ArticlePDF Available

Making a Niche as Promising Health Supplements and Complementary Medicines

Authors:
  • Rayat bahra institute of pharmacy

Abstract and Figures

Hippocrates once quoted, "Let food be thy medicine and medicine be thy food,‰ this ancient statement has certainly become a belief in todayÊs scenario. The live microbial food supplements, called as Probiotics (known to have beneficial effects on human health in various ways) are widely used now a days. Owing to their potential as alternative or complementary therapeutic agent in combating large number of pathological conditions and ability to enhance immune response, an overwhelming interest have developed for probiotics in medical field. Probiotics, being widely used around the globe by consumers and in clinical practice, a thorough understanding of the risks and benefits related to their use are of vital importance. This review addresses the concept of probiotics, historical development, their sources and probable mechanisms of action, clinical applications along with associated risks and contraindications related to the use of probiotics.
Content may be subject to copyright.
International Journal of Drug Delivery 6 (2014) 329-338
http://www.arjournals.org/index.php/ijdd/index
Review Article
Making a Niche as Promising Health Supplements and Complementary
Medicines
Charanjit Kaur1, Amit Sharma1, Ashish K Garg2, Junaid Niazi3, Rajesh Kumar*1
*Corresponding author:
Rajesh Kumar
1Rayat-Bahra Institute of Pharmacy,
Hoshiarpur, Punjab (India)
2Akal College of Pharmacy, Mastuana
Sahib, Sangrur, Punjab (India)
3Bahra Institute of Pharmacy, Patiala-
Sangrur Highway, Patiala, Punjab
(India)
Abstract
Hippocrates once quoted, „Let food be thy medicine and medicine be thy food,‰ this ancient
statement has certainly become a belief in todayÊs scenario. The live microbial food supplements,
called as Probiotics (known to have beneficial effects on human health in various ways) are widely
used now a days. Owing to their potential as alternative or complementary therapeutic agent in
combating large number of pathological conditions and ability to enhance immune response, an
overwhelming interest have developed for probiotics in medical field. Probiotics, being widely used
around the globe by consumers and in clinical practice, a thorough understanding of the risks and
benefits related to their use are of vital importance. This review addresses the concept of probiotics,
historical development, their sources and probable mechanisms of action, clinical applications along
with associated risks and contraindications related to the use of probiotics.
Keywords: Health, GIT, microflora, infants, immunity.
.
Introduction
The human gastrointestinal tract has long been considered for their
main functions of digestion and absorption of nutrients and
excretion of their waste products. However, GIT also fulfills many
other functions which are essential to our sound health. GI mucosa
poses a barrier, excluding and eliminating various antigens
entering the GIT from external environment. [1,2]
The human GIT has got a vast number of microbes and harbors,
many of which live in symbiotic relationship with host. This mutual
beneficial relationship is termed as host-microbes cross-talk. [3,4].
However few of these bacteria also contribute to the development
of various diseases by a variety of mechanisms [5,6]. Disturbance
in the existing balance between host and friendly microbes can
lead to various GI complications by stimulating the host immune
system. A few of methods by which intestinal microflora can be
altered are: administration of antibiotics, dietary supplements
which enhance physiological functions and thus growth of health
friendly bacteria (prebiotics, which act an alternative for probiotics
or their cofactors e.g. lactulose, galactooligosaccharides, inulin and
its hydrolysates etc.) and probiotics. [7]
History
Microbial cultures have been a niche in food and alcoholic
fermentations and for the past few decades have undergone great
scientific scrutiny for their ability to cure a variety of diseases. A
Nobel laureate Russian biologist, also known as father of modern
immunology, Dr. Ellie Metchnikoff in the year 1908 postulated the
prolongation of life with the consumption of large quantities of
cultured food especially yoghurt on the basis of growth of lactic
acid bacteria which could lead to displacement of the disease
causing microbes in the intestine [8]. In 1915, cultured food therapy
was also found to be beneficial in urogenital infections [9-11].
However, this therapy was ignored over 7-8 decades due to
growing interest in antibiotics. In 1965, Lilly and Stillwell coined the
term "probiotic" for the substances secreted by one organism
which stimulate the growth of another. The term probiotics was
derived from Greek word meaning "for life". Marteul
et al.
in 2002,
redefined them as "microbial preparations or components of
microbial cells that have a beneficial effect on health and well
being" [12-13]. WHO defines probiotics as "live microorganisms"
which when administered in adequate amounts confer a health
benefit on the host [14-15]. A breakthrough came in the field of
probiotics with the evolution of friendly bacteria such as
lactobacilli,
bifidobacteria
in various dosage forms like powders, tablets, liquid
suspensions and lyophilized powder filled capsules [16].
Source of Probiotics
Approximately 500 different microbial species have been reported
in the GIT amongst which 20 genera predominate, a few of which
are namely Bacteroides, Lactobacillus, Clostridium,
Fusobacterium, Bifidobacterium, Eubacterium, Peptococcus,
Peptostreptococcus, Escherichia, and Veillonella. Out of these,
bifidobacteria and lactobacilli were reported to have beneficial
properties [17-19]. In the neonates, the microbiota starts
developing immediately after the birth and is dependant mainly on
the maternal microbiota, mode of delivery, birth environment and
ISSN: 0975-0215
This work is licensed under a Creative Commons Attribution 3.0 License.
Kaur
et al
. International Journal of Drug Delivery 6 (4) 329-338 [2014]
PAGE | 330 |
sometimes on genetic factors [20-21]. The vaginal and intestinal
flora of mother constitutes the source of bacteria by colonizing the
intestine of newborn dominating strains being facultative
anaerobes such as enterobacteria, lactobacilli and coliforms. Once
the child starts taking stuff other than mother's milk, the
composition of intestinal microflora gradually start altering to
resemble to that of adult's microflora [1].
Criteria for being Probiotic
For an organism to be considered as probiotic, it should possess
the following properties:
It should be host friendly
It should not have pathogenic and toxic potential
It should be able to survive through the entire length of GIT
It must retain its viability for long periods during storage
The source species should be same as that of intended host
It must have capacity to influence local metabolic activity
It must be able to persist in the intestine even in the absence of gut
colonization by probiotic strain [1,3,13]
Table 1. List of Microorganisms used as probiotics [3,7,13]
Lactobacillus
species
Bifidobacterium
species
Streptococcus
species
Saccharomyces
species
Other
s
L.acidophilus,
L.plantarum,
L.casei
L.salivarius
L.brevis
L.fermentum
L.helveticum
L.delbruekii
L.bulgaricus
L.gasseri
L.Reuteri
L.rhamnosus
L. johnsonii
B.bifidum
B.longum
B.infantis
B.breve
B.lactis
B.adolescentis
S.thermophilu
s
S.salivarius
S.boulardi
i
Lactococcous lacti
s
Bacillus cereus
Clostridium butyricum
C. cremoris
Enterococcus faecium
Eschericia coli
Lactis pedicoccus
acidilactici,
Propionibacterium
freudenreichii.
Clinically beneficial effects of Probiotics
Kaur
et al
. International Journal of Drug Delivery 6 (4) 329-338 [2014]
PAGE | 331 |
Table 2. The clinically beneficial effects of probotics are listed below.
Intestinal effects
Promotes recovery from diarrhea induced by rotavirus and antibiotics
Produces lactase and thus alleviates symptoms of lactose intolerance and mal absorption
Helps in relieving constipation
Beneficial in the treatment of colitis
Immune system effects
Probiotics enhance immune response (both specific and non-specific)
They reduce the chances of infection from pathogenic microorganisms like Salmonella and Shigella
They are helpful in inhibiting pathogenic growth and translocation
Probiotics also stimulate gastrointestinal immunity
Other effects
Detoxification of carcinogens and thus reduced risks of colon and bladder cancer
Suppression of tumors
Lowering of serum cholesterol level
Treatment of food allergies
Enhances nutrient bioavailability
Improves urogenital health
Reduces blood pressure
Synthesis of nutrients like vitamin B2, B3, B6
,
B9 and B12
Mechanism of action
Probiotics act in several ways to protect the host organism.
Competitive inhibition:
Probiotics competitively inhibit the binding of pathogens to the
enteric epithelial surface8. A good example of this mechanism is
inhibition of adhesion and invasion of (enteroinvasion)
E.coli
by
streptococcus
thermophilus
and
lactobacillus acidophilus
in human
intestinal cell lines [23].
Competitive consumption of nutrients:
Probiotics prevents the growth of pathogens by competitively
consuming the nutrients and thus not allowing the pathogens to
Kaur
et al
. International Journal of Drug Delivery 6 (4) 329-338 [2014]
PAGE | 332 |
grow. Probiotics, by consuming the monosaccharides, prevent the
growth and development of
Clostridium difficile
[24].
Working through reduction of luminal pH
Several investigations have shown that probiotics like lactobacillus
exert antagonistic action against intestinal pathogens and related
organisms. Organic acid such as acetic acid and lactic acid are the
byproducts of bacterial metabolism. These acids inhibit the growth
of many pathogens including pathogenic gram negative types [25]
and lower pH value also potential the activity of these acids
because undissociated forms are more bacterial than the
dissociated ones [26].
Through stimulation of immune response to pathogens
Probiotics are shown to promote the endogenous host defense
mechanism which is characterized by stabilization of the gut
microflora [27].
a) Probiotic bacteria have been shown to enhance humoral
immune responses and thereby promote the intestineÊs
immunological barrier [28,29].
b) Probiotic bacteria have been shown to stimulate non-specific
host resistance (proinflammatory cytokines TNF and IL6) to
microbial pathogens and thereby facilitate the exclusion of
pathogens in the gut [30,31].
c) Probiotics modulate the hostÊs immune responses to potentially
harmful antigens with potential to down regulate hypersensitivity
reactions [32,33].
d) Probiotics exhibit protective mechanism by association with gut
intestinal colonization suppression of pathogens growth and/or
invasion
e) Probiotics enhance and modulate innate and adaptive immune
responses in the host.
f) Probiotics modulate the immune activity and epithelial function in
both large as well as small intestine [34].
g) Oral intake of probiotics such as
Lactobacillus casei
and
Lactobacillus bulgaricus
activate the production of macrophages
and administration of
L.casei
and
L. acidophilus
activates
phagocytosis [35].
h) Specific strains of probiotics such as
Bifidobacterium
and
Lactobacillus
give promising results in the treatment and/or
prevention of eczema in infants and children by alteration of
colonization [34].
Integrity of GI mucosa
Probiotics (
Streptococcus faecalis
and
Clostridium butyricum
), by
the synthesis of glutamine in presence of ammonia and glutamic
acid, maintain integrity of GI mucosa. Therefore, it is postulated
that the probiotics administered to humans can have positive effect
in cases such as diarrhea, constipation, colitis, and recolonization
by pathogens, immunostimulation [36].
By producing inhibitory substances
Probiotic bacteria produce a variety of inhibitory substances
include organic acids, hydrogen peroxide and bacteriocins which
are inhibitory to both gram positive and gram negative bacteria. It
may reduce not only the number of viable cells but may also affect
bacterial metabolism or toxin production [37].
Degradation of toxin receptor
By the degradation of the toxin receptor on the intestinal mucosa,
probiotics
(e.g. Saccharomyces boulardii)
protects the intestine
from disease causing microbes (e.g
C.difficile
) [25,38]
Clinical Applications
Role of probiotics on faecal water-induced DNA
damage in human colon adenocarcinoma cells
Probiotics may have protective effects against the early stages of
colon cancer, although there are many studies in laboratory
animals indicating that administration of probiotics and/or prebiotics
reduces the incidence of tumours and precancerous lesions in the
colon [39-44], but there was no evidence clinically. A recent study
on human volunteers has suggested that in comparison to
conventional yoghurt, the strains of
Lactobacillus
acidophilus
and
Bif. longum
has demonstrated a reduction in faecal water
genotoxicity [45]. Thus,
Lact. plantarum
and
Bif. Bb12
have
intrinsic antigenotoxic potential and probiotics certainly are worthy
of further evaluation in
in vivo
assays for anti carcinogenic effects.
Probiotics in infants
One of the fatal intestinal disorder; necrotizing enterocolitis, which
is known to occur in 10-25% of premature infants and very low birth
weight babies and has a high mortality of 20-30%1. Bacterial
colonization or intestinal infection aggravates the risks of
necrotizing enterocolitis. Modification of the intestinal flora by
increasing the predominance of specific nonpathogenic bacteria
would seem a reasonable means of attaining a prophylactic or
therapeutic effect against enteropathogens. A combination of
increased bifidobacterial counts and decreased concentrations of
other enterobacteria and luminal host factors may play a role in
protecting premature babies and newborns from enterocolitis.
Trials showing reduction of necrotizing enterocolitis in population of
premature newborns given supplements of Lactobacillus GG daily
compared with historical control subjects have been reported.
These findings suggested a correlation between the reduction of
lactobacilli and the increased risk of necrotizing enterocolitis
[46,47].
Wang Y
et al.
conducted a double blind randomized controlled trial
on 100 full term infants with critical illness according to scores of
neonatal acute physiology. Out of them, 50 infants received
Probiotics three times a day after birth for 8 days and remaining 50
were kept on placebo. The incidence of sepsis, multiple organ
dysfunction syndrome (MODS), nosocomial pneumonia and
necrotizing enterocolitis were recorded. Prognosis of probiotic
treatmenr was determined on the basis of rate of recovery and
hospitalization days. Serum IgA, IgG and IgM concentrations were
measured on days 4 and 8. Infants receiving Probiotics showed
significantly reduced rate of nosocomial pneumonia and MODS
(due to increase in the level of IgA) compared with the placebo
group of infants [48].
Kaur
et al
. International Journal of Drug Delivery 6 (4) 329-338 [2014]
PAGE | 333 |
Role of Probiotics on Cold and Influenza-Like
Symptoms, Incidence and Duration in Children
Daily dietary supplementation of probiotics during the winter
months was a safe effective way to reduce episodes of fever,
rhinorrhea, and cough, the cumulative duration of those symptoms,
the incidence of antibiotic prescriptions.
L acidophilus
alone was
effective. There was a broader protective effect with the
combination of
L acidophilus
and
B lactis
Bi-07 [15].
Probiotics in pregnancy
Probiotics have been shown to decrease the risks of bacterial
vaginosis (which has been suggested as a factor that increases the
risk of preterm labour and infant mortality) and maintain normal
lactobacilli vaginal flora [49,50]. These strains were found to be
safe in pregnant animal studies and improving the health of
mothers and newborns [51].
Prevention of allergic reactions by probiotics taken during
pregnancy is another area of interest. Ingestion of probiotics like L
rhamnosus GG and B lactis BB12 during pregnancy and in
newborns during first 6 months of birth was found to prevent atopic
dermatitis (a condition that causes severe skin rashes in up to 15%
of babies) in 50% cases [52].
Role of Probiotics in Lowering of serum cholesterol level
in hypercholesterolemia
Lactobacillus acidophilus,
is
the natural
inhabitant of intestine and
possesses bile-salt
hydrolase activity and possibly can be used for
the
manufacture of acidophilus milk and thus recommended as a
source for reducing cholesterol
level. Anderson and Gilliland
et al.
(1985) in their study reported that
Lactobacillus acidophilus
reduces the blood cholesterol by direct breakdown of cholesterol
and deconjugation of bile salt. In the year 1999, they also
examined effects of consumption of one daily serving of yogurt on
serum lipids in two controlled clinical studies and in those studies it
was concluded that regular intake of fermented milks containing an
appropriate strain of
L. acidophilus
has the potential of reducing
risk for coronary heart disease by 6 to 10%.
L.
acidophilus
showed
highest deconjugation ability and BSH activity towards bile
mixtures that resembled the human bile in a study done by Liong
and Shah in which they screened eleven strains of lactobacilli and
analyzed bile salt deconjugation ability, bile salt hydrolase activity
(BSH) and co-precipitation of cholesterol with deconjugated bile.
Lactobacillus acidophilus
strains had higher deconjugation ability
than
L. casei
strains. Cholesterol co-precipitation with
deconjugated bile increased with decreasing pH [53].
Lactobacillus bulgaricus
is an acid-producing bacterium and in
general, it occurs in dairy products and some plant products. A
study conducted on fifty four volunteers in a randomised cross over
trial; revealed reductions of between 5-10% in serum cholesterol
levels after several weeks of moderate consumption of yoghurt
fermented with
Lactobacillus bulgaricus
and
S.thermophilus
[54].
Use of
Bacillus coagulans
(Probiotics
)
as an attractive
alternative to drug therapy in case of
hypercholesterolemia
Bacillus coagulans,
formerly known as
Lactobacillus Sporogenes,
is a shelf stable, nonpathogenic Gram positive spore-forming
bacteria that produces L(+) lactic acid (dextrorotatory) in
homofermentation conditions. In particular,
B. coagulans
strains
were used to reduce serum cholesterol in certain formulations
(Mohan, 1990). Seok, (1987) concluded that
L.sporogenes
lowers
LDL cholesterol by eliminating it directly from inside the intestines
before it can be absorbed into the blood stream. In another clinical
study,
L. sporogenes
not only lowered total serum cholesterol and
LDL cholesterol in humans, it also improved the ratio of „good‰
HDL cholesterol to total cholesterol. US patent was given to a
therapeutic composition including
Bacillus coagulans
, in
combination with bifidogenic oligosaccharides or other cholesterol-
reducing agents for use in reducing LDL cholesterol and serum
triglycerides (US patent 7232571). This unique microorganism
proved effective in lowering cholesterol by 104 points in a three-
month study performed at the G.B. Pant hospital in New Delhi,
India. There was a highly significant reduction in the LDL
cholesterol levels, and a small but significant increase in HDL
cholesterol levels [55].
products without prescription. Lack of awareness about the risk
factors associated with probiotics needs regulation.
Generally probiotics are safe however some rarely observed side
effects associated with probiotics vary from strain to strain
individually such as bacteremia or sepsis from lactobacilli [28],
fungemia with saccharomyces in immune compromised patients or
ICU patients [60,61].
Some cases of liver abcess [62],
Lactobacillemia
[63,64] and
S.
boulardii
fungemia [65,66] have been reported in such patients. On
the other hand, S.boulardii has been used without complications to
treat chronic diarrhea in AIDS patients [67,68].
Another reason of concern is the risk of transfer of resistance via
probiotic agents as these agents have to be resistant to the
antibiotics because they are to be used alongside. In fact, a study
has demonstrated transfer of plasmid pAMb1, from
L. reutri
to
E.
faecium
and from
E. faecium
to
E. Faecalis
in mouse intestinal
tract [69].
In long term study, in children for 1 year, consumption of 106-109
CFU of Lactobacilli and Bifidobacteria daily did not only result in
any adverse effect but also showed that Probiotics taken over a
long time improve gastrointestinal function and reduce the
incidence of diaper rash [70].
Many lactobacillus strains are resistant to vancomycin naturally;
however, it has been revealed in conjugation studies that these
resistant genes are non-transferrrable [54]
Kaur
et al
. International Journal of Drug Delivery 6 (4) 329-338 [2014]
PAGE | 334 |
Table 2. Probiotics associated fungal sepsis cases in human beings
Note:
CVC- central venous catheter COPD- chronic obstructive pulmonary disease GI- gastrointestinal 250 mg S.boulardii= 5.425 X 1013 live cells
Kaur
et al
. International Journal of Drug Delivery 6 (4) 329-338 [2014]
PAGE | 335 |
Table 3. Probiotics associated risks and contraindications
Possible risk factors of Probiotic therapy
Sr. No. Major risk factors
1. Immune compromisation resulting in debilitated state or malignancy
2. Premature infants
Minor risk factors
1. Central venous catheter
2. Diarrhea and intestinal inflammation due to impairment of intestinal epithelial barrier
3. Administration of probiotics by jejunostomy
4. Co-administration of probiotic resistant antibiotics
5. Probiotics with known pathogenic potential
Problems associated with the use of Probiotics
Viability of Probiotics
The maintenance of viability associated with use of probiotics is a
major issue. The Probiotics need to survive in several
environments including processing treatments, storage conditions
and human body conditions especially gastric acidity and action of
bile salts. The ability to survive through stressful acidic conditions
and bile solutions in the body vary among the strains of probiotic
bacteria.
Only those Strains which tolerate these adverse conditions better,
should be preferred. The viability of Probiotics may also be
improved by using a medium, nutrient and temperature, which
supports their survival.
Viability of
Lactobacilli
has been enhanced by
Encapsulation in artificial sesame oil for storage and in simulated
high gastric or bile salt conditions as compared to free cells [71].
Storage at 4ĈC temperature is reported to be a most important
factor in keeping probiotic
Bifidobacterium
viable during 4 weeks
storage time [72].
The use of prebiotics like isomalto-oligosaccharides has been
found to be associated with higher levels of Probiotics like
Lactobaccilus
and
Bifidobacterium
after 1month storage [73].
The issue of viability of Probiotics from the stage of processing till it
is used by consumer to produce benefits still remains a fertile field
for more research [74].
Commercially available Probiotics (single and multiple
strains)
The optimal use of one or more strain cannot be precisely
determined. While it seems intuitive that a combination of strains
might be convenient to suite a range of indications and individual
variation, this is dependent on the optimal probiotic bacterial
numbers in different situations and assuming that the probiotic
constituent of any cocktail are not mutually antagonistic which
requires further study. Furthermore, the activities of individual
components require definitions and optimization before firm
recommendations can be made.
Single-strain-
Saccharomyces boulardi
(Laboratories Biocodex,
Montrouge, France) is commonly given in doses of 2 capsules
containing 250 mg morning and evening, equivalent to
approximately 10 billion live organisms/d. The nonpathogenic E coli
serotype O6:K5:H1 (Mutaflor;Ardeypharm GmbH, Herdecke,
Germany) is referred to as Nissle 1917 is commonly given in doses
of less than 10 billion LAB/d.
Lactobacillus
GG (LGG) (Valio,
Helsinki, Finland) commonly is given in doses of 1 to 5 billion
LAB/d.
L acidophilus
LA1 (LA1) (Nestle, Vevey, Switzerland) is
commonly given in doses of less than 5 billion LAB/d (sometimes
<1 billion LAB/d).
Multistrain- The probiotic cocktail VSL#3 (Sigma-Tau, Pomezia,
Italy, and VSL Pharmaceuticals, Fort Lauderdale, Florida) is the
only multistrain probiotic tried so far. It consists of four
Lactobacillus
strains (
L. acidophilus
,
L. casei
,
L. delbruecki
subsp
bulgaricus
, and
L. plantarum
), three
Bifidobacterium
strains (
B.
longum
,
B. infantis
,
B. breve
), and
S. salivarius
ssp
thermophilus
(5
1011 cells/g). It is commonly given in high doses,usually 1800
billion LAB and more recently up to 3600 billion LAB/d [1].
Conclusion
Probiotics are gaining popularity in consumers for their health
benefits and are recommended by many health care professionals
too. Prebiotic, probiotic and synbiotic treatment is still in its infancy
but is rapidly moving into the mainstream. Several probiotic
preparations seem to have promise in prevention or treatment of
various conditions. Normalization of the properties of unbalanced
indigenous microflora of the intestinal tract by ingestion of specific
Kaur
et al
. International Journal of Drug Delivery 6 (4) 329-338 [2014]
PAGE | 336 |
strains of the healthy microflora forms the rationale of probiotics
therapy. There exists quite a good number of evidences for the
therapeutic use of probiotics in infectious diarrhea in children,
recurrent Clostridium difficile induced infections and post-operative
pouchitis. Evidence is also emerging for the use of probiotics in
other gastrointestinal infections, irritable bowel syndrome and in
ulcerative colitis and CrohnÊs disease. However, most studies have
been in small number of patients and many have important
methodological limitations, making it difficult to make unequivocal
conclusions regarding efficacy, especially when compared with
proven therapies. Furthermore, considerable differences exist in
composition, doses and biologic activity between various
commercial preparations and one consistent feature is that not all
probiotic bacteria have similar therapeutic effects. The enthusiasm
for probiotics has perhaps outpaced scientific support for these
therapeutic approaches. They are generally regarded as safe, but
physicians should monitor their use in high-risk patients. However,
when used appropriately, probiotics represent a potentially
beneficial adjunct to other proven therapies and with more
controlled and larger studies clear data may emerge in the future.
References
[1]. Harish K. & Varghese T. Probiotics in
humans- evidence based review. Calicut
Med. J. 2006; 4: 1-11.
[2]. Sanderson IR, Walker WA. Uptake and
transport of macromolecules by the
intestine: possible role in clinical
diorders (an update). Gastroenterology
1993; 104: 622-39.
[3]. Eswara MS, Kavitha BTVV, Srikanth JG,
Velmani G. Probiotics as Potential
Therapies in Human Gastrointestinal
Health. Int. J. Advances Pharm. Sc.
2010; 1: 96-110.
[4]. Cebera JJ. Influences of microbiota on
intestinal immune system development.
Am J Clin Nutr. 1999; 69: 1046-51.
[5]. Berg RD. The indigenous
gastrointestinal microflora. Trends
Microbiol. 1996; 4: 430-5.
[6]. Guarner F, Malagelada. JR. Gut flora in
health and disease. Lancet, 2003; 361:
512-9.
[7]. Probiotics- Friendly Bacteria with a Host
of Benefits. Dairy Council of California.
2000; 1-4.
[8]. Agarwal RK. Probiotics- The Health
Friendly Gut Bacteria. Indian Pediatrics,
2008; 45: 953-954.
[9]. Anthony JB, Ian RR. Antigenotoxicity of
probiotics and prebiotics on faecal
water-induced DNA damage in human
colon adenocarcinoma cells. Mutation
Res. 2004, 551: 233ă243.
[10]. Guidelines for the Evaluation of
Probiotics in Food. Joint FAO/WHO
Working Group Report on Drafting
Guidelines for the Evaluation of
Probiotics in Food London, Ontario,
Canada, April 30 and May 1, 2002.
[11]. Rial DR. The Role of Probiotic Cultures
in the Control of Gastrointestinal Health.
J Nutr. 2000; 396S-402S.
[12]. Sharma AK, Mohan P, Nayak BB.
Probiotics: Making a comeback. Ind J
Pharmacol. 2005; 37: 358-365.
[13]. Gupta V, Garg R. Probiotics. Ind J Med
Microb. 2009; 27: 202-209.
[14]. Shendy MS & Nihal MS El-Assaly. The
Effect of the Use of Probiotics in Mildly
and Moderately Severe Ulcerative
Colitis. Res J Medicines Med Sc. 2008;
3: 92-99.
[15]. Gregory JL, Shuguang L, Mohamed EM,
Cheryl R, Arthur CO. Probiotic Effects
on Cold and Influenza-Like Symptom
Incidence and Duration in Children.
Pediatrics. 2009; 124: 172-e179.
[16]. Ebtissam SA. Probiotics: An Overview
and their Role in Inflammatory Bowel
Disease. The Saudi J Gastroenterology.
2007; 13: 150-152.
[17]. Jassawala MJ. Probiotics and WomenÊs
Health. J Obstet Gynecol India. 2007;
57: 19-21.
[18]. Robert JB, Roy MRB, Mimi LKT.
Probiotic use in clinical practice: what
are the risks? Am J Clin Nutr. 2006; 83:
1256-1264.
[19]. Gregor RG, Bruce AW. Could probiotics
be an option for treating and preventing
urogenital infections? Medscape
General Medicine (MedGenMed on the
internet). 2001. (Available from:
http:/www.medscape.com/viewarticle/40
8951).
[20]. Marteau P, Cuillerier E, Meance S,
Gerhardt MF, Myara A, Bouvier M,
et al.
Bifidobacterium animalis.
strain DN-
173010 shortens the colonic transit time
in healthy women: a double-blind,
randomised, controlled study. Aliment
Pharmacol Ther 2002; 16: 587-93.
[21]. Food and Agriculture Organisation of the
United Nations and World Health
Organisation 2001. posting date.
Regulatory and clinical aspects of dairy
probiotics. Food and Agriculture
Organisation of the United Nations and
World Health Organisation Expert
Consultation Report. Food and
Agriculture organisation of the United
Nations and World Health Organisation.
Working group Report.
[22]. Cassaro M, Rugge M, Gutierrez O,
Leandro G, Graham DY, Genta RM.
Topographic Patterns of Intestinal
Metaplasia and Gastric Cancer. Am J
Gastroenterol. 2000; 95: 1431-38.
[23]. Glenn RG and Marcel BR. Dietary
Modulation of the Human Colonic
Microbiota: Introducing the Concept of
Prebiotics. J Nutr. 1995; 125: 1401-
1412.
Kaur
et al
. International Journal of Drug Delivery 6 (4) 329-338 [2014]
PAGE | 337 |
[24]. Wullt M, Hagslatt ML, Odenholt.
Lactobacillus plantarum 299v for the
treatment of recurrent Clostridium
difficile-associated diarrhoea: a double-
blind, placebo-controlled trial. Scand J
Infect Dis. 2003; 35: 365-7.
[25]. McFarland LV, Surawicz CM, Greenberg
RN, Fekety R, Elmer GW, Moyer KA
et
al
. A randomised placebo-controlled trial
of Saccharomyces boulardii in
combination with standard antibiotics for
Clostridium difficile disease. JAMA;
1994: 271: 1913-1918.
[26]. Adams MR and Hall CJ. Growth
inhibition of food-borne pathogens by
lactic and acetic acids and their
mixtures. Int J Food Sci Technol. 1988;
23: 287-292.
[27]. Christine VS, Rohan GK, I RB. The
effect of Food Preservatives on pH
Homeostasis in Escherchia coli. J Gen
Microbiol. 1984; 130: 2845-2850.
[28]. Salminen S, Bouley C, Boutron RMC,
Cummings JH, Franck A, Gibson GR
et
al
. Functional food science and
gastrointestinal physiology and function.
Br J Nutr. 1998; 80: 147-71.
[29]. Solauri E, Majamaa H, Arvola T
et al
.
Lactobacillus casei GG reverses
increased intestinal permeability induced
by cow milk in suckling rats.
Gastroenterol. 1993; 105: 1643-50.
[30]. Mosmann TR, Cherwinski H, Bond MW,
Giedlin MA, Coffman RL. Two types of
murine helper T cell clone. I. Definition
according to profiles of lymphokine
activities and secreted proteins. J
Immunol 1986; 136: 2348-57.
[31]. Perdigon G, de Macias ME, Alvarez S,
Oliver G, de Ruiz Holgado AA. Effect on
per orally administered lactobacilli on
macrophage activity in mice. Infect
Immun. 1986; 53: 404-10.
[32]. Perdigon G, de Macias ME, Alvarez S,
Oliver G, de Ruiz Holgado AA. Systemic
augmentation of the immune response
in mice by feeding fermented milks with
Lactobacillus casei and Lactobacillus
acidophilus. Immunol. 1988; 63:17-23.
[33]. Sutas Y, Soppi E, Korhonen H, Syvaoja
EL, Saxelin M
et al
. Suppression of
lymphocyte proliferation in vitro by
bovine caseins hydrolysed with
Lactobacillus GG-derived enzymes. J
Allergy Clin Immunol. 1996; 98: 216-24.
[34]. Sutas Y, Hurme M, Isolauri ES. Down
regulation of anti-CD3 antibody-induced
IL-4 production by bovine caseins
hydrolysed with Lactobacillus GG-
derived enzymes. Immunol. 1996; 43:
687-9.
[35]. Fedork N and Madsen KL. Probiotics
and Prebiotics in gastrointestinal
disorders. Curr Opin Gastroenterol.
2004; 20: 149-55.
[36]. Isolauri E, Sutas Y, Kankaanpaa P,
Arvilommi H, Salminen S. Probiotics:
effect on immunity. Am J Clin Nut. 2001;
73: 444S-50S.
[37]. Hu FB, Stampfer MJ, Manson JE, Rimm
EB Wolk A, Colditz GA
et al
. Dietary
intake of alpha-linolenic acid and risk of
fatal ischemic heart disease among
women. Am J Clin Nutr. 1999; 69: 890-7.
[38]. Castagliuolo I, LaMont JT, Nikulasson
ST, Pothoulakis C. Saccharomyces
boulardii protease inhibit clostridium
difficile toxin A effects in the rat ileum.
Infect Immun. 1996; 64: 5225-32.
[39]. Abdelali H, Cassand P, Soussotte V,
Daubeze M, Bouley D, Narbonne JF.
Effects of dairy products on initiation of
precursor lesions of colon cancer rats.
Nutr Cancer. 1995; 24: 121-132.
[40]. Goldin BR, Gorbach SL. Effects of
Lactobacillus acidophilus dietary
supplements on 1,2-dimethylhydrazine
dichloride-induced intestinal cancer in
rats. J Natl Cancer Inst. 1980; 64: 263-
265.
[41]. Reddy BS, Rivenson A. Inhibitory effects
of Bifidobacterium longum on colon,
mammary and liver carcinogenesis
induced by 2-amino-3-methylimidazo-
[4,5-f]quinoline, a food mutagen. Cancer
Res. 1993; 5: 3914-3918.
[42]. Challa A, Rao DR, Chawan CB,
Shackleford L. Bifidobacterium longum
and lactulose suppress azoxymethane
induced aberrant crypt foci in rats.
Carcinogenesis. 1997; 18: 517-521.
[43]. Rowland IR, Rumney CJ, Coutts JT,
Lievense LC. Effect of Bifidobacterium
longum on gut bacterial metabolism and
carcinogen-induced aberrant crypt foci in
rats. Carcinogenesis 1998; 19: 281-285.
[44]. Bolognani F, Rumney CJ, Pool-Zobel
BL, Rowland I.R. Effect of lactobacilli,
bifidobacteria and inulin on the formation
of aberrant crypt foci in rats. Eur J Nutr.
2001; 40: 293-300.
[45]. Oberreuther Moschner DL, Jahreis G,
Rechkemmer G, Pool-Zobel BL. Dietary
intervention with the probiotics
Lactobacillus acidophilus 145 and
Bifidobcaterium longum 913 modulates
the potential of human faecal water to
induce damage in HT29clone 19A cells,
Brit J Nutr. 2004; 91.
[46]. Hoyos AB. Reduced incidence of
necrotizing enterocolitis associated with
enteral administration of Lactobacillus
acidophilus and Bifidobacterium infantis
to neonates in an intensive care unit. Int
J Infect. Dis. 1999; 3: 197-202.
[47]. Stricker T and Braegger CP. Oral
probiotics prevent necrotizing
enterocolitis. J Pediatr Gastroenterol
Nutr. 2006; 42: 446-7.
[48]. Wang Y, Gao I, Zhang YH, Shi CS, Ren
CM. Efficacy of probiotic therapy in full
term infants with critical illness. Asia Pac
J Clin Nutr. 2014; 23: 575-80.
[49]. Reid G and Bocking A. The potential for
probiotics to prevent bacterial vaginosis
and preterm labor. Am J Obstet
Gynecol. 2003; 189: 1202-8.
[50]. Anukam KC, Osazuwa EO, Reid G.
Improved appetite of pregnant rats and
increased birth weight of newborns
following feeding with probiotic
Lactobacillus rhamnosus GR-1 and L
fermentum RC-14. J Appl Res. 2005; 5:
46-52.
Kaur
et al
. International Journal of Drug Delivery 6 (4) 329-338 [2014]
PAGE | 338 |
[51]. Kalliomaki M, Salminen S, Poussa T,
Arvilommi H, Isolauri E. Probiotics and
prevention of atopic disease: 4-year
follow-up of a. randomised placebo-
controlled trial. Lancet. 2003; 361: 1869-
71.
[52]. Kankaapa PE, Yang B, Kallio HP,
Isolauri E, Salminen SJ. Influence of
probiotic supplemented infant formula on
composition of plasma lipids in atopic
infants. J Nutr Biochem. 2002; 13: 364-
369.
[53]. Sudha MR., Chauhan P., Dixit K., Babu
S., Jamil K. Probiotics as
complementary therapy for
hypercholesterolemia. Biol and Med.
2009; 1: 1-13.
[54]. Kalliomaki M, Salminen S, Arvilommi H,
Kero P, Koshkinen P, Isolauri E.
Probiotics in primary prevention of atopic
disease: a randomied placebo-controlled
trial. Lancet. 2001; 357: 1076-9.
[55]. Tynkkynen S, Singh KV, Varmanen P.
Vancomycin resistance factor of
Lactobacillus rhamnosus GG in relation
to enterococcal vancomycin resistance
(Van) genes. Int J Food Microbiol. 1998;
41: 195-204.
[56]. Pletincx M, Legein J, Vandenplas Y.
Fungemia with Saccharomyces boulardii
in a 1-year old girl with protracted
diarrhea. J Pediatr Gastroenterol Nutr.
1995; 21: 113-115.
[57]. Lee HY, Park JH, Seok SH, Baek MW,
Kim DJ
et al.
Human originated bacteria,
Lactobacillus rhamnosus PL60, produce
conjugated linoleic acid and show anti-
obesity effects in diet-induced obese
mice. Biochimica et Biophysica Acta.
2006; 1761: 736-744.
[58]. Rijinders BJ, Van Wijngaerden E,
Varwaest C, Peetermans WE.
Saccharomyces fungemia complicating
Saccharomyces boulardii treatment in a
non immunocompromised host.
Intensive Care Med. 2000; 26: 825.
[59]. Rautio M, Jousimies-Somer H, Kauma
H, Pieterinen I, Saxelin M, TYnkkynen S
et al
. Liver abscess due to Lactobacilli
rhamnosus strain indistinguishable from
L. rhamnosus strain GG. Clin Infect Dis.
1999; 28: 1159-60.
[60]. Horwitch CA, Furseth HA, Larson AM,
JOnes TL, Olliffe JF, Spach DH,
et al
.
Lactobacillemia in three patients with
AIDS. Clin Infect Dis. 1995; 21: 1460-2.
[61]. Saxelin M, Chuang NH, Chassy B,
Rautelin M, Makela PH, Salminen S
et
al
. Lactobacilli and bacteremia in
southern Finland, 1989-1992. Clin Infect
Dis. 1996; 22: 564-6.
[62]. Piarroux R, Millon L, Bardonnet K,
Vagner O, Koeing H. Are
Saccharomyces yeasts harmful to
patients? Lancet. 1993; 353: 1851-2.
[63]. Hennequin C, Kauffmann-Lacroix C,
Jobert A, Viard JP, Ricour C, Jacquemin
JL
et al
. Possible role of catheters in
Saccharomyces boulardii fungemia. Eur
J Clin Microbiol Infect Dis. 2000; 19: 16-
20.
[64]. Wunderlich PF, Braun L, Fumagalli I,
D'Appuzo V, Heim F, Karly M
et al
.
Double blind report on the efficacy of
lactic acid producing Enterococcus SF68
in the prevention of antibiotic-associated
diarrhoea and in the treatment of acute
diarrhoea. J Int Med Res. 1989; 17: 333-
8.
[65]. Mitra AK and Rabbani GH. A double-
blind, controlled trial of Bioflorin
(Streptococcus faecium S68) in adults
with acute diarrhoea due to Vibrio
Cholerae and enterotoxigenic
Escherchia choli. Gastroenterol. 1990;
99: 1149-52.
[66]. McConnell MA, Mercer AA, Tannock
GW, Transfer of plasmid pAMb1
between members of the normal
microflora inhabiting the murine
digestive tract and modification of the
plasmid in a Lactobacilli reuteri host.
Microb Ecol Health Dis. 1991; 4: 343-55.
[67]. Saavedra JM, Abi-Hanna A, Moore N,
Yolken R. Effect of long term
consumption of infant formulas with
Bifidobacteria and S. thermophilus on
stool pattern and diaper rash in infants. J
Pediatr Gastroenterol Nutr. 1998; 27: 82.
[68]. Hou RC, Lin M, Wang MMC, Tzen JTC.
Increase of viabiity of entrapped cells of
Lactobacillli delbrueckii ssp bulgaricus in
Artificial Sesame oil Emulsions. J Diary
Sci. 2003; 86: 424-8.
[69]. Schrezenmeir J, Vrese M. Probiotics,
prebiotics and synbiotics-approaching a
definition. Am J Clin Nutr. 2001; 73: 361-
4.
[70]. Ming-Ju Chen, Kun-Nan Chen, Chin-
Wen Lin. Optimisation of the viability of
Probiotics in a Fermented Milk Drink by
the Response Surface Method. Asian-
Aust J Anim Sci 2004; 17: 705-11.
[71]. Jacobsson B, Pernevi P, Chidekel L,
Platz-Christensen J. Bacterial vaginosis
in early pregnancy may predispose for
preterm birth and postpartum
endometritis. Acta Obstet Gynecol
Scand. 2002; 81: 1006-1010.
[72]. Susanne Hempel, Sydne J. Newberry,
Alicia R. Maher, Zhen Wang, Jeremy N.
V. Miles
et al.
Probiotics for the
Prevention and Treatment of Antibiotic-
Associated Diarrhea. JAMA 2012; 307:
1959-1969.
[73]. Angelica TV, Mauro MT and Faviano
SM. The role of Probiotics and
Prebiotics in inducing Gut Immunity.
Front Immunol. 2013; 4: 445.
[74]. Hickson M. Probiotics in the prevention
of antibiotic-associated diarrhea and
clostridium difficile infection. Therap Adv
Gastroenterol. 2011; 4: 185-197.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The role of probiotic organisms as alternative or complementary therapy in combating large number of gastro intestinal disorders and their ability to enhance immune response attracts global attention. In addition, their therapeutic use towards cholesterol-lowering activities has further increased their applications as effective probiotics for humans as supplements in milk and yoghurt, since there are no other supplements for hypercholesterolemia, which is the crucial risk factor for cardiovascular diseases. Changes in dietary habits, stressful life and lack of physical activities are the precursors for increasing incidences of hypercholesterolemia and subsequently cardiovascular diseases. The present review focuses on some of the animal studies and clinical trials conducted with probiotic lactobacilli and bifidobacteria. This review may throw some light to prove the ability of these probiotics as a novel alternative or adjuvants to chemical drugs to help fight hypercholesterolemia.
Article
Full-text available
The gut immune system is influenced by many factors, including dietary components and commensal bacteria. Nutrients that affect gut immunity and strategies that restore a healthy gut microbial community by affecting the microbial composition are being developed as new therapeutic approaches to treat several inflammatory diseases. Although probiotics (live microorganisms) and prebiotics (food components) have shown promise as treatments for several diseases in both clinical and animal studies, an understanding of the molecular mechanisms behind the direct and indirect effects on the gut immune response will facilitate better and possibly more efficient therapy for diseases. In this review, we will first describe the concept of prebiotics, probiotics, and symbiotics and cover the most recently well-established scientific findings regarding the direct and indirect mechanisms by which these dietary approaches can influence gut immunity. Emphasis will be placed on the relationship of diet, the microbiota, and the gut immune system. Second, we will highlight recent results from our group, which suggest a new dietary manipulation that includes the use of nutrient products (organic selenium and Lithothamnium muelleri) and probiotics (Saccharomyces boulardii UFMG 905 and Bifidobacterium sp.) that can stimulate and manipulate the gut immune response, inducing intestinal homeostasis. Furthermore, the purpose of this review is to discuss and translate all of this knowledge into therapeutic strategies and into treatment for extra-intestinal compartment pathologies. We will conclude by discussing perspectives and molecular advances regarding the use of prebiotics or probiotics as new therapeutic strategies that manipulate the microbial composition and the gut immune responses of the host.
Article
The fate of the broad-host-range, conjugative plasmid pAMβ1 in the digestive tract of mice was monitored by DNA-DNA hybridisations using a probe containing the ermAM determinant (macrolide-lincosamide-streptogramin type B resistance). Transfer of pAMβ1 from Lactobacillus reuteri to Enterococcus faecium occurred in the digestive tract of infant mice and from E. faecium to E. faecalis and to L. fermentum in the adult intestinal tract. Long-term administration of lincomycin to the animals resulted in a modified form of pAMβ1 in the L. reuteri host. The modified plasmid was present in caecal isolates of the lactobacillus at higher concentration than in the stock culture used to inoculate the mice at the start of the experiment, and was maintained stably by the lactobacilli in vivo . In contrast, pAMβ1 was not stably maintained in the stock strain in the absence of antibiotic administration. The modified plasmid was no longer self-transmissible. Long-term administration of antibiotic to the animals resulted in erythromycin-resistant strains of Escherichia coli and eubacteria. Keywords: Digestive tract; Microflora; Antibiotic; Lactobacillus; Enterococcus; pAMβ1.
Article
This paper provides recommendations, developed by the Working Group (WG) on Probiotics of the ESPGHAN, for the use of probiotics for the prevention of antibiotic-associated diarrhea (AAD) in children based on a systematic review of previously completed systematic reviews and of randomized controlled trials (RCTs) published subsequently to these reviews. The use of probiotics for the treatment of AAD is not covered. The recommendations were formulated only if at least 2 RCTs that used a given probiotic (with strain specification) were available. The quality of evidence (QoE) was assessed using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) guidelines. If the use of probiotics for preventing AAD is considered because of the existence of risk factors such as class of antibiotic(s), duration of antibiotic treatment, age, need for hospitalization, comorbidities, or previous episodes of AAD diarrhea, the WG recommends using Lactobacillus rhamnosus GG (moderate QoE, strong recommendation) or Saccharomyces boulardii (moderate QoE, strong recommendation). If the use of probiotics for preventing C. difficile-associated diarrhea is considered, the WG suggests using S. boulardii (low QoE, conditional recommendation). Other strains or combinations of strains have been tested, but sufficient evidence is still lacking.
Article
Probiotics are nonpathogenic microbes used to confer health benefits to the recipient. The derangement of normal body flora has been held responsible for causation of various disorders. Probiotics have been tried in a number of infective and noninfective disorders and found useful, primarily because of their ability to supplement the normal body flora. Their use offers various advantages over-existing antimicrobial agents in being relatively cheap and safe. However, there are few reports of systemic fungemia associated with their use especially, in immunocompromised and severely debilitated patients. They also carry a risk of transferring resistance to other microbes including pathogens. In conclusion, more studies are required to establish their definite place in therapeutics.
Article
Background: Probiotics are microbial supplements that have shown efficacy in a wide range of applications. To assess the safety and effects of enteral probiotics in critically ill neonates. Methods: A double-blind, randomized controlled trial was conducted in 100 full-term infants with critical illness according to scores of neonatal acute physiology. Fifty neonatal intensive care patients were randomly assigned to receive probiotics three times daily after birth for 8 days, and fifty patients were not given probiotics, but who received a placebo. The incidence of sepsis, multiple organ dysfunction syndrome (MODS), nosocomial pneumonia, and necrotizing enterocolitis were recorded. The prognosis of probiotic treatment was determined based on the rate of recovery and hospital days. Serum IgA, IgG, and IgM concentrations were measured on days 4 and 8. Results: Infants in the probiotics group showed a significantly reduced rate of nosocomial pneumonia (18% versus 36%) and multiple organ dysfunction syndrome (6% versus 16%) compared with the placebo group (p<0.05). Significant results were demonstrated in favour of the probiotics for days of hospital stay (13 ± 3.5 d versus 15.8 ± 5.3 d) (p<0.05). However, there were no significant differences in the occurrence of sepsis, necrotizing enterocolitis, and recovery rate. Patients given probiotics had significantly greater levels of IgA than those in the placebo group (p<0.05). No serious adverse effects in the study population were noted. Conclusions: Supplements of probiotics to critically ill neonates could enhance immune activity, decrease occurrence of nosocomial pneumonia and MODS, and reduce days in hospital.
Article
Growth promoters were added to skim milk to retain the viability of Lactobacillus acidophilus and Bifidobacterium longum to help the product meet the “therapeutic minimum” at the time of consumption. The experiments were divided into two parts. The first part of the study used chicory inulin, isomalto-oligosaccharides and sucrose to investigate the effects of sugars on the activity of L. acidophilus and B. longum. The results indicated that the addition of isomalto-oligosaccharides stimulated growth of L. acidophilus and B. longum, resulting in a higher level of the probiotics after one month storage and yielded better -galactosidase activity during fermentation. The second part studied the effects of three growth promoters on the viability of the probiotic cultures and the response surface method was employed to find the optimal ratio for addition of the growth promoters. The optimal ratio for added calcium gluconate, sodium gluconate and N-acetylglucosamine in fermented milk drinks were established. The response surface method proved to be a very effective way of optimizing the activity of probiotic cultures when developing a new fermented milk drink.
Article
Probiotics constitute an alternative to prevent or treat antibiotics-associated diarrhea (AAD). According to WHO, probiotics are "live microorganisms which, when administered in adequate amounts, confer a health benefit on the host". Recent meta-analyses, including adult and pediatric in- and outpatients, outlined the lack of good quality studies and of the lack of evaluation of side-effects. These meta-analyses showed a positive effect of probiotics in AAD, nevertheless without identifying patients who would benefit the most of them. Severe side-effects have been described in immunocompromised patients, those with abnormalities in their intestinal barrier, and those with central venous catheters.