You are on page 1of 223

Authors Author(s) IDTitle Year Source title

Yasmeen R.,57212210294;55910215000;57209285895;55314060300;56278587400;57412617100;7006235298;57213910714
Zahid B., Alyas
Ameliorative
S., Akhtareffects
R.,2024
Zahra
of Brazilian
Lactobacillus
N., Kouser
Journal
S.,against
Hashmi
of Biology
Aflatoxin
A.S., Athar
B1 M.,
[Efeitos
Tayyab
benéficos
M., Anjum
do Lactobacillus
A.A. contra a A

Fouad M.T.,56559634400;6603502043;36456895600;57059500100;
El-ShenawyImpact
M., Hussein
of Lactic
A.M.S.,
Acid
2023Bacteria
El-Desouky
JournalCells
of T.A.
Chemical
on the Aflatoxin
Health Risks
B1 in Wheat Flour During Manufacture Fino Bread

Rämö S., Kahala


7801639765;6505773164;57191855982;
M., Joutsjoki
Aflatoxin
V. B1 Binding
2022byApplied
Lactic Acid
Sciences
Bacteria
(Switzerland)
in Protein-Rich Plant Material Fermentation
Nahle S., El 57209304617;15071327000;57200273084;6603654093;12786756200;23501410100;
Khoury A., Assaf
A promising
J.C., Louka
innovative
N.,
2022Chokr
Innovative
technique
A., AtouiFood
for
A. mycotoxin
Science and
detoxification
Emerging Technologies
from beverages using biofilms of lactic

Manouchehri
55364700700;57204660732;54383844400;57195419053;57873747200;
A., PirhadiPotential
M., Parsaei
Harmful
P., Alikord
2022
Effects
Journal
M.,
of Boldaji
Aflatoxin
of Chemical
H.S.
M1 inHealth
Milk and
Risks
Milk Products and Novel Methods to Reduction o

Nasrollahzadeh
57364484700;57192890722;36240463700;7004131495;
A., Mokhtari
Mycotoxin
S., Khomeiri
detoxification
2022
M., Saris
International
ofP.
food by lactic
Journal
acid
of bacteria
Food Contamination
Khomeiri M. DepartmentIranof Food Science
email:and
khomeiri@gau.ac.ir"
Technology

Wlazło Ł., Nowakowicz-Dębek


36714523800;6603183234;57204107213;7004497735;35069227900;
Effect ofB.,Fermented
Ossowski
2022
M.,
Rapeseed
Animals
Łukaszewicz
Meal in
M.,
Diets
Czech
forA.
Piglets on Blood Biochemical Parameters and the Micr
Czech A. Department13 ofAkademicka
Biochemistry
Polandand Toxicology
Aalipanah S.,
57748089700;57210985195;25653343300;22939237900;
Fazeli M.R.,
Synergistic
Akhavan effects
Sepahi2022
of
A.,probiotic
Shariatmadari
Letters in
Bifidobacterium
Applied
F. Microbiology
isolated from chicken's intestine in combination with p
Fazeli M.R. DepartmentIranof Drug andemail:
Foodmofazeli@yahoo.com"
Control

Wang L., Wang


57209144811;57861544600;55801439700;56873539700;57193358014;57941627400;8240256600;56704017700
X., Chang
Effect
J., Wang
of theP.,Combined
Liu2022
C., Yuan
Toxins
Compound
L., Yin Q.,Probiotics
Zhu Q., Luwith
F. Glycyrrhinic Acid on Alleviating Cytotoxicity of IPEC

Guo H., Wang


55468734700;56873539700;57193358014;57801718800;55801439700;8240256600;57209144811;57217128218
P., Liu C.,Effects
Zhou T.,
of Chang
Compound
J.,
2022
YinMycotoxin
Q.,
Toxins
Wang L.,
Detoxifier
Jin S., Zhu
onQ.,
Alleviating
Lu F. Aflatoxin B1-Induced Inflammatory Response
Yin Q. College of Animal
China Science
email:
andqqy1964@henau.edu.cn"
Technology
Zamani N., Fazeli
57743644600;57210985195;25653343300;22939237900;
M.R., ASepahi
new probiotic
A.A., Shariatmadari
Lactobacillus
2022 Archives
F. plantarum
of Microbiology
strain isolated from traditional dairy together with nanochito
Fazeli M.R. DepartmentIranof Drug and
email:
Foodmofazeli@yahoo.com"
Control

Mahmoud F.F.,
57219700947;57219605102;57192155991;57895473700;
Ahmed Control
E.A., Ahmed
of Aflatoxin
A.M.,
2022
Ahmed
Residues
Journal
N.I.H.
inofBroiler
Advanced
Chicken
Veterinary
Using Saccharomyces
Research cerevisiae Fortified Ration

Massoud R.,57038566400;23969589100;
Zoghi A. Potential probiotic
2022
strains
Journal
withof
heavy
Applied
metals
Microbiology
and mycotoxins bioremoval capacity for application in fo

Abdolmaleki57212943549;56507162200;52263473800;8291261800;36097575200;56522588000;57203634349;
K., Javanmardi
Emerging
F., Gavahian
technologies
2022
M., Phimolsiripol
International
in combination
Y.,
Journal
Ruksiriwanich
with probiotics
of Food Science
W.,
forMir
aflatoxins
and
S.A.,Technology
Mousavi
removal:
Khaneghah
an updated
A. review
PhimolsiripolY. Faculty of Agro-Industry
Thailand email: yuthana.p@cmu.ac.th
Mousavi Khaneghah
A. Department36
ofRakowiecka
Fruit and
Poland
Vegetable
St Product Technology

León Aguilera
57877934200;57221313245;57878197700;7006309426;
X.E., Manzano
Probiotics
A., Pirela
and Gut
D.,2022
Bermúdez
Microbiota
JournalV.in
of Obesity:
Personalized
Myths
Medicine
and Realities of a New Health Revolution

Rungchaiwattanakul
57914731300;9039942400;57215630268;
T.,Antagonistic
Chareonpornwattana
activity
2022 ofS.,
Agriculture
Wickerhamomyces
Sirisomboon
and C.D.
Natural
anomalus,
Resources
Kluyveromyces marxianus and Lactobacillus para

Pourmohammadi
56242368900;57197315597;55206968000;56574210500;
K., Sayadi
Determining
M., Abedi
theE.,adsorption
2022
Mousavifard
Journal
capacity
of
M.Foodand
Composition
stability of and
Aflatoxin
Analysis
B1, Ochratoxin A, and Zearalenon on sin
Rezasoltani 56891645700;57652450300;55603369500;57554217300;37049489600;
S., EbrahimiDetoxification
N.A., Boroujeni
of R.K.,
2022
aflatoxin
Aghdaei
Gastroenterology
M1 by
H.A.,
probiotics
Norouzinia
andSaccharomyces
Hepatology
M. from
boulardii,
Bed to Lactobacillus
Bench casei, and Lactobac

Chen G., Fang


57710715500;57710201600;35107597800;57711474100;57710456900;57211885549;57710715600;5710239480
Q., Liao Z.,
Detoxification
Xu C., Liang Z.,
of 2022
Aflatoxin
Liu T.,Frontiers
Zhong
B1 by
Q.,in
aWang
Potential
Microbiology
L., Fang
Probiotic
X., Wang
Bacillus
J. amyloliquefaciens WF2020
Wang J. GuangdongChinaProvincial Key
email:
Laboratory
wangjielangjing@126.com"
of Food Quality and Safety

Li J., Hu K., Hu
56662275800;57195304751;57697793300;57696237100;57192697322;56857053500;42360959900;5685729540
L., Hou X.,
Adsorption
Li Q., Liu A.,
Behavior
Chen
2022
S.,
ofAo
International
3-phenoxybenzoic
X., Hu X., He
Journal
L., Tang
Acid
of Molecular
H.,
by Lactobacillus
Huang D.,
Sciences
Yang
Plantarum
Y., Zou L.,and
LiuIts
S. Potential Applicatio
Petrova P., Arsov
15051152600;56006170000;55948138000;25958341700;36964480700;57222094711;13406656700;
A., Tsvetanova
The Complex
F., Parvanova-mancheva
Role2022
of Lactic
Nutrients
Acid Bacteria
T., Vasileva
in Food
E.,Detoxification
Tsigoriyna L., Petrov K.

Ahmed S.A.A.,
57210207460;57219124385;57199177587;57221752950;57338080300;57014822500;57204001011;5600219360
Nada H.S.,
Comparative
Elsheshtawy
antitoxic
H.M.,
2022Ibrahim
potency
Aquaculture
S.M.,
of honey
Fahmyand
E.M.,
natamycin-supplemented
Khedr M.H.E., Moustafa
diets
S.M.,
against
Ismailaflatoxicosis
T.A., Gesrihaand
S., At

Adácsi C., Kovács


57561465000;57489079400;7005576730;55955389200;56470893600;6602242205;
S., Pócsi
Microbiological
I., Győri Z., Dombrádi
and
2022
Toxicological
Agriculture
Z., Pusztahelyi
Evaluation
(Switzerland)
T. of Fermented Forages
Farkas Z., Országh
55902322400;57218659276;57443428100;57281038500;55901981800;6506405700;9846129200;57218862763;
E., Engelhardt
A Systematic
T., Csorba
Review
2022
S.,
ofToxins
Kerekes
the Efficacy
K., Zentai
of Interventions
A., Süth M.,toNagy
Control
A., Miklós
Aflatoxins
G., Molnár
in the Dairy
K., Rácz
Production
C., Dövényi-N
Chai

Khalafalla M.M.,
55902314900;57419408300;57214912002;57205488002;57190869274;56554243100;12784227900;5637343340
Zayed Dietary
N.F.A., Amer
Lactobacillus
A.A.,2022
Soliman
acidophilus
Probiotics
A.A., Zaineldin
ATCC
and Antimicrobial
4356A.I.,
Relieves
Gewaily
Proteins
the
M.S.,
Impacts
Hassan
of Aflatoxin
A.M., VanB1
Doan
Toxicity
H., Tapingkae
on the Grow
W.,
Dawood M.A.O. Animal Production
Egypt Department
email: Mahmoud.dawood@agr.kfs.edu.eg"

Liew W.-P.-P.,
57200647159;55598057100;57204624974;57209973113;
Sabran M.-R.,
Metagenomic
Than L.-T.-L.,
and2022
proteomic
Abd-Ghani
Food and
approaches
F. ChemicalinToxicology
elucidating aflatoxin B1 detoxification mechanisms of pro
Chaudhary H.J.,
57361010500;57203580954;
Patel A.R.
Removal of aflatoxin
2022M1
Food
from
Bioscience
milk and aqueous medium by indigenously isolated strains of W. confus

Pakizeh M.,58010572000;56156064200;25421111100;
Nouri L., Azizi
Probiotic-Based
M.H. Optimization
2022 JournalofofPistachio
Food Quality
Paste Production and Detoxification of Aflatoxin B1 Using Bi
Ahmad K.N.,57220105230;57222763451;57220108918;57207890166;
Samir Z.T.,ESTIMATION
Mustafa A.A.,
OFSaadi
THE
2022A.M.
EFFECTIVENESS
Journal of Hygienic
OF LACTIC
Engineering
ACID BACTERIA
and Design
IN REDUCING THE CONCENTRATIONS O

Mohamed S.I.A.,
57907444700;14055026600;57908761200;36883791000;57205193161;
Shehata
Does
S.A.M.,
the Use
Bassiony
of Different
2022S.M.,
Probiotics
Mahgoub
Types ofand
Probiotics
S.A.M.,
Antimicrobial
Abd
Possess
El-Hack
Proteins
Detoxification
M.E. Properties Against Aflatoxins Con
Bangar S.P.,57117808600;57201952212;57213226217;8291261800;
Sharma N.,Lactic
Bhardwaj
acid bacteria
A., Phimolsiripol
2022 Quality
Y. Assurance and Safety of Crops and Foods

Gonçalves B.L.,
56304071300;57208163899;56649356600;57196346193;57215382226;25222853500;6508389444;
Uliana R.D.,
Use ofLee
scanning
S.H., Coppa
electron
2022C.F.,
Foodmicroscopy
de Science
OLIVEIRAand
and
C.A.F.,
Technology
high-performance
Kamimura
(Brazil)
E.S.,liquid
Corassin
chromatography
C.H. to assess the abilit

Adriansyah 57467206200;55622287500;56422244300;57466630000;
P.N.A., Rahayu
Aflatoxin
W.P., M1
Kusumaningrum
reduction
2022 International
by microorganisms
H.D., Kawamura
Food Research
isolated
O. from
Journal
kefir grains

Escrivá L., Agahi


56083881800;57216039099;56431158100;7006949013;25227937100;36465287000;
F., Vila-Donat
Bioaccessibility
P., Mañes study
2022
J., Meca
ofToxins
aflatoxin
G., Manyes
b1 and
L. ochratoxin a in bread enriched with fermented milk whey and

Emadi A., Eslami


56948170400;57201082050;57194722962;56574264000;
M., Yousefi
In vitro
B.,strain
Abdolshahi
specific
2022A.reducing
Toxin Reviews
of aflatoxin B1 by probiotic bacteria: a systematic review and meta-ana
Ondiek W., 57221703580;38863481800;55508692000;57195420615;7005154186;55776002700;57221693153;
Wang Y., Sun
Removal
L., ZhouofL.,aflatoxin
On S.L.W.,
2022b1Food
and
Zheng
t-2
Science
H.,
toxin
Ravi
and
byG.bacteria
Technology
isolated
International
from commercially available probiotic dairy

Tajik H., Sayadi


23010589600;57197315597;
M. Effects of probiotic
2022
bacteria
Toxinof
Reviews
Lactobacillus acidophilus and Lactobacillus casei on aflatoxin B1 detoxif

Yasmeen R.,57212210294;7006235298;57213910714;55765162000;24767811300;
Hashmi A.S.,
Molecular
Athar M.,
characterization
Tayyab
2021
M.,Journal
Anjum
of lactobacillus
of
A.A.
Animal andfrom
Plant
broiler
Sciences
and in-vitro detoxification of aflatoxin b1

Khorshidi F.,57223026418;57196350170;56948314500;57209010226;23009371700;
Torshizi M.A.K.,
The efficiency
Ahmadi H.,
of probiotic
2021
Arak H.,
Journal
Mojgani
and of
toxin
Veterinary
N.binders Research
(organic and inorganic) in Amelioration of Aflatoxin impac
Guo H.-W., 55468734700;55801439700;56873539700;8240256600;57193358014;57216083300;54991772800;55496165400
Chang J., Wang
Effects
P., of
Yincompound
Q.-Q., 2021
Liu C.-Q.,
probiotics
AMBXuExpress
X.-X.,
and Dang
aflatoxin-degradation
X.-W., Hu X.-F., Wang
enzyme
Q.-L.
on alleviating aflatoxin-induced cyto
Yin Q.-Q. College of Animal
China Scienceemail:
andqqy1964@126.com"
Technology

Fahim K.M.,57216196168;57208414665;57193997308;57202256426;56285494700;
Badr A.N., Innovative
Shehata M.G.,
application
Hassanen
2021 of
Journal
E.I.,
postbiotics,
Ahmed
of Dairy
L.I.
parabiotics
Research and encapsulated Lactobacillus plantarum RM1 and L

Hu L.-F., Lan57226074402;37761678400;57367576400;57253176500;36486793500;
H.-R., Huang
Personalized
D., Li X.-M.,Immunotherapy
Jin2021
K.-T. Frontiers
in Colorectal
in OncologyCancers: Where Do We Stand?
Li X.-M. DepartmentChina
of Hepatobiliary
email: Surgery
lxm2100236@163.com"
Mohammadi 36716645500;57720594900;57720005400;36141248900;12792054500;56536895000;57221518954;
R., Erfani N.,
Aflatoxin
Sohravbvandi
M1 Reduction
2021
S., Mortazavi
Iranian
by Probiotic
Journal
S.A., Mortazavian
Strains
of Chemistry
in Iranian
A.M.,
and
Feta
Sarlak
Chemical
Cheese
Z., Alizadeh
Engineering
Moghadam M.
Mortazavi S.A. DepartmentIran
of Food Science
email:and
morteza1937@yahoo.com"
Technology

Illueca F., Vila-Donat


57214464655;56431158100;57194500681;57172148600;25227937100;56680899300;
P.,Antifungal
Calpe J., Luz
activity
C., Meca
2021
of biocontrol
G.,
Toxins
Quilesagents
J.M. in vitro and potential application to reduce mycotoxins (Aflatox

Paraskeuas 56893886800;57208799938;57215723742;6508387963;56028975600;6505845109;
V., Griela E.,Effects
Bouziotis
of deoxynivalenol
D., Fegeros
2021 K.,
Toxins
and
Antonissen
fumonisins
G., Mountzouris
on broiler gutK.C.
cytoprotective capacity

Pourmohammadi
56242368900;57215275857;55927243400;26431230600;
K., Sohrabi
A kinetic
M.,analysis
Hashemi 2021
ofS.M.B.,
theFEMS
aflatoxin
Amiri
Microbiology
M.J.
detoxification
Letters
potential of lactic acid bacteria in Terxine (a cereal-bas
Jin J., Beekmann
56594052600;54402293000;56212489400;7005116412;55416369100;
K., Ringø
Interaction
E., Rietjens
between
I.M.C.M.,
2021food-borne
Food
XingControl
F. mycotoxins and gut microbiota: A review

Guerrero-Encinas
57221729292;57217203074;56507285000;8731941500;7202282374;6506846713;55989802400;6603013016;56
I., González-González
Protective Effect J.N.,
2021
of Lacticaseibacillus
Santiago-López
Probiotics andL.,
casei
Antimicrobial
Muhlia-Almazán
CRL 431 Postbiotics
Proteins
A., Garcia
on Mitochondrial
H.S., Mazorra-Manzano
Function and
M.A.,
Oxidati
Valle

Zhao Y., Wang


57223417184;57221494949;57189065777;57205696244;36703250600;55138236100;56162573300;9745735500
T., Li P., Bacillus
Chen J., amyloliquefaciens
Nepovimova
2021E.,Ecotoxicology
Long
B10M.,
canWu
alleviate
and
W., Environmental
Kuca
aflatoxin
K. B1-induced
Safety kidney oxidative stress and apoptosis
Wu W. MOE Joint International
China email:
Research
wuwenda@njau.edu.cn"
Laboratory of Animal Health and Food Safety

Muaz K., Riaz


57201301728;57220653976;23973211900;57205636403;6508389444;56304071300;35557654100;
M., RosimInR.E.,
vitroAkhtar
abilityS.,
ofCorassin
nonviable
2021 LWTC.H.,
cellsGonçalves
of lactic acid
B.L.,bacteria
Oliveirastrains
C.A.F. in combination with sorbitan monosteara
Riaz M. Institute of Pakistan
Food Scienceemail:
& Nutrition
riaz@bzu.edu.pk"
Pavlek Ž., Bošnir
57200854311;6602383526;55856293600;57195361778;57195360665;6507280772;6505884869;7102521923;57
J., Ivešić
Investigation
M., Serdar of
S., milk
Kuharić
2021quality
Medica
Ž., Jakopović
after
Jadertina
removal
Ž., Frece
of afm1
J., Markov
using lactic
K., Čanak
acid bacteria
I., Racz A.and beta-glucan [Istraživan

Chibuzor-Onyema
57200962659;57057793600;55893821400;57222259218;24071297500;57217025966;35726731100;7003506571
I.E., Ezeokoli
Metataxonomic
O.T., Sulyok
analysis
2021M.,
Food
of
Notununu
bacterial
Research
I.,communities
Petchkongkaew
Internationaland A.,
mycotoxin
Elliott C.T.,
reduction
Adelekeduring
R.A., Krska
processing
R., Ezekiel
of three
C.N

Møller C.O.D.A.,
56457721300;57191708331;23973211900;57191343877;56041720100;56654845600;57192073087;6508389444
Freire Effect
L., Rosim
of Lactic
R.E., Margalho
Acid
2021Bacteria
Frontiers
L.P.,Strains
Balthazar
in Microbiology
on the
C.F.,Growth
Franco and
L.T.,Aflatoxin
Sant’AnaProduction
A.D.S., Corassin
Potential
C.H.,of
Rattray
Aspergillus
F.P., Ol
p
Zhang Y., Wang
57219261829;55658456700;7102722752;7006597429;
P., KongBiotransformation
Q., Cotty P.J. 2021
of aflatoxin
Food Chemistry
B1 by Lactobacillus helviticus FAM22155 in wheat bran by solid-state fe

Seyedjafarri57292032200;
S. Detoxification of 2021
Aflatoxin
Asian
M1Journal
in MilkofbyDairy
Lactic
and
Acid
Food
Bacteria
Research

Faghihi Shahrestani
57210965349;54391631600;16232338600;57212485436;6508377827;
F., Tajabadi
Identification
Ebrahimi
of dairy
2021
M., fungal
Bayat
Archives
M.,
contamination
of
Hashemi
Razi Institute
J., and
Razavilar
reduction
V. esistant rile bid and cahree tmount by a1 fla
Ragoubi C., 57222729962;36802434800;56009530000;57188667853;55181544200;57397585100;6602427599;6602237002;
Quintieri L.,Mycotoxin
Greco D., Mehrez
Removal
2021
A.,
byMaatouk
Toxins
Lactobacillus
I., D’ascanio
spp. andV.,
Their
Landoulsi
Application
A., Avantaggiato
in Animal Liquid
G. Feed

Fouad M.T.,56559634400;6603502043;57059500100;
El-ShenawyEfficiency
M., El-Desouky
of selected
T.A.
2021lactic
Journal
acidofbacteria
Pure andisolated
Appliedfrom
Microbiology
some dairy products on Aflatoxin B1and Ochrat

Wass Thilakarathna
57201216251;6602253478;7004765931;
W.P.D.,
Mechanisms
Rupasinghe
by which
2021
H.P.V.,
probiotic
International
Ridgwaybacteria
N.D.Journal
attenuate
of Molecular
the riskSciences
of hepatocellular carcinoma
Rupasinghe H.P.V. DepartmentCanada
of Pathology
email: vrupasinghe@dal.ca"
Baker J.T., Duarte
57222146963;57209505600;57196082397;34975075200;
M.E.,Friend
Holanda
or foe?
D.M.,Impacts
Kim
2021
S.W.
Animals
of dietary xylans, xylooligosaccharides, and xylanases on intestinal health and g

mohammadi57221563290;48461092100;35226619500;35559570600;35560048300;57218672617;14012236000;
R., Abbaszadeh
In vitroS.,activity
Sharifzadeh
of2021
encapsulated
A.,Food
Sepandi
Science
lactic
M., and
Taghdir
acidNutrition
bacteria
M., Youseftabar
on aflatoxin
Miri
production
N., Parastouei
and growth
K. of Aspergillus S
AbbaszadehS. DepartmentIran of Nutritionemail:
and Food
abbaszade@ut.ac.ir
Hygiene
Sharifzadeh A. DepartmentIran of Microbiology
email:and
abbaszade@ut.ac.ir"
Immunology

Peles F., Sipos


20734974300;57221940843;57489079400;55955389200;7005576730;6602242205;
P., Kovács
Biological
S., Gyoricontrol
Z., Pócsi
2021
and
I., Pusztahelyi
mitigation
Toxins ofT.aflatoxin contamination in commodities
Azizi M.N., Loh
57221770537;57212574896;57024850600;56714867800;
T.C., FooIsH.L.,
palmChung
kernelE.L.T.
cake
2021
a suitable
Animalsalternative feed ingredient for poultry?
Loh T.C. Institutes ofMalaysia
Tropical Agriculture
email: tcloh@upm.edu.my"
and Food Security

Zoghi A., _Darani


23969589100;23969408200;7801320384;
K.K., Hekmatdoost
Effects of Pretreatments
A. 2021 Probiotics
on Patulin
andRemoval
Antimicrobial
from Apple
Proteins
Juices Using Lactobacilli: Binding Stability in S

de Oliveira C.A.F.,
57215382226;57201301728;57540391000;6508389444;6602698932;
MuazProbiotics
K., de Almeida
and Mycotoxins
Møller
2021 Probiotics
C.O., Corassin
and C.H.,
Prebiotics
Rattray
in Foods:
F.P. Challenges, Innovations, and Advances
Oonaka K., Kobayashi
6603438259;55587041100;57209733218;57201801969;57388967500;7003593746;
N.,
In Uchiyama
vivo and InY.,
vitro
Honda
2021
Mitigation
Journal
M., Miyake
Effects
of the
S., Food
of
Sugita-Konishi
Lactic
Hygienic
Acid Bacteria
Society
Y. of
Derived
Japan from Fresh Vegetables on Aflatoxi

Vasconcelos57370904400;56321139400;57200416866;57209342544;56053271900;55797352000;14626000200;7102639646
R.A.M., Kalschne
Feasibility
D.L.,ofWochner
L. Plantarum
2021K.F.,
Food and
Moreira
Science
prebiotics
M.C.C.,
and Technology
onBecker-Algeri
aflatoxin(Brazil)
b1 detoxification
T.A., Centenaroin A.I.,
cowColla
milk E., Rodrigues P.C.A

Allameh A., 55398907600;57204943740;16425960800;56901444800;


Khanian M.,Hepatoprotective
Karimi-Torshizi M.-A.,
2021
effects
Avian
Kalantari-Hesari
of Lactobacillus
Pathology A.plantarum 299v supplemented via drinking water against afl
Guo H., Chang
55468734700;55801439700;56873539700;8240256600;57193358014;57210814301;56704017700;23974104200
J., WangDetoxification
P., Yin Q., Liu C.,
of 2021
aflatoxin
Li S., Zhu
FoodB1
Q.,Additives
in
Yang
broiler
M.,and
chickens
Hu Contaminants
X. by a triple-action
- Part A Chemistry,
feed additive
Analysis, Control, Exposur
Yin Q. College of Animal
China Science email:
andqqy1964@126.com"
Technology

Abdi M., Asadi


57211532223;57189890183;56120470300;55866535000;55565969000;56119688500;23100555100;5718935381
A., Maleki
Microbiological
F., Kouhsari E.,detoxification
2021
Fattahi
Infectious
A., Ohadi
of mycotoxins:
Disorders
E., Lotfali- E.,
Drug
Focus
Ahmadi
Targets
on mechanisms
A., Ghafouri and
Z. advances

Li F., Huang27171461000;55316560600;57198448271;55317091100;53064776800;57224987426;
L., Chen H.,Effect
Yuan of
X., Clostridium
Wang C., 2021
Wang
onWorld
proliferating
J. Mycotoxin cell Journal
nuclear antigen and ghrelin in the small intestine of fattening
Gámiz-Gracia6601966891;7004918727;
L., García-Campaña
Report of theA.M.
Vth2021
Workshop
Toxinsof the Spanish National Network on Mycotoxins and Toxigenic Fungi and

Guan Y., Chen


57221708596;57205696244;36703250600;55138236100;56162573300;9745735500;
J., Nepovimova
Aflatoxin
E.,Detoxification
Long M.,
2021
WuToxins
W.,
Using
Kuca
Microorganisms
K. and Enzymes
Wu W. DepartmentCzech
of Chemistry
Republic
email: wuwenda@njau.edu.cn
Kuca K. DepartmentCzech
of Chemistry
Republic
email: kamil.kuca@uhk.cz"
da Cruz P.O.,
57223824840;57223810963;57192961648;14052955400;9332736200;26660951400;
de Matos Efficacy
C.J., Nascimento
of Potentially
2021
Y.M.,Probiotic
Toxins
Tavares J.F.,
Fruit-Derived
de Souza E.L.,
Lactobacillus
Magalhães
fermentum,
H.I.F. L. paracasei and L. plantarum t

Danial E.N.,35271748700;57220778062;57220781367;57220777283;57220784446;57220779200;
Lamfon M.Y.,
Removal
Alghamdi
of aflatoxin
L.A.,2021
Alamri
G1Journal
using
A.M., lactic
Alghamdi
of Food
acidProcessing
M.S.,
bacteria
Alghamdi
and Preservation
S.A.
Danial E.N. Chemistry of
Egypt
Natural andemail:
Microbial
enas_mahdy@yahoo.com"
Products Department

Ibitoye O.A.,57219054573;36652982700;57194798638;10042737300;
Olaniyi O.O.,
Lactic
Ogidi
acid
C.O.,
bacteria
Akinyele
2021
bio-detoxified
Toxin
B.J. Reviews
aflatoxins contaminated cereals, ameliorate toxicological effects and

Bata-Vidács55957668100;57194177796;35611669400;6701772622;55957684600;
I., Kosztik J.,
Aflatoxin
Mörtl M.,
B1Székács
and Sterigmatocystin
2020
A., Toxins
Kukolya J. Binding Potential of Non-Lactobacillus LAB Strains
Liu A., Zheng
56857053500;57219150434;57219144773;42360959900;56661522500;57211083468;57077891800;5719187161
Y., Liu L., Chen
Decontamination
S., He L., Ao 2020
X.,
of Aflatoxins
Yang
Current
Y., Liuby
Microbiology
S.Lactic Acid Bacteria
Liu S. College of Food
ChinaScienceemail: lsliang999@163.com"

Khadivi R., Razavilar


57224421832;6508377827;55888936600;15769124600;
V.,Aflatoxin
Anvar S.A.A.,
M1-binding
Akbari-Adergani
2020ability
Archives
of B.
selected
of Razi Institute
lactic acid bacteria strains and saccharomyces boulardii in the

Furukawa T.,
56209841300;7202463945;35262767400;37052547200;57219089612;56864855400;36877753600;56865388300
KatayamaDioctatin
H., Oikawa
Activates
A., Negishi
2020
ClpPCell
L.,toIchikawa
Degrade
ChemicalT.,
Mitochondrial
Biology
Suzuki M., Murase
Components
K., Takayama
and Inhibits
S., Sakuda
Aflatoxin
S. Production

Kumara S.S.,57204524925;55177214500;24723888300;36128020000;57190050429;
Gayathri D.,
In Hariprasad
vivo AFB1 detoxification
P., Venkateswaran
2020 Toxicon
by Lactobacillus
G., Swamy fermentum
C.T. LC5/a with chlorophyll and immunopotentiatin
Ben Salah-Abbès
57218571664;57207105934;57217592129;57217590312;6602076540;13407983200;
J., Belgacem
Immunological
H., Ezdinieffects
K.,
2020
Mannai
of
Immunopharmacology
AFM1
M., in
Oueslati
experimental
R., Abbès
and
subchronic
Immunotoxicology
S. dosing in mice prevented by lactic acid bac

Ramlucken 57213596679;22938011900;57218555858;36515320500;56654945700;13003467800;
U., Lalloo R.,Advantages
Roets Y., Moonsamy
of Bacillus-based
2020 G.,
Livestock
vanprobiotics
Rensburg
ScienceC.J.,
in poultry
Thantsha
production
M.S.

Ugbaja R.N.,8985964500;57218552250;57192960585;57218557535;56602776500;57218558844;6602293601;
Okedairo O.M.,
Probiotics
Oloyede
consortium
A.R.,
2020
Ugwor
synergistically
Toxicon
E.I., Akinloye
ameliorates
D.I., Ojo O.P.,
aflatoxin
Ademuyiwa
B1-induced
O. disruptions in lipid metabolism
Loh Z.H., Ouwerkerk
57219515775;6507592652;6701356845;6603023877;7203082310;
D.,Toxin
Klieve
degradation
A.V., Hungerford
2020
by rumen
Toxins
N.L.,microorganisms:
Fletcher M.T. A review

Gonçalves B.L.,
56304071300;57201301728;57196346193;23973211900;25222853500;35557654100;6508389444;
Muaz K.,Aflatoxin
Coppa C.F.S.C.,
M1 absorption
2020
RosimFood
R.E.,
by non-viable
Research
KamimuraInternational
cells
E.S.,of
Oliveira
lactic acid
C.A.F.,
bacteria
Corassin
andC.H.
Saccharomyces cerevisiae strain

Khoori E., Hakimzadeh


57218097258;14057886800;57218096357;56458107100;
Effect
V., Mohammadi
of ozonation,
Sani
2020
UV
A.,Journal
light
Rashidi
radiation,
ofH.Food and
Processing
pulsed and
electric
Preservation
field processes on the reduction of total afla
Ye L., Wang57194184457;38863481800;55508692000;55944772500;55199703600;57216907453;57210788623;5691519740
Y., Sun L., Fang
The effects
Z., DengofQ.,
removing
Huang
2020 LWT
Y.,
aflatoxin
Zheng P.,
B1 Shi
andQ.,
T-2Liao
toxin
J., by
Zhao
lactic
J. acid bacteria in high-salt fermented fish pro

Yan M., Wang


57188685895;57201644912;57218249649;57218250740;57218253039;57218250422;57218250894;1194011790
B.-H., FuPetunidin-Based
X., Gui M., WangAnthocyanin
2020
G., Zhao
Frontiers
L., LiRelieves
R.,
in You
Microbiology
Oxygen
C., Liu Z.Stress in Lactobacillus plantarum ST-III

Mosallaie F.,57212586876;26323631600;56230539700;14068433700;
JooyandehBiological
H., Hojjatireduction
M., Fazlara
2020of aflatoxin
Food
A. ScienceB1 inand
yogurt
Biotechnology
by probiotic strains of Lactobacillus acidophilus and Lacto

Zhao Q., Qiu57217338728;57200709646;57206604846;57214838162;57214839854;57214836820;57203249114;5720531222


Y., Wang X.,
Inhibitory
Gu Y., Zhao
Effects
Y., 2020
Wang
of Eurotium
Frontiers
Y., Yuecristatum
T.,inYuan
Microbiology
Y.on Growth and Aflatoxin B1 Biosynthesis in Aspergillus flavus
Trela J., Kierończyk
57216920675;56019110500;24080208800;8858796600;
B., Hautekiet
Combination
V., Józefiak
of bacillus
2020D.Animals
licheniformis and salinomycin: Effect on the growth performance and git micro

Ben Taheur57133187900;57194467726;57188692252;57209316005;35072714000;57215688269;56175443800;
F., MansourAflatoxin
C., Ben Jeddou
B1 degradation
K.,
2020
Machreki
Toxicon
by microorganisms
Y., Kouidhi B., Abdulhakim
isolated from
J.A.,
Kombucha
Chaieb K.culture

Chang J., Wang


55801439700;57215498717;56873539700;8240256600;57193358014;56704017700;56704116100;57202389414
T., Wang
Compound
P., Yin Q.,probiotics
Liu C.,2020
Zhualleviating
Q.,
Ecotoxicology
Lu F., Gao
aflatoxin
T.
and Environmental
B1 and zearalenone
Safetytoxic effects on broiler production perfo

Nguyen T., Flint


57212349652;7101692911;7403311082;
S., Palmer
Control
J. of aflatoxin
2020
M1Food
in milk
Chemistry
by novel methods: A review
Afshar P., Shokrzadeh
16300657600;12767398800;55796286300;57206901795;37075334600;
M.,
Aflatoxins
Raeisi S.N.,
biodetoxification
Ghorbani-HasanSaraei
2020 Toxicon
strategiesA.,
based
Nasiraii
on probiotic
L.R. bacteria

Adegbeye M.J.,
57204664443;57197708269;55249697100;57215091132;55549603900;55785996500;56589498600;
Reddy P.R.K.,
Mycotoxin
Chilaka
toxicity
C.A.,
2020
and
Balogun
Toxicon
residue
O.B.,
in Elghandour
animal products:
M.M.M.Y.,
Prevalence,
Rivas-Caceres
consumer
R.R.,
exposure
Salem A.Z.M.
and reduction stra

Kavitake D.,57110397100;57109501500;57197868607;57109959400;7101901277;
Singh S.P., Report
Kandasamy
on aflatoxin-binding
S., Devi
2020P.B.,
3 Biotech
Shetty
activity
P.H.of galactan exopolysaccharide produced by Weissella confusa KR7

Loi M., Paciolla


57190960623;6603035400;7003551567;55981606000;
C., Logrieco
PlantA.F.,
Bioactive
Mulè Compounds
G.2020 Frontiers
in Pre-
in Microbiology
and Postharvest Management for Aflatoxins Reduction
Zhou Y., Zeng
57216617770;57216619982;57219242648;57208134069;56984618300;7004228695;57114669600;57132273900
Z., Xu Y., Application
Ying J., Wang
ofB.,
Bacillus
Majeed
2020 coagulans
Animals
M., Majeed
in animal
S., Pande
husbandry
A., Li W.and its underlying mechanisms

Wacoo A.P.,57053526200;57190385553;57190373879;6602627726;7404050085;57195805870;6701854608;35621200100;7
Atukunda P.,
Aflatoxins:
MuhooziOccurrence,
G., Braster
2020 Microorganisms
M.,
exposure,
Wagnerand
M.,binding
van dentoBroek
lactobacillus
T.J., Sybesma
speciesW.,
from
Westerberg
the gut microbiota
A.C., Iversen
of rural
P.O.

Chlebicz A.,57201796495;8956663900;
Śliżewska K.In Vitro Detoxification
2020 of
Probiotics
Aflatoxinand
B1,Antimicrobial
Deoxynivalenol,
Proteins
Fumonisins, T-2 Toxin and Zearalenone by Prob

Darani K.K.,23969408200;23969589100;23492258700;57221403857;
Zoghi A., Jazayeri
DECONTAMINATION
S., da Cruz2020
A.G.OFJournal
AFLATOXINS
of Microbiology,
WITH A FOCUS
Biotechnology
ON AFLATOXIN
and Food
B1 BY
Sciences
PROBIOTIC BACTERIA AND
Fouad M.T.,56559634400;57059500100;
El-DesoukyAnti-toxigenic
T.A. effect
2020of Open
lactic Microbiology
acid bacteria against
Journal aspergillus spp isolated from wheat grains

Muaz K., Riaz


57201301728;57220653976;
M. Decontamination2020
of aflatoxin
JournalM1
of Animal
in milk through
and Plantintegration
Sciences of microbial cells with sorbitan monostea

Asurmendi P.,
40261031400;40261523300;7005788612;7007077853;
GerbaldoLactic
G., Pascual
acid bacteria
L., Barberis
2020
withJournal
L.
promising
of Environmental
AFB1 binding Science
properties
andasHealth
an alternative
- Part B Pesticides,
strategy toFood
mitigate
Contamin
conta
Zolfaghari H.,
57084779900;57202610795;55194667500;57189798981;
KhezerlouDetoxification
A., Ehsani A., of
Khosroushahi
2020
aflatoxin
Advanced
B1A.Y.
by probiotic
Pharmaceutical
yeasts and
Bulletin
bacteria isolated from dairy products of Iran
KhosroushahiA.Y. Drug AppliedIran
Research email:
Centeryarikhosroushahia@tbzmed.ac.ir"
Rodríguez M.,
7404259384;56260782100;
Núñez F.Novel approaches2020 to minimizing
Toxins mycotoxin contamination

Peles F., Sipos


20734974300;57221940843;55955389200;55295838100;36552291700;6603251591;6601997736;6505963372;7
P., GyőriAdverse
Z., Pfliegler
Effects,
W.P.,Transformation
2019
Giacometti
Frontiers
F., in
Serraino
andMicrobiology
Channeling
A., Pagliuca
of Aflatoxins
G., Gazzotti
IntoT.,
Food
Pócsi
RawI. Materials in Livestock
Kumara S.S.,57204524925;57204526205;36128020000;24723888300;55177214500;
Bashisht A.,
Characterization
Venkateswaranof
2019
G.,
Novel
Hariprasad
Probiotics
Lactobacillus
P.,
andGayathri
Antimicrobial
fermentum
D. from
Proteins
Curd Samples of Indigenous Cows from Malna

Sampaolesi 55980557400;57190250621;55667106800;57190376156;
S., Gamba R.R.,
Potentiality
De Antoni
of yeasts
G.L.,
2019León
obtained
LWTPeláezasÁ.M.
beer fermentation residue to be used as probiotics

Wochner K.F.,
57200416866;57209342544;56321139400;14626000200;34968988400;
Moreira Detoxification
M.C.C., Kalschne
of 2019
Aflatoxin
D.L., Colla
Journal
B1
E.,and
Drunkler
of Food
M1 by
Safety
D.A.
Lactobacillus acidophilus and prebiotics in whole cow's milk

Zhang G., Li57207176469;57216664439;57209341395;57209344562;55607693000;55715265500;


J., Lv J., Liu Decontamination
L., Li C., Liu L. 2019
of aflatoxin
JournalM1
of Food
in yogurt
Safety
using Lactobacillus rhamnosus LC-4
Ben Taheur57133187900;57194467726;35072714000;56175443800;
F., MansourUse
C., of
Kouidhi
lactic B.,
acidChaieb
2019
bacteria
Toxicon
K. for the inhibition of Aspergillus flavus and Aspergillus carbonarius growth an

Huang L., Zhao


57196687532;57200687871;55286262800;57200688715;55349762500;57194217808;57192429339;5535137000
Z., DuanLactobacillus
C., Wang C., plantarum
Zhao2019
Y., Yang
BMC
C88G.,
Microbiology
protects
Gao L.,against
Niu C., Xu
aflatoxin
J., Li S.B1-induced liver injury in mice via inhibition of N

Ahlberg S., Randolph


56663034000;57210357454;24559623500;56754299200;
D.,Aflatoxin
Okoth S.,binders
Lindahl
2019
inJ.foods
Toxins
for human consumption-can this be promoted safely and ethically?
Panwar R., Kumar
56213549300;57206866056;57201556265;7006555189;7003788183;
N., Kashyap
Aflatoxin
V.,M1
Ram
Detoxification
C.,
2019
Kapila
Probiotics
R. Abilityand
of Probiotic
Antimicrobial
Lactobacilli
Proteins of Indian Origin in In vitro Digestion Model

Zakaria A.M.,
57205426150;57202927999;57420236700;57208082387;57209396052;
Amin Y.A.,Rapid
Khalildetection
O.S.F., Abdelhiee
of
2019
aflatoxin
Journal
E.Y.,M1
Elkamshishi
ofresidues
AdvancedinM.M.
market
Veterinary
milkand
in Aswan
AnimalProvince,
ResearchEgypt and effect of probiot

Assaf J.C., Nahle


57200273084;57209304617;12786756200;6603654093;23501410100;15071327000;
S., Chokr
Assorted
A., Louka
methods
N., Atoui
2019
for A.,
decontamination
Toxins
El Khoury A. of aflatoxin M1 in milk using microbial adsorbents
Sevim S., Topal
57190735573;57206243509;57194614848;24780816000;54684245600;
G.G., Tengilimoglu-Metin
Effects of inulin and
2019
M.M.,
lactic
Food
Sancak
acid
Control
bacteria
B., Kizilstrains
M. on aflatoxin M1 detoxification in yoghurt

Poornachandra
56512412000;57189895094;57207551747;57203832887;6602079682;22938985600;
Rao K., Antiaflatoxigenic
Deepthi B.V., Rakesh
2019
Potential
S.,Probiotics
Ganesh
of Cell-Free
T.,
andAchar
Antimicrobial
Supernatant
P., Sreenivasa
Proteins
fromM.Y.
Lactobacillus plantarum MYS44 Against Aspe

Adebo O.A.,56725372400;37037602100;8294774600;
Kayitesi E.,Reduction
Njobeh P.B.
of Mycotoxins
2019 Toxins
during fermentation of Whole Grain Sorghum to Whole Grain ting (A Southe
Śliżewska K.,8956663900;6602868862;6601999477;6602948995;
CukrowskaThe
B., Effect
Smulikowska
of Probiotic
2019
S., Cielecka-Kuszyk
Supplementation
Toxins J. on Performance and the Histopathological Changes in Liver an

Abdel-Kareem
57192405379;57205319501;6603728066;
M.M., Rasmey
The action
A.M.,mechanism
Zohri2019
A.A. Letters
and biocontrol
in Applied
potentiality
Microbiology
of novel isolates of Saccharomyces cerevisiae again
Ferrero F., Prencipe
57206713164;56997351700;7004011153;7003905676;8271246400;36088043700;7801407428;35760137900;
S., Increase
Spadaro D.,
in aflatoxins
Gullino
2019
M.L.,
due
Journal
Cavallarin
to Aspergillus
of Dairy
L., Piano
Science
section
S., Tabacco
Flavi multiplication
E., Borreaniduring
G. the aerobic deterioration of

Khanian M.,57204943740;16425960800;55398907600;
Karimi-Torshizi
Alleviation
M.-A., of
Allameh
aflatoxin-related
2019
A. Toxicon oxidative damage to liver and improvement of growth performance in

Byakika S., Mukisa


57192156416;35790309300;57053526200;7004607886;6506745146;8253852400;
I.M.,Potential
Wacoo A.P.,
application
Kort2019
R., of
Byaruhanga
International
lactic acid Y.B.,
starters
Journal
Muyanja
inof
theFood
C.
reduction
Contamination
of aflatoxin contamination in fermented so
Ahlberg S., Kärki
56663034000;57212132864;57212141157;7103166295;57191855982;
P., Kolmonen
Aflatoxin
M.,
M1Korhonen
binding
2019by
H.,
World
lactic
Joutsjoki
Mycotoxin
acid V.
bacteria
Journal
in milk

Wang J.P., Lin


57202341454;57201090731;57209806234;57209802310;57198965743;
L., Jiang Q.R.,
Probiotics
Huangand
W.L.,
clay
2019
Liudetoxifier
N.Indian protected
Journal of growth
Animal Sciences
performance and intestinal barrier of lambs fed diet co

Ezekiel C.N.,23018377400;57200967927;57057793600;55893821400;55334859800;57159823900;57208301030;5720096265
Ayeni K.I., High-throughput
Ezeokoli O.T., Sulyok
2019
sequence
M.,
Frontiers
Van
analyses
Wykin D.A.B.,
Microbiology
of bacterial
Oyedele
communities
O.A., Akinyemi
and O.M.,
multi-mycotoxin
Chibuzor-Onyema
profilingI.E.,
during
Adelek
pro
Abdelhamid7004856057;57206785221;16230090900;57207848549;
A.M., Salem
Is it
M.F.I.,
possible
El-Shebly
to detoxify
2019
A.A.,Egyptian
Sultan
aflatoxic
A.S.I.
Journal
aquafeed?
of Aquatic Biology and Fisheries

Wacoo A.P.,57053526200;35790309300;57205588414;57192156416;57053318400;6701854608;7004607886;
Mukisa I.M.,
Probiotic
Meemeenrichment
R., Byakika
2019and
S.,
Nutrients
Wendiro
reduction
D.,ofSybesma
aflatoxins
W.,
in Kort
a traditional
R. african maize-based fermented food

Chen Y., Li R.,


56486103500;57197828484;55387731700;57197836479;8855958100;35207500800;
Chang Q.,Lactobacillus
Dong Z., Yangbulgaricus
H.,2019
Xu C.Toxins
or lactobacillus rhamnosus suppresses nf-κb signaling pathway and protects ag
Sokoutifar R.,
57204557755;6508377827;53163142300;57204558820;
RazavilarDegraded
V., Anvar A.A.,
aflatoxin
Shoeiby
2018
M1 Journal
in
S. artificially
of Foodcontaminated
Safety fermented milk using Lactobacillus acidophilus and

Tahir N.I., Hussain


57204471691;37028707100;56300773100;27267961000;57053431100;57369865300;42760904400;
S., Javed
Nature
M.,ofRehman
aflatoxins:
2018
H., Shahzady
Their
Journal
extraction,
T.G.,
of Food
Parveen
analysis,
SafetyB.,and
Ali K.G.
control

Mahmood Fashandi
57190045072;57190049129;57203634349;
H.,The
Abbasi
detoxification
R., Mousavi
2018
ofKhaneghah
aflatoxin
Journal M1
ofA.Food
by Lactobacillus
Processing and
acidophilus
Preservation
and Bifidobacterium spp.: A review
Namvar Rad57203411288;6508377827;55888936600;15769124600;
M., Razavilar
Selected
V., Anvar
bio-physical
S.A.A.,
2018Akbari-Adergani
factors
Journalaffecting
of Food
B.the
Safety
efficiency of Bifidobacterium animalis lactis and Lactobacillu

Liu N., Ding 57198965743;35976020000;50263339700;57197747329;57197748085;57202341454;


K., Wang J.,Effects
Deng Q.,
of lactic
Gu K.,acid
Wang
2018bacteria
J.
Journaland
of smectite
Animal Physiology
after aflatoxin
and Animal
B1 challenge
Nutrition
on the growth performance, nu

Barukčić I., Bilandžić


56196118500;55889165000;7102521923;56595266200;56196372600;
N.,Reduction
Markov K.,inJakopović
aflatoxin
2018 M1
K.L.,
International
concentration
Božanić R.Journal
during
of Dairy
production
Technology
and storage of selected fermented milks

Liew W.-P.-P.,
57200647159;57193452928;57204624974;55598057100;
Nurul-Adilah
The binding
Z., Thanefficiency
L.T.L.,
2018
Mohd-Redzwan
and
Frontiers
interaction
in Microbiology
S. of Lactobacillus casei Shirota toward aflatoxin B1
Gomaa E.Z.,55034005700;57197735726;57197737981;
Abdelall M.F.,
Detoxification
El-Mahdy O.M.
of 2018
Aflatoxin
Probiotics
B1 by Antifungal
and Antimicrobial
Compounds
Proteins
from Lactobacillus brevis and Lactobacillus pa

Saladino F., 57006686200;57193527707;57172148600;17435122700;6602107652;7006949013;25227937100;


Posarelli E.,Influence
Luz C., Luciano
of probiotic
F.B.,
2018Rodriguez-Estrada
microorganisms
Journal of Food on
Composition
M.T.,
aflatoxins
MañesB1and
J., and
Meca
Analysis
B2G.bioaccessibility evaluated with a simula

Vila-Donat P.,
56431158100;7006199090;7006665185;7401770132;
Marín S.,ASanchis
reviewV.,
of Ramos
the mycotoxin
2018
A.J. Foodadsorbing
and Chemical
agents,
Toxicology
with an emphasis on their multi-binding capacity, for anim

Quattrini M.,
57196478475;7004550489;22936157800;23489308200;55908519700;35187857500;6603641528;57214976910;
Bernardi C.,
Functional
Stuknytėcharacterization
M., Masotti
2018 Food
F., Passera
ofResearch
Lactobacillus
A., International
Ricci plantarum
G., Vallone ITEM
L., De17215:
Noni I.,ABrasca
potential
M.,biocontrol
Fortina M.G.
agent of fun
Martínez M.P.,
57195636442;8971322500;15052994300;55257435200;8950477100;
González
Probiotic
Pereyracharacteristics
M.L., Fernandez
2018 Aquaculture
and
Juri
aflatoxin
M.G.,Research
Poloni
B1 binding
V., Cavaglieri
ability ofL.Debaryomyces hansenii and Kazaschtania e

Liu N., Wang57198965743;57196076791;57197747329;57197748085;57202341454;


J., Deng Q.,
Detoxification
Gu K., Wang of
J. 2018
aflatoxin
Livestock
B1 by lactic
Scienceacid bacteria and hydrated sodium calcium aluminosilicate in br

Hamad G.M.,
56677581000;54894287700;24576475000;57189757153;54580874300;
Taha T.H.,Supplementation
Hafez E.E., Ali S.H.,
2018
of Cerelac
El Sohaimy
Journal
baby
ofS.A.
the
foodScience
with yeast–probiotic
of Food and Agriculture
cocktail strains induces high potential for

AL-Ruwaili M.,
53982509400;57210171366;8922029500;57210169949;57210172659;35242300700;
Alkhalaileh
Reduction
N.I., Herzallah
of Aflatoxin
2018
S.M.,B1
Pakistan
Rawashdeh
residues
Veterinary
inA.,meat
Fataftah
Journal
and organs
A., Holley
of broiler
R. chickens by lactic acid bacteria
[No author name
[No author
available]
id
18th
available]
International2018
Multidisciplinary
InternationalScientific
Multidisciplinary
Geoconference
Scientific
SGEM
GeoConference
2018 Surveying Geology and

Florina R., Sofia


57196257803;57205180994;37089288600;56578476300;15070536800;
P., Lia R.,
Effect
Antoanela
of someC.,probiotic
Butnariu
2018 International
bacteria
M. on the
Multidisciplinary
reduction of aflatoxin
ScientificB1
GeoConference
production in Surveying
stored arabica
Geology
coffee
andb

Kuharić Z., Jakopović


57195360665;6507280772;57078628000;6505884869;6602383526;57200854311;55856293600;7102521923;
Z.,Removing
Čanak I., Frece
aflatoxin
J.,2018
Bošnir
M1Arhiv
from
J., Pavlek
za
milk
Higijenu
with
Z., Ivešić
native
RadaM.,
lactic
i Toksikologiju
Markov
acid bacteria,
K. centrifugation, and filtration
Tsogoeva F.N.,
57202302412;57192209939;57202650357;57202647749;57194624556;57202645145;57494509000;
Temiraev
Use
R.B.,
of antioxidant
Mamukaev2018
and
M.N.,
Asian
probiotic
Osikina
Journal
in
R.V.,
diets
ofTletseruk
Microbiology,
of layers
I.R.,
to Kokov
reduce
Biotechnology
T.N.,
the risk
Vasiliadi
of
and
aflatoxicosis
G.K.
Environmental Sciences

Huang W., Chang


57196346386;55801439700;56873539700;57193358014;8240256600;56704017700;56704116100;57202389414
J., Wang
Effect
P., of
Liuthe
C., combined
Yin Q.,
2018ZhuJournal
compound
Q., Lu of
F., Toxicological
Gao
probiotics
T. with
Sciences
mycotoxin–degradation enzyme on detoxifying afla

Adilah Z.N., 57202379432;57200647159;55598057100;35511562200;


Liew W.-P.-P.,
Effect
Redzwan
of HighS.M.,
Protein
2018
Amin
Diet
BioMed
I. and Probiotic
ResearchLactobacillus
Internationalcasei Shirota Supplementation in Aflatoxin B1-In

Marrez D.A.,56288678900;56192003200;57201378847;36480540200;
Shahy E.M.,
Detoxification
El-Sayed H.S.,
ofSultan
2018
Aflatoxin
Journal
Y.Y.B1 inofmilk
Biological
using lactic
Sciences
acid bacteria
Alrabadi N.I.,
55542544700;57200991287;46661943900;57200984563;
Al-Jubury Lactobacillus
E.M., Thalij K.M.,
rhamnosus
2018
HajeejJordan
J.M.
abilityJournal
of aflatoxin
of Biological
detoxification
Sciences

Assaf J.C., Atoui


57200273084;23501410100;15071327000;12786756200;6603654093;
A., Khoury
A comparative
A.E., Chokrstudy
A.,2018
Louka
ofBrazilian
procedures
N. Journal
for binding
of Microbiology
of aflatoxin M1 to Lactobacillus rhamnosus GG

Moretti A.F.,57190252590;57190250621;57196392570;57196391516;6602189336;57190376156;14619455600;
Gamba R.R.,
Incorporation
Puppo J., Malo
of Lactobacillus
2018
N., Gómez-Zavaglia
Journalplantarum
of FoodA.,
Science
and
Peláez
zeolites
and
Á.L.,
Technology
Golowczyc
in poultry feed
M.A.can reduce aflatoxin B1 levels

Damayanti E.,
57190665254;57190661672;57200304183;57096077100;16685017600;
Istiqomah
Characterization
L., Saragih J.E., of
Purwoko
2017
Lactic
IOPAcid
T.,
Conference
Sardjono
Bacteria Series:
as Poultry
Earth
Probiotic
and Environmental
Candidates with
Science
Aflatoxin B1 Binding Activ
Martinez M.P.,
57195636442;8971322500;35798972000;55257435200;25521943500;8950477100;
Gonzalez
Pediococcus
Pereyra M.L.,
acidolactici
Pena
2017G.A.,
Food
and
Poloni
Additives
Pediococcus
V., Fernandez
andpentosaceus
Contaminants
Juri G., isolated
Cavaglieri
- Part Afrom
Chemistry,
L.R.a rainbow
Analysis,
trout ecosystem
Control, Exposur
have

Hamad G.M.,
56677581000;37048145800;24576475000;
Zahran E.,The
Hafez
efficacy
E.E. of bacterial
2017 Journal
and yeasts
of Food
strains
Safety
and their combination to bind aflatoxin B1 and B2 in artifici

Liu N., Ding 57198965743;35976020000;57196076791;56063955100;57202341454;55650350900;


K., Wang J.Q.,
Detoxification,
Jia S.C., Wang
metabolism,
2017
J.P., Xu
Journal
T.S.andofglutathione
Animal Science
pathway activity of aflatoxin B1 by dietary lactic acid bact
Ma Z.X., Amaro
56086487100;57194870717;55825951700;35588129100;57146347400;6701745843;
F.X., Romero
The capacity
J.J., Pereira
of silage
2017
O.G.,inoculant
Journal
Jeong K.C.,
ofbacteria
Dairy
Adesogan
Science
to bind
A.T.aflatoxin B1 in vitro and in artificially contaminated co

Poloni V., Salvato


55257435200;57194334814;24071594400;55181243800;16639647200;8950477100;25925816700;
L., Pereyra
BakeryC.,by-products
Oliveira 2017
A.,based
Rosa
Food
C.,
feeds
and
Cavaglieri
borne-Saccharomyces
Chemical
L., Keller
Toxicology
K.M. cerevisiae strains with probiotic and antimycot

Taheur F.B.,57133187900;56721269700;56175443800;35072714000;18633819500;57219300292;
Fedhila K., Adsorption
Chaieb K., Kouidhi
of aflatoxin
2017
B., Bakhrouf
International
B1, zearalenone
A., Abrunhosa
Journal
and of
ochratoxin
L.Food Microbiology
A by microorganisms isolated from Kefir grains
Karazhyan R.,
23970434300;57201284792;57201288932;57201287769;44761544800;57212978001;
Sheyda I.S.,
Effect
Atash
of Lactobacillus
M.M.S., 2017
Tajjalirhamnosus
Journal
F.F., Mojtahedi
of Veterinary
(PTCCM.,
1637)
Sadegh
Research
on ruminal
M. detoxification of aflatoxin B1

Khaneghah 57203634349;54791865700;56578610300;
A.M., Chaves
Detoxification
R.D., Akbarirad
of 2017
aflatoxin
H. Current
M1 (AFM1)
Nutrition
in dairy
and Food
baseScience
beverages (Acidophilus milk) by using different typ

Ismail A., Levin


56643851900;55949635500;57220653976;57205636403;7402473492;35557654100;
R.E., Riaz
Effect
M., Akhtar
of different
S., Gong
2017
microbial
Y.Y.,
Food deconcentrations
Control
Oliveira C.A.F.on binding of aflatoxin M1 and stability testing

Moustafa M.,
54796802800;54894287700;55917479300;55545062200;56677581000;26324812800;55329337400;
Taha T., Elnouby
PotentialM.,
detoxification
El-Deeb
2017
N.,Biocell
Hamad
of aflatoxin
G., Abu-Saied
b2 using kluyveromyces
M.A., Alrumman
lactis
S. and saccharomyces cerevisiae integr
Yin H.-B., Kollanoor-Johny
56027335500;25926480100;6603674601;
Natural
A., Venkitanarayanan
approaches
2017
forFocus
controlling
K. on Aflatoxins
aflatoxins
Research
in agriculture

Li H., Duan C.,


57193138673;55286262800;55349762500;57192429339;55351370000;55714147700;13103770400;
Zhao Y., Gao
Reduction
L., Niuof
C.,aflatoxin
Xu 2017
J., Li S.
B1
PLoS
toxicity
ONE by Lactobacillus plantarum C88: A potential probiotic strain isolated

Sarlak Z., Rouhi


56536895000;56662884200;36716645500;35210663700;12792054500;12793070400;55962138200;
M., Mohammadi
Probiotic biological
R., Khaksar
2017
strategies
R.,
Food
Mortazavian
Control
to decontaminate
A.M., Sohrabvandi
aflatoxinS.,
M1 Garavand
in a traditional
F. Iranian fermented milk d
Nduti N., McMillan
56754322700;50162039100;6505956133;8601062400;53064238800;57212486716;56601800600;
A., Seney
Investigating
S., Sumarah
probiotic
2016
M., Njeru
yoghurt
International
P., to
Mwaniki
reduce
DairyM.,
an
Journal
aflatoxin
Reid G. B1 biomarker among school children in eastern K

Patra J.K., Das


24076666800;55994808400;6507743603;38663296800;
G., Paramithiotis
Kimchi andS., other
Shin H.-S.
2016
widelyFrontiers
consumedin Microbiology
traditional fermented foods of Korea: A review

Gacem M.A.,
57191619745;56989793100;
Ould El Hadj-Khelil
Toxicology,A. biosynthesis,
2016 Asian
bio-control
Pacific Journal
of aflatoxin
of Tropical
and newBiomedicine
methods of detection

Adibpour N.,57207944766;14022091000;57191281315;55758255700;
Soleimanian-Zad
Effect ofS.,storage
Sarabi-Jamab
time
2016and
Journal
M.,concentration
Tajalli
of Agricultural
F. of aflatoxin
Sciencem1
andon
Technology
toxin binding capacity of L. acidophilus in f
Saladino F., 57006686200;57172148600;36465287000;24512351900;25227937100;
Luz C., Manyes
In vitro
L., Fernández-Franzón
antifungal2016
activity
Foodof
M.,
Control
lactic
Meca acid
G.bacteria against mycotoxigenic fungi and their application in lo

Salah-Abbès26029752600;55819038800;52365038100;57203654333;6602076540;13407983200;
J.B., Jebali Immuno-physiological
R., Sharafi H., Noghabi
2016 alterations
Journal
K.A., Oueslati
of Immunotoxicology
from
R.,AFB1
Abbès
in S.
rats counteracted by treatments with Lactobacillus par

Padmaja P.B.,
57192096274;26639763200;
Periyar Selvam
Determination
S. of2016
antiaflatoxigenic
Biosciences effect
Biotechnology
of probiotic
Research
strainsAsia
in sorghum bicolour

Singh S.P., Sundar


57109501500;56512275900;7101901277;
K., Shetty
Aflatoxin
P.H. B1 binding
2016byJournal
microflora
of Pure
isolated
and Applied
from fermented
Microbiology
foods
Galarza-Seeber
56741290300;56017379700;10641656400;54784677200;10639544300;55497894000;57192809688;1063900350
R., Latorre
Leaky
J.D.,
GutBielke
and Mycotoxins:
L.R.,
2016Kuttappan
Frontiers
Aflatoxin
V.A.,
in Veterinary
Wolfenden
B1 does not
Science
A.D.,
increase
Hernandez-Velasco
gut permeability
X., in
Merino-Guzman
broiler chickensR., Vicent

Prathivadi Bayankaram
57189256239;26639763200;
Antifungal
P., Sellamuthu
and anti-aflatoxigenic
P.S.
2016 Toxin Reviews
effect of probiotics against Aspergillus flavus and Aspergillus parasitic

Nyamete F.A.,
57194390422;7004056222;6602002417;
Bennink Potential
M., Mugulaof lactic
J.K. 2016
acid fermentation
African Journal
in of
reducing
Food, Agriculture,
aflatoxin B1Nutrition
in Tanzania
andmaize-based
Developmentgruel

Ghislaine D.A.,
57192167099;57192155988;57192153073;57192156978;55340024200;36129795600;
Imene B.A.,
Influence
Zohraof
E.F.,
lactic
Samira
2016
ferments
M.,
DerBouziane
Pharmacia
on Aflatoxin
A.,Lettre
Mohammed
M1 in the industrial
B. manufacture of the Algerian Leben
Ansari F., Rezaei
22133462900;36873364300;16042465200;26647795100;
K., Khodaiyan
Optimisation
F., Rahmani
of aflatoxin
2016
A. Quality
B1 reduction
Assurance
in pistachio
and Safetynuts
of Crops
by kefir
andgrains
Foodsusing statistical experimental m

Marhamatizadeh
55213835100;57190405134;
M.H.,The
Goosheh
combined
S.R. effect
2016ofItalian
Thymus Journal
Vulgaris
of Food
extract
Science
and probiotic bacteria (lactobacillus acidophilus and b

Mohd Redzwan
55598057100;55222196400;57189269738;57192590124;
S., Jamaluddin
Probiotics
R.,as
Ahmad
Potential
2016
F.N.,Adsorbent
Probiotics,
Lim Y.-J. of
Prebiotics,
Aflatoxinand Synbiotics: Bioactive Foods in Health Promotion

Abbès S., Ben


13407983200;26029752600;55819038800;7004822781;6602076540;
Salah-Abbès
Interaction
J., JebaliofR.,aflatoxin
Younes
2016 Journal
B1
R.B.,
andOueslati
of
fumonisin
Immunotoxicology
R. B1 in mice causes immunotoxicity and oxidative stress: Possi
Tipu M.K., Saleem
57914159200;55626217900;36674030200;7102137044;57219477301;26322530300;9734001400;7201619518;5
U., Rehman
Protective
T., Aslam
role of2015
M.,
lactobacillus
Muhammad
Journal of
acidophilus
Animal
K., Hussain
and
against
Plant
K., Bukhari
aflatoxin
Sciences
N., B1-
Hassan
induced
S., Hussain
immunosuppression
Z., Faisal M., Ahmad B.

Aiko V., Mehta


55616623000;55617724100;
A. Occurrence, detection
2015 and
Journal
detoxification
of Biosciences
of mycotoxins

Giovati L., Magliani


10142265200;6603706027;35748535000;56358775800;7103105347;7005491896;
W.,AFM1
Ciociola
in milk:
T., Santinoli
Physical,
2015C.,Toxins
biological,
Conti S., Polonelli
and prophylactic
L. methods to mitigate contamination

Fan Y., Zhao55701492600;55646256100;56841115000;55816304900;56230050700;7404513304;55720227200;8533859600;


L., Ji C., Li X.,
Protective
Jia R., Xi effects
L., Zhang
2015
of J.,
Bacillus
Toxins
Ma Q.subtilis ANSB060 on serum biochemistry, histopathological changes and a
Ahlberg S.H.,
56663034000;57191855982;7103166295;
Joutsjoki V.,
Potential
Korhonen
of lactic
H.J. 2015
acid bacteria
International
in aflatoxin
Journalrisk
of Food
mitigation
Microbiology

Poloni V., Dogi


55257435200;14049799400;24071594400;15052994300;56640800600;55552784400;6701436326;8950477100;
C., Pereyra
Potentiation
C.M., Fernández
of the
2015
effect
JuriFood
M.G.,
of aAdditives
commercial
Köhler P.,
and
Rosa
animal
Contaminants
C.A.R.,
feedDalcero
additive
- Part
A.M.,
Amixed
Chemistry,
Cavaglieri
with different
Analysis,
L.R. probiotic
Control, yeast
Exposur
st

Salah-Abbès26029752600;13407983200;55819038800;35363966300;6602076540;
J.B., AbbèsPotential
S., Jebalipreventive
R., Haous
2015
Z.,
role
Journal
Oueslati
of lactic
ofR.Immunotoxicology
acid bacteria against Aflatoxin M1 immunotoxicity and genotoxicity
Okeke C.A.,57055359600;23018377400;39962196300;55893821400;57056418300;35726731100;36639205800;7003506571
Ezekiel C.N.,Bacterial
Nwangburuka
diversity
C.C.,
2015
andSulyok
mycotoxin
Frontiers
M., Ezeamagu
inreduction
Microbiology
C.O.,
during
Adeleke
maize R.A.,
fermentation
Dike S.K.,(steeping)
Krska R. for Ogi production

Serrano-Niño
55508408500;55499865400;56905462600;6603013016;55989802400;56013544800;7202282374;
J.C., Cavazos-Garduño
In vitro reduced
A., availability
Cantú-Cornelio
2015 LWT of aflatoxin
F., Gonzalez-Cordova
B1 and acrylamide
A.F., by
Vallejo-Cordoba
bonding interactions
B., Hernandez-Mendoza
with teichoic acidsA.

Da Silva J.F.M.,
55507028400;35113735200;8389995800;23005689600;56720000600;7005946096;55552784400;6603347120;5
Peluzio Use
J.M.,ofPrado
Probiotics
G., Madeira
2015
to Control
Scientific
J.E.G.C.,
Aflatoxin
World
Silva Production
M.O.,
Journal
De Morais
in Peanut
P.B., Grains
Rosa C.A., Pimenta R.S., Nicoli J.R.
Serrano-Niño
55508408500;55499865400;6603013016;55989802400;56013544800;7202282374;
J.C., Cavazos-Garduño
In vitro Study A.,
of the
González-Córdova
2014
Potential
JournalProtective
of Food
A.F.,Safety
Vallejo-Cordoba
Role of Lactobacillus
B., Hernández-Mendoza
Strains by Acrylamide
A.,Binding
García H.S.

Vosough P.R.,
55773973600;55391206100;55943378300;23970434300;
Sani A.M.,
InMehraban
vitro effectM.,
of Lactobacillus
Karazhyan
2014 Nutrition
R. rhamnosus
and FoodGG
Science
on reduction of aflatoxin B1

Zoghi A., Khosravi-Darani


23969589100;23969408200;12793070400;
Surface
K., Sohrabvandi
binding of2014
toxins
S. Mini-Reviews
and heavy metals
in Medicinal
by probiotics
Chemistry
Mohammadi
55391206100;55363235600;56005218100;
Sani A., Marhamati
Bio-detoxification
Z., Marhamatizade
2014
of aflatoxin
BioTechnology:
M.H.
M1 in kefir
Anusing
IndianLactobacillus
Journal casei

Jans D., Pedrosa


57222105335;57222113959;14023663400;7006781612;37115563500;
K., Schatzmayr
Mycotoxin
D.,reduction
Bertin2014
G.,inGrenier
Mycotoxin
animalB.diets
Reduction in Grain Chains

González Pereyra
8971322500;14049799400;56503857600;6603191036;57212697540;35083400400;22984344200;42962753600;
M.L., Genotoxicity
Dogi C., Torres
andLisa
2014
cytotoxicity
A., Journal
Wittouck
evaluation
ofP.,
Applied
Ortízof
M.,
Microbiology
probiotic
EscobarSaccharomyces
F., Bagnis G., Yaciuk
cerevisiae
R., Poloni
RC016:
L., Torres
A 60-day
A., subch
Dalce
Pleadin J., Markov
13407376300;7102521923;6505884869;24336504900;35316266700;
K., Frece
Bio-prevalence,
J., Vulić A., Perši
determination
2014N.Aflatoxins:
andFood
reduction
Sources,
of aflatoxin
OccurrenceB1 in
andcereals
Toxicological Effects

Sani A.M., Marhamati


55391206100;55363235600;56005218100;
Z.,
Aflatoxin
Marhamatizade
M1 detoxification
2014
M.H.BioTechnology:
in kefir using
AnLactobacillus
Indian Journal
acidophilus

Elsanhoty R.M.,
25631607400;53868358100;7006764090;15730074300;
Salam S.A.,
Detoxification
Ramadan of
M.F.,
2014
aflatoxin
Badr
Food
F.H.
M1Control
in yoghurt using probiotics and lactic acid bacteria
Lim S.-M., Ahn
36489940600;26641677500;
D.-H. Incubation conditions
2013and
Korean
physico-chemical
Journal of Microbiology
factors affecting aflatoxin B1 binding of lactic acid bacteria

Alidad M., Tajali


55918454700;55919325200;55391206100;
F., SaniDetoxification
A.M. of 2013
AFM1BioTechnology:
in yogurt using An
Lactobacillus
Indian Journal
casei

Sezer C., Güven


12243837400;7006151744;19337690600;12244082400;
A., OralDetoxification
N.B., Vatansever
of 2013
aflatoxin
L. Turkish
B1 byJournal
bacteriocins
of Veterinary
and bacteriocinogenic
and Animal Sciences
lactic acid bacteria

Hamidi A., Mirnejad


55840886600;55936169200;55542862500;55840339200;57201953430;55840671300;55840479000;5554262540
R.,The
Yahaghi
aflatoxin
E., Behnod
B1 isolating
2013
V.,Asian
Mirhosseini
potential
Pacificof
A.,
Journal
two
Amani
lactic
of S.,
Tropical
acid
Sattari
bacteria
Biomedicine
S., Darian E.K.
Fan Y., Zhao55701492600;55646256100;8533859600;55816304900;55815654800;35867628600;55720227200;56841115000
L., Ma Q., Li
Effects
X., ShiofH.,
Bacillus
Zhou 2013
T.,
subtilis
Zhang
FoodANSB060
J.,and
Ji C.Chemical
on growth
Toxicology
performance, meat quality and aflatoxin residues in bro

Elsanhoty R.M.,
25631607400;7006764090;36124503700;55946088300;8360160000;
Ramadan
Ability
M.F.,ofEl-Gohery
selected2013
microorganisms
S.S.,Food
Abol-Ela
Control
M.F.,
for Azeke
removing
M.A.aflatoxins invitro and fate of aflatoxins in contaminate

Bovo F., Corassin


23972243100;6508389444;23973211900;35557654100;
C.H., Rosim
Efficiency
R.E.,ofdeLactic
Oliveira
2013
AcidC.A.F.
Food
Bacteria
and Strains
Bioprocess
for Decontamination
Technology of Aflatoxin M1 in Phosphate Buffer Salin

Zuo R.-Y., Chang


33568483400;55801439700;8240256600;56873539700;56658991300;54797035000;56089338700;36027791800
J., Yin Q.-Q.,
Effect of
Wang
the P.,
combined
Yang
2013Y.-R.,
Food
probiotics
Wang
and Chemical
X.,
with
Wang
aflatoxin
Toxicology
G.-Q.,B1-degrading
Zheng Q.-H. enzyme on aflatoxin detoxification, broi
Vosough P.R.,
55773973600;55391206100;55774557800;23970434300;
Sani A.M.,
Assessing
Sangatash
theM.M.,
ability
2013
Karazhyan
of BioTechnology:
Lactobacillus
R. rhamnosus
An IndianGG
Journal
to bind aflatoxin B1 from contaminated cottonsee

Abbès S., Salah-Abbès


13407983200;26029752600;52365038100;55819038800;57203654333;6602076540;
J.B.,
Ability
Sharafi
of Lactobacillus
H., Jebali
2013R.,rhamnosus
Journal
Noghabiof K.A.,
Immunotoxicology
GAF01
Oueslati
to remove
R. AFM1 in vitro and to counteract AFM1 immunoto

Hope J. 54889913200;
A review of the mechanism
2013 The Scientific
of injury and
World
treatment
Journal approaches for illness resulting from exposure to w

Nikbakht Nasrabadi
55598098300;55222196400;55221755200;8076553200;55571918600;55598057100;
E., Reduction
JamaluddinofR.,
aflatoxin
Abdul
2013Mutalib
level
Journal
in M.S.,
aflatoxin-induced
of Applied
Khaza'ai
Microbiology
H., Khalesi
rats by S.,
theMohd
activity
Redzwan
of probiotic
S. Lactobacillus casei stra
Serrano-Niño
55508408500;55499865400;56013544800;7004428985;6603610067;14014168400;7202282374;
J.C., Cavazos-Garduño
Assessment ofA.,probiotic
Hernandez-Mendoza
2013 Food
strains
Control
ability A.,
to reduce
Applegate
the B.,
bioaccessibility
Ferruzzi M.G.,ofSan
aflatoxin
Martin-González
M1 in artificially
M.F., Garcí
conta

Corassin C.H.,
6508389444;23972243100;23973211900;35557654100;
Bovo F., Rosim
Efficiency
R.E.,ofOliveira
Saccharomyces
2013
C.A.F.
Food Control
cerevisiae and lactic acid bacteria strains to bind aflatoxin M1 in UHT skim

Friedman M.,
7403710129;8672437800;
Rasooly R.
Review of the inhibition
2013 Toxins
of biological activities of food-related selected toxins by natural compounds

Bagherzadeh
52363227500;16425960800;55398907600;22939237900;
Kasmani F.,
Aflatoxin
Karimi Torshizi
detoxification
2012
M.A.,Iranian
Allameh
potential
Journal
A.A.,
of lactic
Shariatmadari
of Veterinary
acid bacteria
Research
F. isolated from iranian poultry
Kabak B., Ozbey
55907550400;55256332200;
F. Aflatoxin M 1 in UHT
2012milk
Foodconsumed
Control in Turkey and first assessment of its bioaccessibility using an in vi

Pizzolitto R.P.,
22036529700;35797910000;6507380618;8950477100;6701436326;6507517771;
ArmandoEvaluation
M.R., Combina
of Saccharomyces
M.,
2012Cavaglieri
Journalcerevisiae
of
L.R.,
Environmental
Dalcero
strains
A.M.,
asScience
probiotic
Salvanoand
M.A.
agent
Health
with
- Part
aflatoxin
B Pesticides,
B1 adsorption
Food Contamin
ability f

Kabak B., Ozbey


55907550400;55256332200;
F. Assessment of the2012
bioaccessibility
Journal of Food
of aflatoxins
Composition
fromand
various
Analysis
food matrices using an in vitro digestion m

Armando M.R.,
35797910000;22036529700;14049799400;25635506500;36639949200;55257435200;6701436326;8950477100;
Pizzolitto
Adsorption
R.P., DogiofC.A.,
ochratoxin
2012
Cristofolini
Journal
A andA.,ofzearalenone
Merkis
Applied
C.,Microbiology
Poloni
by potential
V., Dalcero
probiotic
A.M., Saccharomyces
Cavaglieri L.R. cerevisiae strains a
Valencia-Quintana
55989753600;6506854384;23467680400;7401531497;56013749900;55577845100;
R., Sánchez-Alarcón
Preventive strategies
J.,2012
Tenorio
aimed
Toxicology
M.G.,
at reducing
Deng
and Environmental
Y.,the
Waliszewski
health risks
Health
S.M.,
of Aflatoxin
Valera
Sciences
M.Á.
B1

Pizzolitto R.P.,
22036529700;6507517771;6701436326;
Salvano Analysis
M.A., Dalcero
of fumonisin
A.M.
2012BInternational
1 removal byJournal
microorganisms
of Food Microbiology
in co-occurrence with aflatoxin B 1 and the natu

Armando M.R.,
35797910000;14049799400;55552784400;6701436326;8950477100;
Dogi C.A.,
Saccharomyces
Rosa C.A.R., Dalcero
cerevisiae
2012 Food
A.M.,
strains
Additives
Cavaglieri
and the
and
L.R.
reduction
Contaminants
of Aspergillus
- Part A parasiticus growth and aflatoxin B1
Bagherzadeh
52363227500;16425960800;55398907600;22939237900;
Kasmani F.,
A novel
Karimiaflatoxin-binding
Torshizi2012
M.A.,Poultry
Allameh
Bacillus
Science
A.,
probiotic:
Shariatmadari
Performance,
F. serum biochemistry, and immunological par

Kumar M., Verma


24284560600;56294622600;23393366000;56672243400;25960576900;35249756400;57212985655;
V., Nagpal
Anticarcinogenic
R., Kumar A.,
2012
effect
Behare
The
of probiotic
P.V.,
British
Singh
journal
fermented
B., Aggarwal
of nutrition
milkP.K.
and chlorophyllin on aflatoxin-B1-induced liver car

Kumar M., Verma


24284560600;56294622600;23393366000;56672243400;15845469500;25960576900;57216049501;5721298565
V., Nagpal
Effect R.,
of probiotic
Kumar A.,
2011
fermented
Gautam
GeneS.K.,
milkBehare
and chlorophyllin
P.V., Groveron
C.R.,
gene
Aggarwal
expressions
P.K. and genotoxicity during AFB
Fernández-Juri
15052994300;54785900500;6701436326;6603478521;
M.G., Muzzolón
Effect of J.A.,
acid Dalcero
lactic
2011
bacteria
A.M.,
Mycotoxin
Magnoli
isolated
Research
C.E.
from faeces of healthy dogs on growth parameters and aflatoxin B

El Khoury A.,15071327000;23501410100;38962150900;
Atoui A., Yaghi
Analysis
J. of aflatoxin
2011M1Food
in milk
Control
and yogurt and AFM1 reduction by lactic acid bacteria used in Lebanes

Hathout A.S.,
36910157000;57190830831;16834760700;26662036900;7101761439;8087610600;
MohamedAbility
S.R., El-Nekeety
of Lactobacillus
2011
A.A., casei
Toxicon
Hassanand
N.S.,
Lactobacillus
Aly S.E., Abdel-Wahhab
reuteri to protect
M.A.against oxidative stress in rats fed afla

Gao X., Ma 57209148581;8533859600;55646256100;37761828500;39661391700;56841115000;


Q., Zhao L., Isolation
Lei Y., Shan
of Bacillus
Y., Ji C.
2011
subtilis:
European
Screening
Food for
Research
aflatoxins
andB1,
Technology
M1, and G1 detoxification

Hernandez-Mendoza
56013544800;6603013016;55989802400;7202282374;
A.,Effect
González-Córdova
of oral supplementation
2011
A.F.,
Journal
Vallejo-Cordoba
of
of Basic
Lactobacillus
Microbiology
B., Garcia
reuteri
H.S.
in reduction of intestinal absorption of aflatoxin
Dogi C.A., Armando
14049799400;55176329500;55547133071;6506058808;16639647200;6701436326;8950477100;
R., Saccharomyces
Ludueña R., de Moreno
cerevisiae
2011 Food
destrains
LeBlanc
Additives
retain
A., and
Rosa
their
Contaminants
C.A.R.,
viability
Dalcero
and- aflatoxin
Part
A., Cavaglieri
A B1 binding
L. ability under gastrointe

Jard G., Liboz


55082811700;6506378924;7005378155;55499745000;6701842592;
T., Mathieu
Review
F., Guyonvarch
of mycotoxin
2011
A., Lebrihi
reduction
Food Additives
A. in foodand
andContaminants
feed: from prevention
- Part A in the field to detoxification by adso

Guan S., Zhou


7101750446;7402989468;56553540500;56189442400;17344349500;7408528418;
T., Yin Y.,Microbial
Xie M., Ruan
strategies
Z., Young
2011
to World
control
J.C. Mycotoxin
aflatoxins Journal
in food and feed
Mykkänen H.,
7003915985;9242677500;6603690577;
Gratz S.W.,Applied
El-Nezami
Studies
H. with
2010Probiotics:
ProbioticsFundamentals
and Health Claims
for Meeting the Health Claims

Markov K., Frece


7102521923;6505884869;35110351800;57503384600;6603306107;
J., Čvek
Aflatoxin
D., Lovrić
M1N.,
in Delaš
raw
2010
milk
F.Mljekarstvo
and binding of aflatoxin by lactic acid bacteria [Aflatoksin M1 u sirovom mlije
Rawal S., Kim
29567449700;34770378900;57211953905;
J.E., Coulombe
Aflatoxin
R. B1 in poultry:
2010 Research
Toxicology,
in Veterinary
metabolismScience
and prevention

Oluwafemi F.,
6504080751;55338811500;7005269281;57220297133;36553933300;
Kumar M.,
Bio-detoxification
Bandyopadhyay 2010
of
R.,aflatoxin
Ogunbanwo
Toxin Reviews
B1 inT.,
artificially
Ayanwande contaminated
K.B. maize grains using lactic acid bacteria

Chelule P.K.,6506961429;35756404900;57224291913;6602776482;
MbongwaLactic
H.P., Carries
acid fermentation
S., Gqaleni
2010 Food
N.
improves
Chemistry
the quality of amahewu, a traditional South African maize-based po

Topcu A., Bulat


56074520800;35749606800;35763103800;6603211547;
T., Wishah
Detoxification
R., BoyacI I.H.
of 2010
aflatoxin
International
B1 and patulin
Journal
by of
Enterococcus
Food Microbiology
faecium strains
Wu Q., Jezkova
34769157200;24764426800;8935299700;34768801200;10539549300;9745735500;
A., YuanBiological
Z., Pavlikova
degradation
L., 2009
Dohnal
of
Drug
V.,
aflatoxins
Kuca
Metabolism
K. Reviews

Siruguri V., Ganguly


6603868712;35322202200;55417853900;
C., Utilization
Bhat R.V. of mouldy
2009sorghum
African Journal
and Cassia
of Biotechnology
tora through fermentation for feed purposes

Hernandez-Mendoza
56013544800;6602602238;7202282374;
A.,Key
Guzman-De-Peña
role of teichoic
2009
D.,
acids
Journal
Garcia
on aflatoxin
H.S.
of Applied
B1 binding
Microbiology
by probiotic bacteria

Kabak B., Brandon


55907550400;8843143600;6602281131;8050060300;7006376241;
E.F.A.,
Effects
Var I.,ofBlokland
probiotic
2009
M.,
bacteria
Sips
Journal
A.J.A.M.
onof
the
Environmental
bioaccessibility
Science
of aflatoxin
and Health
B1 and
- Part
ochratoxin
B Pesticides,
A using
Food
an in
Contamin
vitro di
Hernandez-Mendoza
56013544800;7202282374;8745046300;
A.,Screening
Garcia H.S.,
of Lactobacillus
Steele
2009J.L.Food
casei
andstrains
Chemical
for their
Toxicology
ability to bind aflatoxin B1

Fazeli M.R.,57210985195;25957688000;25957707100;13610912000;6507598749;9133175000;25957804200;57217973734;
Hajimohammadali
AflatoxinM.,
B1Moshkani
binding
2009capacity
A.,
Journal
Samadi
ofofautochthonous
N.,
FoodJamalifar
Protection
H.,strains
Khoshayand
of lacticM.R.,
acid bacteria
Vaghari E., Pouragahi S.
Carvajal M.7006369561;
Aflatoxin-DNA adducts
2008 ACS
as biomarkers
SymposiumofSeries
cancer: Nature, formation, kinds of AF-DNA adducts, methodo

Khanafari A.,
17434722500;17435937900;17434993700;23034949500;
Soudi H., Miraboulfathi
An in vitro investigation
M.,2007
OsbooPakistan
of
R.K.
aflatoxin
Journal
B1 biological
of Biological
control
Sciences
by Lactobacillus plantarum
Gratz S., Wu9242677500;57220847327;6603690577;7005331813;7003915985;7402096074;
Q.K., El-Nezami
Lactobacillus
H., Juvonen
rhamnosus
2007
R.O., Applied
Mykkänen
strain and
GGH.,
reduces
Environmental
Turneraflatoxin
P.C. Microbiology
B1 transport, metabolism, and toxicity in Caco-2 c

Bueno D.J., 7004849633;7003550411;22036529700;6507517771;16484405700;


Casale C.H.,Physical
Pizzolitto
adsorption
R.P., Salvano
2007
of aflatoxin
Journal
M.A., Oliver
ofB1Food
byG.lactic
Protection
acid bacteria and Saccharomyces cerevisiae: A theoretical

Kabak B., Dobson


55907550400;57203087843;6602281131;
A.D.W.,
Strategies
Var I. to prevent
2006mycotoxin
Critical Reviews
contamination
in Food Science
of food and
and Nutrition
animal feed: A review

Gratz S., Täubel


9242677500;6507850955;7005331813;56186665400;7402096074;7003915985;6603690577;
M., Juvonen
Lactobacillus
R.O., Viluksela
rhamnosus
2006M.,Applied
Turner
strain and
GG
P.C.,
modulates
Environmental
Mykkänenintestinal
H., Microbiology
El-Nezami
absorption,
H. fecal excretion, and toxicily of afla
El-Nezami H.S.,
6603690577;6505944468;55265022900;9237388600;14054110400;7006341181;7005331813;7102912002;6701
Polychronaki
Probiotic
N.N.,
supplementation
Ma J.,2006
Zhu H.,
American
Ling
reduces
W.,Journal
Salminen
a biomarker
of Clinical
E.K.,for
Juvonen
Nutrition
increased
R.O.,risk
Salminen
of liverS.J.,
cancer
Poussa
in young
T., Mykkänen
men fromH.M
So

Gratz S., Mykkänen


9242677500;7003915985;6603690577;
H., Aflatoxin
El-NezamiB1
H.binding
2005byJournal
a mixture
of Food
of Lactobacillus
Protection and Propionibacterium: In vitro versus ex vivo

Donaldson M.S.
7103330819;
Nutrition and cancer:
2004ANutrition
review ofJournal
the evidence for an anti-cancer diet

Gratz S., Mykkänen


9242677500;7003915985;7005440286;7005331813;7102912002;6603690577;
H., Intestinal
Ouwehandmucus
A.C., Juvonen
alters
2004 Applied
theR.,ability
Salminen
andofEnvironmental
probiotic
S., El-Nezami
bacteria
Microbiology
H. to bind aflatoxin B1 in vitro
Var I., Kabak6602281131;55907550400;
B. Removal of aflatoxins
2004by
Archiv
viablefurand
Lebensmittelhygiene
heat-killed lactic acid bacteria and bifidobacteria

Kabak B., Var


55907550400;6602281131;
I. Binding of aflatoxin
2004
M1Milchwissenschaft
by Lactobacillus and Bifidobacterium strains

Mishra H.N.,55539634000;55865801100;
Das C. A Review on Biological
2003 Critical
ControlReviews
and Metabolism
in Food Science
of Aflatoxin
and Nutrition

Surono I.S. 37069818500;


In vitro probiotic 2003
properties
Asian-Australasian
of indigenous Journal
dadih lactic
of Animal
acid bacteria
Sciences
Khajarern J.M.,
6506205790;6506181130;57206509599;55870742000;
Khajarern
Effects
S., Moon
of dietary
T.H., 2003
Lee
supplementation
J.H.
Asian-Australasian
of fermented
Journalchitin-chitosan
of Animal Sciences
(FERMKIT) on toxicity of mycotoxin in

Byun J.R., Yoon


7102897468;7402126645;
Y.H. Binding of Aflatoxin
2003G1,Asian-Australasian
G2 and B2 by Probiotic
Journal
Lactobacillus
of Animal Sciences
spp.

Štyriak I., Conková


7003865149;55889915500;
E. Microbial binding2002
and biodegradation
Veterinary and of
Human
mycotoxins
Toxicology

Turbic A., Ahokas


33768138300;7006308329;6602356857;
J.T., Haskard
SelectiveC.A.
in vitro 2002
binding
Food
of dietary
Additives
mutagens,
and Contaminants
individually or in combination, by lactic acid bacteria
Haskard C.A.,
6602356857;6603690577;6701573026;7102912002;7006308329;
El-NezamiSurface
H.S., Kankaanpää
Binding of2001
Aflatoxin
P.E.,Applied
Salminen
B1 and
byS.,
Lactic
Environmental
Ahokas
AcidJ.T.
Bacteria
Microbiology

Peltonen K.,7005515367;6603690577;6602356857;7006308329;7102912002;
El-Nezami Aflatoxin
H., Haskard
B1 C.,
binding
Ahokas
2001
byJournal
J.,
dairy
Salminen
strains
of Dairy
S.of lactic
Scienceacid bacteria and bifidobacteria

Peltonen K.D.,
7005515367;6603690577;7102912002;7006308329;
El-Nezami
Binding
H.S., Salminen
of aflatoxin
2000
S.J.,B1Ahokas
Journal
by probiotic
J.T.
of thebacteria
Science of Food and Agriculture

Haskard C., 6602356857;57492511200;7006308329;


Binnion C., Factors
Ahokas affecting
J. 2000
the sequestration
Chemico-Biological
of aflatoxin
Interactions
by Lactobacillus rhamnosus strain GG

Oatley J.T., Rarick


6602612888;57210661505;7102392951;7006196730;
M.D.,Binding
Ji G.E., of
Linz
aflatoxin
J.E. 2000
B1 Journal
to bifidobacteria
of Food Protection
in vitro
Kankaanpää6701573026;6602726399;6603690577;7006308329;7102912002;
P., Tuomola
Binding
E., El-Nezami
of aflatoxin
H.,
2000
Ahokas
B1 Journal
alters
J., the
Salminen
of adhesion
Food Protection
S.J. properties of Lactobacillus rhamnosus strain GG in a Caco-2

Lankaputhra6603120845;7401823907;
W.E.V., Shah
Antimutagenic
N.P. properties
1998 Mutation
of probiotic
Research
bacteria
- Fundamental
and of organic
and acids
Molecular Mechanisms of Mutagenesis

El-Nezami H.,
6603690577;6701573026;7102912002;7006308329;
Kankaanpää
Physicochemical
P., Salminen S.,
alterations
1998
Ahokas
Journal
J.enhance
of Foodthe
Protection
ability of dairy strains of lactic acid bacteria to remove aflato

Bolognani F.,
15850161100;6602187738;23129267700;
Rumney C.J.,
Influence
Rowland
of carcinogen
I.R. 1997 Food
binding
andby
Chemical
lactic acid-producing
Toxicology bacteria on tissue distribution and in vivo mut
Thyagaraja N.,
6506689695;57192966661;
Hosono Binding
A. properties
1994
of lactic
Foodacid
andbacteria
Chemicalfrom
Toxicology
'Idly' towards food-borne mutagens

Luchese R.H.,
6507224672;36829732500;
Harrigan Growth
W.F. of, and aflatoxin
1990 Journal
production
of Applied
by Aspergillus
Bacteriology
parasiticus when in the presence of either Lactococ

MARSHALL 7402186443;7006464413;
D.L., BULLERMAN
Antimicrobial
L.B. Activity
1986ofJournal
Sucrose
of Fatty
Food Acid
Science
Ester Emulsifiers
Abstract
Lactobacilli are probiotics with Aflatoxin (AF) detoxification ability, found in fermented products, GIT of animals
and environment. Purpose of this study was to investigate the ability of broiler isolates of Lactobacillus against
Aflatoxin B1 (AFB1). For this purpose, 5 isolates of Lactobacillus from broiler gut were incubated with 100 ppb
AFB1 in aqueous environment and effect of different parameters (cell fractions, time, temperature, pH) on
detoxification was determined by HPLC. The ameliorative effect of Lactobacillus salivarius (LS) against AFB1 was
studied in broiler. The results revealed that LS (CR. 4) showed the best results (in vitro) as compared to other
isolates (L. salivarius (CR. 3, CR, 4), L. agilis (CE. 2.1, CE. 3.1) and L. crispatus (CE. 28). Cell debris of CR. 4 showed
significantly higher detoxification (P<0.05). Maximum amount of AFB1 was detoxified at 30°C (97%), pH 4.0
(99%) and 6 h (99.97%). In vivo study showed that AFB1 decreased weight gain (1,269 ± 0.04 gm/ bird), feed
consumed (2,161 ± 0.08 gm/ bird), serum total protein (2.42 ± 0.34 gm/ dl), serum albumin (0.5 ± 0.2 2 gm/dl)
and antibody titer (4.2 ± 0.83). Liver function enzymes were found (alanine transaminase (ALT): 32 ± 10.7 U/L)
and aspartate transaminase (AST): 314.8 ± 27 U/L) elevated in AFB1 fed broilers. Treatment with 1% LS not only
decreased the toxic effects of AFB1 (group D) but also improved the overall health of broilers due to its probiotic
effects (p<0.05) as compared to control negative (group A). The detoxification ability of LS was better than
Lactic acid bacteria (LAB) play many function roles during the preparation food; one of those roles is to remove
or reduce mycotoxins from contaminated food. Therefore, this study aimed to study the impact of five strains
from LAB (Lactobacillus rhamnosus, Lactobacillus plantarum, Bifidobacterium bifidum, Streptococcus
thermophilus and Lactobacillus reuteri) to reduce aflatoxin B1 (AFB1) during manufacturing Fino bread. Also, this
study has been extended to evaluate the qualities and characteristics of the Fino bread manufactured by treated
wheat flour by LAB cells. The data reflected that the percentages of reduction of AFB1 after mixing ingredients
were 9.7, 8.5, 7.04, 7.4 and 5.5% with addition L. rhamnosus, L. plantarum, Bifidobacterium bifidum, Str.
Thermophilus and L. reuteri, respectively. Moreover, the results indicated that the addition of L. rhamnosus and
L. plantarum cells given the highest percentage of removal AFB1 after fermentation stage were 60.5 and 54.25%,
respectively, while the lowest reduction of AFB1 recorded with the addition of L. reuteri cells was 42.25%. AFB1
reduction has reached 100% in blends treated with L. rhamnosus and L.plantarum cells in the final bread. The
results indicated that the increase in water absorption and the dough development time as well as dough
Featured Application: The current study provides applicable results for enhancing food safety by showing that
certain starter lactic acid bacteria have the potential to mitigate background level mycotoxin risks. This is of
crucial importance in the current global situation, where the mycotoxin contamination of foodstuffs is emerging
due to climate change. The results of this study provide a basis for easily accessible technologies to also ensure
the availability of safer and nutritious food to low-income countries. At the same time as the strong ambition to
improve sustainability and the healthiness of food systems through a transition towards a more plant-based diet,
climate change is increasing the risk of plant diseases. Consequently, mycotoxigenic fungi have become a food
safety issue of major importance. A variety of strategies to suppress fungal growth in the pre- and postharvest
stages of plant production have been established, and the potential of various biological methods has been
assessed to ensure food safety. Of the various food microbes, lactic acid bacteria are known for their capacity to
suppress the growth of toxigenic fungi and adsorb free mycotoxins. The current study showed that lactic acid
fermentation could mitigate aflatoxin risk in plant-based foods through a reduction in free aflatoxin B1. In line
with previous studies, in which Lactiplantibacillus plantarum (formerly Lactobacillus plantarum) was shown to
reduce the level of free aflatoxin B1 in vitro, L. plantarum was shown to achieve up to a 90% reduction in free
aflatoxin B1 in food fermentation. The results showed that traditional lactic acid fermentation, using L.
plantarum as the starter strain, could be applied to mitigate aflatoxin B1 contamination risk in proteinaceous
This study assessed the removal of aflatoxin M1 (AFM1) and ochratoxin A (OTA) from artificially contaminated
whole UHT milk and red grape juice, respectively, using biofilms from Lactobacillus rhamnosus GG. Using ELISA,
the level of AFM1 and OTA removal from beverages was determined depending on various factors. Biofilms of
various ages demonstrated varying degrees of AFM1 removal capacity from phosphate-buffered saline (PBS).
Different levels of AFM1 contaminated whole UHT milk (0.1, 0.2, and 1 μg/L) and OTA contaminated red grape
juice (2 and 4 μg/L) were tested in the detoxification process. The binding ability of mycotoxins was improved by
increasing the biofilm surface area up to 70 cm2. L. rhamnosus GG biofilm was effective in removing mycotoxins
within a short contact time ranging from 1 to 10 min. The proportion of bound AFM1 and OTA by L. rhamnosus
GG biofilm was 64.6 and 98.3% respectively. A new machine has been proposed and used as a trial for
Dairy products are rich sources of vitamins, proteins and calcium that are vital to the human body. Aflatoxin M1
is a hydroxylated metabolite of aflatoxin B1. The presence of this toxin in milk has caused serious concern among
nutritionists. Consumption of aflatoxin-contaminated milk (AFM1) may lead to serious health problems in
humans. AFM1 causes various cancers such as liver cancer, damage to the nervous and DNA, as well as
mutagenicity and teratogenicity. In reducing the amount of aflatoxin in the product, with does not affect the
milk quality is of particular importance. Absorption and restructuring methods such as yeast, lactic acid
(bacteria), enzymes, ozon, and cold plasma have been used for this purpose. In this study, they have been
Today, a few hundred mycotoxins have been identified and the number is rising. Mycotoxin detoxification of
food and feed has been a technically uphill task for the industry. In the twenty-first century, the public demand is
healthy food with minimum use of chemicals and preservatives. Among all the fungal inhibition and mycotoxin
detoxification methods so far developed for food, biopreservation and biodetoxification have been found safe
and reliable. Nowadays, lactic acid bacteria (LAB) are of great interest as biological additives in food owing to
their Generally Recognized as Safe (GRAS) classification and mycotoxin detoxification capability. The occurrence
of fungul growth in the food chain can lead to health problems such as mycotoxicosis and cancer to humans due
to producing mycotoxins such as aflatoxins. Biopreservation is among the safest and most reliable methods for
inhibition of fungi in food. This review highlights the great potential of LAB as biodetoxificant by summarizing
various reported detoxification activities of LAB against fungal mycotoxins released into foods. Mechanisms of
2-s2.0-85122651501
The study assessed the influence of rapeseed meal (RSM) fermented using Bacillus subtilis 87Y on the feed
microbiota, intestinal microbiota, blood biochemical parameters, and content of minerals in the blood plasma
and faeces of piglets. Modulation of the microbial composition of feed containing fermented rapeseed meal
(FRSM) and of the faeces of pigs consuming it was observed. There was a significant increase in the number of
lactic acid bacteria (LAB) and a decrease in the total number of coliforms and Clostridium perfringens in the
faeces of animals from the experimental groups. FRSM in the diet of piglets was shown to improve the mineral
balance by increasing the levels of P, Ca, and Mg in the blood plasma and reducing their amount in the faeces. A
beneficial effect on parameters of protein and lipid metabolism was also noted, resulting in an increase in the
levels of total protein (TP) and albumins (ALB) and a reduction in triacylglycerols (TG) and low-density lipoprotein
(LDL) cholesterol in the blood plasma of the piglets. The research results indicate that the presence of FRSM in
Scopus
Food contamination with aflatoxin is one of the most critical concerns of health professionals. One of the best
ways to reduce aflatoxin content in food is probiotics. Therefore, this study was performed to isolate
Bifidobacterium from the chick's intestine; evaluate its probiotic activities and its application with
Polyvinylpyrrolidone (PVP) to reduce aflatoxin B1 (AFB1) in the medium were investigated. Samples were
isolated from the chick's intestine, and Bifidobacterium was isolated and identified by biochemical and molecular
methods. Next, the potential probiotic characterization was assessed. Afterwards, the effect of selected isolate
and PVP on reducing AFB1 in the medium was studied using ELISA and HPLC. Biochemical and molecular
evaluations indicated isolation of Bifidobacterium bifidum strain from chick's intestine. One of the B. bifidum
strains was selected for the next steps, which showed potential probiotic characterization and the ability to
reduce the concentration of AFB1 in the medium (50% reduction). When used in combination with PVP showed
synergistic effects in reducing the concentration of AFB1 from the medium (up to 90%). In the conclusion, it was
2-s2.0-85134238712
Aflatoxins B1 (AFB1), deoxynivalenol (DON) and zearalenone (ZEA) are the three most prevalent mycotoxins,
whose contamination of food and feed is a severe worldwide problem. In order to alleviate the toxic effects of
multi-mycotoxins (AFB1 + DON + ZEA, ADZ) on inflammation and apoptosis in swine jejunal epithelial cells (IPEC-
J2), three species of probiotics (Bacillus subtilis, Saccharomyces cerevisiae and Pseudomonas lactis at 1 × 105
CFU/mL, respectively) were mixed together to make compound probiotics (CP), which were further combined
with 400 μg/mL of glycyrrhinic acid (GA) to make bioactive materials (CGA). The experiment was divided into
four groups, i.e., the control, ADZ, CGA and ADZ + CGA groups. The results showed that ADZ decreased cell
viability and induced cytotoxicity, while CGA addition could alleviate ADZ-induced cytotoxicity. Moreover, the
mRNA expressions of IL-8, TNF-α, NF-Κb, Bcl-2, Caspase-3, ZO-1, Occludin, Claudin-1 and ASCT2 genes, and
protein expressions of TNF-α and Claudin-1 were significantly upregulated in ADZ group; while the mRNA
abundances of IL-8, TNF-α, NF-Κb, Caspase-3, ASCT2 genes, and protein expressions of TNF-α and Claudin-1 were
significantly downregulated in the ADZ + CGA group. In addition, the protein expressions of COX-2, ZO-1, and
ASCT2 were significantly downregulated in the ADZ group, compared with the control group; whereas CGA co-
In order to alleviate the toxic effects of aflatoxins B1 (AFB1) on inflammatory responses in the intestine, liver,
and kidney of broilers, the aflatoxin B1-degrading enzyme, montmorillonite, and compound probiotics were
selected and combined to make a triple-action compound mycotoxin detoxifier (CMD). The feeding experiment
was divided into two stages. In the early feeding stage (1–21 day), a total of 200 one-day-old Ross broilers were
randomly divided into four groups; in the later feeding stage (22–42 day), 160 broilers aged at 22 days were
assigned to four groups: Group A: basal diet (4.31 μg/kg AFB1); Group B: basal diet with 40 μg/kg AFB1; Group C:
Group A plus 1.5 g/kg CMD; Group D: Group B plus 1.5 g/kg CMD. After the feeding experiment, the intestine,
liver, and kidney tissues of the broilers were selected to investigate the molecular mechanism for CMD to
alleviate the tissue damages. Analyses of mRNA abundances and western blotting (WB) of inflammatory factors,
as well as immunohistochemical (IHC) staining of intestine, liver, and kidney tissues showed that AFB1
aggravated the inflammatory responses through NF-κB and TN-α signaling pathways via TLR pattern receptors,
while the addition of CMD significantly inhibited the inflammatory responses. Phylogenetic investigation showed
that AFB1 significantly increased interleukin-1 receptor-associated kinase (IRAK-1) and mitogen-activated protein
2-s2.0-85140622988
The present study aimed to evaluate the effects of new Lactobacillus plantarum strain isolated from dairy
products as well as chitosan nanoparticles on reducing aflatoxin B1 (AFB1) toxicity In vitro. After collection and
preparation of yogurt, cheese, milk, and whey products, lactic acid bacteria (LABs) were isolated and identified
using biochemical and molecular methods. pH, bile, and salt tolerance tests were used to measure probiotic
activity. Then, the antimicrobial activity of LABs against gastrointestinal pathogens was studied. The strain
isolated from cheese (C1) was selected as the appropriate strain and antibiotic susceptibility test was performed
for this strain. Then, the effect of C1 isolate and chitosan nanoparticles on reducing aflatoxin B1 (AFB1) in the
medium was studied by measuring AFB1 using the enzyme-linked immunosorbent assay (ELISA) and high-
performance liquid chromatography (HPLC). The results of biochemical evaluations indicated the separation of
different strains of L. plantarum. Antimicrobial activity test showed extensive antimicrobial activity of C1 isolate.
The results showed that this strain has good probiotic activities. This strain was shown to be resistant to
erythromycin, fusidic acid, gentamicin, kanamycin, nalidixic acid, neomycin, ofloxacin, and vancomycin
antibiotics. C1 strain together with chitosan nanoparticles was able to reduce AFB1 in the medium and, when
both were used simultaneously, a synergistic effect was seen in reducing AFB1 from the medium. Based on the
2-s2.0-85138143851
The current research was designed to examine the protective effect of probiotic-fortified ration against aflatoxin
B1 (AFB1) toxicity and its residual level in broilers’ edible tissues. Sensitive high-performance liquid
chromatography coupled with a fluorescence detector (HPLC-FLD) was used for measuring the toxin. Ninety, one-
day-old Cobb chicks were allocated into three equal groups (n=30) with three replicates per group. The first
control group (G1) was fed a balanced basal diet only and the second group (G2) received AFB1 (2 mg/ kg basal
diet), while the third group (G3) received a combination of AFB1 (2 mg/kg basal diet) with Saccharomyces
cerevisiae (SC; 1.5 g/Kg basal diet). Experimental birds were monitored for 6 weeks, their growth performance
was then compared. AFB1 residue was assessed in the meat and liver sample. AFB1 resulted in a significant
(P<0.05) reduction of growth performance parameters such as body weight and carcass yield in comparison to
the control and SC supplemented groups. Moreover, AFB1 residue significantly (P<0.05) diminished in SC
fortified group when compared with the AFB1 group. In conclusion, probiotics such as Saccharomyces cerevisiae
Heavy metals and mycotoxins in foodstuffs are one of the major concerns of our world nowadays. Food
decontamination with the help of microbial biomass is a cheap, easy, efficient and green method known as
bioremoval. Probiotics are able to reduce the availability of heavy metals and toxins in food products. The
purpose of this review is to summarize the probiotics and potential probiotics' interesting role in food bio-
decontamination. After a brief glance at the definition of potential probiotic strains with bioremoval ability, LABs
(lactic acid bacteria) are described as they are the most important groups of probiotics. After that, the role of the
main probiotic and potential probiotic strains (Bacillus, Lactobacillus, Lactococcus, Enterococcus,
Bifidobacterium, Pediococcus, Propionibacterium, Streptococcus and Saccharomyces cerevisiae) for heavy
metals and mycotoxins bioremoval are described. Additionally, the bioremoval mechanism and the effect of
some factors in bioremoval efficiency are explained. Finally, the investigations about probiotic and contaminant
Aflatoxins are known for their high toxicity, and their presence in food is associated with a high health risk. The
levels were set for aflatoxin in different foodstuffs to protect public health. Various chemical, physical, and
biological methods have been reported for decontamination and detoxifying aflatoxins. Among biological
methods, probiotics have an important role in the decontamination of aflatoxins. At the same time, some
emerging food processing technologies showed promising results in aflatoxin degradation. This article reviewed
significant studies of the last four years that investigated the effects of probiotics on the degradation rate of
aflatoxins. Discussions on the possible contribution of emerging technologies along with probiotics are also
provided. The results showed that the bioavailability of aflatoxins was reduced by adsorption or binding to
probiotics’ cell walls, which increased their excretion rate. Besides, the amount of aflatoxin absorbed by
probiotics depends on several parameters, including the complexity of the food matrix. Therefore, probiotics can
email: yuthana.p@cmu.ac.th
Scopus
Obesity and its comorbidities are humans’ most prevalent cardio-metabolic diseases worldwide. Recent
evidence has shown that chronic low-grade inflammation is a common feature in all highly prevalent chronic
degenerative diseases. In this sense, the gut microbiota is a complete ecosystem involved in different processes
like vitamin synthesis, metabolism regulation, and both appetite and immune system control. Thus, dysbiosis has
been recognised as one of the many factors associated with obesity due to a predominance of Firmicutes, a
decrease in Bifidobacterium in the gut, and a consequent short-chain fatty acids (SCFA) synthesis reduction
leading to a reduction in incretins action and intestinal permeability increase. In this context, bacteria, bacterial
endotoxins, and toxic bacterial by-products are translocated to the bloodstream, leading to systemic
Importance of the work: The contamination of Aspergillus section Flavi producing aflatoxins during ensiling is a
main problem resulting in corn silage spoilage. Objectives: To evaluate the antagonistic activities of yeasts and a
lactic acid bacterium against A. flavus. Materials &amp; Methods: Individual yeast culture, mixed yeast culture
(Wickerhamomyces anomalus MSCU 0652 and Kluyveromyces marxianus MSCU 0655) and Lactobacillus
paracasei AN3 were tested for their antifungal activities against A. flavus growth and its aflatoxin production
both in vitro and in corn silage. Results: In vitro studies showed inhibitory activities of Lactobacillus paracasei and
mixed yeasts against aflatoxin production but lower activity against fungal growth. The co-cultures of L.
paracasei and mixed yeasts were used as starter for sterile and non-sterile corn ensiling. For the sterile
condition, a substantial reduction of pH was observed 1 day after ensiling. The growth of mixed yeasts and L.
paracasei was not mutually affected, but that of A. flavus was completely inhibited in 7 d with the reduction of
aflatoxin production after 14 d of ensiling. For corn silage, mixed yeasts and L. paracasei at a ratio of 1 × 106:1 ×
108 colony forming units/g inhibited fungal growth and reduced aflatoxin production at 5 d and 7 d of ensiling,
This study assessed the adsorption capacity and stability of Aflatoxin B1 (AFB1), Ochratoxin A (OTA), and
Zearalenon (ZEA) via Lactobacillus acidophilus ATCC 4356 and Lactobacillus rhamnosus ATCC 7469 individually
and in combination with each other. Detoxifying efficiency depends on lactic acid bacteria species
(hydrophobicity of cell wall), pH, temperature, time and mycotoxin type. According to the genetic algorithm
results, the highest adsorption capacity (qe) of toxin for L. acidophilus + L. rhamnosus after washing with ethanol
and chloroform decreased to (8.487 and 10.309 μg kg−1; AFB1), (9.464 and 9.014 μg kg−1; OTA), and (162.020
and 144.047 μg kg−1; ZEA), which is significantly less than before washing (9.872, 9.834, and 180.993) for AFB1,
OTA, and ZEA after 24 h incubation at 37 °C. The surface hydrophobicity in turn toxin binding stability and
capacity of the strains increased by increasing the incubation temperature from 27° to 37°C after 24 h. The
affinity of AFB1 and OTA toxins to surface of LAB showed the most and the least content. For investigation of the
stability binding of the AFB1, OTA and ZEA on LAB surfaces, toxin+LAB complexes were washed with solvents of
different polarities. The bacteria–toxin complex stability showed the trend AFB1 &gt; OTA&gt; ZEA for toxin
release in ethanol and ZEA&gt; OTA&gt; AFB1 for chloroform based on the difference in their polarity. Practical
application: In the present study, the investigation of the efficiency of AFB1, OTA and ZEA adsorption on single
and co-culture L. acidophilus and L. rhamnosus, on the basis of bacterial cell wall hydrophobicity and toxin
polarity was studied. Food composites can have polarity differences that affect the stability of the toxin-bacteria
Aim: The current study aimed to remove aflatoxin from reconstituted milk by adding three probiotics, namely
Saccharomyces boulardii, Lactobacillus casei, and Lactobacillus acidophilus. Background: Aflatoxins are
poisonous substances produced by certain kinds of fungi that are found naturally all over the world. They can
contaminate food crops and pose a serious health threat to humans and livestock. Microbial detoxification is one
method of eliminating aflatoxins, including aflatoxin M1. Methods: For this purpose, about 109 and 107 cfu/ml
of S. boulardii, L. casei, and L. acidophilus were inoculated into skim milk without aflatoxin M1. The samples
were then spiked by aflatoxin M1 in concentrations of 0.5 and 0.75 ng/ml. The concentration of the aflatoxin
residing in supernatant of milk samples after different storage times (30 and 90 minutes) and temperatures of 4 -
and 37°C was measured by ELISA method, and the results were confirmed by HPLC. Results: The results showed
that the highest amount of aflatoxin M1 removal was related to S. boulardii (96.88 ± 3.79c) with a microbial
density concentration of 109 cfu/ml and toxin concentration of 0.75 ng/ml at 37°C for 90 minutes and then to L.
acidophilus (71.46 ± 3.79b) with a microbial density concentration of 107 cfu/ml and toxin concentration 0.75
ng/ml at 4°C for 90 minutes. Furthermore, the maximum level of AFM1 binding to 107 cfu/ml of L. casei with
Microbial degradation is considered as an attractive method to eliminate exposure to aflatoxin B1 (AFB1), the
most toxic mycotoxin that causes great economic losses and brings a serious threat to human and animal health,
in food and feed. In this study, Bacillus amyloliquefaciens WF2020, isolated from naturally fermented pickles,
could effectively degrade AFB1 ranging from 1 to 8 μg/ml, and the optimum temperature and pH value were
37–45°C and 8.0, respectively. Moreover, B. amyloliquefaciens WF2020 was considered to be a potential
probiotic due to the synthesis of active compounds, absence of virulence genes, susceptibility to various
antibiotics, and enhanced lifespan of Caenorhabditis elegans. Extracellular enzymes or proteins played a major
role in AFB1 degradation mediated by B. amyloliquefaciens WF2020 into metabolites with low or no
mutagenicity and toxicity to C. elegans. AFB1 degradation by the cell-free supernatant was stable up to 70°C,
with an optimal pH of 8.0, and the cell-free supernatant could still degrade AFB1 by 37.16% after boiling for 20
min. Furthermore, B. amyloliquefaciens WF2020 caused a slight defect in fungal growth and completely inhibited
AFB1 production when co-incubated with Aspergillus flavus. Additionally, B. amyloliquefaciens WF2020
suppressed the expression of 10 aflatoxin pathway genes and 2 transcription factors (alfR and alfS), suggesting
that B. amyloliquefaciens WF2020 might inhibit AFB1 synthesis in A. flavus. These results indicate that B.
2-s2.0-85130705859
3-PBA is a major degradation intermediate of pyrethroids. Its widespread existence in the environment poses a
severe threat to the ecosystem and human health. This study evaluated the adsorption capacity of L. plantarum
RS20 toward 3-PBA. Batch adsorption experiments indicated that the optimal adsorption conditions were a
temperature of 37 °C and initial pH of 6.0–8.0, under which the removal rate was positively correlated with the
cell concentration. In addition, there was no link between the incubation time and adsorption rate. The kinetic
study showed that the ad-sorption process fitted well with the pseudo-second-order model, and the adsorption
isotherms could be described by both Langmuir and Freundlich equations. Heat and acid treatments showed that
the ability of strain RS20 in removing 3-PBA was independent of microbial vitality. Indeed, it was involved with
chemisorption and physisorption via the cell walls. The cell walls made the highest contribution to 3-PBA
removal, according to the adsorption experiments using different cellular components. This finding was further
reconfirmed by SEM. FTIR spectroscopy analysis indicated that carboxyl, hydroxyl, amino groups, and –C–N were
the functional sites for the binding of 3-PBA. The co-culture experiments showed that the adsorption of strain
RS20 enhanced the degradation of 3-PBA by strain SC-1. Strain RS20 could also survive and effectively remove 3-
Toxic ingredients in food can lead to serious food-related diseases. Such compounds are bacterial toxins
(Shiga-toxin, listeriolysin, Botulinum toxin), mycotoxins (aflatoxin, ochratoxin, zearalenone, fumonisin),
pesticides of different classes (organochlorine, organophosphate, synthetic pyrethroids), heavy metals, and
natural antinutrients such as phytates, oxalates, and cyanide-generating glycosides. The generally regarded safe
(GRAS) status and long history of lactic acid bacteria (LAB) as essential ingredients of fermented foods and
probiotics make them a major biological tool against a great variety of food-related toxins. This state-of-the-art
review aims to summarize and discuss the data revealing the involvement of LAB in the detoxification of foods
from hazardous agents of microbial and chemical nature. It is focused on the specific properties that allow LAB
to counteract toxins and destroy them, as well as on the mechanisms of microbial antagonism toward toxigenic
producers. Toxins of microbial origin are either adsorbed or degraded, toxic chemicals are hydrolyzed and then
used as a carbon source, while heavy metals are bound and accumulated. Based on these comprehensive data,
Dietary aflatoxins are a major risk to the health of aquaculture species and their animal and human consumers.
This study reports a first comparative antitoxic and antifungal activity of natural probiotic (honey) and antifungal
drug (natamycin)-supplemented diets to alleviate the toxicity induced by the fungus (A. flavus) or it's producing
aflatoxins. A set of experiments were primarily conducted in vitro including determining the minimum inhibitory
concentration (MIC) of A. flavus for natamycin and honey and the antifungal activity of the honey, besides
contaminating a fish diet with toxigenic strain A. flavus then detecting the dietary presence of aflatoxins, using
TLC, before and after cultivating the fungus. We further investigated the influence of honey and natamycin on
growth; biochemical parameters; histology and immunohistochemistry; and liver residual deposition (using HPLC
to identify the aflatoxin type and amount). One hundred and eighty Nile tilapia (Oreochromis niloticus) (25.99 ±
3.31 g) were allocated into six groups in triplicates (10 fish/replicate). A control group (G1) and five other groups,
G2, G3, G4, G5, and G6, were fed daily a basal diet and basal diets enriched with honey (70% concentration, 200
ml kgL−1 diet)) (H), natamycin (2.5 g kgL−1) (N), aflatoxins, and combined aflatoxins groups (aflatoxins+H and
aflatoxins+N) respectively for 45 days. Fish received aflatoxins-contaminated diet (G4) showed low growth, and
alterations in immune status, hepatic antioxidant, and hepato-renal biomarkers. Additionally, it revealed
histological changes in liver and kidneys tissues, besides a strong caspase-3 and weak BCL-2
immunohistoreactivity compared to combined groups (G5, G6), and higher tissues bioaccumulation. The dietary
additive of honey (G2) significantly (P &lt; 0.05) elevated growth parameters; serum immunological biomarkers
including lysozyme, nitric oxide (NO), and Myeloperoxidase (MPO); tissue antioxidant indicators including
superoxide dismutase (SOD) and reduced glutathione (GSH) implying oxidative protection; and retrieved the
malondialdehyde (MDA), antistress indicator (cortisol), hepato-renal parameters including ALP, AST, ALT, urea,
creatinine. At the tissue and cellular level, honey sustained the histological architecture, modulated the
immunohistochemical parameters, and efficiently mitigated the aflatoxins residual deposition in hepatic tissues
compared to natamycin (G3) and control (G1). Comparing the antitoxic efficiency of honey and natamycin for
the supplemented groups with aflatoxins (G5 and G6), natamycin was more effective in ameliorating the toxicity
with aflatoxins reflected in modulating all measured parameters and low mortalities compared to honey. The
Several feed preservation methods can ensure lower mycotoxin contamination levels enter the food life cycle,
and a relatively common wet preservation method of forage plant materials is fermentation. This study aimed to
characterize the microbiological state and mycotoxin contamination of fermented silages and haylages (corn,
alfalfa, rye, and triticale), their main microbiota, and isolation of bacteria with mycotoxin resistance. Bacteria
that remain viable throughout the fermentation process and possess high mycotoxin resistance can have a
biotechnological benefit. Lactic acid bacteria, primarily found in corn silage, were Lactiplantibacillus plantarum
isolates. Meanwhile, a high percentage of alfalfa silage and haylage was characterized by Lactiplantibacillus
pentosus. In rye silage and haylage samples, Pediococci were the typical bacteria. Bacterial isolates were
characterized by deoxynivalenol and zearalenon resistance. Some of them were sensitive to aflatoxin B1, while
ochratoxin A caused 33–86% growth inhibition of the cultures. The mycotoxin resistant organisms are under
The study presents a systematic review of published scientific articles investigating the effects of interventions
aiming at aflatoxin reduction at the feed production and animal feeding phases of the milk value chain in order
to identify the recent scientific trends and summarize the main findings available in the literature. The review
strategy was designed based on the guidance of the systematic review and knowledge synthesis methodology
that is applicable in the field of food safety. The Web of Science and EBSCOhost online databases were searched
with predefined algorithms. After title and abstract relevance screening and relevance confirmation with full-text
screening, 67 studies remained for data extraction, which were included in the review. The most important
identified groups of interventions based on their mode of action and place in the techno-logical process are as
follows: low-moisture production using preservatives, acidity regulators, ad-sorbents and various microbiological
additives. The results of the listed publications are summa-rized and compared for all the identified intervention
groups. The paper aimed to help feed pro-ducers, farmers and relevant stakeholders to get an overview of the
Dietary Lactobacillus acidophilus ATCC 4356 was used to relieve the impacts of aflatoxin B1 toxicity on the
performances of Liza ramada. The control diet was without any additives, while the second and third diets were
supplemented with aflatoxin B1 at 0.5 and 1 mg/kg. The fourth diet was supplemented with Lb. acidophilus ATCC
4356 at 1 × 106 CFU/mL per kg diet, while the fifth with aflatoxin B1 at 1 mg/kg and Lb. acidophilus ATCC 4356 at
1 × 106 CFU/mL per kg diet. The growth performance markedly increased (p &lt; 0.05) in L. ramada fed Lb.
acidophilus ATCC 4356, while aflatoxin B1 at 0.5 and 1 mg/kg groups showed a severe reduction. The red blood
cells, hemoglobulin, hematocrit, and white blood cells were markedly increased in L. ramada fed Lb. acidophilus
ATCC 4356 while decreased (p &lt; 0.05) in fish fed aflatoxin B1 at 0.5 and 1 mg/kg. The blood total protein and
albumin were markedly increased (p &lt; 0.05) in L. ramada fed Lb. acidophilus ATCC 4356 while reduced in
aflatoxin B1 at 0.5 and 1 mg/kg groups. The levels of total cholesterol and triglycerides were meaningfully
increased in fish of the Lb. acidophilus ATCC 4356 and aflatoxin B1 at 1 mg/kg groups while decreased in
aflatoxin B1 at 0.5 and 1 mg/kg groups. Alanine aminotransferase, aspartate aminotransferase, creatinine, and
urea levels were markedly decreased (p &lt; 0.05) in fish-fed Lb. acidophilus ATCC 4356 while increased in
aflatoxin B1 at 0.5 and 1 mg/kg groups. The highest levels of blood glucose and cortisol were seen in fish
contaminated with aflatoxin B1 at 1 mg/kg, while the lowest levels were observed in the fish fed Lb. acidophilus
ATCC 4356 group (p &lt; 0.05). The catalase and superoxide dismutase were markedly enhanced in the Lb.
acidophilus ATCC 4356 group and severely declined in aflatoxin B1 at 0.5 and 1 mg/kg groups (p &lt; 0.05). The
malondialdehyde level was markedly reduced in fish fed Lb. acidophilus ATCC 4356 with or without aflatoxin B1
at 1 mg/kg diets while increased in fish contaminated with aflatoxin B1 at 0.5 and 1 mg/kg (p &lt; 0.05). The
control group had lower malondialdehyde levels than the aflatoxin B1 at 1 mg/kg group and higher than the Lb.
acidophilus ATCC 4356 with or without aflatoxin B1 toxicity (p &lt; 0.05). Histopathological examination revealed
2-s2.0-85123214080
The modulation of gut microbiota and proteome due to aflatoxin B1 (AFB1) by probiotics remains unclear. This
study investigated the alterations of gut microbiota and proteome in AFB1-exposed rats treated with probiotic
Lactobacillus casei Shirota (Lcs). Forty male Sprague Dawley rats were randomly divided into five groups (n = 8)
comprised control, AFB1, AFB1+activated charcoal, AFB1+Lcs, and Lcs groups. The rats were subjected to
different treatments via oral gavage for four weeks. Urine and serum were collected for the measurement of
AFB1 biomarkers and organs were harvested for histological analysis. Metagenomic sequencing was performed
on fecal samples to profile gut microbiota. Besides, AFB1 most affected organ i.e. jejunum was subjected to
proteomic analysis. The results indicated that Lcs intervention significantly reduced AFB1 biomarkers. H&amp;E-
stained intestine showed Lcs alleviated AFB1-induced inflammation and abnormal cell growth, particularly at the
jejunum. Although AFB1 increased potentially pathogenic bacteria and reduced beneficial bacteria abundance in
feces, the microbiota composition was normalized with Lcs treatment. The gut proteome analysis of the jejunum
sample showed several pathways of AFB1 toxicity, wherein Lcs treatment demonstrated its protective effect. It
Employing LAB and/or their metabolites to reduce aflatoxins from food products is one of the fascinating areas
of research. Therefore, in the present work an attempt was made to study the effect of isolated lactic acid
bacteria (LAB) on the reduction of aflatoxin M1 (AFM1) from phosphate buffer saline (PBS) and reconstituted
skim milk (RSM). Out of 68 isolates, nine strains of LAB isolated from food products were examined to bind and
reduce AFM1. The isolates were incubated with AFM1 at different time and temperature (24 h, 48 h, and 72 h at
37 °C and 4 °C) and later the residual toxin in the supernatant was determined. The preliminary results through
thin layer chromatography (TLC) revealed that all isolates were able to bind AFM1 in PBS. However, based on the
intensity of spots observed in TLC sheets, it was indicated that two isolates, H1 and S2 genotypically identified as
W. confusa and L. plantarum shown better AFM1 binding ability as compared to other isolates. Further, the heat
treated cells of both the selected strains did not show significant difference to AFM1 binding ability as compared
to live cells. Results of high performance liquid chromatography (HPLC) observed that W. confusa H1 and L.
plantarum had shown 78% and 72% AFM1 binding in PBS at 37 °C after 72 h. There was a significant difference (P
< 0.05) in AFM1 binding ability of L. plantarum S2 at 37 °C & 4 °C, but such difference was not observed for W.
confusa H1. Moreover, these isolates also observed reduction of naturally present AFM1 in 10% RSM. This is the
Pistachio paste is very popular for breakfast or supper thanks to its desirable taste, flavor, and texture. One of
the hazards that are directly related to agricultural practices, processing, storage, and transportation of
pistachios and the byproducts is aflatoxin, which can cause irreversible effects on the consumer. Probiotics are
one of the most effective and safe methods to reduce aflatoxins. The variables under study were temperature
and time, aflatoxin concentration, and probiotic content. In total, 30 treatments were determined through the
rotatable central composite design. This is the first and most comprehensive study to optimize the production of
probiotic pistachio paste and investigate the detoxification effects of aflatoxin B1 using Bifidobacterium lactis
with six treatments and three replications in the pistachio paste matrix. In simple terms, it is possible to remove
a higher percentage of toxins by increasing the number of microorganisms and decreasing the toxin level. The
highest aflatoxin B1 reduction was observed in pistachio paste with aflatoxin B1 contamination of (19.7039
ng/g), which was spiked with Bifidobacterium lactis (109 CFU/g) and then stored at 25°C for 26.1853 days
(aflatoxin B1: 8.00007 ng/g = 59.4% reduction), which is consistent with the permissible limits of the Iran
National Standards Organization and the European Commission Regulation. The results showed a significant
Goat milk is a type of milk widely used either directly or by processing milk for various dairy products. This study
aimed to identify or reduce fungal toxins using lactic acid bacteria and assess their effects on goat milk. Lactic
acid bacteria (Lactobacillus acidophilus) were used to remove mycotoxins, aflatoxin M1, aflatoxin B1, and
ochratoxin A from goat milk that had been infected with these toxins. The removal process depends on linking
mycotoxins to the bacterial cell wall by adsorption of mycotoxins on the bacterial cell wall. Milk samples from
purchased dairy goats were used on a diet with quantities of pre-prepared aflatoxins. Produced aflatoxins
originated from the development of Aspergillus parasiticus cultivated on potato medium; the spore suspension
was collected after the incubation process. Lactobacillus acidophilus was then activation and isolated in 30 mL of
MRS liquid environment at 370C for 24 hours under anaerobic conditions, and the dead bacteria were separated
from the living bacteria by centrifugation of bacterial culture and were suspended (separately) in test tubes
containing 5 mL of sterile milk contaminated with mycotoxins. The tubes were then shaken for two minutes.
Determination of toxins aflatoxin M1 and B1 and ochratoxin A in milk samples were determined by ELISA
according to the fermentation periods of 2, 6, 12, 24, and 48 hours, and temperatures of 4, 25, and 370C, and at
the following different pH values: 5, 5.5, 6, 6.5, 7, and 7.5, which were conditions for both, dead and live
bacteria. The results revealed that the longer the fermentation period, the greater the capacity of Lactobacillus
acidophilus to detoxify goat milk, with removal rates of aflatoxin M1, aflatoxin B1, and ochratoxin A reaching
11.63, 43.33, and 1.98% after 2 hours, and 80.23, 65.33, and 5.87% after 48 hours, respectively. In contrast, the
effects of living bacteria were (9.57, 33.83, and 1.27%) after 2 hours, (78.8, 68.03, and 5.53%) after 48 hours for
aflatoxin M1, aflatoxin B1, and ochratoxin A, respectively. When talking about the effect of temperature on the
ability of Lactobacillus acidophilus to detoxify, high temperatures have increased detoxification, and the result
for dead bacteria was 28.5, 51.17, and 1.19% when fermentation temperature 40C, while the removal rate
reached 83.53, 74.8, and 5.33% when fermentation temperature was 370C, for both aflatoxin M1, aflatoxin B1,
and ochratoxin A respectively, because of adsorption mycotoxins on the bacterial cell wall. The effect of
temperature on living bacteria condition was similar to the state of dead bacteria, where detoxification rises
with high temperatures, with the ratios of 32.8, 47.33, and 3.2%, and 75, 70.83, and 5.07% for both 4 and 37 0 C,
respectively, for aflatoxin M1, aflatoxin B1, and ochratoxin A. pH levels influenced the susceptibility of
Lactobacillus acidophilus to remove toxins such as mycotoxins, meaning, the lower the pH value the greater
The present work was carried out to study the ability of five probiotics on the in vitro degradation of Aflatoxins
B1 (AFB1). The best results of in vitro were tested on the detoxification of AFB1 in rabbits. A total of 40 growing
New Zealand White (NZW) male rabbits were assigned to five experimental groups. Animals were fed the
following diets: basal diet (control), basal diet contaminated with 300 ppb AFB1, basal diet contaminated with
300 ppb AFB1. + probiotic 3 (0.5 g/kg diet), basal diet contaminated with 300 ppb AFB1 + ajowan (0.5 g/kg diet),
and basal diet contaminated with 300 ppb AFB1 + probiotic 3 (0.5 g/kg diet) + ajowan (0.5 g/kg diet). Live body
weight significantly (P &lt; 0.05) decreased in rabbits fed AFB1 contaminated diet compared to the control
rabbits. All additives improved (P &lt; 0.05) the live body weight. The best improvement occurred with probiotics
+ ajowan. The addition of probiotics increased (P &lt; 0.05) daily body weight gain in all weeks except the first
week. Adding ajowan or ajowan + probiotic led to a significant (P &lt; 0.05) increase in live body weight gain and
feed intake compared to rabbits fed AFB1 alone. The digestion coefficients of dry matter (DM), organic matter
(OM), crude fiber (CF), ether extract (EE), nitrogen-free extract (NFE), and digestible crude protein (DCP)
significantly (P &lt; 0.05) decreased in rabbits fed AFB1 contaminated diet. All additives improved (P &lt; 0.05)
the digestibility coefficients of DM, OM, EE, CF, NFE, and total digested nutrients (TDN)%. The best improvement
occurred with probiotics + ajowan. Concentrations of serum total protein, albumin and globulin significantly (P
Mycotoxins produced from Aspergillus, Penicillium, and Fusarium cause food spoilages during handling and
storage, owing to immense economic losses and serious human health concerns including immunosuppression
and carcinogenic effects. Furthermore, these species are also known to produce mycotoxins. Aflatoxin B1
(AFB1), zearalenone (ZEA), ochratoxin A (OTA), and deoxynivalenol (DON) are the most commonly occurring
mycotoxins. The removal of mycotoxins from the contaminated food using lactic acid bacterias (LABs) has been
proposed as a green, inexpensive, safe, and promising mycotoxin decontamination strategy. LABs can control
the mycotoxin production following a series of steps, including, adsorption, metabolite interaction, and
biodegradation. This article provides systematic review of LABs as bio-green preservative with anti-mycotoxin
potential for sustainable food safety. This consolidated review may be of technical importance to understand
The aim of this study was to evaluate the capacity of two strains of lactic acid bacteria (LAB), Lactobacillus
rhamnosus and Lactococcus lactis, and a yeast strain, Saccharomyces cerevisiae, inactivated by heat (121 °C, 10
min), from binding to aflatoxin M1 (AFM1), as well as the interaction between these microorganisms, aflatoxin
M1 and the Minas Frescal cheese matrix after 2 and 30 days of storage. The ability of LABs and S. cerevisiae to
bind AFM1 to Minas Frescal cheese was evaluated by high performance liquid chromatography (HPLC) composed
of a fluorescence detector. The interaction between these microorganisms and AFM1 was evaluated using a
scanning electron microscope composed of a backscattered electron detector with a voltage of 15 kV and
magnifications of 1000 ×, 5000 × and 8000 ×. The use of microorganisms as a biological method is efficient in
reducing AFM1 in Minas Frescal cheese and does not affect the microbiological parameters. AFM1 reduction
varied according to the microorganism used in the treatments. S cerevisiae showed greater capacity to bind
AFM1 over time, compared to LABs. Scanning electron microscopy was especially useful, confirming that lactic
Aflatoxin M1 (AFM1) is a mycotoxin that often contaminates milk. Like other mycotoxins, it is thermostable and
potentially carcinogenic. The present work was carried out to evaluate the ability of microorganisms isolated
from Indonesian kefir grains to reduce AFM1 in contaminated phosphate buffer saline (PBS). Fourteen isolates of
lactic acid bacteria, both aerobic (LAE) and anaerobic (LAN), and nine isolates of yeast (YEA) were used. The
significantly highest AFM1 reduction percentage was shown by the isolate LAE7 (29.3 ± 0.6%) after 4 h
incubation. DNA sequencing of LAE7 and YEA2 isolates showed that these isolates had homology (level of
similarity) with species of Lactobacillus kefiri strain A/K and Saccharomyces cerevisiae NRRL Y-12632,
The presence of mycotoxins in cereals and cereal products remains a significant issue. The use of natural
ingredients such as pumpkin and whey, which contain bioactive compounds, could be a strategy to reduce the
use of conventional chemical preservatives. The aim of the present work was to study the bioaccessibility of
aflatoxin B1 (AFB1) and ochratoxin (OTA) in bread, as well as to evaluate the effect of milk whey (with and
without lactic acid bacteria fermentation) and pumpkin on reducing mycotoxins bioaccessibility. Different bread
typologies were prepared and subjected to an in vitro digestion model. Gastric and intestinal extracts were
analyzed by HPLC–MS/qTOF and mycotoxins bioaccessibility was calculated. All the tested ingredients but one
significantly reduced mycotoxin intestinal bioaccessibility. Pumpkin powder demonstrated to be the most
effective ingredient showing significant reductions of AFB1 and OTA bioaccessibility up to 74% and 34%,
respectively. Whey, fermented whey, and the combination of pumpkin-fermented whey showed intestinal
bioaccessibility reductions between 57–68% for AFB1, and between 11–20% for OTA. These results pointed to
This systematic review and meta-analysis aimed to investigate the capacity of probiotic bacteria for reducing of
aflatoxin B1 as a food hazard. The results indicated that probiotic bacteria significantly decreased aflatoxin B1 by
45.99% (CI: 42.08%, 49.91%) also The rank order of binding subgroups were Lactobacillus 47.96% (CI: 43.51%,
52.40%), Bifidobacterium 43.95% (CI: 34.96%, 53.65%; I 2 = 99.8%), Pediococcus 41.61% (CI: 31.49%, 51.74%; I 2
= 99.3%), Lactococcus 33.56% (CI: 19.19%, 47.94%; I 2 = 99.5%) and Enterococcus 27.14% (CI: 20.70%, 33.58%, I
2 = 96.9%,). Probiotic bacteria could be recommended as a biological tool for decreasing aflatoxin B1 in different
This study isolated lactic acid bacteria from commercially available probiotic foods to determine their capacity to
remove aflatoxin B1 (AFB1) and trichothecene-2 (T-2). The removal rates by original live and heat-treated cells of
lactic acid bacteria (LAB) were compared to test the effect of heat treatment on efficacy. LAB is capable to
remove up to 46% of AFB1 and up to 45% of T-2 toixn. The toxin removal capability increased as toxin
concentration increased despite bacterial cell viability declining. Surprisingly, the denatured LAB removed
greater percentages of AFB1 (up to 62%) and T-2 (up to 52%) than live bacterial cells (P &lt; 0.05), lending
support to the hypothesis that there is higher binding of toxins to the cell membrane of nonviable cells. The
The present study was objected to assess the ability of lactic acid bacteria in the reduction of aflatoxin B1 (AFB1)
in a simulated human gastrointestinal tract. In this work, eight treatments were prepared to compare the ability
of noted probiotic bacteria in AFB1 reduction in different simulated gastrointestinal conditions. The results of
the present study revealed a significant reduction in AFB1 in all of the treatments in which the highest reduction
rate was observed by L. acidophilus in the absence of sterilized milk. Moreover, the findings showed that
bacteria, gastric juice, and small intestine may contribute to the reduction of AFB1. © 2020 Informa UK Limited,
Aflatoxin B1 (AFB1) is the most hazardous toxin in food and feed components. Lactobacilli are extensively
studied probiotic bacteria with detoxification characteristics. This study was conducted to isolate Lactobacillus
from broiler gut, identify them by 16S ribosomal RNA (16S r RNA) gene sequence and evaluate their
detoxification ability against AFB1. A total of fourteen Lactobacillus isolates were obtained from poultry gut and
were identified. Phylogenetic study on the base of 16S rRNA gene showed that isolates from same species and
source are genetically close to each other as compared to other isolates. Out of fourteen isolates, five have
shown detoxification ability against AFB1 (50 ppb), in aqueous environment at 37 °C for 6 hours. Viable cells and
cell walls have removed more concentration of AFB1 as compared to cell metabolites. Isolate Cr. 4 (L. salivarius)
removed the highest amount (92.3%) of AFB1 as compared to other isolates. It was concluded that Lactobacillus
BACKGROUND: Aflatoxins are mainly developed during the storage of feedstuffs, and their destruction is difficult
after the occurrence. The most practical strategy to combat aflatoxins is the use of mycotoxin binders.
OBJECTIVES: Comparison of the efficiency of traditional and lab-synthesized polymeric mycotoxin binder with
gastrointestinal microflora modulating feed additives in amelioration of aflatoxin effects in broiler chicken.
METHODS: A total of 240 1-day old broilers (Ross 308, straight forward) were examined in a completely
randomized design with five treatments and four replicates of 12 birds for 24 days of study duration. Treatments
were: 1. The negative control, feed without aflatoxin or any feed additive, 2. The positive control, aflatoxins
contaminated feed (500 µg/kg), 3. Aflatoxins + probiotic (Hypro Tect), 4. Aflatoxins + molecular imprinted
polymer (MIP), and 5. Aflatoxins + commercial toxin-binder (Zarin-binder). The growth performance of birds was
measured during the experiment. At the end of the experiment, some biochemical and immunological analyses
were performed on blood samples. Some bone characteristics were studied on tibia samples. RESULTS:
Supplementation of probiotics and toxin-binder in aflatoxin-contaminated feed improved the aflatoxin-induced
reduction of feed intake and body weight gain in the first 10 days of the experiment (P<0.05), compared to
positive control group. Aflatoxin alone (the positive control) or with the feed additives did not affect feed
conversion ratio. Aflatoxin reduced the levels of serum total protein, albumin, phosphorus, magnesium, and zinc
(P<0.05). Use of probiotic, MIP and commercial toxin-binder, in aflatoxin-contaminated feeds, has alleviated the
adverse effects of aflatoxin on serum albumin (P<0.05). The tibia weight increased in probiotic and MIP fed
broilers compared to the birds fed aflatoxin-contaminated feed without additives-the positive control (P<0.05).
The highest tibia breaking strength was observed in probiotic fed birds, which was different from that of the
positive control group. The tibia length was decreased by the aflatoxin compared to the negative control birds
(P<0.05). Anti-SRBC titers were decreased in aflatoxin contaminated group without feed additive
supplementation-positive control (P<0.05). CONCLUSIONS: The tested feed additives in present study exerted
just partial protection against some aflatoxicosis effects. The extent of effectiveness of studied feed additives in
Aflatoxin B1 (AFB1) is one of the most dangerous mycotoxins for humans and animals. This study aimed to
investigate the effects of compound probiotics (CP), CP supernatant (CPS), AFB1-degradation enzyme (ADE) on
chicken embryo primary intestinal epithelium, liver and kidney cell viabilities, and to determine the functions of
CP + ADE (CPADE) or CPS + ADE (CPSADE) for alleviating cytotoxicity induced by AFB1. The results showed that
AFB1 decreased cell viabilities in dose-dependent and time-dependent manners. The optimal AFB1
concentrations and reactive time for establishing cell damage models were 200 µg/L AFB1 and 12 h for intestinal
epithelium cells, 40 µg/L and 12 h for liver and kidney cells. Cell viabilities reached 231.58% (p &lt; 0.05) for
intestinal epithelium cells with CP addition, 105.29% and 115.84% (p &lt; 0.05) for kidney and liver cells with CPS
additions. The further results showed that intestinal epithelium, liver and kidney cell viabilities were significantly
decreased to 87.12%, 88.7% and 84.19% (p &lt; 0.05) when the cells were exposed to AFB1; however, they were
increased to 93.49% by CPADE addition, 102.33% and 94.71% by CPSADE additions (p &lt; 0.05). The relative
mRNA abundances of IL-6, IL-8, TNF-α, iNOS, NF-κB, NOD1 (except liver cell) and TLR2 in three kinds of primary
cells were significantly down-regulated by CPADE or CPSADE addition, compared with single AFB1 group (p &lt;
2-s2.0-85101886776
This study aimed to evaluate aflatoxin M1 (AFM1) level in milk powder and infant milk formulae, in addition to
applying innovative methods for AFM1 & AFB1 detoxification. Fifty random samples of milk powder and infant
formulae (25 of each) were collected from the Egyptian markets for assessing AFM1 level using ELISA technique.
Bioactive components comprising cell free supernatants (postbiotic), acid-dead cells (parabiotic) and the
encapsulated-cells of Lactobacillus plantarum RM1 and Lactobacillus paracasei KC39 were evaluated for their
antifungal activity against toxigenic mold strains and their impact on AFB1 and AFM1 reduction in reconstituted
milk powder. AFM1 concentration in unpacked milk powder was higher than that of packed samples and infant
formulae, although these differences were not significant (P > 0.05). About 96.0, 29.4 and 25.0% of the tested
infant formulae, unpacked, and packed milk powder were unacceptable in terms of the AFM1 limit defined by
Egyptian and European standards, while all samples were in accordance with the USA/FDA standard. All tested
mycotoxigenic strains were sensitive to the different treatments of the probiotics with the highest sensitivity
regarding Fusarium strain with L. paracasei KC39 compared to other genera. The degradation ratios of AFM1
using the bioactives of the L. paracasei KC39 were higher than that of L. plantarum RM1 bioactives. Additionally,
KC39 parabiotic manifested the best AFB1 reduction (60.56%). In conclusion, the positive and highly significant
Colorectal cancer (CRC) is the second leading cause of cancer death in the world. Immunotherapy using
monoclonal antibodies, immune-checkpoint inhibitors, adoptive cell therapy, and cancer vaccines has raised
great hopes for treating poor prognosis metastatic CRCs that are resistant to the conventional therapies.
However, high inter-tumor and intra-tumor heterogeneity hinder the success of immunotherapy in CRC. Patients
with a similar tumor phenotype respond differently to the same immunotherapy regimen. Mutation-based
classification, molecular subtyping, and immunoscoring of CRCs facilitated the multi-aspect grouping of CRC
patients and improved immunotherapy. Personalized immunotherapy using tumor-specific neoantigens provides
the opportunity to consider each patient as an independent group deserving of individualized immunotherapy.
In the recent decade, the development of sequencing and multi-omics techniques has helped us classify patients
more precisely. The expansion of such advanced techniques along with the neoantigen-based immunotherapy
could herald a new era in treating heterogeneous tumors such as CRC. In this review article, we provided the
latest findings in immunotherapy of CRC. We elaborated on the heterogeneity of CRC patients as a bottleneck of
2-s2.0-85120921595
Among the fermented products, cheese has a good potential to deliver probiotic microorganisms into the
gastrointestinal system due to its high protein and fat contents. The contamination of milk with aflatoxin M
deserves attention concerning cheese consumption due to the harmful effects on human health. In the present
research, the reduction of aflatoxin M1 (AFM1) by two well-known probiotic strains was studied in artificially
aflatoxin-contaminated Feta cheese. Changes in pH, the viability of the probiotic strains and the level of aflatoxin
in the samples were analyzed during 60-day storage. The results showed that all samples containing probiotics
dramatically reduced the AFM1 levels. From both the health and economic aspects, the B. bifidum species at an
inoculation level of 107 CFU/mL has proven to be the best treatment, due to the lowest cost of probiotics,
2-s2.0-85131051123
Food bio-preservatives are requested as substituents of chemical pesticides in food. The aim of this study was to
carry out a screening of twenty biocontrol agents (BCAs) for their potential fungicidal activity in vitro. Twenty
BCAs were tested against ten pathogenic fungi. Some of the cell-free supernatants (CFS) tested showed in vitro
antifungal activity versus pathogenic fungi. The highest fungicidal activity was observed in the fermented CFS of
Paenibacillus chibensis CECT 375, Bacillus amyloliquefaciens CECT 493, and Pantoea agglomerans CECT 850,
which showed a minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC) values of
125 and 250 g/L, respectively. The compounds responsible for the antifungal activity, such as organic and
phenolic acids, were determined. Lactic acid, acetic acid, benzoic acid, and phenyllactic acid among others can be
related to antifungal activity. HPLC-MS/MS analysis showed a reduction of ochratoxin A (OTA) and aflatoxin B1
(AFB1) up to 26% (Paenibacillus alvei CECT 2) and 55% (Paenibacillus polymyxa CECT 155), respectively. The
present study prompts that metabolism products of BCAs are propitious for the bioconservation of food, due to
Mycotoxins are a crucial problem for poultry production worldwide. Two of the most frequently found
mycotoxins in feedstuffs are deoxynivalenol (DON) and fumonisins (FUM) which adversely affect gut health and
poultry performance. The current knowledge on DON and FUM effects on broiler responses relevant for gut
detoxification, antioxidant capacity, and health is still unclear. The aim of this study was to assess a range of
selected molecular intestinal biomarkers for their responsiveness to the maximum allowable European Union
dietary levels for DON (5 mg/kg) and FUM (20 mg/kg) in broilers. For the experimental purpose, a challenge diet
was formulated, and biomarkers relevant for detoxification, antioxidant response, stress, inflammation, and
integrity were profiled across the broiler intestine. The results reveal that DON significantly (p < 0.05) induced
aryl hydrocarbon receptor (AhR) and cytochrome P450 enzyme (CYP) expression mainly at the duodenum.
Moreover, DON and FUM had specific significant (p < 0.05) effects on the antioxidant response, stress,
inflammation, and integrity depending on the intestinal segment. Consequently, broiler molecular responses to
DON and FUM assessed via a powerful palette of biomarkers were shown to be mycotoxin and intestinal site
Aflatoxin B1 (AFB1) is a hazardous component that can seriously threaten the public health. Terxine is a
component used in traditional soup and found in the western mountainous regions of Iran. Several
microorganisms have been reported to bind or degrade aflatoxins (AFs) in foods and feeds. This research aimed
to investigate the effect of Terxine fermentation by Lactobacillus plantarum strains AF1 and LU5 on AFB1.
Fermentation was carried out, and pH, lactic acid and AFB1 amount and microbial count were further
determined. In addition, the kinetic experimental data of AFB1 by L. plantarum AF1 and LU5 (obtained at 37°C)
were fitted to the zero-order, first-order and parabolic diffusion models. According to the coefficient of
determination (R2) and root mean square of errors (RMSE), the zero-order model best described AF
degradation. The growth of Lactobacillus strains was increased by the rise in the fermentation time; in this
regard, the number of L. plantarum AF1 increased from 4.2 to 5.1 log cfu/g and that of L. plantarum LU5
increased from 4.1 to 5.2 log cfu/g in the first 8 h, reaching 7.2 and 7.4 log cfu/g in the next 8 h, respectively. The
results also showed that the amount of lactic acid increased whereas the pH value decreased during the 24 h
Contamination of food and feed by mycotoxins is considered one of most serious food safety problems in the
world, because these fungal metabolites can be teratogenic, mutagenic, carcinogenic and immunosuppressive,
and can cause serious damages to animal and human health. Mycotoxins may modulate the gut microbiota with
potential consequences for gut and host health. On the other hand, the gut microbiota may metabolize the
mycotoxins thereby converting them to a form with different activity. Chemical-microbial interactions can be
categorized into two classes: Microbiome Modulation of Toxicity (MMT) and Toxicant Modulation of the
Microbiome (TMM). The present review provides a state-of-the-art overview of this bi-directional interaction
between the major food-borne mycotoxins such as aflatoxins, ochratoxins, deoxynivalenols and zearalenone
present in food, feed and the gut microbiota. In addition, the effect of probiotics on gut microbiota in animals
exposed to mycotoxins is summarized. Possible consequences of the role of gut microbiota for the risk
Studies have shown that the intracellular content of probiotic (postbiotics) has antioxidant properties, which can
improve the antioxidant status in vivo. However, its absorption and mechanisms underlying the protective
effects are still unknown. The antioxidant capacity of Lacticaseibacillus casei CRL431 (IC-431) postbiotics was
determined after an in vitro simulated digestive process. Permeability of antioxidant constituents of IC-431 was
determined by an ex vivo everted duodenum assay. Aflatoxin B1–induced oxidative stress rat models were
established and treated with IC-431; biomarkers of hepatic mitochondrial function and H2O2 levels, oxidative
stress, and oxidative stress index (OSi) were examined. The antioxidant capacity of IC-431 (477 ± 45.25 μmol
Trolox Equivalent/L) was reduced by exposure to the simulated digestive process. No difference (p &gt; 0.05)
was found among digested and the permeate fraction of IC-431. A protective effect was observed by significantly
lower OSi and higher liver glutathione peroxidase and catalase activities. Lower H2O2 production, a higher
degree of mitochondrial uncoupling, and lower mitochondrial respiration coefficient were also observed
(p &lt; 0.05). These results suggest that IC-431 antioxidant components permeate intestinal barriers to enter the
bloodstream and regulate antioxidant status during AFB1-induced oxidative stress by reducing hepatic
Aflatoxin B1(AFB1) widely exists in food and feed, which seriously endangers human and animal health. How to
detoxify AFB1 is a research hotspot at present. This study attempts to use the Bacillus amyloliquefaciens B10,
one of probiotics strain as the research object to ascertain whether it can alleviate the kidney injury induced by
AFB1 in mice and its mechanism. Fifty-six mice were divided into four groups (control, AFB1, AFB1 + B10, and
B10). The mice that received intragastric administration for 28 days were euthanised, and serum was collected
for biochemical index detection with fresh kidney tissue taken for HE staining, TUNEL detection, and protein
expression detection. Our results showed that the biochemical indices changed, significant pathological changes
appeared, the number of apoptotic cells increased in the kidney tissue of the AFB1 group mice; the protein
expressions of Nrf2, HO-1,AKT, P-AKT, and Bcl-2 in the AFB1 group were significantly decreased; the protein
expressions of Keap-1, PTEN, Bax, Caspase-9, and Caspase-3 were significantly increased. After B.
amyloliquefaciens B10 co-treatment, compared with the AFB1 group, the biochemical indices, pathological
2-s2.0-85105836534
This study aimed to evaluate the in vitro ability of heat-killed (HK) and acid-killed (AK) cells from three
commercial strains of lactic acid bacteria, alone or in combination with sorbitan monostearate (SM), to bind to
aflatoxin M1 (AFM1) in skimmed milk at concentrations of 0.05, 0.2 and 0.5 ng mL−1. The stability of the AFM1-
bacterial cell bonds and the cell wall and exopolysaccharides (EPS) contribution to AFM1 decontamination were
also investigated. Compared with HK cells, AK cells exhibited higher binding capacities in milk containing the
highest AFM1 level, with maximum removal of 81.4, 56.8, and 50.8% by Lactococcus lactis ssp. cremoris,
Lacticaseibacillus rhamnosus (formerly Lactobacillus rhamnosus), and L. lactis ssp. lactis at 1010 cells mL−1,
respectively. The combination of HK cells with SM increased the AFM1 binding capacity, although no effect was
found regarding AK cells. SM treatment also enhanced the stability of the AFM1 bond formed with both HK and
AK cells. Cell wall isolates and EPS were the major components involved in the AFM1 binding process. Combining
2-s2.0-85105825119
Contamination of milk with aflatoxin M1 (AFM1) is related to the feed for milking cows, which is contaminated
with aflatoxin B1 (AFB1). Feed AFB1 converts to AFM1 by dehydrogenation. In this study, we used Lactic acid
bacteria (LAB) isolated from raw milk and its products and commercial or laboratory-made beta-glucan isolated
from yeast and oats to establish how these mycotoxin binders affect the quality of sterilised, long-life, 2.8% fat
milk contaminated with 0.05 mg/L of AFM1. We took the content of fats, carbohydrates, sugars (lactose), and
proteins, and the calculated energy values for quality parameters. The mean energy value of the milk treated
with AFM1 binders ranged between 85.7% and 101.5% of the control, untreated milk, whereas the fat content
ranged between 65.3% and 100.7%. The protein content ranged between 64.4% and 101.1%, carbohydrates
between 83.1% and 103%, and lactose between 76.3% and 100.8%. The results indicated a good possibility of
binding of AFM1 with Lactobacilus plantarum bacteria, and 0.01% of β-glucan from oats was 0.005% of β-glucan
isolated from yeast from Saccharomyces cerevisiae 20. These findings suggest that milk treated with these
Ogi is a fermented cereal beverage, made primarily from maize (Zea mays) and rarely from millets. Unlike maize-
based ogi, little is known about the bacterial community and mycotoxin profile during the production of millet-
based ogi. Therefore, the bacterial community dynamics and mycotoxin reduction during ogi processing from
three millet varieties were investigated using next-generation sequencing of the 16S rRNA gene and liquid
chromatography-tandem mass spectrometry, respectively. A total of 1163 amplicon sequence variants (ASVs)
were obtained, with ASV diversity across time intervals influenced by processing stage and millet variety. ASV
distribution among samples suggested that the souring stage was more influenced by millet variety than the
steeping stage, and that souring may be crucial for the quality attributes of the ogi. Furthermore, bacterial
community structure during steeping and souring was significantly differentiated (PERMANOVA, P < 0.05)
between varieties, with close associations observed for closely-related millet varieties. Taxonomically,
Firmicutes, followed by Actinobacteria, Bacteroidetes, Cyanobacteria and Proteobacteria phyla were relatively
abundant (>1%). Lactic acid bacteria, such as Burkholderia-Caballeronia-Paraburkholderia, Lactobacillus,
Lactococcus and Pediococcus, dominated most fermentation stages, suggesting their roles as key fermentative
and functional bacteria in relation to mycotoxin reduction. About 52–100%, 58–100% and 100% reductions in
mycotoxin (aflatoxins, beauvericin, citrinin, moniliformin, sterigmatocystin and zearalenone) concentrations
were recorded after processing of white fonio, brown fonio and finger millet, respectively, into ogi. This study
The increased consumption of plant-based foods has intensified the concern related to mycotoxin intoxication.
This study aimed to investigate the effect of selected lactic acid bacteria (LAB) strains on the growth of
Aspergillus parasiticus NRRL 2999 and its production of aflatoxin (AF). The ability of the heat-killed (100°C for 1
h) LAB strains to bind aflatoxin M1 (AFM1) in milk and aflatoxin B1 (AFB1), ochratoxin A (OTA), and zearalenone
(ZEN) in potassium phosphate buffer (PPB) was also evaluated in vitro. Ten LAB strains were tested individually,
by inoculating them simultaneously with the fungus or after incubation of the fungus for 24 or 48 h at 25°C.
Double layer yeast extract sucrose (YES) agar, de Man Rogosa and Sharpe (MRS) agar, and YES broth were
incubated for 7 days at 25°C to follow the development of the fungus. Levilactobacillus spp. 3QB398 and
Levilactobacillus brevis 2QB422 strains were able to delay the growth of A. parasiticus in YES broth, even when
these strains were inoculated 24 h after the fungus. The inhibitory effect of these LAB strains was confirmed by
the reduction of fungus colony size, suggesting dominance of LAB by competition (a Lotka-Voltera effect). The
production of AFB1 by A. parasiticus was inhibited when the fungus was inoculated simultaneously with
Lactiplantibacillus plantarum 3QB361 or L. plantarum 3QB350. No AFB1 was found when Levilactobacillus spp.
2QB383 was present, even when the LAB was inoculated 48 h after the fungus. In binding studies, seven
inactivated LAB strains were able to promote a reduction of at least 50% the level of AFB1, OTA, and ZEN. This
reduction varied depending on the pH of the PPB. In milk, however, only two inactivated LAB strains were able
Lactobacillus helveticus FAM22155 was the most efficient among five lactic acid bacteria at removing aflatoxin
B1 (AFB1) during solid-state fermentation on wheat bran substrate. The mechanism of removal was explored by
comparing different fermentation modes. Liquid fermentation had little effect on the breakdown of AFB1.
However, a protein extract from the fermented bran was equally effective at degrading aflatoxin B1 as living cell
digestion. After treatment with heat and protease K, the degrading capacity of the protein extract was
significantly reduced. Taken together, the observed biotransformation of AFB1 was mainly associated with
proteins produced during bran fermentation. Four products of U-[13C17]-AFB1 were found by mass
spectrometry, including Ⅱ-1 (C11H10O4), Ⅱ-2 (C11H10O4), III (C15H12O5), and IV (C14H10O4). These products
Background: Aflatoxin M1 is a highly toxic and carcinogenic compound which is found in milk and milk products
and it is a hydorxylated metabolite of Aflatoxin B1. When the dairy animal digested AFB1 contaminated feed, it is
changed to aflatoxin M1 and transferred to tissues and milk. Aflatoxin M1 is less carcinogenic than AFB1, but it is
acutely hepatotoxic as AFB1. Liver is their main target organ in the body. It has been calculated approximately
that 0.3- 6.2% of presented AFB1 in animal feed transferred as aflatoxin M1 in milk. Occurrence of AFM1 in milk
and milk products is a big concern. Therefore, several countries have standardized the maximum levels of AFM1
in milk and milk products. Methods: In this experiment, the ability of yoghurt bacteria to degrade AFM1 levels in
milk and yoghurt were analysed. The starter culture of yoghurt contains Streptococcus thermophilus and
Lactobacillus delbrueckii subsp. Bulgaricus (1:1). The experiment is carried out in Glasgow Caledonian University,
department of life science in 2010. Result: These bacteria showed the higher binding ability between 90- 100% in
milk samples whereas no considerable reduction was observed in yoghurt samples. In some yoghurt samples, an
increase of AFM1 level was detected but in overall, concentration of AFM1 was stable in yoghurt. Also, the fat
Aflatoxins (AFs) released by fungi are observed in the cow's milk even after pasteurization. Aflatoxin M1 (AFM1)
has particularly an incredible clinical significance, as a critical carcinogenic agent for humans. Several strategies
have been implemented for lowering the AFM1 amount, such as the employment of probiotics, particularly
lactobacilli or lactic acid bacteria (LAB). However, this strategy has not been applied routinely until today. This
study aimed to evaluate the effect of three LABs on the reduction of AFM1 in traditional milk and cheese
samples. In total, 85 milk (n=45) and cheese (n=40) samples were obtained from the open markets of Shiraz,
Iran, from February to June 2018. Additionally, the AFM1 levels were evaluated, compared to those of the
National Iranian Standard. The data were then analyzed in SPSS software (version 20) through the Chi-square
test. Statistical analysis was performed at a 95% confidence level (p-value of <0.00001). Out of 50 purchased
LABs, the efficient antifungal property and resistance to bile salts were observed in five strains. The mean value
of these five strains was calculated after adding 5 ppm AFM1, compared to natamycin. The strains with a
reduction in AFM1 level were sequenced and registered in the NCBI database. In total, 15 samples with
contamination higher than the allowed limit included Penicillium spp, Aspergillus niger, Saccharomyces cerevisia,
Saccharomyces paradoxus, and Yarrowia lipolytica. The results also showed reduced AFM1 levels in three LAB-
treated strains. Lactobacillus fermentum CECT562 (T), Lactobacillus brevis ATCC14869 (T), and Enterococcus
faecium LMG 11423 (T) had this capability to 0.05, 0.03, and 0.03 respectively. The National Iranian Standard
The removal of mycotoxins from contaminated feed using lactic acid bacteria (LAB) has been proposed as an
inexpensive, safe, and promising mycotoxin decontamination strategy. In this study, viable and heat-inactivated
L. acidophilus CIP 76.13T and L. delbrueckii subsp. bulgaricus CIP 101027T cells were investigated for their ability
to remove aflatoxin B1 (AFB1), ochratoxin A (OTA), zearalenone (ZEA), and deoxynivalenol (DON) from MRS
medium and PBS buffer over a 24 h period at 37◦C. LAB decontamination activity was also assessed in a ZEA-
contaminated liquid feed (LF). Residual mycotoxin concentrations were determined by UHPLC-FLD/DAD analysis.
In PBS, viable L. acidophilus CIP 76.13T and L. delbrueckii subsp. bulgaricus CIP 101027T cells removed up to 57%
and 30% of ZEA and DON, respectively, while AFB1 and OTA reductions were lower than 15%. In MRS, 28% and
33% of ZEA and AFB1 were removed, respectively; OTA and DON reductions were small (≤15%). Regardless of
the medium, heat-inactivated cells produced significantly lower mycotoxin reductions than those obtained with
viable cells. An adsorption mechanism was suggested to explain the reductions in AFB1 and OTA, while
biodegradation could be responsible for the removal of ZEA and DON. Both viable LAB strains reduced ZEA by
23% in contaminated LF after 48 h of incubation. These findings suggest that LAB strains of L. acidophilus CIP
Aflatoxin B1 (AFB1) and ochratoxin A (OTA) are two of the most important of mycotoxins associated with
tropical and subtropical climate as in Egypt. Therefore, this study aimed to isolated lactic acid bacteria (LAB) and
Bifidobacteria from some local dairy products from Egypt and using some strains to reduce/or remove AFB1 and
OTA toxins. Fifty samples, of milk and dairy products, including ten samples each, of Kareish, Damietta cheese
(soft type), buffalo's milk, yoghurt (zabady), and naturally acidified milk (Rayeb)were screened for their load of
Lactobacilli, Lactococci and Bifidobacteria, using MRS agar, M17 agar and (MRS-Cys) agar, respectively. The
obtained data indicated that 38, 31 and 22 isolates belonged to Lactobacillus, Lactococcus and genus
Bifidobacterium, respectively. Fourteen strains isolated from LAB and Bifidobacterium were tested on AFB1 and
OTA in PBs contaminated by10 ppb with three times incubation periods (6, 12 and 36 hours) at 37°C. The results
indicated that all tested strains were able to reduce AFB1 at different rates ranging between 12.1 to 65.4% after
incubation for 6 h. These rates increased to 78.8% and 89.9% after incubation at 37°C for 12 and 36h,
respectively. The same results were with OTA, where percentages of reduction ranging from 81.4 to 80.4% were
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and the second leading cause of
cancer-related deaths worldwide. Chronic infections with hepatitis B virus (HBV) and hepatitis C virus (HCV),
alcoholic liver disease (ALD), and non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH)
are the major extrinsic risk factors of HCC devel-opment. Genetic background is pivotal in HCC pathogenesis, and
both germline mutations and single nucleotide polymorphism (SNP) are intrinsic risk factors of HCC. These HCC
risk factors predispose to hepatic injury and subsequent activation of fibrogenesis that progresses into cirrhosis
and HCC. Probiotic bacteria can mitigate HCC risk by modulating host gut microbiota (GM) to promote growth of
beneficial microbes and inhibit HCC-associated dysbiosis, thus preventing pathogen-associated molecular
patterns (PAMPs)-mediated hepatic inflammation. Probiotics have antiviral activities against HBV and HCV
infections, ameliorate obesity and risk of NAFLD/NASH, and their antioxidant, anti-proliferative, anti-angiogenic,
and anti-metastatic effects can prevent the HCC pathogenesis. Probiotics also upregulate the expression of
tumor suppressor genes and down-regulate oncogene expression. Moreover, metabolites generated by
probiotics through degradation of dietary phytochemicals may mitigate the risk of HCC development. These
2-s2.0-85101980766
This paper discusses the structural difference and role of xylan, procedures involved in the production of
xylooligosaccharides (XOS), and their implementation into animal feeds. Xylan is non-starch polysaccharides that
share a β-(1-4)-linked xylopyranose backbone as a common feature. Due to the myriad of residues that can be
substituted on the polymers within the xylan family, more anti-nutritional factors are associated with certain
types of xylan than others. XOS are sugar oligomers extracted from xylan-containing lignocellulosic materials,
such as crop residues, wood, and herbaceous biomass, that possess prebiotic effects. XOS can also be produced
in the intestine of monogastric animals to some extent when exogenous enzymes, such as xylanase, are added
to the feed. Xylanase supplementation is a common practice within both swine and poultry production to reduce
intestinal viscosity and improve digestive utilization of nutrients. The efficacy of xylanase supplementation varies
widely due a number of factors, one of which being the presence of xylanase inhibitors present in common
feedstuffs. The use of prebiotics in animal feeding is gaining popularity as producers look to accelerate growth
rate, enhance intestinal health, and improve other production parameters in an attempt to provide a safe and
sustainable food product. Available research on the impact of xylan, XOS, as well as xylanase on the growth and
health of swine and poultry, is also summarized. The response to xylanase supplementation in swine and poultry
feeds is highly variable and whether the benefits are a result of nutrient release from NSP, reduction in digesta
viscosity, production of short chain xylooligosaccharides or a combination of these is still in question. XOS
supplementation seems to benefit both swine and poultry at various stages of production, as well as varying
This study aimed to investigate the potential ability of simultaneously used L. acidophilus(LA-5),
L.rhamnosus(LGG), and L.casei(LC-01) in encapsulated (E) and nonencapsulated (NE) forms in mycelial growth of
Aspergillus spp and aflatoxin production by A. flavus. In order to assess the zone of fungal growth inhibition by E
and NE lactic acid bacteria, the agar well diffusion method was applied. Quantification of aflatoxin was
performed using a high-performance liquid chromatography technique. Lactic acid bacteria exhibited high
antifungal activity and significantly reduced AFB1, AFB2, AFG1, and AFG2 production in both E and NE forms
compared to the control group. The percentage of reduction in total AFs production in treated samples with E
and NE lactic acid bacteria was 94.1% and 95.5%, respectively. These results suggested that simultaneously used
email: abbaszade@ut.ac.ir
2-s2.0-85099447524
Aflatoxins (AFs) are toxic secondary metabolites produced mostly by Aspergillus species. AF contamination
entering the feed and food chain has been a crucial long-Term issue for veterinarians, medicals, agroindustry
experts, and researchers working in this field. Although different (physical, chemical, and biological) technologies
have been developed, tested, and employed to mitigate the detrimental effects of mycotoxins, including AFs,
universal methods are still not available to reduce AF levels in feed and food in the last decades. Possible
biological control by bacteria, yeasts, and fungi, their excretes, the role of the ruminal degradation, pre-harvest
biocontrol by competitive exclusion or biofungicides, and post-harvest technologies and practices based on
biological agents currently used to alleviate the toxic effects of AFs are collected in this review. Pre-harvest
biocontrol technologies can give us the greatest opportunity to reduce AF production on the spot. Together with
Palm kernel cake (PKC), a by-product of oil extracted from palm nuts through expeller press or solvent extraction
procedures is one of the highest quantities of locally available and potentially inexpensive agricultural product.
PKC provides approximately 14–18% of crude protein (CP), 12–20% crude fiber (CF), 3–9% ether extract (EE), and
different amounts of various minerals that feasible to be used as a partial substitute of soybean meal (SBM) and
corn in poultry nutrition. Poultry’s digestibility is reported to be compromised due to the indigestion of the high
fiber content, making PKC potentially low for poultry feeding. Nevertheless, solid-state fermentation (SSF) can be
applied to improve the nutritional quality of PKC by improving the CP and reducing CF content. PKC also contains
β-mannan polysaccharide, which works as a prebiotic. However, there is a wide variation for the inclusion level
of PKC in the broiler diet. These variations may be due to the quality of PKC, its sources, processing methods and
value-added treatment. It has been documented that 10–15% of treated PKC could be included in the broiler’s
diets. The inclusion levels will not contribute to a negative impact on the growth performances and carcass yield.
Furthermore, it will not compromise intestinal microflora, morphology, nutrient digestibility, and immune
2-s2.0-85099941624
Recently, researchers have reported the presence of patulin as a mycotoxin in commercial apple products,
especially apple juices. The aim of this study was to assess adsorption of patulin from artificially contaminated
apple juice using two lactic acid bacteria (LAB) strains of Lactobacillus acidophilus ATCC 4356 and Lactobacillus
plantarum ATCC 8014. Furthermore, effects of five physical and chemical pretreatments on the patulin
adsorption were investigated. Results demonstrated that patulin adsorption abilities of both strains increased
with NaOH pretreatment but decreased after autoclaving. The NaOH-treated L. plantarum ATCC 8014 showed
the best removal rate (59.74%) after 48 h of refrigerated storage, compared with the NaOH-treated L.
acidophilus ATCC 4356 (52.36%). Moreover, stability of the LAB-patulin complex was assessed in simulated
gastrointestinal tract conditions and a low quantity of patulin was released into the solution. The patulin
adsorption process by NaOH-treated L. plantarum ATCC 8014 followed Freundlich isotherm model and pseudo-
second-order kinetic model. Fourier transform infrared spectroscopy showed that polysaccharide and protein
components of the L. plantarum ATCC 8014 cell wall played key roles in patulin adsorption. The major functional
groups of the cell wall that were involved in adsorbing patulin included -OH/-NH, -CH2, C=O, and C-O groups. The
Mycotoxins have been a relevant issue worldwide for decades, becoming an emerging health concern and
present a substantial economic loss in several countries. Mycotoxins produced by fungi may influence human
health negatively due to their carcinogenic, mutagenic, estrogenic, nephrotoxic, hepatotoxic, neurotoxic, and
immunosuppressive effects. Mycotoxins are heat-stable compounds that are resistant to conventional food
process technologies, although detoxification by microorganisms has been proposed for their control for
maintaining the quality and safety of foods. The potential of probiotics to reduce the availability of mycotoxins in
foods has been extensively studied, with a special focus on lactic acid bacteria (LAB) species. In this chapter, a
comprehensive description of the most important LAB is presented, along with a discussion of the various
approaches used for the microbiological detoxification of the main mycotoxins. The proposed mechanistic mode
Aflatoxins (AFs) are known to be oncogenic mycotoxins. This study investigated the mitigation effects of lactic
acid bacteria (LAB) isolated from four types of vegetable, cucumber, Chinese cabbage, Japanese radish and
eggplant, which are used to make Japanese traditional fermented pickles, on AFs. Using aflatoxin M1 (AFM1)
binding assay for screening, four representative strains were selected (one from each vegetable) from total 94
LAB strains, based on the highest binding ratio. The ranges of the binding ratio of these representative strains to
aflatoxin B1 (AFB1), aflatoxin B2, aflatoxin G1, aflatoxin G2 and AFM1 were 57.5%–87.9% for the LAB strain
derived from cucumber, 18.9%–43.9% for the LAB strain derived from Chinese cabbage, 26.4%–41.7% for the
LAB strain derived from Japanese radish, and 15.0%–42.6% for the LAB strain derived from eggplant. The
strains isolated from cucumber, Chinese cabbage, Japanese radish and eggplant were identified as Lactococcus
lactis subsp. lactis, Weissella cibaria, Leuconostoc mesenteroides and Leu. mesenteroides, respectively. An in
vitro binding assay of the four strains under acidic conditions showed that the number of living bacteria
decreased, while the binding ratio increased in some strains, suggesting that the LAB maintained their capacity
to bind aflatoxins even in an environment that imitated the stomach. An in vivo experiment using L. lactis subsp.
lactis derived from cucumber revealed that the bacteria significantly inhibited the absorption of AFB1 into blood.
Milk is a key food worldwide prone to mycotoxins contamination. Lactobacillus plantarum and prebiotics
detoxification ability was evaluated by a Plackett-Burman Design considering the reduction of aflatoxin B1
(AFB1) and its bioaccessibility in artificially contaminated ultra-high temperature cow milk. Six variables were
evaluated: AFB1 concentration (from 5.0 to 10.0 μg L−1); incubation time (0 to 6 h); and inulin, oligofructose, β-
glucan, and polydextrose concentrations (from 0.00 to 0.75%). The reduction in AFB1 ranged from 0% to 55.85%
and in vitro bioaccessibility from 15.62% to 51.09%. The greatest reduction in AFB1 occurred by adding L.
plantarum combined with inulin, oligofructose and β-glucan. The greatest reduction in bioaccessibility occurred
by adding inulin or oligofructose and L. plantarum with a 10.0 μg L−1 AFB1 concentration. A sharp reduction in
AFB1 was accompanied by higher bioaccessibility rates, and in this case, bioaccessibility is considered the main
factor to ensure a low AFB1 absorption by the body. The best experimental condition was 10.0 µg L-1 AFB1,
added of L. plantarum and inulin or oligofructose (0.75%), ensuring &gt; 16% final bioaccessibility. Such results
The aim of this study was to examine the impact of intake of a lactic acid bacterium, Lactobacillus plantarum
299v (Lp299v) on aflatoxin-induced hepatotoxicity in broilers. For this, broilers were intoxicated with dietary
aflatoxins and simultaneously treated with live Lp299v in drinking water. One-day-old male broilers were divided
into eight groups (n = 10/group) as follows. Aflatoxin groups fed basal diet contaminated with aflatoxins (200 or
2000 ppb). The probiotic groups received drinking water enriched with live (Lp299v) (108 CFU/ml). A group of
birds was given a commercial mycotoxin binder (2.5 g/kg feed). Control groups received basal diet without
probiotic or aflatoxin binder. The growth performance was calculated for the entire period (0–42 days), and
blood and liver specimens were processed for histology and determination of liver damage markers. Results
showed extensive damage including bile duct hyperplasia, hepatocellular ballooning, and necrosis in chickens fed
aflatoxin alone. However, liver lesions were limited to lobular inflammation and pyknosis in broilers treated with
aflatoxins along with Lp299v. The histology of the liver tissues from the birds on aflatoxin-free diet + probiotic
appeared to be normal when compared to the respective controls. Histopathological indices in different
experimental groups were corroborated with the liver damage markers namely, serum ALT, AST, LD, and γ-GT. It
is concluded that the improvement in the growth performance and prevention of aflatoxin-related liver lesions
could be mainly assigned to the probiotic therapy for the entire period of breeding, although the aflatoxin-
The aim of this study was to evaluate the detoxification of aflatoxin B1 (AFB1) in vitro and in broiler chickens
using a triple-action compound mycotoxin detoxifier (CMD). Response surface methodology (RSM) was used to
evaluate AFB1 detoxification in artificial gastrointestinal fluid (AGIF) in vitro. The AFB1-degradation rate was
41.5% (P &lt; .05) when using a compound probiotic (CP) in which the visible counts of Bacillus subtilis,
Lactobacillus casein, Enterococcus faecalis and Candida utilis were 1.0 × 105, 1.0 × 105, 1.0 × 107 and 1.0 × 105
CFU/mL, respectively. When CP was combined with 0.1% AFB1-degrading enzyme to give CPADE, the AFB1-
degradation rate was increased to 55.28% (P &lt; .05). The AFB1-removal rate was further increased to above
90% when CPADE was combined with 0.03% montmorillonite to make CMD. In vivo, a total of 150 one-day-old
Ross broilers were allotted to 3 groups, 5 replications for each group, 10 broilers in each replication. Group A:
basal diet, Group B: basal diet with 40 μg/kg AFB1, Group C: basal diet with 40 μg/kg AFB1 plus CMD. The
feeding experiment period was 21 d. The results showed that broiler growth was increased, and AFB1 residues in
2-s2.0-85112209580
Some fungal species of the genera Aspergillus, Penicillium, and Fusarium secretes toxic metabolites known as
mycotoxins, have become a global concern that is toxic to different species of animals and humans. Biological
mycotoxins detoxification has been studied by researchers around the world as a new strategy for mycotoxin
removal. Bacteria, fungi, yeast, molds, and protozoa are the main living organisms appropriate for the mycotoxin
detoxification. Enzymatic and degradation sorptions are the main mechanisms involved in microbiological
detoxification of mycotoxins. Regardless of the method used, proper management tools that consist of before-
harvest prevention and after-harvest detoxification are required. Here, in this review, we focus on the
Grains and feed are severely contaminated by deoxynivalenol (DON) globally, threatening both human and
animal health. Research on bio-degradation of DON, in general, is gaining attention. The aim of this research was
to estimate the effect of Clostridium sp. WJ06 as a microbiological detoxification of DON based on the
expression and distribution of proliferating cell nuclear antigen (PCNA) as well as ghrelin in the small intestine. A
total of 24 fattening pigs were randomly divided into three groups. The control group was fed with a basic diet,
the DON group was fed with DON at 5.0 mg/kg in feed, and the DON+C group was provided DON feed with
Clostridium sp. WJ06. Several selected blood parameters, the intestinal morphology, and the expression and
distribution of PCNA and ghrelin, were evaluated. The results proved that the selected blood parameters were
altered, the intestinal villi were damaged, the epithelium was shed, as well as the expression and distribution of
PCNA and ghrelin were changed by DON exposure. These toxic effects were prevented by the addition of
Clostridium sp. WJ06. In short, the addition of Clostridium sp. WJ06 to the feed may eliminate the toxic effects of
[No abstract available]
Mycotoxin contamination causes significant economic loss to food and feed industries and seriously threatens
human health. Aflatoxins (AFs) are one of the most harmful mycotoxins, which are produced by Aspergillus
flavus, Aspergillus parasiticus, and other fungi that are commonly found in the production and preservation of
grain and feed. AFs can cause harm to animal and human health due to their toxic (carcinogenic, teratogenic,
and mutagenic) effects. How to remove AF has become a major problem: biological methods cause no
contamination, have high specificity, and work at high temperature, affording environmental protection. In the
present research, microorganisms with detoxification effects researched in recent years are reviewed, the
detoxification mechanism of microbes on AFs, the safety of degrading enzymes and reaction products formed in
the degradation process, and the application of microorganisms as detoxification strategies for AFs were
email: wuwenda@njau.edu.cn
2-s2.0-85099888451
This study evaluated the efficacy of potentially probiotic fruit-derived Lactobacillus isolates, namely, L. paracasei
108, L. plantarum 49, and L. fermentum 111, to remove aflatoxin M1 (AFM1) from a phosphate buffer solution
(PBS; spiked with 0.15 µg/mL AFM1). The efficacy of examined isolates (approximately 109 cfu/mL) as viable and
non-viable cells (heat-killed; 100◦ C, 1 h) to remove AFM1 was measured after 1 and 24 h at 37◦ C. The recovery
of AFM1 bound to bacterial cells after washing with PBS was also evaluated. Levels of AFM1 in PBS were
measured with high-performance liquid chromatography. Viable and non-viable cells of all examined isolates
were capable of removing AFM1 in PBS with removal percentage values in the range of 73.9–80.0% and
72.9–78.7%, respectively. Viable and non-viable cells of all examined Lactobacillus isolates had similar abilities to
remove AFM1. Only L. paracasei 108 showed higher values of AFM1 removal after 24 h for both viable and non-
viable cells. Percentage values of recovered AFM1 from viable and non-viable cells after washing were in the
range of 13.4–60.6% and 10.9–47.9%, respectively. L. plantarum 49 showed the highest AFM1 retention capacity
after washing. L. paracasei 108, L. plantarum 49, and L. fermentum 111 could have potential application to
Aflatoxin G1 (AFG1), a hepatocarcinogenic mycotoxin, is one of the fundamental supporters of the high pace of
carcinoma hepatocellular. This investigation intended to find the capacity of ten lactic acid bacteria (LAB) species
in expelling or restricting AFG1 from De Mann, Rogosa and Sharpe media (MRS) stock media and to assess the
solidness of LAB/AFG1 complex through storage at 4 and 37°C for 72 and 48 hr individually. The proficiency of
LAB strains in expelling AFG1 from stock media was influenced by the kind of utilized strain and media.
Bifidobacterium longum and Lactobacillus mesenteroides achieved 28% and 27.1% of reduction respectively
within 24 hr at 37°C. The count of LAB cells in MRS broth medium decreased by adding AFG1 (5 µg/L).
Lactobacillus rhamnosus and Lactobacillus mesenteroides favored cold storage to reduce 56.8 and 56% of the
AFG1 content respectively after 72 hr, whereas at 37°C Lactobacillus mesenteroides reduce 35.9% of the AFG1
content after 48 hr. Practical applications: Prevention can only be effective for those mycotoxins that are formed
2-s2.0-85097592798
This research is aimed at bio-detoxifying aflatoxins in artificially contaminated cereals using Lactic Acid Bacteria
(LAB). LAB isolates showed inhibitory activity against toxigenic A. flavus. The in vitro assay revealed noticeable
decrease in the quantity of the aflatoxin B1 (AFB1) and B2 (AFB2) in contaminated millet and sorghum treated
with monoculture and co-culture LAB, respectively. The hematological parameters of the rats fed with
contaminated diets treated with LAB were higher than the group fed with commercial diet. The toxic effect of
aflatoxins on the livers of albino rats fed was ameliorated by the treatment of contaminated cereals with LAB. ©
Research on the ability of lactic acid bacteria (LAB) to bind aflatoxin B1 (AFB1) has mostly been focusing on
lactobacilli and bifidobacteria. In this study, the AFB1 binding capacities of 20 Enterococcus strains belonging to
E. casseliflavus, E. faecalis, E. faecium, E. hirae, E. lactis, and E. mundtii, 24 Pediococcus strains belonging to
species P. acidilactici, P. lolii, P. pentosaceus, and P. stilesii, one strain of Lactococcus formosensis and L. garviae,
and 3 strains of Weissella soli were investigated in MRS broth at 37 °C at 0.2 µg/mL mycotoxin concentration.
According to our results, among non-lactobacilli LAB, the genera with the best AFB1 binding abilities were genus
Pediococcus, with a maximum binding percentage of 7.6% by P. acidilactici OR83, followed by genus
Lactococcus. For AFB1 bio-detoxification purposes, beside lactobacilli, pediococci can also be chosen, but it is
important to select a strain with better binding properties than the average value of its genus. Five Pediococcus
strains have been selected to compare their sterigmatocystin (ST) binding abilities to AFB1 binding, and a 2–3-
fold difference was obtained similar to previous findings for lactobacilli. The best strain was P. acidilactici OR83
Aflatoxins are toxic secondary metabolic products, which exert great hazards to human and animal health.
Decontaminating aflatoxins from food ingredients to a threshold level is a prime concern for avoiding risks to the
consumers. Biological decontamination processes of aflatoxins have received widespread attention due to their
mild and environmental-friendly nature. Many reports have been published on the decontamination of
aflatoxins by microorganisms, especially lactic acid bacteria (LAB), a well-explored probiotic and generally
recognized as safe. The present review aims at updating the decontamination of produced aflatoxins using LAB,
with an emphasis on the decontamination mechanism and influence factors for decontamination. This
comprehensive analysis provides insights into the binding mechanisms between LAB and aflatoxins, facilitating
2-s2.0-85091406405
There is a growing concern regarding the recurrent observation of aflatoxins (AFs) in the milk of lactating
animals. Regarding this, the present study was conducted to assess the aflatoxin M1 (AFM1)-binding ability of
three species, namely Lactobacillus rhamnosus, L. plantarum, and Saccharomyces boulardii, in AFM1-
contaminated milk. The mentioned species were administered at the concentrations of 107 and 109 CFU/mL to
skimmed milk contaminated with 0.5 and 0.75 ng/mL AFM1 within the incubation times of 30 and 90 min at 4°C
and 37°C. Lactobacillus rhamnosus was found to have the best binding ability at the concentrations of 107 and
109 (CFU/ml), rendering 82% and 90% removal in the milk samples with 0.5 and 0.75 ng/ml AFM1, respectively.
Accordingly, this value at 107 and 109 CFU/ml of L. plantarum was obtained 89% and 82% with 0.75 ng/ml of
AFM1, respectively. For S. boulardii at 107 and 109 CFU/ml, the rates were respectively estimated at 75% and
90% with 0.75 ng/ml of AFM1. The best AFM1-binding levels for L. rhamnosus, L. plantarum, and S. boulardii
were 91.82±10.9%, 89.33±0.58%, and 93.20±10.9, respectively, at the concentrations of 1×109, 1×107, and
1×107 CFU/ml at 37, 4, and 37°C, respectively. In this study, the maximum AFM1 binding (100.0±0.58) occurred
while a combination of the aforementioned probiotics was employed at a concentration of 1×107 CFU/ml at
37°C with 0.5 ng/ml AFM1, followed by the combination of L. rhamnosus and L. plantarum (95.86±10.9) at a
Furukawa et al. identified mitochondrial ClpP as the target molecule of dioctatin, an inhibitor of aflatoxin
production of fungi. They revealed that dioctatin activates ClpP to degrade mitochondrial energy-related
proteins, such as a component of respiratory chain complexes, which causes metabolic disturbances and
reduction of aflatoxin production. © 2020 Elsevier LtdAflatoxin contamination of crops is a serious problem
worldwide. Utilization of aflatoxin production inhibitors is attractive, as the elucidation of their modes of action
contributes to clarifying the mechanism of aflatoxin production. Here, we identified mitochondrial protease ClpP
as the target of dioctatin, an inhibitor of aflatoxin production of Aspergillus flavus. Dioctatin conferred
uncontrolled caseinolytic capacity on ClpP of A. flavus and Escherichia coli. Dioctatin-bound ClpP selectively
degraded mitochondrial energy-related proteins in vitro, including a subunit of respiratory chain complex V,
which was also reduced by dioctatin in a ClpP-dependent manner in vivo. Dioctatin enhanced glycolysis and
alcohol fermentation while reducing tricarboxylic acid cycle metabolites. These disturbances were accompanied
by reduced histone acetylation and reduced expression of aflatoxin biosynthetic genes. Our results suggest that
The potential Aflatoxin B1 (AFB1) binding Lactobacillus fermentum (LC5/a) was used for in vivo AFB1 binding and
detoxification in presence of chlorophyll (CL) in male Swiss albino mice. Mice were randomly divided into seven
groups. The control groups (CL, AFB1 and LC5/a) received chlorophyll (250 μg/kg b.w), AFB1 (100 μg/kg b.w) and
LC5/a (1 × 108 CFU) for 21 days. The treatment group (AFB1+LC5/a) received 100 μl of lyophilized bacterial
suspension (1 × 108 CFU) 2 h before the AFB1 dosage (100μg/kg b.w). The chlorophyll mice group (CL + AFB1)
was given single oral dose of CL (250 μg/kg b.w) before AFB1 dosage and last mice group received the
combination of CL + LC5/a before the AFB1 dosage over a period of 21 days. Ballooning of cytoplasm and
necrosis in liver was evident in histopathological examination of AFB1 mice group, while, marked improvement
and nearly normal histology were seen in LC5/a and CL treated mice group. The levels of AST, ALT, GST, and SOD
were increased in AFB1 mice group compared to LC5/a and CL treated mice group. Elevated levels of pro-
inflammatory cytokines, TNF-α, IL-12, IL-6 (324, 506, 117.25 pg/ml) were observed in AFB1 treated mice serum
Aim: Recently, higher contamination by aflatoxin M1 (AFM1) has been detected in many countries.
Unfortunately, many tons of contaminated milk and milk byproducts are removed from the food chain to avoid
human contamination; as a consequence of higher economic losses. Fewest number of studies are interested to
AFM1 detoxification using lactic acid bacteria. Materials and methods: In this study, AFM1-degradation using
Lactobacillus paracasei BEJ01 (LPBEJ01) was tested in vitro. The preventive effect of LPBEJ01 against AFM1
immunobiological effects in mice are treated orally during 3 weeks with 100 µg AFM1, LPBEJ01 (2 ×
109 CFU/ml∼2 mg/kg p.c.) and a mixture of AFM1 and LPBEJ01. Results: In vitro LPBEJ01 was found able to
absorb 98% of AFM1 (100 µg/ml) in liquid medium after 24 h and more than 95% of AFM1 could be eliminated
after 24 h in a solid-state fermentation. Animals treated with AFM1 obtained lower body weight than the control
ones. The mitogenic response of spleen mononuclear cells (SMCs) in vivo was higher in mice treated with AFM1.
The SMC of mice treated with AFM1 produced lower levels of IL-2, higher levels IL-4 and no effect on IL-10
production. The peritoneal macrophages of mice that treated with AFM1 released less H2O2, while mice
exposed orally with the mixture of AFM1 and LPBEJ01 produced higher levels. Conclusion: LPBEJ01 was safe and
Broiler chicken production is one of the most lucrative food production industries globally because of the
demand for poultry products. Stringent regulations pertaining to the use of antibiotic growth promoters (AGP) in
livestock production coupled with the changing consumer trends in terms of an increase in the consumtion of
AGP-free meat pose a challenge to the poultry industry. Probiotics, specifically from the genus Bacillus, are
emerging as a feasible solution to address this challenge because their spore-forming capabilities afford them
several advantages over conventional probiotics. Success of these organisms has been attributed to their
possession of a myriad of mechanisms that elicit probiotic effects, including among others, competitive exclusion
of common poultry pathogens, improvement in digestion and absorption via the production of exogenous
enzymes, improvement of intestinal morphology, immunomodulation, and the reduction of toxic compounds
such as ammonia and aflatoxins. These effects reportedly reduce disease and mortality, improve feed efficiency
up to 5%, enhance health, and assist in the environmental sustainability of poultry production. Bacillus spp. are
easily isolated from environments and have up to 100% survivability in the harsh conditions of the
gastrointestinal tract. Besides these important considerations, the key advantage for the use of Bacilli as feed
probiotics is their robust nature pertaining to industrial production because of the high-density spore
production, spores can be produced in excess of 1 × 1011 spores•mL−1. Furhtermore spores can retain
approximately 90% viability during the probiotic harvesting process. In addition, these spores remain stable at a
concentration of 1 × 109 spores•mL−1 when formulated into probiotic products, with a shelf life potential of 5 yr.
To investigate the effects of oral administration of probiotics consortium on lipid metabolism in aflatoxin B1
(AFB1) exposed rats, ninety female albino rats were first grouped into two: NC (control fed standard feed) and
AF (fed AFB1-contaminated feed at 40 ppb). After eight weeks, baseline animals were sacrificed from both
groups while the others further divided into four groups - NC treated with and without the probiotics
consortium, aflatoxin treated with and without the probiotics consortium (NCT, NCC, AFT, and AFC respectively).
Five animals from each group were sacrificed weekly for four weeks, with the collection of blood, liver, brain,
and the small intestine. Administration of probiotics instigated significant (p < 0.05) reductions in the elevated
plasma and organ lipids as well as HDL-TAG and VLDL + LDL CHO concentrations of animals exposed to AF. AF-
induced hepatic lipogenesis and up-regulation of 3-hydroxy-3-methylglutaryl-CoA reductase activity were also
significantly (p < 0.05) attenuated following treatment with probiotics in a time-dependent manner. Moreover,
neither AF nor probiotics had any effect on glycerol-3-phosphate acyltransferase. Lipid peroxidation was
Animal feeds may contain exogenous compounds that can induce toxicity when ruminants ingest them. These
toxins are secondary metabolites originating from various sources including plants, bacteria, algae and fungi.
Animal feed toxins are responsible for various animal poisonings which negatively impact the livestock industry.
Poisoning is more frequently reported in newly exposed, naïve ruminants while ‘experienced’ ruminants are
observed to better tolerate toxin-contaminated feed. Ruminants can possess detoxification ability through
rumen microorganisms with the rumen microbiome able to adapt to utilise toxic secondary metabolites. The
ability of rumen microorganisms to metabolise these toxins has been used as a basis for the development of
preventative probiotics to confer resistance against the poisoning to naïve ruminants. In this review,
detoxification of various toxins, which include plant toxins, cyanobacteria toxins and plant-associated fungal
mycotoxins, by rumen microorganisms is discussed. The review will include clinical studies of the animal
poisoning caused by these toxins, the toxin mechanism of action, toxin degradation by rumen microorganisms,
reported and hypothesised detoxification mechanisms and identified toxin metabolites with their toxicity
The purpose of this study was to evaluate the ability of heat-killed cells (121 °C, 10 min) from two strains of lactic
acid bacteria (LAB) (Lactobacillus rhamnosus and Lactococcus lactis) and one strain of yeast (Saccharomyces
cerevisiae), alone or in combination, to reduce the levels of aflatoxin M1 (AFM1) in Frescal cheese during 30
days of storage. The experimental design was totally randomized, in a 2 × 2 × 2 factorial arrangement,
corresponding to two levels of LAB (0 and L. rhamnosus at 1010 cells/kg + L. lactis at 1010 cells/kg), two levels of
S. cerevisiae in milk (0 and 1010 yeast cells/kg) and two AFM1 levels (0 and 0.5 µg/kg) added to the cheese curd,
totaling 8 treatments with three replicates per treatment. AFM1 levels in Frescal cheese were evaluated by using
a high-performance liquid chromatography. Cheese fat and protein contents were not affected (P &gt; 0.05) by
any of the treatments, and only pH decreased (P &lt; 0.05) in all treatments from days 2 to 30 of storage (usual
shelf life of this type of cheese). AFM1 levels detected in contaminated cheeses decreased on day 2 of storage,
varying from 0.09 µg/kg (cheese with addition of bacterial cells) to 0.29 µg/kg (no addition of LAB or yeast cells),
this may have occurred due to loss of AFM1 in the Frescal cheese whey. The concentrations of detected AFM1
decreased (P &lt; 0.05) in all treatments from days 2 to 10 of storage, and the maximum percentage reduction of
the detectable levels (100%) was achieved after 10 and 20 days of storage in cheeses containing LAB and yeast
In this study, the application of ozonation, ultraviolet radiation, and pulsed electric field techniques on the
elimination of total Aflatoxins (AF) and AFM1 in probiotic milk (Acidophilus milk) has been investigated. A central
composite response surface analysis was performed to determine the effects of these methods on the reduction
of total AF and AFM1 and also, the viability of bacteria using Design-Expert software. Results showed a
significant and synergistic effect on all independent variables in reducing the amounts of AFM1 and total AF in
acidophilus milk. The implemented numerical optimization showed that the predicted and actual values were
similar and confirmed the possible application of PEF, UV intensity, and ozone for reducing toxins in acidophilus
milk. Bacterial viability also decreased and the level of Lactobacillus acidophilus in the final product was
106 CFU/g. The optimized parameters were 13.15 µsec for pulse duration, 9.99 mg/min for ozone concentration,
and 4.99 J/cm−2 for UV intensity. Practical applications: Dairy products are an important part of people's daily
diet. AFM1 is a derivation of 4-hydroxy AFB1 and AFB2 and contaminated animals ‘milk. This study was
investigated the use of Hurdle technologies (ozonation, UV radiation, and pulsed electric field) to reduce the
amount of AF in milk containing acidophilus bacteria. The results showed a significant and synergistic effect on
The objective of this study was to determine the effects of a high salt culture medium (fish sauce and salted fish)
on the detoxification capacity of Lactobacillus acidophilus (L.a), L. bulgaricus (L.b), and L. casei (L.c). These lactic
acid bacteria (LAB) strains were mixed with dried fish and fish sauce containing aflatoxin B1 (AFB1) and T-2 toxin
(T-2) and cultured at room temperature for 24 h. The toxin concentrations in the media were quantified using LC-
MS/MS following the incubation. Compared with the MRS broth, the growths of L.a, L.b, and L.c were inhibited
in the salted fish and fish sauce media. As the toxin concentration in the MRS broth increased, the toxin removal
rates for L.a, L.b, and L.c increased, especially those for the mixed L.a, L.b, and L.c (L.abc). Moreover, the abilities
of L.a, L.b, and L.c to remove AFB1 in the MRS broth was significantly stronger than that for T-2, indicating the
mycotoxin toxicity enhanced the detoxification ability of LAB. Unlike the MRS broth, salted fish and fish sauce
Application of probiotics in the food industry has been hampered by their sensitivity to challenging conditions
that reduce their vitality in food matrices. A lot of attempts have been made to promote the growth of these
probiotics in the aspect of nutrition demands. Among the other adverse conditions, oxygen stress can restrict
the growth of probiotics and has not yet been paid enough attention to. In this study, the effect of a petunidin-
based anthocyanin (ACN) on the growth of probiotic Lactobacillus plantarum ST-III was investigated under
oxygen stress. The growth of ST-III was analyzed through spot assay on agar plates as well as plating-based
enumeration of the viable cells in the liquid culture. Results indicated that ACN could efficiently improve the
growth of ST-III under oxygen stress, whereas no effect was observed in the absence of oxygen stress. Further
investigations indicated that ACN reduced the oxido-reduction potential of the culture; meanwhile, it exerted a
positive transcriptional regulation on the thioredoxin system of ST-III, leading to a decrease in reactive oxygen
species accumulation within the cells. Moreover, ACN enabled the growth of ST-III in reconstituted skim milk and
promoted the formation of milk clots. These results revealed the role of a petunidin-based ACN in oxygen stress
The present study was conducted to investigate the ability of two probiotic strains, L. acidophilus PTCC 1643 and
L. rhamnosus PTCC 1637, to bind aflatoxin B1 (AFB1, 20 ng/ml) in comparison with yogurt starter cultures, at
equal bacterial count (~ 109 LogCFU/ml) during a 21-day storage period at 4 °C. All assessed treatments
exhibited high percentages of AFB1-binding, ranged from 64.56 to 96.58%. However, the ability of probiotic
bacteria was statistically higher than yogurt starter cultures. Aflatoxin binding ability of the selected lactic acid
bacteria was dependent on both time and bacteria species. The highest and the lowest percentages of AFB1-
removal was observed at 11th day of cold storage by L. rhamnosus (96.58 ± 3.97%) and at the first day of storage
for yogurt starter cultures (64.56 ± 5.32%), respectively. The stability of bacterial cells-AFB1 complex was
Probiotic strain Eurotium cristatum was isolated from Chinese Fuzhuan brick-tea and tested for its in vitro
activity against aflatoxigenic Aspergillus flavus. Results indicated that E. cristatum can inhibit the radial growth of
A. flavus. Furthermore, this inhibition might be caused by E. cristatum secondary metabolites. The ability of
culture filtrate of strain E. cristatum against growth and aflatoxin B1 production by toxigenic A. flavus was
evaluated in vitro. Meanwhile, the influence of filtrate on spore morphology of A. flavus was analyzed by
scanning electron microscopy (SEM). Results demonstrated that both radial growth of A. flavus and aflatoxin B1
production were significantly weakened following increases in the E. cristatum culture filtrate concentration. In
addition, SEM showed that the culture filtrate seriously damaged hyphae morphology. Gas chromatography
mass spectrometry (GC/MS) analysis of the E. cristatum culture supernatant revealed the presence of multiple
antifungal compounds. Real-time quantitative polymerase chain reaction (RT-qPCR) analysis showed that the
expression of aflatoxin biosynthesis-related genes (aflD, aflQ, and aflS) were down-regulated. Importantly, this
latter occurrence resulted in a reduction of the AflS/AflR ratio. Interestingly, cell-free supernatants of E.
cristatum facilitated the effective degradation of aflatoxin B1. In addition, two degradation products of aflatoxin
The aim of the study was to investigate the effect of Bacillus licheniformis and salinomycin supplementation in
broiler diets as individual factors or in combination on the growth performance, GIT morphometry, and
microbiota populations. Four hundred one-day-old Ross 308 chicks were randomly distributed to four dietary
treatments (10 replicates, 10 birds each). The following treatments were applied: NC—no additives; NC +
SAL—salinomycin addition (60 mg/kg diet), NC + PRO—B. licheniformis DSM 28710 preparation (1.6 × 109
CFU/kg; 500 mg/kg diet), and NC + SAL + PRO—combination of salinomycin and B. licheniformis. Probiotic
administration resulted in improvement (p &lt; 0.05) of the performance parameters, including body weight gain
(1–10 d, and 11–22 d) and feed conversion ratio (11–22 d, 1–36 d). An interaction (p &lt; 0.05) between
experimental factors was observed in terms of lower pH values in the crop (tendency, p = 0.053) and ceca. Both
factors lowered the alpha diversity and Enterobacteriaceae and promoted Bacillaceae communities in the
jejunum (p &lt; 0.05). Interactions were also observed in terms of reducing Clostridiaceae in the ceca. In
Aflatoxin B1 (AFB1) is the most harmful mycotoxin. Aflatoxin occurrence in tea makes this beverage unsuitable
for consumption and presented risks to human health. Therefore, researches in aflatoxin microbial degradation
are necessary to overcome this problem. Kombucha beverage is associated with health promoting effects. Thus,
novel strains (Lactic acid bacteria and yeasts) were isolated from a Kombucha culture and assessed for AFB1
degradation in the liquid medium (Man Rogosa and Sharpe broth, yeast extract peptone dextrose broth and
black tea). The main strains involved in AFB1 decontamination were identified based on DNA sequencing and the
toxicity of the new products was evaluated on Hep2 cells and on Brine shrimp (Artemia salina). Our results
showed that after 7 days of fermentation, kombucha was able to degrade 97% of AFB1 in black tea. Moreover,
the effective yeasts present in Kombucha were identified as Pichia occidentalis, Candida sorboxylosa and
Hanseniaspora opuntiae and the highest AFB1 degradation capacity was accorded to P. occidentalis (59%) when
cultivated in black tea. Data on cytotoxicity tests on Hep2 cells and Brine shrimp (Artemia salina) showed that
the biodegraded products were less toxic than pure AFB1. These findings suggest that, kombucha isolated strains
In order to alleviate toxic effects of aflatoxins B1 (AFB1) and zearalenone (ZEA) on broiler production
performance and gut microbiota, three kinds of compound probiotics (CP) were selected. The optimal ratios of
Bacillus subtilis, Lactobacillus casei and Candida utilis in broiler diets were 7, 5 and 6 log CFU/g for ZEA
biodegradation (CP1); 6, 7 and 7 log CFU/g for AFB1 biodegradation (CP2); 7, 6 and 7 log CFU/g for ZEA + AFB1
biodegradation (CP3). A total of 350 1-day-old Ross broilers were randomly divided into 7 groups. Group A was
the basal diet, group B-G contained ZEA, AFB1, ZEA + AFB1, ZEA + CP1, AFB1+CP2, ZEA + AFB1+CP3, respectively.
The experiment showed that AFB1 or AFB1+ZEA significantly decreased broiler production performance,
damaged liver and jejunum, increased mycotoxin residues in broiler body; however, three kinds of compound
probiotics additions could alleviate mycotoxin negative effects on the above parameters (p &lt; 0.05). The gut
microbiota analysis indicated that AFB1+ZEA increased jejunal microbial richness, but which were decreased to
almost the same level as the control group by CP3 addition. CP3 addition significantly increased jejunal
Firmicutes and Lactobacillus aviarius abundances. The correlative analysis showed that gut Lactobacillus aviarius
abundance was positively correlated with average daily gain (ADG) of broilers (p &lt; 0.05), while AFB1+ZEA
addition decreased its relative abundance, indicating that CP3 addition increased broiler growth by increasing
Lactobacillus aviarius abundance. AFB1 and ZEA residues in broiler body were negatively correlated with the gut
beneficial bacterial abundances (p &lt; 0.01), but positively correlated with the potentially harmful bacterial
Aflatoxin M1 (AFM1) in milk and milk products has been recognised as an issue for over 30 years. Controlling
AFM1 in milk is important to protect human health and trade. Preventing contamination by avoiding fungal
contamination of cattle feed is the best method of control, however this is hard to avoid in some countries.
Treating milk containing AFM1 is an alternative control measure, however, there is no single approved method.
The challenge is to select a treatment method that is effective but does not affect the organoleptic quality of
milk. This study reviews the strategies for degrading AFM1 in milk including yeast, lactic acid bacteria, enzyme,
peroxide, ozone, UV light and cold plasma. This review compares the efficacy, influencing factors, (possible)
mechanisms of activity, advantages, limitations and potential future trends of these methods and provides some
Aflatoxins are secondary metabolites of fungi that are the most dangerous mycotoxin and food safety
challenges. Human exposure to mycotoxins occurs directly throughout the intake of contaminated agricultural
products or indirectly throughout the consumption of products prepared with animal origin or obtained from
animals that were fed with contaminated material. For detoxification and reducing threats to public health and
the economic damage caused by the aflatoxins in animal and plants food products, different techniques
(physical, chemical and biological) has been studied. All of these methods, by modifying and destroying the toxin
molecular structure, would inhibit its transfer to the digestive system and could reduce the accessibility of toxins
to the target tissue and eliminate it. In terms of the overarching challenges presented by the aflatoxins (AFs)
contamination in foods and feeds, there is an urgent need to evolve cost-effective and appropriate strategies to
combat this hazard. The review addresses have been noted the pathogenicity of AFs and the plausible
mechanism of their-induced toxicity. Furthermore, assessed the AFs degradation using probiotic bacteria of their
Mycotoxin residues are transferred from feed to animal products, yet, less attention has been paid to it in
developing countries. There is a need to find alternative alleviation material for reducing the impact of
mycotoxin. This review is meant to elucidate different additives that can reduce mycotoxin residue in animal
products in the world, especially in developing countries. There is evidence of relationship between mycotoxin
residue in breast milk of nursing mothers and mycotoxin exposure through crop and animal product (egg and
milk) intake, especially in Asia, Africa, Middle East, Latin America, and some parts of Europe. Younger livestock
tends to have more toxin residues in their tissue compared to older ones. Grazing animal are also exposed to
mycotoxin intake which corresponds to high level of mycotoxins in their products including meat and milk. This
review shows that phytogenic, probiotic, and prebiotic additives can decrease mycotoxin residues in milk, eggs,
meat liver and other tissues of livestock. Specifically, bentonites, difructose anhydride III, yeast (Trichosporon
mycotoxinivorans), Bacillus spp., or their biodegradable products can reduce mycotoxin residue in animal
products. In addition, Ally isothiocyanates from mustard seed were able to mitigate mycotoxins in silo-simulated
system. Evidence shows that there are now low-cost, accessible, and eco-friendly additives, which could alleviate
Galactan exopolysaccharide (EPS) produced by Weissella confusa KR780676 isolated from an Indian traditional
fermented food has been reported earlier. In this manuscript, we have studied aflatoxin-binding ability of this
galactan EPS. Aflatoxin B1 (AFB1) binding ability of galactan EPS was observed in an increasing trend with
increasing EPS concentration (20–100 mg/mL). At lower concentrations (&lt; 20 mg/mL) of EPS, the binding
activity was undetectable, while notable binding was seen from 30 mg/mL. Enhanced AFB1 binding (32.40%) was
recorded at 50 mg/mL of EPS and it increased gradually up to 34.79% at 100 mg/mL concentrations of EPS. The
intensity of bands in high-performance thin-layer chromatography (HPTLC) analysis confirms the AFB1 binding
efficiency of galactan EPS, which shows its potential application for removal of toxins in food and feed industry.
Galactan EPS binding activity to AFB1 is further studied with particle size analysis (PSA). This is the first study
Aflatoxins (AFs) are secondary metabolites produced by Aspergillus spp., known for their hepatotoxic,
carcinogenic, and mutagenic activity in humans and animals. AF contamination of staple food commodities is a
global concern due to their toxicity and the economic losses they cause. Different strategies have been applied
to reduce fungal contamination and AF production. Among them, the use of natural, plant-derived compounds is
emerging as a promising strategy to be applied to control both Aspergillus spoilage and AF contamination in food
and feed commodities in an integrated pre- and postharvest management. In particular, phenols, aldehydes, and
terpenes extracted from medicinal plants, spices, or fruits have been studied in depth. They can be easily
extracted, they are generally recognized as safe (GRAS), and they are food-grade and act through a wide variety
of mechanisms. This review investigated the main compounds with antifungal and anti-aflatoxigenic activity, also
elucidating their physiological role and the different modes of action and synergies. Plant bioactive compounds
are shown to be effective in modulating Aspergillus spp. contamination and AF production both in vitro and in
In recent decades, probiotics have attracted widespread attention and their application in healthcare and animal
husbandry has been promising. Among many probiotics, Bacillus coagulans (B. coagulans) has become a key
player in the field of probiotics in recent years. It has been demonstrated to be involved in regulating the balance
of the intestinal microbiota, promoting metabolism and utilization of nutrients, improving immunity, and more
importantly, it also has good industrial properties such as high temperature resistance, acid resistance, bile
resistance, and the like. This review highlights the effects of B. coagulans in animal husbandry and its underlying
Chronic exposure of children in sub-Saharan Africa to aflatoxins has been associated with low birth weight,
stunted growth, immune suppression, and liver function damage. Lactobacillus species have been shown to
reduce aflatoxin contamination during the process of food fermentation. Twenty-three Lactobacillus strains
were isolated from fecal samples obtained from a cohort of rural Ugandan children at the age of 54 to 60
months, typed by 16S rRNA gene sequencing, and characterized in terms of their ability to bind aflatoxin B1 in
vitro. Evidence for chronic exposure of these children to aflatoxin B1 in the study area was obtained by analysis
of local foods (maize flour and peanuts), followed by the identification of the breakdown product aflatoxin M1 in
their urine samples. Surprisingly, Lactobacillus in the gut microbiota of 140 children from the same cohort at 24
and 36 months showed the highest positive correlation coefficient with stunting among all bacterial genera
identified in the stool samples. This correlation was interpreted to be associated with dietary changes from
The aim of the following research was to determine the detoxification properties of probiotic Lactobacillus sp.
bacteria (12 strains) and S. cerevisiae yeast (6 strains) towards mycotoxins, such as aflatoxin B1, deoxynivalenol,
fumonisins, T-2 toxin and zearalenone, which pose as frequent feed contamination. The experiment involved
analysing changes in concentration of mycotoxins in PBS solutions, after 6, 12 and 24 h of incubation with
monocultures of tested microorganisms, measured by high-performance liquid chromatography (HPLC). We
found that all strains detoxified the mycotoxins, with the highest reduction in concentration observed for the
fumonisin B1 and B2 mixture, ranging between 62 and 77% for bacterial strains and 67–74% for yeast. By
contrast, deoxynivalenol was the most resistant mycotoxin: its concentration was reduced by 19–39% by
Lactobacillus sp. strains and 22–43% by yeast after 24 h of incubation. High detoxification rates for aflatoxin B1,
T-2 toxin and zearalenone were also observed, with concentration reduced on average by 60%, 61% and 57% by
Lactobacillus, respectively, and 65%, 69% and 52% by yeast, respectively. The greatest extent of reduction in the
concentration for all mycotoxins was observed after 6 h of incubation; however, a decrease in concentration was
Food and feed contamination by aflatoxins represents a great challenge for human and animal health. Aflatoxins
detoxification using probiotic bacteria and yeasts has been introduced as an inexpensive and promising method.
This article is organized with an overview of the potential application of probiotic bacteria and yeasts to
eliminate, inactivate or reduce the bioavailability of aflatoxins, especially aflatoxin B1, in vitro and in vivo. Also, a
fast glance to beneficial health effects and preservative properties of probiotics followed by the mechanism of
binding of aflatoxins by probiotics, influence of different probiotic pretreatments, and the stability of aflatoxin-
Introduction: Many fungi infect the wheat grains. Under field and or storage conditions from temperature and
humidity, some fungi can produce aflatoxins (AFs), which may cause acute or chronic diseases. Therefore, there
is a necessary and urgent need to find an effective and safe way to reduce or remove AFs. Objective: The
objective of this study was the evaluation of Lactobacillus rhamnosus, Lactobacillus gasseri, and Lactobacillus
plantarum for their ability to reduce and or remove AFs produced by Aspergillus flavus and Aspergillus
parasiticus, which were isolated from wheat grains, as well as control of AFs produced on affected wheat grain
by A.parasiticus spores only. Methods: LAB, isolated from some local dairy products, were cultured in MRS for
the evaluation of their ability to remove AFs, produced by A. flavus and A. parasiticus on (YES) media, in addition
to the treatment of wheat grains by LAB cells to prevent AFs produced by A. parasiticus. Results: The L.
rhamnosus strain gave the highest reduction rates of AFs produced by A. parasiticus that were 62.6, 44.4, 43.3,
and 52.2% for AFG1, AFB1, AFG2, and AFB2, respectively. While in the case of A. flavus, the reduction was 50.4,
42.7, 40.6, and 36.8% in the same order of toxins. When applied, these strains with wheat grains were affected
by A. parasiticus, the inhibition rates of AFs were ranged between 61.4 and 75.8% with L. rhamnosus strain and
43.7 to 52.1% with L. gasseri, while L. plantarum strain ranged from 55.5 to 66.9%. Conclusion: According to this
Aflatoxin M1 (AFM1) is a highly toxic milk contaminant that poses a grave threat to human health. Among
various strategies proposed for AFM1 decontamination, use of microbes have been considered to be one of the
suitable techniques. However, the use of microbial cells alone had not proven to be efficient at higher AFM1
levels. Moreover, the complex between microbial cell and AFM1 had been suggested to be weaker and is
dissociated soon after its exposure to washings. The objective of this study was to enhance the AFM1 binding
efficiencies of bacterial cells (C) belonging to Lactobacillus paracasei and Bacillus coagulans in the presence of
activated carbon (CAC), bentonite (CBENT) and sorbitan monostearate (CSP60). The reduction of AFM1 was
found to be directly proportional to the concentration of microbial cells. Heat killed and acid treated L. paracasei
successfully reduced AFM1 in milk spiked at 0.2 µg/L to 89% and 100% in CBENT, 84% and 90% in CAC, 59% and
47% in CSP60 and, 51.5% and 42% in C, respectively. Among treatments involving B. coagulans, acid treated
CSP60 proved to be least effective showing 44.6% reduction, while CBENT for both acid and heat treated along
with acid treated CAC proved to be most effective by removing 100% AFM1. CBENT and CAC (acid and heat
killed) among both bacterial strains showed the formation of most stable complex with AFM1 showing no
release of detectable AFM1 after couple of phosphate buffer saline (PBS) washings. Among other treatments,
CSP60 of heat killed cells formed most stable complex for both L. paracasei and B. coagulans with 19% and 22%
Adsorption of molecules to the cell walls of microorganisms plays an important role in helping to prevent animal
exposure to the toxic and carcinogenic effects of aflatoxins (AFs). The aim of this study was to evaluate the
ability of LAB strains, isolated from brewers’ grains, to adsorb aflatoxin B1 (AFB1). All LAB were able to reduce
the bioavailability of AFB1 from phosphate buffered-saline (PBS). In addition, the strains retained their
effectiveness even after heat treatment. The AFB1-LAB complex stability was first evaluated through sequential
washing steps. These assays demonstrated that a low percentage of AFB1 was released after consecutive
washes. After subjecting the complex to different pH and bile salt treatments, the percentage of bound AF
decreased, as compared to the control, but remained at high levels. Finally, to simulate the formation of the
AFB1-LAB complex at conditions similar to those of the gastrointestinal tract, LAB and AFB1 were homogenized
in PBS adjusted at acidic conditions or under different bile salt concentrations. In general, LAB strains showed
the highest AFB1 adsorption at the lowest pH (2) and bile salt concentration (0.05%). In conclusion, the studied
Purpose: The present study was conducted to assess the ability of probiotic bacteria and yeasts strains to reduce
aflatoxin B1 (AFB1) in gastrointestinal simulated conditions. Aflatoxins are potent carcinogenic and
immunosuppressive agents. Acute exposure to a high level of aflatoxins leads to aflatoxicosis, which cause rapid
death due to liver failure. It is anticipated that consumption of probiotic microorganisms capable of binding
aflatoxins can reduce the risk of AFB1 on human health to a certain extent. Methods: For this purpose, the
bacteria (1 × 1010 cfu/mL) and yeasts count (2 × 108 cells/mL) and AFB1 concentration (10 ppb) were adjusted.
Then, the samples were incubated in the simulated medium, human gastric secretions and small intestine. The
concentration of residual AFB1 was determined using enzyme-linked immunosorbent assay (ELISA). The results
were statistically analyzed by SPSS 16 software. Results: The native isolated bacteria and yeasts in the simulated
gastrointestinal tract condition showed a significant effect on AFB1 reduction (P&lt; 0.05). The AFB1 reduction
ability of native probiotic microorganisms was strain dependent. The highest binding ability in bacteria belonged
to Lactobacillus rhamnosus (31.14%) and at yeasts belonged to Saccharomyces cerevisiae (30.46%). Conclusion:
The use of probiotic strains is the appropriate biological method to reduce AFB1 in the human gastrointestinal
tract. Probiotic bacteria could help to decrease the harmful effects of AFB1 in humans through enhancing the
food safety. © 2020 The Author (s). This is an Open Access article distributed under the terms of the Creative
2-s2.0-85088617905
[No abstract available]
Aflatoxins are wide-spread harmful carcinogenic secondary metabolites produced by Aspergillus species, which
cause serious feed and food contaminations and affect farm animals deleteriously with acute or chronic
manifestations of mycotoxicoses. On farm, both pre-harvest and post-harvest strategies are applied to minimize
the risk of aflatoxin contaminations in feeds. The great economic losses attributable to mycotoxin
contaminations have initiated a plethora of research projects to develop new, effective technologies to prevent
the highly toxic effects of these secondary metabolites on domestic animals and also to block the carry-over of
these mycotoxins to humans through the food chain. Among other areas, this review summarizes the latest
findings on the effects of silage production technologies and silage microbiota on aflatoxins, and it also discusses
the current applications of probiotic organisms and microbial products in feeding technologies. After ingesting
contaminated foodstuffs, aflatoxins are metabolized and biotransformed differently in various animals
depending on their inherent and acquired physiological properties. These mycotoxins may cause primary
aflatoxicoses with versatile, species-specific adverse effects, which are also dependent on the susceptibility of
individual animals within a species, and will be a function of the dose and duration of aflatoxin exposures. The
transfer of these undesired compounds from contaminated feed into food of animal origin and the aflatoxin
residues present in foods become an additional risk to human health, leading to secondary aflatoxicoses.
Considering the biological transformation of aflatoxins in livestock, this review summarizes (i) the metabolism of
aflatoxins in different animal species, (ii) the deleterious effects of the mycotoxins and their derivatives on the
animals, and (iii) the major risks to animal health in terms of the symptoms and consequences of acute or
chronic aflatoxicoses, animal welfare and productivity. Furthermore, we traced the transformation and
channeling of Aspergillus-derived mycotoxins into food raw materials, particularly in the case of aflatoxin
Thirty-four isolates of Lactobacillus spp. (LAB) from 34 curd samples were evaluated for their aflatoxin B1 (AFB1)
binding and probiotic properties. Upon characterization, four LAB isolates (LC3/a, LC4/c, LC/5a, and LM13/b)
were found to be effective in removing AFB1 from culture media with a capacity of above 75%. Staining reaction,
biochemical tests, pattern of sugar utilization, and 16s rRNA gene sequence analysis revealed the identity of all
the four isolates as L. fermentum. All of them could tolerate acidic pH, salt, and bile, which promise the use of
these probiotic bacterial isolates for human applications. These isolates showed poor hydrophobicity and higher
auto-aggregation properties. All L. fermentum isolates were found susceptible to gentamycin, chloramphenicol,
cefoperazone, ampicillin, and resistant to ciprofloxacin and vancomycin. Results of hemolytic and DNase activity
indicated their nonpathogenic nature. Though all L. fermentum isolates found inhibiting the growth of
Salmonella ebony, Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa, maximum inhibition
was obtained with isolate LC5/a. Kinetic studies revealed that all four bacteria required a minimum of 2 h to
reach stationary phase of AFB1 binding. AFB1 binding ability varied from 66 to 85.2% among these four isolates.
Bile (0.4%) was significant (P ≤ 0.05) in reducing the AFB1 binding property of isolates LC3/a, LC4/c, and LM13/b,
while increased AFB1 binding ability was recorded at acidic pH (2.0). AFB1 binding properties of isolate LC5/a
were found least affected by acidic pH and bile. The findings of our study revealed the higher efficiency of L.
fermentum isolate LC5/a in reducing the bioavailability of AFB1 in gut, and additionally, it improves the
Beer is the most consumed alcoholic beverage worldwide and brewery is a growing industry. Biomass by-
product of beer production is constituted by viable and non-viable flocculated yeasts which are discarded. To
increase the value of this waste, the potential applications of the beer fermentation residue (BFR) as probiotic
and bio-preservative were studied. Strains isolated from commercial brewing starters and BFRs were identified.
The M6 BFR and its constituent strains, Saccharomyces cerevisiae CMUNLPY6.2 and Pichia kudriavzevii
CMUNLPY6.1, proved to be the most resistant to gastrointestinal conditions in vitro. The cell-free supernatants
obtained from micro-fermentations were capable to reduce Aspergillus flavus and Aspergillus parasiticus
germination, two species well-known to produce the potent carcinogenic aflatoxin B1 (AFB1). A cytoprotective
effect of the BFRs against AFB1 on HepG2 cells was observed. Brewing yeasts bound AFB1 in vitro, thus reducing
the cell damage induced by the toxin. Throughout the study, yeasts grown in brewing wort showed better
The detoxification ability of Lactobacillus acidophilus and prebiotics was evaluated by a Plackett-Burman Design
to examine the reduction of concentration and bioaccessibility of aflatoxin B1 (AFB1) and M1 (AFM1) in
artificially contaminated whole cow's milk. Six variables were evaluated: AFB1 (3.25–6.0 μg L−1) or AFM1
(1.0–2.0 μg L−1) concentration; incubation time (0–6 h); and inulin, oligofructose, β-glucan, and polydextrose
concentrations (each between 0.00 and 0.75%). All runs achieved reductions of AFB1 (13.53–35.53%) and AFM1
(17.65–71.52%). Comparing with the positive control, the AFB1 bioaccessibility ranged from 23.68 to 72.67% and
for AFM1 was 0%. The probiotic, isolated or combined with prebiotics, was efficient in mycotoxin reduction,
while the combination of the two reduced the mycotoxin bioaccessibility. The best experimental condition was
This study aimed to evaluate the ability of the Lactobacillus rhamnosus strain LC-4 to bind aflatoxin M1 (AFM1)
in phosphate-buffered saline (PBS) and yogurt. Bacterial cells were subjected to heat, acid, and alkali treatments.
The AFM1-binding rate of acid-treated bacteria was of 78.63 ± 0.52%. The binding of L. rhamnosus LC-4 to AFM1
was partially reversible and the binding of AFM1 was more stable when treated bacteria were used. The involved
components of the cell wall in AFM1 binding were determined, with peptidoglycan playing a critical role. The
integrity of the bacteria was also highly important for detoxification ability, and this ability was influenced by
different factors such as temperature, pH, and toxin concentration, and so on. L. rhamnosus LC-4 retained its
detoxification ability in yogurt. The pH and bacterial concentration slightly affected the binding of AFM1 to L.
rhamnosus LC-4 during storage time. These results indicate that L. rhamnosus LC-4 may be applied to reduce the
concentration of AFM1 in yogurt. Practical Applications: In the present work, we determined the involved
components of the cell wall in AFM1 binding and different factors influencing the binding process. The integrity
of the cell wall is indispensable for AFM1 binding and peptidoglycans were critical components. Understanding
Almonds and peanuts are a rich source of proteins, vitamins and unsaturated fatty acids. However, they can be
also contaminated by mycotoxigenic fungi; a reason that has enhanced to investigate efficient strategies of
management of these fungal contaminations. Some Lactic acid bacteria have been proven capable of inhibiting
growth and mycotoxin production in livestock and transform it into nontoxic derivatives. In this work, four lactic
acid bacteria (LAB) were tested for their abilities to inhibit the growth and mycotoxin production of Aspergillus
flavus and Aspergillus carbonarius. Antifungal activity was evaluated in agar medium as well as in almonds and
peanuts. Results showed that LAB significantly inhibited Aspergillus flavus and Aspergillus carbonarius in agar
medium but none of the strains were able to completely inhibit fungal growth. The highest fungal growth
inhibition was obtained using L. kefiri FR7 (51.67% and 45.56% growth inhibition of A. flavus and A. carbonarius,
respectively). The cell-free supernatants (CFS) from LAB reduced fungal growth with average growth inhibitions
ranging from 13.33% to 40.56% and 12.78% to 37.78% for A. flavus and A. carbonarius, respectively. We noted
also that cell-free supernatants at pH7 (CFS-pH7) from the entire tested LAB did not inhibit fungal growth. L.
kefiri FR7 was the most effective strain in mycotoxin suppression with a reduction percentage reaching 97.22%,
95.27% and 75.26% for AFB1, AFB2 and OTA respectively. Moreover, the inoculation of L. kefiri FR7 in almonds
artificially contaminated with A. flavus decrease 85.27% of AFB1 and 83.94% of AFB2 content after 7 days of
Background: Probiotics play an important role in the human and animal defense against liver damage. However,
the protective mechanism of Lactobacillus plantarum C88 on chronic liver injury induced by mycotoxin remains
unclear. Results: In this study, the addition of L. plantarum C88 obviously ameliorated the increased contents of
alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), total cholesterol
and triglyceride, the diminish contents of total protein and albumin in serum of mice challenged with AFB1.
Simultaneously, L. plantarum C88 attenuated the inflammatory response via significantly reducing the levels of
pro-inflammatory factors, including interleukin-1β (IL-1β), IL-6, IL-8, interferon-γ(IFN-γ) and tumor necrosis
factor-α (TNF-α) in serum. Furthermore, L. plantarum C88 remarkably down-regulated the nuclear factor kappa B
(NF-κB) signaling pathways by weakening the expression of toll-like receptor 2 (TLR2) and TLR4, and inhibited NF-
κB nuclear translocation through enhancing the expression of NF-κB inhibitor (IκB). Neutralization experiments
confirmed that L. plantarum C88 decreased the levels of some pro-inflammatory factors due to the suppression
of the NF-κB signaling pathways. Besides, L. plantarum C88 decreased the levels of Bax and Caspase-3, elevated
the level of Bcl-2, and reduced mRNA expressions of Fatty acid synthetase receptor (Fas), FAS-associated death
domain (FADD), TNF receptor associated death domain (TRADD) and Caspase-8 in the liver. Conclusions:
Probiotic L. plantarum C88 prevented AFB1-induced secretion of pro-inflammatory cytokines by modulating
Aflatoxins continue to be a food safety problem globally, especially in developing regions. A significant amount
of effort and resources have been invested in an attempt to control aflatoxins. However, these efforts have not
substantially decreased the prevalence nor the dietary exposure to aflatoxins in developing countries. One
approach to aflatoxin control is the use of binding agents in foods, and lactic acid bacteria (LAB) have been
studied extensively for this purpose. However, when assessing the results comprehensively and reviewing the
practicality and ethics of use, risks are evident, and concerns arise. In conclusion, our review suggests that there
are too many issues with using LAB for aflatoxin binding for it to be safely promoted. Arguably, using binders in
Aflatoxin M1 (AFM1) is known to be a potent carcinogen and continues to pose a public health concern through
the consumption of contaminated dairy foods. It is anticipated that consumption of lactic acid bacteria capable
of binding aflatoxins can reduce the risk of AFM1 on human health to a certain extent. Seldom reports have
hinted the possibility of using lactic acid bacteria for the biological detoxification of AFM1. Hence, the present
study was conducted to assess the ability of selected probiotic Lactobacillus strains for their AFM1 binding ability
in PBS and to reduce its bioaccessibility in artificially contaminated skim milk using an in vitro digestion model.
Eleven tested probiotic strains illustrated various degrees of AFM1 binding ability ranging from 4.13 to 64.16%.
Five among the 11 probiotic strains were subsequently selected for detailed studies on the basis of highest
binding potential after 24 h of incubation period. The stability of bacterial-AFM1 complex was assessed by
repeated washings with AFM1 free PBS. The observation on bacterial-AFM1 complex stability showed small
release of AFM1 in first and second wash (17.30 to 0.98%) where as none was detectable in the third wash.
However, upon chloroform extraction, 88.57 to 92.30% of bound AFM1 was released from the bacterial cells
which indicate AFM1 binding to the bacterial cell surface rather than absorption or degradation of AFM1 by
bacterial cells. During the in vitro digestion test in skim milk, bioaccessibility of AFM1 was reduced to a scale of
32.61 to 52.84% in the presence of selected strains of probiotic lactobacilli. The present findings suggest that
Objectives: The objectives of our study were to determine the presence of Aflatoxin M1 (AFM1) in market milk
in Aswan province, Egypt and studying the effect of addition of some strains of probiotics microorganisms on
AFM1 level in milk. Materials and Methods: Between July and October 2018, 90 market milk samples (15 Ultra
Heat Treated (UHT), 75 raw) were collected from different dairy shops in Aswan City, Egypt to be examined for
AFM1 presence by rapid strip test and the results were confirmed by high-performance liquid chromatography
(HPLC). Results: The results revealed that all UHT milk samples were negative, while 37 (49%) raw milk samples
were positive for AFM1 residues. All 37 positive milk samples were examined by HPLC to determine the level of
AFM1. The results showed that the level of AFM1 ranged between 0.053 and 0.207 with mean ± SE of 0.1003 ±
0.008 ppb. Some probiotics strains were used to determine their effect on AFM1 by milk fermentation; the
result showed that the probiotics have significant effect on the reduction of AFM1 level in milk (p < 0.05). Also,
Public health importance of AFM1 was discussed. Conclusion: Presence of AFM1 in 49% of examined raw milk
samples indicate widespread occurrence of AFM1 in market milk in Aswan province, Egypt which considered
possible hazards for consumers, while the absence of AFM1 from UHT milk indicates that type of milk is safer.
So, regular monitoring of AFM1 in market milk is necessary for evaluating their contamination status. Mixed
Aflatoxins (AF) are carcinogenic metabolites produced by different species of Aspergillus which readily colonize
crops. AFM1 is secreted in the milk of lactating mammals through the ingestion of feedstuffs contaminated by
aflatoxin B1 (AFB1). Therefore, its presence in milk, even in small amounts, presents a real concern for dairy
industries and consumers of dairy products. Different strategies can lead to the reduction of AFM1
contamination levels in milk. They include adopting good agricultural practices, decreasing the AFB1
contamination of animal feeds, or using diverse types of adsorbent materials. One of the most effective types of
adsorbents used for AFM1 decontamination are those of microbial origin. This review discusses current issues
about AFM1 decontamination methods. These methods are based on the use of different bio-adsorbent agents
such as bacteria and yeasts to complex AFM1 in milk. Moreover, this review answers some of the raised
concerns about the binding stability of the formed AFM1-microbial complex. Thus, the efficiency of the
The aim of this study was to determine the binding ability of three lactic acid bacteria strains (LABs) (L.
plantarum ATCC 10697, B. animalis ATCC 27672, and B. bifidum ATCC 35914) to aflatoxin M1 (AFM1) from
contaminated yoghurt and phosphate-buffered saline (PBS) either alone or in binary combination for different
periods of cold storage (one or ten days) and to investigate the effect of inulin supplementation at 4%
concentration on the AFM1 binding ability of LABs. The AFM1 binding capacities of cultures varied, ranging
between 49.8 ± 2.53% to 60.8 ± 3.58% and 49.5 ± 2.00% to 55.1 ± 1.67%; the best AFM1 binding was observed
with yoghurt starter (YS) + B. bifidum-B. animalis (60.8%) (p < 0.001) and YS + L. plantarum-B. bifidum mixtures
(55.1%) (p < 0.01) after one day and ten days cold storage periods, respectively. Inulin supplementation led to
increased AFM1 binding capacity of YS + B. bifidum (p < 0.01), YS + B. animalis (p < 0.01), and YS + L. plantarum-
B. bifidum (p < 0.001) mixtures at the end of the one-day storage period. Furthermore, the binding capacity of YS
+ B. bifidum-B. animalis, YS + L. plantarum-B. bifidum, YS + L. plantarum-B. animalis increased with statistical
significance at the end of the ten-day storage period by inulin addition (p < 0.05, p < 0.001 and p < 0.001,
The study aims to evaluate the cell-free supernatant (CFS) from Lactobacillus plantarum strain MYS44 against the
growth and aflatoxin production by Aspergillus parasiticus MTCC 411. Standard in vitro techniques revealed the
potential antifungal activity of CFS of LpMYS44. In poison food technique, it was observed that 6% CFS of
LpMYS44 retarded maximum growth. The inhibition of A. parasiticus on peanuts confirmed the ability of CFS of
LpMYS44 for biopreservation. Further, CFS of LpMYS44 was purified by chromatography and analyzed by GC-MS.
The major antifungal compounds were oleic acid, octanoic acid, butanamide, and decanoic acid derivatives.
Twofold concentrated 80 μL of CFS was found to be minimum inhibitory concentration (MIC) of CFS of LpMYS44.
CFS of LpMYS44 suppressed the germination and growth of the spores of A. parasiticus. Microscopic observation
showed that CFS of LpMYS44 severely affected the hyphal wall of A. parasiticus by the leakage of cytoplasmic
content leading to complete destruction. Acidic condition is favorable for CFS of LpMYS44 activity. In poultry
feed sample, CFS of LpMYS44 reduced the aflatoxin B 1 content by 34.2%, reflecting its potentiality to use as
detoxification agent. The multiple antifungal components in CFS of LpMYS44 exhibited antifungal properties
against aflatoxigenic A. parasiticus resulted in causing overall morphological changes. Furthermore, we also
observed the biopreservative ability of CFS of LpMYS44 against A. parasiticus and AFB 1 reduction in for poultry
Mycotoxins are fungal secondary metabolites that pose health risks to exposed individuals, requiring necessary
measures to reduce them. Using liquid chromatography-tandem mass spectrometry (LC-MS/MS), mycotoxins
were quantified in whole grain sorghum and ting subsequently derived from two sorghum varieties (high and
low tannin). The whole grain (WG) ting samples were obtained by fermenting sorghum with Lactobacillus
fermentum strains (FUA 3165 and FUA 3321). Naturally (spontaneously) fermented WG-ting under the same
conditions were equally analysed. Among the mycotoxins investigated, fumonisin B1 (FB1), B2 (FB2), B3 (FB3), T-
2 toxin (T-2), zearalenone (ZEA), alpha-zearalenol (α-ZOL) and beta-zearalenol (β-ZOL) were detected in
sorghum. Results obtained showed that mycotoxin concentrations significantly (p ≤ 0.05) reduced after
fermentation. In particular, L. fermentum FUA 3321 showed the capability to significantly (p ≤ 0.05) reduce all
the mycotoxins by 98% for FB1, 84% for T-2 and up to 82% for α-ZOL, compared to raw low tannin sorghum.
Fermenting with the L. fermentum strains showed potential to effectively reduce mycotoxin contamination in
The aim of the study was to investigate the toxic effects of aflatoxin B 1 (AFB 1 ) and efficacy of a probiotic
preparation containing L. reuteri, L. plantarum, L. pentosus, L. rhamnosus and L. paracasei and Saccharomyces
cerevisiae yeasts to ameliorate their effects in broiler chickens. A total of 168 one-day-old female Ross 308
broilers were randomly allocated to six groups. Three wheat and soybean meal-based diets were prepared:
Control diet and diets contaminated with 1 or 5 mg/kg AFB 1 supplied in moldy wheat. All diets were
unsupplemented or supplemented with probiotic, cold pelleted and fed from 1 to 35 day of life. Feeding diet
with 1 mg AFB 1 /kg did not affect performance, but a diet with 5 mg AFB 1 resulted in a significant reduction of
feed intake and BWG, both diets induced liver and kidneys enlargement. The probiotic supplementation of the
diets partially ameliorated those negative effects and resulted in a significant increase of AFB 1 excretion. It was
accompanied by the reduced level of AFB 1 residues in the liver from 8.9 to 3.7 and from 11.8 to 5.9 µg/kg, in
kidneys from 7.9 to 2.5 and from 13.7 to 4.1 µg/kg in birds fed the less and more contaminated diets,
respectively. AFB 1 exposure caused many severe histopathological changes in the liver and kidneys of broilers,
Inhibition of Aspergillus flavus growth and its aflatoxins production using the biocontrol agent Saccharomyces
cerevisiae as well as to explore its mode of action was studied. Eight strains of S. cerevisiae strains were able to
suppress the growth of A. flavus Z103. The maximum growth inhibition of A. flavus Z103 was obtained by living
cells of S. cerevisiae EBF101 and S. cerevisiae 117 with 85 and 83%, respectively. The sporulation inhibition and
hyphae deterioration of A. flavus Z103 by S. cerevisiae cells adhesion were observed under SEM; up to 99·8%
inhibition of aflatoxins biosynthesis by A. flavus Z103 was resulted when the fungus was treated by autoclaved
extracellular crude of S. cerevisiae. Also, the tested strains are potential to produce exo-chitinase which could be
suggested as another mode of action for its antifungal activity. GC-MS analysis of S. cerevisiae 117 extracellular
secondary metabolites revealed the existence of 4-Hydroxyphenethyl alcohol (46·32%), 4, 4-Dimethyloxazole
(9·14%) and 1,2-Benzenedicarboxylic acid dioctyl ester (2·8%). Significance and Impact of the Study: The use of
Saccharomyces cerevisiae instead of chemical preservatives in fermented food, animal and fish feed and storage
cereal grains could encourage the food industry to produce organic food free of chemical additives. Overall, our
data suggest the possibility of using S. cerevisiae as an alternative treatment in the food industries to control the
dispersion and aflatoxins production by Aspergillus flavus during storage. This method could provide an
The growth of Aspergillus flavus and the production of aflatoxins (AF) during the aerobic deterioration of corn
silage represent a problem for animal and human health. This experiment was conducted to evaluate whether
the presence of A. flavus and AF production originate from the field or additional AF are produced during the
fermentation phase or during aerobic deterioration of corn silage. The trial was carried out in northern Italy on
corn at a dry matter (DM) level of 34%. The fresh herbage was either not treated (C) or treated with a
Lactobacillus buchneri (LB) NCIMB 40788 [(at 3 × 10 5 cfu/g of fresh matter (FM)], Lactobacillus hilgardii (LH)
CNCM I-4785 (at 3 × 10 5 cfu/g of FM), or their combination (LB+LH; at 1.5 × 10 5 cfu/g of FM of each strain)
ensiled in 20-L silos and opened after 250 d of ensiling. After silo opening, the aerobic stability was evaluated
and samples were taken after 7 and 14 d of air exposure. The pre-ensiled material, the silages at silo opening,
and the aerobically exposed silages were analyzed for DM content, fermentative profiles, microbial count,
nutritive characteristics, DM losses, and AFB 1 , AFB 2 , AFG 1 , and AFG 2 contents. Furthermore, a subsample of
colonies with macromorphological features of Aspergillus section Flavi was selected for AF gene pattern
characterization and in vitro AF production. The presence of A. flavus was below the detection limit (&lt;1.00 log
10 cfu/g) in the fresh forage before ensiling, whereas it was found in 1 out of 16 silage samples at silo opening at
a level of 1.24 log 10 cfu/g. The AF were found in both the fresh forage and at opening in all the samples, with a
predominance of AFB 2 (mean value of 1.71 μg/kg of DM). The inoculation of lactic acid bacteria determined a
reduction in the lactic-to-acetic ratio compared with the control. A larger amount of acetic acid resulted in a
lower yeast count and higher aerobic stability in the treated silages than in the control ones. At the beginning of
aerobic deterioration, the yeasts increased to over 5 log 10 cfu/g, whereas the molds were close to the value
observed at silo opening. When the inhibiting conditions were depleted (pH and temperature higher than 5 and
35°C, respectively), both the total molds and A. flavus reached higher values than 8.00 and 4.00 log 10 cfu/g,
respectively, thus determining the ex novo production of AFB 1 during aerobic deterioration, regardless of
treatments. The analysis of gene pattern showed that 64% of the selected colonies of A. flavus showed the
presence of all 4 AF gene patterns, and 43% of the selected colonies were able to produce AF in vitro. During air
Growing broiler chicks on a diet contaminated with aflatoxins (200 or 2000 ppb) for entire growth period
resulted in increased oxidative stress and liver damage markers. The toxicity was subsided in broilers received a
specific aflatoxin-binding probiotic i.e., Lactobacillus plantarum 299v (Lp299v). There was a substantial (30–90%)
increase in antioxidant activity of plasma, which was suppressed due to dietary aflatoxins. Probiotic also reduced
serum lactate dehydrogenase and alanine amino transferase together with lipid peroxidation products in liver,
which were elevated due to aflatoxin. Because of Lp299v consumption, there was ∼20–55% recovery in body
weight gain in broilers intoxicated with aflatoxins. Comparison of the Lp299v effects with that of a commercial
aflatoxin binder revealed that, improved antioxidant activity of the chicks was associated with growth
performance. These data suggest that aflatoxin-binding probiotics are beneficial with multi-functional effects
Background: Aflatoxin contamination in traditionally fermented cereal-based beverages is a serious food safety
challenge considering that commercialization of these products is rising. The challenge is aggravated by the fact
that aflatoxin elimination from the food chain is almost impossible. This paper focuses on Obushera, a popular
traditional spontaneously fermented sorghum – millet beverage from Uganda. Method: Mold and total aflatoxin
levels in cereal flours and Obushera from different markets in Kampala were determined. Ability of lactic acid
bacteria (LAB) starters from Obushera; Lb. plantarum MNC 21, W. confusa MNC 20 and L. lactis MNC 24 to bind
aflatoxin B1 (AFB1) was evaluated against Lb. rhamnosus yoba 2012 as the reference strain. Results: Mold counts
in sorghum, millet and Obushera ranged between 0.0–2.4 log cfu/g, 2.0–6.5 log cfu/g and 2.0–5.5 log cfu/g,
respectively. None of the flours complied with food safety standards for molds (maximum = 4 log cfu/g) while
88.0% of Obushera did (standard = maximum 1.3 log cfu/g). Total aflatoxin levels in sorghum, millet and
Obushera were 22.3 ± 21.2 ppb, 9.9 ± 10.0 ppb and 10.4 ± 6.1 ppb, respectively. LAB bound 19.3–69.4% of AFB1
in a 1000 ppb solution with binding efficiency in the order of Lb. rhamnosus yoba 2012 = Lb. plantarum MNC 21
> W. confusa MNC 20 = L. lactis MNC 24. The LAB-AFB1complex remained stable under a series of washes with
This research focused on biocontrol solution to increase food safety through studying lactic acid bacteria (LAB)
which can bind aflatoxins in milk. Aflatoxins are toxic contaminants found in feeds and foods. In milk aflatoxin is
found in metabolised form, aflatoxin M1 (AFM1). Three indigenous LAB Lactobacillus strains and one
Lactococcus strain isolated from Kenyan spontaneously fermented foods were tested for their AFM1 binding
abilities in different conditions and after different treatments along with two reference Lactobacillus strains.
Binding of AFM1 in different concentrations was examined with unconcentrated, concentrated, heat treated and
concentrated heat-treated LAB cultures. Observed binding of AFM1 by LAB varied between 11 to 100%, being
approximately at the level of 40% throughout the analysis sets. The results of this study suggest that the
aflatoxin binding ability by LAB strain is not strongly strain specific and depends on many external and condition
Probiotics or clay detoxifier can improve the intestinal health of monogastric animals fed diets contaminated
with aflatoxin B1 (AFB1), but little is known in ruminants. This study aimed to investigate the effect of probiotics
and clay detoxifier on the growth performance, enterotoxigenic bacteria, endotoxins and intestinal barrier of
lambs fed diet contaminated with AFB1. Lambs (24) were randomly allocated into 4 groups with 6 replicates.
Treatments included control, AFB1 (100 µg/kg), probiotics (AFB1 + probiotics @ 3×109 cfu/kg) and clay (AFB1 +
clay @ 4.0 g/kg of feed). The trial lasted for 35 d. Results showed that AFB1 worsened body weight gain and feed
conversion ratio, and these were recovered by probiotics and clay detoxifier supplementation. Also, AFB1
increased cecal counts of Clostridium perfringens, Salmonella, Escherichia coli and gram-negative bacteria,
serum endotoxin and diamine oxidase, but decreased duodenal mRNA expressions of claudin-1, IgA inducing
protein, junctional adhesion molecule 2 (JAM-2), joining chain of multimeric IgA and IgM (J-chain) and occludin.
Probiotics ameliorated these negative effects, but for Clostridium perfringens and J-chain, whereas clay
detoxifier only showed beneficial effects on Escherichia coli, gram-negative bacteria, endotoxins, claudin-1 and
JAM-2. In addition, probiotics were more protective against enterotoxigenic bacteria and enterotoxic markers
Kunu is a traditional fermented single or mixed cereals-based beverage popularly consumed in many parts of
West Africa. Presently, the bacterial community and mycotoxin contamination profiles during processing of
various kunu formulations have never been comprehensively studied. This study, therefore, investigated the
bacterial community and multi-mycotoxin dynamics during the processing of three kunu formulations using high-
throughput sequence analysis of partial 16S rRNA gene (hypervariable V3-V4 region) and liquid chromatography
tandem mass spectrometry (LC-MS/MS), respectively. A total of 2,303 operational taxonomic units (OTUs) were
obtained across six processing stages in all three kunu formulations. Principal coordinate analysis biplots of the
Bray-Curtis dissimilarity between bacterial communities revealed the combined influences of formulations and
processing steps. Taxonomically, OTUs spanned 13 phyla and 486 genera. Firmicutes (phylum) dominated
(relative abundance) most of the processing stages, while Proteobacteria dominated the rest of the stages.
Lactobacillus (genus taxa level) dominated most processing stages and the final product (kunu) of two
formulations, whereas Clostridium sensu stricto (cluster 1) dominated kunu of one formulation, constituting a
novel observation. We further identified Acetobacter, Propionibacterium, Gluconacetobacter, and
Gluconobacter previously not associated with kunu processing. Shared phylotypes between all communities
were dominated by lactic acid bacteria including species of Lactobacillus, Lactococcus, Leuconostoc,
Pediococcus, and Weissella. Other shared phylotypes included notable acetic acid bacteria and potential human
enteric pathogens. Ten mycotoxins [3-Nitropropionic acid, aflatoxicol, aflatoxin B1 (AFB1), AFB2, AFM1,
alternariol (AOH), alternariolmethylether (AME), beauvericin (BEAU), citrinin, and moniliformin] were quantified
at varying concentrations in ingredients for kunu processing. Except for AOH, AME, and BEAU that were retained
at minimal levels of &lt; 2 μg/kg in the final product, most mycotoxins in the ingredients were not detectable
after processing. In particular, mycotoxin levels were substantially reduced by fermentation, although simple
The aim of this experiment was to investigate the effect of aflatoxin B (AFB) in the diets of Nile tilapia
(Oreochromis niloticus) and to examine the detoxification activity of a commercial anti-mycotoxin ARCAVIT®
Bioacid Forte. The experimental period lasted 70 days during September - December 2018. Sixty fish were
chosen with an average initial weight of about 40 g then randomly distributed into three treatments in six glass
aquaria (30 × 60 × 40 cm); each treatment was applied in two aquaria (replicates). Dietary AFB was added at a
concentration of 5 ng / g diet. The anti-mycotoxin ARCAVIT® (pro- and prebiotics) is one of the commercial anti-
mycotoxins in the local market. It is the first test for it in aquaculture, which was added at a concentration of 0.5
g/Kg diet. The experimental diets of the three treatments were; the control treatment (T 1 ): the basal diet (BD)
without any additives. The second treatment (T 2 ): BD with an addition of 5 ng AFB / g diet. The third treatment
(T 3 ): BD with an addition of 5 ng AFB / g diet and 0.5g ARCAVIT® / Kg diet. Fish fed the experimental diets at a
daily rate of 3% of their live body weight, at two meals. The obtained results showed that AFB is very toxic, even
at a low concentration as 5 ng / g diet, for Nile tilapia. It threats fish morphology, gross pathology, liver histology,
growth performance, and internal organs indices, feed utilization, body composition, and properties, as well as
hematological and plasma biochemistry parameters. Moreover, the anti-toxic agent (ARCAVIT®) at the tested
Fermentation of food products can be used for the delivery of probiotic bacteria and means of food
detoxification, provided that probiotics are able to grow, and toxins are reduced in raw materials with minimal
effects on consumer acceptability. This study evaluated probiotic enrichment and detoxification of kwete, a
commonly consumed traditional fermented cereal beverage in Uganda, by the use of starter culture with the
probiotic Lactobacillus rhamnosus yoba 2012 and Streptococcus thermophilus C106. Probiotic kwete was
produced by fermenting a suspension of ground maize grain at 30 ◦ C for a period of 24 h, leading to a decrease
of the pH value to ≤ 4.0 and increase in titratable acidity of at least 0.2% (w/v). Probiotic kwete was acceptable
to the consumers with a score of ≥6 on a 9-point hedonic scale. The products were stable over a month’s study
period with a mean pH of 3.9, titratable acidity of 0.6% (w/v), and Lactobacillus rhamnosus counts &gt;10 8 cfu g
−1 . HPLC analysis of aflatoxins of the water-soluble fraction of kwete indicated that fermentation led to an over
1000-fold reduction of aflatoxins B 1 , B 2 , G 1 , and G 2 spiked in the raw ingredients. In vitro fluorescence
spectroscopy confirmed binding of aflatoxin B 1 to Lactobacillus rhamnosus with an efficiency of 83.5%. This
Aflatoxin B 1 (AFB 1 ), a mycotoxin found in food and feed, is immunotoxic to animals and poses significant
threat to the food industry and animal production. The primary target of AFB 1 is the liver. To overcome
aflatoxin toxicity, probiotic-mediated detoxification has been proposed. In the present study, to investigate the
protective effects and molecular mechanisms of Lactobacillus bulgaricus or Lactobacillus rhamnosus against liver
inflammatory responses to AFB 1 , mice were administered with AFB 1 (300 µg/kg) and/or Lactobacillus
intragastrically for 8 weeks. AML12 cells were cultured and treated with AFB 1 , BAY 11-7082 (an NF-κB
inhibitor), and different concentrations of L. bulgaricus or L. rhamnosus. The body weight, liver index,
histopathological changes, biochemical indices, cytokines, cytotoxicity, and activation of the NF-κB signaling
pathway were measured. AFB 1 exposure caused changes in liver histopathology and biochemical functions,
altered inflammatory response, and activated the NF-κB pathway. Supplementation of L. bulgaricus or L.
rhamnosus significantly prevented AFB 1 -induced liver injury and alleviated histopathological changes and
inflammatory response by decreasing NF-κB p65 expression. The results of in vitro experiments revealed that L.
rhamnosus evidently protected against AFB 1 -induced inflammatory response and decreased NF-κB p65
expression when compared with L. bulgaricus. These findings indicated that AFB 1 exposure can cause
This study was focused on measuring the percent value of degraded aflatoxin M1 (AFM1) in the industrial and
traditional fermented milk product (Doogh in Persian) using Lactobacillus acidophilus and Lactobacillus
plantarum, and a combination of them at concentrations of 104 cfu/ml associated with the selected bio-physical
factors including Temperature of produced Doogh at 4, 21, and 37 °C and Time of Doogh storage (0, 48 hr, 11
days, and 30 days). These results showed that the level of AFM1 binding to L. acidophilus at concentration of 1 ×
104 cfu/ml increased in a temperature-dependent manner from the second day of the research for traditional
produced Doogh (100.0 ± 0.37) compared with those of other days of the experiment. These findings were
repeated for industrially produced Doogh with slight difference at 30 days of experiment (94.28 ± 0.47) with
significant differences (p &lt;.05) compared with those of the second and the 11th days (100.0 ± 0.37 and 100.0
± 0.45, respectively). It is concluded that the use of L. acidophilus and L. plantarum can be useful for binding the
AFM1 presented in fermented milk especially at long storage time and higher degrees of temperature (21 and 37
°C). Practical applications: One of the most reasons for application the probiotics in to milk and dairy products
such as fermented milk (Doogh) has been reducing the amount of aflatoxin M1 (AFM1). In this study, the findings
showed that lactic acid bacteria bacteria including Lactobacillus acidophilus and Lactobacillus plantarum have
Aflatoxins gain entry into food products when proper drying, storage, and transport conditions are not applied.
They comprise of closely related compounds for example, aflatoxins B1, B2, G1, G2, M1, and M2. The order of
toxicity among aflatoxis is B1 > G1 > B2 > G2. In developing countries, the serious illness and deaths are common
due to acute aflatoxicosis and human children are more prone to their attack. Chronic aflatoxicosis affects on
animal nutrition status. Aflatoxins are commonly extracted from foodstuffs by liquid–liquid extraction,
supercritical fluid extraction, and solid phase extraction. They are analyzed by thin layer chromatography, high-
performance liquid chromatography (HPLC), HPLC-mass spectrometry and UV-Visible spectroscopy. They
demonstrate high stability within food or feed stuffs and can be removed by physical, chemical, and biological
means. Physical approaches use heat, sunlight, UV light, gamma rays, or adsorption phenomenon while chemical
methods involve various fungicides, herbal plant extracts, oxidizing/hydrolytic/chlorinating agents, or clay.
Biological approaches involve either probiotic and yeast mixtures or atoxigenic fungi. A proper detoxification
procedure ensures the nutritional value of food/feed stuffs and does not generate new carcinogenic, mutagenic,
or toxic substances. Consideration of hygienic precautions and technical assistance, implementation of
regulatory initiatives, and stricter quality control measures are all important for their control. Practical
applications: Aflatoxins are toxic metablotics produced primarily by means of fungal species. Cereals, spices,
nuts, grapes, apples, dried fruit, dried vegetables (peas, beans), oil seeds, teas, cocoa, and coffees are commonly
infected aflatoxins. Their presence in an animal diet severely affects the kidneys, liver, rate of growth and
reproduction, serious illness, and even death. The aflatoxin problem is more common in developing countries.
One of the most studied mycotoxins is Aflatoxin M1 (AFM1),which as a carcinogen should be eliminated or
limited from food products especially dairy products. Among different detoxification methods of AFM1, several
strains of lactic acid bacteria (LABs) (particularly Lactobacillus acidophilus and Bifidobacterium spp.) can bind
Aflatoxin M1 and furthermore, eliminate them. The principal mechanism of detoxification by LABs is the physical
binding to peptidoglycan and polysaccharides present in the bacterial cell wall. This paper reviews the
detoxification capability of some LABs strains and the critical roles of factors affecting the binding procedures
including LAB strains, optimum incubation time, and the cell concentration of LABs. Practical application: This
study reviewed the impact of lactic acid bacteria on detoxification of aflatoxin M1. As the high toxicity effect of
aflatoxin M1 in food is proved, finding a safe method in order to remove them is crucial. Lactic acid bacterias
(LABs) are vastly applied in food science and technology due to their various beneficial effects, being easy
This study investigated the ability of Bifidobacterium animalis lactis and Lactobacillus delbrueckii bulgaricus at
concentrations of 108 and 109 cfu/ml to detoxify skim milk that was contaminated with 0.25 ng/ml, 0.5 mg/ml,
and 0.75 mg/ml aflatoxin M1 (AFM1) at 30 min, 60 min, 120 min, and 24 hr, and at 4 and 37 °C. The maximum
level of AFM1 binding to 1 × 108 cfu/ml of L. bulgaricus was 57.5 ± 5.2% at 37 °C. Both strains showed the
degrading potential to reduce AFM1 concentrations and, thus, could be used as starter cultures to detoxify milk.
Furthermore, the temperature, bacteria concentration, and vitality have more significant influence on AFM1
binding in contaminated milk than other indices. The results indicated that Bifidobacterium lactis and
Lactobacillus bulgaricus could be used as beneficial probiotics to decrease the destructive properties of AFM1 to
consumers exposed to AFM1-contaminated milk. Practical applications: The upcoming demands are to embrace
probiotic strains that have documented beneficial health effects for the consumer. Bifidobacterium animalis
lactis and Lactobacillus delbrueckii bulgaricus can be used as probiotic to bind selected dietary contaminants
This study aimed to investigate the effect of lactic acid bacteria (LAB) and smectite on the growth performance,
nutrient digestibility and blood parameters of broilers that were fed diets contaminated with aflatoxin B1
(AFB1). A total of 480 newly hatched male Arbor Acres broilers were randomly allocated into four groups with six
replicates of 20 chicks each. The broilers were fed diets with the AFB1 (40 μg/kg) challenge or without (control)
it and supplemented with smectite (3.0 g/kg) or LAB (4.0 × 1010 CFU/kg) based on the AFB1 diet. The trial lasted
for 42 days. The results showed that during days 1–42 of AFB1 challenge, the feed intake (FI) and body weight
gain (BWG) were depressed (p &lt;.05). The inclusion of LAB and smectite increased (p &lt;.05) the BWG by 71.58
and 41.89 g/bird, respectively, which reached the level of the control diet (p ≥.05), but there were no differences
(p ≥.05) in performance between LAB and smectite. LAB and smectite also increased (p &lt;.05) the apparent
total tract digestibility of the crude protein. Regarding the blood parameters, AFB1 decreased (p &lt;.05) the
levels of red blood cell count, haematocrit, mean corpuscular volume, haemoglobin, albumin and total protein.
In the meantime, the AFB1 increased (p &lt;.05) leucocyte counts, urea nitrogen, cholesterol, total bilirubin,
creatinine, glutamic-pyruvic transaminase, glutamic oxaloacetic transaminase and alkaline phosphatase. By
contrast, LAB and smectite affected (p &lt;.05) these parameters in the opposite direction. It can be concluded
This study focused on monitoring changes in aflatoxin M1 (AFM1) concentrations during production and storage
of different fermented milks using selected probiotic and nonprobiotic combined cultures. Milk samples
intended for fermentation were intentionally contaminated by adding a standard of AFM1. All of the tested
cultures caused remarkable reductions in AFM1 concentrations during the fermentation process. Probiotic
cultures were more effective than nonprobiotic cultures, with Lactobacillus caseiLC-01 strain being the most
efficient, achieving a reduction level of approximately 58%. Among the nonprobiotic cultures, yoghurt culture YC-
The use of probiotic as dietary approach to prevent exposure to food contaminant, aflatoxin B1 (AFB1) has
greatly increased. Several studies found that AFB1 binding to the bacterial cell wall is strain-specific. Moreover,
the interaction between AFB1 and bacterial cell wall is not well-understood, thus warrants further investigation.
This research was conducted to assess the ability of Lactobacillus casei Shirota (Lcs) to bind AFB1 at different
concentrations and to determine AFB1 binding efficiency of different Lcs cell components including live cell, heat-
treated, and cell wall. In addition, the interaction between AFB1 and Lcs was also evaluated via scanning electron
microscopy (SEM) and through an animal study. The binding of AFB1 by all Lcs cell components depends on the
concentration of available AFB1. Among all Lcs cell components, the live Lcs cells exhibited the highest binding
efficiency (98%) toward AFB1. Besides, the SEM micrographs showed that AFB1 induced structural changes on
the bacterial cell surface and morphology including rough and irregular surface along with a curve rod-shaped. In
vivo experiment revealed that Lcs is capable to neutralize the toxicity of AFB1 on body weight and intestine
through the binding process. The animal's growth was stunted due to AFB1 exposure, however, such effect was
significantly (p < 0.05) alleviated by Lcs. This phenomenon can be explained by a significant (p < 0.05) decreased
level of blood serum AFB1 by Lcs (49.6 ± 8.05 ng/mL) compared to AFB1-exposed rats without treatment (88.12
Aflatoxins are a large group of highly toxic, mutagenic, and carcinogenic mycotoxins produced by specific species
of fungi. Potential contamination of food commodities by these compounds causes extensive damage that lead
to great economic losses. This study explored the potential use of antifungal compounds, produced by
Lactobacillus brevis and Lactobacillus paracasei, for growth inhibition and subsequent aflatoxin B1 production
from select strains of Aspergillus flavus and Aspergillus parasiticus. Lactobacilli strains were isolated from
traditional Egyptian dairy products, whereas fungal strains were isolated from infected cereal seeds. There were
noticeable decreases in mycelium biomass and aflatoxin production as well. L. brevis exhibited the highest
reduction of aflatoxin B1 production by A. flavus and A. parasiticus, 96.31 and 90.43%, respectively. The
concentrations of amino acids of the antifungal compound produced by L. brevis were significantly higher than
that produced by L. paracasei. Asparagine, glutamine, glycine, alanine, and leucine were the most concentrated
amino acids for both strains. The antifungal compounds produced by L. brevis and L. paracasei were active in a
wide range of pH, heat stable and inactivated by proteolytic enzymes (protease K and trypsin A). The expression
of Omt-A gene that involved in the later step of aflatoxin production was evaluated by real-time PCR. There was
a vigorous reduction at transcriptional level of Omt-A gene observed in A. flavus that is treated by L. brevis and L.
paracasei (80 and 70%, respectively). However, the reduction of Omt-A gene observed in A. parasiticus that is
treated by L. brevis and L. paracasei was 64.5 and 52%, respectively. Treating maize seeds with antifungal
Aflatoxins (AFs) are produced mainly by the molds Aspergillus flavus and Aspergillus parasiticus. Aflatoxin B1
(AFB1) is classified as carcinogenic to humans. The aim of this study was to evaluate the capacity of different
strains of Lactobacilli (Lb.) and Bifidobacteria (Bf.) to reduce the bioaccessibility of AFB1 and aflatoxin B2 (AFB2),
spiked in loaf bread, using a dynamic in vitro simulated gastrointestinal digestion system. Aliquots of 20 mL of
gastric and duodenal fluids were sampled for the determination of the mycotoxins gastric and duodenal
bioaccessibility respectively, by liquid-chromatography coupled to the mass spectrometry in tandem
(LC–MS/MS). A reduction of AFs bioaccessibility compared to the control (digestion without bacterial strains)
was evidenced. The strains that evidenced the highest gastric and duodenal bioaccessibility reductions of AFB1
and AFB2 were Lb. johnsoni CECT 289, Lb. reuteri CECT 725, Lb. plantarum CECT 220 and Lb. casei CECT 4180,
with values ranging from 76.38 to 98.34% for AFB1 and from 77.14 to 98.66% for AFB2. These results suggest
Contamination of animal feed with mycotoxins still occurs very often, despite great efforts in preventing it.
Animal feeds are contaminated, at low levels, with several mycotoxins, particularly with those produced by
Aspergillus and Fusarium genera (Aflatoxin B1 Ochratoxin A, Zearalenone, Deoxynivalenol and Fumonisina B1). In
animal feed, to date, only Aflatoxin B1 is limited through EU regulation. Consequently, mycotoxins cause serious
disorders and diseases in farm animals. In 2009, the European Union (386/2009/EC) approved the use of
mycotoxin-detoxifying agents, as feed additives, to prevent mycotoxicoses in farm animals. The present review
gives an overview of the problem of multi-mycotoxin contamination of feed, and aims to classify mycotoxin
adsorbing agents (minerals, organic, and synthetic) for feed decontamination, focusing on adsorbents with the
In this work, we explored the potential of 25 Lactobacillus plantarum strains isolated from cereals and milk-
based products, testing characteristics related to antifungal activity and to nutritional quality. The tested strains
demonstrated interesting beneficial traits, such as the ability to utilize fructo-oligosaccharides, prebiotic
substances that help probiotic microorganisms to grow in the human gut, and to reduce phytate, an antinutrient
present in cereal sector. Regarding mould inhibition, we highlighted the ability of the strains to inhibit Penicillium
roqueforti, Mucor circinelloides and mycotoxinogenic moulds associated with cereal grains as Aspergillus flavus,
A. niger, Fusarium verticillioides. Moreover, a moderate reduction of the bioavailability of aflatoxin AFB1 was
detected. The selected L. plantarum strain ITEM 17215, showed a strong inhibitory ability towards fungal growth
and was able to produce 1,2-dihydroxybenzene, benzoic acid, p-hydroxyphenyllactic acid and 3-phenyllactic acid.
The latter compound, already described as efficient antifungal inhibitor, was the most abundant and its
concentration was further increased by adding phenylalanine and phenylpyruvic acid in the growth medium. The
metabolites produced by strain ITEM 17215 could also be related to the ability of the strain to induce cereal
The aim of this study was to evaluate probiotic properties and the aflatoxin B1 adsorption ability of yeasts
isolated from rainbow trout intestine and fish feed to assess their use in the formulation of feed additives.
Growth at pH 2, bacterial pathogens inhibition, bacterial pathogens co-aggregation, autoaggregation,
homologous and heterologous inhibition against lactic acid bacteria were evaluated. Moreover, aflatoxin B1
adsorption was tested. All strains were able to maintain viable (107 cells/ml) at pH 2. All strains isolated from
intestine were identified as Kazaschtania exigua, while strains isolated from feed were all identified as
Debaryomyces hansenii. Kazaschtania exigua RC035 and RC037 showed the strongest antimicrobial activity while
K. exigua RC037 and RC038 were the most efficient co-aggregating bacterial pathogens. All strains exhibited
strong autoaggregation. None of the tested yeast strains showed homologous inhibition towards other yeasts
and heterologous inhibition towards lactic acid bacteria strains. Debaryomyces hansenii RC031 demonstrated
aflatoxin B1 adsorption capacity (21%). The results of the present study indicate that select strains of
Aflatoxin contamination is very common in feedstuffs across the world and finding an ideal detoxifier is urgent
because of the toxic action on animals and negative effects on foods, humans, and the environment. To
thoroughly eliminate the toxin, a detoxification method has changed from physical to biological. The objective of
the present study was to investigate the effect of lactic acid bacteria (LAB) and hydrated sodium calcium
aluminosilicate (HSCAS) on detoxification of aflatoxin B1 (AFB1) by assessing growth performance, digestibility,
immune function, and AFB1 residues in tissues and excreta of broiler chickens from d 0 to 21. A total of 480
female broiler chicks on d 0 were randomly allotted to 4 treatments with 6 cages of 20 chicks each for diets:
positive control (PC, undetectable AFB1), negative control (NC, PC + 40 µg AFB1/kg), LAB (NC + 1.5 × 1010 cfu
LAB/kg), and HSCAS (NC + 3.0 g HSCAS/kg). Results showed that the NC treatment reduced (P &lt; 0.05) average
daily gain and feed efficiency, and LAB or HSCAS supplementation improved (P &lt; 0.05) the growth
performance of broiler chickens, and the effect of LAB was greater than HSCAS. The LAB and HSCAS increased (P
&lt; 0.05) the digestibility of dry matter, crude protein, crude fat, and digestible energy by 4.0–15.0%, and
improved (P &lt; 0.05) immune function by modulating the relative weights of immune organs, lymphocyte
BACKGROUND: Aflatoxins (AFs) are a group of toxic, nephrotoxic, hepatotoxic and carcinogenic fungal
metabolites. Heat- and acid-treated yeasts, probiotic bacteria and their combination were used to remove AFB1,
AFB2, AFG1 and AFG2 from human and animal food. RESULTS: The in vitro study revealed that the highest
removal percentage of AFs in phosphate-buffered saline was recorded after 72 h with the yeast–probiotic
coctile, reaching 95.59%. Therefore, this coctile was added to Cerelac contaminated with AFB1, AFB2, AFG1 and
AFG2, and the removal percentages were 8.17%, 36.12%, 44.75%, 64.72% and 93.21% after 6, 12, 24, 48 and 72
h of treatment, respectively. Cerelac yeast–probiotic coctile was administered to female rats and the results
showed that all AFs (AFB1, AFB2, AFG1 and AFG2) were detected in the serum of mother rats for both AF groups
III and IV. On the other hand, AFM1 and AFM2 metabolites were not observed in mothers' sera but were
detected in all infants of groups III and IV. Meanwhile, AFB1, AFB2, AFG1 and AFG2 were not observed in infants'
Aflatoxin B1 (AFB1) contamination of animal feed and food including meat and dairy products, even at very low,
ppb, levels can adversely affect human and animal health. This study assessed the effect of yogurt whey addition
at 5% to the drinking water of broiler chickens fed AFB1-contaminated diets on the resulting AFB1 residues in
organs (liver, kidney), and meat (breast, leg). Groups that received whey in their drinking water showed
reductions in AFB1 of 55.46% (1.19 to 0.43 ng/g), 37.68% (0.69 to 0.53 ng/g), and 59.35% (28.81 to 11.71 ng/g)
in leg and breast, respectively, at the end of week 6. The concentration of AFB1 in liver, kidney, and gizzard was
reduced by 60.68 % (1.88 to 0.74 ng/g), 60.64% (2.06 to 0.81 ng/g), and 52.73% (1.10 to 0.52 ng/g), respectively,
at week 6 compared to those without whey addition. It is suggested that aflatoxin B1 was biodegraded by the
lactic acid bacteria (LAB) in yogurt whey, which included Lactobacillus bulgaricus, Streptococcus thermophilus
The proceedings contain 280 papers. The special focus in this conference is on Scientific Geoconference. The
topics include: Shungite nanopowder additive influence on tensile strength of one-ply particle boards; TiO2
assisted photocatalytic degradation of bisphenol in aqueous media, under UV-VIS irradiation; validation of low-
cost sensor measurements in urban territory for air pollution monitoring. Riga city example; colloidal silica
granules of chicken manure biochar increase plant biomass and microbial count; analysis of the garlic landraces
in the west of Romania using the flowcytometry method; antioxidant capacity of some marine green macroalgae
species fluid extracts; application of neural network model for predicting the antibacterial activity of alginate-
chitosan sponges; biotechnological applications of soil microbial activities in ecological agriculture; chemical
composition and antioxidant properties of dill essential oil; comparative methods for skin and hide waste
capitalisation; comparative morphology, anatomy and biochemistry in prunus spinosa L. Fruits; cost, nutritional
and energy value optimized coarse-grained restructured fish mince development; diffractometric method for
determining the crystallinity degree of cellulose; effect of almond flour on nutritional, sensory and bakery
characteristics of gluten-free muffins; effect of some probiotic bacteria on the reduction of aflatoxin B1
production in stored arabica coffee beans; efficacy of biochar with immobilized microbes for soil fertilization;
estimation of efficiency of use of vegetable oils for re-ception of biofuel by a method of enzymatic catalysis;
Aflatoxin B1 (AFB1) contamination of coffee beans continues to be a serious issue regarding livestock
productivity and human health. AFB1 (secondary metabolites produced by a larger number of Aspergillus spp.) is
classified as a Group I carcinogen by the International Agency for Research on Cancer due to the considerable
risk to human and animal health. Therefore, monitoring of this toxin by using probiotics during storage became a
challenge due to the demand for reduced use of chemical preservatives by consumers. The FAO/WHO defines
probiotics as “live microorganisms which when administered in adequate amounts confer a health benefit on the
host.” They usually belong to the genus Bifidobacterium and Lactobacillus. Principal characteristic of the
probiotics is the antagonistic activity against food pathogen agents. This study evaluated the antagonistic effect
of Bifidobacterium BB-12® and Lactobacillus acidophilus DSM 20242 against spore production by Aspergillus
flavus DSM1959 as well as aflatoxin inhibiting ability in Arabica green coffee beans during two month storage.
Green coffee beans (Coffea arabica L.) samples were collected from a local commercial store. Probiotic
suspensions alone or in combinations were tested by sprinkling on the beans followed by aerobically incubation
at 30°C for 11 days. Quantification of the AFB1 production was determined by an immunoenzymatic method
(ELISA). Both live and inactivated forms significantly reduced A. flavus sporulation, but the best results were
obtained with live cells. A decrease of the AFB1 production was observed for Bifidobacterium (8.24%) and
In order to minimise human exposure to aflatoxin M1 (AFM1) the levels of this highly carcinogenic mycotoxin in
milk, heat-treated milk, and other dairy products have been limited to &lt;0.05 μg kg-1. However, its removal
from dairy products presents a challenge for dairy producers, as commercial additives change organoleptic
properties, and filtration alone yields poor results. The aim of this study was to find a strain of lactic acid bacteria
(LAB) from milk or dairy products that most effectively binds AFM1 and to see whether heat treatment of the
selected LAB affects the binding efficiency. We also wanted to investigate whether centrifugation can improve
filtering of the obtained AFM1-LAB complexes from milk. To do that, we isolated and identified 10 native LAB
species/strains, incubated their viable or heat-treated cells (108 CFU mL-1) in milk spiked with 0.5 μg L-1 of
AFM1 at 4 °C for 0, 2, 4, and 24 h, and quantified the amount of unbound AFM1 with HPLC. AFM1 binding
efficiency ranged from 21 to 92 % for viable cells and from 26 to 94 % for the heattreated ones. Since both viable
and heat-treated Lactobacillus plantarum KM showed the best results, we used it for the next step in AFM1
To neutralize mycotoxins entering the digestive tract with feed, probiotics and antioxidants are increasingly
used. They are introduced into compound feeds to restore the normal microflora in the gut, activate feed
fermentolysis, strengthen the immune system and increase hens’ egg productivity. The research aims to study
the effect of probiotic Bifidum SHG and antioxidant Santochinum on egg productivity, physico-chemical and
incubation properties of eggs of layers whose feed has a tolerant level of aflatoxin B1. The objects of research
were layers of cross “Smena-7”. The results were statistically processed by Student’s t-test using mathematical
analysis software package “Microsoft Excel”. Feeding probiotic and preparation Santochinum as a part of mixed
feed with the tolerant level of aflatoxin B1 contributed per average layer in the third test group to increase egg
production by 13.2%. The highest average eggs weight had hens of the third test group, which in this indicator
exceeded the control counterparts by 2.67%. Combined application of these preparations had the best effect on
biochemical composition of eggs of birds from the compared groups. For layers of the third test group it was as
superiority in the presence in the egg yolk of dry matter by 1.48%, protein – by 2.00%, vitamin A – by 18.2% and
vitamin E – by 12.0%, as well as in the concentration of dry matter in egg white by 1.68% and protein – by 1.25%.
Hens of the third test group outperformed the control counterparts in such morphological indicators of eggs, as
Aflatoxin B1 (AFB1) and zearalenone (ZEA) are the secondary toxic metabolites of fungi which contaminate a
wide range of food and feedstuffs. Limiting exposure of humans and livestock to them is very essential. Among
numerous methods of mycotoxin-degradation, biodegradation by microorganisms and enzymes is an effective
and promising approach to eliminate their hazards. The present study aims to optimize the proportion of
different species of beneficial microbes by means of response surface methodology (RSM) and its combination
with mycotoxin-degradation enzymes. The results indicated that AFB1 and ZEA degradation rates were 38.38%
and 42.18% by individual Bacillus subtilis (P &lt; 0.05); however, AFB1 and ZEA degradation rates reached 45.49%
and 44.90% (P &lt; 0.05) when three probiotic species such as Bacillus subtilis, Lactobacillus casein and Candida
utilis were at a ratio of 1:1:1, corresponding with the predictive value of the RSM model. The further experiment
showed that AFB1 and ZEA degradation rates were 63.95% and 73.51% (P &lt; 0.05) when the compound of
three probiotic species was combined with mycotoxin-degradation enzymes from Aspergillus oryzae at a ratio of
Probiotic Lactobacillus casei Shirota (LcS) is a potential decontaminating agent of aflatoxin B1 (AFB1). However,
few studies have investigated the influence of diet, especially a high protein (HP) diet, on the binding of AFB1 by
probiotics. This research was conducted to determine the effect of HP diet on the ability of LcS to bind AFB1 and
reduce aflatoxin M1 (AFM1) in AFB1-induced rats. Sprague Dawley rats were randomly divided into three
groups: A (HP only), B (HP + 108 CFU LcS + 25 μg AFB1/kg BW), and C (HP + 25 μg AFB1/kg BW). Levels of AST
and ALP were higher in all groups but other liver function's biomarkers were in the normal range, and the liver's
histology showed no structural changes. The urea level of rats in group B (10.02 ± 0.73 mmol/l) was significantly
lower (p&lt;0.05) than that of rats in group A (10.82 ± 0.26 mmol/l). The presence of carcinoma in the small
intestine and colon was more obvious in group C than in group B. Moreover, rats in group B had significantly
(p&lt;0.05) lower AFM1 concentration (0.39 ± 0.01 ng/ml) than rats in group C (5.22 ± 0.28 ng/ml). Through
Background and Objective: The prevalence of liver cancer in Egypt is particularly worrisome. Aflatoxin B1 (AFB1),
ahepatocarcinogenic mycotoxinjs one of the main contributors to the high rate of hepatocellular carcinoma
(HCC). This study aimed to determine the ability of four lactic acid bacteria (LAB) species in removing or binding
aflatoxin B1 (AFB1) from broth media and whole milk and to evaluate the stability of LAB/AFB1 complex during
cold storage at 4°C for 1 week. Materials and Methods: Whole milk and MRS broth media were spiked with 50
ngmL-1 AFB1 and incubated at 37°C for 24 h with four LAB strains followed by storage at 4°C for 24,72 and 196
h.AFBl removal was determined using HPLC. Results: The efficiency of LAB strains in removing AFB1 from both
media was affected by the type of used strain and media. Whole milk was a favorable media for the tested LAB
strain in AFB1 reduction when compared with MRS broth. Lactobacillus acidophilus (L acidophilus) achieved 80%
reduction in milk within 24 h at 37°C, whereas Lactobacillusplantarum (L. plantarum) favored cold storage to
reduce 85% of the AFB1 content after 1 week. The count of LAB cells in whole milk medium raised in MRS broth
The present study was conducted to isolate and identify Lactobacillus rhamnosus from locally fermented dairy
products collected from different markets in Irbid city, Jordan. Thereafter, the ability of Lb. rhamnosus to
detoxify aflatoxins (AFs) was investigated in vitro after incubation on 37°C in MRS medium and in artificial
intestine fluid (AIF). Three Lb. rhamnosus out of nine different species of Lactic Acid Bacteria (LAB) isolated from
15 fermented dairy products samples were identified. The isolates were characterized based on their
morphological, microscopic, cultural and biochemical properties. The selection of isolates as probiotics
depended on their abilities to grow in pH levels between 2 to 6 and their tolerance to grow at 1.0 % bile salts
concentrations, Furthermore, Lb. rhamnosus was able to adhere to mucus onto the intestine surface at 54.7%.
The ability of Lb. rhamnosus of AFs detoxification has significantly (p < 0.05) increased with the increase in
Several strains of lactic acid bacteria (LAB), frequently used in food fermentation and preservation, have been
reported to bind different types of toxins in liquid media. This study was carried out to investigate the effect of
different concentrations of Lactobacillus rhamnosus GG (ATCC 53103) to bind aflatoxin M1 (AFM1) in liquid
media. AFM1 binding was tested following repetitive washes or filtration procedures in combination with
additional treatments such as heating, pipetting, and centrifugation. The mixture of L. rhamnosus GG and AFM1
was incubated for 18 h at 37 °C and the binding efficiency was determined by quantifying the unbound AFM1
using HPLC. The stability of the complexes viable bacteria-AFM1 and heat treated bacteria-AFM1 was tested.
Depending on the bacterial concentration and procedure used, the percentages of bound AFM1 by L. rhamnosus
GG varied from as low as undetectable to as high as 63%. The highest reduction in the level of unbound AFM1
was recorded for the five washes procedure that involved heating and pipetting. Results also showed that
binding was partially reversible and AFM1 was released after repeated washes. These findings highlight the
The aim of this work was to evaluate the incorporation of a freeze-dried probiotic strain (Lactobacillus plantarum
CIDCA 83114) into zeolites. The bacteria-zeolite mixture was added to poultry feed together with thyme, and the
obtained product was stored for 60 days at 25 °C and 60–70% relative humidity. The ability of the obtained
product to remove aflatoxin B1 (AFB1) was studied. The highest bacterial viability was observed when 50% w/w
bacteria were added to zeolites. The bacterial:zeolite mixtures were then incorporated into poultry feed
containing or not thyme. Initial counts of L. plantarum were in the range of 1–2 × 108 CFU/g for all samples. In all
cases, bacterial viability decreased one logarithmic order after 20 days of storage, and three logarithmic orders
after 60 days. No significant viability loss was observed after exposure of the poultry feed to gastro-intestinal
conditions. Freeze-dried L. plantarum and zeolite were able to remove AFB1, with an average reduction of 20
and 80%, respectively. Moreover, the freeze-dried bacteria-zeolite mixture was capable to remove up to 90%
Our previous studies have selected lactic acid bacteria (LAB) with antifungal activities from traditional fermented
foods made from cassava (G7) and silage feed palm leaf (PDS5 and PDS3). In this study we evaluated their ability
to bind aflatoxin B1 (AFB1) and probiotic characteristic. The probiotic characteristic assays of LAB consisted of
resistance to acidic conditions (pH 3), gastric juice and bile salts 0.3%. We also carried out an in vitro evaluation
of LAB aflatoxin binding ability in viable and non-viable cell for 24 and 48 hours of incubation. The measurement
of aflatoxin content was performed by ELISA method using AgraQuant Total Aflatoxin Assay kit. The results
showed that all isolates were potential as probiotics and the G7 isolate had the highest viability among other
isolates in pH 3 (92.61 %) and the bile salts assay (97.71 %). The percentage of aflatoxin reduction between
viable and non-viable cell from each LAB isolate were different. The highest aflatoxin reduction in viable cell
assay was performed by G7 isolate (69.11 %) whereas in non-viable cell assay was performed by PDS3 isolate
(73.75 %) during incubation time 48 hours. In this study, G7 isolate performed the best probiotic characteristics
with the highest viability in acid pH assay, bile salt 0.3% assay and percentage of aflatoxin B1 reduction in viable
cell condition. Molecular identification using 16S rRNA sequence analysis showed that G7 isolate had homology
Probiotics are being used in biological control of bacterial pathogens, as an alternative to antibiotics, to improve
health and production parameters in fish farming. Fish farming production is severely affected by aflatoxins
(AFs), which are a significant problem in aquaculture systems. Aflatoxins exert substantial impact on production,
causing disease with high mortality and a gradual decline of reared fish stock quality. Some aspects of
aflatoxicosis in fish, particularly its effects on the gastrointestinal tract, have not been well documented. The aim
of the present study was to evaluate probiotic properties of lactic acid bacterial (LAB) strains isolated from
rainbow trout intestine and feed. Moreover, AFB1-binding and/or degrading abilities were also evaluated to
assess their use in the formulation of feed additives. Growth at pH 2, the ability to co-aggregate with bacterial
pathogens, inhibition of bacterial pathogens, and determination of the inhibitory mechanism were tested.
Aflatoxin B1 (AFB1) adsorption and degradation ability were also tested. All strains were able to maintain viable
(107 cells ml–1) at pH 2. Pediococcus acidilactici RC001 and RC008 showed the strongest antimicrobial activity,
inhibiting all the pathogens tested. The strains produced antimicrobial compounds of different nature, being
affected by different treatments (catalase, NaOH and heating), which indicated that they could be H2O2, organic
acids or proteins. All LAB strains tested showed the ability to coaggregate pathogenic bacteria, showing
inhibition percentages above 40%. Pediococcus acidilactici RC003 was the one with the highest adsorption
capacity and all LAB strains were able to degrade AFB1 with percentages higher than 15%, showing significant
Aflatoxins could be detoxify by microbes to reduce their toxic effect in contaminated food. Cerelac is the
common baby food in Egypt. This study was conducted to evaluate the efficacy of both probiotics bacteria and
yeasts on aflatoxins B1 and B2 dtoxification. Bacteria (heat acid inactivated); S. thermophilus TH-4, B. bifidum
and yeasts; k. lactis, S. cerevisiae were used to detoxify these aflatoxins. In PBS each bacterial strain, reduced
AFB1 and AFB2 by 28-29% and great reduction of 55% was achieved using both bacterial strains in combination.
Similarly, yeast strains were able to reduce AFB1 and AFB2; 54% with S. cerevisiae and 42% with K. lactis, with
higher reduction of 66.6% using both yeast strains in combination. Using the probiotic combination with Cerelac
exhibited higher level of B1 and B2 reduction by 94.1%, 94.5%, respectively. Scanning Eectron Microscopy
confirmed the ability of these probiotics to adsorb the toxins particles. Practical applications: Probiotic and yeast
mixtures could be the best materials used to reduce and/or eliminate mycotoxins especially aflatoxins from
Lactic acid bacteria (LAB) and the glutathione (GSH) pathway are protective against aflatoxin, but information on
the effect of LAB on aflatoxin metabolism and GSH activity in farm animals is scarce. This study aimed to
investigate the effects of LAB and aflatoxin B1 (AFB1) on growth performance, aflatoxin metabolism, and GSH
pathway activity using 480 male Arbor Acres broiler chickens from d 1 to 35 of age. Diets were arranged in a 2 ×
2 factorial design, including AFB1 at 0 or 40 μg/kg of feed and LAB at 0 or 3 × 1010 cfu/kg of feed, and the LAB
was a mixture of equal amounts of Lactobacillus acidophilus, Lactobacillus plantarum, and Enterococcus faecium.
The results showed that there were highly significant (P &lt; 0.01) effects of AFB1 toxicity, LAB protection, and
their interaction on ADFI, ADG, and G:F of broilers during d 1 to 35. Compared with the AFB1 diet, the LAB diet
reduced (P &lt; 0.05) the residues of AFB1 in the liver, kidney, serum, ileal digesta, and excreta on d 14 by 121.5,
80.6, 43.7, 47.0, and 26.5%, respectively, and on d 35 by 40.6, 60.2, 131.7, 37.9, and 32.9%, respectively,
whereas the LAB diet increased (P &lt; 0.05) the contents of aflatoxin M1, a metabolite of AFB1, in the liver,
kidney, serum, and ileal digesta on d 14 by 98.2, 154.2, 168.6, 19.1, and 34.1%, respectively, and in the kidney
and serum on d 35 by 32.6 and 142.2%, respectively. For the activity of the GSH pathway in the liver and
duodenal mucosa, there were significant (P ≤ 0.01) effects of LAB and AFB1 on reduced GSH, glutathione S-
transferases (GST), and glutathione reductase (GR) on d 14 and 35; compared with the control diet, the LAB diet
increased (P &lt; 0.05) GSH, GST, and GR by a range of 11.6 to 86.1%, and compared with the AFB1 diet, the LAB
diet increased (P &lt; 0.05) GSH, GST, and GR by a range of 24.1 to 146.9%. In the liver, there were interactions
The objectives were to examine the aflatoxin B1 (AFB1)-binding capacity of silage bacteria and factors affecting
the responses. Experiments 1 and 2 examined the effects of bacterial strain and population on the AFB1-binding
capacity of 10 bacteria. When applied at 106 cfu/mL to an in vitro medium, only Lactobacillus plantarum PT5B
bound the AFB1 and the binding capacity was low (4%). When applied at 109 cfu/mL, all 10 bacteria bound AFB1,
but L. plantarum R2014 (Lp) and EQ12, Lactobacillus buchneri R1102 (Lb), and Pediococcus acidilactici R2142 and
EQ01 (Pa) had the greatest capacity (23.9 to 33%). Experiment 3 examined the AFB1-binding capacity of viable
and nonviable (HCl-treated) forms of Lp, Lb, and Pa at different pH. Nonviable Lb and Lp, but not Pa, increased
AFB1 binding. Binding of AFB1 was greatest at pH 2.5 and least at pH 8. As the nonviable Lb and Lp that bound
AFB1 in experiment 3 would not be effective silage inoculants, experiment 4 examined effects of benign versus
severe treatments (85 vs. 100°C; pH 2.5 vs. &lt;1) on the viability of Lp, Lb, and Pa. The population of bacteria
was reduced from 9 to 4 log cfu/mL by treatment with HCl at pH 2.5 and to 2 log cfu/mL by 85 or 100°C, whereas
acidification at pH &lt;1 eliminated the bacteria. Experiment 5 determined the effect of the ensiling duration and
benign treatment methods [37 (viable cells) or 85°C (heated cells) or acidification with HCl at pH 2.5 (acid-
treated cells)] on binding of AFB1 and silage quality during the fermentation of corn forage. Corn forage was
ensiled after treatment with only deionized water (control), AFB1 (30 µg/kg of fresh forage), or a mixture of
AFB1 and 109 cfu/g of each of the treated bacteria. Adding AFB1 alone to corn forage reduced the pH decline
during the first 3 d of ensiling and increased or tended to increase butyric acid concentration and final pH after
ensiling for 21 d. Bacterial inoculation inhibited these negative effects. The fermentation profile of silage treated
The aim of this study was to select S. cerevisiae strains able to exert probiotic and antimycotoxin effects plus
antibiotics resistance properties for use in animal production. S. cerevisiae LL74 and S. cerevisiae LL83 were
isolated from bakery by-products intended for use in animal feed and examined for phenotypic characteristics
and nutritional profile. Resistance to antibiotic, tolerance to gastrointestinal conditions, autoaggregation and
coaggregation assay, antagonism to animal pathogens and aflatoxin B1 binding were studied. S. cerevisiae LL74
and S. cerevisiae LL83 showed resistance to all the antibiotics assayed (ampicillin, streptomycin, neomycin,
norfloxacin, penicillin G, sulfonamide and trimethoprim). The analysis showed that exposure time to acid pH had
a significant impact onto the viable cell counts onto both yeast strains. Presence of bile 0.5% increased
significantly the growth of the both yeast strains. Moreover, they were able to tolerate the simulated
gastrointestinal conditions assayed. In general, the coaggregation was positive whereas the autoaggregation
capacity was not observed. Both strains were able to adsorb AFB1. In conclusion, selected S. cerevisiae LL74 and
A strategy to reduce the deleterious effects of mycotoxins is to use dietary supplements that contain
microorganisms that bind mycotoxins and decrease their gastrointestinal absorption. Novel strains were isolated
from a Kefir culture and assessed for their mycotoxin adsorption and biotransformation ability. The most active
strains were identified using DNA sequencing, and the stability of microorganism/mycotoxin complexes was
evaluated using buffer solutions to simulate the pH conditions in the gastrointestinal tract. Our results showed
that the microorganism consortium of Kefir grains adsorbed 82 to 100% of aflatoxin B1 (AFB1), zearalenone
(ZEA) and ochratoxin A (OTA) when cultivated in milk. The main strains that were capable of mycotoxin
adsorption were identified as Lactobacillus kefiri, Kazachstania servazzii and Acetobacter syzygii. The strain L.
kefiri KFLM3 was the most active, adsorbing 80 to 100% of the studied mycotoxins when cultivated in milk.
Nonetheless, the strain K. servazzii KFGY7 retained more mycotoxin after the desorption experiments (65, 69
and 67% for AFB1, OTA and ZEA, respectively). These findings suggest that Kefir consumption may help to reduce
gastrointestinal absorption of these mycotoxins and consequently reduce their toxic effects. The isolated strains
BACKGROUND: Aflatoxins are secondary metabolites due to the growth of molds in animal feed. Lactic acid
bacteria are microorganisms that can absorb aflatoxins. OBJECTIVES: the effect of the yeast Lactobacillus
rhamnosus (PTCC 1637) on Aflatoxin B1 detoxification and absorption of toxin in in vitro (the cow rumen) was
investigated. METHODS: For this purpose, the bacteria used in various treatments (live-treated, autoclave, heat-
treated, treated with acid 100°C) was prepared and added to the rumen of cattle. Aflatoxin B1 in different doses
(0, 5, 10, 20) ppb in the rumen were added and at times one and two hours were incubated at 37°C. The amount
of toxin residues was measured by ELISA using Europroxima kits. RESULTS: The results showed that
microorganisms have been treated in an autoclave have the largest amount toxin removal (90.5 percent)
(p<0.05). Also with increases the incubation time, the amount of toxin absorbed significantly (78%) increased
(p<0.05) and with increasing concentrations of toxin in vitro the bacteria's ability to absorb toxin increases.
During the last century, notable attention has been sporadically directed on health benefits derived from
consumption of milk products such as acidophilus milk. The tangy flavor and thickened texture of acidophilus
milk can be correlated to added Lactobacillus acidophilus bacteria. Aflatoxin M1 (AFM1) is the result of
secondary metabolites of different fungal strains. Because of the important role of dairy products in human life,
especially children, a huge concern raised regarding the presence of AFM1 in consumed products. Due to serious
threats to both human health and economics in all societies, the control of AFM1 is crucial. However, several
physical or chemical detoxification methods were developed to removing or inactivating of AFM1, nowadays
customers prefer to consume the products with lowest changes in composition as well as highest level of
nutrient value. In this context, biological methods can be considered as one of the possible alternatives for
conventional detoxification methods. Recently, the application of Lactic Acid Bacteria (LAB) for the reduction of
AFM1 by approaching of produced metabolites and binding of AFM1 with LAB was introduced. The major aim of
this present review is to highlight the feasibility of using LAB (Streptococcus thermophilus, Streptococcus
Aflatoxin M1 (AFM1) is a group 2b category carcinogenic compound of global concern due to its occurrence in
milk. Various microbes have been employed for the binding of AFM1 and to reduce its bioavailability in humans.
In the current study three strains of lactic acid bacteria, a strain of Saccharomyces cerevisiae and a mixture of all
four were used to evaluate their binding potentials for AFM1. Milk samples were spiked with two different
concentrations of AFM1, 0.05 and 0.1 μg/l, and four concentrations of microbes, 107, 108, 109 and 1010 cfu/ml,
were tested to evaluate their binding potentials. The concentration of AFM1 and microbes were found to
significantly affect the binding potentials of microbes. Saccharomyces cerevisiae, Lactobacillus helveticus and the
mixture of microbes at the concentration of 1010 cfu/ml resulted in 100% binding of AFM1. Lactobacillus
helveticus was found to have a higher binding potential than any other lactic acid bacteria reported previously.
The binding of AFM1 with microbes was reversible as the washing test resulted in ∼20–∼70% release of AFM1.
Current investigation has shown that human exposure to aflatoxins is not limited to the administration of
contaminated cereals, but water is another possible source. This study was aimed to design easily applicable
method to eliminate aflatoxin B2 (AFB2) from contaminated drinking water. Electrospinning has been used for
preparation of probiotic-coated polyvinyl alcohol (PVA) and cellulose acetate (CA) nanofibers. Both of these
hybrid nanofibers were studied by scanning electron microscopy (SEM) and Fourier-transformed infrared
spectroscopy (FT-IR). SEM showed the proper coating of probiotic strains (Kluyveromyces lactis CBS 2359 and
Saccharomyces cerevisiae ATCC 9763) on both nanofiber types. Different areas (1-5 cm2) of the probiotic-
nanofiber hybrid were used to enhance the removal of 20 ng/ml of aflatoxin B2 (AFB2) from prepared AFB2-
contaminated water over time. Results revealed that a 5 cm2 area of probiotic-coated PVA nanofibers can
eliminate 97.5% of AFB2 as compared to 87.5%, 90.5%, 93.5%, and 95.5%, for 1 cm2, 2 cm2, 3 cm2, and 4 cm2,
respectively, while probiotic-coated CA nanofibers were slightly less effective. Nevertheless, the cytotoxicity of
probiotics-CA treated water on cultured human fibroblasts was almost 10 times lower than the cytotoxicity
recorded in probiotics-PVA treated water. Therefore, results of the current research suggest that probiotics-
Aflatoxins (AF) are fungal toxic metabolites produced by Aspergillus flavus and Aspergillus parasiticus that
frequently contaminate a variety of crops, including peanut, corns, and cottonseed. Aflatoxin contamination
causes an estimated average economic loss of $1 billion annually to the US agriculture. Additionally,
contamination of feed ingredients with AF is a major concern to animal agriculture due to their deleterious
effects on livestock such as reduced animal performance and increased mortality. Moreover, the consumption of
AF-contaminated animal products negatively affects public health due to their carcinogenic and
hepatotoxiceffects. The common practices for detoxification of AF in crops include prolonged heating under
pressure and the use of chemicals such as oxidizing agents and ammonia. However, nutrient losses associated
with heat processing and public health safety concerns on chemical residues have limited the applicability of
these methods. Moreover, the inclusion of AF-binding adsorbents in feed to protect livestock from the harmful
effects of AF has been shown to impair nutrient utilization and mineral absorption in livestock. Thus, it is critical
to develop effective strategies for controlling AF in crops, feed ingredients and livestock. Extensive studies have
identified several natural plant- and animal-derived compounds exhibiting potent antifungal or antitoxigenic
properties against toxin producing molds. This chapter highlights the potential of various natural approaches,
In this study, we investigated the potential of Lactobacillus plantarum isolated from Chinese traditional
fermented foods to reduce the toxicity of aflatoxin B1 (AFB1), and its subsequent detoxification mechanism.
Among all the investigated L. plantarum strains, L. plantarum C88 showed the strongest AFB1 binding capacity in
vitro, and was orally administered to mice with liver oxidative damage induced by AFB1. In the therapy groups,
the mice that received L. plantarum C88, especially heat-killed L. plantarum C88, after a single dose of AFB1
exposure, showed an increase in unabsorbed AFB1 in the feces. Moreover, the effects of L. plantarum C88 on
the enzymes and non-enzymes antioxidant abilities in serum and liver, histological alterations of liver were
assayed. The results indicated that compared to the control group, L. plantarum C88 alone administration
induced significant increase of antioxidant capacity, but did not induce any significant changes in the histological
picture. Compared to the mice that received AFB1 only, L. plantarum C88 treatment could weaken oxidative
stress by enhancing the activity of antioxidant enzymes and elevating the expression of Glutathione S-
transferase (GST) A3 through Nuclear factor erythroid (derived factor 2) related factor 2 (Nrf2) pathway.
Furthermore, cytochrome P450 (CYP 450) 1A2 and CYP 3A4 expression was inhibited by L. plantarum C88, and
urinary aflatoxin B1-N7-guanine (AFB-N7-guanine), a AFB1 metabolite formed by CYP 1A2 and CYP 3A4, was
significantly reduced by the presence of viable L. plantarum C88. Meanwhile, the significant improvements were
showed in histological pictures of the liver tissues in mice orally administered with viable L. plantarum C88.
Collectively, L. plantarum C88 may alleviate AFB1 toxicity by increasing fecal AFB1 excretion, reversing deficits in
Aflatoxin M1 is an important mycotoxin mostly found in milk and dairy products. The main objective of this work
was to study the effects of probiotic strains, a probiotic inoculated population, the physiology of probiotic
bacteria and final fermentation pH at four consecutive stages on the reduction of 0.500 ppb of free aflatoxin M1
(AFM1) in Doogh (a traditional Iranian fermented milk drink). Samples’ biochemical, microbial and AFM1 binding
characteristics were monitored during fermentation and storage (5 °C for 28 days). An immunoaffinity column
was used to extract AFM1 and a high-performance liquid chromatograph with a fluorescence detector was used
to measure it. Results showed that Lactobacillus acidophilus was probiotic strain that most reduced free AFM1.
Inoculation of L. acidophilus at 9 log cfu/mL, despite the higher cost, revealed significantly higher free AFM1
binding capacity than 7 log cfu/mL. Heat-killed (dead) L. acidophilus bacteria reduced less free AFM1 at the end
of storage than viable. Samples with a final fermentation pH of 4.5 bound more free AFM1 during fermentation
and storage than those with a pH of 4.2. It is concluded that inoculation of 7 log cfu/mL L. acidophilus viable cells
Aflatoxin exposure remains a health problem in developing countries. The mean concentration of aflatoxin B1 in
maize meal samples from eastern Kenya of 17.4 ppb verified that the food was contaminated. A probiotic
yoghurt was created containing aflatoxin B1 binding Streptococcus thermophilus, Lactobacillus rhamnosus GR-1
and Weissella cibaria NN20 isolated from fermented kimere, a dough food product made from millet. Forty
primary school children, with maize being a regular part of their diet, were randomly assigned to consume
200 mL yoghurt or control milk daily for 7 days, followed by a 7 day washout and another 7 day treatment. After
both 7 day treatment periods, aflatoxin metabolite 1 concentration in urine samples was significantly lower than
baseline in the probiotic group (P > 0.01), but increased in the milk group. The findings were confirmed using
Different types of fermented foods such as chongkukjang, doenjang, ganjang, gochujang, and kimchi are
plentifully available and widely consumed in north eastern Asian countries including Korea. Among them, kimchi
is one of the most popular Korean traditional food. It is prepared by fermenting the baechu cabbage together
with other vegetables and lactic acid bacteria (LAB) with functional potential. Many types of ingredients are
added to kimchi to enhance its taste, flavor, nutritional value, texture etc. A number of bacteria are involved in
the fermentation of kimchi, but LAB are the dominant species in the fermentation process. The addition of other
sub ingredients and formation of different by-products during fermentation eventually leads to eradication of
putrefactive and pathogenic bacteria, and also increase the functionalities, nutritional and nutraceutical
potential of kimchi. Kimchi possesses anti-inflammatory, antibacterial, antioxidant, anticancer, antiobesity,
probiotic properties, cholesterol reduction, and antiaging properties. In the present review an attempt has been
made to review the different types of fermented foods found in the Korean peninsula with detailed scientific
Mycotoxins and their derivatives since their discoveries and until the present time are behind unspecified
economic and medical damages. Aflatoxins are classified according to their physical–chemical and toxicological
characters in the most dangerous row of the mycotoxins. These aflatoxins are in part responsible, of irreversible
medical disasters that are not easily manageable such as cancer of the liver and kidneys, and in the other part, of
losses in the stored cereal products. Based on these crucial findings, monitoring of this toxin became imperative
in post-harvest food products, during storage, during transformation chain and even during the long phases of
conservation. Vigilance of this toxin is delivered by detection methods using very advanced technologies to
respond in the shortest possible times. In addition, the knowledge of factors supporting the biosynthesis of
aflatoxins such as the temperature, moisture content, concentration of nitrogen and carbon, and the molecules
responsible for the genetic control of the synthesis will be reflected later in the choice of bio-control techniques.
This control is currently based on new strategies using the bioactives substances of the plants, the lactic bacteria
and some strains of actinomycetes that have good inhibiting activity against aflatoxins with fewer side effects on
Aflatoxins are potent carcinogenic and immunosuppressive agents. Acute exposure to high level of aflatoxins
leads to aflatoxicosis, which cause rapid death due to liver failure. Immune modulating effects of probiotic
bacteria have good prospects to detoxification of natural foods. This study was aimed to investigate the ability of
Lactobacillus acidophilus strain LA-5 in the presence and absence of yoghurt starter culture for removing
Aflatoxin M1 (AFM1) in comparison with yoghurt starter cultures (108 CFU ml-1). AFM1 detoxification was
evaluated for 21 days of yoghurt storage at 4°C at different concentrations of Aflatoxin (0.1, 0.5 and 0.75 µg L-1).
The amounts of unbound AFM1 were determined using competitive Enzyme-Linked Immunosorbent Assay
(ELISA). L. acidophilus combined with yoghurt starter culture and alone could significantly (P≤ 0.05) remove
AFM1 compared to control group. The results indicated that increasing initial AFM1 concentration in the yoghurt
Food spoilage caused by mycotoxigenic fungi represents an important food safety problem. Lactic acid bacteria
(LAB) are used as starter cultures in a larger number of food products. In this study, 16 strains of LAB were
cultivated in MRS broth under anaerobiosis. Then, cell free supernatants were obtained by centrifugation and
their antifungal activity against Aspergillus parasiticus and Penicillium expansum was tested using the disc-
diffusion method. Furthermore, the LABs that showed in vitro antifungal activity were used in bread
fermentation with yeast in order to study fungal growth inhibition and aflatoxin (AF) reduction in processed
bread previously inoculated with A. parasiticus. The compounds present in the fermented medium of six LAB
strains induced inhibition of P. expansum growth, whereas five probiotic strains produced antifungal compounds
against A. parasiticus. The analysis by liquid chromatography coupled to mass spectrometry in tandem showed a
High contamination by aflatoxin B1 (AFB1) has been detected in Beja province (Tunisia) in many dairy products
and animal feed, which has resulted in many tons of cereals and cereals being removed from the market, causing
economic loss. While removal represents a means of reducing risk, exposures still occur. Studies have
increasingly focused on means of AFB1 biodegradation/elimination using lactic acid bacteria and clay mineral. In
the study here, Lactobacillus paracasei BEJ01 (LP) and montmorilonite clay (MT) were used to reduce the physio-
/immunotoxicologic disorders that could develop in rats that underwent AFB1 exposures for a total of 7
consecutive days. The results indicated that rats treated with AFB1 (80 mg/kg BW) alone had significant
decreases in lymphocytes in their blood (including B-lymphocytes, CD3+, CD4+, and CD8+ T-lymphocyte
subtypes, and NK cells), immunoglobulins (IgA and IgG) and pro-inflammatory cytokines; these rats also had
altered oxidative stress status. In contrast, in rats treated with LP +MT (2_109 cfu/ml [_ 2 mg/kg] + 0.5mg MT/kg
BW) for a total of 7 days before, concurrent with or after AFB1 treatment, there was a significant
blockade/mitigation of each AFB1-impacted parameter. Moreover, treatment with the mixture at any point in
relation to AFB1 treatment expectedly caused enhanced TNF_ and IL-1_ expression relative to control values; all
other parameters were comparable to values noted in control rats. Alone, the mixture had no impact on host
Sorghum is a food and feed crop grown commonly in Asian and African countries. Aflatoxin contamination of
sorghum is a common problem in all tropical countries. Since even low doses of aflatoxin intake, over a period of
time, can prove to be carcinogenic, removal of aflatoxin from sorghum is necessary. Biodecontamination of food
grains is now gaining popularity since it is considered to be a safer alternative to chemical methods. Probiotic
organisms such as lactic acid bacteria (LAB), have been commonly used in fermentation of various food products.
Studies have been done to prove the ability of the probiotic organisms to bind and remove aflatoxin from food.
Here the ability of four strains of lactic acid bacteria (Lactobacillus plantarum, Lactobacillus rhamnosus,
Lactobacillus brevis, and Lactobacillus delbrueckii subsp. lactis) to reduce aflatoxin levels in sorghum during
fermentation has been studied. The initial and final aflatoxin content in sorghum was analyzed using HPLC and
UV spectrophotometer. It was concluded that L. delbrueckii had the maximum antiaflatoxigenic ability. It
Aflatoxins are major contaminants of wide range of food commodities which are susceptible to infection by
Aspergillus, include cereals, oilseeds, spices and tree nuts. In the present study sixty eight lactic acid bacterial
(LAB) cultures were isolated from fermented foods including idli batter, pickle, and fermented porridge (koozhu)
and were screened for their ability to bind Aflatoxin B1 (AFB1). HPLC were used to quantify the aflatoxin during
binding studies. Out of the 68 isolates tested, 45 isolates exhibited their ability to bind AFB1 and 5 strains were
able to bind more than 60% at 2 μg/ml concentration in Phosphate Buffer Saline (PBS) at pH 7. These five strains
were Leuconostoc lactis (86.36%), Lactococcus lactis (78.7%), Bacillus subtilis (67.2%), Pediococcus pentosaceus
(65.12%) and Weissella confusa (60.93%). This will help in the removal of AFB1 naturally from food and feed by
Previous studies conducted in our laboratory have demonstrated that intestinal barrier function can be
adversely affected by diet ingredients or feed restriction, resulting in increased intestinal inflammation-
associated permeability. Two experiments were conducted in broilers to evaluate the effect of three
concentrations of Aflatoxin B1 (AFB1; 2, 1.5, or 1 ppm) on gastrointestinal leakage and liver bacterial
translocation (BT). In experiment 1, 240 day-of-hatch male broilers were allocated in two groups, each group had
six replicates of 20 chickens (n = 120/group): Control feed or feed + 2 ppm AFB1. In experiment 2, 240 day-of-
hatch male broilers were allocated in three groups, each group had five replicates of 16 chickens (n = 80/group):
Control feed; feed + 1 ppm AFB1; or feed + 1.5 ppm AFB1. In both experiments, chickens were fed starter (days 1-
7) and grower diets (days 8-21) ad libitum and performance parameters were evaluated every week. At day 21,
all chicks received an oral gavage dose of FITC-d (4.16 mg/kg) 2.5 h before collecting blood samples to evaluate
gastrointestinal leakage of FITC-d. In experiment 2, a hematologic analysis was also performed. Liver sections
were aseptically collected and cultured using TSA plates to determine BT. Cecal contents were collected to
determine total colony-forming units per gram of Gram-negative bacteria, lactic acid bacteria (LAB), or
anaerobes by plating on selective media. In experiment 2, liver, spleen, and bursa of Fabricius were removed to
determine organ weight ratio, and also intestinal samples were obtained for morphometric analysis.
Performance parameters, organ weight ratio, and morphometric measurements were significantly different
between Control and AFB1 groups in both experiments. Gut leakage of FITC-d was not affected by the three
concentrations of AFB1 evaluated (P > 0.05). Interestingly, a significant reduction in BT was observed in chickens
that received 2 and 1 ppm AFB1. An increase (P < 0.05) in total aerobic bacteria, total Gram negatives, and total
Aflatoxins are carcinogenic mycotoxin, produced by Aspergillus species. These molds infect food crops in warm
humid conditions causing economic losses and affecting the consumers' health adversely. In this study,
antifungal activity and aflatoxin inhibiting ability of four probiotic strains against Aspergillus flavus and
Aspergillus parasiticus were studied. The aflatoxin secreted was analyzed and quantified by both UV
spectrophotometer and HPLC. It was found that Lactobacillus delbrueckii subsp. lactis showed maximal
antifungal (67.43% reduction) and anti-aflatoxigenic (94.33% reduction) activity against A. flavus whereas A.
parasiticus was inhibited by Lactobacillus brevis with the antifungal reduction of 69.38% and anti-aflatoxigenic
Aflatoxins are toxic by-products of fungi, with harmful effects on human and animal health. Although maize is
known to be highly susceptible to aflatoxin contamination, and a staple in many African countries, there is still
lack of methods to mitigate the effects. The effect of lactic acid fermentation on reduction of aflatoxin B1 in
Tanzania maize-based gruel (togwa) by four monocultures (Lactobacillus plantarum, Pediococcus pentosaceus,
Lactobacillus casei and Lactobacillus fermentum), natural fermentation and back-slopping at 30°C for up to 24 h
was investigated. Monocultures removed 45-55% of aflatoxin B1 while natural fermentation and back-slopping
removed 56% and 68% of aflatoxin B1, respectively. Thus, lactic acid fermentation could be a part of a
comprehensive mycotoxicosis prevention strategy in the commonly consumed maize-based gruels. Consumers
could benefit from enhanced food safety through consumption of gruel less contaminated with mycotoxins and
might also benefit from the probiotic effects of lactic acid bacteria. In the scenario where lactic acid bacteria
The physicochemical and microbiological analysis of two types of milk used for the industrial production of
fermented milk such Leben give results in accordance with Algerian standards. These same characteristics reveal
standards compliance for Leben. The amounts of AFM1 determined by competitive ELISA in both types of milk
used for the manufacture of Leben remain higher than those of many countries that have established regulatory
limits for this toxin in milk and dairy products, which range from 0, 05 μg/L for European countries to 0.5 μg/L in
the USA. The AFM1 levels were comparable (p &lt; 0.05) for raw milk and the recombinant, and were
respectively 115,471.10-3 ±1,096 and 119,057.10-3 ±0,821μg/L. From the sixth hour incubation, the Leben
acidity based raw milk or recombined milk, entered into a free exponential phase from 18 ± 0.87 °D and 20 ±
1.32 ° D to 73 ± 1.51 °D and 78± 0.87°D after 18 hours of incubation corresponding to a decrease of 93.34% and
96.39% respectively. This reduction was highly significant from 06 hours of incubation for both types of milk (P
The presence of aflatoxin B1 (AFB1) in pistachio nuts has a serious impact on public health. Kefir grains (KGs)
contain several probiotic microorganisms well known for their aflatoxin decontamination effects. In this study,
the capacity of KG to remove AFB1 from pistachio nuts was investigated and the detoxification method was
optimised using certain statistical approaches. Five variables (toxin concentration, KG level, contact time,
incubation temperature and pre-treatment heat level) were considered in the study. Pistachio samples were
spiked with 5, 10, 15, 20 and 25 ng/g AFB1 stock solutions and then treated with 5, 10, 15, 20 and 25% (w/w,
based on the amount of pistachio paste) KG already heated at 70 and 110 °C. AFB1 concentrations in the
samples were determined using high-performance liquid chromatography. Results of the study indicated that:
(1) KG caused significant decrease in AFB1; (2) cell viability of KG was not necessary for its decontamination
effect; (3) the amount of AFB1 reduction depended on toxin concentration and contact time; and (4) the
interactions between toxin concentration and KG level, toxin concentration and contact time as well as
incubation temperature and contact time had significant effects on AFB1 reduction. The optimum conditions for
the highest AFB1 reduction (96%, compared to the control sample) included a 20 ng/g toxin concentration, 20%
The current study was conducted to evaluate the effect of different doses of Thymus extracts (0, 2, 4 and 6) gr/L
and various values of lactobacillus acidophilus and bifidobacterium bifidum on decreasing of fixed amount of
aflatoxin in kefir beverage. The results showed that Thyme extracts reduced the amount of Aflatoxin M1. Thyme
extract in combination with probiotic bacteria decreased Aflatoxin M1 more than Thymus alone. The most
reduction of Aflatoxin M1 levels was detected by using 4 gr/L Thymus extract and 1×108 Lactobacillus
This chapter discusses the potential ability of probiotic bacteria to bind aflatoxin, a toxin produced by Aspergillus
species of fungi where long-term exposure to this food contaminant is linked to the development of liver cancer.
Its mechanism is unclear, but it is said that aflatoxin binds to the bacterial cell wall or its associated component
and involves a physical attachment rather than through a metabolism process. Findings from in vitro studies,
animal models, and human clinical trials indicated the applicability of probiotic bacteria as preventive agents to
limit human dietary aflatoxin exposure. Although its use in humans is still in its infancy stage, and further studies
to investigate its efficacy are warranted, probiotic bacteria as functional food are an immense value in the area
Aflatoxins (AF) are important foodborne mycotoxins implicated in human health and have immunocytotoxic
effects. The aims of this study were to evaluate a new aflatoxin B1 (AFB1) and fumonisin B1 (FB1)-
binding/degrading micro-organism for biological detoxification, to examine its ability to degrade AFB1 and FB1 in
liquid medium, and to evaluate its potential in vivo protective role against any combined effects from AFB1 and
FB1 on host splenocyte caspase-3 activity (reflecting DNA damage/cell death) and mRNA levels of select
inflammation-regulating cytokines. Balb/c mice were divided into groups (10/group) and treated daily for 2
weeks by oral gavage with AFB1 (80 g/kg BW), FB1 (100 g/kg), AFB1 + FB1, or lactic acid bacteria (Lactobacillus
paracasei BEJ01, 2 × 109 CFU/L, ∼2 mg/kg) - alone or in combination with the AFB1 and/or FB1. After the
exposures, spleens were collected for measures of caspase-3 activity, lipid peroxidation (LP), and glutathione
(GSH) content, expression of anti-oxidation protective enzymes (GPx and SOD), and mRNA levels of inflammation-
regulating cytokines (e.g. IL-10, IL-4, IFNγ, TNF). Thymii were also removed for analysis of apoptosis. The results
indicated that, in the spleen, exposure to the mycotoxins led to increased caspase-3 activity, LP, and IL-10 and IL-
4 mRNA levels, but decreased GSH content and down-regulated expression of GPx and SOD, and of IFNγ and TNF
mRNA. Co-treatment using Lactic Acid Bacteria (LAB) with AFB1 or FB1 suppressed levels of DNA fragmentation,
normalized splenic LP and increased GSH levels, up-regulated expression of GPx and SOD, and normalized mRNA
Aflatoxin B1 is produced by Aspergillus flavus and Aspergillus parasiticus having hepatotoxic, mutagenic,
carcinogenic and immunosuppressant properties. Current study was designed to investigate the effect of
lactobacilli (as probiotic) on the immunosuppression in broilers with experimentally-induced aflatoxicosis and to
compare outcomes with other established adsorbents. At 21st day of age, broilers were segregated into
treatments: AFB (400 µg/kg feed); AFB+Probiotics (PBT2X: 2×109CFU); AFB+PBTX:109CFU; AFB+Mycosorb (1g/kg
feed); Control (basal diet). Treatments were fed to the birds for 2 wk (week 4th and 5th of age). Data was
collected during and for 2 wk post-exposure (Week 6thand 7th).AFB induced reduction of leukocyte count,
relative weight of spleen and bursa of Fabricius and antibody titer against Newcastle Disease Virus. Lactobacilli
significantly protected against suppressive effects of the toxin on TLC, lymphoid organs and antibody titers that
was comparable with the mycosorb. It is concluded that lactobacilli significantly reduced the immunotoxicity of
Mycotoxins have been identified as important toxins affecting animal species and humans ever since the
discovery of aflatoxin B1 in 1960. Mycotoxigenic fungi are ubiquitous in nature and are held responsible for
economic loss as they decrease crop yield and quality of food. The presence of fungi and their mycotoxins are
reported not only in food grains but also in medicinal herbs and processed foods. Since prevention is not always
possible, detoxification of mycotoxins have been attempted using several means; however, only few have been
accepted for practical use, e.g. ammonia in the corn industry. Organizations such as the World Health
Organization, US Food and Drug Administration and European Union have set regulations and safety limits of
important mycotoxins, viz. aflatoxins, fusarium toxins, ochratoxin, patulin zearalenone, etc., to ensure the safety
of the consumers. This review article is a brief and up-to-date account of the occurrence, detection and
Aflatoxins (AFs) are toxic, carcinogenic, immunosuppressive secondary metabolites produced by some
Aspergillus species which colonize crops, including many dietary staple foods and feed components. AFB1 is the
prevalent and most toxic among AFs. In the liver, it is biotransformed into AFM1, which is then excreted into the
milk of lactating mammals, including dairy animals. AFM1 has been shown to be cause of both acute and chronic
toxicoses. The presence of AFM1 in milk and dairy products represents a worldwide concern since even small
amounts of this metabolite may be of importance as long-term exposure is concerned. Contamination of milk
may be mitigated either directly, decreasing the AFM1 content in contaminated milk, or indirectly, decreasing
AFB1 contamination in the feed of dairy animals. Current strategies for AFM1 mitigation include good
agricultural practices in pre-harvest and post-harvest management of feed crops (including storage) and physical
The aim of this study was to investigate the toxic effects of aflatoxins and evaluate the effectiveness of Bacillus
subtilis ANSB060 in detoxifying aflatoxicosis in broilers. A total of 360 one-week-old male broilers (Ross 308)
were assigned to six dietary treatments for five weeks. The treatment diets were: C0 (basal diet), C1.0 (C0 + 1.0 g
B. subtilis ANSB060/kg diet), M0 (basal diet formulated with moldy peanut meal), M0.5, M1.0 and M2.0 (M0 +
0.5, 1.0 and 2.0 g B. subtilis ANSB060/kg diet, respectively). The contents of aflatoxin B1, B2, G1 and G2 in the
diets formulated with moldy peanut meal were 70.7 ± 1.3, 11.0 ± 1.5, 6.5 ± 0.8 and 2.0 ± 0.3 μg/kg, respectively.
The results showed that aflatoxins increased (p &lt; 0.05) serum aspartate transaminase activity, decreased (p
&lt; 0.05) serum glutathione peroxidase activity, and enhanced (p &lt; 0.05) malondialdehyde contents in both
the serum and liver. Aflatoxins also caused gross and histological changes in liver tissues, such as bile duct
epithelium hyperplasia, vacuolar degeneration and lymphocyte infiltration. The supplementation of ANSB060
reduced aflatoxin levels in the duodenum and counteracted the negative effects of aflatoxins, leading to the
Aflatoxins (AF) are ubiquitous mycotoxins contaminating food and feed. Consumption of contaminated food and
feed can cause a severe health risk to humans and animals. A novel biological method could reduce the health
risks of aflatoxins through inhibiting mold growth and binding aflatoxins. Lactic acid bacteria (LAB) are commonly
used in fermented food production. LAB are known to inhibit mold growth and, to some extent, to bind
aflatoxins in different matrices. Reduced mold growth and aflatoxin production may be caused by competition
for nutrients between bacterial cells and fungi. Most likely, binding of aflatoxins depends on environmental
conditions and is strain-specific. Killed bacteria cells possess consistently better binding abilities for aflatoxin B1
(AFB1) than viable cells. Lactobacilli especially are relatively well studied and provide noticeable possibilities in
binding of aflatoxin B1 and M1 in food. It seems that binding is reversible and that bound aflatoxins are released
later on (Haskard et al., 2001; Peltonen et al., 2001). This literature review suggests that novel biological
This study potentiates the adsorbent effect for aflatoxin B1 (AFB1) of a commercial additive (CA) of animal feed,
containing inactive lysate of three Saccharomyces cerevisiae strains, active enzymes, adsorbents and a
selenium–amino acid complex, when the additive was mixed separately with three S. cerevisiae strains. Levels of
AFB1 of 20 and 50 ng g−1 were used to determine the binding capacity of different concentrations of CA alone
and in the presence of yeast strains, as well as toxin desorption, under gastrointestinal conditions. The viability
of yeasts in the presence of CA was evaluated. The results show that the CA did not affect the viability of the
yeast strains assayed. CA alone showed a low percentage adsorption. At 20 and at 50 ng g−1, CA was highly
efficient in adsorbing AFB1 when combined with RC016 and RC012 strains respectively. Desorption of AFB1 by
CA alone and in combination with the yeasts increased with increasing levels of CA. The results demonstrate the
Aflatoxin M1 (AFM1) is a mycotoxin produced by numerous Aspergillus species in pre- or postharvest cereals and
milk. Exposure to AFM1 imparts potent economic losses in the livestock industry. Toxicologically, it also causes
severe immune system problems. The aims of this study were to evaluate a new AFM1-binding/degrading
microorganism for biologic detoxification, to examine its ability to degrade AFM1 in liquid medium, and to
evaluate its potential for in vivo preventative effects against AFM1-induced immunotoxicity and genotoxicity in
mice. Lactobacillus plantarum MON03 (LP) isolated from Tunisian artisanal butter was found to display
significant binding ability to AFM1 in PBS (93%) within 24 h of incubation. Further, the LP was able to tolerate
gastric acidity, bile salts, and adhere efficiently to Caco-3 cells in vitro. The in vivo study used Balb/c mice that
received either vehicle (control), LP only (at 1 × 109CFU/L, ∼1 mg/kg bw), AFM1 (100mg/kg bw), or AFM1 + LP
daily for 15 days (by gavage); two other groups received a single dose of colchicine (4 mg/kg) or mitomycin C (1
mg/kg) as positive controls for induction of micronuclei and chromosomal aberrations, respectively. The results
showed that, compared to in control mice, AFM1 treatment led to significantly decreased body weight gains,
and caused cytotoxic/genotoxic effects as indicated by increases in frequencies of polychromatic erythrocytes,
as well as those with micronucleation (PCEMN) and chromosomal aberrations, among bone marrow cells. The
concurrent administration of LP with AFM1 strongly reduced the adverse effects of AFM1 on each parameter.
Mice receiving AFM1 + LP co-treatment displayed no significant differences in the assayed parameters as
Bacterial diversity and community structure of two maize varieties (white and yellow) during
fermentation/steeping for ogi production, and the influence of spontaneous fermentation on mycotoxin
reduction in the gruel were studied. A total of 142 bacterial isolates obtained at 24-96 h intervals were
preliminarily identified by conventional microbiological methods while 60 selected isolates were clustered into
39 OTUs consisting of 15 species, 10 genera, and 3 phyla by 16S rRNA sequence analysis. Lactic acid bacteria
constituted about 63% of all isolated bacteria and the genus Pediococcus dominated (white maize = 84.8%;
yellow maize = 74.4%). Pediococcus acidilactici and Lactobacillus paraplantarum were found at all steeping
intervals of white and yellow maize, respectively, while P. claussenii was present only at the climax stage of
steeping white maize. In both maize varieties, P. pentosaceus was found at 24-72 h. Mycotoxin concentrations
(μg/kg) in the unsteeped grains were: white maize (aflatoxin B1 = 0.60; citrinin = 85.8; cyclopiazonic acid = 23.5;
fumonisins (B1/B2/B3) = 68.4-483; zearalenone = 3.3) and yellow maize (aflatoxins (B1/B2/M1) = 22.7-513;
citrinin = 16,800; cyclopiazonic acid = 247; fumonisins (B1/B2/B3) = 252-1,586; zearalenone = 205). Mycotoxins
in both maize varieties were significantly (p &lt; 0.05) reduced across steeping periods. This study reports for the
first time: (a) the association of L. paraplantarum, P. acidilactici, and P. claussenii with ogi production from
Several studies have evidenced the ability of some lactic acid bacteria to bind dietary carcinoges, including
aflatoxin B1 (AFB1) and acrylamide (AA). Cell wall teichoic acids (TAs) appear to be components of the bacterial
that might participate in binding; however, the exact mechanisms are not clear. The aims of this work were to
assess the ability of fourteen Lactobacillus strains to bind either AFB1 or AA, and to determine the possible
molecular bonding interactions among toxins and constituents of TAs. Physical binding was assessed in aqueous
solution, and the compontents of TAs were determined by evaluation of hydrolysis products of TAs. Binding was
strain- and toxin-specific dependent. All AFB1-bacterium interactions were partly reversible, while AA-bacterium
appeared to be irreversible. TAs from the evaluated strains consisted of poly(ribitol phosphate) polymers
decorated with glucose, D-alanine and/or glycerol molecules, thus four simplified structures were proposed.
Based on compositional analysis it was hypothesized that hydroxyl groups of AFB1 as well as carbonyl oxygens of
both AFB1 and AA, might be involved in interactions between both toxins and the hydroxyl groups of either
glucose or glycerol in TAs. The results of this work support the suggestion that specific Lactobacillus strains can
Probiotic microorganisms (Saccharomyces cerevisiae var. boulardii, S. cerevisiae UFMG 905, and Lactobacillus
delbrueckii UFV H2b20) were evaluated as biological control agents to reduce aflatoxin and spore production by
Aspergillus parasiticus IMI 242695 in peanut. Suspensions containing the probiotics alone or in combinations
were tested by sprinkling on the grains followed by incubation for seven days at 25°C. All probiotic
microorganisms, in live and inactivated forms, significantly reduced A. parasiticus sporulation, but the best
results were obtained with live cells. The presence of probiotics also altered the color of A. parasiticus colonies
but not the spore morphology. Reduction in aflatoxin production of 72.8 and 65.8% was observed for S. boulardii
and S. cerevisiae, respectively, when inoculated alone. When inoculated in pairs, all probiotic combinations
reduced significantly aflatoxin production, and the best reduction was obtained with S. boulardii plus L.
delbrueckii (96.1%) followed by S. boulardii plus S. cerevisiae and L. delbrueckii plus S. cerevisiae (71.1 and
66.7%, resp.). All probiotics remained viable in high numbers on the grains even after 300 days. The results of
Acrylamide is a chemical naturally formed in certain foods during processing and/or cooking at high
temperatures. Scientific studies have indicated that significant dietary exposure to acrylamide poses a risk for
several types of cancer and may have harmful effects to the nervous system in humans. Hence, methods that
can contribute to reduce acrylamide exposure in foods are needed. Considering that specific lactic acid bacteria
are able to remove a variety of foodborne mutagens, namely heterocyclic amines, lead, cadmium and aflatoxin
B1 by a binding mechanism, the aim of the present study was to assess the ability of fourteen lactic acid strains
to remove acrylamide in vitro. Acrylamide binding ability ranged from 11.89 to 29.12%, and it was found to be
both concentration- and strain-dependent. Lactobacillus reuteri Northern Regional Research Laboratory 14171
(USDA-ARS) and Lactobacillus caseiShirota were the most efficient binders (24.01 and 24.95%, respectively) after
12h of incubation using an acrylamide concentration of 5μg/mL. Acrylamide binding was also pH-dependent, and
the bacterial-toxin complex exhibited high stability assessed by repeated washings. The results of this work
Purpose: Since a sound detoxification method is needed for controlling aflatoxin B1 (AFB1), as one of the most
harmful mycotoxins in animal production and food industry, this study was performed. The paper aims to discuss
these issues. Design/methodology/approach: This study was conducted to examine the ability of Lactobacillus
rhamnosus strain GG to remove AFB1 from liquid media. The binding of AFB1 to Lb. rhamnosus GG was studied
for viable, heat-killed and acid-killed bacteria. AFB1 at concentrations (5, 10 and 20 μg/l) was added to the
bacterial culture (109 cfu/ml) in MRS broth medium and incubated at 25°C for 4, 12 and 24 h. The aflatoxin-
binding capacity of the strain was quantified by the amount of unbound AFB1 using ELISA technique. Findings:
Results showed the AFB1-binding capacity of viable, heat-killed and acid-killed bacteria was about 43, 49 and 50
percent, respectively. The percentage of AFB1 removed was the highest amount in low (5 μg/l) and high (20 μg/l)
concentrations, and there was no significant difference between them (p=0.05). These findings suggest that
lactic acid bacteria can be exploited as an approach to detoxification of aflatoxins from foods. Practical
implications: This method is safe because non-viable bacteria have more ability to remove toxin than viable
Removal of toxic metals and toxins using microbial biomass has been introduced as an inexpensive, new
promising method on top of conventional methods for decontamination of food, raw material and concentrated.
In this article the potential application of lactic acid bacteria and yeasts as the most familiar probiotics to
eliminate, inactivate or reduce bioavailability of contamination in foods and feed has been reviewed. After fast
glance to beneficial health effects and preservative properties of lactic acid bacteria, the mechanisms which
explain antibacterial and antifungal efficiency as well as their antifungal metabolites are mentioned. Then the
article has been focused on potential application of single strain or combination of lactic acid bacteria for
removal of heavy metals (copper, lead, cadmium, chromium, arsenic), cyanotoxins (microcystin-LR, -RR, -LF) and
mycotoxins (aflatoxin B1, B2, B2a, M1, M2, G1, G2, patulin, ochratoxin A, deoxynivalenol, fumonisin B1 and B2, 3-
acetyldeoxynivalenol, deoxynivalenol, fusarenon, nivalenol, diacetoxyscirpenol, HT-2 and T-2 toxin, zearalenone
and its derivative, etc) from aqueous solutions in vitro. Wherever possible the mechanism of decontamination
and the factors influencing yield of removal are discussed. Some factors which can facilitate metal removal
capacity of lactic acid bacteria including the strains, surface charge, pH, temperature, presence of other cations
Kefir is a fermented dairy product, manufactured by starter culture and nowadays consumed widely around the
world. It may become contaminated with aflatoxin M1 (AFM1) which even in small quantities, have harmful
effects on consumers health. There fore, a practical and effective method is needed for detoxification of AFM1
contaminated milk or decreasing its toxicity, such as using different cultures and probiotic agents. Specific lactic
acid bacteria strains have been reported in removing AFM1 fromliquid media by physical binding. The aimof this
study was to detect the effect of kefir starter and Lacto bacillus casei to bind AFM1 in kefirmade frommilk
spikedwith 500pg AFM1 mL-1. Accordingly, five levels of kefir starter (2%,4%,6%,8%,10%) and five levels of Lb.
casei (0.1%, 0.3%, 0.5%, 0.7% and 0.9%) with constant amount of kefir starter (4%) were used, separately. After
48h, the AFM1 content of kefir samples was measured by competitive ELISA technique. Statistical analyses
showed that the sample containing 6% kefir starter had the most reduction in AFM1 concentration (88.17%)
which was significant (p&lt;0.05).Although there were no significant differences (p&lt;0.05) between AFM1
concentration of samples containing 0.5, 0.7 and 0.9% levels of Lb.casei, but the sample containing 0.9% Lb.casei
and 4% kefir starter, had more AFM1 binding (82.12%). Generally, the effect of kefir starter (alone) was more
Mycotoxins are toxic chemicals produced by fungal species that colonize crops in the field or after harvest and
pose a potential threat to the health of humans and domesticated animals. Even when recommended “good
agricultural practices” are implemented to decrease mycotoxin production during crop growth, harvesting, and
storage, the potential for significant contamination still exists. The significance of these unavoidable, naturally
occurring toxicants to human and animal health, the increase in mycotoxin regulations, and the global trans-
shipment of agricultural commodities all highlight the need for strategies to reduce mycotoxin contamination.
These strategies may include any or all of the physical methods, chemical decontamination, adsorbent
treatments, and biological detoxification to decrease mycotoxin bioavailability. Available treatments may reduce
levels of specific mycotoxins; however, no single method is sufficiently effective against the wide variety of
Aims: To acquire data on the safety-in-use of the probiotic Saccharomyces cerevisiae RC016 and test its ability to
reduce genotoxicity caused by dietary aflatoxins (AFs). Methods and Results: The probiotic was orally
administered to Wistar rats. Six groups (n = 6) were arranged: feed and probiotic controls, two levels of AFs-
contaminated feed and two treatments including both the probiotic and the toxin. Genotoxiciy and cytotoxicity
were evaluated with the bone marrow micronuclei assay and the comet assay and internal organs were
macroscopically and microscopically examined. The tested S. cerevisiae strain did not cause genotoxicity or
cytotoxicity in vivo, and it was able to attenuate AFs-caused genotoxicity. Saccharomyces cerevisiae RC016 did
not cause any impairment on the rats' health and it showed no negative impact on the weight gain. Moreover,
RC016 improved zootechnical parameters in AFs-treated animals. The beneficial effects were likely to be caused
by adsorption of AFs to the yeast cell wall in the intestine and the consequent reduction in the toxin's
bioavailability. Conclusions: The dietary administration of RC016 does not induce genotoxicity or cytotoxicity to
Moulds of Aspergillus genus are among the most important causes of food and feed spoilage and can produce
mycotoxins as toxic secondary metabolites when under adverse conditions. Aflatoxins are a group of mycotoxins
that commonly contaminate maize and groundnuts, and are categorized by the International Agency for
Research on Cancer under Class 1A human carcinogens. From the food safety standpoint, one of the most
important mycotoxins is aflatoxin B1 (AFB1). Due to its potent carcinogenic, teratogenic and mutagenic effects
dependent on the level and length of exposure, the presence of this contaminant in food and feed should be
kept as low as achievable. In order to investigate the occurrence of AFB1, determine its concentrations and
explore the possibility of its reduction using different methods, samples of maize, wheat, barley and oat were
collected from different cultivation fields during a three-year period. The immunoassay (ELISA) as a screening
method and high performance liquid chromatography tandem mass spectrometry (LC-MS/MS) as a confirmatory
method were used to determine AFB1 concentrations. Maize contamination seen with AFB1 concentrations
higher than permitted was associated with climate conditions established in the period of concern, which was
extremely warm and dry, and might had favored mould production and AFB1 formation. Substantial to almost
absolute AFB1 reduction in the maize samples was achieved using gamma radiation. A strong antifungal effect
was also obtained upon the use of essential oils and lactic acid bacteria as biological AFB1-reduction alternatives.
As the presence of AFB1 in cereals could be dangerous for human and animal health, in order to prevent its
harmful effects and huge economic problems, the prevention of formation of this contaminant and consistent
Kefir is fermented milk only made from kefir grains and kefir cultures and nowadays consumed widely around
the world. It may become contaminated with aflatoxin M1 (AFM1) which even in small quantities, have
hazardous effects for human beings. Therefore, a practical and effective method is needed to be developed for
the detoxification of AFM1 contaminated milk or decreased its toxicity. It has been reported that specific lactic
acid bacteria are able to remove or degrade AFM1 from liquid media by physical binding. The objective of this
study was to detect the effect of kefir starter and Lactobacillus acidophilus to bind AFM1 in kefir made from milk
spiked with 500 pg AFM1 mL-1. Accordingly, five levels of kefir starter (2, 4, 6%, 8 and 10%) as group 1 and five
levels of Lb. acidophilus (0.1, 0.3, 0.5, 0.7 and 0.9%) with constant amount of kefir starter (4%) as group 2 were
used. After 48h, the AFM1 content of kefir samples was measured by competitive ELISA technique. Statistical
analyses, in group 1, showed that the sample containing 6% kefir starter had the most reduction in AFM1
concentration (88.17%) which was significant (p&lt;0.05). In group 2 the sample containing 0.9% Lb. acidophilus
and 4% kefir starter had the maximum amount of AFM1 binding (89.04%) and there were no significant
differences (p&lt;0.05) between 0.3, 0.5 and 0.7% levels in AFM1 reduction. Generally, the effect of kefir starter
Probiotics and lactic acid bacteria (LAB) are widely used in food fermentation and preservation. This study was
carried out to assess the potential of five strains of LAB and bifidobacteria to remove aflatoxin M1 (AFM1) from
yoghurt. The stability of the AFM1 complexes formed with them in both viable and non-viable (heat- or acid-
treated) forms was assessed by repetitive aqueous extraction. Strains with high ability in removing AFM1 were
selected to study its ability to remove AFM1 from spiked milk during yoghurt production and storage. Three
treatments from yoghurt that produced from spiked milk were studied. Treatment A was the control using
fermented yoghurt culture (Streptococcus thermophilus and Lactobacillus bulgaricus). Treatment B was
fermented by 50% yoghurt culture (S.thermophilus and L.bulgaricus) and 50% Lactobacillus plantrium.
Treatment C was fermented by yoghurt culture (S. thermophilus and L.bulgaricus) and 50% Lactobacillus
acidophilus. The samples were collected during different storage (5°C) times (1, 3, 5 and 7 days) to determine
the ability of the stains to reduce AFM1. The results indicted that there were significant differences (P<0.05)
between the strains in their ability to reduce AFM1 in MRS broth media in the viable stage, heated stage and
acid treatment. L.plantrium was the highest strain capable of removing AFM1. Yoghurt fermented by 50%
The purpose of this study was to investigate the aflatoxin B1 binding of lactic acid bacteria (LAB) isolated from
Korean traditional soybean paste and to evaluate the effect of incubation conditions and physico-chemical
factors on the binding ability of LAB to this mutagen. The amount of aflatoxin B1 bound by Enterococcus faecium
DJ22, Lactobacillus fermentum DJ35, Lactobacillus rhamnosus DJ42, and Lactobacillus pentosus DJ47 was strain
specific with the percent bound ranging from 19.3% to 52.1%. However, Enterococcus faecalis DJ14,
Lactobacillus panis DJ29, and Pediococcus halophilus DJ50 strains did not exhibit any of the binding ability to
aflatoxin B1. For most strains, the binding ability was significantly affected by the environmental conditions such
as the aflatoxin B1 level, incubation time and temperature, and the initial cell count of LAB. The stability of the
aflatoxin B1-bacteria complexes was significantly more unstable after washing. In addition, the binding stability
between viable and nonviable cells was not statistically significant. Treatment with heating, acidic pH, a-amylase,
protease, lysozyme, or sodium metaperiodate caused a significant (P<0.05) decrease in aflatoxin B1 binding for
the tested strains, suggesting that carbohydrates or proteins in the cell walls may be involved in aflatoxin B1
Yogurt is a fermented dairy product, manufactured by starter culture and consumed widely around the world. It
may become contaminated with aflatoxin M1(AFM1) that causes threats to the health of consumers, especially
young children and adults. There are different methods to detoxify foods from AFM1, but in yogurt, the easiest
way is biodetoxification method using different cultures and probiotic agents. So the aim of this study was to
evaluate the effect of starter culture and Lactobacillus casei in detoxification of AFM1. For this purpose skim milk
powder was contaminated artificially with AFM1 at levels: 0.05, 0.1, 0.5, and 0.75 ppb. Yogurt samples including
control (inoculation just by started culture-YC280) and treatments (inoculation by starter culture and Lb.casei-
431) fermented at 42°C to reach pH&lt;4.6 and consequently the AFM1 content was measured by ELISA
technique. Results showed that in the control samples and treatments, the toxin was removed 94.35 and 94.15
The aim of this study was to investigate the effectiveness of lactic acid bacteria and their bacteriocins in
detoxifying aflatoxins. Aflatoxin B1 detoxification abilities of lactic acid bacteria both in liquid culture and as
concentrated pellets, their bacteriocins, and mixtures of these 3 were evaluated. Mixed cultures of the 2
bacteria were also investigated. Lactobacillus plantarum and Lactococcus lactis were separately able to detoxify
aflatoxin B1 in solutions. Lb. plantarum had a better detoxification rate (46%) than Lc. lactis (27%). After heat
treatment, only the groups that contained pellets released the bound toxin back into the solution. Although
bacteria and their bacteriocins were effective individually at detoxification, their efficacy was increased when
they were used together. When Lc. lactis and Lb. plantarum were incubated in separate tubes and then mixed,
that group had a significantly increased ability to bind toxins (59%) compared to their use alone. When these 2
strains were incubated together in a single broth culture, the most successful detoxification rate (81%) was
Objective: To determine lactic acid bacteria's capability to enhance the process of binding and isolating aflatoxin
B1 and to utilize such lactic acid bacteria as a food supplement or probiotic products for preventing absorption
of aflatoxin B1 in human and animal bodies. Methods: In the present research, the bacteria were isolated from
five different sources. For surveying the capability of the bacteria in isolating aflatoxin B1, ELISA method was
implemented, and for identifying the resultant strains through 16S rRNA sequencing method, universal primers
were applied. Results: Among the strains which were isolated, two strains of Lactobacillus pentosus and
Lactobacillus beveris exhibited the capability of absorbing and isolating aflatoxin B1 by respectively absorbing
and discharging 17.4% and 34.7% of the aforementioned toxin existing in the experiment solution. Conclusions:
Strains of Lactobacillus pentosus and Lactobacillus beveris were isolated from human feces and local milk
This study was conducted to investigate the toxic effects of aflatoxins and the efficacy of Bacillus subtilis
ANSB060 for the amelioration of aflatoxicosis in broiler chickens. Six replicates of ten broilers each were
assigned to one of seven dietary treatments, which were labeled C0 (basal diet); M0 (basal diet containing moldy
peanut meal); C500 and C1000 (C0+500 or 1000g/t aflatoxin biodegradation preparations, composed mainly of
ANSB060); and M500, M1000 and M2000 (M0+500, 1000 or 2000g/t aflatoxin biodegradation preparations). The
concentrations of aflatoxin B1, B2, G1 and G2 in the moldy diets (M0, M500, M100 and M2000) fluctuated
around 70.7±1.3, 11.0±1.5, 6.5±0.8 and 2.0±0.3μg/kg, respectively. The results showed that the M0 diet caused
a significant decrease in average daily weight gain and increased feed requirements, with a gain ratio increasing
from d 8 to 42, deterioration in meat quality and aflatoxin residues in broilers' livers as compared with the C0
diet. The addition of ANSB060 to the aflatoxin-contaminated diets offset these negative effects, leading to the
Aflatoxins (AFs), the secondary metabolites produced by species of Aspergillus, have harmful effects on humans,
animals, and crops that result in illnesses and economic losses. This investigation was designed to assess the
potential of four strains of lactic acid bacteria (LAB) for removing AFs invitro. The stability of AFs complexes
formed with LAB in viable and non-viable (heat treated) forms was assessed. The strain with the highest ability
to bind AFs was selected to study its impact on removing AFs from contaminated wheat flour during baladi bread
baking process. Three treatments of baladi bread produced from contaminated wheatflour were formulated
including treatment (A) as control fermented by bakery yeast, treatment (B)fermented by Lactobacillus
rhamnosus TISTR 541 and treatment (C) fermented by the mixture of bakery yeast and L. rhamnosus. The
samples were collected during different steps of bread baking process. The results indicated that there were
significant differences between the amounts of AFB and AFG detected in each sample and there were significant
differences between the strains in their ability to bind AFB and AFG in the viable and heated stage. L. rhamnosus
The aim of this study was to evaluate the ability of seven lactic acid bacteria (LAB) strains to remove aflatoxin
M1 (AFM1) in phosphate buffer saline (PBS) and in skimmed milk samples. The mean AFM1 removal by LAB in
PBS ranged from 5.60 ± 0.45% to 45.67 ± 1.65% (n = 3). Heat-killed cells showed AFM1 removal percentages
significantly higher than viable cells in contact times of 15 min or 24 h, although there were no significant
differences between those times. AFM1/LAB complex resulted from tests with PBS was unstable, as 40.57 ±
4.66% to 87.37 ± 1.82% of AFM1 retained by bacteria were recovered in solution after washing with PBS. Heat-
killed cells of Lactobacillus rhamnosus, Lactobacillus delbrueckii spp. bulgaricus, and Bifidobacterium lactis had
the highest percentage (&gt;33%) of AFM1 removal in PBS tests. In ultrahigh temperature (UHT) skimmed milk
spiked with AFM1, the three selected LAB strains showed no significant differences in removing AFM1 at 37 °C,
and only B. lactis had greater ability to remove AFM1 at 4 °C. Results demonstrated that AFM1 removal by LAB
In order to degrade aflatoxin B1 (AFB1), AFBl-degrading microbes (probiotics) such as Lactobacillus casei, Bacillus
subtilis and Pichia anomala, and the AFBl-degrading enzyme from Aspergillus oryzae were selected and
combined to make feed additive. Seventy-five 43-day-old male Arbor Acres broilers were randomly divided into
5 groups, 15 broilers for each group. The broilers were given with 5 kinds of diets such as the basal diet,
400μg/kg AFB1 supplement without feed additive, and 200, 400, 800μg/kg AFB1 supplement with 0.15% feed
additive. The feeding experimental period was 30d, which was used to determine production performance of
broilers. In addition, serum, liver and chest muscle were selected for measuring AFB1 residues, gene expressions,
microscopic and antioxidant analyses. The results showed that adding 0.15% feed additive in broiler diets could
significantly relieve the negative effect of AFB1 on chicken's production performance and nutrient metabolic
rates (P&lt;0.05). It could also improve AFB1 metabolism, hepatic cell structure, antioxidant activity, and many
hepatic enzyme gene expressions involved in oxidoreductase, apoptosis, cell growth, immune system and
Biological decontamination of mycotoxins using microorganisms is one of the well known strategies for the
management of mycotoxins in foods and feeds. In this study, the interaction of aflatoxin B1 (AFB1) in cottonseed
with Lactobacillus rhamnosus strain GG was investigated for the first time. AFB1 at concentrations (5, 10 and 20
μg/l) was added to the cottonseed meal in buffer phosphate solution and then bacterial culture (109 CFU/ml) in
MRS broth medium was added to the solution and incubated at 25°C for 4, 12 and 24 hrs. The aflatoxin binding
capacity of the strain was quantified by the amount of unbound AFB1 using ELISA technique. Results showed the
binding capacity of viable, heat killed and acid killed bacteria respectively 44, 47 and 49%. Removal of AFB1 by
this strain was a slow process with approximately 41% AFB1 removal at both 12 and also 24 hrs. The primary
concentration of AFB1 did not influence the efficacy of detoxification (p&gt;0.05). These findings further support
Aflatoxin M1 (AFM1) has been detected in many parts of the world both in raw milk and many dairy products,
causing great economic losses and human disease. Unfortunately, there are few studies dealing with AFM1
immunotoxicity/interactions with lactic acid bacteria for potential application as a natural preventive agent. The
aim of this study was to isolate (from dairy products) food-grade probiotic bacteria able to degrade/bind AFM1
in vitro and evaluate whether the same organism(s) could impart a protective role against AFM1-induced
immunotoxicity in exposed Balb/c mice. Bacteria (Lactobacillus plantarum MON03 and L. rhamnosus GAF01)
were isolated from Tunisian artisanal butter and then tested for abilities to eliminate AFM1 from phosphate-
buffered saline (PBS) and reconstituted milk (containing 0.05, 0.10, and 0.20 g AFM1/ml) after 0, 6, and 24h at
37°C. Results showed that the selected bacteria could 'remove' AFM1 both in PBS and skimmed milk. The binding
abilities of AFM1 by L. plantarum MON03 and L. rhamnosus GAF01 strains (at 10 CFU/ml) in PBS and
reconstituted milk ranged, respectively, from 16.1-78.6% and 15.3-95.1%; overall, L. rhamnosus showed a better
potential for removal than L. plantarum. 'Removal' appeared to be by simple binding; the bacteria/AFM1
complex was stable and only a very small proportion of mycotoxin was released back into the solution. L.
rhamnosus GAF01 had the highest binding capacity and was selected for use in the in vivo study. Those results
indicated that use of the organism prevented AFM1-induced effects on total white and red blood cells, and
lymphocyte subtypes, after 15 days of host treatment. These studies clearly indicated that L. rhamnosus GAF01
Physicians are increasingly being asked to diagnose and treat people made ill by exposure to water-damaged
environments, mold, and mycotoxins. In addition to avoidance of further exposure to these environments and to
items contaminated by these environments, a number of approaches have been used to help persons affected
by exposure to restore their health. Illness results from a combination of factors present in water-damaged
indoor environments including, mold spores and hyphal fragments, mycotoxins, bacteria, bacterial endotoxins,
and cell wall components as well as other factors. Mechanisms of illness include inflammation, oxidative stress,
toxicity, infection, allergy, and irritant effects of exposure. This paper reviews the scientific literature as it relates
to commonly used treatments such as glutathione, antioxidants, antifungals, and sequestering agents such as
Aims: Aflatoxin B1 (AFB1) is considered as the most toxic food contaminant, and microorganisms, especially
bacteria, have been studied for their potential to reduce the bioavailability of mycotoxins including aflatoxins.
Therefore, this research investigated the efficacy of oral administration of Lactobacillus casei Shirota (LcS) in
aflatoxin-induced rats. Methods and Results: Sprague Dawley rats were divided into three groups of untreated
control, the group induced with AFB1 only, and the group given probiotic in addition to AFB1. In the group
induced with AFB1 only, food intake and body weight were reduced significantly. The liver and kidney enzymes
were significantly enhanced in both groups induced with AFB1, but they were lower in the group given LcS. AFB1
was detected from all serum samples except for untreated control group's samples. Blood serum level of AFB1 in
the group induced with AFB1 only was significantly higher than the group which received probiotic as a
treatment (P &lt; 0·05), and there was no significant difference between the control group and the group treated
with probiotic. Conclusions: LcS supplementation could improve the adverse effect of AFB1 induction on rats'
body weight, plasma biochemical parameters and also could reduce the level of AFB1 in blood serum.
Significance and Impact of the Study: This study's outcomes contribute to better understanding of the potential
Aflatoxin M1 (AFM1) is a highly toxic compound, stable during milk processing, cheese ripening and storage.
Hence, it may be found as contaminant in milk and dairy products with hazardous effects for human beings.
Different efforts have been made to detoxify toxin-contaminated food, or to decrease its absorption at intestinal
level. In this regard, several in vitro and in vivo studies have demonstrated the potential of probiotic bacteria to
remove aflatoxins from model systems. Therefore, the aims of this study were to assess the ability of five
probiotic strains to bind AFM1 in PBS, and to reduce its bioaccessibility in artificially contaminated milk using an
in vitro digestive model. All assessed strains exhibited different degrees of aflatoxin binding in PBS, ranging from
19.95 to 25.43%. Moreover, AFM1's bioaccessibility in the in vitro digestive model was reduced from 22.72 to
45.17%, depending on assessed probiotic strain. The results of this work suggest that the probiotic strains tested
The purpose of this study was to evaluate the ability of a Saccharomyces cerevisiae strain and a pool of three
lactic acid bacteria (LAB) strains (Lactobacillus rhamnosus, Lactobacillus delbrueckii spp. bulgaricus and
Bifidobacterium lactis), alone or in combination, to bind aflatoxin M1 (AFM1) in UHT (ultra-high-temperature)
skim milk spiked with 0.5 ng AFM1 mL-1. All the LAB pool (1010 cells mL-1) and S. cerevisiae (109 cells mL-1)
cells were heat-killed (100 °C, 1 h) and then used for checking the effect of contact time (30 min or 60 min) on
toxin binding in skim milk at 37 °C. The mean percentages of AFM1 bound by the LAB pool in milk were 11.5 ±
2.3% and 11.7 ± 4.4% for 30 min and 60 min, respectively. Compared to the LAB pool, S. cerevisiae cells had
higher (P &lt; 0.05) capability to bind AFM1 in milk (90.3 ± 0.3% and 92.7 ± 0.7% for 30 min and 60 min,
respectively), although there were not significant differences between the contact times evaluated. When using
S. cerevisiae + LAB pool, a significant increase (P &lt; 0.05) was observed in the percentage of AFM1 bound
There is a need to develop food-compatible conditions to alter the structures of fungal, bacterial, and plant
toxins, thus transforming toxins to nontoxic molecules. The term 'chemical genetics' has been used to describe
this approach. This overview attempts to survey and consolidate the widely scattered literature on the inhibition
by natural compounds and plant extracts of the biological (toxicological) activity of the following food-related
toxins: aflatoxin B1, fumonisins, and ochratoxin A produced by fungi; cholera toxin produced by Vibrio cholerae
bacteria; Shiga toxins produced by E. coli bacteria; staphylococcal enterotoxins produced by Staphylococcus
aureus bacteria; ricin produced by seeds of the castor plant Ricinus communis; and the glycoalkaloid α-
chaconine synthesized in potato tubers and leaves. The reduction of biological activity has been achieved by one
or more of the following approaches: inhibition of the release of the toxin into the environment, especially food;
an alteration of the structural integrity of the toxin molecules; changes in the optimum microenvironment,
especially pH, for toxin activity; and protection against adverse effects of the toxins in cells, animals, and humans
(chemoprevention). The results show that food-compatible and safe compounds with anti-toxin properties can
be used to reduce the toxic potential of these toxins. Practical applications and research needs are suggested
that may further facilitate reducing the toxic burden of the diet. Researchers are challenged to (a) apply the
available methods without adversely affecting the nutritional quality, safety, and sensory attributes of animal
This study was carried out to examine aflatoxin B1 (AFB1) removal ability of four strains of lactic acid bacteria
(LAB). Three indigenous (Lactobacillus rhamnosus TMU094, Lactobacillus fermentum TMU121 and Pedioccus
pentosaceus TMU457) and a non-indigenous (Labacillus rhamnosus PTCC1637) isolates were studied. The strains
were incubated with (AFB1) at different time. The toxin residual in the supernatant was determined. Reduction
of the toxin quantity was observed by all species. Binding of aflatoxin by the studied LAB varied from 19.41 to
75.06%. Aflatoxin-binding activity showed time dependent trend taking into consideration of different
incubation periods. Lactobacillus rhamnosus TMU094 bound 25.64 to 75.06%, L. fermentum bound 38.63 to
72.15%, P. pentosaceus bound 24.86 to 63.21% and L. rhamnosus PTCC1637 bound 19.41 to 35% of AFB1 during
Contamination of milk and dairy products with aflatoxin M 1 (AFM 1) continues to receive increased attention
because of its potential health hazard to humans. The first aim of this study was to know the occurrence and
levels of AFM 1 in whole UHT milk from main processors in Turkey in order to make a preliminary exposure
assessment. A total of 40 milk samples were analysed for AFM 1 using high performance liquid chromatography
with fluorescence detection (HPLC-FD) after immunoaffinity column clean-up. Aflatoxin M 1 was detected in 20%
of samples at levels ranging from &lt;0.004 to 0.076 μg l -1. Only two samples contained AFM 1 above the EU
limit of 0.05 μg l -1.The second aim of this study was to determine the bioaccessibility of AFM 1 from milk using
an in vitro digestion model. The bioaccessibility of AFM 1 in spiked and naturally contaminated milk samples
ranged from 80.5 to 83.8% and from 81.7 to 86.3%, respectively. No difference (P &gt; 0.05) in AFM 1
bioaccessibility was found between spiked and naturally contaminated milk samples. This study also assessed
the binding of AFM 1 by six probiotic bacteria under simulated gastrointestinal conditions. A 15.5-31.6%
reduction in AFM 1 bioaccessibility was observed in the presence of probiotic bacteria. Based on the results
In this study the aflatoxin B1 (AFB1) removal capacity, the tolerance to salivary and gastrointestinal conditions,
autoaggregation and coaggregation with pathogenic bacteria of Saccharomyces cerevisiae strains isolated from
broiler feces, were evaluated. Only four of twelve isolated strains were identified as Saccharomyces cerevisiae
using molecular techniques. The results obtained in AFB1 binding studies indicated that the amount of AFB1
removed was both strain and mycotoxin-concentration dependent. Therefore, a theoretical model was applied
in order to select the most efficient strain to remove AFB1 in a wide range of mycotoxin concentration. The
results indicated that S. cerevisiae 08 and S. cerevisiae 01 strains were the most efficient microorganisms in the
mycotoxin removal. Viability on simulated salivary and gastrointestinal conditions was investigated and S.
cerevisiae 08 strain showed the best results, achieving 98% of total survival whereas S. cerevisiae 01 reached
only 75%. Autoaggregation and coaggregation assays showed S. cerevisiae 08 as the most appropriate strain,
mainly because it was the unique strain able to coaggregate with the four bacterial pathogens assayed.
Consequently, S. cerevisiae 08 is the best candidate for future in vivo studies useful to prevent aflatoxicosis.
Further quantitative in vitro and in vivo studies are required to evaluate the real impact of yeast-binding activity
The present study aims to investigate the bioaccessibility of aflatoxin (AF) from various spiked food matrices
(peanut, pistachio, hazelnut, dried figs, paprika, wheat and maize) using an in vitro digestion model under fed
conditions. In addition, the effectiveness of six probiotic bacteria in reducing AF bioaccessibility was evaluated
with an in vitro digestion model. The bioaccessibility of AFs from seven food matrices ranged from 85.1 to 98.1%
for aflatoxin B 1 (AFB 1), 83.3 to 91.8% for aflatoxin B 2 (AFB 2), 85.3 to 95.1% for aflatoxin G 1 (AFG 1) and 80.7
to 91.2% for aflatoxin G 2 (AFG 2). The bioaccessibilities of all four compounds were independent of the spiking
level and food matrix. The inclusion of probiotic bacteria showed significant (p&lt;0.05) reduction in the
bioaccessibility of AFs: up to 35.6% for AFB 1, 35.5% for AFB 2, 31.9% for AFG 1 and 33.6% for AFG 2. AF-binding
activity of probiotic bacteria in simulated gastrointestinal conditions was reversible, and 10.3-39.8% of bound
Aims: To examine Saccharomyces cerevisae strains with previously reported beneficial properties and aflatoxin
B1 binding capacity, for their ability to remove ochratoxin A (OTA) and zearalenone (ZEA) and to study the
relation between cell wall thickness and detoxificant ability of yeast strains. Methods and Results: A mycotoxin
binding assay at different toxin concentrations and the effect of gastrointestinal conditions on mycotoxin binding
were evaluated. Ultrastructural studies of yeast cells were carried out with transmission electronic microscopy.
All tested strains were capable of removing OTA and ZEA. Saccharomyces cerevisiae RC012 and RC016 showed
the highest OTA removal percentage, whereas RC009 and RC012 strains showed the highest ZEA removal
percentages. The cell diameter/cell wall thickness relation showed a correlation between cell wall amount and
mycotoxin removal ability. After exposure to gastrointestinal conditions, a significant increase in mycotoxin
binding was observed. Conclusions: All tested Saccharomyces cerevisiae strains were able to remove OTA and
ZEA, and physical adsorption would be the main mechanism involved in ochratoxin A and ZEA removal.
Gastrointestinal conditions would enhance adsorption and not decrease mycotoxin-adsorbent interactions.
Significance and Impact of the Study: Live strains with mycotoxin binding ability and beneficial properties are
The human health impact of AFB1 exposure is widespread in developing countries, is known to cause
teratogenicity, immunotoxicity, hepatotoxicity and even death in farm animals and humans. As information has
accumulated on toxicological effects of AFB1, preventive strategies whose objective is to reduce the risks to
human health have been implemented. Chemical, physical and biological strategies for the control and
prevention of aflatoxicosis and AFB1-associated diseases are described. In a primary prevention trial, the
purpose is to reduce exposure to AFB1 in the diet. In secondary prevention trials, one goal is to modulate the
metabolism of ingested AFB1 to enhance detoxification processes, thereby reducing internal doses and
subsequent risk. This information suggested the probable application of different strategies as an alternative
method to avoid the effects produced by the AFB1. The extent to which these interventions may ultimately
The objectives of this investigation were to evaluate the ability of Saccharomyces cerevisiae CECT 1891 and
Lactobacillus acidophilus 24 to remove fumonisin B 1 (FB 1) from liquid medium; to determine the nature of the
mechanism involved in FB 1-microorganism interaction and to analyze whether the presence of aflatoxin B 1
(AFB 1) interferes with the removal of FB 1 and vice versa. The results obtained indicated that: (i) both
microorganisms were able to remove FB 1 from liquid medium; (ii) the removal was a fast and reversible
process; (iii) cell viability was not necessary; (iv) the amount of FB 1 removed was both toxin- and microorganism
concentration-dependent; (v) the process did not involve chemical modification of FB 1 molecules; and (vi) cell
wall structural integrity of the microorganisms was required for FB 1 removal. Consequently, we propose that
the mechanism involved in the removal of FB 1 is a physical adsorption (physisorption) of the toxin molecule to
cell wall components of the microorganisms. It is highly probable that FB 1 and AFB 1 co-occur in contaminated
foods, since the fungal genera Aspergillus and Fusarium frequently occur simultaneously. Therefore, we analyzed
whether the presence of AFB 1 interferes with the removal of FB 1 by the microorganisms previously evaluated,
and vice versa. Studies of co-occurrence of both mycotoxins clearly showed that they did not compete for
binding sites on the microorganism cell wall and the presence of one toxin did not modify the efficiency of the
The effect of Saccharomyces cerevisiae RC008 and RC016, previously selected based on their aflatoxin B1
binding ability and beneficial properties, against Aspergillus parasiticus under different interacting environmental
conditions was evaluated. Studies concerning the lag phase, growth rate and aflatoxin B1 production were
carried out in vitro under different regimes of aw (0.95 and 0.99), pH (4 and 6), temperature (25 and 37°C), and
oxygen availability (normal and reduced). Both yeast strains showed great antagonistic activity at pH 4,
decreasing growth rate compared with the control. The RC008 strain showed the greatest inhibitory activity at
all assayed conditions. A. parasiticus produced large amounts of AFB1in vitro. A significant decrease of AFB1
levels in comparison with the control were observed with yeast interaction. Differences between control and
treatment values ranged from 130 to 5400 ng ml-1. S. cerevisiae RC008 and RC016 could be considered as
effective agents in reducing growth and AFB1 production at different interacting environmental conditions,
related to that found in stored feedstuff. The importance of the present work lies in the search for live strains
Two experiments were performed to screen bacilli isolated from quails for their aflatoxin removal potential and
to assess the efficiency of their amelioration of experimental aflatoxicosis. Nonhemolytic bacilli were selected
for in vitro aflatoxin B1 (AFB1) removal and conventional probiotic tests. The isolate with the highest scores was
selected for assessment in field experiments and was identified as Berevibacillus laterosporus (Bl). In the second
experiment, 125 male Japanese quails (21 d old) were divided into 5 groups with 5 replications to compare the
toxin removal efficiency of Bl with that of a commercial toxin binder, improved Millbond-TX (IMTX). The
experimental groups were as follows: Control (without any feed additive or AFB1); AFB1 (2.5 mg/kg); AFB1 + Bl
(2.5 mg/kg + 108 cfu/ mL); AFB1+IMTX (2.5 mg/kg + 2.5 g/kg); and Bl (108 cfu/mL). The greatest BW gain and
slaughter and carcass weights were found in the Bl group and the lowest values were observed in the AFB1
group (P &lt; 0.05). Feeding AFB1 alone to the chicks resulted in a significant decrease in serum albumin, total
protein, and glucose and cholesterol levels but a significant increase in serum uric acid, urea, creatinin and
phosphrus (P &lt; 0.05). Treatment of birds on AFB1 with Bl restored these to their original levels (P &lt; 0.05).
AFB1 + Bl-fed birds had serum aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase,
and alkaline phosphatase enzyme activity similar to control birds (P &lt; 0.05). Antibody titer against Newcastle
disease virus was found to be lowest in the AFB1 group but highest in the Bl group (P &lt; 0.05). Antibody
production against sheep red blood cells was lower in the AFB1 group compared with the AFB1 + Bl group (P &lt;
The present investigation was carried out to evaluate the hepatoprotective effect of probiotic fermented milk
(FM) containing Lactobacillus rhamnosus GG and Lactobacillus casei strain Shirota, alone as well as in
combination with chlorophyllin (CHL) as an antioxidant agent in male Wistar rats administered aflatoxin-B1
(AFB1). AFB1 was injected intraperitoneally at the rate of 450 μg/kg body weight per animal twice a week for 6
weeks, maintaining an equal time interval between the two consecutive AFB1 administrations. A total of 125
male Wistar rats were randomly allocated to five groups, each group having twenty-five animals. Group I was
offered FM containing L. rhamnosus GG and L. casei strain Shirota. Group II was administered AFB1 and served
as the control group; group III was administered FM-AFB1, in which besides administering AFB1, FM was also
offered. Group IV was offered CHL and AFB1, and group V was offered both FM and CHL along with AFB1. The
rats were euthanised at the 15th and 25th week of the experiment and examined for the biochemical and
hepatopathological profile. A significant reduction in thiobarbituric acid-reactive substances (TBARS) was
observed in the FM-CHL-AFB1 group compared with the AFB1 control group. FM alone or in combination with
CHL was found to show a significant (P &lt; 0·05) hepatoprotective effect by lowering the levels of TBARS and by
The aim of this study was to investigate the chemopreventive effect of probiotic fermented milk and
chlorophyllin on aflatoxin B 1 (AFB 1) induced hepatocellular carcinoma. In vivo trials were conducted on 200
Wistar rats allocated to eight groups. Rats in the positive control group were given intraperitoneal injection of
aflatoxin B 1 at 450μg/kg body weight twice a week for 6weeks. The rats were sacrificed and dissected at 25th
week of the experiment, and comet assay was carried out in hepatic cells to assess the genotoxicity or DNA
damage. The tumour incidence was decreased by approximately one-third than AFB 1 control group. The
expression of c-myc bax, bcl-2, cyclin D1, p53 and rasp-21 genes was also studied. A significant (P&lt;0.05)
reduction in DNA damage was observed in probiotic fermented milk with chlorophyllin group as compared to
aflatoxin B 1 control group. The c-myc, bcl-2, cyclin D1 and rasp-21 level was found to be highest in AFB 1 control
group as compared to the treatment group. The results advocate the enhanced protective potential of probiotic
The aim of the present study was to evaluate the inhibitory effect of Enterococcus faecium and Lactococcus
lactis subsp. lactis isolated from faeces of healthy dogs on (i) lag phase, (ii) growth rate, and (iii) aflatoxin B 1
production by Aspergillus section Flavi on in vitro assays. Thirteen lactic acid bacteria (LAB) isolates were used as
antagonist microorganisms. Antagonistic activity was assayed against four potentially aflatoxigenic Aspergillus
section Flavi isolates: A. flavus (AF210 and AF281), A. parasiticus (AP245) and A. parasiticus (NRRL 2999). In
general, the longest lag phases of Aspergillus isolates were obtained with E. faecium GJ40. Respecting the
growth rate, no significant reduction was found in this parameter in the interaction assays with A. flavus and
antagonist isolates respecting the control. While in A. parasiticus a significant reduction in growth rate was only
observed in the interaction among reference strain and E. faecium MF5 isolate (p &lt; 0.05). In general, AFB1
production was reduced by most of the LAB isolates assayed, except for E. faecium GJ18, GJ20, MF3 and MF4.
In this study a screening survey was undertaken to determine the presence and levels of aflatoxin M1 (AFM1) in
locally produced dairy products. For this purpose, a total of 138 dairy samples (milk and yogurt) were analyzed
to quantify AFM1 levels in these products. Results obtained showed that AFM1 was found in 40.62% and 32.81%
of milk and yogurt samples respectively. The range of contamination levels varied between lower and higher
than European regulation limit. Lactic acid bacteria (Lactobacillus bulgaricus and Streptococcus thermophilus)
used in the Lebanese traditional industry were screened for their ability to reduce the level of AFM1. Due to the
lack in data on the natural occurrence of AFM1 in Lebanese dairy products, the aim of this work was to report
Lactic acid bacteria (LAB) have been reported to remove mycotoxins from aqueous solutions through a binding
process, which appears to be species and strain specific. The aim of the current study was to evaluate the
protective role of Lactobacillus casei (L1) and Lactobacillus reuteri (L2) against aflatoxin (AFs)-induced oxidative
stress in rats. Sixty female Sprague-Dawley rats were divided into 6 groups including the control group and the
groups treated with L1 or L2 (1 × 10 11/ml) alone at a dose of 10 ml/kg b.w or plus AFs (3 mg/kg diet) for 4
weeks. At the end of the treatments, blood and tissue samples were collected for biochemical and histological
studies. The results indicated that AFs alone induced a significant decrease in food intake and body weight and a
significant increase in transaminase, alkaline phosphatase cholesterol, triglycerides, total lipids, creatinine, uric
acid and nitric oxide in serum and lipid peroxidation in liver and kidney accompanied with a significant decrease
in total antioxidant capacity. Treatments with L1 or L2 succeeded to induce a significant improvement in all the
Aflatoxins are commonly found in cereals worldwide and bring significant threats to the food industry and
animal production. The aim of this research was to search for Bacillus subtilis from animals' guts and soil capable
of transforming aflatoxin B1, M1, and G1 simultaneously. In addition, the selected B. subtilis can inhibit the
growth of pathogen and resist to unfavorable conditions within simulated gut environments. High-performance
liquid chromatography was used to determine the reduction in aflatoxin concentrations. B. subtilis ANSB060
from fish gut had the strongest ability to detoxify toxins, and percentages of aflatoxin B1, M1, and G1
degradation were 81.5%, 60%, and 80.7%, respectively. Moreover, the results implied that the activity of
aflatoxins degradation was mainly in the culture supernatant of ANSB060 rather than its cells or cell extracts.
And ANSB060 showed antimicrobial activities against Escherichia coli, Salmonella typhimurium, Staphylococcus
The goals of this work were to assess the ability of Lactobacillus reuteri to bind aflatoxin B 1 in the intestinal
tract and determine its effect on intestinal absorption of the toxin dispensed in either single or multiple doses in
a murine model. Male Wistar rats were used, and two experiments were conducted after bacteria were
implanted. Experiment one involved a single-oral dose of toxin, and the subsequent flow cytometric analysis of
bacteria isolated from the small intestine and treated with specific FITC-labeled AFB 1 antibodies. The second
experiment was carried out supplying the toxin in 7 oral sub-doses, and the later quantification of AFB 1-Lys
adducts in blood samples by ELISA assay. The results demonstrated that L. reuteri was able to bind AFB 1 in the
intestinal tract, mostly in the duodenum. Furthermore, the AFB 1-Lys adducts were present at significantly lower
levels in those animals receiving AFB 1 plus bacteria than in those receiving only AFB 1. Our findings confirm that
probiotic bacteria could act as biological barriers in normal intestinal conditions thereby reducing the
The aim was to evaluate both the ability of yeast strains to survive and bind AFB1 under ruminant
gastrointestinal conditions and the effect of these yeast strains on ruminal fermentation. Yeast viability was
studied under simulated gastrointestinal conditions. AFB1 binding ability was evaluated at different pH values as
present in the ruminant gastrointestinal tract. The effect of yeast strains on cellulose digestion and volatile fatty
acids production by ruminal bacteria was also evaluated. All yeast strains were able to survive under
gastrointestinal conditions and to adsorb AFB1 at the different pH assayed. The strain RC016 showed the highest
binding percentage at the three tested pH. The number of cellulolytic bacteria in ruminal fluid increased in the
presence of RC008 and RC016 yeast strains. The concentration of acetate and propionate after ruminal
fermentation increased with the addition of RC008 and RC016 strains; this effect was less significant with RC009
strain. Strains RC008 and RC016 are potential probiotic to be included in animal feed: they help to increase
fibber digestibility and could reduce AFB1bioavailability in the gastrointestinal tract. Viable S. cerevisiae RC008
Mycotoxins are secondary metabolites present worldwide in agricultural commodities and produced by
filamentous fungi that cause a toxic response (mycotoxicosis) when ingested by animals. Prevention of
mycotoxicoses includes pre- and post-harvest strategies. The best way to reduce the mycotoxin content in food
and feed is the prevention of mycotoxin formation in the field, but this is often not sufficient, so other methods
are needed. To decontaminate and/or detoxify mycotoxin-contaminated food and feed, the most prevalent
approach in the feed industry is the inclusion of sorbent materials in the feed thus obtaining more or less
selective removal of toxins by adsorption during passage through the gastrointestinal tract. Another reliable
approach is to add enzymes or microorganisms capable of detoxifying some mycotoxins. Through a
comprehensive review of published reports on the strategies for mycotoxin removal, this present work aims to
update our understanding of mycotoxin removal. It provides an insight into the detoxification of mycotoxin
present in food and feed. In the future, more emphasis needs to be placed on adsorption of mycotoxins in the
gastrointestinal tract. Concerning the enzymatic transformation of mycotoxins, further efforts are required in
Aflatoxins are a group of toxic and carcinogenic fungal metabolites. They are commonly found in cereals, nuts
and animal feeds and create a significant threat to the food industry and animal production. Several strategies
have been developed to avoid or reduce harmful effects of aflatoxins since the 1960s. However, prevention of
aflatoxin contamination pre/post harvest or during storage has not been satisfactory and control strategies such
as physical removing and chemical inactivating used in food commodities have their deficiencies, which limit
their large scale application. It is expected that progress in the control of aflatoxin contamination will depend on
the introduction of technologies for specific, efficient and environmentally sound detoxification. The utilisation
of biological detoxification agents, such as microorganisms and/or their enzymatic products to detoxify
aflatoxins in contaminated food and feed can be a choice of such technology. To date, many of the microbial
strategies have only showed reduced concentration of aflatoxins and the structure and toxicity of the detoxified
products are unclear. More attention should be paid to the detoxification reactions, the structure of
[No abstract available]
Aflatoxin M1 (AFM1) is potential human car-cinogen. Its presence in milk and dairy products represents risk for
human health. Therefore, this study was carried out in order to determine the degree of microbiological
contamination by mold, and the potential presence of aflatoxin M1 in 60 raw milk samples, randomly taken from
individual producers from different regions of the continental Croatia. The most common genera isolated fungi
were Geotrichum (78.3 %), Aspergillus (32.4 %) and Penicillium (27.0 %). From total of 60 studied milk samples,
86.66 % were positive for the presence of aflatoxin M1, and 6.66 % of samples were above the prescribed limits.
Lactic acid bacteria used in fermented dairy products as a starter culture may play a role in reduction of aflatoxin
in foods and nutrients. In this paper the ability of lactic acid bacteria: Lactobacillus rhamnosus GG (ATCC 53103),
Lactobacillus delbrueckii S1 and Lactobacillus plantarum A1 to bind aflatoxin M1 was investigated. Standard
Aflatoxins (AF) are ubiquitous in corn-based animal feed and causes hepatotoxic and hepatocarcinogenic effects.
The most important AF in terms of toxic potency and occurrence is aflatoxin B1 (AFB1). Poultry, especially
turkeys, are extremely sensitive to the toxic and carcinogenic action of AFB1, resulting in millions of dollars in
annual losses to producers due to reduced growth rate, increased susceptibility to disease, reduced egg
production and other adverse effects. The extreme sensitivity of turkeys and other poultry to AFB1 is associated
with efficient hepatic cytochrome P450-mediated bioactivation and deficient detoxification by glutathione S-
transferases (GST). Discerning the biochemical and molecular mechanisms of this extreme sensitivity of poultry
to AFB1, will contribute in the development of novel strategies to increase aflatoxin resistance. Since AFB1 is an
unavoidable contaminant of corn-based poultry feed, chemoprevention strategies aimed at reducing AFB1
Aflatoxins are a group of carcinogenic mycotoxins produced by Aspergillus flavus, A. parasiticus, and A. nomius.
Due to the ubiquitous occurrence of aflatoxins, preventive and remediative measures are necessary including
detoxification techniques. Physical and chemical decontamination strategies are inconvenient. In this study a
biological detoxification strategy was tested using bacteria of the Lactobacillus species collected from the
biotechnology laboratory at University of Ibadan, Nigeria. Maize grains with moisture content of 17% were
artificially inoculated with toxigenic A. flavus (LA 32G-28) and atoxigenic A. flavus (LA32-79) at ambient
temperature and four samples of bulk maize grains were prepared at aflatoxin B1 concentrations of 50, 100,
200, and 500ng/g. To evaluate the detoxifying potential of lactic acid bacteria five different cultures consisting of
Lactobacillus acidophilus, L. brevis, L. casei, L. delbruekii, and L. plantarum were used to inoculate the aflatoxin
B1contaminated maize samples at 37°C. After 5 days, the residual aflatoxin B1 on maize was determined. All
treatments showed significant reductions (P<0.05) in aflatoxin B1. Lactic acid bacteria decreased the pH of the
medium from 5.0 to 4.0. Pronounced aflatoxin B1 reduction was observed in maize contaminated at 50ng/g
(44.5%), while maize contaminated at 500ng/g was the least reduced (29.9%). L. plantarum was the most
The ability of traditional amahewu fermentation to increase protein digestibility and detoxify mycotoxins
commonly contaminating maize in southern Africa was investigated. Commercial maize meal, with or without a
range of added ingredients, was fermented, following the traditional way, and the levels of proteins and amino
acids assessed. Traditional amahewu samples (and the maize meal used to prepare them) were also collected
from a neighbouring rural village. Mycotoxin levels (aflatoxin B1, fumonisin B1 and zearalenone) in maize meal
and amahewu were analysed and compared in the two sets of samples. Increased levels of protein were
observed in amahewu, especially in the samples with added yeast and bread flour (up to 149%), in comparison
to the levels in starter maize. In addition, the mycotoxins detected in maize samples were drastically reduced, by
76.5-100%, following fermentation. This observation shows that traditional amahewu fermentation may
Aim of the present study was to investigate the detoxification of aflatoxin B1 and patulin from aqueous solution
by probiotic culture of Enterococcus faecium M74 and commercial culture of E. faecium EF031. The effect of the
bacterial viability, incubation time and pH of the medium on the binding ability was tested. Also, binding stability
was determined by washing the bacteria-mycotoxin complexes with phosphate buffer saline. Both M74 and
EF031 strains have the ability to remove aflatoxin B1 and patulin. While M74 removes 19.3 to 30.5% of aflatoxin
B1 and 15.8 to 41.6% of patulin, EF031 removes 23.4 to 37.5% of aflatoxin B1 and 19.5 to 45.3% of patulin
throughout a 48h incubation period. The removal of aflatoxin B1 and patulin was highest at pH 7.0 and 4.0,
respectively. The stability of the aflatoxin B1 and patulin complexes formed with the bacterial strains was found
to be high. The viability of the bacteria did not have any significant effect on the detoxification of aflatoxin B1
Aflatoxins are cancerogenic compounds produced predominantly by certain strains of the Aspergillus genus. The
ideal solution for minimization of health risk that aflatoxins pose is the prevention of foods and feeds
contamination. Unfortunately, these contaminants can never be completely removed, and on that account,
many studies have been carried out to explore an effective process of their detoxification to a threshold level.
Biological decontamination seems to be attractive because it works under mild, environmentally friendly
conditions. This review is focused on the biological detoxification of aflatoxins, especially aflatoxin B1, by
microorganisms. There are briefly mentioned aflatoxin metabolic pathways in the human and animal body.
Microorganisms such as soil or water bacteria, fungi, and protozoa and specific enzymes isolated from microbial
systems can degrade aflatoxin group members with varied efficiency to less-or nontoxic products. Some
aflatoxin-producing fungi from Aspergillus species have the capability to degrade their own synthesized
mycotoxins. Yeasts and lactic acid bacteria work as biological adsorbents that prevent aflatoxin's transfer to the
intestinal tract of humans and animals. Aflatoxin B1 absorbed into the organism could be metabolized by
Microbial fermentation of mouldy grains brought about by lactic acid bacteria is gaining much significance owing
to their ability to inhibit mould growth and detoxify mycotoxins while improving the nutritive value and safety of
the product. In the present study the potential of developing a probiotic feed ingredient from a combination of
mouldy sorghum and Cassia tora seeds, using spontaneous fermentation was explored. The effect of
fermentation at 0, 24 and 36 h on the microflora, ergosterol, mycotoxins and nutritive value, of mouldy sorghum
was assessed individually and in combination with C. tora seeds. A reduction in mould counts upto 58 and 96%
was observed at 24 and 36 h of fermenting mouldy sorghum. Total plate count increased by 2 fold and
Lactobacillus count increased by 4 fold when mouldy sorghum was fermented singly or with C. tora seeds.
Fermentation decreased ergosterol by 76%, aflatoxin to non-detectable levels at 36 h of fermentation and
fumonisin B1 to non-detectable levels at 24 and 36 h of fermentation of mouldy sorghum. Fermentation resulted
in marginal improvement in nutritive value of mouldy sorghum when estimated in terms of proximate principles
Aims: To assess the ability of five probiotic bacteria to bind aflatoxin B1 and to determine the key role of teichoic
acids in the binding mechanism. Methods and Results: The strains were incubated in aqueous solutions
containing aflatoxin B1 (AFB1). The amount of free toxin was quantified by HPLC.Stability of the bacteriaaflatoxin
complex was evaluated by repeated washes with buffer. In order to understand the binding process, protoplasts,
spheroplasts and cell wall components of two strains were analysed to assess their capacity to bind AFB1.
Additionally, the role of teichoic acids in the AFB1 binding process was assessed. Lactobacillus reuteri strain
NRRL14171 and Lactobacillus casei strain Shirota were the most efficient strains for binding AFB1.The stability of
the AFB1bacteria complex appears to be related to the binding ability of a particular strain; AFB1 binding was
also pH-dependent. Our results suggest that teichoic acids could be responsible for this ability.Conclusions: Our
results provide information concerning AFB1 binding by previously untested strains, leading to enhanced
understanding of the mechanism by which probiotic bacteria bind AFB1. Significance and Impact of the Study:
In the present study, we aimed at determining the release of aflatoxin B1 (AFB1) and ochratoxin A (OTA) from
different food products in the gastro-intestinal tract in the absence and presence of probiotics, a possible
adsorbent. The average bioaccessibility of AFB1 and OTA without probiotics was about 90%, and 30%,
respectively, depending on several factors, such as food product, contamination level, compound and type of
contamination (spiked versus naturally contaminated). The six probiotic bacteria showed varying binding
capacity to AFB1 and OTA depending on the bacterial strain, toxin studied, type of food and contamination level.
A reduction to a maximum of 37% and 73% as observed for the bioaccessibility of AFB1 and OTA in the presence
of probiotic bacteria, respectively. This is the first report on the effect of probiotic bacteria on reducing the
It has been proposed that the consumption of lactic acid bacteria capable of binding or degrading foodborne
carcinogens would reduce human exposure to these deleterious compounds. In the present study, the ability of
eight strains of Lactobacillus casei to bind aflatoxin B1 in aqueous solution was investigated. Additionally, the
effect of addition of bile salts to the growth medium on aflatoxin B1 binding was assessed. The eight strains
tested were obtained from different ecological niches (cheese, corn silage, human feces, fermented beverage).
The strains exhibited different degrees of aflatoxin binding; the strain with the highest AFB1 binding was L. casei
L30, which bound 49.2% of the available aflatoxin (4.6 μg/mL). In general, the human isolates bound the most
aflatoxin B1 and the cheese isolates the least. Stability of the bacterial-aflatoxin complex was assessed by
repeated washings. Binding was to a limited degree (0.6-9.2% release) reversible; the L. casei 7R1-aflatoxin B1
complex exhibited the greatest stability. L. casei L30, a human isolate, was the strain least sensitive to the
Some foods are prone to contamination with aflatoxins, with detrimental effect on human health. Lactic acid
bacteria have been reported to bind aflatoxins and remove them from foods and feeds. Reduction of aflatoxin B
1 (AFB1) from the liquid media by the autochthonous lactic acid bacteria (Lactobacillus casei, Lactobacillus
plantarum, and Lactobacillus fermentum) isolated from traditional Iranian sourdough and dairy products is
reported in the current study. The effect of incubation time on the binding capacity of the strains to AFB1 was
also investigated. Duplicates of individual bacteria with population equivalent to 2 × 1010 CFU/ml were
incubated in the presence of AFB1 at 37°C for a period of 72 h, and the amounts of unbound AFB1 were
quantitated by reverse-phase high-performance liquid chromatography. All the strains were capable of removal
of AFB1, and the reduction of AFB1 ranged from 25 to 61% throughout the incubation period. Removal of AFB 1
was a rapid process, with approximately 61 and 56% of the toxin taken instantly by L. fermentum and L.
plantarum, respectively. Binding was of a reversible nature, and some of the bound AFB1 was released into the
media by the repeated centrifugation and resuspension of the cell pellets. The stability of the bacteria-toxin
complex was strain dependent, and L. casei was a stronger binder of AFB1 compared with the other bacteria. No
Aflatoxins (AF) are potent carcinogens of foods, and the exposure of humans to them is continuous. Aflatoxin-
DNA adduct's nature, chemical reactions, and molecular biology are of primary importance because they are the
source of mutagenicity and risk of cancer in animals and humans. The measurement of aflatoxin-DNA adducts as
biomarkers of long-term risk of disease in people, their proper assessment and the quantification of these active
carcinogens is of great importance, because these adducts are directly related to the AF damaging effects and
can explain the origin of the cancer under study. Relatively few biomarkers of long-term health have been even
partially validated in experimental animals and in people. AF activates in the presence of the cytochrome P450,
as an unstable molecule called AF 8,9-epoxide; later this compound links mainly to the N7 of the guanine
nucleotide forming an adduct which is the active carcinogen itself and behaves as a biomarker, that is to say an
objective measure of human exposure to environmental carcinogens. Adducts represent an integration of
exposure, absorption, distribution, metabolism, DNA repair, and cell turnover, and thus provide a measure of
biologically effective dose. The different issues presented in this chapter are: 1) Factors related to AFB-DNA
adduct formation: age, cigarette smoking, alcohol drinking, organ susceptibility to form adducts (e.g., liver
parenchymal cells, lung tissue, small intestine, tracheal explant cultures, placenta, bile radical intermediates,
etc.), and temperature modulation of AFB1 hepatic metabolism; 2) In vitro and in vivo studies of AFB1-DNA
adduct formation primarily at the N7 position of guanine; 3) Formation of AFB 2 adducts; 4) Dietary AFB1
exposure, development of human primary hepatocellular carcinoma (HCC) and mutations in the p53 tumor
suppressor gene. The effect of diet in the adduct formation (choline-deficient/low methionine diet, food and
caloric restriction, and the role of enzymes); 5) Routes of exposure for DNA adduct formation; 6) Vitamins; 7)
Kinds of AF-DNA adducts (guanine, formamidopyrimidine), protein adducts (hemoglobin, albumin) and lysine,
adenosine and cytosine AFB1 adducts; 8) Methodology: a. reversed-phase high-pressure liquid chromatography
(LC), b. a liquid chromatography electrospray tandem mass spectrometry (LC- ESI-MS/MS) method, c.
polymerase chain reaction, d. radioactive methods, e. electron spin resonance (ESR) spectroscopy, f. enzyme-
linked immunosorbent assay (ELISA) with monoclonal antibodies, g. indirect immunofluorescence analysis, and
h. Ames test; 9) Effects; 10) Control, divided into sections: a) Natural repair rates of adduct removal, b) Induction
of resistance to AFB1, c) Detoxification enzymes (enzyme inhibition by beta-naphthoflavone), cytochrome P450
monooxygenases, d) Role of amino acid residues 209 and 365 of the P450 2A5 in the metabolism and toxicity of
AFB1 using recombinant yeasts. e) Pre-exposure to AFM1, f) Inhibition of AFB1 lesions by different compounds
This study assessed the binding of Aflatoxin B1 (AFB 1) from contaminated solution by Lactobacillus plantarum
PTCC 1058. This strain and AFB1 was incubated (1, 24, 48, 90 h at 37°C) and the amount of unbound AFB1 was
quantities by HPLC. The concentration of AFB1 in solution was 0.5 ppm. The stabilities of the bacteria/AFB1
complexes were evaluated by determining the amount of AFB1 remaining bound following three washes. Effect
of Incubation time on AFB1 Binding on viable and dead cells were evaluated at 1, 24, 48, 72 and 90 h time points.
In 1 h 45% and in 90 h 100% AFB1 was removed from solution by this strain. Autoclaved bacteria didn't remove
AFB1 from solutions efficiently (31% in 1 h and 15% in 24 h). Bacteria in logarithmic growth phase retained 92%
of the AFB1 initially bound after three washes. Bacterial binding of AFB1 by this strain was rapid and they were in
logarithmic growth phase. These findings further support the ability of specific strains of lactic acid bacteria to
The probiotic Lactobacillus rhamnosus GG is able to bind the potent hepatocarcinogen aflatoxin B1 (AFB1) and
thus potentially restrict its rapid absorption from the intestine. In this study we investigated the potential of GG
to reduce AFB1 availability in vitro in Caco-2 cells adapted to express cytochrome P-450 (CYP) 3A4, such that
both transport and toxicity could be assessed. Caco-2 cells were grown as confluent monolayers on
transmembrane filters for 21 days prior to all studies. AFB1 levels in culture medium were measured by high-
performance liquid chromatography. In CYP 3A4-induced monolayers, AFB1 transport from the apical to the
basolateral chamber was reduced from 11.1% ± 1.9% to 6.4% ± 2.5% (P = 0.019) and to 3.3% ± 1.8% (P = 0.002)
within the first hour in monolayers coincubated with GG (1 × 1010 and 5 × 1010 CFU/ml, respectively). GG (1 ×
1010 and 5 × 1010 CFU/ml) bound 40.1% ± 8.3% and 61.0% ± 6.0% of added AFB1 after 1 h, respectively. AFB1
caused significant reductions of 30.1% (P = 0.01), 49.4% (P = 0.004), and 64.4% (P &lt; 0.001) in transepithelial
resistance after 24, 48, and 72 h, respectively. Coincubation with 1 × 1010 CFU/ml GG after 24 h protected
against AFB1-induced reductions in transepithelial resistance at both 24 h (P = 0.002) and 48 h (P = 0.04). DNA
fragmentation was apparent in cells treated only with AFB1 cells but not in cells coincubated with either 1 ×
The ability of lactic acid bacteria (LAB) and Saccharomyces cerevisiae to remove aflatoxin B1 (AFB1) from liquid
medium was tested. The experimental results indicated that (i) AFB1 binding to microorganisms was a rapid
process (no more than 1 min); (ii) this binding involved the formation of a reversible complex between the toxin
and microorganism surface, without chemical modification of the toxin; (iii) the amount of AFB1 removed was
both toxin- and bacteria concentration-dependent; and (iv) quantitatively similar results were obtained with
viable and nonviable (heat-treated) bacteria. According to these details, a physical adsorption model is proposed
for the binding of AFB1 to LAB and S. cerevisiae, considering that the binding (adsorption) and release
(desorption) of AFB1 to and from the site on the surface of the microorganism took place (AFB, + S ↔ S - AFB1).
The model permits the estimation of two parameters: the number of binding sites per microorganism (M) and
the reaction equilibrium constant (Keq) involved, both of which are useful for estimating the adsorption
efficiency (M X Keq) of a particular microorganism. Application of the model to experimental data suggests that
different microorganisms have similar K eq values and that the differences in toxin removal efficiency are mainly
due to differences in M values. The most important application of the proposed model is the capacity to select
the most efficient microorganism to remove AFB1. Furthermore, it allows us to know if a modification of the
Mycotoxins are fungal secondary metabolites that have been associated with severe toxic effects to vertebrates
produced by many important phytopathogenic and food spoilage fungi including Aspergillus, Penicillium,
Fusarium, and Alternaria species. The contamination of foods and animal feeds with mycotoxins is a worldwide
problem. We reviewed various control strategies to prevent the growth of mycotoxigenic fungi as well as to
inhibit mycotoxin biosynthesis including pre-harvest (resistance varieties, field management and the use of
biological and chemical agents), harvest management, and post-harvest (improving of drying and storage
conditions, the use of natural and chemical agents, and irradiation) applications. While much work in this area
has been performed on the most economically important mycotoxins, aflatoxin B1 and ochratoxin A much less
information is available on other mycotoxins such as trichothecenes, fumonisin B1, zearalenone, citrinin, and
patulin. In addition, physical, chemical, and biological detoxification methods used to prevent exposure to the
toxic and carcinogenic effect of mycotoxins are discussed. Finally, dietary strategies, which are one of the most
recent approaches to counteract the mycotoxin problem with special emphasis on in vivo and in vitro efficacy of
In this study, the modulation of aflatoxin B1 (AFB1) uptake in rats by administration of the probiotic Lactobacillus
rhamnosus GG was demonstrated. Fecal AFB1 excretion in GG-treated rats was increased via bacterial AFB1
binding. Furthermore, AFB1-associated growth faltering and liver injury were alleviated with GG treatment.
Background: In vitro and in vivo studies suggest that selected strains of probiotic bacteria can form tight
complexes with aflatoxin B1 and other carcinogens. Objective: The aim of the present study was to determine
whether administration of probiotic bacteria could block the intestinal absorption of aflatoxin B1 and thereby
lead to reduced urinary excretion of aflatoxin B1-N7-guanine (AFB-N 7-guanine), a marker for a biologically
effective dose of aflatoxin exposure. Elevated urinary excretion of this aflatoxin-DNA adduct is associated with
an increased risk of liver cancer. Design: Ninety healthy young men from Guangzhou, China, were randomly
assigned to 2 groups; one group received a mixture of Lactobacillus rhamnosus LC705 and Propionibacterium
freudenreichii subsp. shermanii strains 2 times/d for 5 wk, and the other group received a placebo preparation.
The subjects provided 4 urine samples: at baseline, at 3 and 5 wk after starting the supplementation, and at the
end of the 5-wk postintervention period. Results: The percentage of samples with negative AFB-N7-guanine
values tended to be higher in the probiotic group than in the placebo group during the 5-wk intervention period
(odds ratio: 2.63, P = 0.052), and a statistically significant decrease in the concentration of urinary AFB-N7-
guanine was observed in the probiotic group. The reduction was 36% at week 3 and 55% at week 5. The
geometric means for the probiotic and placebo groups were 0.24 and 0.49 ng AFB-N7-guanine/mL, respectively,
Aflatoxin B1 (AFB1) is a well-known carcinogen and reducing its bioavailability is of great interest for human and
animal health. Several probiotic bacteria are able to bind AFB1 in vitro, including Lactobacillus rhamnosus LC-705
and Propionibacterium freudenreichii subsp. shermanii JS. A mixture of these two probiotics is used by the food
and feed industry as biopreservative (Bioprofit), making it a promising candidate for future applications.
Consequently, this study aims to investigate the in vitro and ex vivo ability of this probiotic mixture to bind AFB1.
For in vitro experiments, probiotic mixture was suspended in an AFB1 solution (5 μM), incubated for 1 to 30 min,
centrifuged, and AFB1 residues were quantitated in supernatant and pellet. For ex vivo experiments, duodenal
loops of chicks were ligated and injected with either AFB1 solution alone or probiotic mixture suspension and
AFB1 solution. Lumen content was centrifuged and AFB1 was quantitated in supernatant and pellet. Additionally,
AFB1 was extracted from duodenal tissue to calculate tissue uptake. In vitro, 57 to 66% of AFB1 was removed
from the solution by the probiotic mixture, but only 38 to 47% could be extracted from the bacterial surface. In
ex vivo experiments, only up to 25% of AFB1 was bound by bacteria, and tissue uptake of AFB1 was significantly
reduced when probiotic bacteria were present in the duodenal loop. Furthermore, the effect of intestinal mucus
on the bacterial binding ability was investigated in vitro and was found to significantly reduce AFB1 binding by
It has been estimated that 30-40 percent of all cancers can be prevented by lifestyle and dietary measures alone.
Obesity, nutrient sparse foods such as concentrated sugars and refined flour products that contribute to
impaired glucose metabolism (which leads to diabetes), low fiber intake, consumption of red meat, and
imbalance of omega 3 and omega 6 fats all contribute to excess cancer risk. Intake of flax seed, especially its
lignan fraction, and abundant portions of fruits and vegetables will lower cancer risk. Allium and cruciferous
vegetables are especially beneficial, with broccoli sprouts being the densest source of sulforophane. Protective
elements in a cancer prevention diet include selenium, folic acid, vitamin B-12, vitamin D, chlorophyll, and
antioxidants such as the carotenoids (α-carotene, β-carotene, lycopene, lutein, cryptoxanthin). Ascorbic acid has
limited benefits orally, but could be very beneficial intravenously. Supplementary use of oral digestive enzymes
and probiotics also has merit as anticancer dietary measures. When a diet is compiled according to the
guidelines here it is likely that there would be at least a 60-70 percent decrease in breast, colorectal, and
Several probiotics are known to bind aflatoxin B1 (AFB 1) to their surfaces and to adhere to intestinal mucus. In
this study, preincubation of two probiotic preparations with either AFB1 or mucus reduced the subsequent
surface binding of mucus and AFB1, respectively, in a strain-dependent manner.
The contamination of food and animal feed with AFB1 and AFM 1 is a worldwide problem. AFB1 and AFM1 are
currently of great interest because of their toxic, carcinogenic and mutagenic potential on human and animal
health. For this reason, there is a great demand for novel strategies to reduce or inactivate AFB1 and AFM 1. The
most recent approach for protecting humans against aflatoxins is the utilization of dairy strains of lactic acid
bacteria which are supposed to bind aflatoxins efficiently in the human gastrointestinal tract. For this purpose,
four dairy strains of lactic acid bacteria and four bifidobacteria, viable as well as heat-killed, were tested for their
AFM1 binding ability both in phosphate-buffered saline (PBS) and skim milk. Viable and heat-killed bacteria were
incubated with 0.1 μg ml-1 AFM 1 both in PBS and skim milk for 24 hours and the AFM1 residue in the
supernatant was measured using enzyme-linked immunosorbent assay. All viable and heat-killed strains were
able to bind AFM1 in PBS and skim milk. The binding abilities of AFM1 by viable test strains were found to range
from 22.9 % to 45.3 % and 19.3 % to 38.3 % for PBS and skim milk, respectively. Abilities of heat-treated strains
to bind AFM1 ranged from 31.3 to 61.9 % and 24.6 to 51.5 % for PBS and skim milk, respectively. Moreover, the
abilities of five strains of viable probiotic bacteria to bind AFB1 after 24 hours incubation with 1 μg ml -1 AFB1
Aflatoxin M1 (AFM1) is a highly toxic compound found in milk and milk products, and its presence in milk is a
potential threat to the health of consumers of dairy products. For this reason biological detoxification techniques
are required for the elimination of AFM1 from contaminated products. The ability of 5 probiotic bacteria, 3
Lactobacillus and 2 Bifidobacterium strains, were studied for their AFM1 binding ability. The strains were
incubated both in phosphate-buffered saline and skim milk for 24 h at 37°C and the toxin residue in the
supernatant was measured using thin-layer chromatography. Abilities of tested strains to bind AFMi were found
The series of events that led to the discovery of aflatoxin as a potent carcinogen, its biosynthesis, mechanism of
action, structure-function relationship provide interesting insight into the economical and technological factors
involved in the development of an effective control measure for the toxin. Scientists all over the world are
making continuous efforts to explore a generalized process of detoxification, which can bring down the toxin
content in heterogeneous commodities to a threshold level. In this article biological control methods with
special emphasis on in vivo and in vitro enzymatic detoxification of aflatoxin have been reviewed. Future areas
The aim of this research was to identify candidate probiotic lactic bacteria among indigenous dadih lactic
isolates. Dadih is an Indonesian traditional fermented milk of West Sumatra which is fermented naturally.
Viability of the strain is critical in determining the capacity of lactic bacteria to induce immune stimulation as
well as to colonize in the intestinal tract. Therefore, LAB are proposed to exert health promoting or probiotic
effects in human, such as inhibition of pathogenic microflora, antimutagenic, and the reduction of cholesterol
levels. This manuscript reports in vitro probiotic properties of indigenous dadih lactic bacteria, especially some
important colonization factors in GI tract, such as lysozyme, acid and bile tolerance. Bile Salt Hydrolase (BSH)
activity, spectrum of bacteriocin, and antimutagenic activity of bacterial cells were also assessed. Twenty dadih
lactic isolates were screened further for their tolerance to low pH, at pH 2 and 3 as well as their bile tolerance.
There were ten isolates classified as acid and bile acid tolerant, and further screened for lysozyme tolerance, BSH
activity. The spectrum of bacteriocin activity of isolates was assayed using cell-free neutralized supernatants by
agar spot test against variety of pathogens. Lc. lactis subsp. lactis IS-10285, IS-7386, IS-16183, IS-11857 and IS-
29862, L. brevis IS-27560, IS-26958 and IS-23427, Leu.mesen.mesenteroides IS-27526, and L. casei IS-7257 each
has good survival rate at low pH values and in the presence of lysozyme, and short lag time in the presence of
0.3 % oxgall. Lc. lactis subsp. lactis IS-11857 and IS-29862 each has high BHS activity, Lc. lactis subsp. lactis IS-
10285 and IS-16183 each had a positive spectrum of bacteriocin activity against E. coli 3301 and Lysteria
Two experiments were conducted to evaluate the efficacy of dietary FERMKIT, a commercial toxin binder
consisting of probiotic-fermented natural product containing chitin, chitosan and chitosan oligosaccharides
(FERMKITO®, EASY-BIO SYSTEM, Inc., Korea), in binding aflatoxin (AF) and zearalenone (ZEN) and ameliorating
their mycotoxicity in meat type ducks. FERMKIT was supplemented to AF contaminated diets (at 120 ppb) at
either 0.3 or 0.6% in experiment 1 and to ZEN contaminated diets (at 150 ppb) at 0.6% in experiment 2. In
experiment 1 body weight gains were reduced by 37% and mortality was increased by 18% in ducks fed diet
contaminated with AF at 120 ppb compared to ducks fed control diet (<10 ppb AF) for the 4-wk experimental
period. However, dietary FERMKIT supplementation effectively alleviated overall toxicity induced by AF. The
significant treatment-related changes in feather growth, web-toe hemorrhage, leg deformity, liver paleness,
organ weights, hematological values and serum biochemical values, as compared to the control, were observed.
The FERMKIT supplementation significantly diminished the adverse effects of AF and restored all the parameters
measured back (<0.05) toward the control values. These findings indicated that FERMKIT, when added at the
levels of 0.3 or 0.6% in the 120 ppb AF diets, could modulate the toxicity of AF with percentage sorption capacity
of 52.70% at the level 0.3% and 79.85% at the level 0.6% of the diets (experiment 1). In experiment 2, FERMKIT,
when added at 0.6% to the 150 ppb ZEN diets for the 4-wk experimental period, diminished the toxicity as
shown by body weight gain, weights of testicles, oviducts, Bursa of Fabricius and cloaca eversion score as
The ability of ten probiotic bacteria to bind a common food carcinogen aflatoxin G1,G2 and B2 was assessed. The
strains were incubated in vitro with aflatoxins and the toxin residues in the supernatant were measured using
high performance liquid chromatography. The aflatoxin G1 binding capacity of the strains was found to strain
dependent, most efficient binding of AFG1 was observed by L. acidophilus CU028 and L. brevis CU06 which
bound approximately 50%. L. acidophilus CU028 was capable of bind approximately 67% of AFG2, difference in
their binding ability showed statistical significance (p&gt;0.05). L. acidophilus CU028 and L. helveticus CU 631
were the best binders and the strains were observed to possess variable AFB2-binding ability in the range was
from 38.0% to 55.9%. Lactobacillus acidophilus CU028 was the best common binders of the three types of food
This contribution provides an overview of literature data on the microbial binding and biodegradation of
mycotoxins. These data and preliminary results from our own laboratory suggest that mycotoxin-bacterial
binding or biodegradation is a realistic process and encourages the screening of bacterial strains and their
Specific strains of lactic acid bacteria possessing antimutagenic properties are suggested to remove mutagenic
contaminants of foods through binding and an investigation of their substrate specificity is required. The ability
of Lactobacillus rhamnosus strains GG and LC-705 in viable and non-viable (heat- and acid-treated) forms to
remove both dietary mutagens and other aromatic dietary substrates from solution was studied using HPLC.
Overall, removal increased in the order: caffeine = vitamin B12 = folic acid &lt; ochratoxin A &lt; aflatoxin B1 =
PhIP (2-amino-1-methyl-6-phenyl-imidazo [4,5-b]pyridine) &lt; Trp-P-1 (3-amino-1, 4-dimethyl-5H-pyrido [4,3-
b]indole) (p &lt; 0.05). Aflatoxin B1, Trp-P-1 and PhIP were removed in high amounts (77-95%) and ochratoxin A
was removed in moderate amounts (36-76%). By contrast, only minimal amounts of caffeine, vitamin B12 and
folic acid were removed (9-28%). The significant removal of selected mutagens, but not other substrates,
suggests these strains may be useful for dietary detoxification. Since exposure to multiple mutagens is likely, the
removal of aflatoxin B1 and Trp-P-1 from a mixture of these substrates was also investigated. Removal of AFB1
significantly increased (p &lt; 0.05) in the presence of Trp-P-1, while removal of Trp-P-1 significantly decreased (p
Specific lactic acid bacterial strains remove toxins from liquid media by physical binding. The stability of the
aflatoxin B1 complexes formed with 12 bacterial strains in both viable and nonviable (heat- or acid-treated)
forms was assessed by repetitive aqueous extraction. By the fifth extraction, up to 71% of the total aflatoxin B1
remained bound. Nonviable bacteria retained the highest amount of aflatoxin B1. Lactobacillus rhamnosus strain
GG (ATCC 53103) and L. rhamnosus strain LC-705 (DSM 7061) removed aflatoxin B1 from solution most
efficiently and were selected for further study. The accessibility of bound aflatoxin B1 to an antibody in an
indirect competitive inhibition enzyme-linked immunosorbent assay suggests that surface components of these
bacteria are involved in binding. Further evidence is the recovery of around 90% of the bound aflatoxin from the
bacteria by solvent extraction. Autoclaving and sonication did not release any detectable aflatoxin B1. Variation
in temperature (4 to 37°C) and pH (2 to 10) did not have any significant effect on the amount of aflatoxin B1
released. Binding of aflatoxin B1 appears to be predominantly extracellular for viable and heat-treated bacteria.
Various food commodities including dairy products may be contaminated with aflatoxins, which, even in small
quantities, have detrimental effects on human and animal health. Several microorganisms have been reported to
bind or degrade aflatoxins in foods and feeds. This study assessed the binding of aflatoxin B1 (AFB1) from
contaminated solution by 20 strains of lactic acid bacteria and bifidobacteria. The selected strains are used in the
food industry and comprised 12 Lactobacillus, five Bifidobacterium, and three Lactococcus strains. Bacteria and
AFB1 were incubated (24 h, +37°C) and the amount of unbound AFB1 was quantitated by HPLC. Between 5.6 and
59.7% AFB1 was bound from solution by these strains. TwoLactobacillus amylovorus strains and one
Lactobacillus rhamnosus strain removed more than 50% AFB1 and were selected for further study. Bacterial
binding of AFB1 by these strains was rapid, and more than 50% AFB1 was bound throughout a 72-h incubation
The ability of six probiotic bacteria to bind a common food carcinogen, aflatoxin B1, was assessed. The studied
strains included Lactobacillus strains and one Bifidobacterium strain. The strains were incubated in vitro with
alfatoxin B1 and the toxin residue in the supernatant was measured using high-performance liquid
chromatography. The aflatoxin-binding capacity of the strains was found to range from 5.8 to 31.3%. The results
further support the observation that a number of probiotic bacteria are able to bind specific dietary
contaminants. Although the extent of binding varies depending on the bacterial strain used, the data may
The interaction of a potent carcinogen, aflatoxin B1 (AFB1), with a probiotic strain of lactic acid bacteria,
Lactobacillus rhamnosus strain GG (GG), has been investigated. The binding of AFB1 to GG in the late exponential-
early stationary phase was studied for viable, heat-killed and acid-killed bacteria. In general, viable, heat-killed
and acid-killed GG responded in a similar manner. The effects of pronase E, lipase and m- periodate on AFB1
binding and release were consistent with AFB1 binding predominantly to carbohydrate components of the
bacteria. The effect of urea suggested hydrophobic interactions play a major role in binding. Increasing
concentration (0.01-1 M) of NaCl or CaCl2 had minor effects on AFB1 binding suggesting some involvement of
electrostatic interactions. An increase in pH from 2.5 to 8.5 had no effect on AFB1 binding but decreased binding
Aflatoxins are mycotoxins that cause health and economic problems when they contaminate food and feed. One
potential method for reducing human health effects due to aflatoxin ingestion is to block uptake via binding by
bacteria that either make up the normal gut flora or are present in fermented foods in our diet. These bacteria
would bind aflatoxin and make it unavailable for absorption in the intestinal tract. Bifidobacteria comprise a
large fraction of the normal gut flora, are thought to provide many probiotic effects and are increasingly used in
fermented dairy products. These qualities targeted bifidobacteria for studies to determine if various strains of
heat-killed bifidobacteria can bind aflatoxin B1 (AFB1) in vitro. The AFB1 binding affinities of various strains of
bifidobacteria, Staphylococcus aureus, and Escherichia coli were quantitated utilizing enzyme-linked
immunosorbent and [3H]AFB1 binding assays. The bacteria analyzed were found to bind significant quantities of
Lactic acid bacteria have been previously reported to possess antimycotoxigenic activities both in vitro and in
vivo. The objective of this study was to investigate the effect of aflatoxin B1 on adhesion capability of
Lactobacillus rhamnosus strain GG using a Caco-2 adhesion model. Removal of aflatoxin B1 by L. rhamnosus
strain GG reduced the adhesion capability of this strain from 30% to 5%. It is therefore concluded that aflatoxins
may influence the adhesion properties of probiotics able to sequester them, and subsequently these bacteria
Antimutagenic activities of live and killed cells of 6 strains of Lactobacillus acidophilus and 9 strains of
bifidobacteria and of organic acids usually produced by these probiotic bacteria were determined using 8 potent
chemical mutagens and promutagens. The mutagens and promutagens used were N-methyl, N'-nitro, N-
nitrosoguanidine; 2-nitroflourene; 4-nitro-O- phenylenediamine; 4-nitroquinoline-N-oxide; Aflatoxin-B; 2-amino-
3-methyl- 3H-imidazoquinoline; 2-amino-1-methyl-6-phenyl-imidazo (4,5-b) pyridine, and 2-amino-3-methyl-9H-
pyrido (3,3-6) indole. The mutagenicity of these mutagens and antimutagenic activity of probiotic bacteria
against the mutagens were determined according to the Ames TA-100 assay using a mutant Salmonella
typhimurium. Efficiency of bacterial cells in binding or inhibiting these mutagens was also investigated. Live cells
of probiotic bacteria showed higher antimutagenic activity and their efficiency in inhibiting the mutagens was
better than killed bacterial cells. Live bacterial cells bound or inhibited the mutagens permanently, whereas
Lactobacillus rhamnosus GG and Lactobacillus rhamnosus LC-705, previously shown to effectively bind to
aflatoxin B1, were subjected to various chemical and physical treatments to examine the effects of these
treatments on the binding affinity of these strains towards aflatoxin B1. Treatment of bacterial pellets of both
strains with hydrochloric acid significantly (P &lt; 0.05) enhanced the binding ability when compared to
nontreated pellets or pellets treated by other methods. An enhancement of bacterial ability to bind aflatoxin B1
was also observed when the bacterial pellets were subjected to heat treatment by either autoclaving or boiling
at 100°C in a water bath, but the impact of these two treatments was not as effective as the acid treatment.
The aims of this investigation were to determine whether viable cultures of lactic acid-producing organisms
(LAB) can bind dietary carcinogens and to assess the consequences of binding for the absorption from the gut,
distribution in the body and in vivo genotoxicity of ingested carcinogens. The carcinogens used in this study were
ones known tobe present inthe human diet, namely benzo[a]pyrene (B(a)P), aflatoxin B1 (AFB1) and the cooked
food carcinogens 2-amino-3-methyl-3H-imidazo[4,5-f]quinoline (IQ), 2-amino-3,4-dimethylimidazo[4,5-
f]quinoline (MeIQ), 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline (MeIQx), 5-phenyl-2-amino-1-
methylimidazo[4,5-f]pyridine (PhIP) and 3-amino-1-methyl-5H-pyrido[4,3-b]indole (Trp-P-2). They represent a
range of structural types so that the specificity of any binding effects could be addressed. Of the carcinogens
tested, B(a)P and Trp-P-2 were bound most effectively by the two LAB strains Bifidobacterium longum and
Lactobacillus acidophilus. AFB1 was poorly bound, while MeIQx, MeIQ, PhIP and IQ were bound to an
intermediate degree. The extend of the binding of the heterocyclic amine carcinogens was dependent on the pH
conditions during incubation and this effect was more apparent with B. longum than with L. acidophilus. Using
the host-mediated assay (HMA), an in vivo bacterial mutation assay, it was demonstrated that the administration
of bacterial cell suspensions of B. longum and L. acidophilus did not lead to a reduction in induced mutagenicity
by MeIQ, MeIQx or Trp-P-2, detectable in the liver of treated mice compared with controls. The lack of a
protective effect could not be attributed to a short period of contact between bacterial cells and mutagens,
since similar results were obtained after preincubating bacteria and mutagens together at pH 5 for 50-60 min, to
maximize the binding, before gavaging the mice. Lack of activity of B(a)P in the HMA prevented the
determination of the effect of LAB on genotoxicity of the polycyclic aromatic hydrocarbon. However, it is clear
from the radiolabel distribution study that the amount of the carcinogen entering the blood was not significantly
reduced by B. longum administration. In addition, the amount of radiolabelled B(a)P that reached the target
Lactic acid bacteria from the fermented non-dairy food 'Idly' were examined in vitro for their ability to bind food-
borne mutagens. They were subjected to high pressures and temperatures to investigate alterations in their
binding potential. All the isolates tested exhibited good binding activity towards the amino acid pyrolysates 3-
amino-4-dimethyl-5H-pyrido[4,3-b]indole and 3-amino-1-methyl-5H-pyrido[4,3-d]indole and a moderate level of
binding of 2-amino-6-methyl-dipyrido[1,2-3′,2′-d]-imidazole. No significant binding activity was observed with
other mutagens, such as aflatoxins. High-pressure and heat treatment did not substantially alter the binding
Luchese, R.H. & Harrigan, W.F. 1990. Growth of, and aflatoxin production by Aspergillus parasiticus when in the
presence of either Lactococcus lactis or lactic acid and at different initial pH values. Journal of Applied
Bacteriology69, 512–519. Aspergillus parasiticus was grown in a modified Lab-Lemco tryptone broth both as a
single culture and in association with Lactococcus lactis. Total aflatoxin (B1 + G1) production was higher in the
mixed cultures. This stimulation persisted when different batches of media, inoculation procedures and makes of
ingredients were used. Aflatoxin yields increased in media with an initial pH of 4.2 compared with a pH close to
neutrality. Hydrochloric and/or lactic acid had little effect. The substitution of half the carbon content of the
medium by lactate resulted in stimulation or reduction on aflatoxin production when the initial pH was 4.2 or
Six sucrose esters substituted to different degrees with a mixture of fatty acids (palmitic and stearic) were
examined for antimicrobial properties. Growth and acid production of several lactic acid bacteria and growth of
Saccharomyces cerevisiae were not inhibited by 0.2% of the sucrose esters in the test medium. Antimycotic
activity was detected against several mold species from Aspergillus, Penicillium, Cladosporium, and Alternaria.
The least substituted sucrose ester was the most active in reducing mold growth. Reduction of mold growth
ranged from 37 to 91% with this ester at a 1% concentration. Inhibitory activity did not appear to be influenced
by changes in pH. Aflatoxin production by Aspergillus parasiticus was not affected by presence of 0.1% sucrose
2-s2.0-85035076143
2-s2.0-84966862738
2-s2.0-85101473506
2-s2.0-84920917506

You might also like