B., Smeianov, V.,

Wechter, W., Barabote, R., Lorca, G., Altermann, E., Barrangou, R.,

Ganesan, B., Xie, Y., Rawsthorne, H., Tamir, D., Parker, C., Breidt, F.,

Broadbent, J., Hutkins, R., O'Sullivan, D., Steele, J., Unlu, G., Saier, M.,

129

Klaenhammer, T., Richardson, P., Kozyavkin, S., Weimer, B., Mills, D.,

2006. Comparative genomics of the lactic acid bacteria. Proceedings of

the National Academy of Sciences of the United States of America 103,

15611-15616.

133. Manafi, M., Umakantha, B., Mohan, K., Swamy, H.N., 2012.

Synergistic effects of two commonly contaminating mycotoxins (aflatoxin

and T-2 toxin) on biochemical parameters and immune status of broiler

chickens. World Appl. Sci. J. 17, 364-367.

134. Marin, D.E., Taranu, I., Bunaciu, R.P., Pascale, F., Tudor, D.S.,

Avram, N., Sarca, M., Cureu, I., Criste, R.D., Suta, V., Oswald, I.P., 2002.

Changes in performance, blood parameters, humoral and cellular immune

responses in weanling piglets exposed to low doses of aflatoxin. Journal of

Animal Science 80, 1250-1257.

135. Marroquín-Cardona, A., Deng, Y., Garcia-Mazcorro, J.F., Johnson,

N.M., Mitchell, N.J., Tang, L., Robinson, A., Taylor, J.F., Wang, J.S.,

Phillips, T.D., 2011. Characterization and safety of uniform particle size

NovaSil clay as a potential aflatoxin enterosorbent. Applied Clay Science

54, 248-257.

136. Matur, E., Ergul, E., Akyazi, I., Eraslan, E., Inal, G., Bilgic, S.,

Demircan, H., 2011. Effects of Saccharomyces cerevisiae extract on

haematological parameters, immune function and the antioxidant defence

system in breeder hens fed aflatoxin contaminated diets. British Poultry

Science 52, 541-550.

137. Mechoud, M.A., Juarez, G.E., de Valdez, G.F., Rodriguez, A.V.,

2012. Lactobacillus reuteri CRL 1098 and Lactobacillus acidophilus CRL

130

1014 differently reduce in vitro immunotoxic effect induced by Ochratoxin

A. Food and Chemical Toxicology 50, 4310-4315.

138. Mehrzad, J., Devriendt, B., Baert, K., Cox, E., 2014. Aflatoxin B1

interferes with the antigen-presenting capacity of porcine.

139. Meissonnier, G.M., Pinton, P., Laffitte, J., Cossalter, A.M., Gong,

Y.Y., Wild, C.P., Bertin, G., Galtier, P., Oswald, I.P., 2008a.

Immunotoxicity of aflatoxin B1: impairment of the cell-mediated response

to vaccine antigen and modulation of cytokine expression. Toxicol. Appl.

Pharmacol 231, 142-149.

140. Meissonnier, G.M., Pinton, P., Laffitte, J., Cossalter, A.M., Gong,

Y.Y., Wild, C.P., Bertin, G., Galtier, P., Oswald, I.P., 2008b.

Immunotoxicity of aflatoxin B1: impairment of the cell-mediated response

to vaccine antigen and modulation of cytokine expression. Toxicol Appl

Pharmacol 231, 142-149.

141. Menkir, A., Brown, R., Bandyopadhyay, R., Chen, Z.-y., Cleveland,

T., 2006. A USA–Africa collaborative strategy for identifying,

characterizing, and developing maize germplasm with resistance to

aflatoxin contamination. Mycopathologia 162, 225-232.

142. Merad, M., Sathe, P., Helft, J., Miller, J., Mortha, A., 2013. The

dendritic cell lineage: ontogeny and function of dendritic cells and their

subsets in the steady state and the inflamed setting. Annual review of

immunology 31.

143. Miazzo, R., Rosa, C.A.R., De Queiroz Carvalho, E.C., Magnoli, C.,

Chiacchiera, S.M., Palacio, G., Saenz, M., Kikot, A., Basaldella, E.,

Dalcero, A., 2000. Efficacy of synthetic zeolite to reduce the toxicity of

aflatoxin in broiler chicks. Poultry Science 79, 1-6.

131

144. Micco, C., Miraglia, M., Onori, R., Brera, C., Mantovani, A., Ioppolo,

A., Stasolla, D., 1988. Long-term administration of low doses of

mycotoxins to poultry. 1. Residues of aflatoxin B1 and its metabolites in

broilers and laying hens. Food Addit Contam 5, 303-308.

145. Mitchell, N.J., Kumi, J., Aleser, M., Elmore, S.E., Rychlik, K.A.,

Zychowski, K.E., Romoser, A.A., Phillips, T.D., Ankrah, N.-A., 2014. Short-

Term Safety and Efficacy of Calcium Montmorillonite Clay (UPSN) in

Children. The American Journal of Tropical Medicine and Hygiene 91,

777-785.

146. Mobeen, A., 2011. Aflatoxins B1 and B2Contamination of Peanut

and Peanut Products and Subsequent Microwave Detoxification. Journal

of Pharmacy and Nutrition Sciences 1.

147. Mogadam, N., Azizpour, A., 2013. Ameliorative effect of

glucomannan-containing yeast product (Mycosorb) and sodium bentonite

on performance and antibody titers against Newcastle disease in broilers

during chronic aflatoxicosis. African Journal of Biotechnology 10, 17372-

17378.

148. Mohamadi Sani, A., Gholampour Azizi, E., Ataye Salehi, E., Rahimi,

K., 2012. Reduction of aflatoxin in rice by different cooking methods.

Toxicology and Industrial Health.

149. Morazzoni, P., Montalbetti, A., Malandrino, S., Pifferi, G., 1993.

Comparative pharmacokinetics of silipide and silymarin in rats. European

Journal of Drug Metabolism and Pharmacokinetics 18, 289-297.

150. Muhammad, D., Chand, N., Khan, S., Sultan, A., Mushtaq, M.,

2012. Hepatoprotective role of milk thistle (Silybum marianum) in meat

132

type chicken fed aflatoxin B1 contaminated feed. Pakistan Veterinary

Journal 32, 443-446.

151. Mulder, J.E., Bondy, G.S., Mehta, R., Massey, T.E., 2014. Up-

regulation of nucleotide excision repair in mouse lung and liver following

chronic exposure to aflatoxin B1 and its dependence on p53 genotype.

Toxicology and Applied Pharmacology 275, 96-103.

152. Muriel, P., Mourelle, M., 1990. Prevention by silymarin of

membrane alterations in acute CCl4 liver damage. J Appl Toxicol 10, 275-

279.

153. Mushtaq, M., Sultana, B., Anwar, F., Khan, M.Z., Ashrafuzzaman,

M., 2012. Occurrence of aflatoxins in selected processed foods from

Pakistan. International journal of molecular sciences 13, 8324-8337.

154. Nabavi, S., Rafraf, M., Somi, M.-h., Homayouni-Rad, A., Asghari-

Jafarabadi, M., 2015. Probiotic yogurt improves body mass index and

fasting insulin levels without affecting serum leptin and adiponectin levels

in non-alcoholic fatty liver disease (NAFLD). Journal of Functional Foods

18, Part A, 684-691.

155. Neeff, D.V., Ledoux, D.R., Rottinghaus, G.E., Bermudez, A.J.,

Dakovic, A., Murarolli, R.A., Oliveira, C.A.F., 2013. In vitro and in vivo

efficacy of a hydrated sodium calcium aluminosilicate to bind and reduce

aflatoxin residues in tissues of broiler chicks fed aflatoxin B1. Poultry

Science 92, 131-137.

156. Neish, A.S., 2009. Microbes in gastrointestinal health and disease.

Gastroenterology 136, 65-80.

133

157. Nesbitt, B.F., O'Kelly, J., Sargeant, K., Sheridan, A., 1962.

Aspergillus flavus and turkey X disease. Toxic metabolites of Aspergillus

flavus. Nature 195, 1062-1063.

158. Ngindu, A., Johnson, B.K., Kenya, P.R., Ngira, J.A., Ocheng, D.M.,

Nandwa, H., Omondi, T.N., Jansen, A.J., Ngare, W., Kaviti, J.N., Gatei, D.,

Siongok, T.A., 1982. Outbreak of acute hepatitis caused by aflatoxin

poisoning in Kenya. Lancet 1, 1346-1348.

159. Nikbakht Nasrabadi, E., Jamaluddin, R., Abdul Mutalib, M.S.,

Khaza'ai, H., Khalesi, S., Mohd Redzwan, S., 2013. Reduction of aflatoxin

level in aflatoxin-induced rats by the activity of probiotic Lactobacillus casei

strain Shirota. Journal of Applied Microbiology 114, 1507-1515.

160. Obuseh, F.A., Jolly, P.E., Kulczycki, A., Ehiri, J., Waterbor, J.,

Desmond, R.A., Preko, P.O., Jiang, Y., Piyathilake, C.J., 2011. Aflatoxin

levels, plasma vitamins A and E concentrations, and their association with

HIV and hepatitis B virus infections in Ghanaians: a cross-sectional study.

J Int AIDS Soc 14, 1758-2652.

161. Pandey, G., 2014. SILYMARIN, A HERBAL DRUG AGAINST

MULTIPLE DISORDERS. INTERNATIONAL JOURNAL OF ANIMAL,

VETERINARY, FISHERY AND ALLIED SCIENCES ISSN-2394-4498 1.

162. Peltonen, K., El-Nezami, H., Haskard, C., Ahokas, J., Salminen, S.,

2001b. Aflatoxin B-1 binding by dairy strains of lactic acid bacteria and

bifidobacteria. Journal of dairy science 84, 2152-2156.

163. Petkov, R., Tacheva, T., Borisova, L., 1985. [Paraclinical research

on broiler chickens treated with aflatoxin B1 and microbiological studies of

the meat and liver obtained from them]. Vet Med Nauki 22, 58-64.

134

164. Phillips, T., Afriyie-Gyawu, E., Williams, J., Huebner, H., Ankrah, N.-

A., Ofori-Adjei, D., Jolly, P., Johnson, N., Taylor, J., Marroquin-Cardona,

A., 2008a. Reducing human exposure to aflatoxin through the use of clay:

a review. Food Additives and Contaminants 25, 134-145.

165. Phillips, T.D., Afriyie-Gyawu, E., Williams, J., Huebner, H., Ankrah,

N.A., Ofori-Adjei, D., Jolly, P., Johnson, N., Taylor, J., Marroquin-Cardona,

A., Xu, L., Tang, L., Wang, J.S., 2008b. Reducing human exposure to

aflatoxin through the use of clay: A review. Food Additives &

Contaminants: Part A 25, 134-145.

166. Phillips, T.D., Lemke, S.L., Grant, P.G., 2002a. Characterization of

clay-based enterosorbents for the prevention of aflatoxicosis. Adv Exp

Med Biol 504, 157-171.

167. Phillips, T.D., Lemke, S.L., Grant, P.G., 2002b. Characterization of

clay-based enterosorbents for the prevention of aflatoxicosis. Advances in

experimental medicine and biology 504, 157-171.

168. Pizzolitto, R.P., Armando, M.R., Salvano, M.A., Dalcero, A.M.,

Rosa, C.A., 2013. Evaluation of Saccharomyces cerevisiae as an

antiaflatoxicogenic agent in broiler feedstuffs. Poultry Science 92, 1655-

1663.

169. Poorbaghi, S.L., Dadras, H., Gheisari, H.R., Mosleh, N., Firouzi, S.,

Roohallazadeh, H., 2014. Effects of Lactobacillus acidophilus and inulin on

faecal viral shedding and immunization against H9N2Avian influenza virus.

Journal of Applied Microbiology 116, 667-676.

170. Prandini, A., Tansini, G., Sigolo, S., Filippi, L., Laporta, M., Piva, G.,

2009. On the occurrence of aflatoxin M< sub> 1</sub> in milk and dairy

products. Food and chemical toxicology 47, 984-991.

135

171. Probst, C., Bandyopadhyay, R., Price, L.E., Cotty, P.J., 2010.

Identification of Atoxigenic Aspergillus flavus Isolates to Reduce Aflatoxin

Contamination of Maize in Kenya. Plant Disease 95, 212-218.

172. Probst, C., Njapau, H., Cotty, P.J., 2007. Outbreak of an acute

aflatoxicosis in Kenya in 2004: Identification of the causal agent. Applied

and environmental microbiology 73, 2762-2764.

173. Qazi, J.I., Fayyaz, Z., 2006. Aflatoxin contaminated foods and

health risk perspective for Pakistani population. Mycopathol 4, 27-34.

174. Quigley, E.M.M., 2010. Prebiotics and probiotics; modifying and

mining the microbiota. Pharmacological Research 61, 213-218.

175. Qureshi, H., Zuberi, S.J., Jafarey, N.A., Zaidi, S.H., 1990.

Hepatocellular carcinoma in Karachi. J Gastroenterol Hepatol 5, 1-6.

176. Raju, M., Devegowda, G., 2000a. Influence of esterified-

glucomannan on performance and organ morphology, serum biochemistry

and haematology in broilers exposed to individual and combined

mycotoxicosis (aflatoxin, ochratoxin and T-2 toxin). British poultry science

41, 640-650.

177. Raju, M.V.L.N., Devegowda, G., 2000b. Influence of esterified-

glucomannan on performance and organ morphology, serum biochemistry

and haematology in broilers exposed to individual and combined

mycotoxicosis (aflatoxin, ochratoxin and T-2 toxin). British Poultry Science

41, 640-650.

178. Rao, S.B.N., Chopra, R.C., 2001. Influence of sodium bentonite and

activated charcoal on aflatoxin M1 excretion in milk of goats. Small

Ruminant Research 41, 203-213.

136

179. Rashid, N., Bajwa, M., Rafeeq, M., Khan, M., Ahmad, Z., Tariq, M.,

Wadood, A., Abbas, F., 2012. Prevalence of aflatoxin B1 in finished

commercial broiler feed from west central Pakistan. J Anim Plant Sci 22, 6-

10.

180. Rashid, N., Bajwa, M.A., Rafeeq, M., Tariq, M.M., Abbas, F., Awan,

M.A., Khan, M.A., Shahzad, I., Rehman, A., Ahmad, Z., 2013. Prevalence

of aflatoxicosis in broiler chickens in Quetta, Pakistan. Pakistan Journal of

Zoology 45.

181. Rastogi, R., Srivastava, A.K., Rastogi, A.K., 2001. Biochemical

changes induced in liver and serum of aflatoxin B1-treated male wistar

rats: preventive effect of picroliv. Pharmacol Toxicol 88, 53-58.

182. Rastogi, R., Srivastava, A.K., Srivastava, M., Rastogi, A.K., 2000.

Hepatocurative Effect of Picroliv and Silymarin Against Aflatoxin B~ 1

Induced Hepatotoxicity in Rats. Planta medica 66, 709-713.

183. Rawal, S., Bauer, M.M., Mendoza, K.M., El-Nezami, H., Hall, J.R.,

Kim, J.E., Stevens, J.R., Reed, K.M., Coulombe Jr, R.A., 2014.

Aflatoxicosis chemoprevention by probiotic Lactobacillius and lack of effect

on the major histocompatibility complex. Research in Veterinary Science

97, 274-281.

184. Rawal, S., Kim, J.E., Coulombe, R., 2010. Aflatoxin B 1 in poultry:

toxicology, metabolism and prevention. Research in veterinary science 89,

325-331.

185. Regulation, E.C.E.C., 2006. European Commission (EC)

Commission Regulation (EC) No 1881/2006 of 19 December 2006 setting

maximum levels for certain contaminants in foodstuffs. Off. J. Eur. Union L

364, 5-24.

137

186. Rizzello, V., Bonaccorsi, I., Dongarra, M.L., Fink, L.N., Ferlazzo, G.,

2011. Role of natural killer and dendritic cell crosstalk in

immunomodulation by commensal bacteria probiotics. BioMed Research

International 2011.

187. Rossi, G.A., Zocchi, E., Sacco, O., Balbi, B., Ravazzoni, C.,

Damiani, G., 1986. Alveolar macrophage stimulation of T-cell proliferation

in autologous mixed lymphocyte reactions. Role of HLA-DR antigens. Am

Rev Respir Dis 133, 78-82.

188. Saad-Hussein, A., Abdalla, M.S., Shousha, W.G., Moubarz, G.,

Mohamed, A.H., 2014. Oxidative Role of Aflatoxin B1 on the Liver of

Wheat Milling Workers. Open Access Macedonian Journal of Medical

Sciences [previously Macedonian Journal of Medical Sciences] 2, 6.

189. Salim, H.M., Kang, H.K., Akter, N., Kim, D.W., Kim, J.H., Kim, M.J.,

Na, J.C., Jong, H.B., Choi, H.C., Suh, O.S., Kim, W.K., 2013.

Supplementation of direct-fed microbials as an alternative to antibiotic on

growth performance, immune response, cecal microbial population, and

ileal morphology of broiler chickens. Poultry Science 92, 2084-2090.

190. Saller, R., Meier, R., Brignoli, R., 2001. The Use of Silymarin in the

Treatment of Liver Diseases. Drugs 61, 2035-2063.

191. Salminen, S., Nybom, S., Meriluoto, J., Collado, M.C., Vesterlund,

S., El-Nezami, H., 2010b. Interaction of probiotics and pathogens--benefits

to human health? Curr. Opin. Biotechnol 21, 157-167.

192. Samuel, A.O., Olubukola, O., Matthew, A.O., 2009. Hematological

and immunological effect on chicken exposed to aflatoxin. Vet. World 2, 5-

7.

193. Schoental, R., 1967. Aflatoxins. Annu Rev Pharmacol 7, 343-356.

138

194. Selim, K., El-hofy, H., Khalil, R., 2014. The efficacy of three

mycotoxin adsorbents to alleviate aflatoxin B1-induced toxicity in

Oreochromis niloticus. Aquaculture International 22, 523-540.

195. Shamsudeen, P., Shrivastava, H., 2013. Interaction of zinc with

aflatoxin with reference to serum biochemical profile of broiler chicken.

Indian Journal of Field Veterinarians (The) 9, 39-42.

196. Shantha, T., 1999. Fungal degradation of aflatoxin B1. Natural

toxins 7, 175-178.

197. Shen, H., Lv, P., Xing, X., Xing, L., Yan, X., Wang, J., Zhang, X.,

2012. Impairment of alveolar type-II cells involved in the toxicity of

Aflatoxin G1 in rat lung. Food and chemical toxicology 50, 3222-3228.

198. Shetty, P.H., Hald, B., Jespersen, L., 2007. Surface binding of

aflatoxin B1 by Saccharomyces cerevisiae strains with potential

decontaminating abilities in indigenous fermented foods. International

Journal of Food Microbiology 113, 41-46.

199. Shotwell, O.L., Hesseltine, C., Stubblefield, R., Sorenson, W., 1966.

Production of aflatoxin on rice. Applied Microbiology 14, 425-428.

200. Smela, M.E., Hamm, M.L., Henderson, P.T., Harris, C.M., Harris,

T.M., Essigmann, J.M., 2002. The aflatoxin B(1) formamidopyrimidine

adduct plays a major role in causing the types of mutations observed in

human hepatocellular carcinoma. Proc Natl Acad Sci U S A 99, 6655-

6660.

201. Smith, E.E., Kubena, L.F., Braithwaite, C.E., Harvey, R.B., Phillips,

T.D., Reine, A.H., 1992. Toxicological evaluation of aflatoxin and

cyclopiazonic acid in broiler chickens. Poult Sci 71, 1136-1144.

139

202. Smith, J.E., Henderson, R.S., 1991. Mycotoxins And Animal Foods.

CRC Press, Boca Raton, FL.

203. Sohooa, R., Khana, A.U., Ameena, K., Rafia-Munire, A., Saleemb,

F., 2015. Outbreak of aflatoxicosis on a local cattle farm in Pakistan.

Veterinaria 3, 13-17.

204. Sporn, M.B., Suh, N., 2002. Chemoprevention: an essential

approach to controlling cancer. Nat Rev Cancer 2, 537-543.

205. Suchý, P., Straková, E., Kummer, V., Herzig, I., Písaříková, V.,

Blechová, R., Mašková, J., 2008. Hepatoprotective effects of milk thistle

(Silybum marianum) seed cakes during the chicken broiler fattening. Acta

Veterinaria Brno 77, 31-38.

206. Sur, E., Celİk, İ., 2003. Effects of aflatoxin B1 on the development

of the bursa of Fabricius and blood lymphocyte acid phosphatase of the

chicken. British Poultry Science 44, 558-566.

207. Tandon, H.D., Tandon, B.N., Ramalingaswami, V., 1978. Epidemic

of toxic hepatitis in India of possible mycotoxic origin. Arch Pathol Lab Med

102, 372-376.

208. Tang, L., Xu, L., Afriyie-Gyawu, E., Liu, W., Wang, P., Tang, Y.,

Wang, Z., Huebner, H., Ankrah, N.-A., Ofori-Adjei, D., 2009. Aflatoxin–

albumin adducts and correlation with decreased serum levels of vitamins A

and E in an adult Ghanaian population. Food additives and contaminants

26, 108-118.

209. Tedesco, D., Steidler, S., Galletti, S., Tameni, M., Sonzogni, O.,

Ravarotto, L., 2004a. Efficacy of silymarin-phospholipid complex in

reducing the toxicity of aflatoxin B1 in broiler chicks. Poult Sci 83, 1839-

1843.

140

210. Tedesco, D., Tameni, M., Steidler, S., Galletti, S., Di Pierro, F.,

2003. Effect of silymarin and its phospholipid complex against AFM1

excretion in an organic dairy herd. Milchwissenschaft 58, 416-419.

211. Thongsong, B., Kalandakanond-Thongsong, S., Chavananikul, V.,

Effects of the addition of probiotic containing both bacteria and yeast or an

antibiotic on performance parameters, mortality rate and antibiotic residue

in broilers.

212. Tumolo, T., Lanfer-Marquez, U.M., 2012. Copper chlorophyllin: A

food colorant with bioactive properties? Food Research International 46,

451-459.

213. Valchev, I., Kanakov, D., Hristov, T., Lazarov, L., Binev, R.,

Grozeva, N., Nikolov, Y., 2014. INVESTIGATIONS ON THE LIVER

FUNCTION OF BROILER CHICKENS WITH EXPERIMENTAL

AFLATOXICOSIS. Bulgarian Journal of Veterinary Medicine 17, 302-313.

214. Vekiru, E., Fruhauf, S., Rodrigues, I., Ottner, F., Krska, R.,

Schatzmayr, G., Ledoux, D.R., Rottinghaus, G.E., Bermudez, A.J., In vitro

binding assessment and in vivo efficacy of several adsorbents against

aflatoxin B&lt;sub&gt;1&lt;/sub&gt. World Mycotoxin Journal.

215. Verbrugghe, E., Croubels, S., Vandenbroucke, V., Goossens, J., De

Backer, P., Eeckhout, M., De Saeger, S., Boyen, F., Leyman, B., Van

Parys, A., Haesebrouck, F., Pasmans, F., 2012. A modified glucomannan

mycotoxin-adsorbing agent counteracts the reduced weight gain and

diminishes cecal colonization of Salmonella Typhimurium in T-2 toxin

exposed pigs. Research in Veterinary Science 93, 1139-1141.

216. Verma, J., Johri, T.S., Swain, B.K., Ameena, S., 2004. Effect of

graded levels of aflatoxin, ochratoxin and their combinations on the

141

performance and immune response of broilers. British Poultry Science 45,

512-518.

217. Viegas, S., Veiga, L., Malta-Vacas, J., Sabino, R., Figueredo, P.,

Almeida, A., Viegas, C., Carolino, E., 2012. Occupational exposure to

aflatoxin (AFB(1)) in poultry production. J Toxicol Environ Health A 75,

1330-1340.

218. Wang, G.H., Xue, C.Y., Chen, F., Ma, Y.L., Zhang, X.B., Bi, Y.Z.,

Cao, Y.C., 2009. Effects of combinations of ochratoxin A and T-2 toxin on

immune function of yellow-feathered broiler chickens. Poultry Science 88,

504-510.

219. Wang, P., Afriyie-gyawu, E., Tang, Y., Johnson, N.M., Xu, L., Tang,

L., Huebner, H.J., Ankrah, N.A., Ofori-adjei, D., Ellis, W., Jolly, P.E.,

Williams, J.H., Wang, J.S., Phillips, T.D., 2008. NovaSil clay intervention in

Ghanaians at high risk for aflatoxicosis: II. Reduction in biomarkers of

aflatoxin exposure in blood and urine. Food Additives & Contaminants:

Part A 25, 622-634.

220. Weaver, A.C., See, M.T., Hansen, J.A., Kim, Y.B., De Souza, A.L.,

Middleton, T.F., Kim, S.W., 2013. The use of feed additives to reduce the

effects of aflatoxin and deoxynivalenol on pig growth, organ health and

immune status during chronic exposure. Toxins 5, 1261-1281.

221. Weaver, M.A., Abbas, H.K., Falconer, L.L., Allen, T.W., Pringle,

H.L., Sciumbato, G.L., 2015. Biological control of aflatoxin is effective and

economical in Mississippi field trials. Crop Protection 69, 52-55.

222. Wei, D.-L., Jong, S.-C., 1986. Production of aflatoxins by strains of

the Aspergillus flavus group maintained in ATCC. Mycopathologia 93, 19-

24.

142

223. Wild, C., Turner, P., 2002. The toxicology of aflatoxins as a basis

for public health decisions. Mutagenesis 17, 471-481.

224. Williams, J.H., Phillips, T.D., Jolly, P.E., Stiles, J.K., Jolly, C.M.,

Aggarwal, D., 2004. Human aflatoxicosis in developing countries: a review

of toxicology, exposure, potential health consequences, and interventions.

Am J Clin Nutr 80, 1106-1122.

225. Wolzak, A., Pearson, A.M., Coleman, T.H., Pestka, J.J., Gray, J.I.,

Chen, C., 1986. Aflatoxin carryover and clearance from tissues of laying

hens. Food Chem Toxicol 24, 37-41.

226. Wu, F., 2008. Field evidence: Bt corn and mycotoxin reduction. ISB

News Report Available: http://www. isb. vt. edu/news/2008/feb08. pdf

[February 2008].

227. Wu, F., Khlangwiset, P., 2010. Health economic impacts and cost-

effectiveness of aflatoxin reduction strategies in Africa: Case studies in

biocontrol and postharvest interventions. Food additives & contaminants.

Part A, Chemistry, analysis, control, exposure & risk assessment 27, 496-

509.

228. Wu, Q., Jezkova, A., Yuan, Z., Pavlikova, L., Dohnal, V., Kuca, K.,

2009. Biological degradation of aflatoxins. Drug Metab Rev 41, 1-7.

229. Xu, L., Eicher, S., Applegate, T., 2011. Effects of increasing dietary

concentrations of corn naturally contaminated with deoxynivalenol on

broiler and turkey poult performance and response to lipopolysaccharide.

Poultry science 90, 2766-2774.

230. Yang, J., Bai, F., Zhang, K., Bai, S., Peng, X., Ding, X., Li, Y.,

Zhang, J., Zhao, L., 2012. Effects of feeding corn naturally contaminated

143

with aflatoxin B1 and B2 on hepatic functions of broilers. Poultry Science

91, 2792-2801.

231. Yildirim, E., Yalçinkaya, L., Kanbur, M., Çinar, M., Oruc, E., 2011.

Effects of yeast glucomannan on performance, some biochemical

parameters and pathological changes in experimental aflatoxicosis in

broiler chickens. Rev. Med. Vet.(Toulouse) 162, 413-420.

232. Yunus, A., Böhm, J., 2011. A simple method for producing aflatoxin

B1 on rice medium for use in experimental animal feeds. The Journal of

Animal and Plant Sciences (Pakistan) 21, 303-304.

233. Yunus, A.W., Razzazi-Fazeli, E., Bohm, J., 2011a. Aflatoxin B1 in

affecting broiler’s performance, immunity, and gastrointestinal tract: A

review of history and contemporary issues. Toxins 3, 566-590.

234. Zhong, L., Zhang, X., Covasa, M., 2014. Emerging roles of lactic

acid bacteria in protection against colorectal cancer. World journal of

gastroenterology: WJG 20, 7878.

235. Zoghi, A., Khosravi-Darani, K., Sohrabvandi, S., 2014. Surface

Binding of Toxins and Heavy Metals by Probiotics. Mini Reviews in

Medicinal Chemistry 14, 84-98.

236. Zychowski, K.E., Hoffmann, A.R., Ly, H.J., Pohlenz, C., Buentello,

A., Romoser, A., Gatlin, D.M., Phillips, T.D., 2013. The Effect of Aflatoxin-

B1 on Red Drum (Sciaenops ocellatus) and Assessment of Dietary

Supplementation of NovaSil for the Prevention of Aflatoxicosis. Toxins 5,

1555-1573.

144

A1 Approval letter of Animal and Ethical Committee