DataPDF Available

Cancer-preventing attributes of probiotics an update

Authors:
International Journal of Food Sciences and Nutrition,
August 2010; 61(5): 473496
Cancer-preventing attributes of probiotics: an update
MANOJ KUMAR
1
, ASHOK KUMAR
2
, RAVINDER NAGPAL
3
, DHEERAJ
MOHANIA
4
, PRADIP BEHARE
1
, VINOD VERMA
5
, PRAMOD KUMAR
1
,
DEV PODDAR
1
, P. K. AGGARWAL
1
, C. J. K. HENRY
6
, SHALINI JAIN
7
&
HARIOM YADAV
7
1
Dairy Microbiology Division, National Dairy Research Institute, Karnal (Haryana), India,
2
Department of Zoology, M.L.K. P.G. College, Balrampur (U.P.), India,
3
Department of
Biotechnology, Lovely Professional University, Phagwara, Punjab, India,
4
Animal Biochemistry
Division, National Dairy Research Institute, Karnal (Haryana), India,
5
AgResearch Ltd,
Ruakura Research Centre, Reproductive Technologies, East Street, Hamilton, New Zealand,
6
School of Biological and Molecular Sciences, Oxford Brookes University, Oxford, UK, and
7
National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health,
Bethesda, Maryland, USA
Abstract
Cancer is a serious global public health problem. Cancer incidence and mortality have been
steadily rising throughout the past century in most places of the world. There are several
epidemiological evidences that support a protective role of probiotics against cancer. Lactic acid
bacteria and their probioactive cellular substances exert many benecial effects in the gastro-
intestinal tract, and also release various enzymes into the intestinal lumen and exert potential
synergistic (LAB) effects on digestion and alleviate symptoms of intestinal malabsorption.
Consumption of fermented dairy products with LAB may elicit anti-tumor effects. These effects
are attributed to the inhibition of mutagenic activity, the decrease in several enzymes implicated
in the generation of carcinogens, mutagens, or tumor-promoting agents, suppression of tumors,
and epidemiology correlating dietary regimes and cancer. Specic cellular components in lactic
acid bacteria seem to induce strong adjuvant effects including modulation of cell-mediated
immune responses, activation of the reticulo-endothelial system, augmentation of cytokine
pathways, and regulation of interleukins and tumor necrosis factors. Studies on the effect of
probiotic consumption on cancer appear promising, since recent in vitro and in vivo studies have
indicated that probiotic bacteria might reduce the risk, incidence and number of tumors of the
colon, liver and bladder. The protective effect against cancer development may be ascribed to
binding of mutagens by intestinal bacteria, may suppress the growth of bacteria that convert
procarcinogens into carcinogens, thereby reducing the amount of carcinogens in the intestine,
reduction of the enzymes b-glucuronidase and b-glucosidase and deconjugation of bile acids, or
merely by enhancing the immune system of the host. There are isolated reports citing that
administration of LAB results in increased activity of anti-oxidative enzymes or by modulating
circulatory oxidative stress that protects cells against carcinogen-induced damage. These
include glutathione-S-transferase, glutathione, glutathione reductase, glutathione peroxidase,
superoxide dismutase and catalase. However, there is no direct experimental evidence for
cancer suppression in human subjects as a result of the consumption of probiotic cultures in
Correspondence: Dr Hariom Yadav, National Institute of Diabetes and Digestive and Kidney Diseases, Diabetes Branch,
5W5872, CRC, Bld 10, Bethesda, MD 20892, USA. E-mail: yadavhariom@gmail.com
ISSN 0963-7486 print/ISSN 1465-3478 online 2010 Informa UK Ltd
DOI: 10.3109/09637480903455971
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
fermented or unfermented dairy products, but there is a wealth of indirect evidence based largely
on laboratory studies.
Keywords: Probiotics, cancer, prebiotics, synbiotics, functional food
Introduction
Probiotics are dened as live microbial food ingredients (supplements) that confer
benecial health effects to the host upon ingestion in adequate amounts (Salminen and
von Wright 1998). The concept of probiotics evolved at the turn of the twentieth
century from a hypothesis rst proposed by Nobel Prize-winning Russian scientist Elie
Metchnikoff, who propounded that the long and healthy life of Bulgarian people
resulted from their consumption of fermented milk products (Bibel 1998). He believed
that, when consumed, the fermenting bacillus (Lactobacillus) positively inuenced the
microbiota of the colon and, decreased their toxic activities. Several lactic acid bacteria
may help prevent initiation of cancer (Table I). It appears that LAB can reduce the
levels of colon enzymes that convert pro-carcinogens to carcinogens. Specically, LAB
can reduce levels of the enzymes b-glucuronidase, nitro-reductase, and azo-reductase.
LAB may also be involved in the direct reduction of pro-carcinogens; for example, by
taking up nitrites and by reducing the levels of secondary bile salts (Fernandes and
Shahani 1990). Changes in enzyme activity in humans have been observed with
Lactobacillus acidophilus and Bidobacterium bidum (Marteau et al. 1990), and
Lactobacillus rhamnosus GG (LGG) (Goldin and Gorbach 1984). Animal studies
showed fewer tumors in those exposed to a carcinogen in the presence of LGG
compared with the animals exposed to the carcinogen without the benet of LGG
(Goldin 1996). Human epidemiological reports have indicated that populations
ingesting fermented dairy products have a decreased risk of colon cancer (Kampman
et al. 1994). An anti-tumor effect has been reported by oral intake of LAB in in vitro
studies but, since colon carcinogenesis is a multistage process, the mechanisms if
validated remain to be elucidated (Hirayama and Rafter 1999). A preventive effect on
malignancy development could be mediated by different mechanisms involved in
detoxication of carcinogens.
In vitro studies have shown that, in general, live cells of probiotic bacteria possessed
higher anti-mutagenic activity. Intake of fermented milk by (L. acidophilus and
B. bidum) inuenced gut ora enzymes, like b-glucuronidase, b-glucosidase and
nitro-reductase (Wollowski et al. 2001). L. acidophilus and B. bidum decreased the
activity of nitro-reductase and b-glucuronidase but increased that of b-glucosidase
(Marteau et al. 1990). This could be an advantage since b-glucosidase may release
avonoids that have anti-mutagenic, anti-oxidative and immune-stimulatory effects
(Stoner and Mukhtar 1995). Most animal and human studies do indicate that feeding
certain LAB decreases fecal enzyme levels that may be involved in formation of
carcinogens. Also, benecial effects can be attributed to immune-potentiating effects
by LAB strains. One specic effect was shown by heat-killed Lactobacillus plantarum
L-137, which restored the inhibited IL-12 production in DBA/2 mice with tumors
(Murosaki et al. 2000). The anti-tumor effects were found to be due to the activation of
macrophage by LC9018 (Okawa et al. 1993). Conjugated linoleic acid has been
highlighted recently because of its effect to reduce carcinogenesis, atherosclerosis
and body fats (Chin et al. 1992). Mycotoxins are well-known contaminants of grains
and other food raw materials in which Ochratoxin A is carcinogenic, genotoxic,
474 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
immunosuppressive and nephrotoxic (Petzinger and Ziegler 2000). In some reports,
strains of Lactobacillus and Lactococcus were shown to be resistant to ochratoxin
A(510 microgram/disc), and just one strain of L. helveticus was sensitive (0.1
microgram/disc) (Piotrowska and Zakowska 2005). Furthermore, all strains tested
reduced the amount of ochratoxin A in the growth medium as measured after 120 h. L.
acidophilus CH-5, LGG and a couple of other strains reduced the initial level of
ochratoxin A by more than 50%. Intake of such LAB may potentially reduce carci-
nogenic/genotoxic effects of consumption of food contaminated by ochratoxin A.
Aatoxin B
1
(AFB1), a mycotoxin produced by the common fungi Aspergillus avus
and Aspergillus parasiticus, is an established human hepato-carcinogen (Vamio et al.
1993). Various strains of LAB isolated from either dairy products or healthy human
microbiota showed that two probiotic L. rhamnosus strains, LGG and LC705, were
found to be highly efcient in binding a range of mycotoxins, including AFB1. Earlier,
a single viable bacterium was found to bind >10
7
AFB1 molecules (Haskard et al.
2000).
Protective and prevention of cancer occurrence
Liver cancer is the sixth most commonly diagnosed cancer in the world, and the third
most common cause of death from cancer, according to Cancer Research UK. Despite
Table I. Potential probiotic bacteria having cancer-preventing properties.
Probiotic bacteria References
B. polyfermenticus Jeong et al. (2003), Park et al. (2004, 2007)
B. longum ATCC 15708 Challa et al. (1997), Vanderhoof (2001), Biasco et al. (1991), Pool-Zobel
et al. (1996), Rowland et al. (1998), Reddy and Rivenson (1993), Singh
et al. (1997), Reddy (1998), Haskard et al. (2000), Lin and Chang (2000)
Lactobacillus GG Goldin et al. (1996), Drisko et al. (2003), Femia et al. (2002), Caro et al.
(2005), Ahotupa et al. (1996), Fang and Polk (2002), Lam et al. (2007)
L. casei (LC9018) Aso et al. (1995), Lidbeck et al. (2000), Okawa et al. (1993)
L. casei Shirota Matsumoto et al. (2009)
L. acidophilus Lidbeck et al. (1992), Bifet al. (1997),Vanderhoof (2001), Biasco et al.
(1991), Pool-Zobel et al. (1996), Arimochi et al. (1997), Goldin et al.
(1980), Haskard et al. (2000)
L. gasseri Vanderhoof (2001), Pool-Zobel et al. (1996)
L. confusus, Vanderhoof (2001), Pool-Zobel et al. (1996)
S. thermophilus Vanderhoof (2001), Pool-Zobel et al. (1996)
B. breve ATCC 15700 Vanderhoof (2001), Pool-Zobel et al. (1996), Onoue et al.
(1997), Chalova et al. (2008)
L. delbrueckeii Rafter (1995)
B. infantis Bifet al. (1997)
B. lactis Femia et al. (2002), Sekine et al. (1995)
L. plantarum L-137,
L. plantarum JCM 1551,
L. plantarum KLAB21,
L. plantarum 2592
Murosaki et al. (2000), Ando et al. (2004), Rhee and Park,
(2001), Kruszewska et al. (2002), Khanafari et al. (2007)
P. pentosaceus 16:1 Kruszewska et al. (2002)
L. paracasei F19 Kruszewska et al. (2002)
L. reuteri Iyer et al. (2008)
B. adoleascentis ATCC 15703 Chalova et al. (2008)
VSL#3 Otte et al. (2009)
Cancer-preventing attributes of probiotics 475
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
these gures, the cancer remains relatively rare, with 18,500 new cases in the USA
every year, and about 3,000 in the UK. The highest incidences of the disease are in east
and southeast Asia, particularly China, and for this reason the current researchers
looked at the effects of probiotic supplements on markers for the disease. The biggest
risk factor for the disease is said to be chronic hepatitis B virus infection, but
consumption of foods (contaminated with aatoxins) are also established causes of
liver cancer.
AFB1 is classied as a group I carcinogen in humans by International Agency for
Research on Cancer. However, the exact mechanisms of AFB1 hepatocarcinogenesis
have not been fully elucidated. But some studies have suggested that oncogenes are
critical molecular targets for AFB1, and AFB1 causes characteristic genetic changes in
the p53 tumor suppressor gene and ras proto-oncogenes. The hepatic carcinogen
AFB1 is metabolized in the liver by at least four different P450s, all of which exhibit
large inter-individual differences in the expression levels. These differences could affect
the individual risk of hepatocellular carcinoma. Some researchers provided additional
evidence that reactive oxygen species (ROS) and oxidative DNA damage may be
involved in AFB1-induced p53 and ras mutations. AFB1glutathione (GSH) conju-
gation is the major pathway for the detoxication of aatoxin metabolites (Figure 1).
This reaction is catalyzed by glutathione-S-transferase (GST) and plays a major role in
modulation of AFB1 adduct formation to nuclear DNA. Changes recorded in hepatic
GST activity during development of rats can alter the balance between AFB1GSH
conjugation and AFB1DNA adduct formation. Some probiotic bacterial strains
successfully bind mycotoxins; L. rhamnosus binds away AFB1 in vivo, thus reducing
bio-absorption of the toxin from the gut, while L. acidophilus and Bidobacterium longum
neutralize AFB1 and AFM1 by a similar binding mechanism (Haskard et al. 2000).
Epidemiological studies in Africa and Asia (where high levels of exposure occur) link
AFB1 exposure and hepato-cellular carcinomas (Bulatao-Jayme et al. 1982; Bosch and
Munoz 1988), and the risk is synergistic with hepatitis viral infections. The mechanism
of action for AFB1 mutagenicity begins with metabolic activation by CYP3A4,
CYP3A5 and/or CYP1A2 (Garner et al. 1972; Reynolds et al. 1987) that forms an
exo-8,9-epoxide and subsequent adduct formation and DNA damage (Essigmann et al.
Aspergillus spp.
Aflatoxin B1
Aflatoxin-exo8,9-epoxide
CYP 1A2
CYP 3A4
DNA
Alatoxin-N7-Guanine
Genotoxicity
Aflatoxin-S-G
AFB1-mercapturate
Aflatoxin-dihydrodiol
Aflatoxin dialdehyde
Aflatoxin-Albumin
Inhibition-Probiotic
by binding mechanism
Detoxification by antioxidants
produced by probiotics
Figure 1. Mechanism of action of probiotics during liver cancer progression induced by AFB1.
476 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
1977; Iyer et al. 1994). This damage has been shown in vitro to cause guanine
nucleotide substitutions (Lilleberg et al. 1992) specically to codon 249 of the p53
gene (Aguilar et al. 1993). Mutations in specic oncogenes and tumor-suppressor
genes (e.g. K-ras, p53) are involved in tumor development in the colon and liver, as
they occur at high frequencies in human colon and liver cancers (Erdman et al. 1997).
The expression levels of these genes can be measured and used to demonstrate the
effect of various dietary components on tumor formation in colonic mucosa.
Addition of the probiotic B. longum to the diet of rats was shown to exert a strong
anti-tumor activity on colonic mucosa by reducing the expression level of ras-p21
expression and cell proliferation (Singh et al. 1997; Reddy 1998). LGG administration
determined the upregulation and downregulation of 334 and 92 genes, respectively,
shown by microarray analysis, and further real-time polymerase chain reaction con-
rmed the reliability of the analysisit mainly affects the expression of genes involved
in immune response and inammation (transforming growth factor-beta and TNF
family members, cytokines, nitric oxide synthase 1, defensin alpha 1), apoptosis, cell
growth and cell differentiation (cyclins and caspases, oncogenes), cellcell signaling
(intracellular adhesion molecules), cell adhesion (cadherins), signal transcription and
transduction (Caro et al. 2005). Colorectal cancer represents a major public health
problem accounting for over 1 million cases and about 0.5 million deaths worldwide
(Chau and Cunningham 2006). Survival from colon cancer at 5 years has been found to
vary demographically and estimated to be 65% in North America, 54% in Western
Europe, 34% in Eastern Europe and 30% in India (Parkin et al. 2005). Dietary
interventions and natural bioactive supplements have now been extensively studied to
reduce the risks of colon cancer as a cause of prevention instead of cure. Postulated
mechanisms include reduction in the activity of several cancer-causing bacteria,
des-mutagenic and anti-carcinogenic properties (Collins and Gibson 1999; Chau
and Cunningham 2006). Probiotics have been found by several researchers to decrease
fecal concentrations of enzymes and secondary bile salts, and to reduce absorption of
harmful mutagens that may contribute to colon carcinogenesis (Rafter 1995). Other
studies suggest that normal intestinal ora can inuence carcinogenesis by producing
enzymes (glycosidase, B-glucuronidase, azoreductase, and nitroreductase) that
transform precarcinogens into active carcinogens (Goldin et al. 1980; Goldin
1990; Marteau et al. 1990; Ling et al. 1994; Pedrosa et al. 1995). Certain probiotics
may protect the host from this activity. L. acidophilus and Lactobacillus casei supple-
mentation in humans helped to decrease levels of these enzymes, as shown by fecal
specimens (Hayatsu and Hayatsu 1993; Lee and Salminen 1995; Lidbeck et al. 1992).
In animal studies, the aforementioned bacterial enzymes have been suppressed with
the administration of LGG (Drisko et al. 2003). Other LAB have shown similar
results; however, the relationship between enzyme activity and cancer risk needs
further investigation.
Several mechanisms have been proposed as to how LAB may inhibit colon cancer,
including enhancing the hosts immune response, altering the metabolic activity of the
intestinal microbiota, binding and degrading carcinogens, producing antimutagenic
compounds, and altering the physiochemical conditions in the colon (Hirayama and
Rafter 2000) (Figure 2). During carcinogenesis in the colon, aberrant crypts can be
recognized as early neoplastic lesions in both rodents and humans (Bird 1995).
A number of natural chemopreventive agents or medicinal plants that inhibit the
development of aberrant crypt foci (ACF) have been demonstrated to prevent colon
Cancer-preventing attributes of probiotics 477
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
cancer in rodents (Finley et al. 2000; Ohishi et al. 2002), thereby suggesting that ACF
assays in the rodent colon can be employed as good biomarkers in colon carcinogen-
esis. Colon carcinogenesis is also known to be a pathological consequence of persistent
oxidative stress, resulting in DNA damage and mutations in cancer-associated genes,
cycle arrest or cell death, and mutations and regeneration in which the cellular
overproduction of reactive oxygen molecules and reactive nitrogen species have
been implicated (Payne et al. 1999; Bartsch and Nair 2002; Korenaga et al.
2002; Nair et al. 2002) In an in vitro experiment, Bacillus polyfermenticus exerted
antioxidative, antigenotoxic, and anticarcinogenic effects (Jeong et al. 2003; Park et al.
2004). Consumption of large quantities of dairy products such as yoghurt and
fermented milk containing Lactobacillus or Bidobacterium may be related to a lower
incidence of colon cancer (Shahani and Ayebo 1980). Oral administration of LAB has
been shown to effectively reduce DNA damage, induced by chemical carcinogens, in
gastric and colonic mucosa in rats. By the comet assay, L. acidophilus,Lactobacillus
gasseri,Lactobacillus confusus,Streptococcus thermophilus,Bidobacterium breve and B.
longum were antigenotoxic toward N¢-nitro-N-nitrosoguanidine (MNNG) (Pool-Zobel
et al. 1996). These bacteria were also protective toward 1,2-dimethylhydrazine
(DMH)-induced genotoxicity. Metabolically active L. acidophilus cells, as well as an
acetone extract of the culture, prevented MNNG-induced DNA damage, while heat-
treated L. acidophilus was not antigenotoxic. Goldin et al. (1996) showed that a specic
strain of L. casei subsp. rhamnosus designated GG can interfere with the initiation or
early promotional stages of DMH-induced intestinal tumor genesis and that this
effect is most pronounced for animals fed a high-fat diet. Feeding of L. acidophilus
and B. longum suppressed the formation of ACF and tumor incidence, induced by
azoxymethane (AOM) (Kulkarni and Reddy 1994; Arimochi et al. 1997; Singh et al.
1997; Challa et al. 1997; Rowland et al. 1998) or DMH (Abdelali et al.
Steps leading to cancer
DMH (Procarcinogen)
Carcinogen (Azoxymethanemethyldiazoniumion)
Reactions with cellular targets (colonocytes)
Carcinogen-DNA adduct
Neoplastic manifestation
Cancer (Adenocarcinoma)
Probiotics by
binding mechanism
Probiotics by
antioxidants
Inhibition and detoxification
Figure 2. Mechanism of action of probiotics during colon cancer progression.
478 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
1995; Gallaher et al. 1996). In addition, it has been reported that colonization of
bacteria with an ability to produce genotoxic compounds and high b-glucuronidase
activity enhanced progression of ACF induced by DMH in rats, and that the additional
colonization of B. breve reduced the number of ACF with four or more crypts/focus and
crypt multiplicity that are reliable predictors of malignancy (Onoue et al. 1997). AOM-
induced colon tumor development was also suppressed with a decrease in colonic
mucosal cell proliferation and tumor ornithine decarboylase and ras-p21 activities
(Hirayama and Rafter 2000). There was a report on the anti-tumorigenic activity of
the prebiotic inulin, enriched with oligofructose, in combination with the probiotics
L. rhamnosus and Bidobacterium lactis in the AOM-induced colon carcinogenesis rat
model (Femia et al. 2002). Reddy and Rivenson (1993) reported that lyophilized
cultures of B. longum administered in the diet to rats inhibited liver, colon and
mammary tumors, induced by the food mutagen 2-amino-3-methyl-3H-imidazo
(4,5-f) quinoline (IQ). Goldin and Gorbach (1980) showed that dietary supplements
of L. acidophilus not only suppressed the incidence of DMH-induced colon carcino-
genesis but also increased the latency period in rats. Feeding of fermented milk
increased the survival rate of rats with chemically induced colon cancer (Shackelford
et al. 1983). B. polyfermenticus was also determined to protect against the DNA damage
induced by MNNG, a direct-acting carcinogen with alkylating properties in CHO-K1
cells and in human lymphocytes, and was also determined to inhibit the growth of
Caco-2 human colon cancer cells in a dose-dependent manner, as shown by the results
of an MTT assay. These in vitro results of B. polyfermenticus were conrmed in the
present in vivo animal study. Our current results indicate clearly that the dietary feeding
of B. polyfermenticus (3.1 x 10
8
cfu/day) effectively suppresses the occurrence of colonic
ACF induced by DMH when administered 1 week prior to treatment with the
carcinogen (Park et al. 2007). Using AOM-induced aberrant crypt foci in rats,
Reddy et al. (1997) found that a stimulated growth of bidobacteria in the colon
could lead to the inhibition of colon carcinogenesis. Reduction in GST activity is
associated with an increased risk for Colorectal Cancer (CRC) (Szarka et al. 1995).
The use of lactulose separately or in conjunction with B. longum has been demonstrated
to signicantly increase GST levels in rat colon (Challa et al. 1997). These results led to
the suggestion that probiotics could suppress colon cancer.
There are 57 cytochrome P450s encoded in the human genome, mainly catalyzing
the metabolism of steroids, bile acids, eicosanoids, drugs and xenobiotic chemicals.
However, some of the P450s are also active carcinogens. Past epidemiological research
has shown increased risk of colon cancer in individuals with high P450-1A2 activity.
The metabolic activation of food-borne heterocyclic amines to colon carcinogens in
humans is hypothesized to occur via N-oxidation followed by O-acetylation to form the
N-acetoxy arylamine that binds to DNA to give carcinogen-DNA adducts. These steps
are catalyzed by hepatic cytochrome P450-1A2 and N-acetyltransferase-2 (NAT-2),
respectively (Lang et al. 1994) (Table II). It has been postulated that probiotics such as
Bidobacterium could lower the risks of colon cancer, by producing metabolites that
could affect the mixed-function of P450s and subsequently affect the conversion of
azoxymethane from proximate to ultimate carcinogen (Campbell and Hayes 1976).
GST is an important family of phase II detoxication enzymes, which play a crucial role
in protecting the colon mucosa from dietary carcinogens.
Burns and Rowland (2000) suggested that increasing the amount of LAB in the
colon decreases the ability of microbiota to produce carcinogens. A randomized,
Cancer-preventing attributes of probiotics 479
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
Table II. Examples of carcinogens, their biomarkers and potential probiotics that reduce their effects.
Target
organ
Biomarkers
Carcinogen Probiotics References Genetic susceptibility Phenotypic biomarker
Activating enzyme with
genetic
polymorphisms
Detoxifying enzyme
with genetic
polymorphisms
Adduct Gene
mutation
AFB1 Liver L. rhamnosus,
L. acidophilus,
B. longum
Haskard et al. (2000) CYP3A4
a
, CYP3A5,
CYP1A2
Glutathione
transferases,
glucuronyl
transferases
Adducts in DNA,
albumin, hemogloblin
and urinary metabolites
p
53
mutation
at codon 249
AGG to AGT
IQ Colon,
breast
B. longum,
S. thermophilus,
B. animalis
Reddy and Rivenson
(1993), Tavan et al. (2002)
CYP1A2, NAT1
b
GST, GPx, NAT2 N-(deoxyguanosin-
8-yl)-2-amino-3-
methylimidazo-[4,5-f]
quinoline (dG-C8-IQ)
k-ras
MEIQ Colon,
breast
S. thermophilus,
B. animalis
Reddy and Rivenson
(1993), Tavan et al. (2002)
CYP1A2, NAT1
b
GST, GPx, NAT2, glucuronyltransferase C-8 guanine
adducts
Kras
PhIP Colon,
breast
S. thermophilus,
B. animalis
Reddy and Rivenson
(1993), Tavan et al. (2002)
CYP1A2, NAT1
b
GST, GPx, NAT2 DNA adduct and
adducts in albumin
C-8 guanine adducts
P53, K-ras
Trp-P-2 Liver,
colon
B. longum Reddy and Rivenson (1993) CYP1A2, CYP1A1, and
CYP2B
GST DNA adduct Kras
DMH Colon L. acidophilus,
L. casei sub sp.
rhamnosus,
B. breve
Goldin et al. (1980,
1996), Onoue et al. (1997)
CYP1A1, CYP2E1 GST, glucuronyl
transferases
O6-Methylguanine
(O6-MeGua) adduct
Kras, c-myc
AOM Colon L. acidophilus,
B. longum
Arimochi et al. (1997),
Kulkarni et al. (1994),
Challa et al. (1997),
Rowland et al. (1998)
CYP1A1, CYP2E1 GST, glucuronyl
transferases
O6-Methylguanine
(O6-MeGua) adduct
Kras, c-myc
MNNG Colon L. acidophilus,
L. gasseri,
L. confusus,S.
Pool et al. (1996),
Park et al. (2007),
Caldini et al. (2005)
CYP1A2, CYP1A3 GST N7-Methyl
deoxyguanosine
Kras
480 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
Table II (Continued)
Target
organ
Biomarkers
Carcinogen Probiotics References Genetic susceptibility Phenotypic biomarker
thermophilus,
B. breve,
B. longum,
B. polyfermenticus
3¢-monophosphate
(N7-MedGp)
DMBA Mammary
cancer,
liver
cancer
L. bulgaricus
191 R
Nadathur et al. (1995) CYP1A1 and CYP1B1 GST DNA adduct K-ras
4NQO Tongue
cancer
L. bulgaricus
191 R, B. longum
(ATCC 15708),
L. acidophilus
(ATCC 4356)
Nadathur et al. (1995), Lin
and Chang, (2000)
CYP1A1 GST, GPx DNA adduct H-RAS
DMAB Colon,
prostate,
pancreas,
urinary
bladder
cancer
L. bulgaricus
191 R
Nadathur et al. (1995) CYP1A2, NAT1
b
GST DNA adduct K-ras
MC Lung
cancer,
liver
cancer,
mammary
cancer
L. casei strain
shirota
Takagi et al. (1999) CYP1A1, CYP1A2 GST-a, UDP,
glucuronyl
transferase, quinone
oxido-reductase I
(NQO1), aldehyde
dehydrogenase
(ALDH)
DNA adduct K-ras
BaP Lung
cancer
B. adoleascentis
ATCC 15703,
B. breve
Chalova et al. (2008) CYP1A1, CYP1B1 GST, glucuronyl
transferase
Benzo[a]pyrene diol
epoxide
Kras, p53
a
CYP, cytochrome P450.
b
NAT, N-acetyltransferase.
Cancer-preventing attributes of probiotics 481
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
controlled study by Aso and Akazan (1992) demonstrated that the recurrence of
bladder tumors was delayed with daily intake of L. casei. They performed another study
that was larger (125 patients) and placebo-controlled, and found that L. casei reduced
the recurrence of tumors in all patients except those with more than one recurrent
tumor (Aso et al. 1995). In an animal model with DMH-induced colon cancer, it was
shown that LGG signicantly reduced the incidence of colon tumors (Goldin et al.
1996). LAB administered to animals have been shown to prevent carcinogen-induced
preneoplastic tumors and lesions (Wollowski et al. 2001) and also reduced the growth
and viability of the HT-29 human colon cancer line (Hirayama and Rafter 2000). L.
gasseri,L. confusus,S. thermophilus,B. breve,B. longum, and L. acidophilus strains
showed an antigenotoxic effect after MNNG administration (Vanderhoof 2001), and
L. acidophilus,L. confusus,S. thermophilus,L. delbrueckeii,L. gasseri,B. longum, and B.
breve inhibited DNA damage from DMH (Rafter 1995).
Supplementation of the chow with 0.5% lyophilized B. longum caused pronounced
inhibition of the incidence of 3-amino-1-methyl-5H-pyrido[4,3-6]indole (Trp-P-2)-
induced liver and colon tumors (i.e. 80% and 100% reduction in tumor frequencies,
respectively) (Reddy and Rivenson 1993); however, the concentration of tryptophan
pyrolysates in meats are much lower than those of other heterocyclic amines (HCAs)
(Felton et al. 2000). Fermented milks prepared by S. thermophilus or Bidobacterium
animalis inhibited the induction of preneoplastic lesions (ACF in the colon) and
DNA migration caused by a HCA mixture containing IQ, 2-amino-3,4-dimethyl-
3H-imidazo[4,5-f]quinoline (MeIQ) and 2-amino-1-methyl-6-phenylimidazo[4,5-b]
pyridine (PhIP) (ratio 1:1:1) (Tavan et al. 2002). Anti-mutagenicity of an acetone
extract of a L. bulgaricus 191R fermented yoghurt observed a signicant dose-depen-
dent anti-mutagenic activity against several mutagens including MNNG,
4-nitroquinoline-N-oxide, 3,2-dimethyl-4-aminobiphenyl, 9,10-dimethyl-1,2-benz[a]
anthracene and Trp-P2. This study suggests that a metabolite of the LAB may be
responsible for their anti-carcinogenic properties (Nadathur et al. 1995). Bidobacter-
ium infantis and L. acidophilus bacteria strains inhibited the growth of the MCF7 breast
cancer cell line (Bifet al. 1997). Studies have suggested that the hosts immune
response may be stimulated by B. infantis, leading to tumor suppression or regression
(Hirayama and Rafter 2000). The metabolic activity of the intestinal microbiota may
also be altered with administration of LAB (Goldin and Gorbach 1984). The alteration
of the physicochemical conditions in the colon may inuence colon cancer (Modler
et al. 1990), and suggest that reducing the intestinal pH may prevent the growth of
putrefactive bacteria. In a 3-month study, L. acidophilus and B. bidum were admin-
istered to patients with colonic adenomas. The result was a decrease in fecal pH and
cell proliferative activity in the upper colon (Biasco et al. 1991). The mechanisms of the
links of probiotics to anti-tumor activity are not completely clear, but offer useful
potential material for future cancer studies.
Possible modes of actions of probiotics: as evidenced by various probiotic
strains
Reduction of reactive oxygen species load
Oxidative stress results when the balance between the production of ROS overrides the
antioxidant capability of the target cell; oxidative damage from the interaction of
482 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
reactive oxygen with critical cellular macromolecules may occur. ROS may interact
with and modify cellular protein, lipid, and DNA, which results in altered target cell
function. The accumulation of oxidative damage has been implicated in both acute and
chronic cell injury including possible participation in the formation of cancer. Oxidative
stress occurs in a cell or tissue when the concentration of ROS generated exceeds the
antioxidant capability of that cell (Sies 1991). ROS can be produced both endoge-
nously and exogenously (Figure 3). Endogenous oxidative stress can be the result of
normal cellular metabolism and oxidative phosphorylation. The metabolism of sub-
stances by the P450 enzyme system generates oxygen free radicals through normal or
futile cycling mechanisms (Parke and Ioannides. 1990). Exogenous sources of ROS
can also impact on the overall oxidative status of a cell. Drugs, hormones, and other
xenobiotic chemicals can produce ROS by either direct or indirect mechanisms (Trush
and Kensler 1991; Halliwell 1996). Alternatively, oxidative stress can also occur when
there is a decrease in the antioxidant capacity of a cell. Non-enzymatic antioxidant
levels (vitamin E, vitamin C, GSH, etc.) and enzymatic antioxidant levels (superoxide
dismutase, GSH peroxidase, and catalase) in the cell can be decreased through
modication in gene expression, decreased in their uptake in the diet, or can be
overloaded in ROS production, which creates a net increase in the amount of oxygen
free radicals present in the cell (Vuillaume 1987; Barber and Harris 1994). Several
human chronic disease states, including cancer, have been associated with oxidative
stress produced through either an increased free radical generation and/or a decreased
antioxidant level in the target cells and tissues (Trush and Kensler 1991; Rice-Evans and
Burdon 1993). Also it is well known that LGG and LGG-fermented milk were potent
scavengers of superoxide anion and inhibitors of lipid peroxidation reactions in vitro
(Ahotupa et al. 1996). Glutathione peroxidase (GPx) and glutathione-S-transferase
Endogenous source of
ROS
mitochondria
cytochrome P450
peroxisomes
inflammatary cells
Exogenous source of
ROS
radiation
ozone
hyperoxia
xenobiotics
Protein damage
(DNA repair
proteins,
Caspases)
DNA damage and
mutation
8-oxo-dG
nitrosamines
8-nitroguanine
S-nitrosothiol
DNA-strand breako
Lipid
peroxidation
4HNE
MDA
Arachidonic
acid cascade
Eicosanoids
Cell
proliferation
Probiotic
bacteria
Source
Enzymatic
(SOD, CAT, GSH
Peroxidase)
Nonenzymatic
(Vit E, GSH, Vit C)
Cell membrane
Figure 3. Effect of probiotics on ROS production that are produced by endogenous and exogenous sources.
Cancer-preventing attributes of probiotics 483
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
(GST) are detoxication/biotransformation enzymes that are involved in the detoxi-
cation of toxic substances such as xenobiotics, carcinogens, free radicals and peroxides
by conjugating these substances with GSH (Hayes and Pulford 1995). Since the ultimate
carcinogenic form of DMH and AFB1 is a toxic electrophile (methyldiazonium ion and
carbonium ion) and exo-8,9-epoxide, GPx and GST take considerable signicance in
promoting carcinogen detoxication (Beckett and Hayes, 1993). ROS are produced
during passage of nutrients through the gastrointestinal tract. The natural production of
host antioxidants decreases rostrally. It is well known that oxidative damage forms part in
the pathogenesis of cancer, cirrhosis, atherosclerosis and other chronic diseases.
B. longum (ATCC 15708) and L. acidophilus (ATCC 4356) showed antioxidative activity,
inhibiting linoleic acid peroxidation by 2848% and also showed the ability to scavenge
the a-diphenyl-b-picrylhydrazyl free radical, scavenging 2152%. The intact cells of
these two intestinal bacteria demonstrated a high inhibitory effect on the cytotoxicity of
4-nitroquinoline-N-oxide (4NQO). Cytotoxicity of 4NQO was reduced by L. acidophilus
by approximately one-half, and by almost 90% by B. longum. Nevertheless, no inhibition
of cytoxicity was observed for intracellular cell-free extracts of 10
9
cells of B. longum and
L. acidophilus (Lin and Chang 2000). Pediococcus pentosaceus 16:1 and L. plantarum 2592
produced antioxidants after 18 h growth corresponding to 100 microgram vitamin C,
and L. paracasei F19 slightly less but another L. paracasei did not exert antioxidative
activity, again emphasizing that these characteristics are strain dependent (Kruszewska
et al. 2002). In a recent study, obligatory homofermentative lactobacilli produced high
antioxidant activity whereas this was highly strain dependent among facultatively and
obligate heterofermentative lactobacilli (Annuk et al. 2003).
Binding/adsorption of carcinogens
Several carcinogens such as heterocyclic amines and AFB1 are reportedly adsorbed or
bound in vitro by LAB and other intestinal bacteria. A concomitant decrease in
mutagenicity is often reported. (Orrhage 1994; Bolognani et al. 1997). In-depth
investigations have also showed that cultured milk possessed des-mutagenicity and
this activity increased with increasing numbers of viable cells, indicating that probiotics
could play an important role in the inhibition of mutagenicity (Usman 1998). Thya-
garaja and Hosono (1993) found that probiotic isolated from idly, a traditional cereal
pulse product of India, could exert des-mutagenicity on various spice mutagens,
heterocyclic amines and aatoxins. Subsequent studies on the des-mutagenicity
properties of probiotics suggested that the des-mutagenic substances may reside in
the cellular envelope of the bacterial cell wall (Singh et al. 1997). A cell wall preparation
of B. infantis was found to inhibit tumor activity in mouse peritoneal cells (Sekine et al.
1995), while a cell wall preparation of heat-killed L. casei (LC9018) was found to
induce immunity against tumor induction in a randomized, controlled and compar-
ative study involving 223 patients with stage III cervical cancer. Also there are some
potential probiotics, Bidobacterium adoleascentis ATCC 15703 and B. breve ATCC
15700, that exhibited higher antimutagenicity against benzo[a]pyrene (BaP) and
sodium azide (Chalova et al. 2008). Strains of B. lactis were shown to express anti-
mutagenic properties, probably linked to cell wall constituents. The anti-mutagenic
effect occurred even after acid and bile treatment, mimicking the gastrointestinal
transport; interestingly, enhanced in the presence of whole milk (Lo et al. 2004).
One mechanism for this effect could be binding of mutagens, as heterocyclic aromatic
484 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
amines were shown to be bound to the cell wall of certain bacteria such as B. longum
and other LAB, and thereby be detoxied (Orrhage et al. 1994). However, the anti-
mutagenic effect of L. plantarum KLAB21 was mediated by three glycoproteins that are
secreted extracellularly (Rhee and Park 2001).
Antigenotoxic activity of several Lactobacillus species against 4-nitroquinoline-
1-oxide was shown in in vitro tests, whereas only one L. acidophilus strain inhibited
MNNG. All strains also showed anti-mutagenic properties and were viable after the
tests (Caldini et al. 2005). Also, mutagens were suggested to be bound to the cell wall
of probiotics.
Effect on production of bacterial enzymes and metabolites
Oral administration of some LAB/fermented milks to animals and humans leads to a
decrease in certain metabolites and enzymes purported to be involved in synthesis or
activation of carcinogens, genotoxins and tumor promoters such as b-glucuronidase,
b-glucosidase, nitrate reductase and ammonia. Such changes in enzyme activities
concomitantly suppressed tumors. This would appear to be due to the low specic
activity of these enzymes in LAB (Saito and Rowland 1992). Such changes in enzyme
activity or metabolite concentration have been suggested to be responsible for the
decreased level of pre-neoplastic lesions or tumors seen in carcinogen-treated rats given
prebiotics and probiotics (Reddy and Rivenson 1993; Rowland et al. 1998).
Stimulation of host enzymes involved in carcinogen inactivation
There are isolated reports citing that administration of LAB results in increased activity
of antioxidative enzymes or processes that protect cells against carcinogen-induced
damage. These include GST, GSH, GSH reductase, and GSH peroxidase, which
consequently reduce the apoptotic rate and the necrobiotic changes in liver. Such
mechanisms of protection could be effective against a wide range of dietary carcinogens
possibly inuencing several cancer sites. Lowering of hepatic GSH levels and enhanced
lipid peroxidation was studied in animals dosed with AFB1 and other carcinogens. It
may be useful to employ hepatic lipid peroxidation as a biochemical marker during
AFB1-initiated hepato-carcinogenesis (Corrales et al. 1992; Aguilar-Delnetal.
1996; Gasso et al. 1996). Many food-borne carcinogens such as heterocyclic amines
and polycyclic aromatic hydrocarbons are known to be conjugated to GSH, which
appears to result in inactivation. The enzyme involved, GST, is found in the liver and in
other tissues including the gut. A study of the effect of B. longum and lactulose on AOM-
induced ACF in the colon showed that the activity of GSH in the colonic mucosa was
inversely related to the ACF numbers (Challa et al. 1997). Such a mechanism of
protection would be effective against a wide range of dietary carcinogens that the various
LAB can inhibit genotoxicity of dietary carcinogens. The degree of inhibition was
strongly species dependent. For example, Pool-Zobel et al. (1993) demonstrated that
L. casei and L. lactis inhibited the mutagenic activity of nitrosated beef by over 85%,
whereas L. confusus and Lactobacillus sake had no effect. Pre-treatment of mice with
L. casei 1 h before the administration of genotoxins can lead to a moderate reduction
(P<0.05) in the genotoxicity of pro-carbazine and urethane (Abraham and Paul 2001).
Using the technique of single-cell microgel electrophoresis (Comet assay), Pool-Zobel
et al. (1993) investigated the ability of range of species of LAB to inhibit DNA damage in
Cancer-preventing attributes of probiotics 485
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
the colon mucosa of rats treated with the carcinogens MNNG or DMH. All the strains of
lactobacilli and bidobacteria testedL.acidophilus (isolated from a yoghurt sample), L.
gasseri,L. confusus,B. breve and B. longumprevented MNNG-induced DNA damage
when given at a dose of 10
10
cells/kg body weight 8 h before feeding the carcinogen. In
most cases the DNA damage was reduced to a level similar to that in untreated rats. S.
thermophilus was not as effective as the other LAB strains. The comet assay has also been
used to evaluate the effect of a prebiotic, lactulose on DNA damage in the colonic
mucosa. Rats that were fed a diet containing 3% lactulose and given DMH exhibited less
DNA damage in colon cells than similarly treated animals fed a sucrose diet. In the latter
animals, the percentage of cells with severe DNA damage comprised 33% of the total,
compared with only 12.6% in the lactulose-fed rats (Rowland et al. 1998).
Enhancing the hosts immune response
In animal and human studies with probiotic treatments (L. casei,L. acidophilus,or
B. bidus) shown to inuence several aspects of immune function, enhancing secretory
IgA production was observed in which L. casei is most effective in stimulating secretory
IgA (Perdigon et al. 1991) and increasing the systemic immune response in malnour-
ished animals (Perdigon 1992). There are several other examples of probiotics (L. casei,
LGG, and other strains) that can affect cytokine production and promote a non-
specic immune response by enhancing phagocytosis of pathogens (Erickson and
Hubbard 2000). Another study showed that mice fed LAB had higher splenocyte
proliferation in response to mitogens for T cells and B cells (De Simone et al.
1993; Erickson and Hubbard 2000). LAB shows anti-tumor activity against sarcoma
180, a transplantable mouse tumor (Yokokura et al. 1981). One explanation for tumor
suppression by LAB may be mediated through an immune response of the host. Sekine
et al. (1985) suggested that B. infantis stimulates the host-mediated response, leading to
tumor suppression or regression. In addition, there are studies to suggest that LAB play
an important role and function in the hosts immunoprotective system by increasing
specic and non-specic mechanisms to have an anti-tumor effect (Kato et al.
1983; De Simone et al. 1993; Schiffrin et al. 1995). L. casei Shirota (LcS) has been
shown to have potent anti-tumor and anti-metastatic effects on transplantable tumor cells
and to suppress chemically induced carcinogenesis in rodents. Also, intra-pleural
administration of LcS into tumor-bearing mice has been shown to induce the production
of several cytokines, such as IFN-g, IL-1 and TNF-a, in the thoracic cavity of mice,
resulting in the inhibition of tumor growth and increased survival (Matsuzaki 1998). An
additional study has indicated that oral administration of BLP, a preparation of viable
L. casei YIT 9018, potentiated systemic immune responses that modied T-cell func-
tions in tumor-bearing mice (Kato et al. 1994). It has also been demonstrated that
B. longum and B. animalis promote the induction of inammatory cytokines (IL-6,
TNF-a) in mouse peritoneal cells (Sekine et al. 1994). Suppression of colon tumor
growth by yoghurt in DMH-treated mice was associated with suppression of the
inammatory immune response (Wollowski et al. 2001). Many case studies have shown
that consumption of fermented milk resulted in reduction of risk of breast, liver and
colon cancer (Aso and Akazan 1992).
Among them, LcS showed especially high potency. LcS is a strain of LAB that has
been selected for its specic biological activity in humans. This strain is not directly
cytotoxic to tumor cells in vitro; it has been postulated that its anti-tumor action may be
486 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
mediated by augmentation of the hosts immune system (Kato et al. 1981) and also
exhibited marked anti-tumor activity against human malignant cancer cells in clinical
trials (Aso and Akazan 1992; Aso et al. 1995). This idea stimulated further studies of
the anti-tumor and immunoregulatory actions of LcS in various experimental models
(Matsuzaki et al. 1985, 1988; Miake et al. 1985), and it has been demonstrated that oral
administration of LcS has benecial effects in both humans and animals. It has also
been reported that LcS induced recovery of host immune responses that were decreased
by treatment with carcinogens and augmented the natural killer activity and T-cell
functions of host immune cells. After LcS is ingested by the host, it is incorporated
into M cells in Peyers patches and digested to form active components. In Peyers
patches, macrophages or dendritic cells that phagocytosed LcS gained the ability to
produce several cytokines, especially TNF. The components of LcS digested in Peyers
patches were then recognized through Toll-like receptor 2 in antigen-presenting cells,
resulting in the production of several cytokines that elicited varied responses in host
immune cells (Takeshi et al. 2007). LcS exerted a strong inhibitory effect on carcino-
genesis in mice through the regulation of host immune cells (Takagi et al. 1999) in a
3-methylcholanthrene (MC)-induced carcinogenesis model (Noguchi et al. 1996) that
induced various tumors such as colon, liver, lung, uterine cervix, and mammary gland
cancer models (Rao and Hussain 1988; Baral and Maity 1992). Oral feeding of mice with
LcS inhibits MC-induced tumorigenesis by modulating the host immune responses that
are disrupted during MC carcinogenesis. A possible mechanism of the prevention of the
carcinogenesis is the activation of natural killer (NK) cells. NK cells are large granular
lymphocytes derived from bone marrow, and these cells display non-MHC-restricted
cytotoxicity against a variety of tumors. It is well recognized that NK cells act as cytolytic
effector cells of the innate immune system. Oral feeding of LcS to MC-treated mice
rendered their NK cells tumoricidal in terms of both quality and quantity, resulting in the
suppression of tumor incidence (Takagi et al. 2001). Other possible mechanism of action
in which effector cells that may respond to LcS are dendritic cells. Dendritic cells are
thought to be one of the most important types of cells involved in the presentation of
several antigens and in the production of cytokines (Banchereau and Steinman
1998; Rescigno et al. 1999). Recent studies showed Lactobacillus strain-specic activity
in prevention of murine tumorigenesis and in induction of IL-12 release by bone
marrow-derived dendritic cells in vitro (Takagi et al. 2008). Recently, Matsumoto
et al. (2009) described the role of the LAB strain LcS for prevention of colon cancer
by targeting immune system. They reported that L. casei produces polysaccharide, which
suppresses colon carcinogenesis by suppressing synthesis of IL-6 and signal transducer
and activator of transcription 3 in a mouse model.
These ndings suggest that treatment with probiotics has the potential to ameliorate
or prevent tumorigenesis through modulation of the hosts immune system, specically
cellular immune responses. Elucidating the precise mechanism of cancer prevention by
probiotic bacteria may help identify a novel and effective molecular target for cancer
prevention.
Production of conjugated linoleic acid from castor oil
Conjugated linoleic acid (CLA) 4 is a term dening a group of positional (e.g. 7:9,
9:11, 10:12, and 11:13) and geometric (i.e. cis or trans) isomers of linoleic acid
(C18, cis-9:cis-12) that have been shown to exert numerous health benets, including
Cancer-preventing attributes of probiotics 487
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
anti-atherogenic, anti-diabetic, anti-inammatory and anticarcinogenic properties
(Maggiora et al. 2004). In vitro, CLA inhibits the growth of HT-29 and Caco-2 colon
cancer cells (Lampen et al. 2005). CLA-treated SW480 colonic tumor cells possess
increased caspase-3 and caspase-9 activities and reduced Bcl-2 expression compared
with controls (Miller et al. 2002). CLA has been shown to reduce the incidence of
colonic, skin, mammary, and prostate carcinogenesis in animal models (Belury 2002).
Rats supplemented with CLA showed reduced incidence of colonic tumors and
increased apoptotic indices in response to the administering of DMH (Kim and
Park 2003). Some probiotic strains of bacteria have demonstrated protective effects
against tumor production (Sekine et al. 1995; Bifet al. 1997; OMahony et al.
2001; Saikali et al. 2004). L. acidophilus and B. longum have been shown to reduce
incidence of colonic tumors and aberrant crypt foci, respectively, in animal models
(Rowland et al. 1996; McIntosh et al. 1999). A cocktail of probiotic strains was recently
demonstrated to increase the colonic apoptotic index in normal rats (Linsalata et al.
2005). Several strains of bacteria that are considered to have probiotic effects
(i.e. lactobacilli and bidobacteria) are capable of converting linoleic acid to CLA.
In this study, we investigated a previously unreported mechanism of probiotic action:
the production of CLA. We demonstrated that probiotic strains in VSL3 (L. casei,
L. plantarum,L. acidophilus,L. delbrueckii subsp. bulgaricus,B. infantis,B. breve,
B. longum,S. salivarius subsp. thermophilus) have the capacity to convert linoleic
acid to CLA, inducing the upregulation of PPARg, a reduction in cancer cell viability,
and the induction of apoptosis. This linoleic acid-conjugating capacity is maintained
in vivo (Julia et al. 2006). One LAB strain, L. plantarum JCM 1551, was shown to
efciently produce conjugated linoleic acid from castor oil in the presence of lipase,
which is an interesting side effectof anti-tumor activities of LAB (Ando et al. 2004).
By regulation of apoptosis
Apoptosis is a genetically regulated active process abolishing cell populations in both
physiological and pathological processes. Impaired apoptosis, either due to expression
of oncogenes or mutations of tumor suppressor genes, leads to an uncontrolled
accumulation of malignant cells, and eventually to cancer.
Caspase-3 is one of the cysteine proteases that play a major role in the execution of
apoptosis (Nicholson 1999). A number of genetic and biochemical studies suggest that
caspase activation is essential for the occurrence of the apoptotic phenotype of cell
death (Janicke et al. 1998) A variety of caspase substrates are involved in the regulation
of DNA structure, repair and replication. Caspase-3 substrate cleavage has been
observed under oxidative stress in different pathological conditions (Nicholson and
Thornberry 1997). Aatoxins are one of the most dangerous mycotoxins known, owing
to their high toxicity to both animals and human. AFB1, a metabolite of Aspergillus
avus, is a potent hepatotoxic and hepatocarcinogenic mycotoxin. One of manifesta-
tions of AFB1-induced toxicity is oxidative stress (Souza et al. 1999). Recently, it is
accepted that oxidative stress is an apoptosis inducer (Chandra et al 2000). Many
agents that induce apoptosis are either oxidants or stimulators of cellular oxidative
metabolism. Conversely, many inhibitors of apoptosis have antioxidant activities or
enhance cellular antioxidant defenses (Freeman and Grapo 1982). The GSH redox
cycle is an important component of the antioxidant machinery in cells. In normal cells,
a primary defense against oxidative damage is provided by antioxidants such as GSH
488 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
and the onset of apoptosis is associated with a fall in intracellular GSH in numerous
cellular systems (Ratan et al. 1994).
There are isolated reports citing that administration of LAB results in increased
activity of antioxidative enzymes or process that protect cells against carcinogen-
induced damage. These include GST, GSH, GSH reductase, and GSH peroxidase,
which consequently reduce the apoptotic rate and the necrobiotic changes in
liver. Such mechanisms of protection could be effective against a wide range of
dietary carcinogens possibly inuencing several cancer sites. Some probiotics
(e.g. LGG) prevent cytokine-induced apoptosis in intestinal epithelial cell models.
Also it is well known that LGG and LGG-fermented milk are potent scavengers of
superoxide anion and inhibitors of lipid peroxidation reactions in vitro (Ahotupa et al.
1996).
Culture of LGG with either mouse or human colon cells activates the anti-apoptotic
Akt/protein kinase B. This model probiotic also inhibits activation of the pro-apoptotic
p38/mitogen-activated protein kinase by TNF, IL-1aor IFN-g. Furthermore, products
recovered from LGG culture broth supernatant showed concentration-dependent
activation of Akt and inhibition of cytokine-induced apoptosis. These observations
suggest a novel mechanism of communication between probiotics and colon epithelia
that increases survival of intestinal cells normally found in an environment of
pro-apoptotic cytokines (Fang and Polk 2002). LGG enhanced gastric ulcer healing
via attenuation of the cell apoptosis to cell proliferation ratio and an increase in
angiogenesis. Regulators of these processes such as ornithine decarboxylase, B-cell
lymphoma 2 (Bcl-2), vascular endothelial growth factor and epidermal growth factor
receptor are likely to be involved in the healing action of LGG for gastric ulcer
(Lam et al. 2007). B. longum is a probiotic, known for its benecial effects to the
human gut and even for its immune-modulatory and anti-tumor activities. Recently,
many studies have indicated an intimate relation between probiotic bacteria and the
gut mucosa and its inuence on human cellular homeostasis. Bacteria were incubated
with Caco-2 cells to investigate apoptotic deletion. Co-cultures of Caco-2 cells with
adherent strains (B12 and B18) of B. longum induced DNA fragmentation, indicating
that it can induce apoptotic deletion of Caco-2 cells. Thus it helps in restoring the
ecology of damaged colon tissues. Lactobacillus reuteri secretes factors that potentiate
apoptosis in myeloid leukaemia-derived cells induced by TNF, as indicated by
intracellular esterase activity, terminal deoxynucleotidyl transferase-mediated
deoxyuridine triphosphate nick end-labeling assays and poly(ADP-ribose) polymer-
ase cleavage. L. reuteri downregulated nuclear factor-kB-dependent gene products
those mediate cell proliferation (Cox-2, cyclin D
1
) and cell survival (Bcl-2, Bcl-xL).
Moreover Otte et al. (2009) demonstrated that a probiotic combination call VSL#3
signicantly suppressed the COX-2 expression in a cancer cell line (Colo320 and
SW480 intestinal epithelial cells). L. reuteri suppressed TNF-induced nuclear
factor-kB activation, including nuclear factor-kB-dependent reporter gene
expression, in a dose-dependent and time-dependent manner. L. reuteri may
regulate cell proliferation by promoting apoptosis of activated immune cells via
inhibition of IkBaubiquitination and enhancing pro-apoptotic mitogen-activated
protein kinase signaling. An improved understanding of L. reuteri-mediated effects
on apoptotic signaling pathways may facilitate development of future probiotic-
based regimens for prevention of colorectal cancer and inammatory bowel disease
(Iyer et al. 2008).
Cancer-preventing attributes of probiotics 489
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
Conclusions
Although there is no direct evidence for the prevention or treatment of cancer
malignancies in humans using probiotics, numerous in vivo animal studies have been
carried out. These have demonstrated that probiotics are capable of reducing the
incidence of tumor formation and aberrant crypt formation, thereby suppressing bac-
terial enzyme activities and reducing DNA damage. Mechanistic studies indicate that
probiotics may exert anticarcinogenic properties by altering colonic metabolism, degrad-
ing carcinogens, producing antimutagenic compounds, and enhancing host immune
responses and by preventing cytokine-induced apoptosis of colonic epithelial cells.
Therefore LAB offer potential as chemo-protective agents and thus further research
is clearly needed to quantify the benecial effects for prevention of human cancer.
Declaration of interest: The authors report no conicts of interest. The authors alone
are responsible for the content and writing of the paper.
References
Abdelali H, Cassand P, Soussotte V, Daubeze M, Bouley C, Narbonne JF. 1995. Effect of dairy products on
initiation of precursor lesions of colon cancer in rats. Nutr Cancer 24:121123.
Abraham SK, Paul J. 2001. Protective effects of lactic acid bacteria against genotoxicity in mice. Curr Sci
80:1310-1312.
Aguilar F, Hussain SP, Cerutti P. 1993. Aatoxin B1 induces the transversion of G! T in codon 249 of the
p53 tumor suppressor gene in human hepatocytes. Proc Natl Acad Sci USA 90:85868590.
Aguilar-Deln I, Lopez-Barrera F, Hernandez-Munoz R. 1996. Selective enhancement of lipid peroxidation
in plasma membrane in two experimental models of liver regeneration: Partial hepatectomy and acute
CCl4 administration. Hepatology 2(4):657662.
Ahotupa M, Saxelin M, Korpela R. 1996. Antioxidant properties of lactobacillus GG. Nutr Today Suppl
31:5152.
Ando A, Ogawa J, Kishino S, Shimizu S. 2004. Conjugated linoleic acid production from castor oil by
Lactobacillus plantarum JCM 1551. Enzyme Microb Technol 35:4045.
Annuk H, Shchepetova J, Kullisaar T, Songisepp E, Zilmer M, Mikelsaar M. 2003. Characterization of
intestinal lactobacilli as putative probiotic candidates. J Appl Microbiol 94:403412.
Arimochi H, Kinouchi T, Kataoka K, Kuwahara T, Ohnishi Y. 1997. Effect of intestinal bacteria on
formation of azoxymethane-induced aberrant crypt foci in the rat colon. Biochem Biophys Res Commun
238:753757.
Aso Y, Akazan H. 1992. Prophylactic effect of a Lactobacillus casei preparation on the recurrence of supercial
bladder cancer. BLP Study Group. Urol Int 49:125129.
Aso Y, Akaza H, Kotake T, Tsukamoto T, Imai K, Naito S 1995. Preventive effect of a Lactobacillus casei
preparation on the recurrence of supercial bladder cancer in a double-blind trial. The BLP Study Group.
Eur Urol 27:104109.
Banchereau J, Steinman RM. 1998. Dendritic cells and the control of immunity. Nature 392:245252.
Baral RN, Maity P. 1992. Induction of colorectal cancer in rats by 20-methylcholanthrene. Cancer Lett
61:177183.
Barber DA, Harris SR. 1994. Oxygen free radicals and antioxidants: A review. Am Pharm 34:2635.
Bartsch H, Nair J. 2002. Potential role of lipid peroxidation derived DNA damage in human colon
carcinogenesis: Studies on exocyclic base adduct as stable oxidative stress markers. Cancer Detect
Prev 26:308312.
Beckett GJ, Hayes JD. 1993. Glutathione S-transferases: biomedical applications. Adv Clin Chem 30:
281380.
Belury MA. 2002. Inhibition of carcinogenesis by conjugated linoleic acid: Potential mechanisms of action.
J Nutr 132:29952998.
Biasco G, Paganelli GM, Brandi G, Brillanti S, Lami F, Callegari C, Gizzi G. 1991. Effect of Lactobacillus
acidophilus and Bidobacterium bidum on rectal cell kinetics and fecal pH. Ital J Gastroenterol 23:142147.
Bibel DJ. 1998. Elie Metchnikoffs bacillus of long life. ASM News 54:661665.
490 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
BifA, Coradini D, Larsen R, Riva L, Di Fronzo G. 1997. Antiproliferative effect of fermented milk on the
growth of a human breast cancer cell line. Nutr Cancer 28:9399.
Bird RP. 1995. Role of aberrant crypt foci in understanding the pathogenesis of colon cancer. Cancer Lett
93:5571.
Bolognani F, Rumney CJ, Rowland IR. 1997. Inuence of carcinogen binding by lactic acid producing
bacteria on tissue distribution and in-vivo mutagenicity of dietary carcinogens. Food Chem Toxicol
35:535545.
Bosch FX, Munoz N. 1988. Prospects for epidemiological studies on hepatocellular cancer as a model for
assessing viral and chemical interactions. IARC Sci Publ 89:427438.
Bulatao-Jayme J, Almero EM, Castro MC, Jardeleza MT, Salamat LA. 1982. A casecontrol dietary study of
primary liver cancer risk from aatoxin exposure. Int J Epidemiol 11:112119.
Burns AJ, Rowland IR. 2000. Anti-carcinogenicity of probiotics and prebiotics. Curr Issues Intest Microbiol
1:1324.
Caldini G, Trotta F, Villarini M, Moretti M, Pasquini R, Scassellati-Sforzolini G, Cenci G. 2005. Screening
of potential lactobacilli antigenotoxicity by microbial and mammalian cell-based tests. Int. J Food
Microbiol 102:3747.
Campbell TC, Hayes JR. 1976. The effect of quantity and quality of dietary protein on drug metabolism. Fed
Proc 35:24702474.
Challa A, Rao DR, Chawan CB, Shackelford L. 1997. Bidobacterium longum and lactulose suppress
azoxymethane-induced colonic aberrant crypt foci in rats. Carcinogenesis 18:517521.
Chalova VI, Lingbeck JM, Kwon YM, Ricke SC. 2008. Extracellular antimutagenic activities of selected
probiotic Bidobacterium and Lactobacillus spp. as a function of growth phase. J Environ Sci Health Part B
43:193198.
Chandra J, Samali A, Orrenius S. 2000. Triggering and modulation of apoptosis by oxidative stress. Free
Radic Biol Med 29:323333.
Chau I, Cunningham D. 2006. Adjuvant therapy in colon cancerwhat, when and how? Ann Oncol
17:13471359.
Chin SE, Liu W, Storkson JM, Ha YL, Pariza MW. 1992. Dietary sources of conjugated dienoic isomers of
linoleic acid, a newly recognized class of anticarcinogens. J Food Comp Anal 5:185197.
Collins MD, Gibson GR. 1999. Probiotics, prebiotics, and synbiotics: approaches for modulating the
microbial ecology of the gut. Am J Clin Nutr 69:1052S-1057S.
Corrales F, Gimenez A, Alvarez L, Caballeria J, Pajares MA, Andreu H, Pares A, Mato JM, Rodes J. 1992.
S-adenosylmethionine treatment prevents carbon tetrachloride-induced S-adenosylmethionine synthetase
inactivation and attenuates liver injury. Hepatology 16:10221027.
De Simone C, Vesely R, Bianchi SB, Salvadori B, Jirillo E. 1993. The role of probiotics in modulation of the
immune system in man and in animals. Int J Immunother 9:2328.
Di Caro S, Tao H, Grillo A, Elia C, Gasbarrini G, Sepulveda AR, Gasbarrini A. 2005. Effects of Lactobacillus
GG on genes expression pattern in small bowel mucosa. Digest Liver Dis 37:320332.
Drisko JA, Giles CK, Bischoff BJ. 2003. Probiotics in health maintenance and disease prevention. Alt Med
Rev 8:143155.
Erdman SH, Wu HD, Hixson LJ, Ahnen DJ, Gerner EW. 1997. Assessment of mutations in Ki-ras and
p53 in colon cancers from azoxymethane- and dimethylhydrazine-treated rats. Mol Carcinogen 19:137
144.
Erickson KL, Hubbard NE. 2000. Probiotic immuno-modulation in health and disease. J Nutr 130:
403S409S.
Essigmann JM, Croy RG, Nadzan AM, Busby WF Jr, Reinhold VN, Buchi G, Wogan GN. 1977. Structural
identication of the major DNA adduct formed by aatoxin B1 in-vitro. Proc Natl Acad Sci USA 74:
18701874.
Fang Y, Polk DB. 2002. Probiotic bacterium prevents cytokine-induced apoptosis in intestinal epithelial cells.
J Biol Chem 277:50959-50965.
Felton JS, Jagerstad M, Knize MG, Skog K, Wakabayashi K. 2000. Contents in foods, beverages and tobacco.
In: Nagao M, Sugimura T, editors. Food borne carcinogenesis: Heterocyclic amines. New York: Wiley.
pp 529.
Femia AP, Luceri C, Dolara P, Giannini A, Biggeri A, Salvadori M, Clune Y, Collins KJ, Paglierani M,
Caderni G. 2002. Antitumorigenic activity of the prebiotic inulin enriched with oligofructose in
combination with the probiotics Lactobacillus rhamnosus and Bidobacterium lactis on azoxymethane-
induced colon carcinogenesis in rats. Carcinogenesis 23:19531960.
Cancer-preventing attributes of probiotics 491
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
Fernandes CF, Shahani KM. 1990. Anticarcinogenic and immunological properties of dietary lactobacilli.
J Food Protect 53:704710.
Finley JW, Davis CD, Feng Y. 2000. Selenium from high-selenium broccoli protects rats from colon cancer.
J Nutr 130:23842389.
Freeman BA, Grapo JD. 1982. Biology of disease: Free radical and tissue injury. Lab Invest 47:412426.
Gallaher DD, Stallings WH, Blessing LL, Busta FF, Brady LJ. 1996. Probiotics, cecal microbiota, and
aberrant crypts in the rat colon. J Nutr 126:13621371.
Garner RC, Miller EC, Miller JA. 1972. Liver microsomal metabolism of aatoxin B1 to a reactive derivative
toxic to Salmonella typhimurium TA 1530. Cancer Res 32:20582066.
Gasso M, Rubio M, Varela G, Cabre M, Caballeria J, Alonso E, Deulofem R, Camps J, Gimenez A, Pajares
M, Pares A, et al 1996. Effects of S-adenosylmethionine on lipid peroxidation and liver brogenesis in
carbon tetrachloride-induced cirrhosis. J Hepatol 25:200205.
Goldin B. 1996. The metabolic activity of the intestinal microbiota and its role in colon cancer. Nutr Today
31:24S27S.
Goldin BR. 1990. Intestinal microbiota: Metabolism of drugs and carcinogens. Ann Med 22:4348.
Goldin BR, Gorbach SL. 1980. Effect of Lactobacillus acidophilus dietary supplements on
1,2-dimethylhydrazine dihydrochloride-induced intestinal cancer in rats. J Natl Cancer Inst 64:263265.
Goldin BR, Gorbach SL. 1984. The effect of milk and lactobacillus feeding on human intestinal bacterial
enzyme activity. Am J Clin Nutr 39:756761.
Goldin BR, Swenson L, Dwyer J, Sexton M, Gorbach SL. 1980. Effect of diet and Lactobacillus acidophilus
supplements on human fecal bacterial enzymes. J Natl Cancer Inst 64:255261.
Goldin BR, Gualtieri LJ, Moore RP. 1996. The effect of Lactobacillus GG on the initiation and promotion of
DMH-induced intestinal tumours in the rat. Nutr Cancer 25:197204.
Halliwell B. 1996. Mechanisms involved in the generation of free radicals. Pathol Biol 44:613.
Haskard C, Binnion C, Ahokas J. 2000. Factors affecting the sequestration of aatoxin by Lactobacillus
rhamnosus strain GG. Chem Biol Interact 128:3949.
Hayatsu H, Hayatsu T. 1993. Suppressing effect of Lactobacillus casei administration on the urinary
mutagenicity arising from ingestion of fried ground beef in the human. Cancer Lett 73:173179.
Hayes JD, Pulford DJ. 1995. The glutathione S-transferase supergene family: Regulation of GST and
contribution of the isoenzymes to cancer chemoprevention and drug resistance. Crit Rev Biochem Mol
Biol 31:445600.
Hirayama K, Rafter J. 1999. The role of lactic acid bacteria in colon cancer prevention: Mechanistic
considerations. Ant V Leeuwenhoek 76:391394.
Hirayama K, Rafter J. 2000. The role of probiotic bacteria in cancer prevention. Microbes Infect 2:681686.
Iyer C, Kosters A, Sethi G, Kunnumakkara AB, Aggarwal BB, Versalovic J. 2008. Probiotic Lactobacillus
reuteri promotes TNF-induced apoptosis in human myeloid leukemia-derived cells by modulation of
NF-kB and MAPK signalling. Cellular Microbiol 10:14421452.
Iyer RS, Coles BF, Raney KD, Their R, Guengerich FP, Harris TM. 1994. DNA adduction by the potent
carcinogen aatoxin B-1-mechanistic studies. J Am Chem Soc 116:16031609.
Janicke RU, Spregart ML, Wati MR, Porter AG. 1998. Caspase-3 is required for DNA fragmentation and
morphological changes associated with apoptosis. J Biol Chem 273:93579360.
Jeong HY, Kim TH, Park JS, Kim KT, Paik HD. 2003. Antioxidative and cholesterol-reducing activity of
Bacillus polyfermenticus SCD. Korean J Biotechol Bioeng 18:371376.
Julia B, Ewaschuk JW, Walker H, Diaz H, Madsen KL. 2006. Bioproduction of conjugated linoleic acid by
probiotic bacteria occurs in-vitro and in-vivo in mice. J Nutr 136:14831487.
Kampman E, Goldbohm RA, van den Brandt PA. 1994. Fermented dairy products, calcium and colorectal
cancer in the Netherlands cohort study. Cancer Res 54:31863190.
Kato I, Kobayashi S, Yokokura T, Mutai M. 1981. Antitumor activity of Lactobacillus casei in mice. Gann
72:517523.
Kato I, Yokokura T, Mutai M. 1983. Macrophage activation by Lactobacillus casei in mice. Microbiol
Immunol 27:611618.
Kato I, Endo K, Yokokura T, 1994. Effects of oral administration of Lactobacillus casei on antitumor
responses induced by tumor resection in mice. Int J Immunopharmacol 16:2936.
Khanafari A, Soudi H, Miraboulfathi M, Osbo RK. 2007. An in vitro investigation of aatoxin B1 biological
control by Lactobacillus plantarum. Pakistan J Biol Sci 10:25532556.
Kim KH, Park HS. 2003. Dietary supplementation of conjugated linoleic acid reduces colon tumor incidence
in DMH-treated rats by increasing apoptosis with modulation of biomarkers. Nutrition 19:772777.
492 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
Korenaga D, Takesue F, Kido K, Yasuda M, Inutsuka S, Honda M, Nagahama S. 2002. Impaired
antioxidant defense system of colonic tissue and cancer development in dextran sulfate sodium-
induced colitis in mice. J Surg Res 102:144149.
Kruszewska D, Lan J, Lorca G, Yanagisawa N, Marklinder I, Ljungh A. 2002. Selection of lactic acid bacteria
as probiotic strains by in-vitro tests. Microb Ecol Health Dis 29:3749.
Kulkarni N, Reddy BS. 1994. Inhibitory effect of Bidobacterium longum cultures on the azoxymethane
induced aberrant crypt foci formation and fecal bacterial b-glucuronidase. Proc Soc Exp Biol Med
207:278283.
Lam EKY, Tai EKK, Koo MWL, Wong HPS, Wu WKK, Yu L, So WHL, Cho CH. 2007. Enhancement of
gastric mucosal integrity by Lactobacillus rhamnosus GG. Life Sci 80:21282136.
Lampen A, Leifheit M, Voss J, Nau H. 2005. Molecular and cellular effects of cis-9, trans-11-conjugated
linoleic acid in enterocytes: Effects on proliferation, differentiation, and gene expression. Biochim Biophys
Acta 1735:3040.
Lang NP, Butler MA, Massengill J, Lawson M, Stotts RC, Maurer-Jensen M, Kadlubar FF. 1994. Rapid
metabolic phenotypes for acetyltransferase and cytochrome P4501A2 and putative exposure to food-borne
heterocyclic amines increase the risk for colorectal cancer or polyps. Cancer Epidemiol Biomark Prevent
3:675682.
Lee YK, Salminen S. 1995. The coming age of probiotics. Trends Food Sci Technol 6:241245.
Lidbeck A, Nord CE, Gustafsson JA, Rafter J. 1992. Lactobacilli, anticarcinogenic activities and human
intestinal microbiota. Eur J Cancer Prev 1:341353.
Lilleberg SL, Cabonce MA, Raju NR, Wagner LM, Kier LD. 1992. Alterations in the p53 tumor suppressor
gene in rat liver tumors induced by aatoxin B1. Prog Clin Biol Res 376:203222.
Lin MY, Chang FJ. 2000. Antioxidative effect of intestinal bacteria Bidobacterium longum ATCC 15708 and
Lactobacillus acidophilus ATCC 4356. Dig Dis Sci 45:16171622.
Ling WH, Korpela R, Mykkanen H, Salminen S, Hanninen O. 1994. Lactobacillus strain GG
supplementation decreases colonic hydrolytic and reductive enzyme activities in healthy female adults.
J Nutr 124:1823.
Linsalata M, Russo F, Berloco P, Valentini AM, Caruso ML, De Simone C, Barone M, Polimeno L, Di Leo
A. 2005. Effects of probiotic bacteria (VSL#3) on the polyamine biosynthesis and cell proliferation of
normal colonic mucosa of rats. In Vivo 19:989995.
Lo PR, Yu RC, Chou CC, Huang EC. 2004. Determinations of the antimutagenic activities of several
probiotic bidobacteria under acidic and bile conditions against benzo[a]pyrene by a modied Ames test.
Int J Food Microbiol 93:249257.
Maggiora M, Bologna M, Ceru MP, Possati L, Angelucci A, Cimini A, Miglietta A, Bozzo F, Margiotta C,
Muzio G, Canuto RA. 2004. An overview of the effect of linoleic and conjugated-linoleic acids on the
growth of several human tumor cell lines. Int J Cancer 112:909919.
Marteau P, Rochart P, Flourie B, Pellier P, Santos L, Desjeux JF, Rambaud JC. 1990. Effect of chronic
ingestion of a fermented dairy product containing Lactobacillus acidophilus and Bidobacterium bidum on
metabolic activities of the colonic ora in humans. Am J Clin Nutr 52:685688.
Matsuzaki T. 1998. Immunomodulation by treatment with Lactobacillus casei strain Shirota. Int J Food
Microbiol 41:133140.
Matsuzaki T, Yokokura T, Azuma I. 1985. Anti-tumor activity of Lactobacillus casei on Lewis lung carcinoma
and line-10 hepatoma in syngeneic mice and guinea pigs. Cancer Immunol Immunother 20:1822.
Matsuzaki T, Yokokura T, Mutai M. 1988. Antitumor effect of intrapleural administration of Lactobacillus
casei in mice. Cancer Immunol Immunother 26:209214.
McIntosh GH, Royle PJ, Playne MJ. 1999. A probiotic strain of L. acidophilus reduces DMH-induced large
intestinal tumors in male SpragueDawley rats. Nutr Cancer 35:153159.
Miake S, Nomoto K, Yokokura T, Yoshikai Y, Mutai M, Nomoto K. 1985. Protective effect of Lactobacillus
casei on Pseudomonas aeruginosa infection in mice. Infect Immun 48:480485.
Miller A, Stanton C, Devery R. 2002. Cis 9, trans 11- and trans 10, cis 12-conjugated linoleic acid isomers
induce apoptosis in cultured SW480 cells. Anticancer Res 22:38793887.
Modler GW, McKellar RC, Yaguchi M. 1990. Bidobacteria and bidogenic factors. Can Inst Food Sci
Technol J 23:2941.
Murosaki S, Muroyama K, Yamamoto Y, Yoshikai Y, 2000. Antitumor effect of heat-killed Lactobacillus
plantarum L-137 through restoration of impaired interleukin-12 production in tumor-bearing mice.
Cancer Immunol Immunother 49:157164.
Cancer-preventing attributes of probiotics 493
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
Matsumoto S, Hara T, Nagaoka M, Mike A, Mitsuyama K, Sako T, Yamamoto M, Kado S, Takada T. 2009.
A component of polysaccharide peptidoglycan complex on Lactobacillus induced an improvement of
murine model of inammatory bowel disease and colitis-associated cancer. Immunology 128:e170e180.
Nadathur S, Gould S, Bakalinsky A. 1995. Antimutagenicity of an acetone extract of yoghurt. Mutat Res
334:213224.
Nair S, Norkus EP, Hertan H, Pitchumoni CS. 2002. Serum and colon mucosa micronutrient antioxidants:
Differences between adenomatous polyp patients and controls. Am J Gastroenterol 96:34003405.
Nicholson DW. 1999. Caspase structure, proteolytic substrates and function during apoptotic cell death. Cell
Death Diff 6:10281042.
Nicholson DW, Thornberry NA. 1997. Caspases: Killer proteases. Trends Biochem Sci 22:299306.
Noguchi Y, Jungbluth A, Richards EC, Old LJ. 1996. Effect of interleukin 12 on tumor induction by
3-methylcholanthrene. Proc Natl Acad Sci USA 93:1179811801.
OMahony L, Feeney M, OHalloran S, Murphy L, Kiely B, Fitzgibbon J, Lee G, OSullivan G, Shanahan F,
Collins JK. 2001. Probiotic impact on microbial ora, inammation and tumour development in IL-10
knockout mice. Aliment Pharmacol Ther 15:12191225.
Ohishi T, Kishimoto Y, Miura N, Shiota G, Kohri T, Hara Y, Hasegawa J, Isemura M. 2002. Synergistic
effects of ()-epigallocatechin gallate with sulindac against colon carcinogenesis of rats treated with
azoxymethane. Cancer Lett 177:4956.
Okawa T, Niibe H, Arai T, Sekiba K, Noda K, Takeuchi S, Hashimoto S, Ogawa N. 1993. Effect of LC9018
combined with radiation therapy on carcinoma of the uterine cervix. A phase III, multicenter, randomized,
controlled study. Cancer 72:1949-1954.
Onoue M, Kado S, Sakaitani Y, Uchida K, Morotomi M. 1997. Specic species of intestinal bacteria
inuence the induction of aberrant crypt foci by 1,2-dimethylhydrazine in rats. Cancer Lett 113:179186.
Orrhage K, Sillerstrom E, Gustafsson JA, Nord CE, Rafter J. 1994. Binding of mutagenic heterocyclic amines
by intestinal and lactic acid bacteria. Mutat Res 311:239248.
Otte JM, Mahjurian-Namari R, Brand S, Werner I, Schmidt WE, Schmitz F. 2009. Probiotics regulate the
expression of COX-2 in intestinal epithelial cells. Nutr Cancer 61:103113.
Park E, Kim KT, Kim CJ, Paik HD. 2004. Anticarcinogenic and antigenotoxic effects of Bacillus
polyfermenticus. J Microbiol Biotechnol 14:852858.
Park E, Jeon GI, Park JS, Paik HD. 2007. A probiotic strain of Bacillus polyfermenticus reduces DMH induced
precancerous lesions in F344 male rat. Biol Pharm Bull 30(3):569574.
Parke DV, Ioannides C. 1990. Role of cytochrome P-450 in mouse liver tumor production. Prog Clin Biol
Res 331:215230.
Parkin DM, Bray F, Ferlay J, Pisani P. 2005. Global cancer statistics 2002. CA Cancer J Clin 55:74108.
Payne CM, Bernstein C, Bernstein H, Gerner EW, Garewal H. 1999. Reactive nitrogen species in colon
carcinogenesis. Antioxid Redox Signal 1:449467.
Pedrosa MC, Golner BB, Goldin BR, Barakat S, Dallal GE, Russell RM. 1995. Survival of yogurt-containing
organisms and Lactobacillus gasseri (ADH) and their effect on bacterial enzyme activity in the
gastrointestinal tract of healthy and hypochlorhydric elderly subjects. Am J Clin Nutr 61:353359.
Perdigon G. 1992. Probiotics and the immune state. In: Fuller R, editor. Probiotics: The scientic basis.
London: Chapman and Hall. p 145.
Perdigon G, Alvarez S, Pesce de Ruiz Holgado A. 1991. Immunoadjuvant activity of oral Lactobacillus casei:
Inuence of dose on the secretory immune response and protective capacity in intestinal infections. J Dairy
Res 58:485496.
Petzinger E, Weidenbach A. 2000. Mycotoxins in the food chain: The role of ochratoxins (2002) Livestock
Production Science, 76, pp 245250.
Petzinger E, Ziegler K. 2000. Ochratoxin A from a toxicological perspective. J Vet Pharmacol Ther. 23:
9198.
Piotrowska M, Zakowska Z. 2005. The elimination of Ochratoxin A by lactic acid bacteria strains. Polish J
Microbiol 54:279286.
Pool-Zobel BL, Munzner R, Holzaapfel H. 1993. Antigenotoxic properties of lactic acid bacteria in the
S. typhimurium mutagenicity assay. Nutr Cancer 20:261270.
Pool-Zobel BL, Neudecker C, Domizlaff I, Ji S, Schillinger U, Rumney C, Moretti M, Vilarini I, Scasellati-
Sforzolini R, Rowland IR. 1996. Lactobacillus and Bidobacterium mediated antigenotoxicity in the colon of
rats. Nutr Cancer 26:365380.
Rafter JJ. 1995. The role of lactic acid bacteria in colon cancer prevention. Scand J Gastroenterol 30:497502.
Rao AR, Hussain SP. 1988. Modulation of methylcholanthrene-induced carcinogenesis in the uterine cervix
of mouse by indomethacin. Cancer Lett 43:1519.
494 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
Ratan RR, Murphy TH, Baraban JM. 1994. Oxidative stress induces apoptosis in embryonic cortical
neurons. J Neurochem 62:376379.
Reddy BS. 1998. Prevention of colon cancer by pre- and probiotics: Evidence from laboratory studies. Br J
Nutr 80:S219S223.
Reddy BS, Hamid R, Rao CV. 1997. Effect of dietary oligofructose and inulin on colonic preneoplastic
aberrant crypt foci inhibition. Carcinogenesis 18:13711374.
Reddy BS, Rivenson A. 1993. Inhibitory effect of Bidobacterium longum on colon, mammary, and liver
carcinogenesis induced by 2-amino-3-methylimidazo[4,5-f]quinoline, a food mutagen. Cancer Res
53:39143918.
Rescigno M, Granucci F, Citterio S, Foti M, Ricciardi-Castagnoli P. 1999. Coordinated events during
bacteria-induced DC maturation. Immunol Today 20:200203.
Reynolds SH, Stowers SJ, Patterson RM, Maronpot RR, Aaronson SA, Anderson MW. 1987. Activated
oncogenes in B6C3F1 mouse liver tumors: Implications for risk assessment. Science 237:13091316.
Rhee CH, Park HD. 2001. Three glycoproteins with antimutagenic activity identied in Lactobacillus
plantarum KLAB21. Appl Environ Microbiol 67:34453449.
Rice-Evans C, Burdon R. 1993. Free radicallipid interactions and their pathological consequences. Prog
Lipid Res 32:71110.
Rowland IR, Bearne CA, Fischer R, Pool-Zobel BL. 1996. The effect of lactulose on DNA damage induced
by DMH in the colon of human ora-associated rats. Nutr Cancer 26:3747.
Rowland IR, Rumney CJ, Coutts JT, Lievense LC. 1998. Effect of Bidobacterium longum and inulin
on gut bacterial metabolism and carcinogen-induced aberrant crypt foci in rats. Carcinogenesis
19:281285.
Saikali J, Picard C, Freitas M, Holt P. 2004. Fermented milks, probiotic cultures, and colon cancer. Nutr
Cancer 49:1424.
Saito YT, Rowland IR. 1992. Effects of soybean oligosaccharides on the human gut microbiota in in-vitro
culture. Microb Ecol Health Dis 5:105110.
Salminen S, Von Wright A. 1998. Current probioticssafety assured? Microbial Ecol Health Dis 10:6877.
Schiffrin EJ, Rochat F, Link-Amster H, Aeschlimann JM, Donnet-Hughes A. 1995. Immunomodulation of
human blood cells following the ingestion of lactic acid bacteria. J Dairy Sci 78:491496.
Sekine K, Toida T, Saito M, Kuboyama M, Kawashima T, Hashimoto Y. 1985. A new morphologically
characterized cell wall preparation (whole peptidoglycan) from Bidobacterium infantis with a higher
efcacy on the regression of an established tumor in mice. Cancer Res 45:13001307.
Sekine K, Kawashima T, Hashimoto, Y. 1994. Comparison of the TNF-alevels induced by human-derived
Bidobacterium longum and rat-derived Bidobacterium animalis in mouse peritoneal cells. Bidobacteria
Microbiota 13:7989.
Sekine K, Ohta J, Onishi M, Tatsuki T, Shimokawa Y, Toida T, Kawashima T, Hashimoto Y. 1995. Analysis
of antitumor properties of effector cells stimulated with a cell wall preparation (WPG) of Bidobacterium
infantis. Biol Pharm Bull 18:148153.
Shackelford LA, Rao DR, Chawan CB, Pulusani SR. 1983. Effect of feeding fermented milk on the incidence
of chemically-induced colon tumours in rats. Nutr Cancer 5:159164.
Shahani KM, Ayebo AD. 1980. Role of dietary lactobacilli in gastrointestinal microecology. Am J Clin Nutr
33:24482457.
Sies H. 1991. Oxidative stress: Introduction. In: Sies H, editor. Oxidative stress: Oxidants and antioxidants.
San Diego, CA: Academic Press. pp 1522.
Singh J, Rivenson A, Tomita M, Shimamura S, Ishibashi N, Reddy BS. 1997. Bidobacterium longum, a lactic
acid-producing intestinal bacterium inhibits colon cancer and modulates the intermediate biomarkers of
colon carcinogenesis. Carcinogenesis 18:833841.
Souza MF, Tome AR, Rao VS. 1999. Inhibition by the bioavonoid ternatin of aatoxin B1-induced lipid
peroxidation in rat liver. J Pharm Pharmacol 51:125129.
Stoner GD, Mukhtar H, 1995. Polyphenols as cancer chemopreventive agents. J Cell Biochem 22:169180.
Szarka CE, Pfeiffer GR, Hum ST, Everley LC, Balshem AM, Moore DF, Litwin S, Goosenberg EB, Frucht
H, Engstrom PF. 1995. Glutathione S-transferase activity and glutathione S-transferase mu expression in
subjects with risk for colorectal cancer. Cancer Res 55:27892793.
Takagi A, Matsuzaki T, Sato M, Nomoto K, Morotomi M, Yokokura T. 1999. Inhibitory effect of oral
administration of Lactobacillus casei on 3-methylcholanthrene-induced carcinogenesis in mice. Med
Microbiol Immunol (Berl) 188:111116.
Takagi A, Matsuzaki T, Sato M, Nomoto K, Morotomi M, Yokokura T. 2001. Enhancement of natural killer
cytotoxicity delayed murine carcinogenesis by a probiotic microorganism. Carcinogenesis 22:599605.
Cancer-preventing attributes of probiotics 495
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
Takagi A, Ikemura H, Matsuzaki T, Sato M, Nomoto K, Morotomi M, Yokokura T. 2008. Relationship
between the in-vitro response of dendritic cells to Lactobacillus and prevention of tumorigenesis in the
mouse. J Gastroenterol 43:661669.
Takeshi M, Akimitsu T, Haruo I, Tetsuya M, Teruo Y. 2007. Intestinal microbiota: Probiotics and
autoimmunity. J Nutr 137:798S802S.
Tavan E, Cayuela C, Antoine JM, Trugnan G, Chaugier C, Cassand P. 2002. Effects of dairy products on
heterocyclic aromatic amine-induced rat colon carcinogenesis. Carcinogenesis 23:477483.
Thyagaraja N, Hosono A. 1993. Antimutagenicity of lactic acid bacteria from Idlyagainst food related
mutagens. J Food Protect 56:10611066.
Trush MA, Kensler TW. 1991. An overview of the relationship between oxidative stress and chemical
carcinogenesis. Free Radic Biol Med 10:201209.
Usman HA. 1998. Desmutagenicity of milk cultured with Lactobacillus acidophilus strains against mutagenic
heated tauco. Food Chem Toxicol 36:805810.
Vanderhoof JA. 2001. Probiotics: Future directions. Am J Clin Nutr 73:1152S1155S.
Vuillaume M. 1987. Reduced oxygen species, mutation, induction and cancer initiation. Mutat Res
186:4372.
Wollowski I, Rechkemmer G, Pool-Zobel BL. 2001. Protective role of probiotics and prebiotics in colon
cancer. Am J Clin Nutr 73:451S455S.
Yokokura T, Kato I, Mutai M. 1981. Antitumor effect of Lactobacillus casei (LC 9018). In: Mitsuoka T,
editor. Intestinal ora and carcinogenesis. Tokyo: Gakkai-Syuppan Center. pp 7288.
496 M. Kumar et al.
Int J Food Sci Nutr Downloaded from informahealthcare.com by Massey University on 09/19/11
For personal use only.
Article
Full-text available
The ingestion of certain probiotics has been suggested as a promising nutritional strategy to improve aging. The objective of this work was to evaluate the effects of the daily intake, for a month, of a new probiotic Akkermansia muciniphila (AKK) (2 × 108 cfu/100µL PBS) on behavior, as well as function and redox state of immune cells of old female ICR-CD1 mice (OA group). For this, several behavioral tests were performed, and function and oxidative-inflammatory stress parameters of peritoneal leukocytes were analyzed in OA group, in a group of the same age that did not take AKK (old control, OC group) and in another adult control (AC) group. The results showed, in OA group, a significant improvement of several behavioral responses (coordination, balance, neuromuscular vigor, exploratory ability and anxiety like-behaviors), as well as in immune functions (chemotaxis, phagocytosis, NK activity and lymphoproliferation) and in oxidative stress parameters (glutathione peroxidase and reductase activities, oxidized glutathione and lipid oxidation concentrations) of the peritoneal leukocytes in comparison to those observed in OC group. In addition, peritoneal immune cells from the OA group released lower basal concentrations of pro-inflammatory cytokines (IL-2, IL-6 and TNF-α) compared to those from the OC group. The values of parameters in OA were similar to those in AC group. These improvements in the old mice receiving the probiotic were reflected in an increase in their lifespan. In conclusion, our data indicate that AKK supplementation for a short period could be a good nutritional strategy to promote healthy longevity.
Article
Full-text available
Purpose of Review The role and importance of biocatalysis in the production and modification of functional food and its ingredients have been discussed. The challenges in the field and future scope have been highlighted. Recent Findings The incorporation of advanced synthetic biology and computational tools with recent research developments in food biotechnology has led to a better understanding of the mechanism of health benefits of functional foods. Studies at the molecular level are still a potential area of future research. Summary The challenges in this area are limited knowledge about the structure and functionality of the enzymes and their reaction mechanism. Efforts should be directed towards the expansion of unrevealed aspects of the biocatalytic toolbox, to reduce the production and downstream operations costs and improve the low yields of the products. Alternative substrates with higher efficiency should be explored. Cutting-edge research is required in nanotechnology for the identification of food-grade, non-toxic nanoparticles for increasing the bioavailability of functional food ingredients for health benefits.
Article
Full-text available
It has been demonstrated that epigenetic modifications of histone (acetylation/deacetylation) participate in a critical role in cancer progression by the regulation of gene expression. Several processes could be regulated by deacetylation of histone and non-histone proteins such as apoptosis, proliferation, cell metabolism, differentiation, and DNA repair. Hence, histone deacetylase inhibitors (HDACis) are employed as a hopeful group of anti-cancer drugs that could inhibit tumor cell proliferation or apoptosis. The elimination of the acetylation marks that take place as an essential epigenetic change in cancer cells is associated to HDAC expression and activity. In this regard, it has been reported that class I HDACs have a vital role in the regulation of tumor cell proliferation. Objectives In this review, we discuss whether gut origin microorganisms could promote cancer or tumor resistance and explain mechanisms of these processes. Conclusions According to the enormous capacity of the metabolism of the intestine microbiota, bacteria are likely to convert nutrients and digestive compounds into metabolites that regulate epigenetic in cancer. The effect of the food is of interest on epigenetic changes in the intestinal mucosa and colonocytes, as misleading nucleotide methylation may be a prognostic marker for colorectal cancer (CRC). Since epigenetic changes are potentially reversible, they can serve as therapeutic targets for preventing CRC. However, various mechanisms have been identified in the field of prevention, treatment, and progression of cancer by probiotics, which include intestinal microbiota modulation, increased intestinal barrier function, degradation of potential carcinogens, protective effect on intestinal epithelial damage, and increased immune function.
Article
Full-text available
Apoptosis induction in cancer cells is one of the most efficient ways to treat cancer and find anticancer compounds. The aim of this study was to evaluate the cytotoxic effects of heat-killed indigenous probiotic bacteria and apoptosis induction in the HT-29 human colon adenocarcinoma cell line. The growth-inhibitory effects of probiotic heat-killed Lactobacillus brevis and Lactobacillus paracasei isolated from the traditional Iranian food “Terxine” on the HT-29 cell line were determined by 3-(4,5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide (MTT) assay. Flow cytometry by Annexin-FITC kit, DNA fragmentation assay, 4,6-diamidino-2-phenylindole staining, and the expression of Bax, Bcl2, caspase-3, and caspase-9 were used to analyze apoptosis. MTT results demonstrated that the heat-killed bacteria inhibited the proliferation of HT-29 cells and induced apoptosis in a time-, dose-, and strain-dependent manner. The results demonstrated that both bacteria could induce apoptosis in the HT-29 cell line. Heat-killed probiotic bacteria increased the expression of Bax, caspase-3, and caspase-9 mRNA levels in HT-29 cell lines. Also, heat-killed probiotic bacteria reduced the expression of Bcl2 in HT-29 cells. The heat-killed probiotic bacteria in this study exhibited potent growth inhibitory effects on cancer cells. The results demonstrated that L. brevis has a greater ability to inhibit the growth of HT-29 cells and induce apoptosis, compared with L. paracasei. It is proposed that these bacteria can be used as biological products for the treatment and prevention of cancer, pending further investigation.
Article
Full-text available
Breast cancer is one of the most important causes of cancer related morbidity and mortality in the world. Along with genetic, environmental factors also play a multifaceted role in the development of disease. Breast contains several bacterial species performing specialized functions. Probiotics, as functional food, play pivotal role against breast cancer development in vivo and in vitro. Current review summarized all the available data related to diet, probiotics, and their association with breast cancer risk along with underlying mechanisms. Presently, it was believed that many of the commercially available probiotic products were safe to use and had some beneficial health effects for the host. Probiotics had a potential to act against breast cancer progression evidenced by many animal model and cell-based experiments. Some probiotics strains may be useful as an adjuvant therapy for breast cancer prevention or treatment, by modulating immune response or breast microbial community. However, large-scale clinical trials and intense research are mandatory to explore probiotics-related metabolic and molecular mechanisms in breast cancer.
Article
Full-text available
Managing arsenic intoxication with conventional metal chelators is a global challenge. The present study demonstrated the therapeutic role of probiotics against arsenic-induced oxidative stress and female reproductive dysfunction. Sodium arsenite-treated (1.0 mg/100 g body weight) Wistar female rats were followed up by a post-treatment of commercially available probiotic mixture in powder form (0.25 mg/100 g body weight) orally. Rats that experienced arsenic ingestion showed a significant lessening in the activities of uterine superoxide dismutase (SOD), catalase activities, and the level of non-protein soluble thiol (NPSH) with a concomitant increase in malondialdehyde (MDA) and conjugated dienes (CD). Exposure to arsenic significantly lowered the levels of vitamin B12 and estradiol. Exposure to arsenic highly expressed the inflammatory marker and transcription factor NF-κB. Arsenic-mediated instability of these above parameters was controlled by the probiotics with a rebuilding of better function of anti-oxidant components. Besides its function in regulating endogenous anti-oxidant system, probiotics were able to augment the protection against mutagenic uterine DNA-breakage, necrosis, and ovarian-uterine tissue damages in arsenicated rats.
Article
Full-text available
Previously, we reported that Lactobacillus gasseri BNR17 (BNR17), a probiotic strain isolated from human breast milk, inhibited increases in body weight and adipocyte tissue weight in high-sucrose diet-fed Sprague-Dawley (SD) rats and reduced glucose levels in type 2 diabetes mice. In the current study, we conducted further experiments to extend these observations and elucidate the mechanism involved. C57BL/6J mice received a normal diet, high-sucrose diet or high-sucrose diet containing L. gasseri BNR17 (10(9) or 10(10) CFU) for 10 weeks. The administration of L. gasseri BNR17 significantly reduced the body weight and white adipose tissue weight regardless of the dose administered. In BNR17-fed groups, mRNA levels of fatty acid oxidation-related genes (ACO, CPT1, PPARα, PPARδ) were significantly higher and those of fatty acid synthesis-related genes (SREBP-1c, ACC) were lower compared to the high-sucrose-diet group. The expression of GLUT4, main glucose transporter-4, was elevated in BNR17-fed groups. L. gasseri BNR17 also reduced the levels of leptin and insulin in serum. These results suggest that the anti-obesity actions of L. gasseri BNR17 can be attributed to elevated expression of fatty acid oxidation-related genes and reduced levels of leptin. Additionally, data suggested the anti-diabetes activity of L. gasseri BNR17 may be to due elevated GLUT4 and reduced insulin levels.
Article
Four probiotic bacteria (Lactobacillus paracasei L26, L. casei-01, L. acidophilus Ki, and Bifidobacterium animalis BB-12®) were encapsulated in plain alginate or alginate supplemented with L-cysteine·HCl, and resulting microcapsules were stored at different temperatures, namely 21, 4, −20, or −80°C for a period of up to 6 months. The results showed that the encapsulation in calcium alginate microcapsules was only effective in promoting protection at freezing temperatures, independently of the sensitivity of the strain. Storage of calcium alginate microcapsules at −80°C indicated a protective effect upon viability of all four probiotic strains and the presence of L-cysteine·HCl in the alginate matrix improved protection upon cell viability of B. animalis BB-12®. An increase in storage temperature of encapsulated bacteria caused an increase in rate of loss in their viability that was strain dependent. This study suggests that microencapsulation of probiotic cells in calcium alginate can be suitable for sustaining the viability of probiotics in food products that require storage below freezing temperatures, even in the absence of cryoprotectors, contributing to an increased shelf life.
Article
Full-text available
Data from literature suggest the possible use of probiotics as chemopreventive agents against colon cancer, but few investigations are available on their effects on gastric cancer proliferation. In our previous study, a specific Lactobacillus, strain L. paracasei IMPC2.1, was demonstrated to colonize the human gut and positively affect fecal bacteria and biochemical parameters. The aims of the present study were to investigate the effects of L. paracasei IMPC2.1, comparing them with those of Lactobacillus rhamnosus GG (L.GG), either as viable or heat-killed cells, on cell proliferation and apoptosis in a gastric cancer (HGC-27) and a colorectal cancer cell line (DLD-1). Both the gastric and colon cancer cells were sensitive to the growth inhibition and apoptosis induction by both viable or heat-killed cells from L. paracasei IMPC2.1 and L.GG. These findings suggest the possibility for a food supplement, based on dead probiotics, including L. paracasei IMPC2.1 cells, which could represent an effective component of a functional food strategy for cancer growth inhibition, with potential for cancer prevention.
Article
Full-text available
Cardiovascular diseases are one of the major causes of deaths in adults in the western world. Elevated levels of certain blood lipids have been reported to be the principal cause of cardiovascular disease and other disabilities in developed countries. Several animal and clinical trials have shown a positive association between cholesterol levels and the risks of coronary heart disease. Current dietary strategies for the prevention of cardiovascular disease advocate adherence to low-fat/low-saturated-fat diets. Although there is no doubt that, in experimental conditions, low-fat diets offer an effective means of reducing blood cholesterol concentrations on a population basis, these appear to be less effective, largely due to poor compliance, attributed to low palatability and acceptability of these diets to the consumers. Due to the low consumer compliance, attempts have been made to identify other dietary components that can reduce blood cholesterol levels. Supplementation of diet with fermented dairy products or lactic acid bacteria containing dairy products has shown the potential to reduce serum cholesterol levels. Various approaches have been used to alleviate this issue, including the use of probiotics, especially Bifidobacterium spp. and Lactobacillus spp.. Probiotics, the living microorganisms that confer health benefits on the host when administered in adequate amounts, have received much attention on their proclaimed health benefits which include improvement in lactose intolerance, increase in natural resistance to infectious disease in gastrointestinal tract, suppression of cancer, antidiabetic, reduction in serum cholesterol level, and improved digestion. In addition, there are numerous reports on cholesterol removal ability of probiotics and their hypocholesterolemic effects. Several possible mechanisms for cholesterol removal by probiotics are assimilation of cholesterol by growing cells, binding of cholesterol to cellular surface, incorporation of cholesterol into the cellular membrane, deconjugation of bile via bile salt hydrolase, coprecipitation of cholesterol with deconjugated bile, binding action of bile by fibre, and production of short-chain fatty acids by oligosaccharides. The present paper reviews the mechanisms of action of anti-cholesterolemic potential of probiotic microorganisms and probiotic food products, with the aim of lowering the risks of cardiovascular and coronary heart diseases.
Article
Full-text available
Lactic acid bacteria from “Idly”, a traditional cereal pulse product of southern India, were evaluated for antimutagenic properties. Effect of their presence in Salmonella mutagenicity assay with foodborne mutagens like spice extracts, amino acid pyrolysates, and aflatoxins was examined. Variation of antimutagenic potential at different growth stages of these lactic acid bacteria was examined in time-course studies. The enzyme profile was examined to determine any possible relationship between antimutagenicity and enzymes in these lactic acid bacteria. Most of the lactic acid bacteria tested were found to decrease mutagenicities exerted by these mutagens significantly. Time-course studies showed that antimutagenic ability decreased in stationary phase of growth of lactic acid bacteria. There was no correlation between antimutagenicity and enzyme profile quantifying proteolytic, lipolytic, and other enzymes of carbohydrate metabolism. Lactic acid bacteria from “Idly” were found to be effective in suppressi...
Article
Full-text available
Consumption of Lactobacillus products and supplements containing viable lactic acid bacteria results in their establishment in the gastrointestinal tract. Their presence in the intestinal tract has been suggested to be prophylactic. They may reduce risk associated with dietary onset of carcinogenesis directly due to the reduction of procarcinogenic substances or indirectly due to the reduction in the level of enzymes that convert procarcinogens to carcinogens. In short-term studies, feeding as well as intraperitoneal administration of lactic acid bacteria suppress tumors implanted in rodents. The suppression is probably mediated by lactic acid bacteria through activation of host defense system. The activation of host defense system has been hypothesized to augment the immune system. Transient increase in the host immune defense system has been observed following feeding of cultured dairy products, but the biological significance of these changes has not yet been elucidated.
Article
The possible protective effects of lactic acid bacteria (LAB) against in vivo genotoxicity were assessed in mice. Culture broth containing Lactobacillus casei was orally administered to mice either 1 h before intake of the genotoxins, cyclophosphamide (CPH), procarbazine (PCB) and urethane (URE) or together with PCB. In addition, an experiment was carried out in which heat-inactivated L. casei was orally administered to mice 1 h before PCB. Control animals received cell-free culture broth. Genotoxic effects were assessed in the mouse bone-marrow micronucleus test. The results suggest that pretreatment of mice with L. casei 1 h before the administration of genotoxins can lead to a moderate reduction (P < 0.05) in the genotoxicity of PCB and URE, but not CPH. However, co-administration of PCB with L. casei did not show any significant effect on genotoxicity. Similarly, no significant change in genotoxicity was observed after administration of heat-inactivated LAB. From this investigation, there is limited evidence for the protective effect of LAB against in vivo genotoxicity.
Article
The aim of the present paper is to review the authors' studies on the influence of yogurt and yogurt bacteria on immune responses in man and animals. Lactic acid bacteria present in yogurt play a role in modulating the translocation of the Gram-negative bacteria present in the gut, increase the survival of mice challenged with Salmonella typhimurium and stimulate local immune responses at the level of Peyer's patches. In man, yogurt modulates gamma-interferon production in vitro and in vivo. The presence of membrane receptors for LAB on human lymphocytes probably represents a potent stimulus for lymphoid-cell activation.
Article
The morbidity and mortality of colon cancer are increasing, because of the westernization of food habit. Probiotics such as lactic acid bacteria (LAB) have been known to play an important role in retarding colon carcinogenesis by possibly influencing metabolic, immunologic, and protective functions in the colon. In this study, we evaluated the effect of B. polyfermenticus SCD on N-methyl-N′-nitro-N-nitrosoguanidine (MNNG) induced DNA damage in CHO-K1 cells and human lymphocytes, and on proliferation of human colon cancer cell. Using the Comet assay to detect DNA damage, we found that B. polyfermenticus SCD protected cells from the DNA damage induced by MNNG in CHO-K1 cells and in human lymphocytes. B. polyfermenticus SCD was also found to inhibit We growth of colon cancer cells in a dose-dependent manner, detected by the MTT assay. These results indicate that B. polyfermenticus SCD has the potential to inhibit not only DNA damage induced by a carcinogen, but also the proliferation of colon cancer cells.
Article
Article
Bifidobacteria are harbored in the intestine of specific animals and humans at different ages. In this study, we examined the immune responses affected by Bifidobacterium longum and B. animalis, focusing upon the cytokines produced by mouse peritoneal cells. Both B. longum and B. animalis induced the expression of IL-1β, IL-6, IL-10 and TNF-α mRNA in mouse peritoneal cells, as determined by RTPCR. The amount of IL-6 secreted by thioglycollate-induced peritoneal exudate cells (TG-PEC) in the presence of these bacteria were similarly augmented, as shown by ELISA. More TNF-α was induced in B. longum than in B. animalis. A difference in the level of TNF production was also suggested from the in vivo tumor suppression test using TNF-sensitive Meth A tumors, although both species of bacteria profoundly suppressed the tumor incidence and tumor growth. Taken together, both B. longum and B. animalis promote the induction and/or secretion of inflammatory cytokines (IL-6 and TNF-α) in mouse peritoneal cells. However, the level of TNF-α production induced by B. animalis was lower than that by B. longum.